Next Article in Journal
Dietary Patterns and Their Relationship with Frailty in Functionally Independent Older Adults
Next Article in Special Issue
Lutein and Zeaxanthin Are Positively Associated with Visual–Spatial Functioning in Older Adults: An fMRI Study
Previous Article in Journal
Folinic Acid Increases Protein Arginine Methylation in Human Endothelial Cells
Previous Article in Special Issue
Macular Xanthophylls Are Related to Intellectual Ability among Adults with Overweight and Obesity
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Rationale for Dietary Antioxidant Treatment of ADHD

by
Annelies A. J. Verlaet
1,*,
Carlijn M. Maasakkers
1,2,3,
Nina Hermans
1 and
Huub F. J. Savelkoul
3
1
Department of Pharmaceutical Sciences, Laboratory of Nutrition and Functional Food Science, University of Antwerp, Universiteitsplein 1 (A104), 2610 Wilrijk, Belgium
2
Department of Geriatric Medicine, Radboud Institute for Health Sciences, Radboud University Medical Center, P.O. Box 9101, 6500 HB Nijmegen, The Netherlands
3
Cell Biology and Immunology Group, Wageningen University, De Elst 1 Building 122, 6709 PG Wageningen, The Netherlands
*
Author to whom correspondence should be addressed.
Nutrients 2018, 10(4), 405; https://doi.org/10.3390/nu10040405
Submission received: 15 February 2018 / Revised: 12 March 2018 / Accepted: 21 March 2018 / Published: 24 March 2018
(This article belongs to the Special Issue Nutrition and the Function of the Central Nervous System)

Abstract

:
Increasing understanding arises regarding disadvantages of stimulant medication in children with ADHD (Attention-Deficit Hyperactivity Disorder). This review presents scientific findings supporting dietary antioxidant treatment of ADHD and describes substantial alterations in the immune system, epigenetic regulation of gene expression, and oxidative stress regulation in ADHD. As a result, chronic inflammation and oxidative stress could develop, which can lead to ADHD symptoms, for example by chronic T-cell-mediated neuroinflammation, as well as by neuronal oxidative damage and loss of normal cerebral functions. Therefore, modulation of immune system activity and oxidant-antioxidant balance using nutritional approaches might have potential in ADHD treatment. The use of natural antioxidants against oxidative conditions is an emerging field in the management of neurodegenerative diseases. Dietary polyphenols, for example, have antioxidant capacities as well as immunoregulatory effects and, therefore, appear appropriate in ADHD therapy. This review can stimulate the development and investigation of dietary antioxidant treatment in ADHD, which is highly desired.

1. Introduction: Attention-Deficit Hyperactivity Disorder

1.1. Prevalence and Diagnosis

Attention-Deficit Hyperactivity Disorder (ADHD) is a common neurocognitive behavioral disorder with a worldwide prevalence of 5.9–7.1%. It is associated with other psychiatric disorders, such as oppositional defiant disorder (ODD), autism and anxiety [1,2]. The 5th edition of the Diagnostic and Statistical Manual of Mental Disorders (DSM-5TM), used for the formal diagnosis of ADHD, describes ADHD as “a persistent and pervasive pattern of inattention and/or hyperactivity-impulsivity that interferes with functioning or development” and resulting in “a negative impact on social and academic/occupational activities”. The symptoms must have an onset before the age of twelve, persist for half a year, and be present in the home and school environment. Symptoms related to the inattentive subtype include difficulties in organizing tasks, often losing things needed for activities, and difficulties in sustaining attention. With regard to the hyperactivity-impulsivity subtype, symptoms include excessive fidgeting with hands or squirming in seat, talking excessively, and having difficulties in waiting for a turn. The combination-type of ADHD contains symptoms from both manifestations [3,4].

1.2. Etiology

Despite its high prevalence, no straightforward indication can yet be given about the underlying mechanisms of ADHD due to its complex etiology [5]. Especially, the heterogeneity and interchangeability of the involved factors make the pathophysiology unclear [6]. However, multiple factors related to the cause of the disorder have generally been accepted, such as genetic, biochemical, psychological and environmental factors [7].
Moreover, developmental trajectory studies show a delay in cortical maturation in ADHD, most profound in prefrontal regions which are important for attention and motor planning [8]. In addition, an overall dysfunction in the dopaminergic and noradrenergic system is involved in ADHD [9,10,11]. These catecholamines are active in brain areas involved in the regulation of attention and behavior [12]. Indeed, destruction of dopamine pathways in the prefrontal cortex resulted in hyperactivity [13], while concentrations of catecholamines are higher in the urine of ADHD patients compared to controls [14,15]. Moreover, ADHD medication modifies the uptake of catecholamines by neurons, thereby enhancing the activity of neurotransmitters and reducing the symptoms of ADHD [16]. Environmental factors, including prenatal alcohol exposure and maternal smoking, but also malnutrition (e.g., imbalance in omega 3 and 6 polyunsaturated fatty acids (PUFAs)) and deficiencies (e.g., zinc, magnesium) are implicated in the risk to develop ADHD [17].

1.2.1. Genetics

Generally, a strong genetic contribution (with a heritability ranging from 0.6 to 0.9) is found for ADHD, with several candidate genes, including genes involved in dopamine, noradrenaline, and serotonin transmission and metabolism, but each of these genes individually only slightly increases the risk of ADHD [5]. Genome-wide analyses demonstrated that many traits genetically correlated as well as that shared genetic susceptibilities exist between different somatic and psychiatric disorders, e.g., the SLC9A9 gene has been implicated in both ADHD and multiple sclerosis [18,19].
As an example, genetic variations are seen in ADHD for the dopamine active transporter (DAT) gene. Dopamine is released by activated neurons into the synaptic cleft to function as a neurotransmitter and can subsequently be taken up by DAT, which stops the signal transduction [17]. On average, the activity of this transporter is 14% higher in ADHD patients than in controls [5]. As a result, the time for dopamine to exert an effect in the synaptic cleft is significantly shorter in patients than in controls [11]. Medication such as methylphenidate (MPH), e.g., Ritalin®, targets this protein [5]. By binding to DAT1, MPH blocks this transporter, thereby increasing extracellular dopamine and norepinephrine levels [20,21].
Furthermore, dopamine receptors are involved, located on the dendrite of the adjacent neuron and signaling when dopamine is present in the synaptic cleft. In ADHD patients, DRD4 (dopamine receptor D4) gene is expressed less abundantly in brain areas involved in attention, such as the anterior cingulate cortex. Consequently the signal is transduced less to the next neuron [5].
Other genes mentioned in relation to ADHD are DRD1, DRD2 and DRD5 [16]. The norepinephrine transporter gene (NET1), which is responsible for the reuptake of norepinephrine, and the alpha-2A-adrenergic receptor (ADRA2A), which regulates epinephrine release, also potentially play a role [5].

1.2.2. Perinatal Influences

Also related to pregnancy, various factors have been associated with an increased risk on ADHD, such as maternal smoking, low birth weight, preterm birth and small size for gestational age. In addition, it is also increasingly accepted that a robust association exists between ADHD in the offspring and various maternal disorders, such as viral and bacterial infections, allergy and specific autoimmune diseases. Associated pro-inflammatory cytokines could affect neuronal development directly or via epigenetic pathways. Other factors that could mediate the effect between maternal diseases and ADHD are medication, more contact with health care services and genetics [19]. Indeed, use of specific medications during pregnancy, like anti-asthmatic drugs, acetaminophen, antipsychotics and selective serotonin reuptake inhibitors, is associated with an increased risk of birth defects, hyperkinetic disorder and altered neurobehavioral development in offspring. Acetaminophen may for instance act as a hormone disruptor, interfering with thyroid function, which is important for brain development. Women with chronic immunological disorders use such medications more frequently during pregnancy than women without such disorders [22,23,24].

1.3. Treatment Options

The conventional treatment for ADHD consists of behavioral and educational interventions, often combined with psychostimulant medication. The psychostimulants MPH and dextroamphetamine, and non-stimulating prefrontal cortex noradrenaline reuptake inhibitor atomoxetine, are the standard treatments for ADHD. MPH is a central nervous system stimulant as it increases attention and reduces hyperactivity and impulsivity by inhibition of dopamine reuptake in the striatum, without triggering its release [25]. However, neurotransmitters enable synaptic communication, but are also involved in central nervous system (CNS) development, including morphogenesis and the proliferation, migration and differentiation of neurons. Therefore, any interference with neurotransmitter systems potentially produces acute and/or long-lasting alterations in CNS structure and function. In fact, neurotoxicants associated with ADHD were found to damage dopaminergic neurons and dopaminergic and noradrenergic dysfunction is considered to underlie ADHD behavior, as the volume and activity of these brain areas, typically involved in attention, emotion and behavior, are reduced in patients [26].
Moreover, while ADHD medication does reduce symptoms in the majority of patients, most of them experience side effects within the first 6 months of MPH use [25,27]. A review reports adverse effects, like insomnia and decreased appetite, in about 25% of patients using MPH [28]. Other adverse effects include weight loss, abdominal pain, sleep disturbances, headaches, irritability and depressed mood and appetite, with some reports of stimulant induced psychosis. MPH is prescribed for chronic use to a large proportion of ADHD patients, but is linked to possible publication bias in reported efficacy. In addition, parents are often disinclined to use MPH due to its negative publicity and its frequent side effects, and subsequently, non-adherence to therapy is high [17,27,29,30,31,32,33,34,35]. Furthermore, pharmacotherapy may be unable to normalize functioning, with only small academic improvements, and within two years, the mind and body apparently get used to the medication, so that most patients no longer experience any difference in symptoms. Additionally, possibly part of the efficacy of the medication is a result of the extra attention that has been given to the child or a placebo response [25,36].
Increasing understanding regarding disadvantages of stimulant medication in children stimulates the investigation of alternative therapeutics [37]. Development of interventions that successfully target functional deficits is vital to improve the long-term outcomes of children with ADHD. More specifically, as previous research points at substantial alterations in the immune system and oxidative stress regulation in ADHD, a novel therapy based on improvement of those systems might have more potential than conventional therapy. This review therefore presents an overview of existing literature on oxidative stress, immunity and nutritional factors in ADHD and an update on present insights supporting antioxidant treatment in ADHD.

2. Oxidative Stress in ADHD

2.1. Oxidants

Reactive oxygen species (ROS) are oxygen-derived radicals and non-radicals that are oxidising agents and/or easily converted into radicals [38]. Similarly, reactive nitrogen species (RNS) are derived from oxygen and nitrogen. Different mechanisms generate ROS and RNS in vivo. Mitochondria are the most important source of ROS, generated during energy production, but also enzymatic mechanisms, exposure to UV-radiation, pollutants, cigarette smoke and the immune system during the development of inflammation contribute substantially. The two main sources for ROS production within the central nervous system are mitochondria and activated microglia (resident innate immune cells in the brain) to destroy pathogens or attack abnormal proteins accumulating in the brain [12]. Low levels of ROS are involved in regulating gene expression and signaling pathways, among other functions, and are therefore required for normal cellular function. As radicals are highly reactive molecules which can damage carbohydrates, nucleic acids, lipids and proteins and thereby alter their functions, an excess of ROS can damage cells [39,40,41,42].

