Header Image
3. Physical ADHD risk factors

Sitemap

3. Physical ADHD risk factors

Author: Ulrich Brennecke
Review: Dipl.-Psych. Waldemar Zdero

Stressful physical or psychological childhood experiences can be a contributory cause of ADHD.

For infants:
Bottle feeding increases the risk of ADHD, while breastfeeding reduces the risk. Crying babies, feeding and sleeping problems in infants, subependymal pseudocysts and antihistamines in the first years of life increase the risk of ADHD.

For infants and children:
Exposure to passive smoking, air pollution (especially particulate matter and nitrogen oxides) and various pollutants such as lead, manganese or phthalates is associated with an increased risk of ADHD.
Surgical procedures under anesthesia, neurodermatitis, bacterial infections, concussions are just as ADHD risk factors as stressful psychological childhood experiences such as trauma, chronic stress or growing up in a home.
Poor bonding behavior of the mother or parents in childhood, stress of the mother in childhood or mental health problems of the parents increase the risk of ADHD in children, as do low socio-economic status, a low level of education or unemployment of the parents.
Earlier school entry and the young age of a child within a class are further risk factors.

During puberty:
A high level of stress during puberty is considered a risk factor for the persistence of ADHD into adulthood.

The % values indicate how much higher the ADHD risk increase correlates with the respective factor. The figures do not indicate that the respective factor is a causal factor.

3.1. Newborns, infants

3.1.1. Screaming children (up to + 1181 %)

3.1.1.1. Increased risk of ADHD in crying children

Cry babies have a significantly increased risk of ADHD[{Schmid, Wolke (2014): Preschool regulatory problems and attention-deficit/hyperactivity and cognitive deficits at school age in children born at risk: different phenotypes of dysregulation? Early Hum Dev. 2014 Aug;90(8):399-405. doi: 10.1016/j.earlhumdev.2014.05.001. n = 1120}}1 The risk of developing hyperactivity at the age of 8 to 10 years was 11.8 times higher (plus 1181%). Behavioral problems and a negative emotional orientation were reported twice as often as in unaffected individuals.2

3.1.1.2. Factors that increase the risk of crying children

If the parents are heavy smokers, or if the mother smokes during pregnancy, the risk of a crying child increases by 30 to 150% (several studies); the largest study on this (n = 5845) states a 69% increased risk.3
In addition, there are several other possible causes that should be systematically ruled out.4

Details Screaming children

With a usual prevalence of 5 to 10 % for ADHD (all subtypes), an 11.8-fold increase in risk would mean that 60 to 100 % of all crying children develop a form of ADHD.
In addition, studies report that (former) crying children with 3.5 years of age have more frequent behavioral problems according to their mothers, but no problems with attention span, behavior regulation and sociability.5 Crying children cause considerable stress for their parents. 5.6% of all crying children bring their parents to the point of abuse and neglect, including serious bodily harm (shaking, hitting).3

This is evidence of the considerable stress experienced by the baby with ADHD over and above the actual cause that makes them cry. A self-reinforcing system develops: the child’s stress causes crying, which causes stress in the parents, which in turn increases the child’s stress.

Crying is not currently considered a separate, first symptom of ADHD.

3.1.2. Febrile convulsions in premature babies (+ 540 %)

Febrile convulsions increased the risk of ADHD in premature infants 6.4-fold and the risk of ASD 16.9-fold.6

3.1.3. Bottle feeding increases (up to +200%), breastfeeding reduces ADHD risk (-23% to -74%)

The % figures were calculated without the highest and lowest values.

The WHO recommends exclusively breastfeeding newborns for the first 6 years.7
A cohort study in Taiwan found that of Taiwanese mothers7

  • 82.1 % breastfed after delivery
  • the average duration of breastfeeding was 2 months (59 days)
  • 26 % breastfed for 3 months
  • 9.8 % breastfed for 6 months
  • 21.9% have ever exclusively breastfed their child (average duration of breastfeeding here 125 days)
  • only 3.8% adhered to the WHO recommendation of exclusive breastfeeding for up to 6 months

Infants who were not breastfed showed an increased risk of ADHD as children, while children who were breastfed as infants showed a decreased risk of ADHD 8 9 10 1112

  • 3.71-fold ADHD risk (+ 271 %, OR = 3.71; meta-analysis of k = 11 studies).13
  • almost 3 times the risk of ADHD (+ almost 200%)14
  • approx. 1.55-fold ADHD risk (+ 55 %, OR = 1.55; METASTUDY)15

Children with ADHD were 1.51 times more likely to be exclusively breastfed for less than 3 months and 52% less likely to be exclusively breastfed for more than 3 months. Children with ADHD had a significantly (2.44 months) shorter duration of breastfeeding (SMD: Hedges’ g = - 0.36) (meta-analysis of k = 11 studies). 13

In breastfed children (without distinction as to whether exclusively or partially and regardless of duration), the risk of ADHD was

  • by 83 %16 and for major depression by 74 %, and for anxiety disorders by 4 %. The effects on ASD, bipolar Disorder and schizophrenia were not significant.
  • reduced by 74 % (OR = 0.263, small study)17
  • by 30 % (meta-analysis, k = 12, n = 106,907).18
  • reduced by 28% (raw value) to 18% (adjusted value) by the age of 57 Since ADHD is usually diagnosed later, the impact is likely to be much greater,
  • unchanged19

Children who were exclusively breastfed had a higher risk of ADHD compared to children who were exclusively bottle-fed,

  • reduced by 38% with exclusive breastfeeding for at least 6 months, after adjustment for potential confounding factors20
  • hyperactivity reduced by 35% at 8 years of age if breastfeeding was exclusive for at least 3 months21
  • reduced by 18 %22
  • reduced by 7 %23
  • unchanged ADHD symptoms at the age of 1621

Exclusive breastfeeding for more than 6 months increased the risk of ADHD by 15%.7

Partially breastfed children had a

  • aDHD risk reduced by 44% at the age of 4 years, after breastfeeding for more than 12 weeks24
  • aDHD symptoms reduced by 32% at age 16 if breastfed for at least 6 months21
  • hyperactivity reduced by 24% at 8 years, but not statistically significant25
  • aSS risk reduced by 11 % 22
  • aDHD risk reduced by 9 %22
  • improved executive functions at the age of 4 years (increase of 4.9 points after breastfeeding for more than 20 weeks)24
  • improved social skills at the age of 4 years (43% reduced risk of being in the worst 20% after breastfeeding for more than 12 weeks)24

The risk of ADHD decreases with the duration of breastfeeding2627 28 (25-year observational cohort study, n = 16,365).29. One study found no significant values for ADHD.30
The risk decreased by

  • 20% if breastfed for more than 1 month compared to less than 1 month (meta-analysis, k = 12, n = 106,907).18
  • 33% if breastfed for more than 3 months compared to less than 3 months (meta-analysis, k = 12, n = 106,907).18
  • 50% if breastfed for more than 6 months compared to less than 6 months (meta-analysis, k = 12, n = 106,907).18
  • 55% if breastfed for more than 12 months compared to less than 12 months (meta-analysis, k = 12, n = 106,907).18

Each additional month

  • the duration of breastfeeding reduced the risk of ADHD by 8%20
  • of exclusive breastfeeding (without complementary food) reduced the risk of ADHD by approx. 8 % (but not statistically significant)20

Prolonged breastfeeding improved cognitive performance,30

Complementary food at the age of 6 months

  • Complementary food up to 6 months reduced the risk of ADHD by 28%7
    • by 6 % with fruit/vegetable juice (not statistically significant)
    • by 14% with fruit/vegetable porridge (not statistically significant)
    • by 19 % with traditional rice porridge (Taiwan) (not statistically significant)
    • by 27% with rice-based diets without traditional rice porridge (Taiwan)
    • increased by 8 % with solid food made from wheat (not statistically significant)
  • had no influence2220

The risk of ASA was reduced by breastfeeding

  • by 36 %22
  • significantly due to prolonged breastfeeding30
  • not significant16

Breast milk contains many substances that are essential for the development of babies, such as

  • polyunsaturated fatty acids.10
  • Prebiotics31
    • Oligosaccharides
    • promote the growth and activity of beneficial microorganisms

The substances contained in breast milk promote31
- antimicrobial active ingredients
- bacterial growth
- secreted IgA for a regulatory immune system

Stilen in the first three months of life influences the intestinal microbiotics:31

  • increases the frequency of
    • Lactobacillus (Bacillota strain)
    • Bifidobacterium
    • Enterococcus
    • Corynebacterium
    • Propiobacterium
    • Streptococcus
    • Sneathia
  • reduces the frequency of
    • Bacteroides (strain Bacteroidota)
    • Staphylococcus

Bottle-fed infants show a different gut microbiome:31

  • increased frequency of
    • Atopobium (strain Actinomycetota)
    • Bacteroides (strain Bacteroidota)
    • Bilophila (strain Thermodesulfobacteriota)
    • Enterobacter (strain Pseudomonadota)
    • Escherichia (strain Pseudomonadota)
    • Citrobacter (strain Pseudomonadota)
    • Clostridium (Bacillota strain)
    • Enterococcus (Bacillota strain)
    • Lactobacillus (Bacillota strain)
    • Granulicatella (strain Bacillota)

More on the influence of the gut-brain axis on ADHD at Gut-brain axis and ADHD

Bisphenol A is known to increase the risk of ADHD. Exposure to PAHs, particularly bisphenol A (BPA), is associated with ADHD symptoms in primary school children and a significant reduction in the volume of the caudate nucleus3233 Bisphenol A was much more common in baby bottles in 2007 than in 2011, which could explain why a study found a five-fold increase in the risk of ADHD in children fed by bottle in 2007, but no increase in risk in children fed by bottle in 2011.34

After all, breastfeeding has its own influence on the well-being and positive development of children, independent of food intake.
Maternal care is the infant’s first social experience and has a decisive influence on its survival, proper development and social skills throughout life.
Infants and puppies do not yet have a high inherent value for their respective mothers at birth.35. Breastfeeding in humans and suckling puppies in animals are rewarding and reinforcing stimuli that promote maternal behavior and bonding. Suckling puppies activates the mesocorticolimbic dopamine system in mothers, which reinforces bonding and nurturing behavior. The extracellular dopamine levels in the nucleus accumbens (the reward center of the brain) increase and correlate with the time spent caring for the offspring.

3.1.4. First-born status (+ 31 to + 100 %)

The risk of ADHD in first-born children was

  • By 200% compared to children with younger and older siblings, according to a large Spanish registry study.36
  • Increased by almost 100 % compared to non-firstborns37
  • Increased by 70 % if there was only one (1) sibling38
  • Increased by 35% according to a Spanish study of n = 1,104 primary school children39
  • According to the German KiGGS study (n = 13,488), the risk of ADHD was 31% higher for first-born children compared to last-born children and children without siblings.40
  • Increased by 22 %38
  • A very large Swedish registry study found an increased risk of ADHD and depression for firstborns.41
  • An Indian study found a high proportion of first-born children among people with ADHD42
  • A study of Ethiopian students found a significant ADHD risk increase for first-time mothers43 and explained this conclusively by increased perinatal risk factors and birth-related complications, which are a major contributor to ADHD and are so much more common in first-time mothers that they were labeled high-risk by the authors:44
  • low birth weight 57% more frequently (evidence that this increases the risk of ADHD)
  • Emergency caesarean sections 57 % more frequent
  • pregnancy-induced high blood pressure 15.38 % more frequently
  • intrauterine growth retardation 19.23 % more frequently
  • premature labor 9.61 % more often
  • fetal distress 19.23 % more frequent
  • Oligohydraminos 17.30 % more frequent

It remains to be seen whether there are other risk factors.

No influence of first-born status on the risk of ADHD found

  • a study of n = 598 children and adolescents with ADHD from relatively large families (more than 47% had at least 4 siblings)45
  • an Iranian study on n = 400 children46,
  • a study of n = 387 Iranian students.47
  • two smaller studies (n = 100 and n = 32)48
  • a smaller study (n = 173)49

First-born status increased the risk

  • By 45 % for aggressive behavior. The risk was even higher if there were older siblings.50
  • By 28 % for severe externalizing symptoms51

Children with ASD had a significantly reduced risk of ADHD if they had an older sibling52

One study did not ask about first-born status, but about the presence of siblings (at 18 months, 3 years and 5 years of age). The siblings could be older and or younger. Among the newborns, about 50.4% were their mothers’ first child and 38.4% were their second child. The remaining newborns were other children.53
Siblings reduced the risk of ADHD by 35%.7

3.1.5. Type of complementary food (wheat + 8 %, rice - 26 %)

Complementary food at the age of 6 months

  • Complementary food up to 6 months reduced the risk of ADHD by 28%7
    • by 6 % with fruit/vegetable juice (not statistically significant)
    • by 14% with fruit/vegetable porridge (not statistically significant)
    • by 19 % with traditional rice porridge (Taiwan) (not statistically significant)
    • by 26% with rice-based diets without traditional rice porridge (Taiwan)
    • increased by 8 % with solid food made from wheat (not statistically significant)
  • had no influence2220

3.1.6. Feeding problems with infants

Feeding problems in infants correlate with ADHD in adolescence and adulthood.1
Frequently skipped meals correlated with an increased risk of ADHD.12

3.1.7. Sleep problems in infants

Sleep problems in infants correlate with ADHD in adolescence and adulthood.1

3.1.8. Subependymal pseudocysts

Subependymal pseudocysts in newborns increase the risk of ADHD and autism.54

3.1.9. Valproate

Studies in mice suggest that valproate administration in newborns could cause permanent damage similar to that caused by ASA and, in some cases, ADHD.55

3.1.10. D-3 insufficiency in the first 12 months

A vitamin D3 level of less than 25 ng/ml in the first 12 months of life was dose-dependently associated with an increased risk of ADHD in childhood. This also applied to ASD and emotional behavioral disorders56
This is consistent with the findings of an increased risk of ADHD if the mother has a D3 deficiency during pregnancy (see there).

3.1.11. Uncontrolled metabolic decompensation

Uncontrolled metabolic decompensation, e.g. as a result of MCAD deficiency (Medium-Chain Acyl-Coenzyme A Dehydrogenase Deficiency), increases the risk of missing developmental milestones and developing aphasia and ADHD.57

3.2. Children

3.2.1. Infectious diseases

An infection is the entry of microorganisms (viruses, fungi, bacteria, parasites) into an organism and their colonization and multiplication.
In a broader sense, infectious diseases are also referred to imprecisely as “infections”.58

3.2.1.1. Infections in the first year of life (+ 16 %)

A register-based cohort study (n = 2,885,662 of which n = 1,864,660 were full siblings) found a slight increase of 16% in the risk of ADHD due to infections in the child in the first year of life (ASD + 12%; tics + 12%, mental retardation + 63%), which was lost when twins were controlled for ADHD and tics.59

3.2.1.2. Bacterial infections (up to + 593 %)

Severe bacterial infectious diseases in childhood or adolescence massively increase the risk of severe mental disorders (HR):60

  • ASS: 13,80
  • ADHD: 6.93
    • ADHD medication use: 11.81
  • Tic Disorder: 6.19
  • OCD: 3.93
  • Bipolar Disorder: 2.50
  • Depressive disorders: 1.93
    • Antidepressant intake: 2.96
    • Mood stabilizer intake: 4.51
    • Atypical antipsychotic use: 4.23
  • Schizophrenia61

Of the bacterial species examined (streptococci, staphylococci, pseudomonas, klebsiella, haemophilus, mycoplasma, tuberculosis, meningococci, escherichia, chlamydia and scrub typhus), streptococci were associated with the most disorders. ADHD was associated with eight bacterial pathogen infections.60

The prevalence of Streptococcus agalactiae infection (Group B Streptococcus, GBS) in infants was 0.07%.
GBS has brought about:62

  • increased infant mortality (19.41-fold)
  • long-term neurological developmental disorders (3.49-fold)

GBS meningitis increased the risk of62

  • ADHD
  • cerebral palsy
  • Epilepsy
  • Hearing impairment
  • profound and specific developmental disorders
3.2.1.2.1. Bacterial meningitis (meningitis) (+ 40 % to + 180 %)

Consequences, children with bacterial meningitis (but not enteroviral meningitis) had an increased risk of ADHD or ADHD medication use:63

  • Meningitis in the first 90 days of life:
    • ADHD risk 2.8-fold (aHR 2.8, + 180 %)
    • ADHD medication intake 2.2-fold
    • ASS risk 1.9-fold
    • Behavioral and emotional disorders 2-fold
    • Learning and intellectual development disorders 4.2-fold
  • Meningitis between day 90 and 18 years:
    • ADHD risk 1.4-fold (+ 40 %)
    • ADHD medication intake 1.5-fold
    • Learning and intellectual development disorders 1.5-fold
3.2.1.2.2. Antibiotics in the first years of life (up to + 33 %)

In a very large study, antibiotic administration in the second year of life increased the risk of ADHD by 20 to 33 % and of sleep problems by 24 to 50 %.64 An even larger study from Korea found a 10% dose-dependent increase in the risk of ADHD, with prenatal and early childhood administration together further increasing the risk65
A smaller study found more frequent behavioral difficulties and depressive symptoms in 3 1/2 year old children who had received antibiotics in the first year of life.66 Two other studies found no increased risk of mental disorders when antibiotics were given in the first 1 67 to 268 years of life.
One study found no increase in risk for ADHD, but for asthma, food allergies and allergic rhinitis,69 which are known to be associated with an increased risk of ADHD.

