Submitted:
16 July 2025
Posted:
17 July 2025
You are already at the latest version
Abstract
Keywords:
1. Neuroglial Insights into Autism Spectrum Disorder
- –
- Molecular level: regulation of ions, protons, reactive oxygen species, neurotransmitters, and metabolites
- –
- Cellular level: astrocyte roles in neurogenesis and axon guidance
- –
- Synaptic network level: astrocyte and microglial functions in synaptogenesis, synaptic development, and pruning; myelinating functions of oligodendrocytes and Schwann cells
- –
- Organ level: astrocyte role in the blood–brain barrier
- –
- Systemic level: glial cells acting as central chemoreceptors [13].
2. The Role of Neuroglia in Intellectual Disability
- Deficits in intellectual functioning – Including limitations in reasoning, problem-solving, planning, abstract thinking, judgment, academic learning, and learning from experience.
- Impairments in adaptive functioning – Manifesting as significant limitations in personal independence and social responsibility across communication, social participation, and independent living.
- Onset during the developmental period – Symptoms must be evident before the age of 18.
- Mild ID: IQ between 50–70 (comprising approximately 85% of cases)
- Moderate ID: IQ between 35–50 (about 10%)
- Severe ID: IQ between 20–35 (around 4%)
- Profound ID: IQ below 20 (approximately 1%)
2.1. Astrocytes
2.2. Microglias
2.3. Oligodendrocytes
2.4. Glial Coordination: The Microenvironmental Basis of Intellectual Disability
2.5. Clinical Implications and Therapeutic Targets of Glial Dysfunction
| Biomarker | Glial Origin | Associated ID Subtypes | Clinical Relevance |
|---|---|---|---|
| GFAP | Astrocytes | Down syndrome | Neuroinflammation marker |
| S100B | Astrocytes | Fragile X, DS | Oxidative stress, neurotoxicity |
| IL-1β | Microglia | Rett syndrome | Pro-inflammatory signature |
| CX3CL1 | Neuron–Microglia axis | Various | Synaptic modulation, immune crosstalk |
3. Attention Deficit/Hyperactivity Disorder and Neuroglia
3.1. Astrocytes and ADHD
3.2. Oligodendrocytes and ADHD
3.3. Microglias and ADHD
4. Tic Disorders And Neuroglia
4.1. Epidemiology
4.2. Etiology
| Risk Factor | Associated Findings / Studies |
|---|---|
| Exposure to maternal stress during pregnancy | Interview-based study [291] |
| Nausea and vomiting during the first trimester of pregnancy | Interview-based study [291] |
| Absence of prenatal care in the first trimester | Retrospective review study [292] |
| Higher number of prenatal visits | Retrospective review study [292] |
| Low Apgar score at 5 minutes | Retrospective review study [292] |
| Low birth weight | Systematic review [293,294] |
| Maternal smoking during pregnancy | Systematic review [293,294] |
| Maternal alcohol consumption during pregnancy | Prospective prenatal cohort study [295] |
| Maternal cannabis use during pregnancy | Prospective prenatal cohort study [295] |
| Parity (number of previous births by the mother) | Prospective prenatal cohort study [295] |
| Inadequate weight gain during pregnancy | Prospective prenatal cohort study [295] |
| History of psychiatric disorder in either parent | Case-control study [296] |
| Poor parental relationship within the nuclear family | Survey-based study [297] |
4.3. Glial Cells
4.4. Microglia and Neurogenesis
4.5. Neuroinflammation and Tourette Syndrome
4.6. Inflammatory Factors
4.7. Production of Inflammatory Mediators via Peripheral Immune System Activation
4.8. Peripheral Immune System Overactivation
4.9. Microglial Activation Mediated by Inflammatory Factors
- Elevated Blood CCL5 Levels: Pathogenic infections can lead to overactivation of T lymphocytes and increased CCL5 secretion by immune cells. CCL5 can cross the BBB via its receptors CCR1 and CCR5 to enter the central nervous system. Elevated serum CCL5 levels in TS patients suggest a possible role of this chemokine in neuroinflammation [351,352]. The CCL5-CCR1 interaction may promote microglial activation via the CCR1/TPR1/ERK1/2 pathway, while CCL5-CCR5 signaling may induce neuronal pyroptosis through the CCR5/PKA/CREB/NLRP1 axis, contributing to neural dysfunction and tic expression [353,354].
- Upregulated Immune-Related Genes: Several hub genes, such as ICAM1, CCL2, HMOX1, MYC, and SOCS3, have been found to be upregulated in TS. These genes are involved in immune and inflammatory processes, particularly those related to interleukin and interferon signaling pathways [355]. Gene expression analyses in the caudate nucleus and putamen reveal that the majority of upregulated genes in these regions are immune-related and may enhance microglial activation and inflammatory responses [356].
- Histamine Deficiency: Histamine deficiency increases the secretion of pro-inflammatory mediators such as IL-1β, sensitizing microglia to inflammatory stimuli and promoting their polarization toward the M1 phenotype [357]. Histamine normally acts through H4 receptors to suppress microglial inflammatory responses and regulate their function [357,358]. A deficiency of the Hdc gene, known to be associated with TS, results in a reduction of IGF-1-positive protective microglia, increasing susceptibility to neuronal damage [358,359]. Enhanced M1 polarization, especially in the striatum, may lead to dopaminergic neuroinflammation and dysfunction, potentially triggering the development of tics [360,361,362]. These findings indicate a potential interaction between immune responses and dopaminergic dysregulation in TS pathophysiology.
4.10. Other Mechanisms of Immune-Neural Crosstalk Disruption Mediated by Inflammatory Factors
4.11. Anti-Neuronal Antibodies
4.12. Signaling Pathways Involved in Neuroimmune Interaction
4.13. CaMKII Signaling Pathway
4.14. JAK2/STAT3 Pathway
4.15. NF-κB Pathway
4.16. Other TS-Related Pathways
4.17. Postmortem and Imaging Findings
5. Neuroglial Dysfunction in Disruptive Mood Dysregulation Disorder and Irritability
6. Specific Learning Disorder in Terms of Neuroglia
6.1. Microglial Activity and Synaptic Regulation
6.2. Astrocytes and Dyslexia
6.3. Oligodendrocytes and the Relationship of Myelinization with Dyslexia
6.4. The Role of Neuron-Glia Interaction Disorders in Dyslexia
| Glial Cell Type | Possible Dysfunction | Impact on Learning Function |
|---|---|---|
| Microglia | Excessive or insufficient synaptic regulation | Lack or excess of connectivity in optimal reading network formation |
| Astrocyte | Slowness in glutamate reuptake, inadequate metabolic support | Prolongation of synaptic signals, neuronal over/underexcitation, mental fatigue and processing inefficiency during reading |
| Oligodendrocyte | Delayed/deficient myelination | Decreased neural conduction velocity, impaired synchrony between brain regions, decreased reading speed and fluency |
7. Cognitive and Neuropsychological Impacts
Intervention Approaches and Therapeutic Strategies
8. Conclusions
References
- A. American Psychiatric Association and A. P. Association, Diagnostic and statistical manual of mental disorders: DSM-IV, vol. 4. American psychiatric association Washington, DC, 1994.
- M. L. Pennington, D. Cullinan, and L. B. Southern, “Defining autism: variability in state education agency definitions of and evaluations for autism spectrum disorders,” Autism Res Treat, vol. 2014, no. 1, p. 327271, 2014.
- A. Narzisi et al., “Prevalence of Autism Spectrum Disorder in a large Italian catchment area: A school-based population study within the ASDEU project,” Epidemiol Psychiatr Sci, vol. 29, p. e5, 2020.
- M. S. Durkin and B. L. Wolfe, “Trends in autism prevalence in the US: A lagging economic indicator?,” J Autism Dev Disord, vol. 50, pp. 1095–1096, 2020.
- N. Salari et al., “The global prevalence of autism spectrum disorder: a comprehensive systematic review and meta-analysis,” Ital J Pediatr, vol. 48, no. 1, p. 112, 2022.
- A. G. Lever and H. M. Geurts, “Psychiatric co-occurring symptoms and disorders in young, middle-aged, and older adults with autism spectrum disorder,” J Autism Dev Disord, vol. 46, pp. 1916–1930, 2016.
- M. Kumar et al., “Altered cerebellar lobular volumes correlate with clinical deficits in siblings and children with ASD: evidence from toddlers,” J Transl Med, vol. 21, no. 1, p. 246, 2023.
- F. Sun, Y. Chen, Q. Gao, and Z. Zhao, “Abnormal gray matter structure in children and adolescents with high-functioning autism spectrum disorder,” Psychiatry Res Neuroimaging, vol. 327, p. 111564, 2022.
- N. Krumm, B. J. O’Roak, J. Shendure, and E. E. Eichler, “A de novo convergence of autism genetics and molecular neuroscience,” Trends Neurosci, vol. 37, no. 2, pp. 95–105, 2014.
- D. Ebrahimi-Fakhari and M. Sahin, “Autism and the synapse: emerging mechanisms and mechanism-based therapies,” Curr Opin Neurol, vol. 28, no. 2, pp. 91–102, 2015.
- S. M. Matta, E. L. Hill-Yardin, and P. J. Crack, “The influence of neuroinflammation in Autism Spectrum Disorder,” Brain Behav Immun, vol. 79, pp. 75–90, 2019.
- J. Lamanna and J. Meldolesi, “Autism Spectrum disorder: brain areas involved, neurobiological mechanisms, diagnoses and therapies,” Int J Mol Sci, vol. 25, no. 4, p. 2423, 2024.
- A. Verkhratsky, M. S. Ho, R. Zorec, and V. Parpura, “The concept of neuroglia,” Neuroglia in Neurodegenerative Diseases, pp. 1–13, 2019.
- Y. Xiong, J. Chen, and Y. Li, “Microglia and astrocytes underlie neuroinflammation and synaptic susceptibility in autism spectrum disorder,” Front Neurosci, vol. 17, p. 1125428, 2023.
- S. M. Matta, Z. Moore, F. R. Walker, E. L. Hill-Yardin, and P. J. Crack, “An altered glial phenotype in the NL3R451C mouse model of autism,” Sci Rep, vol. 10, no. 1, p. 14492, 2020.
- K. Gzielo and A. Nikiforuk, “Astroglia in autism spectrum disorder,” Int J Mol Sci, vol. 22, no. 21, p. 11544, 2021.
- N. Nassir et al., “Single-cell transcriptome identifies molecular subtype of autism spectrum disorder impacted by de novo loss-of-function variants regulating glial cells,” Hum Genomics, vol. 15, pp. 1–16, 2021.
- B. Djukic, K. B. Casper, B. D. Philpot, L.-S. Chin, and K. D. McCarthy, “Conditional knock-out of Kir4. 1 leads to glial membrane depolarization, inhibition of potassium and glutamate uptake, and enhanced short-term synaptic potentiation,” Journal of Neuroscience, vol. 27, no. 42, pp. 11354–11365, 2007.
- C. Scuderi and A. Verkhratsky, “The role of neuroglia in autism spectrum disorders,” Prog Mol Biol Transl Sci, vol. 173, pp. 301–330, 2020.
- A. K. Simhal, Y. Zuo, M. M. Perez, D. V Madison, G. Sapiro, and K. D. Micheva, “Multifaceted changes in synaptic composition and astrocytic involvement in a mouse model of fragile X syndrome,” Sci Rep, vol. 9, no. 1, p. 13855, 2019.
- L. Ferrucci, I. Cantando, F. Cordella, S. Di Angelantonio, D. Ragozzino, and P. Bezzi, “Microglia at the tripartite synapse during postnatal development: Implications for autism spectrum disorders and schizophrenia,” Cells, vol. 12, no. 24, p. 2827, 2023.
- Q. Wang et al., “Impaired calcium signaling in astrocytes modulates autism spectrum disorder-like behaviors in mice,” Nat Commun, vol. 12, no. 1, p. 3321, 2021.
- Q. Wang et al., “Impaired calcium signaling in astrocytes modulates autism spectrum disorder-like behaviors in mice,” Nat Commun, vol. 12, no. 1, p. 3321, 2021.
- L. Qin et al., “Astrocytic Neuroligin-3 influences gene expression and social behavior, but is dispensable for synapse number,” Mol Psychiatry, vol. 30, no. 1, pp. 84–96, 2025.
- D. Guneykaya et al., “Sex-specific microglia state in the Neuroligin-4 knock-out mouse model of autism spectrum disorder,” Brain Behav Immun, vol. 111, pp. 61–75, 2023.
- N. Usui, H. Kobayashi, and S. Shimada, “Neuroinflammation and oxidative stress in the pathogenesis of autism spectrum disorder,” Int J Mol Sci, vol. 24, no. 6, p. 5487, 2023.
- L. Weinhard et al., “Microglia remodel synapses by presynaptic trogocytosis and spine head filopodia induction,” Nat Commun, vol. 9, no. 1, p. 1228, 2018.
- A. Borreca et al., “Loss of interleukin 1 signaling causes impairment of microglia-mediated synapse elimination and autistic-like behaviour in mice,” Brain Behav Immun, vol. 117, pp. 493–509, 2024.
- Y. S. Kim, J. Choi, and B.-E. Yoon, “Neuron-glia interactions in neurodevelopmental disorders,” Cells, vol. 9, no. 10, p. 2176, 2020.
- E. Coutinho et al., “Persistent microglial activation and synaptic loss with behavioral abnormalities in mouse offspring exposed to CASPR2-antibodies in utero,” Acta Neuropathol, vol. 134, pp. 567–583, 2017.
- Z.-X. Xu et al., “Elevated protein synthesis in microglia causes autism-like synaptic and behavioral aberrations,” Nat Commun, vol. 11, no. 1, p. 1797, 2020.
- H. Zhu, A. Guan, J. Liu, L. Peng, Z. Zhang, and S. Wang, “Noteworthy perspectives on microglia in neuropsychiatric disorders,” J Neuroinflammation, vol. 20, no. 1, p. 223, 2023.
- N. C. Derecki et al., “Wild-type microglia arrest pathology in a mouse model of Rett syndrome,” Nature, vol. 484, no. 7392, pp. 105–109, 2012.
- I. Maezawa and L.-W. Jin, “Rett syndrome microglia damage dendrites and synapses by the elevated release of glutamate,” Journal of Neuroscience, vol. 30, no. 15, pp. 5346–5356, 2010.
- E. Bar and B. Barak, “Microglia roles in synaptic plasticity and myelination in homeostatic conditions and neurodevelopmental disorders,” Glia, vol. 67, no. 11, pp. 2125–2141, 2019.
- C. Bagni and R. S. Zukin, “A synaptic perspective of fragile X syndrome and autism spectrum disorders,” Neuron, vol. 101, no. 6, pp. 1070–1088, 2019.