2.2. Antioxidants

Multiple mechanisms exist to reduce ROS accumulation. First of all, by creating a high oxygen gradient between intracellular and extracellular compartments, oxygen diffuses out of the cell, thereby preventing intracellular ROS formation. In addition, free radicals can be detoxified by non-enzymatic and enzymatic antioxidants [43]. Other mechanisms exist to repair, in a direct and indirect way, oxidative damage to cellular structures. During direct repair, specific enzymes reduce oxidized products, such as proteins. During indirect repair, the damaged part is recognized and eliminated [44].
Different types of antioxidants protect different parts of the body, as some are water soluble (e.g., vitamin C), while some are fat soluble (vitamin E, coenzyme Q10). In addition, some antioxidants can penetrate membranes easily, including the ability to cross the blood-brain barrier (BBB), while others cannot. Antioxidant enzymes include catalase (CAT), superoxide dismutase (SOD) and glutathione peroxidases (GSH-Px), among others [45,46]. The intracellular antioxidant glutathione (GSH) is a major non-enzymatic antioxidant, present in all cell compartments. The ratio of reduced and oxidized glutathione (GSH/GSSG) is the major determinant of oxidative stress. Other non-enzymatic antioxidants include vitamin C, vitamin E, vitamin A, carotenoids, coenzyme Q, uric acid, albumin and bilirubin [43,47]. Total antioxidant status (TAS) represents the cumulative effect of all antioxidants present in plasma. The same is true for total oxidant status (TOS), respectively for oxidants present. The oxidative stress index (OSI, TOS/TAS) represents the current oxidative stress status. In certain cases, no differences can be found in individual antioxidant levels, though a difference in total antioxidant status is found. This underlines the cumulative effect of certain enzymes and substances or the involvement of other antioxidant mechanisms [7].

2.3. Oxidative Stress and Oxidative Damage

The shift in the balance between oxidants and antioxidant mechanisms in favor of oxidants is termed “oxidative stress”. Oxidative stress can generate damage to, e.g., lipids, proteins and DNA, thereby for example altering signal transduction and gene expression, inhibiting protein function, and promoting cell death [48,49]. Biomarkers of oxidative damage formed are more reflective of the actual oxidative stress situation compared to antioxidant levels, since even relative high antioxidant levels can still be too low to balance high oxidant levels [50].
Lipid peroxidation is a free-radical-mediated oxidative deterioration to which PUFAs are especially vulnerable due to their unconjugated double bonds [51]. By damaging fatty acid side chains, lipid peroxidation affects both membranes and lipoproteins. It decreases membrane fluidity, increases membrane leakiness and inactivates membrane bound enzymes and receptors [41]. In addition, when free radicals react with PUFAs via their carbon-carbon double bond, lipid hydroperoxide is formed, which is unstable and therefore subjected to fragmentation into different products, such as malondialdehyde (MDA) and 4-hydroxy-2-nonenal [52]. Therefore, MDA is used as an oxidative damage marker [47,53]. Besides MDA, F2-isoprostanes are a marker of oxidative lipid damage, formed when essential fatty acids, especially arachidonic acid, are oxidized [47].
3-nitrotyrosine is a marker of oxidative damage to proteins, and is formed by a reaction of RNS, such as peroxynitrite (ONOO), with tyrosine [54]. Oxidative damage to proteins includes primary damage to enzymes, receptors and transport proteins, as well as secondary damage to other biomolecules (e.g., increased mutation frequency due to damaged DNA-repair enzymes) [55,56].
Of all nucleobases, guanine is most prone to oxidation [57]. When deoxyguanosine is oxidized, a hydroxyl group is added to the 8th position of guanine forming 8-hydroxy-2′-deoxyguanosine (8-OHdG) [47,57]. This oxidation causes a shift of a G-C base pair into an A-T base pair in DNA [47]. When DNA is repaired, 8-OHdG is excreted in urine [57,58]. Metabolites of oxidized RNA, such as the oxidized nucleoside 8-hydroxy-2-guanosine (8-OHG), are found in urine as well [57]. Oxidative stress can disturb DNA structure by degradation or modification of bases, single or double strand breaks, or cross-linking with proteins. Such damage can lead to mutations and problems with DNA replication, arrest or induction of transcription and genomic instability, and thereby to alteration of gene expression, unless adjusted sufficiently by DNA repair mechanisms [48].

2.4. Oxidative Stress in ADHD

Multiple effects of oxidative stress on the brain can be related to ADHD aetiology [10]. The brain is particularly sensitive to oxidative stress partly due to its high metabolic rate, while oxidative stress can impair neuronal proliferation and mediate apoptosis. Elevated oxidative stress can therefore lead to progressive neuronal damage and deterioration of normal cerebral functions. For instance, increased oxidative stress could lead, through peroxidation of membrane lipids and membrane-associated proteins (e.g., neurotransmitter receptors), to structural neuronal damage with decreased membrane fluidity and altered neurotransmitter receptor and transporter functioning, and thereby to abnormal neurotransmitter regulation [10,12,59]. Furthermore, oxidative stress may interfere with dopamine synthesis, neuronal cell migration, and neuronal plasticity, for example via mutations in genes needed for proper brain development or by limiting brain-derived neurotrophic factors required for neurogenesis and neuronal plasticity [10,59,60,61]. In addition, dopamine is highly susceptible to auto-oxidation when antioxidant defence is weak [10,62]. Resulting dopamine cytotoxicity can cause brain damage in, e.g., the prefrontal cortex and basal ganglia, involved in attention and activity [10,63]. Oxidative stress and dopamine dysfunction thus appear interrelated in ADHD, possibly leading to a vicious cycle.
Various studies demonstrated, for example, decreased activity of antioxidant enzymes like CAT in ADHD, as well as increased levels of oxidative stress markers like plasma MDA [43,46,63,64,65]. In addition, increased levels of the oxidative enzymes xanthine oxidase (XO) and nitric oxide synthase (NOS) were observed, as well as decreased levels of the antioxidant enzymes glutathione-S-transferase (GST) and paraoxonase-1 (PON-1).

3. Immune Dysfunction in ADHD

Various types of research results point at the role of (allergic) immunopathology in ADHD, with indications of chronic neurological inflammation involving cellular immunity, probably requiring a predisposing genetic background [66,67,68].

3.1. Hypersensitivity

The potential involvement of the immune system in ADHD has long been suspected due to the increased prevalence of allergic diseases including atopic dermatitis, asthma and rhinitis in patients with ADHD [69]. Allergic reactions could deregulate cholinergic and adrenergic activity in the CNS, leading to symptoms of ADHD. The increased prevalence of allergies in ADHD was not dependent on environmental or lifestyle factors [70,71]. In addition, children with atopic dermatitis displayed more ADHD related symptoms, such as attention problems and impulsivity [72].
Several hypotheses were put forward to explain the association between atopic eczema and ADHD, which could be the result of atopic eczema influencing the development of ADHD, or ADHD increasing the vulnerability to develop atopic eczema. Mutual risk factors connecting both diseases include genetic background and perinatal stress. The underlying mechanism is probably related to an increased secretion of IgE, increased eosinophilic activity and predominantly TH2 type cytokine secretion, leading to an overproduction of inflammatory cytokines that pass the BBB and activate neuro-immune mechanisms involved in neuronal circuits that relate to behavioral and emotional modulation. Next to elevated cytokine levels, increased stress levels and thereby raised glucocorticoids can interfere with the development of the brain and neurotransmitter systems and increase the risk of developing atopic eczema [67].

3.2. Antibodies

No association was found between the severity of IgE-mediated atopy and ADHD scores, based on history of anaphylaxis and skin-prick tests [73,74]. The relationship between certain foods and ADHD was also not related to IgE, which is normally involved in food allergies. Involvement of IgG was not found either [75]. ADHD might thus be a (non) allergic hypersensitivity disorder caused by an environmental trigger, based on a non-IgE dependent histamine release from mast cells and basophilic granulocytes, since the histamine H3 receptor is involved in hyperactivity and promotes dopamine release in the frontal cortex. Moreover, polymorphisms in the histamine N-methyl transferase (HNMT) gene, impairing histamine clearance, were found to affect the behavioral responses to food additives, which increase histamine levels [76].

3.3. Cytokines

ADHD patients are reported to have a cerebrospinal fluid cytokine profile intermediate to that of obsessive-compulsive disorder patients (characterized by a skewing to TH1 mediated cytokines) and schizophrenics (skewing to TH2 mediated cytokines), with somewhat elevated concentrations of TNF-α and reduced levels of IFN-γ [77]. However, serum levels of IFN-γ and IL-13 were significantly higher in ADHD patients compared to healthy controls. Levels of IL-2, IL-6, IL-16 and IL-10 were slightly increased in the ADHD group, while IL-1β showed a small decrease. These altered levels of all cytokines were normalized by medication, though no consistent dysfunction in the balance of pro- and anti-inflammatory cytokines was found [68]. Nevertheless, an association has been observed between ADHD and TH1 and TH2 cell-mediated disorders, such as ear infections and eczema [6]. In addition, the underlying mechanisms of the association between specific autoimmune disorders and ADHD could also be based on higher levels of pro-inflammatory cytokines, including TNF-α, IL-6 and IL-8, which are related to ulcerative colitis [70]. Moreover, STAT6 (signal transducer and activator of transcription, involved in the expression of cytokines) is linked to IL-4 signaling [78] and plays an important role in brain development, as STAT6-deficient mice have fewer dopamine transporters and increased locomotor activity [79].
Additionally, cortisol production was found to be reduced in ADHD, which is suggested to be a consequence of early programming of the HPA-axis [80]. Though contradicted by another study [81], reduced cortisol production might lead to stimulation of TH1 derived inflammation and a defective regulatory T cell (TReg) immunomodulation. This would lead to an elevation of pro-inflammatory cytokines and reduction of anti-inflammatory IL-10, which has a major role in the pathophysiology of brain white matter damage, as chronic elevation of TNF-α and IL-6 primes cells to undergo apoptosis or necrosis [6]. More specifically, in pro-inflammatory conditions, TH1 and TH17 cells and their associated cytokines (IFN-γ, TNF-α, IL-17) can infiltrate into the brain parenchyma through lesioned BBB and have neurotoxic potential by promoting glial activation. The resulting neuro-inflammation could result in cognitive and behavioral impairments [82].
Cellular immunity therefore appears involved as a pathophysiological mechanism in ADHD. Furthermore, a defective TReg regulation potentially results in TH2-driven inflammation, making the child “allergy-prone” (thus more likely to have asthma, allergic rhinitis, atopic dermatitis and allergic conjunctivitis) by inducing activated B-cells, mast cells and eosinophils [83].

3.4. Prenatal Infections

During development, cytokines are involved in neuronal proliferation, differentiation, migration and synaptogenesis [84]. Accordingly, elevated levels of fetal and/or placental pro-inflammatory cytokines like IL-6, IL-1β and TNF-α in response to maternal immune activation can cause a CNS inflammatory response in the fetus and promote oxidative stress and apoptosis. Dependent on timing, the increase in pro-inflammatory cytokine levels can thus produce pathological changes in brain morphology akin to those observed in autism [84,85,86]. Additionally, maternal immune activation can affect neurodevelopment indirectly: an increase in placental IL-6 leads to a reduction of growth hormone and insulin-like growth factor, while simultaneously activating the Janus kinase (JAK)-signal transducer and activator of transcription 3 (STAT3) pathway, resulting in the expression of acute phase genes. These placental changes in gene expression mediated by maternal IL-6 can have downstream effects on fetal (neuro)development [87]. There is indeed evidence suggesting neuroanatomical abnormalities in the main dopaminergic, GABAergic and glutamatergic systems after prenatal infection, which could contribute to the development of ADHD [88].

3.5. Microbiome

Commensal microbiota play a crucial role in the development of intestinal immunity, e.g., via the induction of TRegs [6,89]. Converging evidence suggests that the gut microbiome is altered in ADHD. For instance, in ADHD patients versus controls, increases in the genus Bifidobacterium were found, which increased metabolic function of cyclohexadienyl dehydratase. This enzyme is involved phenylalanine synthesis, an essential amino acid and dopamine precursor. These metabolic changes were related to decreased ventral striatal signals to reward anticipation, a neural hallmark of ADHD [90,91,92].