The administration of antibiotics in the first three years of life reduces the diversity, stability and composition of the microbiome:31

  • reduced frequency of genera
    • Bacteroides
    • Bifidobacterium
    • Lactobacillus
    • Staphylococcus
    • Sediminibacterium
  • increased frequency of
    • Members of the Enterobacteriaceae family
    • of the genus Enterococcus

During the maturation of the microbiome, several processes of the central nervous system take place, such as synaptogenesis, myelination and synaptic pruning, which can be influenced by microbiome-associated metabolites.31

Exposure to antibiotics in the first years of life increased the child’s risk of: (meta-analysis, k = 30, n = 7,047,853)70

  • ADHD: + 33 %
    • not significant in studies that used sibling control groups
  • Depression: + 29 %
    • not significant in studies that used sibling control groups
  • ASS: + 19 %

A cohort study found no significant increase in the risk of ADHD or ASD due to antibiotic administration in the first two years of life.71

3.2.1.3. Viral infections

Powassan flavivirus is transmitted by ticks and in rare cases causes encephalitis in humans. 6 children with Powassan virus encephalitis, diagnosed between the ages of 14 months and 11 years, all suffered permanent neurological sequelae, including72

  • Seizures
  • Movement disorders
  • Behavioral problems
  • ADHD
  • Learning problems
  • States of anxiety
  • Sleep disorders

3.2.2. Craniosynostosis (+ 500 %)

Craniosynostosis is a premature ossification of cranial sutures. Maternal smoking during pregnancy significantly increases the risk of craniosynostosis. In one study, the majority of children with craniosynostosis also showed symptoms of ADHD.73

Around one in two children who had surgery for metopic synostosis (trigonocephaly or metopic suture craniosynostosis) at 9.5 (± 7.9) months of age showed at least borderline hyperactivity and inattention scores at 10.3 (± 3.5) years of age.74 Older age at surgery was associated with poorer executive functions.

3.2.3. Epilepsy (+ 470 %)

Epilepsy is a physical neurological disorder,
One study found a 5.7-fold risk of ADHD in children with epilepsy (41.5 % vs. 7.3 %). ADHD was usually diagnosed in these children after the epilepsy had subsided and the epilepsy medication had been discontinued.75 Another study found ADHD or a learning disorder in 43.3% of children with epilepsy.76

Of children with infection-related epilepsy (FIRES), 33.3% showed symptoms of inattention, 53.3% aggression and 20% mood problems.77

3.2.4. Metals

3.2.4.1. 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.78 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.

3.2.4.2. Lead (+ 160 % to + 310 %)

According to several meta-analyses, lead exposure during development increased the risk of ADHD79 by

  • 433 % in the half of Spanish children (8 to 15 years) with the higher urinary lead exposure.80
  • 310 % increased) for the 20 % with the highest blood lead level compared to those with the lowest 20 %81
  • 239 to 306 %82
  • 160 to 260 %83
  • 95 %84
  • Every doubling of blood lead levels increased the risk of ADHD in children by 35%85 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.86
Elevated lead urine levels correlated with increased inattention and hyperactivity symptoms and reduced IQ in children.87
Children with ADHD had significantly higher lead hair levels than children without ADHD.88

3.2.4.3. Cadmium (+ 269 % ?)

The risk of ADHD was 269% higher in the half of Spanish children (8 to 15 years) with higher urinary cadmium levels.78 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.87 Cadmium correlated negatively with IQ.
0 %: Cadmium exposure during development did not significantly increase the risk of ADHD, according to a meta-analysis84

3.2.4.4. Manganese (+ 163 % to + 257 %)

According to a meta-analysis, exposure to manganese during development increased the risk of ADHD by

  • 163 %89
  • 79 %84
  • Early exposure to manganese causes permanent attention problems via the mTOR pathway and an alteration of the catecholaminergic system90 as well as sensorimotor problems.91
    Manganese intoxication shows a correlation with certain CYP2D6 gene variants92

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.91

In persons with ADHD, elevated manganese levels were only found in the hair, but not in the blood93, another study also found elevated manganese blood levels in schoolchildren with ADHD.94 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%).88

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.95
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.96

In children with ADHD, urinary manganese levels were slightly lower than in children without ADHD.87

An Israeli study addressed the fact that manganese levels in infant formula were often higher than in breast milk and recommended stricter limits.97

3.2.4.5. Antimony (up to + 204 %)

ADHD risk 204% higher in the half of Spanish children (8 to 15 years) with higher urinary antimony exposure.78
In a study of children with and without ADHD, children with ADHD-H/I showed the highest antimony urine levels.87 Antimony urine levels correlated with the severity of ADHD symptoms according to the teacher rating.

3.2.4.6. Mercury (up to + 168 %)

Exposure to methylmercury during development increased the risk of ADHD by 168% (meta-analysis, k = 47)98 One review found an increased risk of ADHD and ASD.99 Two reviews describe causality.10083
Two meta-analyses found no significant increase in risk from mercury.8479
Mercury urine levels were not significantly elevated in children with ADHD.87 Mercury blood levels were unchanged in children with ADHD.85
The correlation between mercury exposure and ADHD appears to be influenced by DNA methylation.101

3.2.4.7. Arsenic (+ 53 %)

According to a meta-analysis, exposure to arsenic during development increased the risk of ADHD by 53%84

Wistar rats given arsenic (50% of the lethal dose, 8 mg/kg) developed hyperactivity.102

3.2.4.8. Zinc

One study found elevated zinc blood levels in school children with ADHD.94

3.2.5. Chemicals

3.2.5.1. Phthalates (+ 212% for girls to + 254% for boys)

According to a meta-analysis, exposure to phthalates during development increased the risk of ADHD by 212% for girls and 254% for boys.89
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.103

3.2.5.2. Perfluorooctane sulfonate (PFOS) (+ 77 %)

Perfluorooctane sulfonate (PFOS) in breast milk correlated with a 77% increased risk of ADHD per higher interquartile range.104
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.105

3.2.5.3. β-Hexachlorocyclohexane (β-HCH) (+ 75 %)

Peβ-hexachlorocyclohexane (β-HCH) in breast milk correlated with a 75% increased risk of ADHD per higher interquartile range.104

3.2.5.4. Chlorinated kerosenes (+ 57 % / quartile)

Chlorinated kerosenes (CP) are widespread environmental and industrial pollutants.
A large study (n = 122,965, including 7,139 with ADHD) examined 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.106

3.2.5.5. Polychlorinated biphenyls (PCBs)

Exposure to polychlorinated biphenyls (PCBs) in childhood correlated with an increased risk of ADHD.83
Prenatal PCB exposure slightly reduced internalizing symptoms by 17% and slightly reduced ADHD risk by 16%.107
Rats that were exposed perinatally to A1221 (a PCB mixture) showed108

  • 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

3.2.6. Gastroesophageal reflux (+ 248 %)

Gastroesophageal reflux disease (GERD) increased the risk of ADHD at 3 to 5 years of age (HR = 3.48) (registry study, n = 631,695).109

It is not clear from the abstract whether it is really about GERD in the children themselves. The full text of the study was not yet available to us.

3.2.7. Passive smoking - smokers in the environment in the first years of life (+ 42 % to + 170 %)

Passive nicotine exposure increases the risk of ADHD.110 Nicotine exposure in children is associated with a 1.42-fold111 to 2.7-fold89 increase in ADHD.27 In one study, children with ADHD were twice as likely to have smokers in the family as children who were not affected.112
In the case of passive smoking, a connection is made to certain MAO-A gene variants that cause a lower level of serotonin degradation.113

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.114

One study found no link between smoke exposure after birth and ADHD.81

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.115 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).

3.2.8. D-3 insufficiency (+ 157 %)

A meta-analysis of 10,334 children and adolescents found a 2.57-fold risk of ADHD with a D3 insufficiency (between 10 and 30 mg/nl in blood plasma).116

3.2.9. Air pollution in childhood

Air pollution consists of a complex mixture of117

  • 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)118

  • Children in the top third of exposure to organic substances showed a 1.51-fold ADHD risk (+51%) compared to the bottom third119

  • Children in the top third of soot exposure showed a 1.29-fold ADHD risk (+29%) compared to the bottom third119

  • Children in the top third of sulfate exposure showed a 1.20-fold ADHD risk (+20%) compared to the bottom third119

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:120

  • Home renovations
  • Incense burning
  • Cooking oil vapors
  • Smokers in the household

Pathways of air pollution on ADHD

Pathways of air pollution on ADHD:117

  • 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
    • Oxidative stress
      • thereby, among other things, increased peripheral noradrenaline121
    • reduced central noradrenaline122
    • Promotion of a parasympathetically dominated state by NO123

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
As particles up to a maximum of 1000 nanometers = 1 micrometer can cross the blood-brain barrier (microplastics up to 200 nanometers124, 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.125 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.126 Estimates that a person ingests 1 to 5 grams of microplastics (plastic up to a maximum of 5 mm) per week127 are criticized as being orders of magnitude too high128. 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 129128

9 % of microplastic dust up to 5 mm in Austria results from tire abrasion.130
Around 4% of the particles inhaled by humans are the result of microplastics, the other 96% are of natural origin.131
Mineral water from plastic bottles did not contain more microplastics than mineral water from glass bottles.128
The proportion of microplastics in cosmetics fell by 97% between 2012 and 2017.128 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.

3.2.9.1. 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.117

A study of single nucleotide polymorphisms associated with air pollutants found132

  • 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 %)
3.2.9.1.1. 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 PM10.133

  • ADHD was found in134
    • 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 tercile135
    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.136 A Taiwanese register study came to comparable results.137
3.2.9.1.2. 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.138139

  • A review of k = 45 meta-analyses found a 1.82-fold ADHD risk (+82%) from PM2.5 exposure.140
  • There is clear evidence of a link between PM2.5 and NO2 levels
    • in early childhood and ASD141
    • in childhood and ADHD141; (meta-analysis; k = 25)142
    • PM2.5 causally increased the risk of ADHD 2.0-fold (+100%)143
    • likewise for PM10 (meta-analysis; k = 25)142
  • Children in the highest quintile of PM2.5 exposure had a 1.70-fold risk of ADHD (+70%) compared to children from the lowest quintile144
  • Children in the top third of PM2.5 exposure showed a 1.37-fold ADHD risk (+37%) compared to the bottom third 119

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.145
A meta-analysis of 12 studies found a correlation between particulate matter and ADHD in children in 9 of them.146

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 oxide147
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.148
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.149

3.2.9.1.3. 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) caused150

  • 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
3.2.9.1.4. 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 as117

  • 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 pathways151
    • 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.152 PAHs influence the gene regulation of NMDAR subunits.
    • PAH disrupt Ca2+ homeostasis, which impairs BDNF signaling153
  • Heavy metals
  • organic and inorganic compounds
  • reactive gases

PM2.5 exposure causes154

  • 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 lungs138
  • hypoxia-related damage
  • Inflammatory reactions

PM2.5 exposure155

  • 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 PM2.5155

  • 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 schizophrenia149

Exposure to particulate matter can117

  • 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

Prenatal exposure to PM caused156157

  • dysregulation of dopamine and its metabolites in various regions of the brain
    • Dopamine increased in the PFC158159
    • Noradrenaline increased in PFC in mice158, unchanged in rabbits159
    • Reduced dopamine and noradrenaline turnover in the striatum
  • altered serotonin159
  • reduced spontaneous motor skills158
  • Impulsiveness changes

3.2.9.2. 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.160

  • Children in the highest third of NO exposure had a 2.1-fold risk of ADHD (+ 110%) compared to children in the lowest tercile135
  • Children in the highest quintile of NO2 exposure had 1.63 times the risk of ADHD (+ 63%) compared to children from the lowest quintile144

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.144 A smaller cohort study confirmed this for particulate matter, but not for nitrogen dioxide161
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 majority162
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 18.163
One study found a 32% increase in the risk of ADHD in children due to a 10 μg/m3 increase in nitrogen oxide.133
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:164
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 [10-700 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%.165 Another study found similar results.166
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 disorders167

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.144
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 decreased168

  • 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 allele169

  • Behavioral problems,
  • a smaller decrease in inattention over time
  • a smaller caudate volume

A Polish study found a correlation between170

  • 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%)171

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.172
Emissions of nitrogen oxides in Germany fell by almost 2/3 between 1990 and 2020.173

Acute NO2 inhalation causes174

  • 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 exposure117

  • reversibly inhibits human alpha-7 nicotinic acetylcholine receptors (α7-nAChRs). α7-nAChRs regulate synaptic neurotransmitter release in the CNS175 and the enhancement of glutamatergic activity that regulates working memory and attention-related pathways in the dlPFC176.
  • Inhibition of α7-nAChR by N2O can trigger oxidative stress, which is characterized by increased NO in peripheral regions121
    α7-nAChR on sympathetic nerves normally increase the release of noradrenaline, which leads to neuronal vasodilation. N2O-mediated inhibition of α7-nAChR thus reduces sympathetic activity, contributing to ADHD117

NO promotes parasympathetic activity.117
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.117

NO2 emissions in the USA have halved from 15 million tons in 2011 to just over 7.5 million tons in 2020117

3.2.9.3. Nitrous oxide (laughing gas)

Increasing exposure to N2O from air pollution could contribute significantly to the development of ADHD118
Environmentally relevant concentrations of N2O, even at trace levels, impair cognitive functions such as working memory in adult males117

3.2.9.4. 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.28

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) 177117
High ozone exposure is a risk factor for neurological diseases.178 Long-term exposure to ozone leads to cognitive impairments such as memory problems or impaired executive functions.179

3.2.10. Hyperthyroidism / hyperthyroidism (+ 70 %)

One study found a 1.7-fold prevalence of ADHD in children with hyperthyroidism.180
Children with ADHD showed

  • significantly higher T4 levels (total thyroxine).181
  • significantly reduced FT4 levels (free thyroxine) and TT3 levels (total triiodothyronine) (meta-analysis, k = 12, n = 11,836)182, children with ASA showed reduced FT4 and unchanged TT3.

Hyperthyroidism can cause ADHD-like symptoms, including anxiety, nervousness, irritability and physical hyperactivity. One study found a correlation between elevated TSH levels and hyperactivity in ADHD.183

For the effects of abnormal thyroid hormone levels during pregnancy, see Prenatal stressors as ADHD environmental causes

3.2.11. Concussions and traumatic brain injuries (+ 68 %)

Craniocerebral trauma can trigger SDHD (secondary ADHD).184
Brain injury severity correlates with significantly higher ADHD symptomatology. Brain injury-altered default mode network (DMN) morphometry predicted higher ADHD symptomatology 12 months after injury, while salience network (SN) and central executive network (CEN) morphometry were not significant independent predictors.185

ADHD correlated with an increased incidence of brain injury trauma186187 188 and otorhinologic trauma.189
One study examined mild (concussion) and severe traumatic brain injuries before the age of 10. The incidence was 1,156 per 100,000 person-years. At the age of 19, the risk of ADHD was increased by 68% and the risk of learning disability by 29%.190
In more severe cases of traumatic brain injury, the correlation was not statistically significant. In an analysis of cases with possible traumatic brain injury (equivalent to a concussion), the result was significant (risk of ADHD increased by 105%, risk of learning disability increased by 42%). The risk in adulthood was particularly increased in children with the least severe injuries.
In pure correlation studies, however, the extent to which craniocerebral injuries are a consequence of ADHD (increased accident risk) or ADHD is a consequence of craniocerebral injuries (secondary ADHD) is an open question.

Among 1,709 ice hockey players aged 11 to 17, the rate of concussions correlated with higher self-reported and parent-reported scores for attention problems. Only self-reported hyperactivity, not parent-reported hyperactivity, also correlated significantly with concussion. A T-score ≥ 60 combining attention problems and hyperactivity scores (an estimate of probable attention deficit hyperactivity disorder) was not significantly associated with incidence of injury or concussion191

A Finnish nationwide retrospective cohort study (1998 to 2018) of pediatric traumatic brain injury patients (n > 126,000) found a significant association between pediatric traumatic brain injury and post-traumatic ADHD medication use during a 20-year follow-up, with the association being particularly pronounced after 4 years.192
Neurodegeneration due to craniocerebral trauma, subsequent neuroinflammation and oxidative stress was assumed to be the pathway of action, which could impair brain development and neurotransmitters and increase the risk of neurodevelopmental disorders.193

3.2.12. Febrile convulsions (+ 28 % to + 66 %)

According to clinical and animal studies, febrile convulsions have harmful effects on neurodevelopment, which can lead to ADHD, increased susceptibility to epilepsy, hippocampal sclerosis and cognitive decline in adulthood194
Febrile convulsions in children increased the risk of ADHD by 28%195 to 66%.196

3.2.13. Nutrition

3.2.13.1. Diet high in sugar (+ 41 %)

A diet high in refined sugar and saturated fats during childhood increased the risk of ADHD by 41%, while a healthy diet reduced the risk of ADHD by 35%. (Meta-analysis, k = 14 studies)197 Compared to a healthy diet, an unhealthy diet therefore increases the risk by 117%.

One study found a correlation between sugar intake at 30 months and the risk of ADHD, sleep disorders and anxiety. No correlation was found at the age of 12 months.198
In an Egyptian study, a child’s daily consumption of sweets correlated with a 6.82-fold risk of ADHD (+ 582%).88 Whether this is a causal cause or a consequence of changed food preferences due to the predisposition to the disorder remains to be seen.