- H. Zhu, A. Guan, J. Liu, L. Peng, Z. Zhang, and S. Wang, “Noteworthy perspectives on microglia in neuropsychiatric disorders,” J Neuroinflammation, vol. 20, no. 1, p. 223, 2023.
- H. Zhu, A. Guan, J. Liu, L. Peng, Z. Zhang, and S. Wang, “Noteworthy perspectives on microglia in neuropsychiatric disorders,” J Neuroinflammation, vol. 20, no. 1, p. 223, 2023.
- G. Albertini et al., “Serotonin sensing by microglia conditions the proper development of neuronal circuits and of social and adaptive skills,” Mol Psychiatry, vol. 28, no. 6, pp. 2328–2342, 2023.
- R. Coomey, R. Stowell, A. Majewska, and D. Tropea, “The role of microglia in neurodevelopmental disorders and their therapeutics,” Curr Top Med Chem, vol. 20, no. 4, pp. 272–276, 2020.
- X. Liao, J. Yang, H. Wang, and Y. Li, “Microglia mediated neuroinflammation in autism spectrum disorder,” J Psychiatr Res, vol. 130, pp. 167–176, 2020.
- A. Mordelt and L. D. de Witte, “Microglia-mediated synaptic pruning as a key deficit in neurodevelopmental disorders: Hype or hope?,” Curr Opin Neurobiol, vol. 79, p. 102674, 2023.
- L. Shi, S. E. P. Smith, N. Malkova, D. Tse, Y. Su, and P. H. Patterson, “Activation of the maternal immune system alters cerebellar development in the offspring,” Brain Behav Immun, vol. 23, no. 1, pp. 116–123, 2009.
- V. X. Han, S. Patel, H. F. Jones, and R. C. Dale, “Maternal immune activation and neuroinflammation in human neurodevelopmental disorders,” Nat Rev Neurol, vol. 17, no. 9, pp. 564–579, 2021.
- K. Canada, T. M. Evans, and K. A. Pelphrey, “Microglial regulation of white matter development and its disruption in autism spectrum disorder,” Cerebral Cortex, vol. 35, no. 4, p. bhaf109, 2025.
- H. Khelfaoui, C. Ibaceta-Gonzalez, and M. C. Angulo, “Functional myelin in cognition and neurodevelopmental disorders,” Cellular and Molecular Life Sciences, vol. 81, no. 1, p. 181, 2024.
- M. Graciarena, A. Seiffe, B. Nait-Oumesmar, and A. M. Depino, “Hypomyelination and oligodendroglial alterations in a mouse model of autism spectrum disorder,” Front Cell Neurosci, vol. 12, p. 517, 2019.
- W. Xin and J. R. Chan, “Myelin plasticity: sculpting circuits in learning and memory,” Nat Rev Neurosci, vol. 21, no. 12, pp. 682–694, 2020.
- M. Wedel et al., “Transcription factor Tcf4 is the preferred heterodimerization partner for Olig2 in oligodendrocytes and required for differentiation,” Nucleic Acids Res, vol. 48, no. 9, pp. 4839–4857, 2020.
- A. U. Carbonell et al., “Haploinsufficiency in the ANKS1B gene encoding AIDA-1 leads to a neurodevelopmental syndrome,” Nat Commun, vol. 10, no. 1, p. 3529, 2019.
- C. H. Cho et al., “ANKS1B encoded AIDA-1 regulates social behaviors by controlling oligodendrocyte function,” Nat Commun, vol. 14, no. 1, p. 8499, 2023.
- I. Fischer et al., “Shank3 mutation impairs glutamate signaling and myelination in ASD mouse model and human iPSC-derived OPCs,” Sci Adv, vol. 10, no. 41, p. eadl4573, 2024.
- American Psychiatric Association, Diagnostic and Statistical Manual of Mental Disorders. American Psychiatric Association, 2013. [CrossRef]
- D. R. Patel, R. Apple, S. Kanungo, and A. Akkal, “Narrative review of intellectual disability: definitions, evaluation and principles of treatment,” Pediatric Medicine, vol. 1, pp. 11–11, Dec. 2018. [CrossRef]
- D. R. Patel, D. E. Greydanus, J. Merrick, and I. L. Rubin, “Introduction to Intellectual and Developmental Disabilities,” in Health Care for People with Intellectual and Developmental Disabilities across the Lifespan, Cham: Springer International Publishing, 2016, pp. 5–12. [CrossRef]
- M. E. Snell et al., “Characteristics and Needs of People With Intellectual Disability Who Have Higher IQs,” Intellect Dev Disabil, vol. 47, no. 3, pp. 220–233, Jun. 2009. [CrossRef]
- K. Lee, M. Cascella, and R. Marwaha, Intellectual Disability. 2025.
- O. Obi, K. Van Naarden Braun, J. Baio, C. Drews-Botsch, O. Devine, and M. Yeargin-Allsopp, “Effect of Incorporating Adaptive Functioning Scores on the Prevalence of Intellectual Disability,” Am J Intellect Dev Disabil, vol. 116, no. 5, pp. 360–370, Sep. 2011. [CrossRef]
- S. L. Milne, J. L. McDonald, and E. J. Comino, “Adaptive function in preschoolers in relation to developmental delay and diagnosis of autism spectrum disorders: Insights from a clinical sample,” Autism, vol. 17, no. 6, pp. 743–753, Nov. 2013. [CrossRef]
- K. Lee, M. Cascella, and R. Marwaha, Intellectual Disability. 2025.
- C. Chen et al., “Role of astroglia in Down’s syndrome revealed by patient-derived human-induced pluripotent stem cells,” Nat Commun, vol. 5, no. 1, p. 4430, Jul. 2014. [CrossRef]
- N. A. Neuman and E. P. Henske, “Non-canonical functions of the tuberous sclerosis complex-Rheb signalling axis,” EMBO Mol Med, vol. 3, no. 4, pp. 189–200, Apr. 2011. [CrossRef]
- L. K. K. Pacey, S. Guan, S. Tharmalingam, C. Thomsen, and D. R. Hampson, “Persistent astrocyte activation in the fragile <scp>X</scp> mouse cerebellum,” Brain Behav, vol. 5, no. 10, Oct. 2015. [CrossRef]
- T. Andoh-Noda et al., “Differentiation of multipotent neural stem cells derived from Rett syndrome patients is biased toward the astrocytic lineage,” Mol Brain, vol. 8, no. 1, p. 31, Dec. 2015. [CrossRef]
- O. Purugganan, “Intellectual Disabilities,” Pediatr Rev, vol. 39, no. 6, pp. 299–309, Jun. 2018. [CrossRef]
- T. R. Hammond, D. Robinton, and B. Stevens, “Microglia and the Brain: Complementary Partners in Development and Disease,” Annu Rev Cell Dev Biol, vol. 34, no. 1, pp. 523–544, Oct. 2018. [CrossRef]
- B. Wang, L. Zou, M. Li, and L. Zhou, “Astrocyte: A Foe or a Friend in Intellectual Disability-Related Diseases,” Front Synaptic Neurosci, vol. 14, Jun. 2022. [CrossRef]
- Á. Fernández-Blanco and M. Dierssen, “Rethinking Intellectual Disability from Neuro- to Astro-Pathology,” Int J Mol Sci, vol. 21, no. 23, p. 9039, Nov. 2020. [CrossRef]
- C. Murphy-Royal, S. Ching, and T. Papouin, “Contextual guidance: An integrated theory for astrocytes function in brain circuits and behavior,” Nov. 2022. [CrossRef]
- B. S. Khakh and M. V Sofroniew, “Diversity of astrocyte functions and phenotypes in neural circuits,” Nat Neurosci, vol. 18, no. 7, pp. 942–952, Jul. 2015. [CrossRef]
- M. V. Sofroniew and H. V. Vinters, “Astrocytes: biology and pathology,” Acta Neuropathol, vol. 119, no. 1, pp. 7–35, Jan. 2010. [CrossRef]
- M. V Sofroniew, “Astrocyte Reactivity: Subtypes, States, and Functions in CNS Innate Immunity.,” Trends Immunol, vol. 41, no. 9, pp. 758–770, Sep. 2020. [CrossRef]
- M. A. Anderson, Y. Ao, and M. V Sofroniew, “Heterogeneity of reactive astrocytes.,” Neurosci Lett, vol. 565, pp. 23–9, Apr. 2014. [CrossRef]
- C. Escartin et al., “Reactive astrocyte nomenclature, definitions, and future directions.,” Nat Neurosci, vol. 24, no. 3, pp. 312–325, Mar. 2021. [CrossRef]
- S. E. Khoshnam, W. Winlow, M. Farzaneh, Y. Farbood, and H. F. Moghaddam, “Pathogenic mechanisms following ischemic stroke.,” Neurol Sci, vol. 38, no. 7, pp. 1167–1186, Jul. 2017. [CrossRef]
- R. Dringen, M. Brandmann, M. C. Hohnholt, and E.-M. Blumrich, “Glutathione-Dependent Detoxification Processes in Astrocytes,” Neurochem Res, vol. 40, no. 12, pp. 2570–2582, Dec. 2015. [CrossRef]
- J. Frade et al., “Glutamate induces release of glutathione from cultured rat astrocytes--a possible neuroprotective mechanism?,” J Neurochem, vol. 105, no. 4, pp. 1144–52, May 2008. [CrossRef]
- G. Carmignoto and M. Gómez-Gonzalo, “The contribution of astrocyte signalling to neurovascular coupling.,” Brain Res Rev, vol. 63, no. 1–2, pp. 138–48, May 2010. [CrossRef]
- P. Kowiański, G. Lietzau, A. Steliga, M. Waśkow, and J. Moryś, “The astrocytic contribution to neurovascular coupling--still more questions than answers?,” Neurosci Res, vol. 75, no. 3, pp. 171–83, Mar. 2013. [CrossRef]
- S. Liebner, R. M. Dijkhuizen, Y. Reiss, K. H. Plate, D. Agalliu, and G. Constantin, “Functional morphology of the blood–brain barrier in health and disease,” Acta Neuropathol, vol. 135, no. 3, pp. 311–336, Mar. 2018. [CrossRef]
- P. J. Magistretti and I. Allaman, “A Cellular Perspective on Brain Energy Metabolism and Functional Imaging,” Neuron, vol. 86, no. 4, pp. 883–901, May 2015. [CrossRef]
- N. Cresto, L.-E. Pillet, P. Billuart, and N. Rouach, “Do Astrocytes Play a Role in Intellectual Disabilities?,” Trends Neurosci, vol. 42, no. 8, pp. 518–527, Aug. 2019. [CrossRef]
- E. Giacometti, S. Luikenhuis, C. Beard, and R. Jaenisch, “Partial rescue of MeCP2 deficiency by postnatal activation of MeCP2.,” Proc Natl Acad Sci U S A, vol. 104, no. 6, pp. 1931–6, Feb. 2007. [CrossRef]
- J. Guy, J. Gan, J. Selfridge, S. Cobb, and A. Bird, “Reversal of Neurological Defects in a Mouse Model of Rett Syndrome,” Science (1979), vol. 315, no. 5815, pp. 1143–1147, Feb. 2007. [CrossRef]
- N. Ballas, D. T. Lioy, C. Grunseich, and G. Mandel, “Non–cell autonomous influence of MeCP2-deficient glia on neuronal dendritic morphology,” Nat Neurosci, vol. 12, no. 3, pp. 311–317, Mar. 2009. [CrossRef]
- C. Cheng, S. K. M. Lau, and L. C. Doering, “Astrocyte-secreted thrombospondin-1 modulates synapse and spine defects in the fragile X mouse model,” Mol Brain, vol. 9, no. 1, p. 74, Dec. 2016. [CrossRef]
- S. Jacobs and L. C. Doering, “Astrocytes Prevent Abnormal Neuronal Development in the Fragile X Mouse,” The Journal of Neuroscience, vol. 30, no. 12, pp. 4508–4514, Mar. 2010. [CrossRef]
- B. Wang, L. Zou, M. Li, and L. Zhou, “Astrocyte: A Foe or a Friend in Intellectual Disability-Related Diseases,” Front Synaptic Neurosci, vol. 14, Jun. 2022. [CrossRef]
- C. Cheng, S. K. M. Lau, and L. C. Doering, “Astrocyte-secreted thrombospondin-1 modulates synapse and spine defects in the fragile X mouse model,” Mol Brain, vol. 9, no. 1, p. 74, Dec. 2016. [CrossRef]
- B. Ponroy Bally and K. K. Murai, “Astrocytes in Down Syndrome Across the Lifespan,” Front Cell Neurosci, vol. 15, Aug. 2021. [CrossRef]
- L. K. K. Pacey and L. C. Doering, “Developmental expression of FMRP in the astrocyte lineage: Implications for fragile X syndrome,” Glia, vol. 55, no. 15, pp. 1601–1609, Nov. 2007. [CrossRef]
- K. E. Reynolds, C. R. Wong, and A. L. Scott, “Astrocyte-mediated purinergic signaling is upregulated in a mouse model of Fragile X syndrome,” Glia, vol. 69, no. 7, pp. 1816–1832, Jul. 2021. [CrossRef]
- I. Maezawa, S. Swanberg, D. Harvey, J. M. LaSalle, and L.-W. Jin, “Rett Syndrome Astrocytes Are Abnormal and Spread MeCP2 Deficiency through Gap Junctions,” The Journal of Neuroscience, vol. 29, no. 16, pp. 5051–5061, Apr. 2009. [CrossRef]