4. Nutrition and ADHD

4.1. Dietary Micronutrients

If oxidative stress and immunity are involved in ADHD, also dietary micronutrients might be of influence on ADHD [50].
Children with iron deficiencies showed ADHD-related behavior, though serum iron levels were overall not different between ADHD and control subjects [5,93,94,95]. Iron has an important role in the structure and function of the CNS. A potential relation with ADHD could be due to its role as co-factor for tyrosine hydroxylase, the rate-limiting enzyme in dopamine synthesis. In addition, iron has neuroprotective effects against lead exposure [17]. However, iron supplementation has not been proven effective in treating ADHD in general, possibly because iron also catalyzes reactions that increase oxidative stress. Ferritin, an intracellular protein, stores iron and regulates its release, thereby limiting the amount of free iron that can produce oxidative radicals [50,96]. In ADHD patients, lower serum ferritin levels were however found compared to controls. This implies a lower regulation of iron, resulting in more free iron and thereby potentially creating more oxidative stress [93,94].
Another trace mineral related to ADHD is magnesium. In 33.6% of the ADHD children a magnesium deficiency in serum was present compared to 15.8% in healthy children. In hair, a 77.6% deficiency rate was found in ADHD children versus 20.7% in healthy children [97]. This is supported by several other studies [98,99,100] and contradicted by one [101]. Magnesium is needed for brain energy metabolism, synaptic signal transduction and general blood flow, but also for magnesium dependent enzyme systems that protect against oxidative DNA damage, such as 8-oxoG endonuclease [100,102].
Lower plasma zinc levels were found in ADHD patients [17,103,104]. Zinc functions as a co-factor in antioxidant defense systems, such as in the enzyme copper-zinc-SOD [50,105,106]. It is also needed for dopamine transport, structure and function of the brain and membranes, and the formation and modulation of melatonin that regulates dopamine metabolism [17,103]. Due to these functions, zinc is of influence on multiple factors involved in ADHD [15,107]. Zinc deficiency causes the brain to be more vulnerable to oxidative stress, also due to a reduced immune defense system and disruption of the BBB, and could therefore be associated with behavioral problems [5,61,108]. Still, trials on zinc supplementation in ADHD have provided conflicting results [109,110,111].
So far, no differences in plasma selenium levels have been found in ADHD patients [61,103]. Still, selenium supports the functional activity of the most important intracellular antioxidant GSH, e.g., in neurons to protect the brain from oxidative stress [61]. GSH-Px, of which selenium is a cofactor, catalyzes reduction of hydrogen peroxide by GSH [103].
Copper concentrations have been found higher in plasma of ADHD patients than in controls. This metal is involved in the metabolism and degradation of catecholamines via dopamine β-hydroxylase and monoamine oxidase [103], with higher copper concentrations leading to reduced dopamine levels in rats [105]. Also the GSH system is affected by copper as well as by mercury. Both micronutrients reduce available GSH present in the cell, which lowers the capacity to reduce hydrogen peroxide particles [61]. Furthermore, copper and mercury produce free radicals, but on the other hand, copper is part of the antioxidant enzyme SOD [103].
Lead is involved in free radical production and affects neurotransmitter generation [50,103]. Higher blood lead levels were found in ADHD patients compared to controls, associated with the severity of hyperactivity symptoms [112]. Hair lead levels were associated with teacher ratings on attention-deficit behavior and physician-diagnosed ADHD [113], although others did not find different lead concentrations in ADHD subjects [103].
There is increasing awareness of the relevance of vitamin D in immune regulation and circulating vitamin D levels were significantly lower in ADHD patients compared to controls [114,115]. This fat-soluble vitamin is involved in bone health and muscle strength and has a beneficial effect on intestinal absorption of minerals, such as calcium and phosphorus, though neither of these minerals have not been linked to ADHD [116]. Lower levels of vitamin D in ADHD patients might result from decreased sunlight exposure, as the prevalence of ADHD is lower in areas with a higher solar intensity [115,117].

4.2. Food, Food Constituents and Food Additives

Concentrations of micronutrients, including zinc, iron and copper, can be altered by food additives, and in extreme cases artificial food colorings result in a zinc deficiency [12,61]. As these minerals are cofactors of antioxidant enzymes, more oxidative stress could be present due to the intake of food additives [12]. In fact, food additives like sodium benzoate preservatives promoted hyperactivity in children [17,118,119]. In addition, the intake of specific foods was shown to cause behavioral problems in specific patients [120].
Therefore, studies have been performed with elimination diets, in which participants restrict the intake of certain foods such as dairy products, corn, soy and chocolate. Beneficial effects were found in children with ADHD when they were restricted from certain foods and food colorings. Atopic subjects responded more often, which points towards allergic hypersensitivity [75,121]. Moreover, maternal reports suggested an increased risk in individuals with food allergy to have symptoms of depression, anxiety or ADHD. In self-reports this increased risk was not significant [122].
Currently, the strongest evidence for an effect of nutrition on ADHD is related to omega-3 fatty acids, like docosahexaenoic acid (DHA) and eicosapentaenoic acid (EPA) [17,123]. DHA is needed as a structural component for neuronal membranes and for the myelination of nerve cells, necessary for neural transmission [124], and is associated with dopamine function [125]. Besides, DHA is a precursor for EPA, which is a precursor for eicosanoids with anti-inflammatory, anti-thrombotic and vasodilation properties [17]. EPA also regulates the uptake of neurotransmitters as well as neuronal signaling by acting as a second messenger [61,124]. Omega-3 fatty acids exert anti-inflammatory effects by inducing apoptosis in TH1 cells, promoting TH2 effector cell differentiation and enhancing TReg development [126,127], by which they possibly improve symptoms related to ADHD and atopic diseases.
In ADHD patients, lower levels of omega-3 fatty acids were found as well as a higher omega-6/omega-3 (AA/EPA) ratio, and supplementation had a beneficial effect on ADHD symptomatology in various studies [7,17,125,128], corroborated in a meta-analysis [129]. Another meta-regression analysis concluded that PUFA supplementation might have beneficial effects in decreasing inattention, but not hyperactive-impulsive symptoms. Still, meta-analyses support the use of EPA, γ-linolenic acid (GLA) and DHA in the management of ADHD symptoms [128] and for their ability to suppress allergic reactions [130], though a randomized placebo-controlled clinical trial did find higher EPA and DHA levels after twelve weeks of supplementation, but no beneficial effect on the Conner’s Teacher Rating Scales of aggression, impulsivity, depression and anxiety in ADHD was found [131]. There have however been inconsistencies in sample sizes, selection criteria, type and dosage of PUFA supplementation, follow-up times, etc. [123,132], while the duration and composition of PUFA supplementation have a profound influence on their effect [128].
Also amino acids are apparently related to ADHD development. In ADHD patients a methionine deficiency had a negative impact on behavior and learning capabilities. Methionine is an essential amino acid required for, among others, GSH production. Since GSH is part of the antioxidant defense, methionine deficiency may create even more oxidative stress [61].

5. Correction by Dietary Supplementation

Various data thus indicate the involvement of oxidative changes in ADHD and also cellular immunity is increasingly recognized as a pathophysiological mechanism behind the disorder. There are several ways by which oxidative stress and immune mechanisms could alter neuronal development and/or injure the developing structures, including altered gene expression, direct DNA damage and promotion of cell death, eventually leading to neuronal damage and loss of function. Still, specific immune biomarkers other than antibodies have not been systematically studied in ADHD. In addition, immune and oxidative effects of both standard therapy and nutritional supplementation in ADHD are neglected topics in research.
Yet, immune and oxidative imbalances linked with ADHD offer potential for nutritional supplementation in ADHD therapy. For example, TH1 and TH2 cytokine balancing and TReg stimulation is a nutritional target for a number of health conditions, with nutritional supplements including plant sterols, polyphenols, pre- and probiotics, vitamins, minerals and omega-3 fatty acids.
Polyphenols, composed of several phenol groups and found abundantly in, e.g., fruits, vegetables, green tea and red wine, are subdivided into different groups, including flavonoids, lignans, stilbenes, hydroxybenzoic acids and hydroxycinnamic acids, and have both antioxidant and immune modulating effects [133,134]. In addition, polyphenols can exert other biological effects, like influencing neuronal survival and regeneration [135]. Natural food constituents, like quercetin, epigallocatechin gallate and cyanidin-3-glucoside, which possess neuroprotective properties, could constitute a promising approach once they reach the brain [136,137].

5.1. Antioxidant Effects

The most well-known characteristic of polyphenols is their antioxidant capacity [138]. Polyphenols are able to scavenge free radicals by their aromatic rings with one or more hydroxyl groups [139], thereby forming resonance-stabilized phenoxyl radicals [140]. However, due to an absorption rate of 5–10% in the gastro-intestinal tract, a direct effect on antioxidant defense might be small [141]. Still, their effects can go beyond their antioxidant capacities and polyphenols contribute to the prevention of cardiovascular diseases, cancers, neurodegenerative diseases and diabetes mellitus [138]. In addition, low doses of phytochemicals like polyphenols can activate signaling pathways leading to increased expression of cytoprotective genes. One example is induction of phase II xenobiotic metabolizing enzymes like GST by antioxidants, e.g., via the transcription factor nuclear factor erythroid 2 (NFE2)-related factor 2 (Nrf2) [142]. This further increases antioxidant capacity and provides more long-lived protective effects as opposed to direct antioxidant effects [143,144].

5.2. Immune Modulating Effects

Epigallocatechin gallate (EGCG), curcumin and Pycnogenol® are examples of effective immunoregulatory and anti-inflammatory polyphenols or polyphenol rich extracts modulating the immune system by influencing both activation and differentiation of multiple immune cell types including TReg cells, TH1 cells, natural killer (NK) cells, dendritic cells (DCs) and macrophages [133,137,145].
Mechanisms associated with immune regulation include epigenetic modifications like DNA methylation of regulatory sequences. Polyphenols are able to regulate such epigenetic factors by oxidant and thiol-redox-mediated signaling modulation [146]. In addition, in vivo and in vitro studies have demonstrated that one of the main effects of polyphenols on macrophages is the inhibition of key regulators of the inflammatory response, including cyclooxygenase-2 (COX-2), TNF-α, IL-1β and IL-6 [147,148,149]. In vitro experiments using Jurkat T cells treated with EGCG found increased Foxp3 and IL-10 gene expression, required for TReg functioning. In vitro assays using naïve T cells from C57BL/6 mice under TH17 differentiation conditions and Baicalin treatment showed a decreased expression of IL-17 [150]. Treatment of U266 cells with green tea showed a dose- and time-dependent decrease of IgE production [151], demonstrating that polyphenols are also relevant in the modulation of B cell function [152]. These findings indicate that, independent of treatment or the cellular model used, the modulation of proinflammatory cytokine production appears a common factor in the immunomodulatory effects of polyphenols. Finally, polyphenols and their metabolites can affect host microbial composition, e.g., by inhibition of Clostridium or stimulation of Bifidobacterium, Lactobacillus, and Eubacterium species [153,154,155].

