Dear reader of ADxS.org, please excuse the disruption.

ADxS.org needs about $63500 in 2024. In 2023 we received donations of about $ 32200. Unfortunately, 99.8% of our readers do not donate. If everyone who reads this request makes a small contribution, our fundraising campaign for 2024 would be over after a few days. This donation request is displayed 23,000 times a week, but only 75 people donate. If you find ADxS.org useful, please take a minute and support ADxS.org with your donation. Thank you!

Since 01.06.2021 ADxS.org is supported by the non-profit ADxS e.V..

$8975 of $63500 - as of 2024-02-29
14%
Header Image
Immune reactions and inflammation as the cause of ADHD?

Sitemap

Immune reactions and inflammation as the cause of ADHD?

The role of neuroinflammation in ADHD is becoming increasingly clear.1 There is a frequent comorbidity of ADHD with inflammatory and autoimmune diseases. Studies show an association of ADHD with elevated cytokine levels in serum and cerebrospinal fluid. Untreated ADHD is associated with elevated cytokine levels, which decrease with treatment.

ADHD-HI (predominantly hyperactive/impulsive) is often associated with hyporeactivity of the HPA axis to acute stressors, which is reflected in a flattened cortisol stress response, among other things, while ADHD-I (predominantly inattentive) very often exhibits hyperreactivity of the HPA axis (stress axis) and the endocrine stress response. The stress systems and in particular cortisol as one of the hormones of the 3rd stage of the HPA axis have a decisive effect on the immune system. While the first stages of the HPA axis use CRH and ACTH to promote the intracellular fight against inflammation through pro-inflammatory cytokines (defense against viruses and internal disruptive factors), cortisol inhibits the fight against inflammation and instead increases anti-inflammatory cytokines, which switch the immune system to fighting extracellular foreign bodies (bacteria, foreign bodies). This cortisol effect is called TH1/TH2 shift. Growth hormone, on the other hand, causes a TH2/TH1 shift.
Bacterial and viral infections during pregnancy through to infancy increase the risk of ADHD.

To date, there have only been a few studies on inflammatory markers in ADHD. Blood tests have found elevated IL-6 and TNF-alpha levels in obese children with ADHD-HI and high hyperactivity/impulsivity scores. A meta-study found no clear correlation between cytokines and ADHD subtypes.
In animal models of ADHD-HI, increased levels of reactive oxygen species and decreased levels of TNF-α and IL-10 were found in the brain. Treatment with dexmedetomidine improved hyperactivity, memory deficits and altered the gut microbiota.

Parkinson’s and ADHD not only appear to share a lack of dopamine, but also have similarities in terms of neuroinflammation.

1. Neuroinflammation in ADHD

There is increasing evidence for a role of neuroinflammation in ADHD pathophysiology:23456

  • There is frequent comorbidity of ADHD with inflammatory and autoimmune diseases:
    • Unaffected twins of ADHD sufferers have a 19% increased risk of inflammatory diseases.7
    • The increased comorbidity of ADHD and asthma has been repeatedly investigated6
  • Studies show an association of ADHD with increased inflammatory markers8 and cytokine levels in serum and cerebrospinal fluid. Studies show increased cytokine levels in untreated ADHD, which decrease in treated ADHD.
  • Studies show associations between polymorphisms in genes associated with inflammatory pathways and ADHD.
  • Early stress increases the risk of ADHD through inflammatory mechanisms. Conversely, prenatal exposure to inflammation causes changes in brain development, e.g. a reduction in volume in the cortex, which also occurs in ADHD, as well as changes in the neurotransmitter system involved in ADHD.
  • In animal models, offspring of mothers with immune activation show similarities with ADHD in terms of behavior and neurophysiological changes.

The hypothesis that ADHD is a (non-)allergic high sensitivity reaction9 correctly recognizes that inflammation and / or allergies occur more frequently than average in ADHD and that high sensitivity is a central factor in ADHD. In our opinion, however, the increased tendency to inflammation and allergies is the result of the frequently flattened endocrine stress response in ADHD-HI and regularly increased endocrine stress response in ADHD-I, which leads to an imbalance of the immune system in the form of a TH1 overbalance in ADHD-HI and a TH2 overbalance in ADHD-I due to the correspondingly low or high cortisol stress response. We therefore do not consider the increased occurrence of inflammation and allergies to be causal, but rather a reaction of the immune system to the endocrine imbalance.

Simulation of maternal viral infections causes increased subcortical dopamine function in rat offspring in adulthood but not in adolescence1011 12 with

  • Deficits in latent inhibition
  • Deficits in prepulse inhibition
  • Increased sensitivity to amphetamine
  • Cognitive impairments
  • Increased dopamine turnover
  • Changes in DA receptor binding

A meta-study on blood tests for ADHD found:13

  • IL-6 tends to be elevated
  • TNF-α reduced
  • unchanged:
    • CRP
    • IL-
    • IL-10
    • Interferon-γ

Another study found that IL-6, but not CRP, modulated the correlation between sleep problems in early childhood and ADHD at age 10.14

2. Studies on the immune system and cytokines in ADHD

2.1. Inflammatory markers in people with ADHD

Unfortunately, there are very few studies that have treated cytokines in ADHD.15
One study compared children with schizophrenia, obsessive-compulsive disorder and ADHD for IL-2, IFN-gamma, TNF-betaLT, IL-4, IL-5, IL-10 and TNF-alpha in the cerebrospinal fluid.
Obsessive-compulsive disorder correlated highly with TH-1 cytokines. IL-4 was rarely present, IL-10 not at all.
Schizophrenia correlated highly with TH-2 cytokines. IL-10 and IFN-gamma were rare.
ADHD did not have a clear TH1 or TH2 picture*.16 In the children with ADHD15

  • At 90 %: IL-2 detectable
  • Detectable at 70 % TNF
  • IL-5 detectable in 62
  • Detectable at 60 % IFN-γ
  • Detectable at 7 % IL-10

In a small study (n = 40), adolescents with ADHD showed elevated blood plasma levels of:17

  • in total
    • Platelet distribution width
  • for men
    • IL-
    • IL-6
    • TNF
    • M1 profile
  • for women
    • TNF
    • pro-/anti-inflammatory ratio

*Unfortunately, the full text of the article by Mittleman et al. has not yet been made available to us, which is why we do not know the composition of the ADHD subject group. In our opinion, however, it is not surprising that ADHD cannot be clearly assigned to one camp, considering that ADHD-HI / ADHD-C is characterized more by hypocortisolism and ADHD-I more by hypercortisolism, and cortisol causes the TH-1/TH-2 shift, which is why ADHD-HI is likely to be TH1-heavy and ADHD-I TH2-heavy. However, other studies that differentiated according to subtypes have not yet found any clear correlations. However, these studies used blood serum, which may be misleading with regard to the neuropsychological effects of cytokines.

In blood serum, one study found elevated IL-6 and TNF-alpha levels in obese children with ADHD-HI and high hyperactivity/impulsivity scores.18

One study found BDNF in blood serum increased in boys with ADHD (which correlated with poorer IQ test scores) and decreased in girls (which correlated with more omission errors on the Conners’ Continuous Performance Test). Contactin-1 (CNTN1) serum levels were unchanged in ADHD.19

One study found (unlike for epilepsy and autism spectrum disorders) no increased risk of ADHD due to brain infections, primarily with20

  • Enteroviruses
  • Streptococcus group B
  • Streptococcus pneumoniae
  • Herpes simplex

The intestinal microbiota in ADHD and ASD are quite similar in both alpha and beta diversity and differ significantly from non-affected individuals.
In addition, a subgroup of ADHD and ASD cases showed an increased concentration of lipopolysaccharide-binding protein, which correlated positively with interleukin IL-8, IL-12 and IL-13, compared to unaffected children. This indicates a disturbance of the intestinal barrier and a dysregulation of the immune system in a subgroup of children with ADHD or ASD.21

Adenosine deaminase (ADA) and dipeptidyl peptidase IV (DPP-IV, DPP4) are T cell-related enzymes.
ADA catalyzes the conversion of adenosine to inosine and deoxyadenosine to deoxyinosine. DPP-IV is expressed on the cell membrane of activated T lymphocytes and other cells as CD 26 and plays a role in diabetes.
ADA and DPP-IV serum activity were significantly increased in ADHD. There was no correlation with the Conners’ Teacher Rating Scales (CTRS-R-L) or the Conners’ Parent Rating Scales (CPRS-R-L).22

2.2. Inflammatory markers in the animal model of ADHD

In the animal model of ADHD-C, the Spontaneous(ly) hypertensive rat (SHR) found in the brain regions (not in the peripheral blood) of adult male animals:23

  • Increased levels of reactive oxygen species (ROS) in the cortex, striatum and hippocampus
  • Reduced glutathione peroxidase activity in the PFC and hippocampus
  • Reduced TNF-α levels in the PFC, the rest of the cortex, hippocampus and striatum
  • Reduced IL-1β levels in the cortex
  • Reduced IL-10 levels in the cortex

SHR treated with taurine showed reduced serum levels of C-reactive protein (CRP) and IL-1β.24 While low doses of taurine increased motor activity, high doses of taurine decreased it.

In SHR, treatment with dexmedetomidine was effective:25

  • Hyperactivity improved
  • Deficits in spatial working memory improved
  • Theta EEG rhythms normalized
  • Composition of the intestinal microbiota changes
  • enriched with beneficial intestinal bacterial genera associated with anti-inflammatory effects in SHR
  • pathological values, intestinal permeability, intestinal inflammation values and brain inflammation values significantly improved

Transplantation of the fecal microbiota from DEX-treated SHR to untreated SHR resulted in similar improvements
Dexmedetomidine is an α2-adrenoceptor agonist (like guanfacine) and acts as a short-term sedative.

3. Prenatal stress and the immune system

Prenatal stress increases levels of immune response genes, including the proinflammatory cytokines IL-6 and IL-1β, particularly in male placentas. Male infants show stress-induced locomotor hyperactivity, a hallmark of dopaminergic dysregulation, which was ameliorated by maternal treatment with nonsterioid anti-inflammatory drugs. The expression of dopamine D1 and D2 receptors was altered by prenatal stress in male offspring.26

4. Neuroinflammation and Parkinson’s disease

Like ADHD, Parkinson’s is characterized by a dopamine deficiency. In Parkinson’s, this is caused by the death of dopaminergic neurons, whereas the cause of the (probably slightly lower) dopamine deficiency in ADHD appears to have more diverse causes.
Neuroinflammation, i.e. inflammation within the CNS, is a relevant cause of Parkinson’s disease.27282930

Oxidative stress has a considerable influence on dopamine metabolism and at the same time on neuroinflammation and the neurodegeneration underlying Parkinson’s disease.31


  1. Dunn GA, Nigg JT, Sullivan EL (2019): Neuroinflammation as a risk factor for attention deficit hyperactivity disorder. Pharmacol Biochem Behav. 2019 Jul;182:22-34. doi: 10.1016/j.pbb.2019.05.005. PMID: 31103523; PMCID: PMC6855401. REVIEW

  2. Dunn, Nigg, Sullivan (2019): Neuroinflammation as a risk factor for attention deficit hyperactivity disorder. Pharmacol Biochem Behav. 2019 Jul;182:22-34. doi: 10.1016/j.pbb.2019.05.005. REVIEW

  3. Leffa, Torres, Rohde (2018): A Review on the Role of Inflammation in Attention-Deficit/Hyperactivity Disorder. Neuroimmunomodulation. 2018;25(5-6):328-333. doi: 10.1159/000489635. REVIEW

  4. Mitchell, Goldstein (2014): Inflammation in children and adolescents with neuropsychiatric disorders: a systematic review. J Am Acad Child Adolesc Psychiatry. 2014 Mar;53(3):274-96. doi: 10.1016/j.jaac.2013.11.013. REVIEW

  5. Anand, Colpo, Zeni, Zeni, Teixeira (2017): Attention-Deficit/Hyperactivity Disorder And Inflammation: What Does Current Knowledge Tell Us? A Systematic Review. Front Psychiatry. 2017 Nov 9;8:228. doi: 10.3389/fpsyt.2017.00228. eCollection 2017. REVIEW

  6. Leffa, Horta, Barros, Menezes, Martins-Silva, Hutz, Bau, Grevet, Rohde, Tovo-Rodrigues (2022): Association between Polygenic Risk Scores for ADHD and Asthma: A Birth Cohort Investigation. J Atten Disord. 2022 Mar;26(5):685-695. doi: 10.1177/10870547211020111. PMID: 34078169.

  7. Chang, Tai, Dai, Chang, Chen, Chen (2019): Risk of Atopic Diseases among Siblings of Patients with Attention-Deficit Hyperactivity Disorder: A Nationwide Population-Based Cohort Study. Int Arch Allergy Immunol. 2019;180(1):37-43. doi: 10.1159/000500831.

  8. Abdel Samei AM, Mahmoud DAM, Salem Boshra B, Abd El Moneam MHE (2023): The Interplay Between Blood Inflammatory Markers, Symptom Domains, and Severity of ADHD Disorder in Children. J Atten Disord. 2023 Sep 16:10870547231197213. doi: 10.1177/10870547231197213. PMID: 37715696.

  9. Pelsser, Buitelaar, Savelkoul (2009): ADHD as a (non) allergic hypersensitivity disorder: A hypothesis, Pediatric Allergy and Immunology, Volume 20, Issue 2, https://doi.org/10.1111/j.1399-3038.2008.00749.x

  10. Feleder, Tseng, Calhoon, O’Donnell (2009): Neonatal intrahippocampal immune challenge alters dopamine modulation of prefrontal cortical interneurons in adult rats. Biol Psychiatry. 2010 Feb 15;67(4):386-92. doi: 10.1016/j.biopsych.2009.09.028. PMID: 19914600; PMCID: PMC2900781.

  11. Ozawa, Hashimoto, Kishimoto, Shimizu, Ishikura, Iyo (2006): Immune activation during pregnancy in mice leads to dopaminergic hyperfunction and cognitive impairment in the offspring: a neurodevelopmental animal model of schizophrenia. Biol Psychiatry. 2006 Mar 15;59(6):546-54. doi: 10.1016/j.biopsych.2005.07.031. PMID: 16256957.

  12. Zuckerman, Rehavi, Nachman, Weiner (2003): Immune activation during pregnancy in rats leads to a postpubertal emergence of disrupted latent inhibition, dopaminergic hyperfunction, and altered limbic morphology in the offspring: a novel neurodevelopmental model of schizophrenia. Neuropsychopharmacology. 2003 Oct;28(10):1778-89. doi: 10.1038/sj.npp.1300248. PMID: 12865897.

  13. Misiak, Wójta-Kempa, Samochowiec, Schiweck, Aichholzer, Reif, Samochowiec, Stańczykiewicz (2022): Peripheral blood inflammatory markers in patients with attention deficit/hyperactivity disorder (ADHD): A systematic review and meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2022 Aug 30;118:110581. doi: 10.1016/j.pnpbp.2022.110581. PMID: 35660454.

  14. Morales-Muñoz I, Upthegrove R, Lawrence K, Thayakaran R, Kooij S, Gregory AM, Marwaha S (2023): The role of inflammation in the prospective associations between early childhood sleep problems and ADHD at 10 years: findings from a UK birth cohort study. J Child Psychol Psychiatry. 2023 Jan 3. doi: 10.1111/jcpp.13755. PMID: 36597271.

  15. Anand, Colpo, Zeni, Zeni, Teixeira (2017): Attention-Deficit/Hyperactivity Disorder And Inflammation: What Does Current Knowledge Tell Us? A Systematic Review. Front Psychiatry. 2017 Nov 9;8:228. doi: 10.3389/fpsyt.2017.00228. eCollection 2017.

  16. Mittleman, Castellanos, Jacobsen, Rapoport, Swedo, Shearer (1997): Cerebrospinal fluid cytokines in pediatric neuropsychiatric disease. J Immunol September 15, 1997, 159 (6) 2994-2999

  17. Ferencova N, Visnovcova Z, Ondrejka I, Hrtanek I, Bujnakova I, Kovacova V, Macejova A, Tonhajzerova I (2023): Peripheral Inflammatory Markers in Autism Spectrum Disorder and Attention Deficit/Hyperactivity Disorder at Adolescent Age. Int J Mol Sci. 2023 Jul 20;24(14):11710. doi: 10.3390/ijms241411710. PMID: 37511467; PMCID: PMC10380731. n = 40

  18. Cortese, Angriman, Comencini, Vincenzi, Maffeis (2019): Association between inflammatory cytokines and ADHD symptoms in children and adolescents with obesity: A pilot study. Psychiatry Res. 2019 May 21;278:7-11. doi: 10.1016/j.psychres.2019.05.030.

  19. Wang, Wu, Lee, Chou, Lee, Chou (2019): Peripheral Brain-Derived Neurotrophic Factor and Contactin-1 Levels in Patients with Attention-Deficit/Hyperactivity Disorder. J Clin Med. 2019 Sep 2;8(9). pii: E1366. doi: 10.3390/jcm8091366.

  20. Lin, Lin, Chou, Lee, Hong (2019): Epilepsy and Neurodevelopmental Outcomes in Children With Etiologically Diagnosed Central Nervous System Infections: A Retrospective Cohort Study. Front Neurol. 2019 May 15;10:528. doi: 10.3389/fneur.2019.00528. eCollection 2019.

  21. Bundgaard-Nielsen C, Lauritsen MB, Knudsen JK, Rold LS, Larsen MH, Hindersson P, Villadsen AB, Leutscher PDC, Hagstrøm S, Nyegaard M, Sørensen S (2023): Children and adolescents with attention deficit hyperactivity disorder and autism spectrum disorder share distinct microbiota compositions. Gut Microbes. 2023 Jan-Dec;15(1):2211923. doi: 10.1080/19490976.2023.2211923. PMID: 37199526; PMCID: PMC10197996.

  22. Naralan YS, Doğan Ö, Elgün S, Öztop DB, Kılıç BG (2023): The Activity of Adenosine Deaminase and Dipeptidyl Peptidase IV in Children With Attention Deficit Hyperactivity Disorder. J Atten Disord. 2023 Sep 11:10870547231197212. doi: 10.1177/10870547231197212. PMID: 37695015.

  23. Leffa, Bellaver, de Oliveira, de Macedo, de Freitas, Grevet, Caumo, Rohde, Quincozes-Santos, Torres (2017): Increased Oxidative Parameters and Decreased Cytokine Levels in an Animal Model of Attention-Deficit/Hyperactivity Disorder. Neurochem Res. 2017 Nov;42(11):3084-3092. doi: 10.1007/s11064-017-2341-6.

  24. Chen, Hsu, Chen, Chou, Weng, Tzang (2017):Effects of taurine on resting-state fMRI activity in spontaneously hypertensive rats. PLoS One. 2017 Jul 10;12(7):e0181122. doi: 10.1371/journal.pone.0181122. eCollection 2017.

  25. Xu X, Zhuo L, Zhang L, Peng H, Lyu Y, Sun H, Zhai Y, Luo D, Wang X, Li X, Li L, Zhang Y, Ma X, Wang Q, Li Y (2023): Dexmedetomidine alleviates host ADHD-like behaviors by reshaping the gut microbiota and reducing gut-brain inflammation. Psychiatry Res. 2023 May;323:115172. doi: 10.1016/j.psychres.2023.115172. PMID: 36958092.

  26. Bronson, Bale (2014): Prenatal stress-induced increases in placental inflammation and offspring hyperactivity are male-specific and ameliorated by maternal antiinflammatory treatment. Endocrinology. 2014 Jul;155(7):2635-46. doi: 10.1210/en.2014-1040.

  27. Hirsch, Hunot (2009): Neuroinflammation in Parkinson’s disease: a target for neuroprotection? Lancet Neurol. 2009 Apr;8(4):382-97. doi: 10.1016/S1474-4422(09)70062-6. PMID: 19296921. REVIEW

  28. Whitton (2007): Inflammation as a causative factor in the aetiology of Parkinson’s disease. Br J Pharmacol. 2007 Apr;150(8):963-76. doi: 10.1038/sj.bjp.0707167. PMID: 17339843; PMCID: PMC2013918. REVIEW

  29. Beal (2003): Mitochondria, oxidative damage, and inflammation in Parkinson’s disease. Ann N Y Acad Sci. 2003 Jun;991:120-31. doi: 10.1111/j.1749-6632.2003.tb07470.x. PMID: 12846981. REVIEW

  30. Kim, de Vellis (2005): Microglia in health and disease. J Neurosci Res. 2005 Aug 1;81(3):302-13. doi: 10.1002/jnr.20562. PMID: 15954124. REVIEW

  31. Meiser, Weindl, Hiller (2013): Complexity of dopamine metabolism. Cell Commun Signal. 2013 May 17;11(1):34. doi: 10.1186/1478-811X-11-34. PMID: 23683503; PMCID: PMC3693914. REVIEW