- M. B. Ramocki and H. Y. Zoghbi, “Failure of neuronal homeostasis results in common neuropsychiatric phenotypes,” Nature, vol. 455, no. 7215, pp. 912–918, Oct. 2008. [CrossRef]
- G. O. Mizuno et al., “Aberrant Calcium Signaling in Astrocytes Inhibits Neuronal Excitability in a Human Down Syndrome Stem Cell Model,” Cell Rep, vol. 24, no. 2, pp. 355–365, Jul. 2018. [CrossRef]
- R. Ballestín et al., “Astrocytes of the murine model for Down Syndrome Ts65Dn display reduced intracellular ionic zinc,” Neurochem Int, vol. 75, pp. 48–53, Sep. 2014. [CrossRef]
- M. Wong, “The role of glia in epilepsy, intellectual disability, and other neurodevelopmental disorders in tuberous sclerosis complex,” J Neurodev Disord, vol. 11, no. 1, p. 30, Dec. 2019. [CrossRef]
- L. A. Jansen, E. J. Uhlmann, P. B. Crino, D. H. Gutmann, and M. Wong, “Epileptogenesis and Reduced Inward Rectifier Potassium Current in Tuberous Sclerosis Complex-1–Deficient Astrocytes,” Epilepsia, vol. 46, no. 12, pp. 1871–1880, Dec. 2005. [CrossRef]
- D. H. Woo et al., “TREK-1 and Best1 Channels Mediate Fast and Slow Glutamate Release in Astrocytes upon GPCR Activation,” Cell, vol. 151, no. 1, pp. 25–40, Sep. 2012. [CrossRef]
- L. Xing, T. Yang, S. Cui, and G. Chen, “Connexin Hemichannels in Astrocytes: Role in CNS Disorders,” Front Mol Neurosci, vol. 12, Feb. 2019. [CrossRef]
- M. L. Olsen, B. S. Khakh, S. N. Skatchkov, M. Zhou, C. J. Lee, and N. Rouach, “New Insights on Astrocyte Ion Channels: Critical for Homeostasis and Neuron-Glia Signaling,” The Journal of Neuroscience, vol. 35, no. 41, pp. 13827–13835, Oct. 2015. [CrossRef]
- N. L. Pacheco et al., “RNA sequencing and proteomics approaches reveal novel deficits in the cortex of Mecp2-deficient mice, a model for Rett syndrome,” Mol Autism, vol. 8, no. 1, p. 56, Dec. 2017. [CrossRef]
- H. Higashimori et al., “Astroglial FMRP-dependent translational down-regulation of mGluR5 underlies glutamate transporter GLT1 dysregulation in the fragile X mouse,” Hum Mol Genet, vol. 22, no. 10, pp. 2041–2054, May 2013. [CrossRef]
- L. Wang et al., “Imbalance between Glutamate and GABA in Fmr1 Knockout Astrocytes Influences Neuronal Development,” Genes (Basel), vol. 7, no. 8, p. 45, Aug. 2016. [CrossRef]
- R. Höftberger et al., “Distribution and cellular localization of adrenoleukodystrophy protein in human tissues: Implications for X-linked adrenoleukodystrophy,” Neurobiol Dis, vol. 28, no. 2, pp. 165–174, Nov. 2007. [CrossRef]
- M. Wong et al., “Impaired glial glutamate transport in a mouse tuberous sclerosis epilepsy model,” Ann Neurol, vol. 54, no. 2, pp. 251–256, Aug. 2003. [CrossRef]
- Y. Okabe, T. Takahashi, C. Mitsumasu, K. Kosai, E. Tanaka, and T. Matsuishi, “Alterations of Gene Expression and Glutamate Clearance in Astrocytes Derived from an MeCP2-Null Mouse Model of Rett Syndrome,” PLoS One, vol. 7, no. 4, p. e35354, Apr. 2012. [CrossRef]
- A. M. Iyer et al., “Metabotropic Glutamate Receptor 5 in Down’s Syndrome Hippocampus During Development: Increased Expression in Astrocytes,” Curr Alzheimer Res, vol. 11, no. 7, pp. 694–705, Aug. 2014. [CrossRef]
- C. Cheng, S. K. M. Lau, and L. C. Doering, “Astrocyte-secreted thrombospondin-1 modulates synapse and spine defects in the fragile X mouse model,” Mol Brain, vol. 9, no. 1, p. 74, Dec. 2016. [CrossRef]
- A. Verkhratsky et al., “Astrocytes in human central nervous system diseases: a frontier for new therapies,” Signal Transduct Target Ther, vol. 8, no. 1, p. 396, Oct. 2023. [CrossRef]
- Q. Li and B. A. Barres, “Microglia and macrophages in brain homeostasis and disease,” Nat Rev Immunol, vol. 18, no. 4, pp. 225–242, Apr. 2018. [CrossRef]
- D. A. Menassa and D. Gomez-Nicola, “Microglial Dynamics During Human Brain Development,” Front Immunol, vol. 9, May 2018. [CrossRef]
- I. Lago-Baldaia, V. M. Fernandes, and S. D. Ackerman, “More Than Mortar: Glia as Architects of Nervous System Development and Disease,” Front Cell Dev Biol, vol. 8, Dec. 2020. [CrossRef]
- K. Sharma, K. Bisht, and U. B. Eyo, “A Comparative Biology of Microglia Across Species,” Front Cell Dev Biol, vol. 9, Apr. 2021. [CrossRef]
- O. Staszewski and N. Hagemeyer, “Unique microglia expression profile in developing white matter,” BMC Res Notes, vol. 12, no. 1, p. 367, Dec. 2019. [CrossRef]
- A. Wlodarczyk et al., “A novel microglial subset plays a key role in myelinogenesis in developing brain.,” EMBO J, vol. 36, no. 22, pp. 3292–3308, Nov. 2017. [CrossRef]
- L. Liu, J. Liu, J. Bao, Q. Bai, and G. Wang, “Interaction of Microglia and Astrocytes in the Neurovascular Unit,” Front Immunol, vol. 11, Jul. 2020. [CrossRef]
- M. S. Thion et al., “Microbiome Influences Prenatal and Adult Microglia in a Sex-Specific Manner.,” Cell, vol. 172, no. 3, pp. 500-516.e16, Jan. 2018. [CrossRef]
- L. Chye, S. Riek, A. de Rugy, R. G. Carson, and T. J. Carroll, “Unilateral movement preparation causes task-specific modulation of TMS responses in the passive, opposite limb,” J Physiol, vol. 596, no. 16, pp. 3725–3738, Aug. 2018. [CrossRef]
- M. W. Salter and B. Stevens, “Microglia emerge as central players in brain disease,” Nat Med, vol. 23, no. 9, pp. 1018–1027, Sep. 2017. [CrossRef]
- R. C. Paolicelli et al., “Synaptic Pruning by Microglia Is Necessary for Normal Brain Development,” Science (1979), vol. 333, no. 6048, pp. 1456–1458, Sep. 2011. [CrossRef]
- C. Arcuri, C. Mecca, R. Bianchi, I. Giambanco, and R. Donato, “The Pathophysiological Role of Microglia in Dynamic Surveillance, Phagocytosis and Structural Remodeling of the Developing CNS,” Front Mol Neurosci, vol. 10, Jun. 2017. [CrossRef]
- I. Maezawa and L.-W. Jin, “Rett Syndrome Microglia Damage Dendrites and Synapses by the Elevated Release of Glutamate,” The Journal of Neuroscience, vol. 30, no. 15, pp. 5346–5356, Apr. 2010. [CrossRef]
- N. C. Derecki et al., “Wild-type microglia arrest pathology in a mouse model of Rett syndrome,” Nature, vol. 484, no. 7392, pp. 105–109, Apr. 2012. [CrossRef]
- U. Kahanovitch, K. C. Patterson, R. Hernandez, and M. L. Olsen, “Glial Dysfunction in MeCP2 Deficiency Models: Implications for Rett Syndrome,” Int J Mol Sci, vol. 20, no. 15, p. 3813, Aug. 2019. [CrossRef]
- K. E. Reynolds, M. Napier, F. Fei, K. Green, and A. L. Scott, “Dysregulated Purinergic Signalling in Fragile X Syndrome Cortical Astrocytes,” Neuromolecular Med, vol. 26, no. 1, p. 36, Sep. 2024. [CrossRef]
- B. Hajizadeh Maleki and B. Tartibian, “RETRACTED: High-intensity interval training modulates male factor infertility through anti-inflammatory and antioxidative mechanisms in infertile men: A randomized controlled trial,” Cytokine, vol. 125, p. 154861, Jan. 2020. [CrossRef]
- R. Rahimian, K. Perlman, C. Canonne, and N. Mechawar, “Targeting microglia–oligodendrocyte crosstalk in neurodegenerative and psychiatric disorders,” Drug Discov Today, vol. 27, no. 9, pp. 2562–2573, Sep. 2022. [CrossRef]
- A. Boerboom, C. Reusch, A. Pieltain, A. Chariot, and R. Franzen, “KIAA1199: A novel regulator of MEK/ERK-induced Schwann cell dedifferentiation,” Glia, vol. 65, no. 10, pp. 1682–1696, Oct. 2017. [CrossRef]
- K.-A. Nave and H. B. Werner, “Myelination of the Nervous System: Mechanisms and Functions,” Annu Rev Cell Dev Biol, vol. 30, no. 1, pp. 503–533, Oct. 2014. [CrossRef]
- M. Cichorek, P. Kowiański, G. Lietzau, J. Lasek, and J. Moryś, “Neuroglia — development and role in physiological and pathophysiological processes,” Folia Morphol (Warsz), vol. 80, no. 4, pp. 766–775, Dec. 2021. [CrossRef]
- A. M. Butt and A. Kalsi, “Inwardly rectifying potassium channels (Kir) in central nervous system glia: a special role for Kir4.1 in glial functions,” J Cell Mol Med, vol. 10, no. 1, pp. 33–44, Jan. 2006. [CrossRef]
- A. M. Butt, R. F. Fern, and C. Matute, “Neurotransmitter signaling in white matter,” Glia, vol. 62, no. 11, pp. 1762–1779, Nov. 2014. [CrossRef]
- C. Escartin et al., “Reactive astrocyte nomenclature, definitions, and future directions.,” Nat Neurosci, vol. 24, no. 3, pp. 312–325, Mar. 2021. [CrossRef]
- S. Kuhn, L. Gritti, D. Crooks, and Y. Dombrowski, “Oligodendrocytes in Development, Myelin Generation and Beyond,” Cells, vol. 8, no. 11, p. 1424, Nov. 2019. [CrossRef]
- O. Giotakos, “Is psychosis a dysmyelination-related information-processing disorder?,” Psychiatriki, vol. 30, no. 3, pp. 245–255, 2019. [CrossRef]
- H.-G. Bernstein, G. Keilhoff, H. Dobrowolny, P. C. Guest, and J. Steiner, “Perineuronal oligodendrocytes in health and disease: the journey so far.,” Rev Neurosci, vol. 31, no. 1, pp. 89–99, Dec. 2019. [CrossRef]
- J. Janowska, J. Gargas, M. Ziemka-Nalecz, T. Zalewska, and J. Sypecka, “Oligodendrocyte Response to Pathophysiological Conditions Triggered by Episode of Perinatal Hypoxia-Ischemia: Role of IGF-1 Secretion by Glial Cells,” Mol Neurobiol, vol. 57, no. 10, pp. 4250–4268, Oct. 2020. [CrossRef]
- I. Pijuan, E. Balducci, C. Soto-Sánchez, E. Fernández, M. J. Barallobre, and M. L. Arbonés, “Impaired macroglial development and axonal conductivity contributes to the neuropathology of DYRK1A-related intellectual disability syndrome,” Sci Rep, vol. 12, no. 1, p. 19912, Nov. 2022. [CrossRef]
- J. Maiuolo et al., “Environmental and Nutritional ‘Stressors’ and Oligodendrocyte Dysfunction: Role of Mitochondrial and Endoplasmatic Reticulum Impairment,” Biomedicines, vol. 8, no. 12, p. 553, Nov. 2020. [CrossRef]
- T. A. Forbes et al., “Environmental enrichment ameliorates perinatal brain injury and promotes functional white matter recovery,” Nat Commun, vol. 11, no. 1, p. 964, Feb. 2020. [CrossRef]
- C. Arcuri, C. Mecca, R. Bianchi, I. Giambanco, and R. Donato, “The Pathophysiological Role of Microglia in Dynamic Surveillance, Phagocytosis and Structural Remodeling of the Developing CNS,” Front Mol Neurosci, vol. 10, Jun. 2017. [CrossRef]
- T. A. Forbes et al., “Environmental enrichment ameliorates perinatal brain injury and promotes functional white matter recovery,” Nat Commun, vol. 11, no. 1, p. 964, Feb. 2020. [CrossRef]
- S. R. PLISZKA et al., “The Texas Children’s Medication Algorithm Project: Revision of the Algorithm for Pharmacotherapy of Attention-Deficit/Hyperactivity Disorder,” J Am Acad Child Adolesc Psychiatry, vol. 45, no. 6, pp. 642–657, Jun. 2006. [CrossRef]
- E. S. Vitola et al., “Exploring DSM-5 ADHD criteria beyond young adulthood: phenomenology, psychometric properties and prevalence in a large three-decade birth cohort,” Psychol Med, vol. 47, no. 4, pp. 744–754, Mar. 2017. [CrossRef]
- E. G. Willcutt, “The Prevalence of DSM-IV Attention-Deficit/Hyperactivity Disorder: A Meta-Analytic Review,” Neurotherapeutics, vol. 9, no. 3, pp. 490–499, Jul. 2012. [CrossRef]