5.3. Pycnogenol

The herbal extract Pycnogenol®, derived from the bark of the French maritime pine (Pinus pinaster Aiton) [140], is composed of phenolic compounds, such as the monomers catechin and epicatechin, as well as oligomeric and polymeric procyanidins [16,156]. An example of a procyanidin present in Pycnogenol® is procyanidin B1 (epicatechin-(4β→8)-catechin) [157]. This extract is standardized to contain 70 ± 5% (w/w) procyanidins [139,158].
Studies indicate high bioavailability of the individual components of Pycnogenol®. For example, 46% of catechin metabolites (glucuronides and sulphates) were recovered in urine [139]. Oral Pycnogenol® ingestion was detectable in plasma after a single dose of 300 mg and multiple doses of 200 mg although detection times in plasma differ per compound. Metabolite M1 (δ-(3,4-dihydroxy-phenyl)-γ-valerolactone) is formed by bacterial metabolism from catechin [157,159]. This indicates that the components of Pycnogenol® can be modified during digestion and absorption as well as by the liver [139], which can both increase and decrease the actual effect in vivo. Therefore, in vitro results need to be interpreted with caution.
Pycnogenol® has multiple pharmacological effects such as antihypertensive, anti-inflammatory and anti-diabetic effects [158]. Due to its antioxidant effect, it reduces oxidative stress and might be beneficial in ADHD [140]. Moreover, it was found to reduce histamine release from rat peritoneal mast cells [152]. Additionally, M1 is able to cross the BBB and other cell membranes, probably mediated by the GLUT-1 transporter [16,157]. This facilitated uptake also causes accumulation of M1, which is metabolized to glutathione adducts, in erythrocytes [157]. Most other external antioxidants are not effective in reducing oxidative stress in the brain, because they lack the ability to cross the BBB [62,160].
In children, Pycnogenol® administration caused positive effects on ADHD symptoms compared to placebo. Statistically significant enhanced concentration and reduced hyperactivity, as rated by the Child Attention Problems teacher rating scale, were found after 1 month of Pycnogenol® supplementation as well as improved visual-motoric coordination as rated by psychologists [16,140]. This effect could be related to a reduction of elevated catecholamine levels, as dopamine levels decreased in the urine of ADHD patients using Pycnogenol® and a trend of decreased epinephrine and norepinephrine levels was seen [5,14,16]. This could be linked to the stimulation of the enzyme endothelial nitric oxide synthase (eNOS), involved in the regulation of the release and uptake of norepinephrine and dopamine [16]. NO increase can also improve blood circulation in cerebral areas, which is impaired in ADHD [17]. However, on the contrary, inflammation is reduced by a suppression of iNOS [161]. In addition, Pycnogenol® increased the activity and expression of SOD, increased GSH levels and decreased GSSG levels, pointing towards less oxidative stress [162]. Also, an increase in GSH reductase was found, which could explain the higher ratio of GSH/GSSG [140]. TAS levels of children with ADHD were increased to a normal level by Pycnogenol® administration [140,162], while increased damage to DNA was lowered. It is therefore tempting to suggest that Pycnogenol® can be beneficial in ADHD because of its direct scavenging ability, chelating activity, stimulation of the DNA repair system and/or combinations thereof [140], but also due to its immune regulatory effects. However, this study had various limitations, including a short supplementation period and small placebo group [16]. In addition, Pycnogenol® standardization is limited to total procyanidin content without determination of major polyphenolic constituents and the actual bioavailability of this complex mixture is still unknown [139]. Though Pycnogenol® appears promising for the treatment of ADHD, further research is therefore required.

5.4. Other Polyphenolic Extracts

Other polyphenolic extracts with antioxidant and immune modulating properties have been investigated in the treatment of ADHD, though most studies were relatively small or conducted over a short time span [163,164,165,166,167,168].
One example is Ginkgo biloba extract, found to improve cerebrovascular blood flow and to affect several central neurotransmitter systems. In a double blind randomized study including fifty ADHD patients, 80–120 mg/day Ginkgo biloba extract (made from the leaves of the Ginkgo tree) for six weeks was much less effective than MPH treatment. Still, Ginkgo caused fewer adverse events than MPH [169]. A three to five-week supplementation with a higher dose (max. 240 mg) in an open study improved ADHD symptoms as well as brain electrical activity [170].
Hypericum perforatum extract (St. John’s wort, 900 mg/day for 8 weeks), containing flavonoids like quercitrin, rutin and quercetin, did not significantly improve ADHD symptoms as compared to placebo in a randomized double-blind trial with 54 patients [171,172]. In another randomized controlled trial with 34 ADHD patients, Passiflora incarnata (0.4 mg/kg/day pass twice daily), containing the flavonoids luteolin, quercetin and rutin, among other constituents, and MPH (1 mg/kg/day twice daily) performed equally well regarding parent and teacher rating scale scores, with both demonstrating a significant clinical benefit over 8 weeks [173].

6. Conclusions

Elevated oxidative stress and immune dysfunction, eventually leading to neuronal damage, appear to play a role in the pathophysiological process of neurodevelopmental disorders. Evidence on the association of ADHD with immune and oxidant-antioxidant imbalances offers potential for antioxidant and/or immunomodulatory nutritional supplements (e.g., polyphenols) as ADHD therapy. One example is Pycnogenol®, a herbal, polyphenol-rich extract with potent antioxidant and anti-inflammatory properties, which is considered to have therapeutic benefits in ADHD, as it increased antioxidant levels, reduced oxidative damage and improved neurochemical status.

Acknowledgments

A.A.J.V. received a PhD scholarship from the Fund for Scientific Research Flanders (Fonds Wetenschappelijk Onderzoek (FWO); FWO MAND 2013-11U8316N 5 W).