3.2.13.2. Highly processed foods (up to + 25 %)

A high percentage of highly processed foods at age 3 to 4 years increased the risk of ADHD at age 12 to 13 years by 25% (RR 1.25).199

3.2.14. Early puberty (pubertas praecox) (+ 40 %)

Among girls with early puberty (onset of sexual maturation before the age of 8 for girls and before the age of 9 for boys), the prevalence of ADHD was found to be 13.5%.200

3.2.15. Surgical procedures under anesthesia (+ 25 to 39 %)

Anesthesia in childhood correlates with an increased risk of ADHD201
Children who underwent a single surgical procedure under anesthesia up to the age of 5 were 37% more likely to take ADHD medication in later years.202 A Korean cohort study found a 41% increased risk of ADHD as a consequence of general anesthesia in early childhood. In addition, the duration of general anesthesia correlated with an increased risk of ADHD.203
One study found a 37% increase in the risk of ADHD with a single anesthetic during surgery up to the age of 5 years, and a 75% increase with multiple anesthetics204
A cohort study of n = 15,072 children, half of whom had received anesthetics between the ages of 0-3 years, found a 39% increased risk of ADHD.Frequency of anesthetic exposure, duration of exposure, male sex, and central nervous system surgery were significant risk factors for ADHD in the future.205
Other studies have come to similar conclusions.206207
A meta-analysis found a 25% increase in the later risk of ADHD due to general anesthesia in childhood (RR = 1.26).208

  • 38 % (RR = 1.38) due to single general anesthesia of max. 60 minutes in children
  • 55 % (RR = 1.55) by single general anesthesia of max. 61 to 120 minutes or more than 120 minutes in children
  • 61 % (RR = 1.61) after several general anesthetics

In contrast, a cohort study in Taiwan found no increased risk of ADHD due to anesthetics in the first 3 years of life.209

It remains to be seen to what extent the probability of a surgical procedure under anesthesia is already influenced by the increased likelihood of accidents among people with ADHD. For more information, see Consequences of ADHD.

However, there is evidence that general anesthesia using fentanyl and sevoflurane can cause ADHD symptoms in newborn mice, such as ADHD-like behaviors, cognitive impairments, fine motor skill disorders, as well as disrupt neurogenesis and alter gene expression. These could be reduced by prior administration of vitamin K2. K2 administration before general anesthesia led to significantly different gene expression changes210

3.2.16. Atopic disorders (immunoglobulin E deficiency)

In low birth weight infants, a history of T2 inflammatory diseases such as asthma and atopic dermatitis increased the risk of:211

  • ADHD by 81 %
  • Learning disability by 74 %
  • ASS by 47 %
  • mental disability by 35 %

Atopic disorders correlated not only with an increased risk of ADHD, but also with the severity of ADHD symptoms.212

3.2.16.1. Asthma (+ 34 % to + 296 %)

A review (k = 21 meta-analyses, n = 348,405,029) found an increase in the risk of ADHD due to asthma of 34% (OR 1.34) with convincing evidence (evidence class I).213
With highly suggestive evidence (class II) increased

  • Asthma the risk
    • for depression by 64 % (OR 1.64)
    • for anxiety by 95 % (OR 1.95)
    • for tic disorders by 90 % (OR 1.90),
    • for suicidal thoughts by 52 % (OR 1.52)
    • for suicide attempts by 60 % (OR 1.60)
  • allergic rhinitis the risk of
    • for Tic Disorders by 161 % (OR 2.61)
    • for sleep disorders by 171 % (OR 2.17)

Allergic rhinitis increased the risk of ADHD

  • by a factor of 3.96 (+ 296 %, OR 3.96). (METASTUDY, k = 2, n = 132,561)214
  • by a factor of 2.08 (+ 108 %)215
  • by a factor of 1.83 (+ 83 %; meta-analysis, k = 18, n = 4,289,444)216
    • Women 1.86 times as often as men (+ 86 %)
    • Children up to 8 years 1.75 times the risk of older children (+ 75%)
    • ASA: 1.90-fold risk (+ 90 %)

A genetic association study found no causal risk increase for ADHD due to asthma217

ADHD increased the risk of allergic rhinitis

  • correlatively by 1.85-fold (+ 85 %, OR 1.84) (meta-analysis, k = 10, n = 397,799)214
  • correlative by 1.38-fold across all studies (+ 38 %; METASTUDY, k = 18, n = 4,289,444)216
    • Women compared to men by a further + 86 % (meta-analysis, k = 18, n = 4,289,444)216
    • Children up to 8 years compared to older children by a further + 75 % (meta-analysis, k = 18, n = 4,289,444)216
    • correlative by 1.90-fold in cohort study (+ 90 %; meta-analysis, k = 18, n = 4,289,444)216
  • causally by 1.27-fold (+ 27 %) in a genetic association study217

3.2.16.2. Atopic eczema (+ 72 %)

Eczema increased the risk of ADHD 1.72-fold (+72%).215

3.2.16.3. Neurodermatitis / atopic dermatitis (up to + 28 %)

Neurodermatitis / atopic eczema / atopic dermatitis in childhood correlates with an increased risk of ADHD.218
In contrast, a cohort study found no significant (+2%) increased risk of ADHD in childhood atopic dermatitis.219
A cohort study of n = 69,732,807 people found an increased risk of learning difficulties (OR = 1.77) and memory problems (OR = 1.69) with atopic dermatitis.
The increase in risk was unevenly distributed: In children with neurodevelopmental disorders such as ADHD, the risk of memory or learning difficulties was increased 2 to 3-fold. In contrast, atopic dermatitis did not change the risk of learning or memory difficulties in children without neurodevelopmental disorders.220

A review (k = 21 meta-analyses, n = 348,405,029) found an increase in risk in atopic dermatitis with highly suggestive evidence (class II)213

  • for ADHD by 28 % (OR 1.28)
  • for depression by 60 % (OR 1.60)
  • for anxiety by 62 % (OR 1.62)
  • for suicidal thoughts by 44 % (OR 1.44)

One study found evidence that susceptibility to atopic dermatitis causally increased the risk of ADHD (+ 11.6 %) and ASD (+ 13.1 %). Conversely, susceptibility to ADHD (+ 11.2 %) and anorexia nervosa (+ 10 %) caused an increased risk of atopic dermatitis. Only the causal relationship between AD and ASD was independent of the reverse effect bias.221
Urticaria increased the risk of ADHD by 9%222

People with ADHD had a 45% increased risk of atopic dermatitis (OR = 1.45).
People with atopic dermatitis had a 34% to 42% increased risk of ADHD (OR = 1.34; HRs = 1.42), with severe atopic dermatitis the risk of ADHD was increased by 162% (OR = 2.62), with multiple allergic diseases by 189% (OR = 2.89), with atopic dermatitis and concurrent sleep disorders by 143% (ORs = 2.43). (METASTUDY, k = 49)223

3.2.17. Selective immunoglobulin A deficiency (+ 30 %)

Selective immunoglobulin A deficiency correlated with a 30% (OR 1.30) higher risk of ADHD and an increased incidence of respiratory and intestinal infections.224.

3.2.18. Antihistamines in the first years of life

A large cohort study found that taking antihistamines (especially first-generation antihistamines) in the first years of life significantly increased the risk of later ADHD. Disorder of REM sleep was cited as a possible cause, which secondarily impaired brain maturation.225

3.2.19. 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.226

3.2.19.1. Chlorpyrifos

In children between 1 and 6 years of age, chlorpyrifos residues in the blood correlated with the risk of ADHD.227 Vitamin D reduced the risk.
Chlorpyrifos also significantly increases the risk of ADHD in the case of prenatal contamination of the mother during pregnancy.

3.2.19.2. Pyrethroid pesticides

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 function228

The annual number of ADHD cases caused by pyrethroids per million inhabitants was estimated at229

  • 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)89

3.2.19.3. Organophosphates

Exposure to organophosphates in childhood correlated with an increased risk of ADHD.83

One study examined the urine of 186 Taiwanese children with and without ADHD for230

  • 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 pathway231

3.2.20. Lack of sleep

Short sleep correlated with increased risk of anxiety, attention deficit hyperactivity disorder and activity-limiting emotional and psychological states after adjusting for ethnicity, deprivation, age and gender.232
Ethnicity and socioeconomic disadvantage in the neighborhood were independently correlated with short sleep and snoring/noisy breathing during sleep.
Long sleep correlated independently with an increased risk of depression.

3.2.21. Acne vulgaris

One study found moderately elevated levels in adolescents (12 to 17 years) with acne vulgaris for233

  • Hyperactivity
  • Hyperactivity/impulsivity
  • Inattention
  • ADHD total score

Both acne vulgaris and ADHD are thought to be partly caused by increased androgen hormone levels in the womb.

3.2.22. 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.234

3.2.23. Pets

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.235

3.2.24. 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.88 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.

3.2.25. Commercially packaged pasta

A child’s consumption of commercially packaged pasta 3 or more times per week correlated with a 57-fold risk of ADHD.88
Whether this is related to ingredients in commercially packaged noodles in Egypt, food preferences due to ADHD, or a consequence of low family socioeconomic status is an open question.

3.2.26. Unpackaged flour

The use of unpackaged flour in cooking correlated with a 44-fold risk of ADHD in an Egyptian study.88

3.2.27. Loud snoring

A study of n = 512 snoring children and adolescents (70% of whom were overweight or obese and 35% with an elevated obstructive AHI >3/h) found that of the snoring children236

  • 36 % showed hyperactivity/impulsivity
  • 24 % showed emotional dysregulation

This would indicate a 300% increased risk of ADHD.
Surprisingly, the reduced oxygen supply due to sleep apnea did not correlate significantly with hyperactivity/impulsivity or emotional dysregulation, while the measure correlated with the volume of snoring.

3.3. Age-independent physical factors

3.3.1. Toxins (up to + 900 %)

3.3.1.1. Phthalates (+ 200 % to + 900 %)

Higher phthalate metabolites in children’s urine correlated with increased likelihood of ADHD by 3 to 9 times.237
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.238
Boys with ADHD showed significantly elevated levels of mono-n-butyl phthalate and ethyl paraben, whereas girls with ADHD did notNeither boys nor girls showed significant differences in testosterone, free testosterone, FSH, LH, estradiol, progesterone or SHBG compared to controls. Boys with ADHD correlated elevated urinary levels of monobenzyl phthalate and monoethylhexyl phthalate with serum testosterone. In girls with ADHD, urinary levels of monoethyl phthalate correlated positively with serum levels of LH, testosterone and free testosterone.239

3.3.1.2. 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.140

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.240 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 hyperactivity241 and cognitive problems.242 Another study found similar results.243 A review summarizes the results.244
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.245
A smaller long-term study in Sweden found an increased risk of ADHD due to fluorides.246

In Germany, 90% of drinking water has a fluoride content of 0.3 mg/liter. Drinking water is not fluoridated in Germany.247 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.248 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.249

3.3.1.3. Lead (+96 % to +133 %)

Elevated blood lead levels lead to an increased risk of ADHD.250251252253254 With a blood lead level of ≥ 5 μg/dl, the risk of ADHD was found to be 2.33 times higher (OR 2.33).255
A review of k = 45 meta-analyses found a 1.96-fold ADHD risk.140

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.256

Lead influenced the dopamine balance in many studies.

  • Reduced dopamine signaling
    • caused cognitive deficits with delayed spatial alternation, which could be corrected by L-dopa and without L-dopa only ended 8 years after the 2-year lead exposure257
    • in the nucleus accumbens258
  • Increased dopamine signaling
    • in mesolimbic pathways (nucleus accumbens)259
    • Lead increases dopaminergic activity and has been linked to attention deficits, Alzheimer’s disease and increased drug sensitivity.260

The DRD2 gene variant rs1800497r is said to promote a link between ADHD and lead.261 A connection to certain MAO-A gene variants is also mentioned, which causes lower serotonin degradation.113 A study in rats suggests interactions between lead exposure and early stress on the dopaminergic system.262 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 addiction263

In one study, lead altered neostriatal serotonin and norepinephrine levels, increased anxiety and decreased open-field activity.264

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.265
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.96

3.3.1.4. Inorganic arsenic (+ 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).255

3.3.1.5. 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.266

3.3.1.6. Smoking by parents (+ 30 %)

Postnatal smoking by parents correlates with a 1.3-fold risk (increased by 30%)267 of ADHD in offspring.
This could be related to genetic factors, as people with ADHD are significantly more likely to smoke. The co-morbidity of smoking with ADHD is 40%.268 In contrast, around 25 % fewer of the total population smoke, namely 26.9 % of women and 32.6 % of men.269

3.3.1.7. 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.270271

Even low levels of PCB exposure during development impair neurobiological, cognitive and behavioral functions.271
One study found a 26% to 92% increased risk of ADHD.272 Individual studies found contradictory or weak impairments,273274 however, the vast majority show evidence of relevance in ADHD.267275

Polychlorinated biphenyls affect the dopamine system.276 PCBs inhibit dopamine synthesis as well as the storage of dopamine in the vesicles and its release, thereby causing low dopamine levels277253 in the basal ganglia and PFC278279 277 280 281 282 , as well as reduced DAT in the striatum283, which overall corresponds quite closely to the picture of ADHD.

Prenatal exposure to PCBs has an adverse effect:

  • Hyperactivity (in rats even at subtoxic doses)277253
  • IQ, memory, attention 284
  • Memory, attention285
  • Impulsivity (via corpus callosum)286287 in rats even at subtoxic doses277253
  • 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.276
  • no effect on sustained attention273

Possible path of action: Gap junctions288

3.3.1.8. Polyvinyl chloride (PVC)

One review describes a suspected correlation between PVC exposure and ADHD.289

3.3.1.9. Pesticides

With regard to pesticides (especially organochlorine compounds, pyrethroids, organophosphates), there are indications of relevance in ADHD.267253

For pesticides during pregnancy and ADHD, see there.

3.3.1.9.1. Organochlorine compounds

With regard to organochlorine compounds, there are indications of relevance in ADHD.267253290

A study of Greek schoolchildren with ADHD found no elevated blood serum levels of291

  • Dichlorodiphenyltrichloroethane (DDT) Metabolites
  • Hexachlorocyclohexane (HCH) isomers
  • Cyclodienes
  • Methoxychlorine

Other organochlorine compounds are:288

  • Lindane
  • Dieldrin
  • Chlordane, endosulfan, heptachlor, aldrin
  • Chlordecone
  • Mirex

Possible path of action: Gap junctions288

3.3.1.9.2. Organophosphates

According to a large number of studies, organophosphate pesticides have a correlation between prenatal and postnatal exposure and ADHD 271292 293 294 290 295 or a theoretically possible increase in ADHD risk.296 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.113
In contrast, two larger studies found no influence 297298
With regard to hyperactivity, 2 studies found an association between organophosphates and hyperactivity, 4 studies found no association.299
One study found no correlation of chlorpyrifos with hyperactivity in rats300 while another found it in females.301
A study in rats was able to induce ADHD-like behaviors in Wystar 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.302
Blood values were measured in Egyptian adolescents, some of whom used pesticides, and the parents were asked about ADHD symptoms in the adolescents:303 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.295

3.3.1.9.3. 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 304 Other studies also found an association with ADHD305290 , ASD or developmental delay.271
Blood levels were measured in Egyptian adolescents, some of whom used pesticides, and the parents were asked about ADHD symptoms in the adolescents:303 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).

3.3.1.9.4. Carbamate (-)

One review found no associations between carbamates and ADHD.290

3.3.1.9.5. Neonicotinoids (- ?)

One review found no associations between neonicotinoids and ADHD, although there were few studies on this topic.290

3.3.1.10. Mercury / Amalgam (Mercury)

There is weak evidence (= not proven) of relevance in ADHD.267253306
A large study with n = 2073 participants was unable to establish a connection between amalgam and ADHD.307
Mercury is also suspected of being a possible contributory cause of Parkinson’s disease.308 This would be a clear indication of a damaging effect on the dopamine system.

3.3.1.11. Manganese

There is weak evidence of relevance in ADHD, although elevated manganese levels were only found in the hair, but not in blood levels, of people with ADHD.26793
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.96
A doubling of the manganese content in teeth from the prenatal and postnatal period increased the risk of attention problems and ADHD symptoms in the school years by 5%. Manganese from the childhood period showed no influence95
A study reports benefits of choline supplementation during pregnancy in rats to prevent manganese-induced developmental disorders in the offspring309

3.3.1.12. Bisphenol A

Bisphenol A is suspected of increasing the risk of ADHD.253 A connection with certain MAO-A gene variants that cause lower serotonin degradation113 and an influence on the thyroid balance is being discussed.310
A meta-analysis found a clear link between bisphenol exposure and ADD(H)S.311

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.238

Possible path of action: Gap junctions288

3.3.1.13. Perfluoroalkyl compounds

Elevated levels of perfluoroalkyl compounds have been observed in ADHD.312

3.3.1.14. Triclosan

Prolonged exposure to the environmental pollutant triclosan induced ADHD symptoms in rats. Triclosan appears to cause a reduction in dopamine levels in the PFC.313

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.314

3.3.1.15. Synergy effects of neurotoxins

The synergistic effects of neurotoxins must be taken into account:267315

  • Formaldehyde increases the toxicity of mercury.
  • Amalgam increases the toxicity of PCBs and formaldehyde.
  • Mercury and PCBs potentiate each other’s effects.

3.3.2. Craniocerebral trauma (up to + 529 %)

Traumatic brain injuries (TBI) are estimated internationally at 349 per 100,000 person-years. Every second person suffers a traumatic brain injury in the course of their life. Traumatic brain injuries have the highest incidence and prevalence of all common neurological disorders and have been linked in studies to Consequences such as neurodegenerative diseases, cognitive impairment, stroke, psychiatric disorders and increased mortality.316

A review of k = 24 METASTUDY found a correlation of ADHD risk with previous traumatic brain injury:316

  • mild traumatic brain injury: + 18 %, not statistically significant, n = 4,098 subjects
  • moderate traumatic brain injury: + 266 %, not statistically significant, n = 117
  • severe traumatic brain injury: + 529 %, statistically significant, n = 5,092

The extent to which traumatic brain injury in ADHD is the result of the increased likelihood of accident and injury in ADHD remains an open question.
In one study, 30% of boys and 15% of girls with traumatic brain injury had previous ADHD.317 This study also suggests that girls should integrate physical activity and resources for coping with school stress into their rehabilitation program. For boys, cognitive support and strategies for coping with ADHD may be more effective.