- R. Thomas, S. Sanders, J. Doust, E. Beller, and P. Glasziou, “Prevalence of Attention-Deficit/Hyperactivity Disorder: A Systematic Review and Meta-analysis,” Pediatrics, vol. 135, no. 4, pp. e994–e1001, Apr. 2015. [CrossRef]
- S. V. Faraone et al., “Attention-deficit/hyperactivity disorder,” Nat Rev Dis Primers, vol. 1, no. 1, p. 15020, Aug. 2015. [CrossRef]
- V. Simon, P. Czobor, S. Bálint, Á. Mészáros, and I. Bitter, “Prevalence and correlates of adult attention-deficit hyperactivity disorder: meta-analysis,” British Journal of Psychiatry, vol. 194, no. 3, pp. 204–211, Mar. 2009. [CrossRef]
- A. Rodriguez and G. Bohlin, “Are maternal smoking and stress during pregnancy related to ADHD symptoms in children?,” Journal of Child Psychology and Psychiatry, vol. 46, no. 3, pp. 246–254, Mar. 2005. [CrossRef]
- Q. Chen et al., “Familial aggregation of attention-deficit/hyperactivity disorder,” Journal of Child Psychology and Psychiatry, vol. 58, no. 3, pp. 231–239, Mar. 2017. [CrossRef]
- S. V. Faraone et al., “Molecular Genetics of Attention-Deficit/Hyperactivity Disorder,” Biol Psychiatry, vol. 57, no. 11, pp. 1313–1323, Jun. 2005. [CrossRef]
- H. Larsson, Z. Chang, B. M. D’Onofrio, and P. Lichtenstein, “The heritability of clinically diagnosed attention deficit hyperactivity disorder across the lifespan,” Psychol Med, vol. 44, no. 10, pp. 2223–2229, Jul. 2014. [CrossRef]
- S. E. Bergen, C. O. Gardner, and K. S. Kendler, “Age-Related Changes in Heritability of Behavioral Phenotypes Over Adolescence and Young Adulthood: A Meta-Analysis,” Twin Research and Human Genetics, vol. 10, no. 3, pp. 423–433, Jun. 2007. [CrossRef]
- T. J. C. Polderman et al., “Meta-analysis of the heritability of human traits based on fifty years of twin studies,” Nat Genet, vol. 47, no. 7, pp. 702–709, Jul. 2015. [CrossRef]
- J. C. Corona, “Role of Oxidative Stress and Neuroinflammation in Attention-Deficit/Hyperactivity Disorder,” Antioxidants, vol. 9, no. 11, p. 1039, Oct. 2020. [CrossRef]
- R. D. Oades, A.-M. Myint, M. R. Dauvermann, B. G. Schimmelmann, and M. J. Schwarz, “Attention-deficit hyperactivity disorder (ADHD) and glial integrity: an exploration of associations of cytokines and kynurenine metabolites with symptoms and attention,” Behavioral and Brain Functions, vol. 6, no. 1, p. 32, 2010. [CrossRef]
- A. H. Darwish, T. M. Elgohary, and N. A. Nosair, “Serum Interleukin-6 Level in Children With Attention-Deficit Hyperactivity Disorder (ADHD),” J Child Neurol, vol. 34, no. 2, pp. 61–67, Feb. 2019. [CrossRef]
- A. Buske-Kirschbaum, J. Schmitt, F. Plessow, M. Romanos, S. Weidinger, and V. Roessner, “Psychoendocrine and psychoneuroimmunological mechanisms in the comorbidity of atopic eczema and attention deficit/hyperactivity disorder,” Psychoneuroendocrinology, vol. 38, no. 1, pp. 12–23, Jan. 2013. [CrossRef]
- J.-M. Beaulieu and R. R. Gainetdinov, “The Physiology, Signaling, and Pharmacology of Dopamine Receptors,” Pharmacol Rev, vol. 63, no. 1, pp. 182–217, Mar. 2011. [CrossRef]
- L.-J. Wang et al., “Attention deficit–hyperactivity disorder is associated with allergic symptoms and low levels of hemoglobin and serotonin,” Sci Rep, vol. 8, no. 1, p. 10229, Jul. 2018. [CrossRef]
- J. Bauer et al., “Hyperactivity and impulsivity in adult attention-deficit/hyperactivity disorder is related to glutamatergic dysfunction in the anterior cingulate cortex,” The World Journal of Biological Psychiatry, vol. 19, no. 7, pp. 538–546, Oct. 2018. [CrossRef]
- S. Bollmann et al., “Developmental changes in gamma-aminobutyric acid levels in attention-deficit/hyperactivity disorder,” Transl Psychiatry, vol. 5, no. 6, pp. e589–e589, Jun. 2015. [CrossRef]
- J. Cheng, A. Liu, M. Y. Shi, and Z. Yan, “Disrupted Glutamatergic Transmission in Prefrontal Cortex Contributes to Behavioral Abnormality in an Animal Model of ADHD,” Neuropsychopharmacology, vol. 42, no. 10, pp. 2096–2104, Sep. 2017. [CrossRef]
- B. S. da Silva, E. H. Grevet, L. C. F. Silva, J. K. N. Ramos, D. L. Rovaris, and C. H. D. Bau, “An overview on neurobiology and therapeutics of attention-deficit/hyperactivity disorder,” Discover Mental Health, vol. 3, no. 1, p. 2, Jan. 2023. [CrossRef]
- F. Passarelli et al., “Anti-Purkinje cell antibody as a biological marker in attention deficit/hyperactivity disorder: A pilot study,” J Neuroimmunol, vol. 258, no. 1–2, pp. 67–70, May 2013. [CrossRef]
- R. Donfrancesco et al., “Anti-Yo Antibodies in Children With ADHD: First Results About Serum Cytokines,” J Atten Disord, vol. 24, no. 11, pp. 1497–1502, Sep. 2020. [CrossRef]
- G. Giana et al., “Detection of auto-antibodies to DAT in the serum: Interactions with DAT genotype and psycho-stimulant therapy for ADHD,” J Neuroimmunol, vol. 278, pp. 212–222, Jan. 2015. [CrossRef]
- M. Toto et al., “Antibasal Ganglia Antibodies and Antistreptolysin O in Noncomorbid ADHD,” J Atten Disord, vol. 19, no. 11, pp. 965–970, Nov. 2015. [CrossRef]
- S. Gnanavel, P. Sharma, P. Kaushal, and S. Hussain, “Attention deficit hyperactivity disorder and comorbidity: A review of literature,” World J Clin Cases, vol. 7, no. 17, pp. 2420–2426, Sep. 2019. [CrossRef]
- T. J. Spencer, J. Biederman, and E. Mick, “Attention-Deficit/Hyperactivity Disorder: Diagnosis, Lifespan, Comorbidities, and Neurobiology,” J Pediatr Psychol, vol. 32, no. 6, pp. 631–642, May 2007. [CrossRef]
- J.-C. Seo et al., “Prevalence and Comorbidities of Attention Deficit Hyperactivity Disorder Among Adults and Children/Adolescents in Korea,” Clinical Psychopharmacology and Neuroscience, vol. 20, no. 1, pp. 126–134, Feb. 2022. [CrossRef]
- C. M. Jensen and H.-C. Steinhausen, “Comorbid mental disorders in children and adolescents with attention-deficit/hyperactivity disorder in a large nationwide study,” ADHD Attention Deficit and Hyperactivity Disorders, vol. 7, no. 1, pp. 27–38, Mar. 2015. [CrossRef]
- M. K. Akmatov, T. Ermakova, and J. Bätzing, “Psychiatric and Nonpsychiatric Comorbidities Among Children With ADHD: An Exploratory Analysis of Nationwide Claims Data in Germany,” J Atten Disord, vol. 25, no. 6, pp. 874–884, Apr. 2021. [CrossRef]
- F. Catalá-López et al., “The pharmacological and non-pharmacological treatment of attention deficit hyperactivity disorder in children and adolescents: A systematic review with network meta-analyses of randomised trials,” PLoS One, vol. 12, no. 7, p. e0180355, Jul. 2017. [CrossRef]
- National Institute for Health and Care Excellence, “Attention deficit hyperactivity disorder: diagnosis and management,” London, 2018.
- V. Nimmo-Smith et al., “Non-pharmacological interventions for adult ADHD: a systematic review,” Psychol Med, vol. 50, no. 4, pp. 529–541, Mar. 2020. [CrossRef]
- B. Bolea-Alamañac et al., “Evidence-based guidelines for the pharmacological management of attention deficit hyperactivity disorder: Update on recommendations from the British Association for Psychopharmacology,” Journal of Psychopharmacology, vol. 28, no. 3, pp. 179–203, Mar. 2014. [CrossRef]
- S. Cortese, “Pharmacologic Treatment of Attention Deficit–Hyperactivity Disorder,” New England Journal of Medicine, vol. 383, no. 11, pp. 1050–1056, Sep. 2020. [CrossRef]
- T. Fawns, “Attention Deficit and Hyperactivity Disorder,” Primary Care: Clinics in Office Practice, vol. 48, no. 3, pp. 475–491, Sep. 2021. [CrossRef]
- M. Cichorek, P. Kowiański, G. Lietzau, J. Lasek, and J. Moryś, “Neuroglia — development and role in physiological and pathophysiological processes,” Folia Morphol (Warsz), vol. 80, no. 4, pp. 766–775, Dec. 2021. [CrossRef]
- I. Lago-Baldaia, V. M. Fernandes, and S. D. Ackerman, “More Than Mortar: Glia as Architects of Nervous System Development and Disease,” Front Cell Dev Biol, vol. 8, Dec. 2020. [CrossRef]
- J. Martins-Macedo, A. C. Lepore, H. S. Domingues, A. J. Salgado, E. D. Gomes, and L. Pinto, “Glial restricted precursor cells in central nervous system disorders: Current applications and future perspectives,” Glia, vol. 69, no. 3, pp. 513–531, Mar. 2021. [CrossRef]
- F. Aloisi, “Immune function of microglia,” Glia, vol. 36, no. 2, pp. 165–179, Nov. 2001. [CrossRef]
- M. V. Sofroniew, “Astrocyte barriers to neurotoxic inflammation,” Nat Rev Neurosci, vol. 16, no. 5, pp. 249–263, May 2015. [CrossRef]
- S. Elkabes, E. DiCicco-Bloom, and I. Black, “Brain microglia/macrophages express neurotrophins that selectively regulate microglial proliferation and function,” The Journal of Neuroscience, vol. 16, no. 8, pp. 2508–2521, Apr. 1996. [CrossRef]
- K. Nakajima, S. Honda, Y. Tohyama, Y. Imai, S. Kohsaka, and T. Kurihara, “Neurotrophin secretion from cultured microglia,” J Neurosci Res, vol. 65, no. 4, pp. 322–331, Aug. 2001. [CrossRef]
- M. Greter and M. Merad, “Regulation of microglia development and homeostasis,” Glia, vol. 61, no. 1, pp. 121–127, Jan. 2013. [CrossRef]
- M. M. Eftekharian et al., “Cytokine profile in autistic patients,” Cytokine, vol. 108, pp. 120–126, Aug. 2018. [CrossRef]
- S. L. Patterson, “Immune dysregulation and cognitive vulnerability in the aging brain: Interactions of microglia, IL-1β, BDNF and synaptic plasticity,” Neuropharmacology, vol. 96, pp. 11–18, Sep. 2015. [CrossRef]
- P. Ashwood, P. Krakowiak, I. Hertz-Picciotto, R. Hansen, I. Pessah, and J. Van de Water, “Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome,” Brain Behav Immun, vol. 25, no. 1, pp. 40–45, Jan. 2011. [CrossRef]
- B. S. Khakh and M. V Sofroniew, “Diversity of astrocyte functions and phenotypes in neural circuits,” Nat Neurosci, vol. 18, no. 7, pp. 942–952, Jul. 2015. [CrossRef]
- L. C. Dury, C. M. Yde Ohki, K.-P. Lesch, S. Walitza, and E. Grünblatt, “The role of astrocytes in attention-deficit hyperactivity disorder: An update,” Psychiatry Res, vol. 350, p. 116558, Aug. 2025. [CrossRef]
- G. Séjourné and C. Eroglu, “Astrocyte-neuron crosstalk in neurodevelopmental disorders,” Curr Opin Neurobiol, vol. 89, p. 102925, Dec. 2024. [CrossRef]
- S. Dejardin et al., “PET study of human voluntary saccadic eye movements in darkness: effect of task repetition on the activation pattern,” European Journal of Neuroscience, vol. 10, no. 7, pp. 2328–2336, Jul. 1998. [CrossRef]
- S. , Y. B. E. , B. K. , O. S. J. , P. H. , S. H. S. , A. G. J. , L. C. J. Lee, “Channel-mediated tonic GABA release from glia,” Science (1979), pp. 790–796, 2010.
- R. D. and B. K. N. Todd, “Is Attention-Deficit/Hyperactivity Disorder an Energy Deficiency Syndrome?,” Biol Psychiatry, vol. 50, no. 4, pp. 151–158, 2001.