Author Contributions

A.A.J.V., C.M.M., N.H. and H.F.J.S. wrote the paper. A.A.J.V. and H.F.J.S. finalized the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Polanczyk, G.; de Lima, M.S.; Horta, B.L.; Biederman, J.; Rohde, L.A. The Worldwide Prevalence of ADHD: A Systematic Review and Metaregression Analysis. Am. J. Psychiatry 2007, 164, 942–948. [Google Scholar] [CrossRef] [PubMed]
  2. Moffitt, T.E.; Melchior, M. Why does the worldwide prevalence of childhood attention deficit hyperactivity disorder matter? Am. J. Psychiatry 2007, 164, 856–858. [Google Scholar] [CrossRef] [PubMed]
  3. APA American Psychiatric Association: DSM-5 Attention Deficit/Hyperactivity Disorder Fact Sheet. Available online: http://www.dsm5.org/Documents/ADHD%20Fact%20Sheet.pdf (accessed on 22 November 2017).
  4. Milich, R.; Ballentine, A.C.; Lynam, D. ADHD combined type and ADHD predominantly inattentive type are unrelated disroders. Clin. Psychol. Sci. Pract. 2001, 8, 463–488. [Google Scholar] [CrossRef]
  5. Faraone, S.V.; Bonvicini, C.; Scassellati, C. Biomarkers in the diagnosis of ADHD-promising directions. Curr. Psychiatry Rep. 2014, 16, 1–20. [Google Scholar] [CrossRef] [PubMed]
  6. Verlaet, A.A.J.; Noriega, D.B.; Hermans, N.; Salvelkoul, H.F.J. Nutrition, immunological mechanisms and dietary immunomodulation in ADHD. Eur. Child Adolesc. Psychiatry 2014, 23, 519–529. [Google Scholar] [CrossRef] [PubMed]
  7. Guney, E.; Cetin, F.H.; Alisik, M.; Tunca, H.; Torun, Y.T.; Iseri, E.; Taner, Y.I.; Cayci, B.; Erel, O. Attention Deficit Hyperactivity Disorder and oxidative stress: A short term follow up study. Psychiatry Res. 2015, 229, 310–317. [Google Scholar] [CrossRef] [PubMed]
  8. Shaw, P.; Gilliam, M.; Liverpool, M.; Weddle, C.; Malek, M.; Sharp, W.; Greenstein, D.; Evans, A.; Rapoport, J.; Giedd, J. Cortical development in typically developing children with symptoms of hyperactivity and impulsivity: Support for a dimensional view of attention deficit hyperactivity disorder. Am. J. Psychiatry 2011, 168, 143–151. [Google Scholar] [CrossRef] [PubMed]
  9. Bulut, M.; Selek, S.; Gergerlioglu, H.S.; Savas, H.A.; Yilmaz, H.R.; Yuce, M.; Ekici, G. Malondialdehyde levels in adult attention-deficit hyperactivity disorder. J. Psychiatry Neurosci. 2007, 32, 435–438. [Google Scholar] [PubMed]
  10. Ceylan, M.; Sener, S.; Bayraktar, A.; Kavutcu, M. Changes in oxidative stress and cellular immunity serum markers in attention-deficit/hyperactivity disorder. Psychiatry Clin. Neurosci. 2012, 66, 220–226. [Google Scholar] [CrossRef] [PubMed]
  11. Oztop, D.; Altun, H.; Baskol, G.; Ozsoy, S. Oxidative stress in children with attention deficit hyperactivity disorder. Clin. Biochem. 2012, 45, 745–748. [Google Scholar] [CrossRef] [PubMed]
  12. Lopresti, A.L. Oxidative and nitrosative stress in ADHD: Possible causes and the potential of antioxidant-targeted therapies. ADHD 2015, 7, 237–247. [Google Scholar] [CrossRef] [PubMed]
  13. Konrad, K.; Gauggel, S.; Schurek, J. Catecholamine functioning in children with traumatic brain injuries and children with attention-deficit/hyperactivity disorder. Cogn. Brain Res. 2003, 16, 425–433. [Google Scholar] [CrossRef]
  14. Dvořáková, M.; Ježová, D.; Blažíček, P.; Trebatická, J.; Skodácek, I.; Suba, J.; Iveta, W.; Rohdewald, P.; Duracková, Z. Urinary catecholamines in children with attention deficit hyperactivity disorder (ADHD): Modulation by a polyphenolic extract from pine bark (Pycnogenol). Nutr. Neurosci. 2007, 10, 151–157. [Google Scholar] [CrossRef] [PubMed]
  15. Scasselati, C.; Bonvicini, C.; Faraone, S.V.; Gennarelli, M. Biomarkers and Attention-Deficit/Hyperactivity Disorder: A Systematic Review and Meta-analyses. J. Am. Acad. Child Adolesc. Psychiatry 2012, 51, 1003–1019. [Google Scholar] [CrossRef] [PubMed]
  16. Trebatická, J.; Kopasová, S.; Hradečná, Z.; Cinovský, K.; Skodácek, I.; Suba, J.; Muchová, J.; Zitnanová, I.; Waczulíková, I.; Rohdewald, P.; et al. Treatment of ADHD with French maritime pine bark extract, Pycnogenol. Eur. Child Adolesc. Psychiatry 2006, 15, 329–335. [Google Scholar] [CrossRef] [PubMed]
  17. Sinn, N. Nutritional and dietary influences on attention deficit hyperactivity disorder. Nutr. Rev. 2008, 66, 558–568. [Google Scholar] [CrossRef] [PubMed]
  18. Zayats, T.; Athanasiu, L.; Sonderby, I.; Djurovic, S.; Westlye, L.T.; Tamnes, C.K. Genome-wide analysis of attention deficit hyperactivity disorder in Norway. PLoS ONE 2015, 10, e0122501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Instanes, J.T.; Halmøy, A.; Engeland, A.; Haavik, J.; Furu, K.; Klungsøyr, K. Attention-Deficit/Hyperactivity Disorder in Offspring of Mothers with Inflammatory and Immune System Diseases. Biol. Psychiatry 2017, 81, 452–459. [Google Scholar] [CrossRef] [PubMed]
  20. Fagundes, A.O.; Rezin, G.T.; Zanette, F.; Grandi, E.; Assis, L.C.; Dal-Pizzol, F.; Quevedo, J.; Streck, E.L. Chronic administration of methylphenidate activates mitochondrial respiratory chain in brain of young rats. Int. J. Dev. Neurosci. 2007, 25, 47–51. [Google Scholar] [CrossRef] [PubMed]
  21. Pliszka, S.R. The neuropsychopharmacology of attention-deficit/hyperactivity disorder. Biol. Psychiatry 2005, 57, 1385–1390. [Google Scholar] [CrossRef] [PubMed]
  22. Liew, Z.; Ritz, B.; Rebordosa, C.; Lee, P.C.; Olsen, J. Acetaminophen use during pregnancy, behavioral problems, and hyperkinetic disorders. JAMA Pediatr. 2014, 168, 313–320. [Google Scholar] [CrossRef] [PubMed]
  23. Blais, L.; Kettani, F.Z.; Forget, A.; Beauchesne, M.F.; Lemiere, C. Asthma exacerbations during the first trimester of pregnancy and congenital malformations: Revisiting the association in a large representative cohort. Thorax 2015, 70, 647–652. [Google Scholar] [CrossRef] [PubMed]
  24. Bauer, A.Z.; Kriebel, D.; Herbert, M.R.; Bornehag, C.G.; Swan, S.H. Prenatal paracetamol exposure and child neurodevelopment: A review. Horm. Behav. 2018. [Google Scholar] [CrossRef] [PubMed]
  25. Volz, T.J. Neuropharmacological mechanisms underlying the neuroprotective effects of methylphenidate. Curr. Neuropharmacol. 2008, 6, 379–385. [Google Scholar] [CrossRef] [PubMed]
  26. Heyer, D.B.; Meredith, R.M. Environmental toxicology: Sensitive periods of development and neurodevelopmental disorders. NeuroToxicol. 2017, 58, 23–41. [Google Scholar] [CrossRef] [PubMed]
  27. Schachter, H.M.; Pham, B.; King, J.; Langford, S.; Moher, D. How efficacious and safe is shortacting methylphenidate for the treatment of attention-deficit disorder in children and adolescents? A meta-analysis. CMAJ 2011, 165, 1475–1488. [Google Scholar]
  28. Storebø, O.J.; Krogh, H.B.; Ramstad, E.; Moreira-Maia, C.R.; Holmskov, M.; Skoog, M.; Nilausen, T.D.; Magnusson, F.L.; Zwi, M.; Gillies, D.; et al. Methylphenidate for attention-deficit/hyperactivity disorder in children and adolescents: Cochrane systematic review with meta-analyses and trial sequential analyses of randomised clinical trials. BMJ 2015, 351. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  29. Antshel, K.M.; Hargrave, T.M.; Simonescu, M.; Kaul, P.; Hendricks, K.; Faraone, S.V. Advances in understanding and treating ADHD. BMC Med. 2011, 9, 72. [Google Scholar] [CrossRef] [PubMed]
  30. Biederman, J.; Faraone, S.V. Attention-deficit hyperactivity disorder. Lancet 2005, 366, 237–248. [Google Scholar] [CrossRef]
  31. Shibib, S.; Chalhoub, N. Stimulant induced psychosis. Child Adolesc. Ment. Health 2009, 14, 20–23. [Google Scholar] [CrossRef]
  32. The British Psychological Society and the Royal College of Psychiatrist. NICE Attention Deficit Hyperactivity Disorder: Diagnosis and Management of ADHD in Children, Young People and Adults. National Clinical Practice Guideline; The British Psychological Society and The Royal College of Psychiatrist: London, UK, 2009. [Google Scholar]
  33. Auci, D.L.; Fikrig, S.; Rodriquez, J. Methylphenidate and the immune system. J. Am. Acad. Child Adolesc. Psychiatry 1997, 36, 1015–1016. [Google Scholar] [CrossRef] [PubMed]
  34. Steer, C.R. Managing attention deficit/hyperactivity disorder: Unmet needs and future directions. Arch Dis. Child 2005, 90, i19–i25. [Google Scholar] [CrossRef] [PubMed]
  35. Taylor, M.; O’Donoghue, T.; Houghton, S. To medicate or not to medicate? The decision-making process of Western Australian parents following their child’s diagnosis with an attention deficit hyperactivity disorder. Int. J. Disabil. Dev. Educ. 2006, 53, 111–128. [Google Scholar] [CrossRef]
  36. Kortekaas-Rijlaarsdam, A.F.; Luman, M.; Sonuga-Barke, E.; Oosterlaan, J. Does methylphenidate improve academic performance? A systematic review and meta-analysis. ECAP 2018. [Google Scholar] [CrossRef] [PubMed]
  37. Sarris, J.; Kean, J.; Schweitzera, I.; Lake, J. Complementary medicines (herbal and nutritional products) in the treatment of Attention Deficit Hyperactivity Disorder (ADHD): A systematic review of the evidence. Complement. Ther. Med. 2011, 19, 216–227. [Google Scholar] [CrossRef] [PubMed]
  38. Halliwell, B. Oxidative stress, nutrition and health. Experimental strategies for optimization of nutritional antioxidant intake in humans. Free Radic. Res. 1996, 25, 57–74. [Google Scholar] [CrossRef] [PubMed]
  39. Firuzi, O.; Miri, R.; Tavakkoli, M.; Saso, L. Antioxidant Therapy: Current Status and Future Prospects. Curr. Med. Chem. 2011, 18, 3871–3888. [Google Scholar] [CrossRef] [PubMed]
  40. Emiliani, F.E.; Sedlak, T.W.; Sawa, A. Oxidative stress and schizophrenia: Recent breakthroughs from an old story. Curr. Opin. Psychiatry 2014, 27, 185–190. [Google Scholar] [CrossRef] [PubMed]
  41. Halliwell, B.; Gutteridge, J.M.C. Free Radicals in Biology and Medicine; Press, C., Ed.; Oxford University Press: Oxford, UK, 1999. [Google Scholar]
  42. Willcox, J.K.; Ash, S.L.; Catignani, G.L. Antioxidants and prevention of chronic disease. Crit. Rev. Food Sci. 2004, 44, 275–295. [Google Scholar] [CrossRef] [PubMed]
  43. Ng, F.; Berk, M.; Dean, O.; Bush, A. Oxidative stress in psychiatric disorders: Evidence base and therapeutic implications. Int. J. Neuropsychopharmacol. 2008, 21, 1–26. [Google Scholar] [CrossRef] [PubMed]
  44. Davies, K.J.A. Oxidative stress, antioxidant defences, and damage removal, repair, and replacement systems. IUBMB Life 2000, 50, 279–289. [Google Scholar] [CrossRef] [PubMed]
  45. Chelikani, P.; Fita, I.; Loewen, P.C. Diversity of structures and properties among catalases. Cell Mol. Life Sci. 2004, 61, 192–208. [Google Scholar] [CrossRef] [PubMed]