See also Concussions and traumatic brain injury in children as a risk for ADHD

3.3.3. Food intolerances, allergies

It is certain that ADHD is not caused by individual, specific foods, phosphates or additives.

However, individual food intolerances or allergies are just as much stressors as illnesses, toxins or psychological stress and can therefore worsen the stress situation of people with ADHD to such an extent that symptoms develop. This is not a finding specific to ADHD. For example, in a group of children with schizophrenia problems, dietary treatment of an existing gluten intolerance was able to eliminate the schizophrenia symptoms in the children affected by this.318319 The same was found in people with ADHD and non-affective psychosis.320

Food additives (here: Sun yellow, carmoisine, tartrazine, ponceau 4R; quinoline yellow, allura red, sodium benzoate) can cause histamine release from circulating basophils. This is not allergic, i.e. not dependent on immunoglobulin E. The increased release of histamine can - in carriers of certain gene variants of the genes that encode histamine-degrading enzymes - increase ADHD symptoms321

To identify rare food intolerances (which, unlike allergies, cannot be detected by blood tests), an elimination diet can be helpful. However, such a diet is very difficult to implement and maintain and is barely adhered to, especially in younger children. In particular, any benefits must be weighed against the sometimes serious social consequences.

In other cases, such a diet can help to alleviate the symptoms of existing intolerances.

When assessing the effectiveness of diets (and other “desirable” therapies), parents’ assessments are often far higher than what tests or teacher evaluations can confirm.

A review (k = 21 meta-analyses, n = 348,405,029) with highly suggestive evidence (evidence class II) found an increased risk of ASA by 179% (OR 2.79) for food allergies.213

More information at Nutrition and diet for ADHD.

3.3.4. Gut-brain axis, gut bacteria, gut flora

See under Gut-brain axis

3.3.5. Polycystic ovary syndrome (PCOS)

Women with polycystic ovary syndrome (PCOS) appear to have an increased risk of mental disorders, primarily anxiety disorders and depression, but also ADHD.322
Adolescents with PCOs also showed an increased risk of ADHD.323

3.3.6. (Untreated) type 1 diabetes (+ 145 %)

A study of persons with ADHD with and without insulin pump treatment found a 2.45-fold increased risk of ADHD in untreated people with type 1 diabetes, with ADHD considered a risk factor for inconsistent diabetes treatment.324
Another study found no evidence of causality of autoimmune diseases such as diabetes 1 for ADHD.325
Among people with ADHD, 15.9% had an existing ADHD diagnosis and a further 31.9% met the ADHD criteria of the ASRS.326 The subjects were those who had responded to the ASRS sent to them, so that a bias towards an inflated ADHD score can be expected here,

3.3.7. Phenylketonuria (PKU)

Phenylketonuria (Følling disease, phenylpyruvic acid oligophrenia) is a genetically caused metabolic disorder in which the amino acid phenylalanine cannot be broken down into tyrosine due to the lack of the enzyme phenylalanine hydroxylase (PAH). Tyrosine, in turn, is required for the synthesis of dopamine, so dopamine deficiency is a consequence of PKU.327 PKU has a prevalence of 1 in 8000 people.

One study found an ADHD rate of 38% in phenylketonuria despite adequate treatment.328
ADHD is also associated with dopamine deficiency.

3.3.8. Anabolic androgenic steroids (AAS)

Strength athletes who take anabolic androgenic steroids are significantly more likely to have ADHD than strength athletes who do not take them.329

3.3.9. Infections

3.3.9.1. Susceptibility to infection and infection burden

A higher burden of infection may have a cumulative association with psychiatric disorders beyond what has been described for individual infections. Susceptibility to infections is reflected in the infection burden (the number of specific infection types or sites). An increased burden of infection correlates with an increased risk of330

  • ADHD
  • ASS
  • bipolar disorders
  • Depression
  • Schizophrenia
  • psychiatric diagnoses overall.
    A modest but significant heritability was found for the burden of infection (h2 = 0.0221) and a high degree of genetic correlation between it and the overall psychiatric diagnosis (rg = 0.4298). There was also evidence of genetic causality of the overall infection for the overall psychiatric diagnosis.

3.3.9.2. Viral infections

3.3.9.2.1. Enteroviruses in general (up to + 182 %)

(Non-polio) enteroviruses cause a good half of all cases of aseptic meningitis and are therefore among the most important known causes.331 In addition to encephaltitis332, (non-polio) enteroviruses also frequently cause febrile illnesses, hand-foot-and-mouth disease, herpangina, aseptic meningitis and encephalitis, as well as sometimes serious and threatening infections such as myocarditis or neonatal sepsis.

Enteroviruses are being discussed as a possible cause of ADHD.333
One study found an increased risk of ADHD due to mild enterovirus infections (16%) and severe enterovirus infections (182%).334

3.3.9.2.2. Enterovirus A71 (EV-A71) (+ 200 %)

A longitudinal study of 43 adolescents who had a central nervous system infection with enterovirus A71 (EV-A71) between the ages of 6 and 18 found that 34.9% had ADHD. This more than tripled the risk of ADHD. There was also an increase in autistic symptoms. Other psychiatric diagnoses were not elevated.335336 Another study found ADHD particularly common when the A71 infection was accompanied by cardiopulmonary failure.337
EV-A71 often causes weakness, limb atrophy, seizures, hand-foot-and-mouth disease, encephalitis and reduced intelligence.

3.3.9.2.3. HIV

A study of children and adolescents with HIV in stable health found ADHD symptoms in 20%.338

3.3.9.2.4. Zoster encephalitis

In one isolated case, ADHD was mentioned in association with zoster encephalitis.339

3.3.9.2.5. Human endogenous retroviruses (HERV)

The topic Human endogenous retroviruses (HERV) and ADHD Is presented due to its high heritability in the chapter Development in the article Genetic and epigenetic causes of ADHD - Introduction

3.3.9.3. Bacterial infections

Periodontal disease is a bacterial inflammation of the gums caused by the bacterium P. gingivalis, which secretes toxins. Periodontal disease and is described as a risk factor for ADHD.340

3.3.9.4. Parasitic infections

A study of 100 children with ADHD and 100 healthy children found a correlation of ADHD with:341

  • Toxoplasma
  • Toxocara
  • Cryptosporidium parvum
  • Giardia lamblia
  • Entamoeba histolytica
    No difference was found with regard to Schistosoma (coccidia parasites).

3.3.10. Glucose-6-phosphate dehydrogenase deficiency (G6PD)

Glucose-6-phosphate dehydrogenase (G6PD) deficiency increased the risk of ADHD by 16%342

G6PD deficiency is an X-linked genetic disorder and affects around 4.9% of all people.
The enzyme glucose-6-phosphate dehydrogenase (G6PD) facilitates the synthesis of nicotinamide adenine dinucleotide phosphate (NADPH) and glutathione (GSH), which are involved in oxidation-reduction equilibrium regulation. G6PD deficiency causes reduced GSH levels and thus increased oxidative stress.

G6PD deficiency is mostly food-related (favism; hemolytic reaction to the consumption of fava beans) and sometimes genetic (more common in the Mediterranean region and the Middle East, partly in Asia and Africa).
G6PD deficiency can trigger (especially in children):

  • severe hemolysis
  • Hyperbilirubinemia
  • Jaundice
  • Hearing disorders
  • Behavioral disorders
  • lead to long-lasting neurological damage
  • increased production of reactive oxygen species (ROS)
    • resulting in activation of astrocytes and microglia, increased proinflammatory chemokines and cytokines, neuroinflammation, impaired brain development
  • Imbalance in the antioxidant system
    • this leads to impairment of astrocytes, neuronal death and DNA damage
    • oxidative cell death of leukocytes, myocytes and other immunological players.

3.3.11. Kawasaki syndrome

A meta-analysis of k = 4 studies with n = 1,454,499 subjects found a 76% increased risk of ADHD in people with ADHD.343
One study found an increased prevalence of ADHD in people with ADHD344, other studies found only a tendency345 or no association.346

3.3.12. Lipodystrophy (lack of fatty tissue)

One study found evidence of a greatly increased prevalence of ADHD in lipodystrophy.347

3.3.13. Dystrophinopathy (muscular dystrophy, muscle weakness)

In dystrophinopathy (e.g. Duchenne muscular dystrophy - complete absence of dystrophin in muscle tissue 348 or Becker muscular dystrophy - dystrophin levels too low349 ), there was evidence of a greatly increased prevalence of ADHD of 18.4%350351 , of ASD of 6%350 to 12.73%351 and of mental retardation of 22%.350
There are also links between ADHD gene candidates and genes associated with dystrophies. See there.

3.3.14. Hyperthyroidism / hypothyroidism

In addition to other cognitive deficits, hyperthyroidism can also cause inattention and hyperarousal. Depending on the degree of hypothyroidism, the cognitive effects can range from mild impairment of memory and attention to complete dementia.352353

The THRA gene encodes the thyroid receptor alpha, TRα1, TRHB the thyroid receptor isoforms TRβ1 and TRβ2.
The pituitary hormone TSH (thyroid-stimulating hormone) stimulates the thyroid gland to produce thyroxine (T4; prohormone) and then triiodothyronine (T3). The thyroid hormones (T3 and T4) in the blood in turn regulate the pituitary release of TSH within the hypothalamic-pituitary-thyroid axis, which is mediated by the receptor isoform TRβ2.

In the case of a (rarely occurring genetic) resistance to thyroid hormone β, this negative feedback loop, which stabilizes the TH level in the blood, is disrupted. This leads to increased TH and unsuppressed, i.e. normal TSH levels354

3.3.15. Interrupted breathing during sleep (sleep apnea)

Breathing interruptions in children’s sleep can trigger cognitive stress, causing symptoms that resemble ADHD.355

It remains to be seen whether breathing interruptions during sleep can cause such stress that they can contribute to ADHD through epigenetic changes, or whether they merely cause symptoms that are similar to those of ADHD. In the latter case, people who did not previously have ADHD and who have developed ADHD (similar) symptoms as a result of breathing interruptions during sleep should see these symptoms disappear completely once the breathing interruptions during sleep have been eliminated. We are not yet aware of any studies on this.

In contrast, a study of n = 512 snoring children and adolescents (70% of whom were overweight or obese and 35% with an increased obstructive AHI >3/h) found that of the snoring children236

  • 36 % showed hyperactivity/impulsivity
  • 24 % showed emotional dysregulation

This would indicate a 300% increased risk of ADHD.
Surprisingly, the reduced oxygen supply due to sleep apnea did not correlate significantly with hyperactivity/impulsivity or emotional dysregulation, while the measure correlated with the volume of snoring.

See also under Differential diagnosis: Primary organic disorders

3.3.16. Gender diversity

A multinational study found evidence that the frequency and severity of ADHD symptoms was particularly high in gender-diverse individuals.356

3.3.17. Lipid metabolism, fatty acids

To avoid redundancies, we have placed this section under Fatty acids, probiotics and more for ADHD In the chapter Treatment: Medication for ADHD in the section Vitamins, minerals, dietary supplements for ADHD.

3.3.18. Inflammatory bowel disease (IBD)

Inflammatory bowel disease (IBD) correlates with an increased risk of ADHD and an increased intake of stimulants.357
People with an onset of IBD in childhood showed an increased risk of358

  • ADHD: + 20 %
  • ASS: + 40 %
  • psychiatric disorders overall: + 60 %
  • Anxiety disorders: + 90 %
  • Affective disorders: + 60 %
  • Eating disorders: + 60 %
  • Personality disorders: + 40 %
  • Suicide attempts: + 40 %

One study found no significant increase in ADHD from IBD overall, but from Crohn’s disease (CD) (+6.5%) and ulcerative colitis (UC) (+5%).359

A study of individuals with onset of IBD in childhood found a 25% reduced risk of MPH use. Ulcerative colitis was associated with a 37% reduced risk of MPH use and a reduced - although not statistically significant - risk of an ADHD diagnosis. The risk of depression was increased by 50% in IBD and by 73% in people with both IBD subtypes (Crohn’s disease and ulcerative colitis.360

Some studies suggest that IBD in the mother during pregnancy can inflame the central nervous system, which increases the risk of ADHD in the offspring.361

3.3.19. Mitochondrial disorder

Mitochondrial dysfunction or mitochondrial disorders are being discussed as a possible cause of ADHD.362363 So far, no concrete findings have emerged.

3.4. Factors without risk increase for ADHD

  • Dichlorodiphenyldichloroethylene did not influence the risk of ADHD107
  • 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.104 Another study found no effect on ADHD.107
  • Organic pollutants (OP pesticides, PCBs, pyrethroid insecticides and trichlorophenol (TCP)) did not increase the odds ratio for ADHD (0.99)89
  • Bismuth urine levels were slightly lower in children with ADHD than in children without ADHD.87
  • Aluminum blood levels were unchanged in children with ADHD.85
  • The prevalence of microcytic anemia was found to be 14% lower in children with ADHD (OR: 0.86)364
  • Growing up bilingual did not increase the risk of ADHD365
  • Autoimmune diseases showed no evidence of causality of autoimmune diseases for ADHD. The study examined325
    • Lupus erythematosus
      • in contrast, a retrospective matched cohort study (n = 11,144) found that a lupus diagnosis 15 years or more later correlated with a 61% higher probability of psychiatric disorders and psychiatric disorders correlated with a 120% increased risk of lupus 10 years or more later. A lupus diagnosis correlated with an increased prescription of ADHD medications 10 years earlier.366
    • Crohn’s disease
    • Ulcerative colitis
    • Type 1 diabetes (opposite study: see above)
    • rheumatoid arthritis
    • Psoriasis
    • Ankylosing spondylitis
    • Multiple sclerosis
  • High blood pressure
    • One study found no statistical significance for a genetic link between high blood pressure and ADHD.367 This is at least countered by the main ADHD model animal, the SHR, which develops hypertension with age.
  • COVID-19 gene therapy
    • Gene predisposition, which makes people more susceptible to COVID-19, showed no signs of an increased risk of ADHD. Conversely, however, ADHD and Tourette’s are associated with an increased risk of COVID-19 and a more severe course of COVID-19.368
  • T1w/T2w ratio
    • The T1w/T2w ratio is a magnetic resonance imaging (MRI)-based indicator of intracortical myelin. There were no differences in the T1w/T2w ratio between ADHD, ASD and controls.369
  • Obesity
    • adjusted, there was no significant causal influence of obesity on ADHD370
  • Platinum- and taxane-based chemotherapy in children371

3.5. Factors with risk reduction for ADHD

  • Immigrant status of parents causes a reduced risk of ADHD372 within the first 2 generations.373
  • p,p’-dichlorodiphenyltrichloroethane (p,p’-DDT) was associated with a 36% lower likelihood of ADHD104
  • Lyme disease correlated with a 10% reduction in ADHD prevalence374

  1. Bilgin, Baumann, Jaekel, Breeman, Bartmann, Bäuml, Avram, Sorg, Wolke (2018): Early Crying, Sleeping, and Feeding Problems and Trajectories of Attention Problems From Childhood to Adulthood. Child Dev. 2018 Oct 6. doi: 10.1111/cdev.13155. Langzeitstudie, n = 342

  2. Wolke, Rizzo, Woods: Persistent Infant Crying and Hyperactivity Problems in Middle Childhood, (2002), Pediatrics 109;1054-1060; n = 64

  3. Reijneveld, van der Wal, Brugman, Sing, Verloove-Vanhorick (2004): Infant crying and abuse. Lancet 364: 1340-1342, zitiert nach Blanca Tro y Baumann, Diss 2010: Exzessives Schreien beim gesunden Säugling – Ein Vergleich der Schreikindprävalenz in fünf europäischen Ländern

  4. Blanca Tro y Baumann, Diss 2010: Exzessives Schreien beim gesunden Säugling – Ein Vergleich der Schreikindprävalenz in fünf europäischen Ländern

  5. Blanca Tro y Baumann, Diss 2010: Exzessives Schreien beim gesunden Säugling – Ein Vergleich der Schreikindprävalenz in fünf europäischen Ländern unter Verweis auf Forsyth und Canny 1991

  6. Lin CH, Lin WD, Chou IC, Lee IC (2021): Hong SY. Is Preterm Birth a Risk Factor for Subsequent Autism Spectrum Disorder and Attention Deficit Hyperactivity Disorder in Children with Febrile Seizure?-A Retrospective Study. Life (Basel). 2021 Aug 20;11(8):854. doi: 10.3390/life11080854. PMID: 34440598; PMCID: PMC8398685.

  7. Chen CY, Shih PY, Su CT, Cheng CF, Lee MC, Lane HY (2025): Association between infant feeding and ADHD development in childhood: a birth cohort study in Taiwan. J Child Psychol Psychiatry. 2025 Jun;66(6):881-891. doi: 10.1111/jcpp.14100. PMID: 39707757; PMCID: PMC12062847. n = 19.721

  8. Park, Kim, Kim, Shin, Yoo, Cho (2014): Protective effect of breastfeeding with regard to children’s behavioral and cognitive problems. Nutr J. 2014 Nov 29;13(1):111. doi: 10.1186/1475-2891-13-111. n = 874

  9. Mimouni-Bloch, Kachevanskaya, Mimouni, Shuper, Raveh, Linder (2013): Breastfeeding may protect from developing attention-deficit/hyperactivity disorder. Breastfeed Med. 2013 Aug;8(4):363-7. doi: 10.1089/bfm.2012.0145.