- U. S. Sandau, Z. Alderman, G. Corfas, S. R. Ojeda, and J. Raber, “Astrocyte-Specific Disruption of SynCAM1 Signaling Results in ADHD-Like Behavioral Manifestations,” PLoS One, vol. 7, no. 4, p. e36424, Apr. 2012. [CrossRef]
- P. J. Espenshade, “SREBPs: sterol-regulated transcription factors,” J Cell Sci, vol. 119, no. 6, pp. 973–976, Mar. 2006. [CrossRef]
- H. Won et al., “GIT1 is associated with ADHD in humans and ADHD-like behaviors in mice,” Nat Med, vol. 17, no. 5, pp. 566–572, May 2011. [CrossRef]
- M. R. DeLong and T. Wichmann, “Circuits and Circuit Disorders of the Basal Ganglia,” Arch Neurol, vol. 64, no. 1, p. 20, Jan. 2007. [CrossRef]
- J. Nagai et al., “Hyperactivity with Disrupted Attention by Activation of an Astrocyte Synaptogenic Cue,” Cell, vol. 177, no. 5, pp. 1280-1292.e20, May 2019. [CrossRef]
- L. C. Dury, C. M. Yde Ohki, K.-P. Lesch, S. Walitza, and E. Grünblatt, “The role of astrocytes in attention-deficit hyperactivity disorder: An update,” Psychiatry Res, vol. 350, p. 116558, Aug. 2025. [CrossRef]
- K. W. Lee, S. Lim, and K. D. Kim, “The Function of N-Myc Downstream-Regulated Gene 2 (NDRG2) as a Negative Regulator in Tumor Cell Metastasis,” Int J Mol Sci, vol. 23, no. 16, p. 9365, Aug. 2022. [CrossRef]
- R. Vegt et al., “Genome-wide linkage analysis in a Dutch multigenerational family with attention deficit hyperactivity disorder,” European Journal of Human Genetics, vol. 18, no. 2, pp. 206–211, Feb. 2010. [CrossRef]
- T. Takeichi et al., “The effect of Ndrg2 expression on astroglial activation,” Neurochem Int, vol. 59, no. 1, pp. 21–27, Aug. 2011. [CrossRef]
- A. Yin et al., “NDRG2 Protects the Brain from Excitotoxicity by Facilitating Interstitial Glutamate Uptake,” Transl Stroke Res, vol. 11, no. 2, pp. 214–227, Apr. 2020. [CrossRef]
- L. Shen et al., “Immunohistochemical detection of Ndrg2 in the mouse nervous system,” Neuroreport, vol. 19, no. 9, pp. 927–931, Jun. 2008. [CrossRef]
- Y. Li et al., “Deficiency of tumor suppressor NDRG2 leads to attention deficit and hyperactive behavior,” Journal of Clinical Investigation, vol. 127, no. 12, pp. 4270–4284, Oct. 2017. [CrossRef]
- Y. Li et al., “Deficiency of tumor suppressor NDRG2 leads to attention deficit and hyperactive behavior,” Journal of Clinical Investigation, vol. 127, no. 12, pp. 4270–4284, Oct. 2017. [CrossRef]
- C. Cavaliere et al., “Methylphenidate administration determines enduring changes in neuroglial network in rats,” European Neuropsychopharmacology, vol. 22, no. 1, pp. 53–63, Jan. 2012. [CrossRef]
- A. D. Bacchi, “Beyond the Neuron: The Integrated Role of Glia in Psychiatric Disorders,” Neuroglia, vol. 6, no. 2, p. 15, Mar. 2025. [CrossRef]
- B. R. Ransom, A. M. Butt, and J. A. Black, “Ultrastructural identification of HRP-injected oligodendrocytes in the intact rat optic nerve,” Glia, vol. 4, no. 1, pp. 37–45, Jan. 1991. [CrossRef]
- A. M. Butt, R. F. Fern, and C. Matute, “Neurotransmitter signaling in white matter,” Glia, vol. 62, no. 11, pp. 1762–1779, Nov. 2014. [CrossRef]
- A. M. Butt and A. Kalsi, “Inwardly rectifying potassium channels (Kir) in central nervous system glia: a special role for Kir4.1 in glial functions,” J Cell Mol Med, vol. 10, no. 1, pp. 33–44, Jan. 2006. [CrossRef]
- A. D. Rivera, I. Chacon-De-La-Rocha, F. Pieropan, M. Papanikolau, K. Azim, and A. M. Butt, “Keeping the ageing brain wired: a role for purine signalling in regulating cellular metabolism in oligodendrocyte progenitors,” Pflugers Arch, vol. 473, no. 5, pp. 775–783, May 2021. [CrossRef]
- M. Ribasés et al., “Association Study of 10 Genes Encoding Neurotrophic Factors and Their Receptors in Adult and Child Attention-Deficit/Hyperactivity Disorder,” Biol Psychiatry, vol. 63, no. 10, pp. 935–945, May 2008. [CrossRef]
- S. Pasquin, M. Sharma, and J.-F. Gauchat, “Ciliary neurotrophic factor (CNTF): New facets of an old molecule for treating neurodegenerative and metabolic syndrome pathologies,” Cytokine Growth Factor Rev, vol. 26, no. 5, pp. 507–515, Oct. 2015. [CrossRef]
- D. Anand, G. D. Colpo, G. Zeni, C. P. Zeni, and A. L. Teixeira, “Attention-Deficit/Hyperactivity Disorder And Inflammation: What Does Current Knowledge Tell Us? A Systematic Review,” Front Psychiatry, vol. 8, Nov. 2017. [CrossRef]
- K. Lesch, “Editorial: Can dysregulated myelination be linked to <scp>ADHD</scp> pathogenesis and persistence?,” Journal of Child Psychology and Psychiatry, vol. 60, no. 3, pp. 229–231, Mar. 2019. [CrossRef]
- D. Demontis et al., “Discovery of the first genome-wide significant risk loci for attention deficit/hyperactivity disorder,” Nat Genet, vol. 51, no. 1, pp. 63–75, Jan. 2019. [CrossRef]
- S. Yoo et al., “Sialylation regulates brain structure and function,” The FASEB Journal, vol. 29, no. 7, pp. 3040–3053, Jul. 2015. [CrossRef]
- I. Lago-Baldaia, V. M. Fernandes, and S. D. Ackerman, “More Than Mortar: Glia as Architects of Nervous System Development and Disease,” Front Cell Dev Biol, vol. 8, Dec. 2020. [CrossRef]
- K. Sharma, K. Bisht, and U. B. Eyo, “A Comparative Biology of Microglia Across Species,” Front Cell Dev Biol, vol. 9, Apr. 2021. [CrossRef]
- L. J. Lawson, V. H. Perry, P. Dri, and S. Gordon, “Heterogeneity in the distribution and morphology of microglia in the normal adult mouse brain,” Neuroscience, vol. 39, no. 1, pp. 151–170, Jan. 1990. [CrossRef]
- D. P. Pelvig, H. Pakkenberg, A. K. Stark, and B. Pakkenberg, “Neocortical glial cell numbers in human brains,” Neurobiol Aging, vol. 29, no. 11, pp. 1754–1762, Nov. 2008. [CrossRef]
- V. A. K. Rathinam and K. A. Fitzgerald, “Inflammasome Complexes: Emerging Mechanisms and Effector Functions,” Cell, vol. 165, no. 4, pp. 792–800, May 2016. [CrossRef]
- N. Kayagaki et al., “Caspase-11 cleaves gasdermin D for non-canonical inflammasome signalling,” Nature, vol. 526, no. 7575, pp. 666–671, Oct. 2015. [CrossRef]
- J. Shi et al., “Cleavage of GSDMD by inflammatory caspases determines pyroptotic cell death,” Nature, vol. 526, no. 7575, pp. 660–665, Oct. 2015. [CrossRef]
- J. N. Crawley, “Mouse Behavioral Assays Relevant to the Symptoms of Autism*,” Brain Pathology, vol. 17, no. 4, pp. 448–459, Oct. 2007. [CrossRef]
- D. Daley, “Attention deficit hyperactivity disorder: a review of the essential facts,” Child Care Health Dev, vol. 32, no. 2, pp. 193–204, Mar. 2006. [CrossRef]
- I. Rauch et al., “NAIP-NLRC4 Inflammasomes Coordinate Intestinal Epithelial Cell Expulsion with Eicosanoid and IL-18 Release via Activation of Caspase-1 and -8,” Immunity, vol. 46, no. 4, pp. 649–659, Apr. 2017. [CrossRef]
- Y. Wolf, S. Yona, K.-W. Kim, and S. Jung, “Microglia, seen from the CX3CR1 angle,” Front Cell Neurosci, vol. 7, 2013. [CrossRef]
- M. Heiman et al., “A Translational Profiling Approach for the Molecular Characterization of CNS Cell Types,” Cell, vol. 135, no. 4, pp. 738–748, Nov. 2008. [CrossRef]
- H.-C. Chuang et al., “Defective cell death of distinct microglial subsets contributes to ADHD-like behavior in mice,” Aug. 30, 2019. [CrossRef]
- C. L. Murray, P. Obiang, D. Bannerman, and C. Cunningham, “Endogenous IL-1 in Cognitive Function and Anxiety: A Study in IL-1RI−/− Mice,” PLoS One, vol. 8, no. 10, p. e78385, Oct. 2013. [CrossRef]
- D. Anand, G. D. Colpo, G. Zeni, C. P. Zeni, and A. L. Teixeira, “Attention-Deficit/Hyperactivity Disorder And Inflammation: What Does Current Knowledge Tell Us? A Systematic Review,” Front Psychiatry, vol. 8, Nov. 2017. [CrossRef]
- D. T. Leffa, I. L. S. Torres, and L. A. Rohde, “A Review on the Role of Inflammation in Attention-Deficit/Hyperactivity Disorder,” Neuroimmunomodulation, vol. 25, no. 5–6, pp. 328–333, 2018. [CrossRef]
- U. S. Sandau, Z. Alderman, G. Corfas, S. R. Ojeda, and J. Raber, “Astrocyte-Specific Disruption of SynCAM1 Signaling Results in ADHD-Like Behavioral Manifestations,” PLoS One, vol. 7, no. 4, p. e36424, Apr. 2012. [CrossRef]
- N. Ferencova et al., “Peripheral Inflammatory Markers in Autism Spectrum Disorder and Attention Deficit/Hyperactivity Disorder at Adolescent Age,” Int J Mol Sci, vol. 24, no. 14, p. 11710, Jul. 2023. [CrossRef]
- S. Asadi, M. Jamali, and Z. Gholizadeh, “Assessment of Genetic Mutations in Genes DSM-IV, DRD4, SERT, HTR1B, SNAP25, GRIN2A, ADRA2A, TPH2 and BDNF Induced Attention Deficit Disorder and Hyperactivity in Children,” J Neurol Disord, vol. 04, no. 08, 2016. [CrossRef]
- J. Camberos-Barraza, A. M. Guadrón-Llanos, and A. K. De la Herrán-Arita, “The Gut Microbiome-Neuroglia Axis: Implications for Brain Health, Inflammation, and Disease,” Neuroglia, vol. 5, no. 3, pp. 254–273, Aug. 2024. [CrossRef]
- E. Carias et al., “Chronic oral methylphenidate treatment increases microglial activation in rats,” J Neural Transm, vol. 125, no. 12, pp. 1867–1875, Dec. 2018. [CrossRef]
- S. Sadasivan, B. B. Pond, A. K. Pani, C. Qu, Y. Jiao, and R. J. Smeyne, “Methylphenidate Exposure Induces Dopamine Neuron Loss and Activation of Microglia in the Basal Ganglia of Mice,” PLoS One, vol. 7, no. 3, p. e33693, Mar. 2012. [CrossRef]
- J. McAfoose and B. T. Baune, “Evidence for a cytokine model of cognitive function,” Neurosci Biobehav Rev, vol. 33, no. 3, pp. 355–366, Mar. 2009. [CrossRef]
- L. Brydon, N. A. Harrison, C. Walker, A. Steptoe, and H. D. Critchley, “Peripheral Inflammation is Associated with Altered Substantia Nigra Activity and Psychomotor Slowing in Humans,” Biol Psychiatry, vol. 63, no. 11, pp. 1022–1029, Jun. 2008. [CrossRef]
- P. Buderath et al., “Postural and gait performance in children with attention deficit/hyperactivity disorder,” Gait Posture, vol. 29, no. 2, pp. 249–254, Feb. 2009. [CrossRef]
- C. Ganos, A. Münchau, and K. P. Bhatia, “The Semiology of Tics, Tourette’s, and Their Associations,” Mov Disord Clin Pract, vol. 1, no. 3, pp. 145–153, Sep. 2014. [CrossRef]
- K. Ueda, S. Kim, D. J. Greene, and K. J. Black, “Correlates and Clinical Implications of Tic Suppressibility,” Curr Dev Disord Rep, vol. 8, no. 2, pp. 112–120, Jun. 2021. [CrossRef]
- Movement Disorders in Childhood. Elsevier, 2016. [CrossRef]
- C. Ganos, J. Bongert, L. Asmuss, D. Martino, P. Haggard, and A. Münchau, “The somatotopy of tic inhibition: Where and how much?,” Movement Disorders, vol. 30, no. 9, pp. 1184–1189, Aug. 2015. [CrossRef]
- J. Jankovic, “Tourette’s Syndrome,” New England Journal of Medicine, vol. 345, no. 16, pp. 1184–1192, Oct. 2001. [CrossRef]
- J. Jankovic, “PHENOMENOLOGY AND CLASSIFICATION OF TICS,” Neurol Clin, vol. 15, no. 2, pp. 267–275, May 1997. [CrossRef]
- J. Kaczyńska and P. Janik, “Tonic Tics in Gilles de la Tourette Syndrome,” Neuropediatrics, vol. 52, no. 05, pp. 370–376, Oct. 2021. [CrossRef]
- J. Jankovic and R. Kurlan, “Tourette syndrome: Evolving concepts,” Movement Disorders, vol. 26, no. 6, pp. 1149–1156, May 2011. [CrossRef]
- American Psychiatric Association, Diagnostic and Statistical Manual of Mental Disorders. American Psychiatric Association, 2013. [CrossRef]
- K. R. Müller-Vahl, T. Sambrani, and E. Jakubovski, “Tic disorders revisited: introduction of the term ‘tic spectrum disorders,’” Eur Child Adolesc Psychiatry, vol. 28, no. 8, pp. 1129–1135, Aug. 2019. [CrossRef]
- V. Roessner, P. J. Hoekstra, and A. Rothenberger, “Tourette’s disorder and other tic disorders in DSM-5: a comment,” Eur Child Adolesc Psychiatry, vol. 20, no. 2, pp. 71–74, Feb. 2011. [CrossRef]
- K. J. Black, E. R. Black, D. J. Greene, and B. L. Schlaggar, “Provisional Tic Disorder: What to tell parents when their child first starts ticcing,” F1000Res, vol. 5, p. 696, Apr. 2016. [CrossRef]
- V. Eapen, A. E. Cavanna, and M. M. Robertson, “Comorbidities, Social Impact, and Quality of Life in Tourette Syndrome,” Front Psychiatry, vol. 7, Jun. 2016. [CrossRef]
- E. Cubo, “Review of Prevalence Studies of Tic Disorders: Methodological Caveats,” Tremor and Other Hyperkinetic Movements, vol. 2, no. 0, p. 02, May 2012. [CrossRef]
- T. Knight, T. Steeves, L. Day, M. Lowerison, N. Jette, and T. Pringsheim, “Prevalence of Tic Disorders: A Systematic Review and Meta-Analysis,” Pediatr Neurol, vol. 47, no. 2, pp. 77–90, Aug. 2012. [CrossRef]
- E. Cubo et al., “Prevalence of Tics in Schoolchildren in Central Spain: A Population-Based Study,” Pediatr Neurol, vol. 45, no. 2, pp. 100–108, Aug. 2011. [CrossRef]
- R. Canitano and G. Vivanti, “Tics and Tourette syndrome in autism spectrum disorders,” Autism, vol. 11, no. 1, pp. 19–28, Jan. 2007. [CrossRef]
- C. Ganos, “Tics and Tourette’s: update on pathophysiology and tic control,” Curr Opin Neurol, vol. 29, no. 4, pp. 513–518, Aug. 2016. [CrossRef]