  46. Ceylan, M.; Senerb, S.; Bayraktarc, A.C.; Kavutcuc, M. Oxidative imbalance in child and adolescent patients with attention-deficit/hyperactivity disorder. Prog. Neuropsychopharmacol. Biol. Psychiatry 2010, 34, 1491–1494. [Google Scholar] [CrossRef] [PubMed]
  47. Hermans, N.; Cos, P.; Maes, L.; De Bruyne, T.; Vanden Berghe, D.; Vlietinck, A.J.; Pieters, L. Challenges and pitfalls in antioxidant research. Curr. Med. Chem. 2007, 14, 417–430. [Google Scholar] [CrossRef] [PubMed]
  48. Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S.; Kalayci, O. Oxidative stress and antioxidant defense. World Allergy Organ. J. 2012, 5, 9–19. [Google Scholar] [CrossRef] [PubMed]
  49. Dröge, W. Free Radicals in the Physiological Control of Cell Function. Physiol. Rev. 2002, 82, 47–95. [Google Scholar] [CrossRef] [PubMed]
  50. Joseph, N.; Zhang-James, Y.; Perl, A.; Faraone, S.V. Oxidative Stress and ADHD: A Meta-Analysis. J. Atten. Disord. 2015, 19, 915–924. [Google Scholar] [CrossRef] [PubMed]
  51. Farmamozaïek, I. De Peroxidatie van Lipiden, Celmembraandestructie en Atherosclerose. Available online: http://www.farmamozaiek.be/farmamozaiek/?q=node/38355 (accessed on 15 November 2017).
  52. Grotto, D.; Maria, L.S.; Valentini, J.; Paniz, C.; Schmitt, G.; Garcia, S.C.; Farina, M. Importance of the lipid peroxidation biomarkers and methodological aspects for malondialdehyde quantification. Quim. Nova 2009, 32, 169–174. [Google Scholar] [CrossRef]
  53. Bulut, M.; Selek, S.; Bez, Y.; Cemal Kaya, M.; Gunes, M.; Karababa, F.; Celik, H.; Savas, H.A. Lipid peroxidation markers in adult attention deficit hyperactivity disorder: New findings for oxidative stress. Psychiatry Res. 2013, 209, 638–642. [Google Scholar] [CrossRef] [PubMed]
  54. Hensley, K.; Williamson, K.S.; Floyd, R.A. Measurement of 3-nitrotyrosine and 5-nitro-gamma-tocopherol by high-performance liquid chromatography with electrochemical detection. Free Radic. Biol. Med. 2000, 28, 520–528. [Google Scholar] [CrossRef]
  55. Dalle-Donne, I.; Rossi, R.; Giustarini, D.; Milzani, A.; Colombo, R. Protein carbonylation in human diseases. Trends Mol. Med. 2003, 9, 169–176. [Google Scholar] [CrossRef]
  56. Dalle-Donne, I.; Rossi, R.; Colombo, R.; Giustarini, D.; Milzani, A. Biomarkers of oxidative damage in human disease. Clin. Chem. 2006, 52, 601–623. [Google Scholar] [CrossRef] [PubMed]
  57. Wu, L.L.; Chioiu, C.; Chang, P.; Wu, J.T. Urinary 8-OHdG: A marker of oxidative stress to DNA and a risk factor for cancer, atherosclerosis and diabetics. Clin. Chim. Acta 2004, 339, 1–9. [Google Scholar] [CrossRef] [PubMed]
  58. Chiou, C.-C.; Chang, P.-Y.; Chan, E.-C.; Wu, T.-L.; Tsao, K.-C.; Wu, J.T. Urinary 8-hydroxydeoxyguanosine and its analogs as DNA marker of oxidative stress: Development of an ELISA and measurement in both bladder and prostate cancers. Clin. Chim. 2003, 334, 87–94. [Google Scholar] [CrossRef]
  59. Kul, M.; Unal, F.; Kandemir, H.; Sarkarati, B.; Kilinc, K.; Kandemir, S.B. Evaluation of oxidative metabolism in child and adolescent patients with attention deficit hyperactivity disorder. Psychol. Invest. 2015, 12, 361–366. [Google Scholar] [CrossRef] [PubMed]
  60. Wu, A.; Ying, Z.; Gomez-Pinilla, F. Dietary curcumin counteracts the outcome of traumatic brain injury on oxidative stress, synaptic plasticity, and cognition. Exp. Neurol. 2006, 197, 309–317. [Google Scholar] [CrossRef] [PubMed]
  61. Dufault, R.; Schnoll, R.; Lukiw, W.; LeBlanc, B.; Cornett, C.; Patrick, L.; Wallinga, D.; Gilbert, S.; Crider, R. Mercury exposure, nutritional deficiencies and metabolic disruptions may affect learning in children. Behav. Brain Funct. 2009, 5, 44–59. [Google Scholar] [CrossRef] [PubMed]
  62. Iuga, C.; Alvarez-Idaboy, J.R.; Vivier-bunge, A. ROS initiated oxidation of dopamine under oxidative stress conditions in aqueous and lipid environments. J. Phys. Chem. 2011, 115, 12234–12246. [Google Scholar] [CrossRef] [PubMed]
  63. El Adham, E.K.; Hassan, A.I.; El Aziz El-Mahdy, A.A. Nutritional and Metabolic Disturbances in Attention Deficit Hyperactivity Disease. Res. J. Med. Med. Sci. 2011, 6, 10–16. [Google Scholar]
  64. Ross, B.M.; McKenzie, I.; Glen, I.; Bennett, C.P. Increased levels of ethane, a non-invasive marker of n-3 fatty acid oxidation, in breath of children with attention deficit hyperactivity disorder. Nutr. Neurosci. 2003, 6, 277–281. [Google Scholar] [CrossRef] [PubMed]
  65. Kawatani, M.; Tsukahara, H.; Mayumi, M. Evaluation of oxidative stress status in children with pervasive developmental disorder and attention deficit hyperactivity disorder using urinary-specific biomarkers. Redox Rep. 2011, 16, 45–46. [Google Scholar] [CrossRef] [PubMed]
  66. Martino, M.; Rocchi, G.; Escelsior, A.; Fornaro, M. Immunomodulation mechanism of antidepressants: Interactions between serotonin/norepinephrine balance and Th1/Th2 balance. Curr. Neuropharmacol. 2012, 10, 97–123. [Google Scholar] [CrossRef] [PubMed]
  67. Buske-Kirschbaum, A.; Schmitt, J.; Plessowa, F.; Romanos, M.; Weidinger, S.; Roessner, V. Psychoendocrine and psychoneuroimmunological mechanisms in the comorbidity of atopic eczema and attention deficit/hyperactivity disorder. Psychoneuroendocrinol 2013, 38, 12–23. [Google Scholar] [CrossRef] [PubMed]
  68. Oades, R.; Dauvermann, M.; Schimmelmann, B.; Schwarz, M.; Myint, A.-M. Attention-deficit hyperactivity disorder (ADHD) and glial integrity: S100B, cytokines and kynurenine metabolism-effects of medication. Behav. Brain Funct. 2010, 6, 2–14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Pelsser, L.M.J.; Frankena, K.; Toorman, J.; Savelkoul, H.F.J.; Pereira, R.R.; Buitelaar, J.K. A randomised controlled trial into the effects of food on ADHD. Eur. Child Adolesc. Psychiatry 2009, 18, 12–19. [Google Scholar] [CrossRef] [PubMed]
  70. Chen, M.; Su, T.; Chen, Y.; Hsu, J.; Huang, K.; Chang, W.; Bai, Y. Comorbidity of allergic and autoimmune diseases among patients with ADHD: A nationwide population-based study. J. Atten. Disord. 2013, 21, 219–227. [Google Scholar] [CrossRef] [PubMed]
  71. Chen, M.; Su, T.; Chen, Y.; Hsu, J.; Huang, K.; Chang, W.; Bai, Y. Attention deficit hyperactivity disorder, tic disorder, and allergy: Is there a link? A nationwide population-based study. J. Child Psychol. Psychiatry 2013, 54, 545–551. [Google Scholar] [CrossRef] [PubMed]
  72. Roth, N.; Beyreiss, J.; Schlenzka, K.; Beyer, H. Coincidence of Attention Deficit Disorder and Atopic Disorders in Children: Empirical Findings and Hypothetical Background. J. Abnorm. Child Psychol. 1991, 19, 1–13. [Google Scholar] [CrossRef] [PubMed]
  73. Gaitens, T.; Kaplan, B.J.; Freigang, B. Absence of an Association between IgE-mediated Atopic Responsiveness and ADHD Symptomatology. J. Child Psychol. Psychiatry 1998, 39, 427–431. [Google Scholar] [CrossRef] [PubMed]
  74. Stevenson, J.; Sonuga-Barke, E.; McCann, D.; Grimshaw, K.; Parker, K.M.; Rose-Zerilli, M.J.; Warner, J.O. The role of histamine degradation gene polymorphisms in moderating the effects of food additives on children’s ADHD symptoms. Am. J. Psychiatry 2010, 167, 1108–1115. [Google Scholar] [CrossRef] [PubMed]
  75. Pelsser, L.M.J.; Frankena, K.; Toorman, J.; Savelkoul, H.F.J.; Dubois, A.E.; Pereira, R.R.; Haagen, T.A.; Rommelse, N.N.; Buitelaar, J.K. Effects of a restricted elimination diet on the behaviour of children with attention-deficit hyperactivity disorder (INCA study): A randomised controlled trial. Lancet 2011, 377, 494–503. [Google Scholar] [CrossRef]
  76. Stevenson, J.; Buitelaar, J.; Cortese, S.; Ferrin, M.; Konofal, E.; Lecendreux, M.; Sonuga-Barke, E. Research review: The role of diet in the treatment of attention-deficit/hyperactivity disorder-an appraisal of the evidence on efficacy and recommendations on the design of future studies. J. Child Psychol. Psychiatry 2014, 55, 416–427. [Google Scholar] [CrossRef] [PubMed]
  77. Mittleman, B.B.; Castellanos, F.X.; Jacobsen, L.K.; Rapoport, J.L.; Swedo, S.E.; Shearer, G.M. Cerebrospinal Fluid Cytokines in Pediatric Neuropsychiatric Disease. J. Immunol. 1997, 159, 2994–2999. [Google Scholar] [PubMed]
  78. Abbas, A.K.; Lichtman, A.H.; Pillai, S. Cellular and Molecular Immunology; Elsevier Saunders: Philadelphia, PA, USA, 2015. [Google Scholar]
  79. Tsai, S.J. Signal transducer and activator of transcription 6 (STAT6) and attention-deficit hyperactivity disorder: A speculative hypothesis. Med. Hypotheses 2006, 67, 1341–1343. [Google Scholar] [CrossRef] [PubMed]
  80. Isaksson, J.; Nilsson, K.W.; Lindblad, F. Early psychosocial adversity and cortisol levels in children with attention-deficit/hyperactivity disorder. ECAP 2013, 22, 425–432. [Google Scholar] [CrossRef] [PubMed]
  81. Işık, U.; Bilgiç, A.; Toker, A.; Kılınç, I. Serum levels of cortisol, dehydroepiandrosterone, and oxytocin in children with attention-deficit/hyperactivity disorder combined presentation with and without comorbid conduct disorder. Psychol. Res. 2018, 261, 212–219. [Google Scholar] [CrossRef] [PubMed]
  82. Sonar, S.A.; Lal, G. Differentiation and Transmigration of CD4 T Cells in Neuroinflammation and Autoimmunity. Front. Immunol. 2017, 8, 1–9. [Google Scholar] [CrossRef] [PubMed]
  83. Miyazaki, C.; Koyama, M.; Ota, E.; Swa, T.; Mlunde, L.B.; Amiya, R.M.; Tachibana, Y.; Yamamoto-Hanada, K.; Mori, R. Allergic diseases in children with attention deficit hyperactivity disorder: A systematic review and meta-analysis. BMC Psychiatry 2017, 17, 120. [Google Scholar] [CrossRef] [PubMed]
  84. Goines, P.E.; Ashwood, P. Cytokine dysregulation in autism spectrum disorders (ASD): Possible role of the environment. Neurotoxicol. Teratol. 2013, 36, 67–81. [Google Scholar] [CrossRef] [PubMed]
  85. Amiri, A.; Turner-Henson, A. The Roles of Formaldehyde Exposure and Oxidative Stress in Fetal Growth in the Second Trimester. J. Obstet. Gynecol. Neonatal Nurs. 2017, 46, 51–62. [Google Scholar] [CrossRef] [PubMed]
  86. Fatemi, S.H.; Folsom, T.D.; Reutiman, T.J.; Huang, H.; Oishi, K.; Mori, S. Gene expression is altered in hippocampus following prenatal viral infection in mouse at E16. Eur. Neuropsychopharmacol. 2009, 19, 648–653. [Google Scholar] [CrossRef] [PubMed]
  87. Hsiao, E.Y.; Patterson, P.H. Activation of the maternal immune system induces endocrine changes in the placenta via IL-6. Brain Behav. Immun. 2011, 25, 604–615. [Google Scholar] [CrossRef] [PubMed]
  88. Baharnoori, M.; Brake, W.G.; Srivastava, L.K. Prenatal immune challenge induces developmental changes in the morphology of pyramidal neurons of the prefrontal cortex and hippocampus I rats. Schizophr. Res. 2009, 107, 99–109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Tillisch, K. The effects of gut microbiota on CNS function in humans. Gut Microbes 2014, 5, 404–410. [Google Scholar] [CrossRef] [PubMed]