  10. Golmirzaei, Namazi, Amiri, Zare, Rastikerdar, Hesam, Rahami, Ghasemian, Namazi, Paknahad, Mahmudi, Mahboobi, Khorgoei, Niknejad, Dehghani, Asadi (2013): Evaluation of attention-deficit hyperactivity disorder risk factors. Int J Pediatr. 2013;2013:953103. doi: 10.1155/2013/953103.

  11. Lee YS, Sprong ME, Shrestha J, Smeltzer MP, Hollender H (2024): Trajectory Analysis for Identifying Classes of Attention Deficit Hyperactivity Disorder (ADHD) in Children of the United States. Clin Pract Epidemiol Ment Health. 2024 May 21;20:e17450179298863. doi: 10.2174/0117450179298863240516070510. PMID: 39130191; PMCID: PMC11311732.

  12. Elvin ÖD, Aydan ÇV, Güzel EÇ, Topçu B (2024): Evaluation of the relationship between the frequency of attention deficit, hyperactivity disorder symptoms and nutritional habits in children. Med J Malaysia. 2024 Sep;79(5):538-546. PMID: 39352155.

  13. Tseng PT, Yen CF, Chen YW, Stubbs B, Carvalho AF, Whiteley P, Chu CS, Li DJ, Chen TY, Yang WC, Tang CH, Liang HY, Yang WC, Wu CK, Lin PY (2019): Maternal breastfeeding and attention-deficit/hyperactivity disorder in children: a meta-analysis. Eur Child Adolesc Psychiatry. 2019 Jan;28(1):19-30. doi: 10.1007/s00787-018-1182-4. PMID: 29907910. METASTUDY

  14. Aliye K, Tesfaye E, Soboka M (2023): High rate of attention deficit hyperactivity disorder among children 6 to 17 years old in Southwest Ethiopia findings from a community-based study. BMC Psychiatry. 2023 Mar 8;23(1):144. doi: 10.1186/s12888-023-04636-9. PMID: 36890504; PMCID: PMC9993367. n = 504

  15. Jenabi E, Ayubi E, Farashi S, Bashirian S, Mehri F (2023): The neonatal risk factors associated with attention-deficit/ hyperactivity disorder: an umbrella review. Clin Exp Pediatr. 2023 Jul 14. doi: 10.3345/cep.2022.01396. PMID: 37448127. METASTUDY

  16. Zhang Z, Yu J, Li Q, Zhao Y, Tang L, Peng Y, Liu Y, Gan C, Liu K, Wang J, Chen L, Luo Q, Qiu H, Ren H, Jiang C (2025): Unraveling the causal pathways of maternal smoking and breastfeeding in the development of neuropsychiatric disorders: A Mendelian randomization perspective. J Affect Disord. 2025 Mar 15;373:35-43. doi: 10.1016/j.jad.2024.12.075. PMID: 39716673. n = 352.094

  17. Assaf M, Rouphael M, Bou Sader Nehme S, Soufia M, Alameddine A, Hallit S, Landry M, Bitar T, Hleihel W (2024): Correlational Insights into Attention-Deficit/Hyperactivity Disorder in Lebanon. Int J Environ Res Public Health. 2024 Aug 5;21(8):1027. doi: 10.3390/ijerph21081027. PMID: 39200638; PMCID: PMC11353674. n = 119

  18. Zeng Y, Tang Y, Tang J, Shi J, Zhang L, Zhu T, Xiao D, Qu Y, Mu D (2020): Association between the different duration of breastfeeding and attention deficit/hyperactivity disorder in children: a systematic review and meta-analysis. Nutr Neurosci. 2020 Oct;23(10):811-823. doi: 10.1080/1028415X.2018.1560905. PMID: 30577717. METASTUDY

  19. Schwenke, Fasching, Faschingbauer, Pretscher, Kehl, Peretz, Keller, Häberle, Eichler, Irlbauer-Müller, Dammer, Beckmann, Schneider (2018): Predicting attention deficit hyperactivity disorder using pregnancy and birth characteristics. Arch Gynecol Obstet. 2018 Sep 8. doi: 10.1007/s00404-018-4888-0.

  20. Soled D, Keim SA, Rapoport E, Rosen L, Adesman A (2021): Breastfeeding Is Associated with a Reduced Risk of Attention-Deficit/Hyperactivity Disorder Among Preschool Children. J Dev Behav Pediatr. 2021 Jan 1;42(1):9-15. doi: 10.1097/DBP.0000000000000854. PMID: 33347037. n = 12.703

  21. Jallow J, Hurtig T, Kerkelä M, Miettunen J, Halt AH (2024): Prenatal maternal stress, breastfeeding and offspring ADHD symptoms. Eur Child Adolesc Psychiatry. 2024 Nov;33(11):4003-4011. doi: 10.1007/s00787-024-02451-5. PMID: 38691181; PMCID: PMC11588867. n = 7.910

  22. Cha JH, Cho Y, Moon JH, Lee J, Na JY, Kim YJ (2023): Feeding practice during infancy is associated with attention-deficit/hyperactivity disorder and autism spectrum disorder: a population-based study in South Korea. Eur J Pediatr. 2023 May 23. doi: 10.1007/s00431-023-05022-z. PMID: 37219627. n = 1.173.448

  23. {Chen CY, Shih PY, Su CT, Cheng CF, Lee MC, Lane HY (2025): Association between infant feeding and ADHD development in childhood: a birth cohort study in Taiwan. J Child Psychol Psychiatry. 2025 Jun;66(6):881-891. doi: 10.1111/jcpp.14100. PMID: 39707757; PMCID: PMC12062847. n = 19.721

  24. Julvez J, Ribas-Fitó N, Forns M, Garcia-Esteban R, Torrent M, Sunyer J (2007): Attention behaviour and hyperactivity at age 4 and duration of breast-feeding. Acta Paediatr. 2007 Jun;96(6):842-7. doi: 10.1111/j.1651-2227.2007.00273.x. PMID: 17537012. n = 500

  25. Jallow J, Hurtig T, Kerkelä M, Miettunen J, Halt AH (2024): Prenatal maternal stress, breastfeeding and offspring ADHD symptoms. Eur Child Adolesc Psychiatry. 2024 Nov;33(11):4003-4011. doi: 10.1007/s00787-024-02451-5. PMID: 38691181; PMCID: PMC11588867. = 7.910

  26. Stadler, Musser, Holton, Shannon, Nigg (2016): Recalled Initiation and Duration of Maternal Breastfeeding Among Children with and Without ADHD in a Well Characterized Case-Control Sample. J Abnorm Child Psychol. 2016 Feb;44(2):347-55. doi: 10.1007/s10802-015-9987-9.

  27. 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

  28. 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

  29. Lebeña A, Faresjö Å, Jones MP, Bengtsson F, Faresjö T, Ludvigsson J (2024): Early environmental predictors for attention-deficit hyperactivity disorder (ADHD), autism spectrum disorder (ASD) and their co-occurrence: The prospective ABIS-Study. Sci Rep. 2024 Jun 26;14(1):14759. doi: 10.1038/s41598-024-65067-4. PMID: 38926504; PMCID: PMC11208583.

  30. Boucher O, Julvez J, Guxens M, Arranz E, Ibarluzea J, Sánchez de Miguel M, Fernández-Somoano A, Tardon A, Rebagliato M, Garcia-Esteban R, O’Connor G, Ballester F, Sunyer J (2017): Association between breastfeeding duration and cognitive development, autistic traits and ADHD symptoms: a multicenter study in Spain. Pediatr Res. 2017 Mar;81(3):434-442. doi: 10.1038/pr.2016.238. PMID: 27846197.

  31. Borrego-Ruiz A, Borrego JJ (2024): Neurodevelopmental Disorders Associated with Gut Microbiome Dysbiosis in Children. Children (Basel). 2024 Jun 28;11(7):796. doi: 10.3390/children11070796. PMID: 39062245; PMCID: PMC11275248. REVIEW

  32. Perera FP, Chang HW, Tang D, Roen EL, Herbstman J, Margolis A, Huang TJ, Miller RL, Wang S, Rauh V (2014): Early-life exposure to polycyclic aromatic hydrocarbons and ADHD behavior problems. PLoS One. 2014 Nov 5;9(11):e111670. doi: 10.1371/journal.pone.0111670. PMID: 25372862; PMCID: PMC4221082.

  33. Mortamais M, Pujol J, van Drooge BL, Macià D, Martínez-Vilavella G, Reynes C, Sabatier R, Rivas I, Grimalt J, Forns J, Alvarez-Pedrerol M, Querol X, Sunyer J (2017): Effect of exposure to polycyclic aromatic hydrocarbons on basal ganglia and attention-deficit hyperactivity disorder symptoms in primary school children. Environ Int. 2017 Aug;105:12-19. doi: 10.1016/j.envint.2017.04.011. PMID: 28482185.

  34. Adesman, Soled, Rosen (2017): Formula Feeding as a Risk Factor for Attention-Deficit/Hyperactivity Disorder: Is Bisphenol A Exposure a Smoking Gun? J Dev Behav Pediatr. 2017 Sep;38(7):545-551. doi: 10.1097/DBP.0000000000000468.

  35. Sagheddu C, Muntoni AL, Pistis M, Melis M (2015): Endocannabinoid Signaling in Motivation, Reward, and Addiction: Influences on Mesocorticolimbic Dopamine Function. Int Rev Neurobiol. 2015;125:257-302. doi: 10.1016/bs.irn.2015.10.004. PMID: 26638769. REVIEW

  36. Carballo JJ, García-Nieto R, Alvarez-García R, Caro-Cañizares I, López-Castromán J, Muñoz-Lorenzo L, de Leon-Martinez V, Baca-García E (2013): Sibship size, birth order, family structure and childhood mental disorders. Soc Psychiatry Psychiatr Epidemiol. 2013 Aug;48(8):1327-33. doi: 10.1007/s00127-013-0661-7. PMID: 23407902.n = 16.823

  37. Marín AM, Seco FL, Serrano SM, García SA, Gaviria Gómez AM, Ney I (2014): Do firstborn children have an increased risk of ADHD? J Atten Disord. 2014 Oct;18(7):594-7. doi: 10.1177/1087054712445066. PMID: 22826511. n = 213

  38. AlOmar RS, AlShamlan NA, Al-Johani WM, Almayyad AH, Alotaibi HK, Alsamin SI, Alfrayyan NY, Boubshait LA, Alghamdi LM, Al-Shammari MA (2023): Adult ADHD Positive Screening and Sibship, Birth Order and Academic Achievement: Implications for Epidemiologists and Physicians in the Kingdom of Saudi Arabia. Psychol Res Behav Manag. 2023 Oct 5;16:4069-4081. doi: 10.2147/PRBM.S426715. PMID: 37817910; PMCID: PMC10561610.

  39. Canals J, Morales-Hidalgo P, Jané MC, Domènech E (2018): ADHD Prevalence in Spanish Preschoolers: Comorbidity, Socio-Demographic Factors, and Functional Consequences. J Atten Disord. 2018 Jan;22(2):143-153. doi: 10.1177/1087054716638511. PMID: 27009923. n = 1.104

  40. Reimelt C, Wolff N, Hölling H, Mogwitz S, Ehrlich S, Martini J, Roessner V (2021): Siblings and Birth Order-Are They Important for the Occurrence of ADHD? J Atten Disord. 2021 Jan;25(1):81-90. doi: 10.1177/1087054718770020. PMID: 29720025. n = 13.488

  41. Björkegren E, Svaleryd H (2023): Birth order and health disparities throughout the life course. Soc Sci Med. 2023 Feb;318:115605. doi: 10.1016/j.socscimed.2022.115605. PMID: 36630815.

  42. Venkatesh C, Ravikumar T, Andal A, Virudhagirinathan BS (2012): Attention-deficit/Hyperactivity Disorder in Children: Clinical Profile and Co-morbidity. Indian J Psychol Med. 2012 Jan;34(1):34-8. doi: 10.4103/0253-7176.96155. PMID: 22661805; PMCID: PMC3361840.

  43. Tilahun WM, Wolde HF, Gebreegziabher ZA, Abebaw WA, Simegn MB, Tadesse AA (2022): Magnitude, relationship and determinants of attention deficit hyperactivity disorder and depression among University of Gondar undergraduate students, Northwest Ethiopia, 2022: Non-recursive structural equation modeling. PLoS One. 2023 Oct 5;18(10):e0291137. doi: 10.1371/journal.pone.0291137. PMID: 37796847; PMCID: PMC10553242.

  44. Kaur J, Kaur K (2012) Obstetric complications: Primiparity Vs. Multiparity. European Journal of Experimental Biology. 2012 Sep; 2(5): 1462-1468.

  45. Berger I, Felsenthal-Berger N (2009): Attention-deficit hyperactivity disorder (ADHD) and birth order. J Child Neurol. 2009 Jun;24(6):692-6. doi: 10.1177/0883073808330763. PMID: 19211923.

  46. Keshavarzi Z, Bajoghli H, Mohamadi MR, Holsboer-Trachsler E, Brand S (2014): Attention deficit hyperactivity disorder in children is found to be related to the occurrence of ADHD in siblings and the male gender, but not to birth order, when compared to healthy controls. Int J Psychiatry Clin Pract. 2014 Oct;18(4):272-9. doi: 10.3109/13651501.2014.957704. PMID: 25152020. n = 400

  47. Jahangard L, Haghighi M, Bajoghli H, Holsboer-Trachsler E, Brand S (2013): Among a sample of Iranian students, adult attention deficit hyperactivity disorder is related to childhood ADHD, but not to age, gender, socioeconomic status, or birth order–an exploratory study. Int J Psychiatry Clin Pract. 2013 Oct;17(4):273-8. doi: 10.3109/13651501.2013.800555. PMID: 23808614. n = 387

  48. Masana Marín A, López Seco F, Martí Serrano S, Acosta García S (2011): Correspondence on ‘‘Attention-deficit hyperactivity disorder (ADHD) and birth order’’. J Child Neurol. 2011 Mar;26(3):395; author reply 395-6. doi: 10.1177/0883073810397546. PMID: 21383230.

  49. Rahman MS, Takahashi N, Iwabuchi T, Nishimura T, Harada T, Okumura A, Takei N, Nomura Y, Tsuchiya KJ (2021): Elevated risk of attention deficit hyperactivity disorder (ADHD) in Japanese children with higher genetic susceptibility to ADHD with a birth weight under 2000 g. BMC Med. 2021 Sep 24;19(1):229. doi: 10.1186/s12916-021-02093-3. PMID: 34556092; PMCID: PMC8461893.

  50. Hsu YC, Chen CT, Yang HJ, Chou P (2019): Family structure, birth order, and aggressive behaviors among school-aged boys with attention deficit hyperactivity disorder (ADHD). Soc Psychiatry Psychiatr Epidemiol. 2019 Jun;54(6):661-670. doi: 10.1007/s00127-018-1624-9. PMID: 30535676.

  51. Hsu YC, Chen CT, Yang HJ, Chou P (2022): Family, personal, parental correlates and behavior disturbances in school-aged boys with attention-deficit/hyperactivity disorder (ADHD): a cross-sectional study. Child Adolesc Psychiatry Ment Health. 2022 Apr 19;16(1):30. doi: 10.1186/s13034-022-00467-w. PMID: 35440036; PMCID: PMC9019941. n = 1.855

  52. Montes G (2018): Having Older Siblings Is Associated with Lower Rates of Depression, ADD/ADHD, Anxiety and Behavior Problems Among Children with ASD. Matern Child Health J. 2018 May;22(5):642-647. doi: 10.1007/s10995-018-2459-4. PMID: 29429133. n = 1.624

  53. Persönliche Mitteilung von Chiuying Chen

  54. Chang, Tsai, Hou, Tseng, Lee, Tsai (2018): Multiple subependymal pseudocysts in neonates play a role in later attention deficit hyperactivity and autistic spectrum disorder. J Formos Med Assoc. 2018 Sep 4. pii: S0929-6646(17)30885-9. doi: 10.1016/j.jfma.2018.08.007.

  55. Norton, Gifford, Pawlak, Derbaly, Sherman, Zhang, Wagner, Kusnecov (2020): Long-lasting Behavioral and Neuroanatomical Effects of Postnatal Valproic Acid Treatment. Neuroscience. 2020 May 10;434:8-21. doi: 10.1016/j.neuroscience.2020.02.029. PMID: 32112916.

  56. Awadu JE, Giordani B, Sikorskii A, Abbo C, Fenton JI, Zalwango S, Ezeamama AE (2023): Vitamin D and Probability of Developmental Disorders among Perinatally HIV-Affected and Unaffected Ugandan Children. Nutrients. 2023 Apr 22;15(9):2020. doi: 10.3390/nu15092020. PMID: 37432158; PMCID: PMC10181402.

  57. Chang IJ, Lam C, Vockley J (2000): Medium-Chain Acyl-Coenzyme A Dehydrogenase Deficiency. 2000 Apr 20 [updated 2024 Sep 26]. In: Adam MP, Feldman J, Mirzaa GM, Pagon RA, Wallace SE, Amemiya A, editors. GeneReviews® [Internet]. Seattle (WA): University of Washington, Seattle; 1993–2025. PMID: 20301597. REVIEW

  58. DocCheck Flexikon: Infektion

  59. Lin MC, Pan YJ, Wu CS, Liu CL, Chen PC, Thompson WK, Fan CC, Wang SH (2025): Prenatal and early childhood infections requiring hospitalization and risk of neurodevelopmental disorders in offspring: a population-based birth cohort study in Taiwan. Mol Psychiatry. 2025 May;30(5):1791-1800. doi: 10.1038/s41380-024-02787-z. PMID: 39390224.