- M. H. Bloch, J. F. Leckman, H. Zhu, and B. S. Peterson, “Caudate volumes in childhood predict symptom severity in adults with Tourette syndrome,” Neurology, vol. 65, no. 8, pp. 1253–1258, Oct. 2005. [CrossRef]
- D. J. Greene, B. L. Schlaggar, and K. J. Black, “Neuroimaging in Tourette Syndrome: Research Highlights from 2014 to 2015,” Curr Dev Disord Rep, vol. 2, no. 4, pp. 300–308, Dec. 2015. [CrossRef]
- S. Kim et al., “Hippocampal Volume in Provisional Tic Disorder Predicts Tic Severity at 12-Month Follow-up,” J Clin Med, vol. 9, no. 6, p. 1715, Jun. 2020. [CrossRef]
- H. P. Sigurdsson, S. R. Jackson, L. Jolley, E. Mitchell, and G. M. Jackson, “Alterations in cerebellar grey matter structure and covariance networks in young people with Tourette syndrome,” Cortex, vol. 126, pp. 1–15, May 2020. [CrossRef]
- M. I. Makki, R. Munian Govindan, B. J. Wilson, M. E. Behen, and H. T. Chugani, “Altered Fronto-Striato-Thalamic Connectivity in Children with Tourette Syndrome Assessed with Diffusion Tensor MRI and Probabilistic Fiber Tracking,” J Child Neurol, vol. 24, no. 6, pp. 669–678, Jun. 2009. [CrossRef]
- J. Muellner et al., “Altered structure of cortical sulci in gilles de la Tourette syndrome: Further support for abnormal brain development,” Movement Disorders, vol. 30, no. 5, pp. 655–661, Apr. 2015. [CrossRef]
- B. Cheng et al., “Altered intrahemispheric structural connectivity in Gilles de la Tourette syndrome,” Neuroimage Clin, vol. 4, pp. 174–181, 2014. [CrossRef]
- S. Ramkiran, L. Heidemeyer, A. Gaebler, N. J. Shah, and I. Neuner, “Alterations in basal ganglia-cerebello-thalamo-cortical connectivity and whole brain functional network topology in Tourette’s syndrome,” Neuroimage Clin, vol. 24, p. 101998, 2019. [CrossRef]
- J. F. Abelson et al., “Sequence Variants in SLITRK1 Are Associated with Tourette’s Syndrome,” Science (1979), vol. 310, no. 5746, pp. 317–320, Oct. 2005. [CrossRef]
- A. A. Stillman et al., “Developmentally regulated and evolutionarily conserved expression of SLITRK1 in brain circuits implicated in Tourette syndrome,” Journal of Comparative Neurology, vol. 513, no. 1, pp. 21–37, Mar. 2009. [CrossRef]
- D. Yael, E. Vinner, and I. Bar-Gad, “Pathophysiology of tic disorders,” Movement Disorders, vol. 30, no. 9, pp. 1171–1178, Aug. 2015. [CrossRef]
- C. L. Baym, B. A. Corbett, S. B. Wright, and S. A. Bunge, “Neural correlates of tic severity and cognitive control in children with Tourette syndrome,” Brain, vol. 131, no. 1, pp. 165–179, Jan. 2008. [CrossRef]
- Z. Wang, T. V. Maia, R. Marsh, T. Colibazzi, A. Gerber, and B. S. Peterson, “The Neural Circuits That Generate Tics in Tourette’s Syndrome,” American Journal of Psychiatry, vol. 168, no. 12, pp. 1326–1337, Dec. 2011. [CrossRef]
- K. W. McCairn, A. Iriki, and M. Isoda, “Global Dysrhythmia of Cerebro-Basal Ganglia–Cerebellar Networks Underlies Motor Tics following Striatal Disinhibition,” The Journal of Neuroscience, vol. 33, no. 2, pp. 697–708, Jan. 2013. [CrossRef]
- R. H. Tobe et al., “Cerebellar morphology in Tourette syndrome and obsessive-compulsive disorder,” Ann Neurol, vol. 67, no. 4, pp. 479–487, Apr. 2010. [CrossRef]
- A. Lerner et al., “Neuroimaging of neuronal circuits involved in tic generation in patients with Tourette syndrome,” Neurology, vol. 68, no. 23, pp. 1979–1987, Jun. 2007. [CrossRef]
- P. Zhuang, M. Hallett, X. Zhang, J. Li, Y. Zhang, and Y. Li, “Neuronal activity in the globus pallidus internus in patients with tics,” J Neurol Neurosurg Psychiatry, vol. 80, no. 10, pp. 1075–1081, Oct. 2009. [CrossRef]
- I. Neuner et al., “Imaging the where and when of tic generation and resting state networks in adult Tourette patients,” Front Hum Neurosci, vol. 8, May 2014. [CrossRef]
- E. Stern et al., “A Functional Neuroanatomy of Tics in Tourette Syndrome,” Arch Gen Psychiatry, vol. 57, no. 8, p. 741, Aug. 2000. [CrossRef]
- A. E. Cavanna, K. J. Black, M. Hallett, and V. Voon, “Neurobiology of the Premonitory Urge in Tourette’s Syndrome: Pathophysiology and Treatment Implications,” J Neuropsychiatry Clin Neurosci, vol. 29, no. 2, pp. 95–104, Apr. 2017. [CrossRef]
- R. Morgenstern, T. Mende, R. Gold, P. Lemme, and W. Oelssner, “Drug-induced modulation of locomotor hyperactivity induced by picrotoxin in nucleus accumbens,” Pharmacol Biochem Behav, vol. 21, no. 4, pp. 501–506, Oct. 1984. [CrossRef]
- D. L. Jones, G. J. Mogenson, and M. Wu, “Injections of dopaminergic, cholinergic, serotoninergic and gabaergic drugs into the nucleus accumbens: effects on locomotor activity in the rat,” Neuropharmacology, vol. 20, no. 1, pp. 29–37, Jan. 1981. [CrossRef]
- “Quantitative morphology of the caudate nucleus in attention deficit hyperactivity disorder,” American Journal of Psychiatry, vol. 151, no. 12, pp. 1791–1796, Dec. 1994. [CrossRef]
- B. S. Peterson et al., “A Functional Magnetic Resonance Imaging Study of Tic Suppression in Tourette Syndrome,” Arch Gen Psychiatry, vol. 55, no. 4, Apr. 1998. [CrossRef]
- A. Draper et al., “Increased GABA Contributes to Enhanced Control over Motor Excitability in Tourette Syndrome,” Current Biology, vol. 24, no. 19, pp. 2343–2347, Oct. 2014. [CrossRef]
- J. F. LECKMAN et al., “Perinatal Factors in the Expression of Tourette’s Syndrome: An Exploratory Study,” J Am Acad Child Adolesc Psychiatry, vol. 29, no. 2, pp. 220–226, Mar. 1990. [CrossRef]
- L. Burd, R. Severud, M. G. Klug, and J. Kerbeshian, “Prenatal and perinatal risk factors for Tourette disorder,” J Perinat Med, vol. 27, no. 4, Jan. 1999. [CrossRef]
- T.-K. Chao, J. Hu, and T. Pringsheim, “Prenatal risk factors for Tourette Syndrome: a systematic review,” BMC Pregnancy Childbirth, vol. 14, no. 1, p. 53, Dec. 2014. [CrossRef]
- E. Ayubi, K. Mansori, and A. Doosti-Irani, “Effect of maternal smoking during pregnancy on Tourette syndrome and chronic tic disorders among offspring: a systematic review and meta-analysis,” Obstet Gynecol Sci, vol. 64, no. 1, pp. 1–12, Jan. 2021. [CrossRef]
- C. A. Mathews, J. M. Scharf, L. L. Miller, C. Macdonald-Wallis, D. A. Lawlor, and Y. Ben-Shlomo, “Association between pre- and perinatal exposures and Tourette syndrome or chronic tic disorder in the ALSPAC cohort,” British Journal of Psychiatry, vol. 204, no. 1, pp. 40–45, Jan. 2014. [CrossRef]
- S. Leivonen et al., “Parental Psychopathology and Tourette Syndrome/Chronic Tic Disorder in Offspring: A Nationwide Case-Control Study,” J Am Acad Child Adolesc Psychiatry, vol. 56, no. 4, pp. 297-303.e4, Apr. 2017. [CrossRef]
- P. Zhu, M. Wu, P. Huang, X. Zhao, and X. Ji, “Children from nuclear families with bad parental relationship could develop tic symptoms,” Mol Genet Genomic Med, vol. 8, no. 7, Jul. 2020. [CrossRef]
- D. Mataix-Cols et al., “Familial Risks of Tourette Syndrome and Chronic Tic Disorders,” JAMA Psychiatry, vol. 72, no. 8, p. 787, Aug. 2015. [CrossRef]
- M. Georgitsi, A. J. Willsey, C. A. Mathews, M. State, J. M. Scharf, and P. Paschou, “The Genetic Etiology of Tourette Syndrome: Large-Scale Collaborative Efforts on the Precipice of Discovery,” Front Neurosci, vol. 10, Aug. 2016. [CrossRef]
- A. J. M. H. Verkerk, C. A. Mathews, M. Joosse, B. H. J. Eussen, P. Heutink, and B. A. Oostra, “Cntnap2 is disrupted in a family with gilles de la tourette syndrome and obsessive compulsive disorder,” Genomics, vol. 82, no. 1, pp. 1–9, Jul. 2003. [CrossRef]
- A. Lawson-Yuen, J.-S. Saldivar, S. Sommer, and J. Picker, “Familial deletion within NLGN4 associated with autism and Tourette syndrome,” European Journal of Human Genetics, vol. 16, no. 5, pp. 614–618, May 2008. [CrossRef]
- A. G. Ercan-Sencicek et al., “L-Histidine Decarboxylase and Tourette’s Syndrome,” New England Journal of Medicine, vol. 362, no. 20, pp. 1901–1908, May 2010. [CrossRef]
- C. Patel, L. Cooper-Charles, D. J. McMullan, J. M. Walker, V. Davison, and J. Morton, “Translocation breakpoint at 7q31 associated with tics: further evidence for IMMP2L as a candidate gene for Tourette syndrome,” European Journal of Human Genetics, vol. 19, no. 6, pp. 634–639, Jun. 2011. [CrossRef]
- E. Petek et al., “Disruption of a Novel Gene (IMMP2L) by a Breakpoint in 7q31 Associated with Tourette Syndrome,” The American Journal of Human Genetics, vol. 68, no. 4, pp. 848–858, Apr. 2001. [CrossRef]
- S. K. George, Y. Jiao, C. E. Bishop, and B. Lu, “Mitochondrial peptidase IMMP2L mutation causes early onset of age-associated disorders and impairs adult stem cell self-renewal,” Aging Cell, vol. 10, no. 4, pp. 584–594, Aug. 2011. [CrossRef]
- V. A. Bjerregaard, B. Schönewolf-Greulich, L. Juel Rasmussen, C. Desler, and Z. Tümer, “Mitochondrial Function in Gilles de la Tourette Syndrome Patients With and Without Intragenic IMMP2L Deletions,” Front Neurol, vol. 11, Mar. 2020. [CrossRef]
- S. K. Sundaram, A. M. Huq, B. J. Wilson, and H. T. Chugani, “Tourette syndrome is associated with recurrent exonic copy number variants,” Neurology, vol. 74, no. 20, pp. 1583–1590, May 2010. [CrossRef]
- A. Y. Huang et al., “Rare Copy Number Variants in NRXN1 and CNTN6 Increase Risk for Tourette Syndrome,” Neuron, vol. 94, no. 6, pp. 1101-1111.e7, Jun. 2017. [CrossRef]
- S. Wang et al., “De Novo Sequence and Copy Number Variants Are Strongly Associated with Tourette Disorder and Implicate Cell Polarity in Pathogenesis,” Cell Rep, vol. 24, no. 13, pp. 3441-3454.e12, Sep. 2018. [CrossRef]
- S. Liu et al., “Mutations in ASH1L confer susceptibility to Tourette syndrome,” Mol Psychiatry, vol. 25, no. 2, pp. 476–490, Feb. 2020. [CrossRef]
- C. Depienne et al., “Association of Rare Genetic Variants in Opioid Receptors with Tourette Syndrome,” Tremor and Other Hyperkinetic Movements, vol. 9, no. 0, Nov. 2019. [CrossRef]
- J. M. Scharf et al., “Genome-wide association study of Tourette’s syndrome,” Mol Psychiatry, vol. 18, no. 6, pp. 721–728, Jun. 2013. [CrossRef]
- S. Liu et al., “Support of positive association in family-based genetic analysis between COL27A1 and Tourette syndrome,” Sci Rep, vol. 5, no. 1, p. 12687, Aug. 2015. [CrossRef]
- J. F. Leckman et al., “Increased serum levels of interleukin-12 and tumor necrosis factor-alpha in Tourette’s syndrome,” Biol Psychiatry, vol. 57, no. 6, pp. 667–673, Mar. 2005. [CrossRef]
- I. Kawikova et al., “Decreased Numbers of Regulatory T Cells Suggest Impaired Immune Tolerance in Children with Tourette Syndrome: A Preliminary Study,” Biol Psychiatry, vol. 61, no. 3, pp. 273–278, Feb. 2007. [CrossRef]
- D. Martino et al., “Soluble adhesion molecules in Gilles de la Tourette’s syndrome,” J Neurol Sci, vol. 234, no. 1–2, pp. 79–85, Jul. 2005. [CrossRef]
- N. G. P. Bos-Veneman et al., “Altered immunoglobulin profiles in children with Tourette syndrome,” Brain Behav Immun, vol. 25, no. 3, pp. 532–538, Mar. 2011. [CrossRef]
- N. Szejko, C. Fremer, K.-W. Sühs, P. Macul Ferreira de Barros, and K. R. Müller-Vahl, “Intravenous Immunoglobulin Treatment Did Not Improve Tics in a Patient With Gilles de la Tourette Syndrome and Intrathecal Antibody Synthesis,” Front Neurol, vol. 11, Feb. 2020. [CrossRef]
- C. Baumgaertel et al., “Immunity in Gilles de la Tourette-Syndrome: Results From a Cerebrospinal Fluid Study,” Front Neurol, vol. 10, Jul. 2019. [CrossRef]
- C. Wenzel, U. Wurster, and K. R. Müller-Vahl, “Oligoclonal bands in cerebrospinal fluid in patients with Tourette’s syndrome,” Movement Disorders, vol. 26, no. 2, pp. 343–346, Feb. 2011. [CrossRef]
- D. Martino, P. Zis, and M. Buttiglione, “The role of immune mechanisms in Tourette syndrome,” Brain Res, vol. 1617, pp. 126–143, Aug. 2015. [CrossRef]
- S. Herculano-Houzel, “The glia/neuron ratio: How it varies uniformly across brain structures and species and what that means for brain physiology and evolution,” Glia, vol. 62, no. 9, pp. 1377–1391, Sep. 2014. [CrossRef]
- M. Sun et al., “Microglia–Astrocyte Interaction in Neural Development and Neural Pathogenesis,” Cells, vol. 12, no. 15, p. 1942, Jul. 2023. [CrossRef]
- K. Saijo and C. K. Glass, “Microglial cell origin and phenotypes in health and disease,” Nat Rev Immunol, vol. 11, no. 11, pp. 775–787, Nov. 2011. [CrossRef]
- A. Verkhratsky and M. Nedergaard, “Physiology of Astroglia,” Physiol Rev, vol. 98, no. 1, pp. 239–389, Jan. 2018. [CrossRef]
- S. G. Torres-Platas et al., “Morphometric characterization of microglial phenotypes in human cerebral cortex,” J Neuroinflammation, vol. 11, no. 1, p. 12, Dec. 2014. [CrossRef]
- R. Kongsui, S. B. Beynon, S. J. Johnson, and F. R. Walker, “Quantitative assessment of microglial morphology and density reveals remarkable consistency in the distribution and morphology of cells within the healthy prefrontal cortex of the rat,” J Neuroinflammation, vol. 11, no. 1, p. 182, Dec. 2014. [CrossRef]
- M. Y. Batiuk et al., “Identification of region-specific astrocyte subtypes at single cell resolution,” Nat Commun, vol. 11, no. 1, p. 1220, Mar. 2020. [CrossRef]
- S. L. Forrest et al., “Distribution Patterns of Astrocyte Populations in the Human Cortex,” Neurochem Res, vol. 48, no. 4, pp. 1222–1232, Apr. 2023. [CrossRef]
- B. Zhou, Y. Zuo, and R. Jiang, “Astrocyte morphology: Diversity, plasticity, and role in neurological diseases,” CNS Neurosci Ther, vol. 25, no. 6, pp. 665–673, Jun. 2019. [CrossRef]
- M. Fakhoury, “Microglia and Astrocytes in Alzheimer’s Disease: Implications for Therapy,” Curr Neuropharmacol, vol. 16, no. 5, pp. 508–518, May 2018. [CrossRef]
- H. S. Kwon and S.-H. Koh, “Neuroinflammation in neurodegenerative disorders: the roles of microglia and astrocytes,” Transl Neurodegener, vol. 9, no. 1, p. 42, Dec. 2020. [CrossRef]
- W. Liu, Y. Tang, and J. Feng, “Cross talk between activation of microglia and astrocytes in pathological conditions in the central nervous system,” Life Sci, vol. 89, no. 5–6, pp. 141–146, Aug. 2011. [CrossRef]
- J. Frey and I. A. Malaty, “Tourette Syndrome Treatment Updates: a Review and Discussion of the Current and Upcoming Literature,” Curr Neurol Neurosci Rep, vol. 22, no. 2, pp. 123–142, Feb. 2022. [CrossRef]
- N. Szejko et al., “European clinical guidelines for Tourette syndrome and other tic disorders—version 2.0. Part I: assessment,” Eur Child Adolesc Psychiatry, vol. 31, no. 3, pp. 383–402, Mar. 2022. [CrossRef]
- C.-Y. Chou, J. Agin-Liebes, and S.-H. Kuo, “Emerging therapies and recent advances for Tourette syndrome,” Heliyon, vol. 9, no. 1, p. e12874, Jan. 2023. [CrossRef]
- B. S. PETERSON, D. S. PINE, P. COHEN, and J. S. BROOK, “Prospective, Longitudinal Study of Tic, Obsessive-Compulsive, and Attention-Deficit/Hyperactivity Disorders in an Epidemiological Sample,” J Am Acad Child Adolesc Psychiatry, vol. 40, no. 6, pp. 685–695, Jun. 2001. [CrossRef]
- V. X. Han, S. Patel, H. F. Jones, and R. C. Dale, “Maternal immune activation and neuroinflammation in human neurodevelopmental disorders,” Nat Rev Neurol, vol. 17, no. 9, pp. 564–579, Sep. 2021. [CrossRef]
- Y. Li, X. Wang, H. Yang, Y. Li, J. Gui, and Y. Cui, “Profiles of Proinflammatory Cytokines and T Cells in Patients With Tourette Syndrome: A Meta-Analysis,” Front Immunol, vol. 13, May 2022. [CrossRef]
- S. E. Swedo et al., “Pediatric Autoimmune Neuropsychiatric Disorders Associated With Streptococcal Infections: Clinical Description of the First 50 Cases,” American Journal of Psychiatry, vol. 155, no. 2, pp. 264–271, Feb. 1998. [CrossRef]
- G. Lepri et al., “Clinical-Serological Characterization and Treatment Outcome of a Large Cohort of Italian Children with Pediatric Autoimmune Neuropsychiatric Disorder Associated with Streptococcal Infection and Pediatric Acute Neuropsychiatric Syndrome,” J Child Adolesc Psychopharmacol, vol. 29, no. 8, pp. 608–614, Oct. 2019. [CrossRef]
- X. Wu, J. Hao, K. Jiang, M. Wu, X. Zhao, and X. Zhang, “Neuroinflammation and pathways that contribute to tourette syndrome,” Ital J Pediatr, vol. 51, no. 1, p. 63, Feb. 2025. [CrossRef]
- I. Kawikova et al., “Decreased Numbers of Regulatory T Cells Suggest Impaired Immune Tolerance in Children with Tourette Syndrome: A Preliminary Study,” Biol Psychiatry, vol. 61, no. 3, pp. 273–278, Feb. 2007. [CrossRef]
- E. Li et al., “Streptococcal infection and immune response in children with Tourette’s syndrome,” Child’s Nervous System, vol. 31, no. 7, pp. 1157–1163, Jul. 2015. [CrossRef]
- Y. Li, X. Wang, H. Yang, Y. Li, J. Gui, and Y. Cui, “Profiles of Proinflammatory Cytokines and T Cells in Patients With Tourette Syndrome: A Meta-Analysis,” Front Immunol, vol. 13, May 2022. [CrossRef]
- J. F. Leckman et al., “Increased serum levels of interleukin-12 and tumor necrosis factor-alpha in Tourette’s syndrome,” Biol Psychiatry, vol. 57, no. 6, pp. 667–673, Mar. 2005. [CrossRef]
- V. Gabbay et al., “A cytokine study in children and adolescents with Tourette’s disorder,” Prog Neuropsychopharmacol Biol Psychiatry, vol. 33, no. 6, pp. 967–971, Aug. 2009. [CrossRef]
- X. Ma et al., “Regulation of IL-10 and IL-12 production and function in macrophages and dendritic cells,” F1000Res, vol. 4, p. 1465, Dec. 2015. [CrossRef]
- J. C. Möller et al., “Immunophenotyping in Tourette syndrome – a pilot study,” Eur J Neurol, vol. 15, no. 7, pp. 749–753, Jul. 2008. [CrossRef]
- T. Brunner et al., “Cell-autonomous Fas (CD95)/Fas-ligand interaction mediates activation-induced apoptosis in T-cell hybridomas,” Nature, vol. 373, no. 6513, pp. 441–444, Feb. 1995. [CrossRef]
- E. Weidinger et al., “Impaired activation of the innate immune response to bacterial challenge in Tourette syndrome,” The World Journal of Biological Psychiatry, vol. 15, no. 6, pp. 453–458, Aug. 2014. [CrossRef]
- A. Cauwels et al., “The origin and function of soluble CD14 in experimental bacterial meningitis.,” J Immunol, vol. 162, no. 8, pp. 4762–72, Apr. 1999.
- K. Riazi, M. A. Galic, J. B. Kuzmiski, W. Ho, K. A. Sharkey, and Q. J. Pittman, “Microglial activation and TNFα production mediate altered CNS excitability following peripheral inflammation,” Proceedings of the National Academy of Sciences, vol. 105, no. 44, pp. 17151–17156, Nov. 2008. [CrossRef]
- A. R. Prossin et al., “Cytomegalovirus Antibody Elevation in Bipolar Disorder: Relation to Elevated Mood States,” Neural Plast, vol. 2015, pp. 1–6, 2015. [CrossRef]
- S. Alshammery et al., “Common targetable inflammatory pathways in brain transcriptome of autism spectrum disorders and Tourette syndrome,” Front Neurosci, vol. 16, Dec. 2022. [CrossRef]
- J. B. Lennington et al., “Transcriptome Analysis of the Human Striatum in Tourette Syndrome,” Biol Psychiatry, vol. 79, no. 5, pp. 372–382, Mar. 2016. [CrossRef]
- R. Ferreira et al., “Histamine modulates microglia function,” J Neuroinflammation, vol. 9, no. 1, p. 90, Dec. 2012. [CrossRef]
- L. Frick, M. Rapanelli, E. Abbasi, H. Ohtsu, and C. Pittenger, “Histamine regulation of microglia: Gene-environment interaction in the regulation of central nervous system inflammation,” Brain Behav Immun, vol. 57, pp. 326–337, Oct. 2016. [CrossRef]
- A. G. Ercan-Sencicek et al., “L-Histidine Decarboxylase and Tourette’s Syndrome,” New England Journal of Medicine, vol. 362, no. 20, pp. 1901–1908, May 2010. [CrossRef]
- X. Wang, X. Liu, L. Chen, and X. Zhang, “The inflammatory injury in the striatal microglia-dopaminergic-neuron crosstalk involved in Tourette syndrome development,” Front Immunol, vol. 14, Apr. 2023. [CrossRef]
- S. Guo, H. Wang, and Y. Yin, “Microglia Polarization From M1 to M2 in Neurodegenerative Diseases,” Front Aging Neurosci, vol. 14, Feb. 2022. [CrossRef]
- M. K. Jha, W.-H. Lee, and K. Suk, “Functional polarization of neuroglia: Implications in neuroinflammation and neurological disorders,” Biochem Pharmacol, vol. 103, pp. 1–16, Mar. 2016. [CrossRef]
- Y. Wang, H. Xu, M. Jing, X. Hu, J. Wang, and Y. Hua, “Gut Microbiome Composition Abnormalities Determined Using High-Throughput Sequencing in Children With Tic Disorder,” Front Pediatr, vol. 10, May 2022. [CrossRef]
- Q. Ma, C. Xing, W. Long, H. Y. Wang, Q. Liu, and R.-F. Wang, “Impact of microbiota on central nervous system and neurological diseases: the gut-brain axis,” J Neuroinflammation, vol. 16, no. 1, p. 53, Dec. 2019. [CrossRef]
- D. L. Krause et al., “Infectious agents are associated with psychiatric diseases,” Ment Illn, vol. 4, no. 1, pp. 38–42, Jan. 2012. [CrossRef]
- R. DALE, P. CANDLER, A. CHURCH, R. WAIT, J. POCOCK, and G. GIOVANNONI, “Neuronal surface glycolytic enzymes are autoantigen targets in post-streptococcal autoimmune CNS disease,” J Neuroimmunol, vol. 172, no. 1–2, pp. 187–197, Mar. 2006. [CrossRef]
- K. Blum et al., “Hypothesizing That Pediatric Autoimmune Neuropsychiatric Associated Streptococcal (PANDAS) Causes Rapid Onset of Reward Deficiency Syndrome (RDS) Behaviors and May Require Induction of ‘Dopamine Homeostasis,’” Open J Immunol, vol. 12, no. 03, pp. 65–75, 2022. [CrossRef]
- C. Baumgaertel et al., “Immunity in Gilles de la Tourette-Syndrome: Results From a Cerebrospinal Fluid Study,” Front Neurol, vol. 10, Jul. 2019. [CrossRef]
- L. Hongyan, Z. Mengjiao, W. Chunyan, and H. Yaruo, “Rhynchophyllin attenuates neuroinflammation in Tourette syndrome rats via JAK2/STAT3 and NF-κB pathways,” Environ Toxicol, vol. 34, no. 10, pp. 1114–1120, Oct. 2019. [CrossRef]
- J. Park, K. M. Jang, and K.-K. Park, “Effects of Apamin on MPP+-Induced Calcium Overload and Neurotoxicity by Targeting CaMKII/ERK/p65/STAT3 Signaling Pathways in Dopaminergic Neuronal Cells,” Int J Mol Sci, vol. 23, no. 23, p. 15255, Dec. 2022. [CrossRef]
- D. Bairamian et al., “Microbiota in neuroinflammation and synaptic dysfunction: a focus on Alzheimer’s disease,” Mol Neurodegener, vol. 17, no. 1, p. 19, Dec. 2022. [CrossRef]
- Q. Zhu, H. Zhang, J. Wang, Y. Wu, and X. Chen, “Associations of TNF-α -238G/A, TNF-α -308G/A, and IL-6 -174G/C polymorphisms with the risk of asthma: Evidence from a meta-analysis,” Pediatr Pulmonol, vol. 55, no. 11, pp. 2893–2900, Nov. 2020. [CrossRef]
- J. Wu et al., “Bergaptol Alleviates LPS-Induced Neuroinflammation, Neurological Damage and Cognitive Impairment via Regulating the JAK2/STAT3/p65 Pathway,” J Inflamm Res, vol. Volume 15, pp. 6199–6211, Nov. 2022. [CrossRef]
- Y. Yan et al., “Immunolocalization of antibacterial peptide S100A7 in mastitis goat mammary gland and lipopolysaccharide induces the expression and secretion of S100A7 in goat mammary gland epithelial cells via TLR4/NFκB signal pathway,” Anim Biotechnol, vol. 34, no. 7, pp. 2701–2713, Dec. 2023. [CrossRef]
- F. Tsetsos et al., “Synaptic processes and immune-related pathways implicated in Tourette syndrome,” Transl Psychiatry, vol. 11, no. 1, p. 56, Jan. 2021. [CrossRef]
- J. B. Lennington et al., “Transcriptome Analysis of the Human Striatum in Tourette Syndrome,” Biol Psychiatry, vol. 79, no. 5, pp. 372–382, Mar. 2016. [CrossRef]
- A. Kumar, M. T. Williams, and H. T. Chugani, “Evaluation of Basal Ganglia and Thalamic Inflammation in Children With Pediatric Autoimmune Neuropsychiatric Disorders Associated With Streptococcal Infection and Tourette Syndrome,” J Child Neurol, vol. 30, no. 6, pp. 749–756, May 2015. [CrossRef]
- M. J. Toohey and R. DiGiuseppe, “Defining and measuring irritability: Construct clarification and differentiation.,” Clin Psychol Rev, vol. 53, pp. 93–108, Apr. 2017. [CrossRef]
- A. K. Roy, V. Lopes, and R. G. Klein, “Disruptive mood dysregulation disorder: A new diagnostic approach to chronic irritability in youth,” American Journal of Psychiatry, vol. 171, no. 9, pp. 918–924, Sep. 2014. [CrossRef]
- J. L. Wiggins et al., “Neural correlates of irritability in disruptive mood dysregulation and bipolar disorders,” American Journal of Psychiatry, vol. 173, no. 7, pp. 722–730, Jul. 2016. [CrossRef]
- A. Brænden, M. Coldevin, P. Zeiner, J. Stubberud, and A. Melinder, “Executive function in children with disruptive mood dysregulation disorder compared to attention-deficit/hyperactivity disorder and oppositional defiant disorder, and in children with different irritability levels,” Eur Child Adolesc Psychiatry, vol. 33, no. 1, pp. 115–125, Jan. 2024. [CrossRef]
- E. Bell, P. Boyce, R. J. Porter, R. A. Bryant, and G. S. Malhi, “Irritability in Mood Disorders: Neurobiological Underpinnings and Implications for Pharmacological Intervention,” CNS Drugs, vol. 35, no. 6, pp. 619–641, Jun. 2021. [CrossRef]
- T. P. Beauchaine and J. L. Tackett, “Irritability as a Transdiagnostic Vulnerability Trait:Current Issues and Future Directions,” Behav Ther, vol. 51, no. 2, pp. 350–364, Mar. 2020. [CrossRef]
- I. C. M. Hoogland, C. Houbolt, D. J. van Westerloo, W. A. van Gool, and D. van de Beek, “Systemic inflammation and microglial activation: Systematic review of animal experiments,” J Neuroinflammation, vol. 12, no. 1, Jun. 2015. [CrossRef]
- R. Martins-Ferreira et al., “The Human Microglia Atlas (HuMicA) unravels changes in disease-associated microglia subsets across neurodegenerative conditions,” Nat Commun, vol. 16, no. 1, p. 739, Jan. 2025. [CrossRef]
- D. Gosselin et al., “An environment-dependent transcriptional network specifies human microglia identity,” Science (1979), vol. 356, no. 6344, pp. 1248–1259, Jun. 2017. [CrossRef]
- D. Brites and A. Fernandes, “Neuroinflammation and depression: Microglia activation, extracellular microvesicles and microRNA dysregulation,” Front Cell Neurosci, vol. 9, no. DEC, pp. 1–20, Dec. 2015. [CrossRef]
- H. Wang et al., “Microglia in depression: an overview of microglia in the pathogenesis and treatment of depression,” J Neuroinflammation, vol. 19, no. 1, Dec. 2022. [CrossRef]
- A. M. Klawonn et al., “Microglial activation elicits a negative affective state through prostaglandin-mediated modulation of striatal neurons,” Immunity, vol. 54, no. 2, pp. 225-234.e6, Feb. 2021. [CrossRef]
- L. Frick, M. Rapanelli, E. Abbasi, H. Ohtsu, and C. Pittenger, “Histamine regulation of microglia: Gene-environment interaction in the regulation of central nervous system inflammation,” Brain Behav Immun, vol. 57, pp. 326–337, Oct. 2016. [CrossRef]
- A. Badimon et al., “Negative feedback control of neuronal activity by microglia,” Nature, vol. 586, no. 7829, pp. 417–423, Oct. 2020. [CrossRef]
- A. D. Umpierre and L. J. Wu, “How microglia sense and regulate neuronal activity,” Glia, vol. 69, no. 7, pp. 1637–1653, Jul. 2021. [CrossRef]
- G. Singhal and B. T. Baune, “Microglia: An interface between the loss of neuroplasticity and depression,” Front Cell Neurosci, vol. 11, Sep. 2017. [CrossRef]
- B. Li, W. Yang, T. Ge, Y. Wang, and R. Cui, “Stress induced microglial activation contributes to depression,” Pharmacol Res, vol. 179, May 2022. [CrossRef]
- Y. F. Wang, C. Y. Chen, L. Lei, and Y. Zhang, “Regulation of the microglial polarization for alleviating neuroinflammation in the pathogenesis and therapeutics of major depressive disorder,” Life Sci, vol. 362, Feb. 2025. [CrossRef]
- J. Bin Kang, D. J. Park, M. A. Shah, M. O. Kim, and P. O. Koh, “Lipopolysaccharide induces neuroglia activation and NF-κB activation in cerebral cortex of adult mice,” Lab Anim Res, vol. 35, no. 1, Dec. 2019. [CrossRef]
- L. Zhang et al., “Comparison of chronic restraint stress-and lipopolysaccharide-induced mouse models of depression: Behavior, c-Fos expression, and microglial and astrocytic activation,” Journal of Neurorestoratology, vol. 12, no. 3, Sep. 2024. [CrossRef]
- T. Qiu et al., “Dynamic microglial activation is associated with LPS-induced depressive-like behavior in mice: An [18F] DPA-714 PET imaging study,” Bosn J Basic Med Sci, vol. 22, no. 4, pp. 649–659, Jul. 2022. [CrossRef]
- S. U. Rehman et al., “Ferulic Acid Rescues LPS-Induced Neurotoxicity via Modulation of the TLR4 Receptor in the Mouse Hippocampus,” Mol Neurobiol, vol. 56, no. 4, pp. 2774–2790, Apr. 2019. [CrossRef]
- L. Joshi et al., “Inhibition of autotaxin and lysophosphatidic acid receptor 5 attenuates neuroinflammation in lps-activated bv-2 micro-glia and a mouse endotoxemia model,” Int J Mol Sci, vol. 22, no. 16, Aug. 2021. [CrossRef]
- T. Muhammad, M. Ikram, R. Ullah, S. U. Rehman, and M. O. Kim, “Hesperetin, a citrus flavonoid, attenuates LPS-induced neuroinflammation, apoptosis and memory impairments by modulating TLR4/NF-κB signaling,” Nutrients, vol. 11, no. 3, Mar. 2019. [CrossRef]
- C. Sun et al., “NLRC5 Deficiency Reduces LPS-Induced Microglial Activation via Inhibition of NF-κB Signaling and Ameliorates Mice’s Depressive-like Behavior,” Int J Mol Sci, vol. 24, no. 17, Sep. 2023. [CrossRef]
- L. Zhang et al., “Comparison of chronic restraint stress-and lipopolysaccharide-induced mouse models of depression: Behavior, c-Fos expression, and microglial and astrocytic activation,” Journal of Neurorestoratology, vol. 12, no. 3, Sep. 2024. [CrossRef]
- A. D. Bacchi, “Beyond the Neuron: The Integrated Role of Glia in Psychiatric Disorders,” Neuroglia, vol. 6, no. 2, Jun. 2025. [CrossRef]
- Y. Tizabi, B. Getachew, S. R. Hauser, V. Tsytsarev, A. C. Manhães, and V. D. A. da Silva, “Role of Glial Cells in Neuronal Function, Mood Disorders, and Drug Addiction,” Brain Sci, vol. 14, no. 6, Jun. 2024. [CrossRef]
- M. Albini, A. Krawczun-Rygmaczewska, and F. Cesca, “Astrocytes and brain-derived neurotrophic factor (BDNF),” Neurosci Res, vol. 197, pp. 42–51, Dec. 2023. [CrossRef]
- J. Nagai et al., “Behaviorally consequential astrocytic regulation of neural circuits,” Neuron, vol. 109, no. 4, pp. 576–596, Feb. 2021. [CrossRef]
- J. Goenaga, A. Araque, P. Kofuji, and D. Herrera Moro Chao, “Calcium signaling in astrocytes and gliotransmitter release,” Front Synaptic Neurosci, vol. 15, 2023. [CrossRef]
- S. Guerra-Gomes, N. Sousa, L. Pinto, and J. F. Oliveira, “Functional roles of astrocyte calcium elevations: From synapses to behavior,” Front Cell Neurosci, vol. 11, Jan. 2018. [CrossRef]
- G. Imrie and I. Farhy-Tselnicker, “Astrocyte regulation of behavioral outputs: the versatile roles of calcium,” Front Cell Neurosci, vol. 19, 2025. [CrossRef]
- N. Ahmadpour, M. Kantroo, and J. L. Stobart, “Extracellular calcium influx pathways in astrocyte calcium microdomain physiology,” Biomolecules, vol. 11, no. 10, Oct. 2021. [CrossRef]
- N. Bazargani and D. Attwell, “Astrocyte calcium signaling: The third wave,” Nat Neurosci, vol. 19, no. 2, pp. 182–189, Jan. 2016. [CrossRef]
- Y. Bozzi, “Unraveling white matter alterations in autism: the role of oligodendrocytes, microglia, and neuroinflammation,” Cerebral Cortex, vol. 35, no. 4, Apr. 2025. [CrossRef]
- M. L. Pfau, C. Ménard, and S. J. Russo, “Inflammatory Mediators in Mood Disorders: Therapeutic Opportunities,” Annu Rev Pharmacol Toxicol, vol. 58, pp. 411–428, Jan. 2018. [CrossRef]
- T. Zhao et al., “Folic Acid Attenuates Glial Activation in Neonatal Mice and Improves Adult Mood Disorders Through Epigenetic Regulation,” Front Pharmacol, vol. 13, Feb. 2022. [CrossRef]
- M. Banasr et al., “Glial pathology in an animal model of depression: Reversal of stress-induced cellular, metabolic and behavioral deficits by the glutamate-modulating drug riluzole,” Mol Psychiatry, vol. 15, no. 5, pp. 501–511, May 2010. [CrossRef]
- B. Czéh and B. Di Benedetto, “Antidepressants act directly on astrocytes: Evidences and functional consequences,” European Neuropsychopharmacology, vol. 23, no. 3, pp. 171–185, Mar. 2013. [CrossRef]
- G. Sanacora and M. Banasr, “From pathophysiology to novel antidepressant drugs: Glial contributions to the pathology and treatment of mood disorders,” Biol Psychiatry, vol. 73, no. 12, pp. 1172–1179, Jun. 2013. [CrossRef]
- “Navigating the Complex Terrain of Dysregulated Microglial Function in Depressive Disorders: Insights, Challenges and Future Directions,” Aging Dis, 2024. [CrossRef]
- R. K. Wagner, F. A. Zirps, A. A. Edwards, S. G. Wood, R. E. Joyner, and B. BJ, “The prevalence of dyslexia: A new approach to its estimation,” J Learn Disabil, vol. 53, no. 5, pp. 354–365.
- C. Doust, P. Fontanillas, E. Eising, S. D. Gordon, Z. Wang, and A. G, “Discovery of 42 genome-wide significant loci associated with dyslexia,” Nat Genet, vol. 54, no. 11, pp. 1621–1629.
- F. Richlan, “The functional neuroanatomy of developmental dyslexia across languages and writing systems,” Front Psychol, vol. 11, no. 155.
- M. Luciano, A. J. Gow, A. Pattie, T. C. Bates, and I. J. Deary, “The influence of dyslexia candidate genes on reading skill in old age,” Behav Genet, vol. 48, pp. 351–360.
- R. D. Fields, A. Araque, H. Johansen-Berg, S. S. Lim, G. Lynch, and N. KA, “Glial biology in learning and cognition,” The neuroscientist, vol. 20, no. 5, pp. 426–431.
- A. Mordelt and L. D. Witte, “Microglia-mediated synaptic pruning as a key deficit in neurodevelopmental disorders: Hype or hope?,” Curr Opin Neurobiol, vol. 79, no. 102674.
- L. Sancho, M. Contreras, and N. J. Allen, “Glia as sculptors of synaptic plasticity,” Neurosci Res, vol. 167, pp. 17–29.
- A. Badimon, H. J. Strasburger, P. Ayata, X. Chen, A. Nair, and I. A, “Negative feedback control of neuronal activity by microglia,” Nature, vol. 586, no. 7829, pp. 417–423.
- Y. Du, F. H. Brennan, P. G. Popovich, and M. Zhou, “Microglia maintain the normal structure and function of the hippocampal astrocyte network,” Glia, vol. 70, no. 7, pp. 1359–1379.
- C. Cserép et al., “Microglia monitor and protect neuronal function through specialized somatic purinergic junctions,” Science (1979), vol. 367, no. 6477, pp. 528–537, Jan. 2020.
- D. P. Schafer et al., “Microglia sculpt postnatal neural circuits in an activity and complement-dependent manner,” Neuron, vol. 74, no. 4, pp. 691–705, May 2012.
- J. L. Stobart and C. M. Anderson, “Multifunctional role of astrocytes as gatekeepers of neuronal energy supply,” Front Cell Neurosci, vol. 7, no. 38.
- X. Verkhratsky and A. Parpura, “Astrocytes: Physiology and Pathophysiology,” Physiology and Pathophysiology. Progress in Neurobiology, vol. 188, 2020.
- X. Patel, “Alterations in glutamate transport and metabolism in neurodevelopmental disorders,” Neurochem Int, vol. 145, 2021.
- L. Hertz, “Astrocytic energy metabolism and glutamate formation—relevance for 13C-NMR spectroscopy and importance of cytosolic/mitochondrial trafficking,” Magn Reson Imaging, vol. 29, no. 10, pp. 1319–1329.
- B. Kossowski, K. Chyl, A. Kacprzak, P. Bogorodzki, and K. Jednoróg, “Dyslexia and age related effects in the neurometabolites concentration in the visual and temporo-parietal cortex.”.
- X. Liu, “NAA/Cho ratio alterations in developmental dyslexia: a meta-analysis of MRS studies,” Neuroimage Clin, vol. 33, 2022.
- X. Colombo and E. Farina, “Astrocytes: Key regulators of neuroinflammation,” Trends Immunol, vol. 37, no. 9, pp. 608–620, 2016.
- X. Sofroniew and M. V Vinters, “Astrocytes: biology and pathology,” Acta Neuropathol, vol. 119, no. 1, pp. 7–35, 2010.
- X. Verkhratsky and A. Nedergaard, “Physiology of astroglia,” Physiol Rev, vol. 98, no. 1, pp. 239–389, 2018.
- X. Burda and J. E. Sofroniew, “Reactive gliosis and the multicellular response to CNS damage and disease,” Neuron, vol. 81, no. 2, pp. 229–248, 2014.
- R. L. Peterson and B. F. Pennington, “Developmental dyslexia,” The lancet, vol. 379, no. 9830, pp. 1997–2007.
- R. D. Fields, “White matter in learning, cognition and psychiatric disorders,” Trends Neurosci, vol. 31, no. 7, pp. 361–370, 2008.
- M. A. Skeide, P. L. Bazin, R. Trampel, A. Schäfer, C. Männel, and K. K, “Hypermyelination of the left auditory cortex in developmental dyslexia,” Neurology, vol. 90, no. 6.
- M. Carreiras, M. L. Seghier, S. Baquero, A. Estévez, A. Lozano, and D. JT, “An anatomical signature for literacy,” Nature, vol. 461, no. 7266, pp. 983–986.
- M. Makinodan, K. M. Rosen, S. Ito, and G. Corfas, “A critical period for social experience-dependent oligodendrocyte maturation and myelination,” Science (1979), vol. 337, no. 6100, pp. 1357–1360, 2012.
- X. Schafer and D. P. Stevens, “Microglia function in central nervous system development and plasticity,” Cold Spring Harb Perspect Biol, 2015.
- R. Farah, S. Ionta, and T. Horowitz-Kraus, “Neuro-behavioral correlates of executive dysfunctions in dyslexia over development from childhood to adulthood,” Front Psychol, vol. 12, no. 708863.
- C. Stüber, M. Morawski, A. Schäfer, C. Labadie, M. Wähnert, and L. C, “Myelin and iron concentration in the human brain: a quantitative study of MRI contrast,” Neuroimage, vol. 93, pp. 95–106.
- C. Beaulieu, “Myelin water imaging demonstrates lower brain myelination in children and adolescents with poor reading ability,” Front Hum Neurosci, vol. 14, 2020.
- J. B. D. Sinisa Pajevicdietmar Plenzpeter, “Oligodendrocyte-mediated myelin plasticity and its role in neural synchronization eLife,” vol. 12, 2023.
- Y. Hattori, “The multifaceted roles of embryonic microglia in the developing brain,” Front Cell Neurosci, vol. 17, 2023.
- C. Doust, “Discovery of 42 genome-wide significant loci associated with dyslexia,” Nat Genet, vol. 54, pp. 1621–1629, 2022.
- R. D. Fields, “Glial regulation of synaptic function and plasticity in health and disease,” Physiol Rev, vol. 102, no. 3, pp. 1251–1286, 2022.
- C. W. Mount and M. Monje, “Wrapped to adapt: experience-dependent myelination,” Neuron, vol. 95, no. 4, pp. 743–756, 2017.
- F. Costanzo, C. Varuzza, S. Rossi, S. Sdoia, P. Varvara, and O. M, “Evidence for reading improvement following tDCS treatment in children and adolescents with dyslexia,” Restor Neurol Neurosci, vol. 34, no. 2, pp. 215–226.
- A. Cancer and A. Antonietti, “tDCS modulatory effect on reading processes: A review of studies on typical readers and individuals with dyslexia,” Front Behav Neurosci, vol. 12, no. 162.
- A. Battisti, G. Lazzaro, C. Varuzza, S. Vicari, and D. Menghini, “Effects of online tDCS and hf-tRNS on reading performance in children and adolescents with developmental dyslexia: a study protocol for a cross sectional, within-subject, randomized, double-blind, and sham-controlled trial,” Front Neurol, vol. 15, no. 1338430.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