  90. Mülek, M.; Fekete, A.; Wiest, J.; Holzgrabe, U.; Mueller, M.J.; Högger, P. Profiling a gut microbiota-generated catechin metabolite’s fate in human blood cells using a metabolomic approach. J. Pharm. Biomed. Anal. 2015, 114, 71–81. [Google Scholar] [CrossRef] [PubMed]
  91. Wang, Y.; Kasper, L.H. The role of microbiome in central nervous system disorders. Brain Behav. Immun. 2014, 38, 1–12. [Google Scholar] [CrossRef] [PubMed]
  92. Aarts, E.; Ederveen, T.H.A.; Naaijen, J.; Zwiers, M.P.; Boekhorst, J.; Timmerman, H.M.; Smeekens, S.P.; Netea, M.G.; Buitelaar, J.K.; Franke, B.; et al. Gut microbiome in ADHD and its relation to neural reward anticipation. PLoS ONE 2017, 12, e0183509. [Google Scholar] [CrossRef] [PubMed]
  93. Konofal, E.; Lecendreux, M.; Arnulf, I.; Mouren, M.C. Iron deficiency in children with attention-deficit/hyperactivity disorder. Arch. Pediatr. Adolesc. Med. 2004, 158, 1113–1115. [Google Scholar] [CrossRef] [PubMed]
  94. Wang, Y.; Huang, L.; Zhang, L.; Qu, Y.; Mu, D. Iron status in attention-deficit/hyperactivity disorder: A systematic review and meta-analysis. PLoS ONE 2017, 12, e0169145. [Google Scholar] [CrossRef] [PubMed]
  95. Tseng, P.T.; Cheng, Y.S.; Yen, C.F.; Chen, Y.W.; Stubbs, B.; Whiteley, P.; Carvalho, A.F.; Li, D.J.; Chen, T.Y.; Yang, W.C.; et al. Peripheral iron levels in children with attention-deficit hyperactivity disorder: A systematic review and meta-analysis. Sci. Rep. 2018, 8, 788–798. [Google Scholar] [CrossRef] [PubMed]
  96. Hariri, M.; Azadbakht, L. Magnesium, iron and zinc supplementation for the treatment of attention deficit hyperactivity disorder: A systematic review on the recent lieterature. Int. J. Prev. Med. 2015, 6. [Google Scholar] [CrossRef]
  97. Kozielec, T.; Starobrat-Hermelin, B. Assessment of magnesium levels in children with attention deficit hyperactivity disorder (ADHD). Magn. Res. 1997, 10, 143–148. [Google Scholar]
  98. Antalis, C.J.; Stevens, L.J.; Campbell, M.; Pazdro, R.; Ericson, K.; Burgess, J.R. Omega-3 fatty acid status in attention deficit/hyperactivity disorder. Prostagland Leukot. Essent. Fat. Acids 2006, 75, 299–308. [Google Scholar] [CrossRef] [PubMed]
  99. Mahmoud, M.M.; El-Mazary, A.A.; Maher, R.M.; Saber, M.M. Zinc, ferritin, magnesium and copper in a group of Egyptian children with attention deficit hyperactivity disorder. Ital. J. Pëdiatr. 2011, 37. [Google Scholar] [CrossRef] [PubMed]
  100. Archana, E.; Pai, P.; Prabhu, B.K.; Shenoy, R.P.; Prabhu, K.; Rao, A. Altered Biochemical parameters in saliva of pediatric attention deficit hyperactivity disorder. Neurochem. Res. 2012, 37, 330–334. [Google Scholar] [CrossRef] [PubMed]
  101. Irmisch, G.; Thome, J.; Reis, O.; Hässler, F.; Weirich, S. Modified magnesium and lipoproteins in children with attention deficit hyperactivity disorder (ADHD). World J. Biol. Psychiatry 2011, 12, S63–S65. [Google Scholar] [CrossRef] [PubMed]
  102. Croteau, D.L.; Bohr, V.A. Repair of oxidative damage to nuclear and mitochondrial DNA in mammalian cells. J. Biol. Chem. 1997, 272, 25409–25412. [Google Scholar] [CrossRef] [PubMed]
  103. Viktorinova, A.; Ursinyova, M.; Trebaticka, J.; Uhnakova, I.; Durackova, Z.; Masanova, V. Changed plasma levels of zinc and copper to zinc ratio and their possible associations with parent- and teacher-rated symptoms in children with attention-deficit hyperactivity disorder. Biol. Trace Elem. Res. 2016, 169, 1–7. [Google Scholar] [CrossRef] [PubMed]
  104. Zhou, F.; Wu, F.; Zou, S.; Chen, Y.; Feng, C.; Fan, G. Dietary, nutrient patterns and blood essential elements in Chinese children with ADHD. Nutrients 2016, 8, 352. [Google Scholar] [CrossRef] [PubMed]
  105. Yu, W.; Jiang, H.; Wang, J.; Xie, J. Copper (Cu2+) induces degeneration of dopaminergic neurons in the nigrostriatal system of rats. Neurosci. Bull. 2008, 24, 73–78. [Google Scholar] [CrossRef] [PubMed]
  106. Jomova, K.; Valko, M. Advances in metal-induced oxidative stress and human disease. Toxicol 2011, 283, 65–87. [Google Scholar] [CrossRef] [PubMed]
  107. Arnold, L.E.; DiSilvestro, R.A. Zinc in attention-deficit/hyperactivity disorder. J. Child Adolesc. Psychopharmacol. 2005, 15, 619–627. [Google Scholar] [CrossRef] [PubMed]
  108. Rosenkranz, E.; Hilgers, R.D.; Uciechowski, P.; Petersen, A.; Plumäkers, B.; Rink, L. Zinc enhances the number of regulatory T cells in allergen-stimulated cells from atopic subjects. Eur. J. Nutr. 2017, 56, 557–567. [Google Scholar] [CrossRef] [PubMed]
  109. Ghanizadeh, A.; Berk, M. Zinc for treating of children and adolescents with attention-deficit hyperactivity disorder: A systematic review of randomized controlled clinical trials. Eur. J. Clin. Nutr. 2013, 67, 122–124. [Google Scholar] [CrossRef] [PubMed]
  110. Rucklidge, J.J.; Johnstone, J.; Kaplan, B.J. Nutrient supplementation approaches in the treatment of ADHD. Expert Rev. Neurother. 2009, 9, 461–476. [Google Scholar] [CrossRef] [PubMed]
  111. Arnold, L.E.; DiSilvestro, R.A.; Bozzolo, D.; Bozzolo, H.; Crowl, L.; Fernandez, S.; Ramadan, Y.; Thompson, S.; Mo, X.; Abdel-Rasoul, M.; et al. Zinc for attention-deicit/hyperactivity disorder: Placebo-controlled double-blind pilot trial alone and combined with amphetamine. J. Child Adolesc. Psychopharmacol. 2011, 21, 1–19. [Google Scholar] [CrossRef] [PubMed]
  112. Nigg, J.T.; Knottnerus, G.M.; Martel, M.M.; Nikolas, M.; Cavanagh, K.; Karmaus, W.; Rappley, M.D. Low blood lead levels associated with clinically diagnosed attention-deficit/hyperactivity disorder and mediated by weak cognitive control. Biol. Psychiatry 2008, 63, 325–331. [Google Scholar] [CrossRef] [PubMed]
  113. Tuthill, R.W. Hair lead levels related to children’s classroom attention-deficit behaviour. Arch. Environ. Health 1996, 51, 214–220. [Google Scholar] [CrossRef] [PubMed]
  114. Issazadeh-Navikas, S.; Teimer, R.; Bockermann, R. Influence of dietary components on regulatory T cells. Mol. Med. 2012, 18, 95–100. [Google Scholar] [CrossRef] [PubMed]
  115. Sharif, M.R.; Madani, M.; Tabatabaei, F.; Tabatabaee, Z. The relationship between serum vitamin D level and attention deficit hyperactivity disorder. Iran J. Child Neurol. 2015, 9, 48–53. [Google Scholar] [PubMed]
  116. Holick, M.F. High prevalence of vitamin D inadequacy and implications for health. Mayo Clin. Proc. 2006, 81, 353–373. [Google Scholar] [CrossRef] [PubMed]
  117. Arns, M.; van der Heijden, K.B.; Arnold, L.E.; Kenemans, J.L. Geographic variation in the prevalence of attention-deficit/hyperactivity disorder: The sunny perspective. Biol. Psychiatry 2013, 74, 585–590. [Google Scholar] [CrossRef] [PubMed]
  118. Bateman, B.; Warner, J.O.; Hutchinson, E.; Dean, T.; Rowlandson, P.; Gant, C.; Grundy, J.; Fitzgerald, C.; Stevenson, J. The effects of a double blind, placebo controlled, sample of preschool children challenge on hyperactivity in a general population artificial food colourings and benzoate preservative. J. Arch. Dis. Child 2004, 89, 506–511. [Google Scholar] [CrossRef]
  119. McCann, D.; Barrett, A.; Cooper, A.; Crumpler, D.; Dalen, L.; Grimshaw, K.; Kitchin, E.; Lok, K.; Porteous, L.; Prince, E.; et al. Food additives and hyperactive behaviour in 3-year-old and 8/9-year-old children in the community: A randomised, double-blinded, placebo-controlled trial. Lancet 2007, 370, 1560–1567. [Google Scholar] [CrossRef]
  120. Stevens, L.J.; Zentall, S.S.; Deck, J.L.; Abate, M.L.; Watkins, B.A.; Lipp, S.R.; Burgess, J.R. Essential fatty acid metabolism in boys with attention-deficit hyperactivity disorder. Am. J. Clin. Nutr. 1995, 62, 761–768. [Google Scholar] [CrossRef] [PubMed]
  121. Boris, M.; Mandel, F. Foods and additives are common causes of the attention deficit hyperactive disorder in children. Ann. Allergy 1994, 73, 462–467. [Google Scholar]
  122. Ferro, M.A.; van Lieshout, R.J.; Ohayon, J.; Scott, J.G. Emotional and behavioural problems in adolescents and young adults with food allergy. Allergy 2015, 71, 532–540. [Google Scholar] [CrossRef] [PubMed]
  123. Richardson, A.; Puri, B. A randomized double-blind, placebo-controlled study of the effects of supplementation with highly unsaturated fatty acids on ADHD-related symptoms in children with specifc learning diffculties. Prog. Neupopsychopharmacol. Biol. Psychiatry 2002, 26, 233–239. [Google Scholar] [CrossRef]
  124. Richardson, A.J. The importance of omega-3 fatty acids for behaviour, cognition and mood. Scand. J. Nutr. 2003, 47, 92–98. [Google Scholar] [CrossRef]
  125. Bos, D.J.; van Montfort, S.J.T.; Oranje, B.; Durston, S.; Smeets, P.A.M. Effects of omega-3 polyunsaturated fatty acids on human brain morphology and function: What is the evidence? Eur. Neuropsychopharmacol. 2016, 26, 546–561. [Google Scholar] [CrossRef] [PubMed]
  126. Kim, S.J.; Zhang, Z.; Saha, A.; Sarkar, C.; Zhao, Z.; Xu, Y.; Mukherjee, A.B. Omega-3 and omega-6 fatty acids suppress ER- and oxidative stress in cultured neurons and neuronal progenitor cells from mice lacking PPT1. Neurosci. Lett. 2010, 479, 292–296. [Google Scholar] [CrossRef] [PubMed]
  127. Han, S.C.; Kang, G.J.; Ko, Y.J.; Kang, H.K.; Moon, S.W.; Ann, Y.S.; Yoo, E.S. Fermented fish oil suppresses T helper 1/2 cell response in a mouse model of atopic dermatitis via generation of CD4+CD25+Foxp3+ T cells. BMC Immunol. 2012, 13, 44. [Google Scholar] [CrossRef] [PubMed]
  128. Puri, B.K.; Martins, J.G. Which polyunsaturated fatty acids are active in children with attention-deficit hyperactivity disorder receiving PUFA supplementation? A fatty acid validated meta-regression analysis of randomized controlled trials. Prostaglandins Leukot. Essent. Fat. Acids 2014, 90, 179–189. [Google Scholar] [CrossRef] [PubMed]
  129. Hawkey, E.; Nigg, J.T. Omega-3 fatty acid and ADHD: Blood level analysis and meta-analytic extension of supplementation trials. Clin. Psychol. Rev. 2014, 34, 496–505. [Google Scholar] [CrossRef] [PubMed]
  130. Kremmyda, L.S.; Vlachava, M.; Noakes, P.S.; Diaper, N.D.; Miles, E.A.; Calder, P.C. Atopy risk in infants and children in relation to early exposure to fish, oily fish, or long-chain omega-3 fatty acids: A systematic review. Clin. Rev. Allergy Immunol. 2011, 41, 36–66. [Google Scholar] [CrossRef] [PubMed]
  131. Matsudaira, T.; Gow, R.V.; Kelly, J.; Murphy, C.; Potts, L.; Sumich, A.; Ghebremeskel, K.; Crawford, M.A.; Taylor, E. Biochemical and psychological effects of omega-3/6 supplements in male adolescents with Attention-Deficit/Hyperactivity Disorder: A randomized, placebo-controlled, clinical trial. J. Child Adolesc. Psychopharmacol. 2015, 25, 775–782. [Google Scholar] [CrossRef] [PubMed]