  60. Hsu TW, Chu CS, Tsai SJ, Bai YM, Su TP, Chen TJ, Chen MH, Liang CS (2022): Risk of Major Mental Disorder after Severe Bacterial Infections in Children and Adolescents: A Nationwide Longitudinal Study. Neuropsychobiology. 2022;81(6):539-549. doi: 10.1159/000526984. PMID: 36404700. n = 14.024

  61. Feigenson KA, Kusnecov AW, Silverstein SM (2014): Inflammation and the two-hit hypothesis of schizophrenia. Neurosci Biobehav Rev. 2014 Jan;38:72-93. doi: 10.1016/j.neubiorev.2013.11.006. PMID: 24247023; PMCID: PMC3896922. REVIEW

  62. Mynarek M, Vik T, Andersen GL, Brigtsen AK, Hollung SJ, Larose TL, Lydersen S, Olsen LC, Strøm MS, Afset JE (2023): Mortality and neurodevelopmental outcome after invasive group B streptococcal infection in infants. Dev Med Child Neurol. 2023 Jun 12. doi: 10.1111/dmcn.15643. Epub ahead of print. PMID: 37306102.

  63. Graham EE, Tetens MM, Bodilsen J, Dessau R, Ellermann-Eriksen S, Andersen NS, Jørgensen CS, Pedersen M, Søgaard KK, Bangsborg J, Nielsen AC, Møller JK, Obel D, Lebech AM, Nygaard U, Omland LH, Obel N (2024): Risk of psychiatric neurodevelopmental disorders after meningitis in childhood: a nationwide, population-based cohort study. Infect Dis (Lond). 2024 Sep 4:1-11. doi: 10.1080/23744235.2024.2399101. PMID: 39230517. n = 20.300

  64. Lavebratt, Yang, Giacobini, Forsell, Schalling, Partonen, Gissler (2019): Early exposure to antibiotic drugs and risk for psychiatric disorders: a population-based study. Transl Psychiatry. 2019 Nov 26;9(1):317. doi: 10.1038/s41398-019-0653-9. n = 1 Mio.

  65. Oh J, Woo HG, Kim HJ, Park J, Lee M, Rahmati M, Rhee SY, Min C, Koyanagi A, Smith L, Fond G, Boyer L, Kim MS, Shin JI, Lee SW, Yon DK (2024): Prenatal and infant exposure to antibiotics and subsequent risk of neuropsychiatric disorders in children: A nationwide birth cohort study in South Korea. Psychiatry Res. 2024 Oct;340:116117. doi: 10.1016/j.psychres.2024.116117. PMID: 39128166. n = 3,2 Mio infants, 2,3 Mio mothers

  66. Slykerman, Thompson, Waldie, Murphy, Wall, Mitchell (2017): Antibiotics in the first year of life and subsequent neurocognitive outcomes. Acta Paediatr. 2017 Jan;106(1):87-94. doi: 10.1111/apa.13613. n = 526

  67. Hamad, Alessi-Severini, Mahmud, Brownell, Kuo (2019): Antibiotic Exposure in the First Year of Life and the Risk of Attention-Deficit/Hyperactivity Disorder: A Population-based Cohort Study. Am J Epidemiol. 2019 Sep 5. pii: kwz178. doi: 10.1093/aje/kwz178.

  68. Axelsson, Clausen, Petersen, Hageman, Pinborg, Kessing, Bergholt, Rasmussen, Keiding, Løkkegaard (2018): Investigating the effects of cesarean delivery and antibiotic use in early childhood on risk of later attention deficit hyperactivity disorder. J Child Psychol Psychiatry. 2018 Aug 23. doi: 10.1111/jcpp.12961. n = 671.592 Geburten

  69. Beier MA, Setoguchi S, Gerhard T, Roy J, Koffman D, Mendhe D, Madej J, Strom BL, Blaser MJ, Horton DB (2025): Early childhood antibiotics and chronic pediatric conditions: a retrospective cohort study. J Infect Dis. 2025 Apr 16:jiaf191. doi: 10.1093/infdis/jiaf191. PMID: 40237450.

  70. Green JE, Wrobel A, Todd E, Marx W, Berk M, Lotfaliany M, Castle D, Cryan JF, Athan E, Hair C, Nierenberg AA, Jacka FN, Dawson S (2025): Early antibiotic exposure and risk of psychiatric and neurocognitive outcomes: systematic review and meta-analysis. Br J Psychiatry. 2025 Mar;226(3):171-183. doi: 10.1192/bjp.2024.121. PMID: 39658347. REVIEW

  71. Yang KL, Yen TA, Lin FJ, Hsu CN, Wang CC (2024): Antibiotic use and risk of autism spectrum disorder and attention-deficit/hyperactivity disorder: a population-based cohort study. Child Adolesc Psychiatry Ment Health. 2024 Jul 11;18(1):82. doi: 10.1186/s13034-024-00774-4. PMID: 38992772; PMCID: PMC11241894. n = 2.441.886

  72. Greenlaw C, MacRae R, Wilson-Murphy M (2025): Powassan Virus Encephalitis in Pediatric Patients. J Child Neurol. 2025 Apr 27:8830738251333465. doi: 10.1177/08830738251333465. PMID: 40289568.

  73. Almeida MN, Alper DP, Barrero C, Parikh N, Hauc SC, Moscarelli J, Golinko M, Persing J, Swanson J, Alperovich M (2024): Radiographic severity is associated with worse executive function in metopic craniosynostosis. Childs Nerv Syst. 2024 Jun 21. doi: 10.1007/s00381-024-06493-7. PMID: 38904768.

  74. Almeida MN, Alper DP, Long AS, Barrero C, Williams MC, Boroumand S, Glahn J, Shah J, Swanson J, Alperovich M (2023): Risk of ADHD, Autism Spectrum Disorder, and Executive Function Impairment in Metopic Craniosynostosis. Plast Reconstr Surg. 2023 Dec 19. doi: 10.1097/PRS.0000000000011249. PMID: 38113367. n = 60

  75. Vanderwiel SL, Jones B, Nickels KC, Wong-Kisiel LC, Fine A, Mandrekar J, Wirrell EC (2024): Risk of ADHD in children with childhood absence epilepsy versus controls: A population-based study. Epilepsy Behav. 2024 Dec;161:110143. doi: 10.1016/j.yebeh.2024.110143. PMID: 39515005.

  76. Brabcová DB, Otáhalová A, Kohout J, Suleková M, Mašková I, Bělohlávková A, Kršek P (2025): Quality of life, academic self-concept, and mental health in children with epilepsy: The possible role of epilepsy comorbidities. Epilepsy Behav. 2025 Sep;170:110471. doi: 10.1016/j.yebeh.2025.110471. PMID: 40383004.

  77. Teo JH, Lim WC, Wong MS, Lin JB, Han VX, Ng ZM, Thomas T (2025): Comparative clinical outcomes in children with Febrile Infection-Related Epilepsy Syndrome (FIRES). Epilepsy Behav. 2025 Sep;170:110467. doi: 10.1016/j.yebeh.2025.110467. PMID: 40382999.

  78. 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

  79. 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

  80. 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.

  81. 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)

  82. 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

  83. 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

  84. 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

  85. 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.

  86. 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.

  87. 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

  88. 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

  89. 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

  90. 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.

  91. 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.

  92. 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.

  93. 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.

  94. 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.

  95. 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.

  96. 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.

  97. 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.

  98. 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

  99. 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

  100. 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

  101. 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.

  102. 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.

  103. 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.

  104. 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

  105. 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.

  106. 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.

  107. 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

  108. 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.

  109. Gurgel W, Garcia-Argibay M, D’Onofrio BM, Larsson H, Polanczyk GV (2025): Predictors of preschool attention-deficit/hyperactivity disorder diagnosis: a population-based study using national registers. J Child Psychol Psychiatry. 2025 Jun;66(6):834-845. doi: 10.1111/jcpp.14093. PMID: 39676220. n = 631.695

  110. 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

  111. 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

  112. 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.

  113. 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

  114. 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

  115. 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.

  116. Khoshbakht Y, Bidaki R, Salehi-Abargouei A. Vitamin D Status and Attention Deficit Hyperactivity Disorder: A Systematic Review and Meta-Analysis of Observational Studies. Adv Nutr. 2018 Jan 1;9(1):9-20. doi: 10.1093/advances/nmx002. PMID: 29438455; PMCID: PMC6333940. METASTUDY

  117. 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

  118. 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.

  119. 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

  120. 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

  121. 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

  122. 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.

  123. 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.

  124. 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.

  125. Deutscher Wetterdienst; Größenverteilung luftgetragener Partikel und ihre Entstehungsprozessenach Whitby und Cantrell, 1976 german

  126. 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

  127. WWF-Studie zur Aufnahme von Mikroplastik

  128. Deutsches Bundesinstitut für Risikobewertung: Mikroplastik: Fakten, Forschung und offene Fragen german

  129. 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.

  130. 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.

  131. 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.

  132. 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.

  133. 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

  134. 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.

  135. 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.

  136. 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

  137. 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.

  138. 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.

  139. 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

  140. 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

  141. 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

  142. 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

  143. 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

  144. 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

  145. 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

  146. 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

  147. 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

  148. 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.

  149. 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.

  150. 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.

  151. 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.

  152. 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

  153. 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.

  154. 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.

  155. 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.

  156. 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.

  157. 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.

  158. 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.

  159. 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.

  160. 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.

  161. 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.

  162. 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

  163. 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

  164. 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

  165. 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

  166. 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.

  167. 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.

  168. 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.

  169. 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.

  170. 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.

  171. 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.

  172. 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

  173. Umweltbundesamt, abgerufen 30.11.23

  174. 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.

  175. 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.

  176. 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.

  177. 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

  178. 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

  179. 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.

  180. Zader SJ, Williams E, Buryk MA (2019); Mental Health Conditions and Hyperthyroidism. Pediatrics. 2019 Nov;144(5):e20182874. doi: 10.1542/peds.2018-2874. PMID: 31582535. n = 2.479

  181. Ozturk Y, Hangül Z, Demir N, Tufan AE (2024): THE RELATIONS BETWEEN THYROID HORMONES AND CLINICAL FEATURES OF TURKISH CHILDREN AND ADOLESCENTS WITH ADHD AND ADHD-NOS: A PRELIMINARY STUDY. Psychiatr Danub. 2024 Dec;36(3-4):333-339. doi: 10.24869/psyd.2024.333. PMID: 39724120. n = 81

  182. Meng H, Bigambo FM, Gu W, Wang X, Li Y (2024): Evaluation of thyroid function tests among children with neurological disorders. Front Endocrinol (Lausanne). 2024 Dec 9;15:1498788. doi: 10.3389/fendo.2024.1498788. PMID: 39717108; PMCID: PMC11663650. METASTUDY

  183. Chen G, Gao W, Xu Y, Chen H, Cai H (2023): Serum TSH Levels are Associated with Hyperactivity Behaviors in Children with Attention Deficit/Hyperactivity Disorder. Neuropsychiatr Dis Treat. 2023 Mar 7;19:557-564. doi: 10.2147/NDT.S402530. PMID: 36915908; PMCID: PMC10007977.

  184. Ornstein TJ, Sagar S, Schachar RJ, Ewing-Cobbs L, Chapman SB, Dennis M, Saunders AE, Yang TT, Levin HS, Max JE (2014): Neuropsychological performance of youth with secondary attention-deficit/hyperactivity disorder 6- and 12-months after traumatic brain injury. J Int Neuropsychol Soc. 2014 Nov;20(10):971-81. doi: 10.1017/S1355617714000903. PMID: 25489810; PMCID: PMC5010863.

  185. Ryan, Catroppa, Ward, Yeates, Crossley, Hollenkamp, Hearps, Beauchamp, Anderson (2022): Association of neurostructural biomarkers with secondary attention-deficit/hyperactivity disorder (ADHD) symptom severity in children with traumatic brain injury: a prospective cohort study. Psychol Med. 2022 Aug 25:1-10. doi: 10.1017/S0033291722002598. PMID: 36004807.

  186. Chang, Hsu, Wu, Huang, Chang, Bai, Chen, Chen (2018): Traumatic Brain Injury in Early Childhood and Risk of Attention-Deficit/Hyperactivity Disorder and Autism Spectrum Disorder: A Nationwide Longitudinal Study.; J Clin Psychiatry. 2018 Oct 16;79(6). pii: 17m11857. doi: 10.4088/JCP.17m11857.

  187. Vrijsen JN, Tendolkar I, Onnink M, Hoogman M, Schene AH, Fernández G, van Oostrom I, Franke B (2018): ADHD symptoms in healthy adults are associated with stressful life events and negative memory bias. Atten Defic Hyperact Disord. 2018 Jun;10(2):151-160. doi: 10.1007/s12402-017-0241-x. Epub 2017 Oct 28. PMID: 29081022; PMCID: PMC5973996.

  188. Sanchez EO, Bangasser DA (2022): The effects of early life stress on impulsivity. Neurosci Biobehav Rev. 2022 Jun;137:104638. doi: 10.1016/j.neubiorev.2022.104638. PMID: 35341796; PMCID: PMC9119952. REVIEW

  189. Karayağmurlu, Aytaç, Gülşen (2019): Relationship between otorhinologic trauma and Attention Deficit Hyperactivity Disorder symptoms in children. Int J Pediatr Otorhinolaryngol. 2019 Feb 8;120:89-92. doi: 10.1016/j.ijporl.2019.02.016.

  190. Brown AW, Esterov D, Zielinski MD, Weaver AL, Mara KC, Ferrara MJ, Immermann JM, Moir C (2023): Incidence and risk of attention-deficit/hyperactivity disorder and learning disability by adulthood after traumatic brain injury in childhood: a population-based birth cohort study. Child Neuropsychol. 2022 Oct 24:1-17. doi: 10.1080/09297049.2022.2136645. PMID: 36278854.

  191. Gerschman T, Brooks BL, Mrazik M, Eliason PH, Bonfield S, Yeates KO, Emery CA, Schneider KJ (2023): Are Self-Reported and Parent-Reported Attention Problems and Hyperactivity Associated With Higher Rates of Concussion in Youth Ice Hockey Players? Clin J Sport Med. 2022 Oct 5. doi: 10.1097/JSM.0000000000001080. PMID: 36731042.

  192. Laaksonen J, Ponkilainen V, Möttönen J, Mattila VM, Kuitunen I (2024): Paediatric traumatic brain injury and attention-deficit/hyperactivity disorder medication in Finland: a nationwide register-based cohort study. BMJ Ment Health. 2024 May 30;27(1):e301083. doi: 10.1136/bmjment-2024-301083. PMID: 39093719; PMCID: PMC11141179. n = 128.006

  193. Cortese S, Moreno C (2025): Advancing the evidence base for child and adolescent psychopharmacology. BMJ Ment Health. 2025 Mar 3;28(1):e301634. doi: 10.1136/bmjment-2025-301634. PMID: 40032555; PMCID: PMC11877239.

  194. Yi Y, Zhong C, Wei-Wei H (2023): The long-term neurodevelopmental outcomes of febrile seizures and underlying mechanisms. Front Cell Dev Biol. 2023 May 25;11:1186050. doi: 10.3389/fcell.2023.1186050. PMID: 37305674; PMCID: PMC10248510. REVIEW

  195. Bertelsen EN, Larsen JT, Petersen L, Christensen J, Dalsgaard S (2016): Childhood Epilepsy, Febrile Seizures, and Subsequent Risk of ADHD. Pediatrics. 2016 Aug;138(2):e20154654. doi: 10.1542/peds.2015-4654. PMID: 27412639.

  196. Ku YC, Muo CH, Ku CS, Chen CH, Lee WY, Shen EY, Chang YJ, Kao CH (2014): Risk of subsequent attention deficit-hyperactivity disorder in children with febrile seizures. Arch Dis Child. 2014 Apr;99(4):322-6. doi: 10.1136/archdischild-2013-304647. PMID: 24307684.

  197. Del-Ponte B, Quinte GC, Cruz S, Grellert M, Santos IS (2019): Dietary patterns and attention deficit/hyperactivity disorder (ADHD): A systematic review and meta-analysis. J Affect Disord. 2019 Jun 1;252:160-173. doi: 10.1016/j.jad.2019.04.061. PMID: 30986731. METASTUDY

  198. Voltas N, Jardí C, Hernández-Martínez C, Arija V, Canals J (2022): Association between free sugars intake and early psychopathological problems. J Child Health Care. 2022 Oct 25:13674935221135106. doi: 10.1177/13674935221135106. PMID: 36282888. n = 86

  199. Ferreira RC, Marin AH, Vitolo MR, Campagnolo PDB (2024): Early ultra-processed foods consumption and hyperactivity/inattention in adolescence. Rev Saude Publica. 2024 Oct 18;58:46. doi: 10.11606/s1518-8787.2024058005636. Erratum in: Rev Saude Publica. 2025 Jan 17;58:46err. doi: 10.11606/s1518-8787.2024058005636err. PMID: 39442121; PMCID: PMC11548911.

  200. Lee J, Lee SI, Lee YM, Hong YH (2023): Prevalence of Attention Deficit Hyperactivity Disorder in Girls With Central Precocious Puberty. J Atten Disord. 2023 Jul 14:10870547231180116. doi: 10.1177/10870547231180116. PMID: 37449382.

  201. Xu J, Wang F, Gao Y, Qi C, Chen T, Yan J (2024): The Relationship between Early Exposure to General Anesthesia and Neurobehavioral Deficits. Dev Neurosci. 2024 Oct 14:1-17. doi: 10.1159/000542005. PMID: 39401493. REVIEW

  202. Ing, Ma, Sun, Lu, Wall, Olfson, Li (2020): Exposure to Surgery and Anesthesia in Early Childhood and Subsequent Use of Attention Deficit Hyperactivity Disorder Medications. Anesth Analg. 2020 Jan 8. doi: 10.1213/ANE.0000000000004619. n = 256.122

  203. Song JY, Cha HR, Lee SW, Ha EK, Kim JH, Han MY (2023): Association Between Receipt of General Anesthesia During Childhood and Attention Deficit Hyperactive Disorder and Neurodevelopment. J Korean Med Sci. 2023 Feb 13;38(6):e42. doi: 10.3346/jkms.2023.38.e42. PMID: 36786086; PMCID: PMC9925326. n = 93.717

  204. Shi, Dykhoff, Guevara, Sangaralingham, Schroeder, Flick, Zaccariello, Warner (2021): Moderators of the association between attention-deficit/hyperactivity disorder and exposure to anaesthesia and surgery in children. Br J Anaesth. 2021 Sep 6:S0007-0912(21)00497-9. doi: 10.1016/j.bja.2021.07.025. PMID: 34503832. n = 185.002

  205. Sun M, Chen WM, Fu S, Wu SY (2023): Zhang J. Early childhood general anesthesia and risk of Attention Deficit Hyperactivity Disorder. J Child Psychol Psychiatry. 2023 Aug 3. doi: 10.1111/jcpp.13871. PMID: 37537781.

  206. Ing, Bellinger (2022): Long-term cognitive and behavioral outcomes following early exposure to general anesthetics. Curr Opin Anaesthesiol. 2022 Jul 6. doi: 10.1097/ACO.0000000000001155. PMID: 35788121.

  207. Holst, Kronborg, Jepsen, Christensen, Vejlstrup, Juul, Bjerre, Bilenberg, Ravn (2020): Transposition of the Great Arteries, and Tetralogy of Fallot. Attention-deficit/hyperactivity disorder symptoms in children with surgically corrected Ventricular Septal Defect, Cardiol Young. 2020 Jan 13:1-8. doi: 10.1017/S1047951119003184.

  208. Zhou H, Sun W, Ning L, Kang J, Jin Y, Dong C (2024): Early exposure to general anesthesia may contribute to later attention-deficit/hyperactivity disorder (ADHD): A systematic review and meta-analysis of cohort studies. J Clin Anesth. 2024 Nov;98:111585. doi: 10.1016/j.jclinane.2024.111585. PMID: 39153353. METASTUDY

  209. Yang, Wang, Chang, Ho, Kuo (2021): A National Population Cohort Study Showed That Exposure to General Anesthesia in Early Childhood Is Associated with an Increase in the Risk of Developmental Delay. Children (Basel). 2021 Sep 24;8(10):840. doi: 10.3390/children8100840. PMID: 34682104; PMCID: PMC8534755.

  210. Hou Y, Li S, Hou Q, Wang R, Xu X, Li Z, Ma L, Liu Q, Shen Y, Zheng H (2025): Vitamin K2 mitigates cognitive, and motor impairments induced by multiple surgery with anesthesia and analgesia in neonatal stage. Biochem Biophys Res Commun. 2025 Jun 8;765:151784. doi: 10.1016/j.bbrc.2025.151784. PMID: 40273626.

  211. Huang H, Zhang KP, Sun KK, Yu G (2024): Association between type 2 inflammatory diseases and neurodevelopmental disorders in low-birth-weight children and adolescents. Front Psychol. 2024 Feb 22;15:1292071. doi: 10.3389/fpsyg.2024.1292071. PMID: 38455122; PMCID: PMC10918750. n = 11.260

  212. Chuang YC, Wang CY, Huang WL, Wang LJ, Kuo HC, Chen YC, Huang YJ (2022): Two meta-analyses of the association between atopic diseases and core symptoms of attention deficit hyperactivity disorder. Sci Rep. 2022 Mar 1;12(1):3377. doi: 10.1038/s41598-022-07232-1. PMID: 35232975; PMCID: PMC8888762. METASTUDY

  213. Xu X, Li S, Chen Y, Deng X, Li J, Xiong D, Xie H (2025): Association between allergic diseases and mental health conditions: An umbrella review. J Allergy Clin Immunol. 2025 Mar;155(3):701-713. doi: 10.1016/j.jaci.2024.10.030. PMID: 39521284. REVIEW

  214. Wang Q, Wang R, Li M, Liang J, Zhan X, Lu Y, Huang G, Gu Q (2024): The relationship between allergic rhinitis and attention deficit hyperactivity disorder: a systematic review and meta-analysis. PeerJ. 2024 Oct 18;12:e18287. doi: 10.7717/peerj.18287. PMID: 39434793; PMCID: PMC11493030. METASTUDY

  215. Wang LJ, Yu YH, Fu ML, Yeh WT, Hsu JL, Yang YH, Chen WJ, Chiang BL, Pan WH (2018): Attention deficit-hyperactivity disorder is associated with allergic symptoms and low levels of hemoglobin and serotonin. Sci Rep. 2018 Jul 6;8(1):10229. doi: 10.1038/s41598-018-28702-5. PMID: 29980754; PMCID: PMC6035203.

  216. Wei J, Li Y, Wu Q, Lei B, Gui X (2025): Bidirectional association between allergic rhinitis and attention-deficit/hyperactivity disorder: A systematic review and meta-analysis. J Affect Disord. 2025 Jan 15;369:499-507. doi: 10.1016/j.jad.2024.10.032. PMID: 39389122. METASTUDY

  217. Jiang Z, Long X, Die X, Hou J, Wang Y, Wang Y, Feng W (2025): Causal effects of pediatric asthma on psychiatric disorders: a bidirectional Mendelian randomization study. J Asthma. 2025 Mar;62(3):445-455. doi: 10.1080/02770903.2024.2409418. PMID: 39320275.

  218. Buske-Kirschbaum, Schmitt, Plessow, Romanos, Weidinger, Roessner (2012): Psychoendocrine and psychoneuroimmunological mechanisms in the comorbidity of atopic eczema and attention deficit/hyperactivity disorder. Psychoneuroendocrinology. 2013 Jan;38(1):12-23. doi: 10.1016/j.psyneuen.2012.09.017. PMID: 23141851. REVIEW

  219. Wan J, Shin DB, Syed MN, Abuabara, Lemeshow, Gelfand (2022): Atopic dermatitis and risk of major neuropsychiatric disorders in children: a population-based cohort study. J Eur Acad Dermatol Venereol. 2022 Aug 26. doi: 10.1111/jdv.18564. PMID: 36018560. n = 2,4 Mio

  220. Ma EZ, Chang HR, Radtke S, Wan J (2024): Symptoms of Cognitive Impairment Among Children With Atopic Dermatitis. JAMA Dermatol. 2024 Mar 6:e240015. doi: 10.1001/jamadermatol.2024.0015. PMID: 38446433; PMCID: PMC10918570.

  221. Cao S, Zhang Z, Liu L, Li Y, Li W, Li Y, Wu D (2024): Causal relationships between atopic dermatitis and psychiatric disorders: a bidirectional two-sample Mendelian randomization study. BMC Psychiatry. 2024 Jan 3;24(1):16. doi: 10.1186/s12888-023-05478-1. PMID: 38172785; PMCID: PMC10763185.

  222. Liu Z, Wang Y, Wang S, Wu J, Jia C, Tan X, Liu X, Huang X, Zhang L (2024): Unraveling the causative connection between urticaria, inflammatory cytokines, and mental disorders: Perspectives from genetic evidence. Skin Res Technol. 2024 Sep;30(9):e13906. doi: 10.1111/srt.13906. PMID: 39300828; PMCID: PMC11413335.

  223. Wang HF, Liu S, Cao Y, Li QS (2025): Bidirectional association between atopic dermatitis and attention deficit hyperactivity disorder: a systematic review and meta-analysis. Ann Med. 2025 Dec;57(1):2483370. doi: 10.1080/07853890.2025.2483370. PMID: 40159827; PMCID: PMC11960313. METASTUDY

  224. Merzon E, Farag R, Ashkenazi S, Magen E, Manor I, Weizman A, Green I, Golan-Cohen A, Genshin A, Vinker S, Israel A (2024): Increased Prevalence of Attention Deficit Hyperactivity Disorder in Individuals with Selective Immunoglobulin A Deficiency: A Nationwide Case-Control Study. J Clin Med. 2024 Oct 12;13(20):6075. doi: 10.3390/jcm13206075. PMID: 39458025; PMCID: PMC11508521. n = 4.632

  225. Fuhrmann, Tesch, Romanos, Abraham, Schmitt (2020): ADHD in school-age children is related to infant exposure to systemic H1-antihistamines. Allergy. 2020 Nov;75(11):2956-2957. doi: 10.1111/all.14411. PMID: 32441335. n = 41.484

  226. 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.

  227. 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.

  228. 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.

  229. 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.

  230. 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.

  231. 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.

  232. Muller D, Signal TL, Shanthakumar M, Paine SJ (2023): Sleep inequities and associations between poor sleep and mental health for school-aged children: findings from the New Zealand Health Survey. Sleep Adv. 2023 Nov 18;4(1):zpad049. doi: 10.1093/sleepadvances/zpad049. PMID: 38084299; PMCID: PMC10710543.

  233. Koç HA, Önal BS (2024): Relationship between acne vulgaris and attention deficit hyperactivity disorder in adolescent: A cross-sectional study. J Dermatol. 2024 Aug 30. doi: 10.1111/1346-8138.17435. PMID: 39212308. n = 194

  234. 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.

  235. 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

  236. Joseph M, Lajili M, Dudoignon B, Delclaux C, Bokov P (2025): Loudness of snoring, not apnea-hypopnea index, is associated with hyperactivity behavior in snoring children and adolescents. Sleep Med. 2025 Mar;127:43-48. doi: 10.1016/j.sleep.2025.01.006. PMID: 39787820.

  237. 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.

  238. 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

  239. 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.

  240. 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

  241. 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.

  242. 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.

  243. 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.

  244. 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

  245. 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

  246. 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.

  247. Bundesinstitut für Risikobewertung: Information Nr. 037/2005 des BfR vom 12. Juli 2005: Durchschnittlicher Fluoridgehalt in Trinkwasser ist in Deutschland niedrig

  248. 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.

  249. 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.

  250. 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

  251. 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

  252. 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.

  253. van de Bor (2019): Fetal toxicology. Handb Clin Neurol. 2019;162:31-55. doi: 10.1016/B978-0-444-64029-1.00002-3.

  254. 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.

  255. 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.

  256. 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.

  257. 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.

  258. 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

  259. 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.

  260. 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

  261. 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.

  262. 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.

  263. 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.

  264. 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.

  265. http://www.adhs.org/genese/ mit weiteren Nachweisen

  266. 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

  267. http://www.adhs.org/genese/

  268. Müller, Candrian, Kropotov (2011): ADHS – Neurodiagnostik in der Praxis, Springer, Seite 88

  269. http://de.statista.com/statistik/daten/studie/261015/umfrage/praevalenz-des-rauchens-in-deutschland-nach-geschlecht/

  270. 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

  271. 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

  272. 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.

  273. 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.

  274. 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

  275. Pessah, Lein, Seegal, Sagiv (2019): Neurotoxicity of polychlorinated biphenyls and related organohalogens. Acta Neuropathol. 2019 Apr 11. doi: 10.1007/s00401-019-01978-1.

  276. 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.

  277. Chishti, Fisher, Seegal (1996): Aroclors 1254 and 1260 reduce dopamine concentrations in rat striatal slices. Neurotoxicology. 1996 Fall-Winter;17(3-4):653-60.

  278. 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

  279. 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.

  280. 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.

  281. 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.

  282. 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.

  283. 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.

  284. 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.

  285. 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.

  286. 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.

  287. 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.

  288. 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

  289. 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

  290. 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

  291. 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.

  292. 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.

  293. (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.

  294. 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

  295. 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.

  296. 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.

  297. 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

  298. 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

  299. 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.

  300. 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.

  301. 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.

  302. 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.

  303. 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

  304. 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.

  305. 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.

  306. 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.

  307. 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

  308. 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

  309. 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.

  310. 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.

  311. 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

  312. 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.

  313. 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.

  314. 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.

  315. 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.

  316. Ogonah MGT, Botchway S, Yu R, Schofield PW, Fazel S (2025): An umbrella review of health outcomes following traumatic brain injury. Nat Ment Health. 2025;3(1):83-91. doi: 10.1038/s44220-024-00356-5. PMID: 39802934; PMCID: PMC11717692. UMBRELLASTUDY

  317. Kaldoja ML, Nadendla KD, Ploetz D, Caldwell CT, Suskauer SJ, Svingos A, Busch TA, Slomine BS (2025): Exploring sex differences in pediatric mild traumatic brain injury: clinical characteristics and recovery patterns of pediatric mTBI patients with non-credible effort. Child Neuropsychol. 2025 Apr 15:1-17. doi: 10.1080/09297049.2025.2492134. PMID: 40235056.

  318. Niederhofer (2011): Association of Attention-Deficit/Hyperactivity Disorder and Celiac Disease: A Brief Report; Prim Care Companion CNS Disord. 2011; 13(3): PCC.10br01104; doi: 10.4088/PCC.10br01104; PMCID: PMC3184556, n = 67

  319. ähnlich: Okusaga, Yolken, Langenberg, Sleemi, Kelly, Vaswani, Giegling, Hartmann, Konte, Friedl, Mohyuddin, Groer, Rujescu, Postolache (2013): Elevated gliadin antibody levels in individuals with schizophrenia. World J Biol Psychiatry. 2013 Sep;14(7):509-15. doi: 10.3109/15622975.2012.747699.

  320. Lachance, McKenzie (2013): Biomarkers of gluten sensitivity in patients with non-affective psychosis: a meta-analysis. Schizophr Res. 2014 Feb;152(2-3):521-7. doi: 10.1016/j.schres.2013.12.001.

  321. Stevenson J, Sonuga-Barke E, McCann D, Grimshaw K, Parker KM, Rose-Zerilli MJ, Holloway JW, Warner JO (2010): The role of histamine degradation gene polymorphisms in moderating the effects of food additives on children’s ADHD symptoms. Am J Psychiatry. 2010 Sep;167(9):1108-15. doi: 10.1176/appi.ajp.2010.09101529. PMID: 20551163. RCT

  322. Rodriguez-Paris, Remlinger-Molenda, Kurzawa, Głowińska, Spaczyński, Rybakowski, Pawełczyk, Banaszewska (2019): Psychiatric disorders in women with polycystic ovary syndrome. Psychiatr Pol. 2019 Aug 31;53(4):955-966. doi: 10.12740/PP/OnlineFirst/93105.

  323. Trivedi C, Rizvi A, Ashraf S, Husain K, Yadav G, Adams E, Perugula M, Nabi S, Mansuri Z, Jain S (2024): Psychiatric Comorbidities in Hospitalized Adolescents With Polycystic Ovary Syndrome: A Cross-Sectional Study Using the National Inpatient Sample. Prim Care Companion CNS Disord. 2024 Nov 7;26(6):24m03760. doi: 10.4088/PCC.24m03760. PMID: 39546358.

  324. Merzon, Grossman, Vinker, Merhasin, Levit, Golan-Cohen (2020): Factors associated with withdrawal from insulin pump therapy: a large-population-based study. Diabetes Metab Res Rev. 2020 Jan 10. doi: 10.1002/dmrr.3288. n = 707

  325. Zhou Y, Jin B, Qiao K (2025): Investigating the causal relationships between attention-deficit/hyperactivity disorder and autoimmune diseases: Evidence from Mendelian randomization study. Medicine (Baltimore). 2025 Jan 3;104(1):e41157. doi: 10.1097/MD.0000000000041157. PMID: 40184135; PMCID: PMC11709194.

  326. Zhang-James Y, Draytsel D, Carguello B, Faraone SV, Weinstock RS (2025): Attention-Deficit/Hyperactivity Disorder Symptoms Are Common and Associated with Worse Glycemic Control in Adults with Type 1 Diabetes. J Clin Med. 2025 May 21;14(10):3606. doi: 10.3390/jcm14103606. PMID: 40429601; PMCID: PMC12112195.

  327. Welsh (1996): A prefrontal dysfunction model of early-treated phenylketonuria. Eur J Pediatr. 1996 Jul;155 Suppl 1:S87-9. doi: 10.1007/pl00014259. PMID: 8828618.

  328. Beckhauser, Beghini Mendes Vieira, Moehlecke Iser, Rozone, Luca, Rodrigues Masruha, Lin, Luiz Streck (2020): Attention Deficit Disorder with Hyperactivity Symptoms in Early-Treated Phenylketonuria Patients. Iran J Child Neurol. 2020 Winter;14(1):93-103. PMID: 32021633; PMCID: PMC6956970. n = 34

  329. Kildal, Hassel, Bjørnebekk (2022): ADHD symptoms and use of anabolic androgenic steroids among male weightlifters. Sci Rep. 2022 Jun 8;12(1):9479. doi: 10.1038/s41598-022-12977-w. PMID: 35676515; PMCID: PMC9178025.

  330. Nudel R, Hougaard DM, Werge T, Benros ME (2023): Genetic and epidemiological analyses of infection load and its relationship with psychiatric disorders. Epidemiol Infect. 2023 May 18;151:e93. doi: 10.1017/S0950268823000687. PMID: 37197974; PMCID: PMC10311684.

  331. Michos AG, Syriopoulou VP, Hadjichristodoulou C, Daikos GL, Lagona E, Douridas P, Mostrou G, Theodoridou M. (2007): Aseptic meningitis in children: analysis of 506 cases. PLoS One. 2007 Aug 1;2(7):e674. doi: 10.1371/journal.pone.0000674. PMID: 17668054; PMCID: PMC1933255.

  332. Fowlkes AL, Honarmand S, Glaser C, Yagi S, Schnurr D, Oberste MS, Anderson L, Pallansch MA, Khetsuriani N. (2008): Enterovirus-associated encephalitis in the California encephalitis project, 1998-2005. J Infect Dis. 2008 Dec 1;198(11):1685-91. doi: 10.1086/592988. PMID: 18959496.

  333. Balestrieri E, Matteucci C, Cipriani C, Grelli S, Ricceri L, Calamandrei G, Vallebona PS (2019): Endogenous Retroviruses Activity as a Molecular Signature of Neurodevelopmental Disorders. Int J Mol Sci. 2019 Nov 30;20(23):6050. doi: 10.3390/ijms20236050. PMID: 31801288; PMCID: PMC6928979. REVIEW

  334. (Chou IC, Lin CC, Kao CH (2015): Enterovirus Encephalitis Increases the Risk of Attention Deficit Hyperactivity Disorder: A Taiwanese Population-based Case-control Study. Medicine (Baltimore). 2015 Apr;94(16):e707. doi: 10.1097/MD.0000000000000707. PMID: 25906098; PMCID: PMC4602682.

  335. Lin HY, Chen YL, Chou PH, Gau SS, Chang LY (2022): Long-term psychiatric outcomes in youth with enterovirus A71 central nervous system involvement. Brain Behav Immun Health. 2022 Jun 1;23:100479. doi: 10.1016/j.bbih.2022.100479. PMID: 35694176; PMCID: PMC9184869.

  336. Goh M, Joy C, Gillespie AN, Soh QR, He F, Sung V (2023): Asymptomatic viruses detectable in saliva in the first year of life: a narrative review. Pediatr Res. 2023 Dec 22. doi: 10.1038/s41390-023-02952-0. PMID: 38135726. REVIEW

  337. Chang LY, Huang LM, Gau SS, Wu YY, Hsia SH, Fan TY, Lin KL, Huang YC, Lu CY, Lin TY (2007): Neurodevelopment and cognition in children after enterovirus 71 infection. N Engl J Med. 2007 Mar 22;356(12):1226-34. doi: 10.1056/NEJMoa065954. PMID: 17377160.

  338. Nozyce, Lee SS, Wiznia, Nachman, Mofenson, Smith, Yogev, McIntosh, Stanley, Pelton (2006): A behavioral and cognitive profile of clinically stable HIV-infected children. Pediatrics. 2006 Mar;117(3):763-70. doi: 10.1542/peds.2005-0451. PMID: 16510656. n = 274

  339. Dale, Church, Heyman (2003): Striatal encephalitis after varicella zoster infection complicated by Tourettism. Mov Disord. 2003 Dec;18(12):1554-6. doi: 10.1002/mds.10610. PMID: 14673900.

  340. Wilson, Thomas (2022): Peridontitis as a Risk Factor for Attention Deficit Hyperactivity Disorder: Possible Neuro-inflammatory Mechanisms. Neurochem Res. 2022 Jun 28. doi: 10.1007/s11064-022-03650-9. PMID: 35764847.

  341. Elmehy DA, Elmansory BM, Gamea GA, Abdelhai DI, Abd-Elsalam SM, Salamah AM, Ata DS, Mahmoud EF, Ibrahim HA, Salama AM (2023): Parasitic infections as potential risk factors for attention deficit hyperactivity disorder (ADHD) in children. J Parasit Dis. 2023 Mar;47(1):82-92. doi: 10.1007/s12639-022-01542-x. PMID: 36910322; PMCID: PMC9998788.

  342. Merzon E, Magen E, Ashkenazi S, Weizman A, Manor I, Krone B, Green I, Golan-Cohen A, Vinker S, Faraone SV, Israel A (2023): The Association between Glucose 6-Phosphate Dehydrogenase Deficiency and Attention Deficit/Hyperactivity Disorder. Nutrients. 2023 Nov 29;15(23):4948. doi: 10.3390/nu15234948. PMID: 38068806; PMCID: PMC10708268.

  343. Hsu CW, Lin YW, Chen YB, Wang LJ, Kuo HC (2025): Association of Kawasaki disease with intellectual disability, attention deficit hyperactivity disorder, and autism spectrum disorder: a systematic review and meta-analysis. Ital J Pediatr. 2025 Feb 21;51(1):52. doi: 10.1186/s13052-025-01897-w. PMID: 39985017; PMCID: PMC11846269. METASTUDY

  344. Daniels LB, Roberts S, Moreno E, Tremoulet AH, Gordon JB, Burns JC (2022): Long-term health outcomes in young adults after Kawasaki disease. Int J Cardiol Heart Vasc. 2022 May 4;40:101039. doi: 10.1016/j.ijcha.2022.101039. PMID: 35573651; PMCID: PMC9096130. n = 406

  345. Kuo HC, Chang WC, Wang LJ, Li SC, Chang WP (2016): Association of Attention deficit hyperactivity disorder and Kawasaki disease: a nationwide population-based cohort study. Epidemiol Psychiatr Sci. 2016 Dec;25(6):573-580. doi: 10.1017/S2045796015000840. PMID: 26392050; PMCID: PMC7137668. n = 651

  346. Lin CH, Lin WD, Chou IC, Lee IC, Hong SY (2019): Heterogeneous neurodevelopmental disorders in children with Kawasaki disease: what is new today? BMC Pediatr. 2019 Nov 4;19(1):406. doi: 10.1186/s12887-019-1786-y. PMID: 31684911; PMCID: PMC6827201. n = 612

  347. Demir T, Simsir IY, Tuncel OK, Ozbaran B, Yildirim I, Pirildar S, Ozen S, Akinci B (2024): Impact of lipodystrophy on health-related quality of life: the QuaLip study. Orphanet J Rare Dis. 2024 Jan 5;19(1):10. doi: 10.1186/s13023-023-03004-w. PMID: 38183080; PMCID: PMC10768358.

  348. Keselman D, Glanzman A, Thelen MY, Prosser LA, McGuire J, Matesanz SE (2025): Motor function testing rates and outcomes in Duchenne muscular dystrophy with comorbid autism and attention-deficit/hyperactivity disorder. Neuromuscul Disord. 2025 Mar;48:105281. doi: 10.1016/j.nmd.2025.105281. PMID: 39946777.

  349. Shen F, Zhou H (2024): Methylphenidate treatment of a Chinese boy with Becker muscular dystrophy combined with attention deficit hyperactivity disorder: a case report. Front Neurosci. 2024 Nov 26;18:1459582. doi: 10.3389/fnins.2024.1459582. PMID: 39659883; PMCID: PMC11628498.

  350. Vaillend C, Aoki Y, Mercuri E, Hendriksen J, Tetorou K, Goyenvalle A, Muntoni F (2025): Duchenne muscular dystrophy: recent insights in brain related comorbidities. Nat Commun. 2025 Feb 3;16(1):1298. doi: 10.1038/s41467-025-56644-w. PMID: 39900900; PMCID: PMC11790952. REVIEW

  351. Diehl E, O’Neill M, Gray L, Schwaede A, Kuntz N, Rao VK (2024): Prevalence of Attention-Deficit/Hyperactivity Disorder and Autism Spectrum Disorder in Individuals With Dystrophinopathy at a Tertiary Care Center in Chicago. Pediatr Neurol. 2024 May 19;158:94-99. doi: 10.1016/j.pediatrneurol.2024.05.011. PMID: 39024712.

  352. Göbel A, Heldmann M, Göttlich M, Dirk AL, Brabant G, Münte TF (2016): Effect of Mild Thyrotoxicosis on Performance and Brain Activations in a Working Memory Task. PLoS One. 2016 Aug 18;11(8):e0161552. doi: 10.1371/journal.pone.0161552. PMID: 27536945; PMCID: PMC4990413.

  353. Bauer M, Goetz T, Glenn T, Whybrow PC (2008): The thyroid-brain interaction in thyroid disorders and mood disorders. J Neuroendocrinol. 2008 Oct;20(10):1101-14. doi: 10.1111/j.1365-2826.2008.01774.x. PMID: 18673409. REVIEW

  354. Göttlich M, Chatterjee K, Moran C, Heldmann M, Rogge B, Cirkel A, Brabant G, Münte TF (2024): Altered brain functional connectivity in patients with resistance to thyroid hormone ß. PLoS One. 2024 Aug 22;19(8):e0306538. doi: 10.1371/journal.pone.0306538. PMID: 39172991; PMCID: PMC11341041.

  355. Smith, Gozal, Hunter, Kheirandish-Gozal (2017): Parent-Reported Behavioral and Psychiatric Problems Mediate the Relationship between Sleep-Disordered Breathing and Cognitive Deficits in School-Aged Children. Front Neurol. 2017 Aug 11;8:410. doi: 10.3389/fneur.2017.00410. eCollection 2017.

  356. Lewczuk K, Marcowski P, Wizła M, Gola M, Nagy L, Koós M, Kraus SW, Demetrovics Z, Potenza MN, Ballester-Arnal R, Batthyány D, Bergeron S, Billieux J, Briken P, Burkauskas J, Cárdenas-López G, Carvalho J, Castro-Calvo J, Chen L, Ciocca G, Corazza O, Csako RI, Fernandez DP, Fujiwara H, Fernandez EF, Fuss J, Gabrhelík R, Gewirtz-Meydan A, Gjoneska B, Grubbs JB, Hashim HT, Islam MS, Ismail M, Jiménez-Martínez MC, Jurin T, Kalina O, Klein V, Költő A, Lee SK, Lin CY, Lin YC, Lochner C, López-Alvarado S, Lukavská K, Mayta-Tristán P, Miller DJ, Orosová O, Orosz G; Sungkyunkwan University’s research team; Ponce FP, Quintana GR, Quintero Garzola GC, Ramos-Diaz J, Rigaud K, Rousseau A, Tubino Scanavino M, Schulmeyer MK, Sharan P, Shibata M, Shoib S, Sigre-Leirós V, Sniewski L, Spasovski O, Steibliene V, Stein DJ, Ünsal BC, Vaillancourt-Morel MP, Claire Van Hout M, Bőthe B (2024): Cross-Cultural Adult ADHD Assessment in 42 Countries Using the Adult ADHD Self-Report Scale Screener. J Atten Disord. 2024 Feb;28(4):512-530. doi: 10.1177/10870547231215518. PMID: 38180045.

  357. Nadeem A, Donohue S, Shah FZ, Perez JA, Harper E, Sinh P, Padival R, Cooper G, Katz J, Cominelli F, Regueiro M, Mansoor E (2024): Early Onset Active Inflammatory Bowel Disease Is Associated With Psychiatric Comorbidities: A Multi-Network Propensity-Matched Cohort Study. Crohns Colitis 360. 2024 Dec 20;7(1):otae066. doi: 10.1093/crocol/otae066. PMID: 39749210; PMCID: PMC11694680.

  358. Butwicka A, Olén O, Larsson H, Halfvarson J, Almqvist C, Lichtenstein P, Serlachius E, Frisén L, Ludvigsson JF (2019): Association of Childhood-Onset Inflammatory Bowel Disease With Risk of Psychiatric Disorders and Suicide Attempt. JAMA Pediatr. 2019 Oct 1;173(10):969-978. doi: 10.1001/jamapediatrics.2019.2662. PMID: 31424531; PMCID: PMC6704748.

  359. Yang X, Yang L, Zhang T, Zhang H, Chen H, Zuo X (2023): Causal atlas between inflammatory bowel disease and mental disorders: a bi-directional 2-sample Mendelian randomization study. Front Immunol. 2023 Oct 13;14:1267834. doi: 10.3389/fimmu.2023.1267834. PMID: 37901213; PMCID: PMC10611497.

  360. Kappel RK, Bisgaard TH, Poulsen G, Jess T (2024): Risk of Anxiety, Depression, and Attention-Deficit/Hyperactivity Disorder in Pediatric Patients With Inflammatory Bowel Disease: A Population-Based Cohort Study. Clin Transl Gastroenterol. 2024 Apr 1;15(4):e00657. doi: 10.14309/ctg.0000000000000657. PMID: 37934192; PMCID: PMC11042764.

  361. Shero N, Pandeya DR (2022): Impact of Crohn’s disease during pregnancy on children with attention deficit hyperactivity disorder: A review. Ann Med Surg (Lond). 2022 Feb 11;75:103369. doi: 10.1016/j.amsu.2022.103369. PMID: 35198194; PMCID: PMC8851270. REVIEW

  362. Pinto Payares DV, Spooner L, Vosters J, Dominguez S, Patrick L, Harris A, Kanungo S (2024): A systematic review on the role of mitochondrial dysfunction/disorders in neurodevelopmental disorders and psychiatric/behavioral disorders. Front Psychiatry. 2024 Jun 28;15:1389093. doi: 10.3389/fpsyt.2024.1389093. PMID: 39006821; PMCID: PMC11239503.

  363. Zayats T, Johansson S, Haavik J (2015): Expanding the toolbox of ADHD genetics. How can we make sense of parent of origin effects in ADHD and related behavioral phenotypes? Behav Brain Funct. 2015 Oct 16;11(1):33. doi: 10.1186/s12993-015-0078-4. PMID: 26475699; PMCID: PMC4609130. REVIEW

  364. Yackobovitch-Gavan M, Ben-Hefer D, Feldhamer I, Meyerovitch J (2024): The association between infantile microcytic anemia and attention deficit hyperactivity disorder, a case-control study. Heliyon. 2024 Jun 22;10(12):e33430. doi: 10.1016/j.heliyon.2024.e33430. PMID: 39015811; PMCID: PMC11250855.

  365. Köder F, Sharma C, Cameron S, Garraffa M (2022):The effects of bilingualism on cognition and behaviour in individuals with attention deficits: A scoping review. Front Psychol. 2022 Dec 23;13:1057501. doi: 10.3389/fpsyg.2022.1057501. PMID: 36619112; PMCID: PMC9816333. METASTUDIE

  366. Blasbalg U, Paz Toren (2024): The association between systemic lupus erythematosus (SLE) and early psychiatric background. J Psychiatr Res. 2024 Dec;180:349-354. doi: 10.1016/j.jpsychires.2024.10.029. PMID: 39520766.

  367. Huangfu N, Lu Y, Ma H, Hu Z, Cui H, Yang F (2023): Genetic liability to mental disorders in relation to the risk of hypertension. Front Cardiovasc Med. 2023 Feb 27;10:1087251. doi: 10.3389/fcvm.2023.1087251. PMID: 36923957; PMCID: PMC10008891.

  368. Chen F, Cao H, Baranova A, Zhao Q, Zhang F (2023): Causal associations between COVID-19 and childhood mental disorders. BMC Psychiatry. 2023 Dec 8;23(1):922. doi: 10.1186/s12888-023-05433-0. PMID: 38066446; PMCID: PMC10704772.

  369. Norbom LB, Syed B, Kjelkenes R, Rokicki J, Beauchamp A, Nerland S, Kushki A, Anagnostou E, Arnold P, Crosbie J, Kelley E, Nicolson R, Schachar R, Taylor MJ, Westlye LT, Tamnes CK, Lerch JP (2025): Probing Autism and ADHD subtypes using cortical signatures of the T1w/T2w-ratio and morphometry. Neuroimage Clin. 2025;45:103736. doi: 10.1016/j.nicl.2025.103736. PMID: 39837011; PMCID: PMC11788868.

  370. Zhang YX, Yang H, Guo Q, Zhang RF (2024): Investigating the causal relationship between childhood obesity and attention deficit hyperactivity disorder: A Mendelian randomization study. Asian J Surg. 2024 Dec 2:S1015-9584(24)02614-9. doi: 10.1016/j.asjsur.2024.11.012. PMID: 39627048.

  371. Van Assche IA, Van Calsteren K, Huis In ’t Veld EA, van Gerwen M, Heylen L, LeJeune CL, Cardonick E, Halaska MJ, Fruscio R, Fumagalli M, van Dijk-Lokkart EM, Lemiere J, van Grotel M, Lagae L, van den Heuvel-Eibrink MM, Amant F (2024): Child outcomes after prenatal exposure to platinum and taxane-based chemotherapy: an unplanned interim analysis of the international network on cancer, infertility, and pregnancy study. EClinicalMedicine. 2024 Nov 11;78:102922. doi: 10.1016/j.eclinm.2024.102922. PMID: 39588212; PMCID: PMC11585789.

  372. Hansen, Qureshi, Gele, Hauge, Biele, Surén, Kjøllesdal (2023): Developmental disorders among Norwegian-born children with immigrant parents. Child Adolesc Psychiatry Ment Health. 2023 Jan 6;17(1):3. doi: 10.1186/s13034-022-00547-x. PMID: 36609392; PMCID: PMC9825022.

  373. Chang J, Lee YJ, Lex H, Kerns C, Lugar K, Wright M (2023): Attention-Deficit Hyperactivity Disorder among children of immigrants: immigrant generation and family poverty. Ethn Health. 2023 Dec 17:1-13. doi: 10.1080/13557858.2023.2293657. PMID: 38105627. n = 83.362

  374. Tetens MM, Graham EE, Andersen NS, Bangsborg J, Bodilsen J, Dessau RB, Ellermann-Eriksen S, Jørgensen CS, Møller JK, Nielsen ACY, Pedersen M, Søgaard KK, Obel D, Nygaard U, Obel N, Lebech AM, Omland LH (2025): No associations between neuroborreliosis in children and psychiatric neurodevelopmental disorders: a nationwide, population-based, matched cohort study. J Child Psychol Psychiatry. 2025 May;66(5):716-724. doi: 10.1111/jcpp.14079. PMID: 39618006. n = 12.452

Diese Seite wurde am 13.09.2025 zuletzt aktualisiert.