  132. Gillies, D.; Sinn, J.K.H.; Lad, S.S.; Leach, M.J.; Ross, M.J. Polyunsaturated fatty acids (PUFA) for attention deficit hyperactivity disorder (ADHD) in children and adolescents (review). Cochrane Database Syst. Rev. 2012, 7. [Google Scholar] [CrossRef]
  133. Manach, C.; Donovan, J.L. Pharmacokinetics and metabolism of dietary flavonoids in humans. Free Radic. Res. 2004, 38, 771–785. [Google Scholar] [CrossRef] [PubMed]
  134. Kim, J.; Kim, J.; Shim, J.; Lee, C.Y.; Lee, K.W.; Lee, H.J. Cocoa phytochemicals: Recent advances in molecular mechanisms on health. Crit. Rev. Food Sci. Nutr. 2014, 54, 1458–1472. [Google Scholar] [CrossRef] [PubMed]
  135. Trebatická, J.; Ďuračková, Z. Psychiatric Disorders and Polyphenols: Can They Be Helpful in Therapy? Oxid. Med. Cell. Longev. 2015, 2015. [Google Scholar] [CrossRef] [PubMed]
  136. Pae, M.; Wu, D. Immunomodulating effects of epigallocatechin-3-gallate from green tea: Mechanisms and applications. Food Funct. 2013, 4, 1287–1303. [Google Scholar] [CrossRef] [PubMed]
  137. Pogačnik, L.; Pirc, K.; Palmela, I.; Skrt, M.; Kim, K.S.; Brites, D.; Brito, M.A.; Ulrih, N.P.; Silva, R.F. Potential for brain accessibility and analysis of stability of selected flavonoids in relation to neuroprotection in vitro. Brain Res. 2015, 1651, 17–26. [Google Scholar] [CrossRef] [PubMed]
  138. Scalbert, A.; Johnson, I.T.; Saltmarsh, M. Polyphenols: Antioxidants and beyond. Am. J. Clin. Nutr. 2005, 81, 215s–217s. [Google Scholar] [CrossRef] [PubMed]
  139. Packer, L.; Rimbach, G.; Virgili, F. Antioxidant activity and biologic properties of a procyanidin-rich extract from pine (Pinus maritima) bark, Pycnogenol. Free Radic. Biol. Med. 1999, 27, 704–724. [Google Scholar] [CrossRef]
  140. Chovanová, Z.; Muchová, J.; Sivoňová, M.; Dvoráková, M.; Zitnanová, I.; Waczulíková, I.; Trebatická, J.; Skodácek, I.; Duracková, Z. Effect of polyphenolic extract, Pycnogenol, on the level of 8-oxoguanine in children suffering from attention deficit/hyperactivity disorder. Free Radic. Res. 2006, 40, 1003–1010. [Google Scholar] [CrossRef] [PubMed]
  141. Clifford, M.N. Diet-derived Phenols in plasma and tissues and their implications for health. Planta Med. 2004, 70, 1103–1114. [Google Scholar] [CrossRef] [PubMed]
  142. Ma, Q. Role of Nrf2 in Oxidative Stress and Toxicity. Annu. Rev. Pharmacol. Toxicol. 2013, 53, 401–426. [Google Scholar] [CrossRef] [PubMed]
  143. Tumer, T.B.; Rojas-Silva, P.; Poulev, A.; Raskin, I.; Waterman, C. Direct and Indirect Antioxidant Activity of Polyphenol- and Isothiocyanate-Enriched Fractions from Moringa oleifera. J. Agric. Food Chem. 2015, 63, 1505–1513. [Google Scholar] [CrossRef] [PubMed]
  144. Lee, M.T.; Lin, W.C.; Yu, B.; Lee, T.T. Antioxidant capacity of phytochemicals and their potential effects on oxidative status in animals—A review. Asian-Australas. J. Anim. Sci. 2017, 30, 299–308. [Google Scholar] [CrossRef] [PubMed]
  145. Bayeta, E.; Lau, B.H.S. Pycnogenol inhibits generation of inflammatory mediators in macrophages. Nutr. Res. 2000, 20, 249–259. [Google Scholar] [CrossRef]
  146. Herken, H.; Akyol, O.; Yilmaz, H.R.; Tutkun, H.; Savas, H.A.; Ozen, M.E.; Kalenderoglu, A.; Gulec, M. Nitric oxide, adenosine deaminase, xanthine oxidase and superoxide dismutase in patients with panic disorder: Alterations by antidepressant treatment. Hum. Psychopharmacol. 2006, 21, 53–59. [Google Scholar] [CrossRef] [PubMed]
  147. Selek, S.; Savas, H.A.; Gergerlioglu, H.S.; Bulut, M.; Yilmaz, H.R. Oxidative imbalance in adult attention deficit/hyperactivity disorder. Biol. Psychol. 2008, 79, 256–259. [Google Scholar] [CrossRef] [PubMed]
  148. Selek, S.; Bulut, M.; Ocak, A.R.; Kalenderoglu, A.; Savas, H.A. Evaluation of total oxidative status in adult attention deficit hyperactivity disorder and its diagnostic implications. J. Psychiatr. Res. 2012, 46, 451–455. [Google Scholar] [CrossRef] [PubMed]
  149. Canali, R.; Comitato, R.; Schonlau, F.; Virgili, F. The anti-inflammatory pharmacology of Pycnogenol in humans involves COX-2 and 5-LOXmRNA expression in leukocytes. Int. Immunopharmacol. 2009, 9, 1145–1149. [Google Scholar] [CrossRef] [PubMed]
  150. Yang, J.; Yang, X.; Chu, Y.; Li, M. Identification of Baicalin as an immunoregulatory compound by controlling T(H)17 cell differentiation. PLoS ONE 2011, 6, e17164. [Google Scholar]
  151. Hassanain, E.; Silverberg, J.I.; Norowitz, K.B.; Chice, S.; Bluth, M.H.; Brody, N.; Joks, R.; Durkin, H.G.; Smith-Norowitz, T.A. Green tea (Camelia sinensis) suppresses B cell production of IgE without inducing apoptosis. Ann. Clin. Lab. Sci. 2010, 40, 135–143. [Google Scholar] [CrossRef] [PubMed]
  152. Choi, Y.H.; Yan, G.H. Pycnogenol inhibits immunoglobulin E-mediated allergic response in mast cells. Phytother. Res. 2009, 23, 1691–1695. [Google Scholar] [CrossRef] [PubMed]
  153. Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef] [PubMed]
  154. Marín, L.; Miguélez, E.M.; Villar, C.J.; Lombó, F. Bioavailability of dietary polyphenols and gut microbiota metabolism: Antimicrobial properties. Biomed. Res. Int. 2015, 905215. [Google Scholar] [CrossRef] [PubMed]
  155. Duda-Chodak, A.; Tarko, T.; Satora, P.; Sroka, P. Interaction of dietary compounds, especially polyphenols, with the intestinal microbiota: A review. Eur. J. Nutr. 2015, 54, 325–341. [Google Scholar] [CrossRef] [PubMed]
  156. Ellam, S.; Williamson, G. Cocoa and human health. Ann. Rev. Nutr. 2013, 33, 105–128. [Google Scholar] [CrossRef] [PubMed]
  157. Kurlbaum, M.; Mülek, M.; Högger, P. Facilitated uptake of a bioactive metabolite of maritime pine bark extract (pycnogenol) into human erythrocytes. PLoS ONE 2013, 8. [Google Scholar] [CrossRef] [PubMed]
  158. Rohdewald, P.J. Pycnogenol, French Maritime Pine Bark Extract. In Encyclopedia of Dietary Supplements, 1st ed.; Coates, P.M., Cragg, G.M., Blackman, M.R., Levine, M., White, J.D., Moss, J., Eds.; Marcel Dekker, Taylor and Francis: Boca Raton, FL, USA, 2005; Volume 1, pp. 545–553. [Google Scholar]
  159. Grimm, T.; Skrabala, R.; Chovanová, Z.; Muchová, J.; Sumegová, K.; Liptáková, A.; Ďuračková, Z.; Högger, P. Single and multiple dose pharmacokinetics of maritime pine bark extract (Pycnogenol) after oral administration to healthy volunteers. BMC Clin. Pharmacol. 2006, 6. [Google Scholar] [CrossRef] [PubMed]
  160. Gilgun-Sherki, Y.; Melamed, E.; Offen, D. Oxidative stress induced-neurodegenerative diseases: The need for antioxidants that penetrate the blood brain barrier. Neuropharmacol 2001, 40, 959–975. [Google Scholar] [CrossRef]
  161. Uhlenhut, K.; Högger, P. Facilitated cellular uptake and suppression of inducible nitric oxide synthase by a metabolite of maritime pine bark extract (Pycnogenol). Free Radic. Biol. Med. 2012, 53, 305–313. [Google Scholar] [CrossRef] [PubMed]
  162. Dvořáková, M.; Sivoňová, M.; Trebatická, J.; Skodácek, I.; Waczuliková, I.; Muchová, J.; Duracková, Z. The effect of polyphenolic extract from pine bark, Pycnogenol, on the level of glutathione in children suffering from attention deficit hyperactivity disorder (ADHD). Redox Rep. 2006, 11, 163–172. [Google Scholar] [CrossRef] [PubMed]
  163. Droy-Lefaix, M.T. Effect of the antioxidant action of Ginkgo biloba extract (EGb 761) on aging and oxidative stress. Age 1997, 20, 141–149. [Google Scholar] [CrossRef] [PubMed]
  164. Puebla-Pérez, A.M.; Lozoya, X.; Villaseñor-García, M.M. Effect of Ginkgo biloba extract, EGb 761, on the cellular immune response in a hypothalamic-pituitary-adrenal axis activation model in the rat. Int. Immunopharmacol. 2003, 3, 75–80. [Google Scholar] [CrossRef]
  165. Aghili, T.; Arshami, J.; Tahmasbi, A.M.; Haghparast, A.R. Effects of Hypericum perforatum extract on IgG titer, leukocytes subset and spleen index in rats. Avicenna J. Phytomed. 2014, 4, 413–419. [Google Scholar] [PubMed]
  166. Orčić, D.Z.; Mimica-Dukić, N.M.; Francišković, M.M.; Petrović, S.S.; Jovin, E.D. Antioxidant activity relationship of phenolic compounds in Hypericum perforatum L. Chem. Cent. J. 2011, 5, 34. [Google Scholar] [CrossRef] [PubMed]
  167. Ingale, S.P.; Kasture, S.B. Antioxidant and antiparkinsonian activity of Passiflora incarnata leaves. Orient. Pharm. Exp. Med. 2014, 14, 231–236. [Google Scholar] [CrossRef]
  168. Beaumont, D.M.; Mark, T.M.J.; Hills, R.; Dixon, P.; Veit, B.; Garrett, N. The effects of chrysin, a Passiflora incarnata extract, on natural killer cell activity in male Sprague-Dawley rats undergoing abdominal surgery. AANA J. 2008, 76, 113–117. [Google Scholar] [PubMed]
  169. Salehi, B.; Imani, R.; Mohammadi, M.R.; Fallah, J.; Mohammadi, M.; Ghanizadeh, A.; Tasviechi, A.A.; Vossoughi, A.; Rezazadeh, S.A.; Akhondzadeh, S. Ginkgo biloba for attention-deficit/hyperactivity disorder in children and adolescents: A double blind, randomized controlled trial. Prog. Neuro-Psychopharmacol. Biol. Psychiatry 2010, 34, 76–80. [Google Scholar] [CrossRef] [PubMed]
  170. Uebel-von Sandersleben, H.; Rothenberger, A.; Albrecht, B.; Rothenberger, L.G.; Klement, S.; Bock, N. Ginkgo biloba extract EGb 761 in children with ADHD. Z. Kinder Jugendpsychiatrie Psychother. 2014, 42, 337–347. [Google Scholar] [CrossRef] [PubMed]
  171. Weber, W.; Vander Stoep, A.; McCarty, R.L.; Weiss, N.S.; Biederman, J.; McClellan, J. Hypericum perforatum (St John’s Wort) for attention-deficit/hyperactivity disorder in children and adolescents: A randomized controlled trial. JAMA 2008, 229, 2633–2641. [Google Scholar] [CrossRef] [PubMed]
  172. Maksyutina, N.P.; Koget, T.A. Polyphenols of the herb Hypericum perforatum and the preparation novoimanin. Chem. Nat. Compd. 1971, 7, 338–341. [Google Scholar] [CrossRef]
  173. Akhondzadeh, S.; Mohammadi, M.R.; Momeni, F. Passiflora incarnata in the treatment of attention-deficit hyperactivity disorder in children and adolescents. Therapy 2005, 2, 609–614. [Google Scholar] [CrossRef]

Share and Cite

MDPI and ACS Style

Verlaet, A.A.J.; Maasakkers, C.M.; Hermans, N.; Savelkoul, H.F.J. Rationale for Dietary Antioxidant Treatment of ADHD. Nutrients 2018, 10, 405. https://doi.org/10.3390/nu10040405

AMA Style

Verlaet AAJ, Maasakkers CM, Hermans N, Savelkoul HFJ. Rationale for Dietary Antioxidant Treatment of ADHD. Nutrients. 2018; 10(4):405. https://doi.org/10.3390/nu10040405

Chicago/Turabian Style

Verlaet, Annelies A. J., Carlijn M. Maasakkers, Nina Hermans, and Huub F. J. Savelkoul. 2018. "Rationale for Dietary Antioxidant Treatment of ADHD" Nutrients 10, no. 4: 405. https://doi.org/10.3390/nu10040405

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop