Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 1794 2023-04-24 10:11:04 |
2 format correct -11 word(s) 1783 2023-04-25 05:29:15 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Senarathne, U.D.; Indika, N.R.; Jezela-Stanek, A.; Ciara, E.; Frye, R.E.; Chen, C.; Stepien, K.M. Biochemical Investigations of Autism Spectrum Disorders. Encyclopedia. Available online: https://encyclopedia.pub/entry/43372 (accessed on 27 July 2024).
Senarathne UD, Indika NR, Jezela-Stanek A, Ciara E, Frye RE, Chen C, et al. Biochemical Investigations of Autism Spectrum Disorders. Encyclopedia. Available at: https://encyclopedia.pub/entry/43372. Accessed July 27, 2024.
Senarathne, Udara D., Neluwa-Liyanage R. Indika, Aleksandra Jezela-Stanek, Elżbieta Ciara, Richard E. Frye, Cliff Chen, Karolina M. Stepien. "Biochemical Investigations of Autism Spectrum Disorders" Encyclopedia, https://encyclopedia.pub/entry/43372 (accessed July 27, 2024).
Senarathne, U.D., Indika, N.R., Jezela-Stanek, A., Ciara, E., Frye, R.E., Chen, C., & Stepien, K.M. (2023, April 24). Biochemical Investigations of Autism Spectrum Disorders. In Encyclopedia. https://encyclopedia.pub/entry/43372
Senarathne, Udara D., et al. "Biochemical Investigations of Autism Spectrum Disorders." Encyclopedia. Web. 24 April, 2023.
Biochemical Investigations of Autism Spectrum Disorders
Edit

The main biochemical mechanisms proposed in autism spectrum disorders (ASD) include mitochondrial dysfunction, oxidative stress, impaired methylation capacity, and altered amino acid metabolism.

autism ASD WES

1. Introduction

Autism spectrum disorders (ASD) are defined by the revised version of the Diagnostic and Statistical Manual of Mental Disorders-5 (DSM-5-TR) as neuro-developmental disorders characterized by persistent deficits in reciprocal social communication and social interaction (Criterion A) and restricted and/or repetitive patterns of behaviour, interests, or activities (Criterion B), present from early childhood (Criterion C) causing clinically significant impairment in social, occupational, or other important areas of current functioning (Criterion D). The symptoms under criteria A and B are not better explained by intellectual disability or global developmental delay (Criterion E). It is known as a “spectrum” disorder because there is wide variability in the severity and pattern of symptoms, progression of the disease, and prognosis. ASD is accompanied by varying degrees of intellectual impairment and is associated with many genetic, neurodevelopmental, mental, and behavioural disorders and environmental factors [1].
The global prevalence of ASD is estimated to be 0.4–1%, with American and European countries having a higher prevalence (1%) compared to Asian (0.4%) countries [2]. The biannual estimates by ASD and the Autism and Developmental Disabilities Monitoring (ADDM) Network report a two-fold increase in ASD prevalence among 8-year-old children in the United States during the past decade, from 2008 (1.1%) to 2018 (2.3%), with boys being four times more affected than girls [3].
The etiological factors and the proposed pathogenic mechanisms in ASD are intricate and involve the interaction of genetic, epigenetic, and environmental elements [4]. The main dysregulations, including intestinal dysbiosis [5], immune dysfunction, metabolic dysfunction [6], and metal dyshomeostasis [7], have been identified using molecular biomarkers, such as gut microbiome-related metabolites, cytokine profile, autoantibodies, metabolites, and vitamin and mineral profiles [8]. These are interconnected; hence, an ASD individual can exhibit several dysfunctions. For instance, a child with gut microbial dysbiosis may exhibit core symptoms of ASD along with immune dysfunction, altered microbial metabolites, and epigenetic changes [9][10]. The gut microbiome may modulate the gut–brain axis through microbiota-derived signalling molecules, immune mediators, gastrointestinal hormones, etc. [10][11].
Although each inherited metabolic disorder (IMD) is rare in isolation, collectively, all IMDs have a combined incidence of 1:800 to 1:2500 [12][13][14]. Contributing to the heterogeneity, a number of IMD, such as 22q11.2 deletion, Angelman syndrome, Cohen syndrome, Noonan syndrome, and fragile X syndrome, are known to associate with autistic symptoms, corroborating the evidence for genetic etiologies of ASD [15][16][17][18][19][20][21]. It should also be noted that the symptoms of ASD can be present in association with many IMDs [16]. The prevalence of IMDs among ASD individuals has been estimated in different studies, ranging from 0.7% to 2.7% [22][23][24]. However, the true prevalence of IMDs among ASD patients has been speculated to be higher [25], corroborating the finding of more than 30% of ASD individuals having some form of metabolic derangement by Spilioti et al. [24]. Furthermore, more than 50% of the IMDs present with neurodevelopmental symptoms, and ASD primarily being a neurodevelopmental disorder, the rational investigation of ASD individuals for probable IMDs is appropriate, especially in communities with a high level of consanguinity [26][27][28].
To date, more than 100 autism-risk genes have been identified with a genetic cause found in 10–20% of the cases of ASD during the investigation process [22][29]. A whole-exome sequencing (WES) study identified that 52% were nuclear sequence-level variants, 46% were nuclear structural variants, and 2% were mitochondrial variants [30]. Exome sequencing is identified as a first-tier clinical diagnostic test for individuals with neurodevelopmental disorders, including ASD, with a molecular diagnostic yield of 16% (CI: 11–24%) in ASD [31]. Additionally, chromosomal microarray (CMA) has revealed definitively pathogenic copy number variants (CNVs) in 5.4% to 14% of individuals with ASD [32]. Even though ASD-associated biallelic genetic variants are commonly reported in consanguineous families, some were observed in non-consanguineous families. Siblings from consanguineous and/or multiplex families who share identical homozygous biallelic mutation may show variable ASD phenotypes with or without intellectual disability, epilepsy, and other clinical features [33]. Moreover, it is estimated that de novo variants in protein-coding genes contribute to risk in approximately 30% of simplex families. At the same time, de novo variants in non-coding regions of the genome (particularly gene promoters) also contribute to ASD pathogenesis [34]. De novo variants in the non-coding region highlight the current need to perform ASD genetic studies using whole genome sequencing (WGS) instead of traditional exome studies [35].
Large population-based studies on the outcome of metabolic investigations in screening for IMDs among ASD individuals are lacking; hence, accurate prevalence and diagnostic yield estimates are not available. Despite the observed low yield of routine metabolic testing in ASD individuals, the positive impact on ASD management may be significantly high as it paves the path to better understanding the underlying pathophysiology, inheritance pattern, and availability of treatment [32].
With the advancement of tandem mass spectrometry, most developed countries employ expanded newborn screening (NBS) as an important public health strategy, though it is yet to become a priority in developing countries [36][37]. Therefore, children who receive an expanded NBS have the advantage of receiving the diagnosis of an IMD, if present, before they would develop any autistic symptoms, whereas, in developing countries, ASD patients may only undergo targeted/selective screening for IMDs following the establishment of the primary diagnosis of a neurological condition or ASD. Selective screening for IMDs in ASD subjects may be a cost-effective strategy to identify the patients with IMDs that remain undiagnosed in countries where mass NBS for IMDs is unavailable. If the diagnosis of an IMD was established, treatment targeted at specific metabolic abnormalities in children with ASD has potential benefits [24][38]. For instance, identifying mitochondrial dysfunction enables the implementation of targeted therapies such as vitamins, carnitine, Coenzyme Q10, etc. [39][40]. In addition, research on advancing medical treatments, such as hematopoietic stem cell transplantation, is underway, offering hope for IMDs associated with ASD [41][42].
Investigating for IMDs associated with ASD should take a multidisciplinary approach led by a clinician (psychiatrist/neurologist/paediatrician/metabolic physician) with a well-informed team, including a chemical pathologist, clinical biochemist, and clinical geneticist. The primary aim of this endeavour is to understand the underlying pathophysiology, to provide genetic counselling where appropriate, and to determine the potential therapies. The success of investigating for IMDs in ASD patients depends on the understanding of different phenotypes of known syndromes and IMDs that overlap with ASD and defining a tailored biochemical and molecular evaluation plan catering to the needs of the individual patient based on their unique clinical information [43]. The assays to be considered as a screening panel should be based on common IMDs associated with ASD, the local epidemiology of IMDs in a given country, and available resources. In addition, the coverage of the existing NBS program should be considered in determining redundant investigations.

2. Biochemical Investigations

The main biochemical mechanisms proposed in ASD include mitochondrial dysfunction [39][44], oxidative stress [45], impaired methylation capacity [46], and altered amino acid metabolism [47]. Interestingly, ASD patients had these metabolic abnormalities in the brain regions involved in speech and auditory processing, social behaviour, sensory and motor coordination, and memory, the core symptoms of ASD [48]. Corroborating the presence of mitochondrial dysfunction in ASD, a 2020 study demonstrated atypical mitochondrial morphology with mitochondrial electron transport chain abnormalities in the fibroblasts of children with ASD [49]. Furthermore, several mitochondrial functional biomarkers, such as lactate, pyruvate, carnitine, and ubiquinone, are significantly altered in ASD, while some even correlate with severity [39][44]. The impaired methylation capacity is evident by the significantly reduced S-adenosylmethionine/S-adenosylhomocysteine ratio (methylation index) in ASD, while reduced methionine levels and increased homocysteine levels indicate the impaired remethylation of homocysteine to methionine [45]. The reduced availability of the cofactors for the remethylation pathway in the brain may be attributed to reduced blood folate [45], vitamin B12 levels [45], low-activity variants of the genes (MTHFR, DHFR, FOLR1) affecting folate and cobalamin metabolism [50][51], and cerebral folate deficiency due to folate receptor α autoantibodies [52]. Deth et al. proposed a “redox/methylation hypothesis of autism” describing the pathogenesis of oxidative stress, precipitated by environmental factors in genetically vulnerable individuals, which limits the activity of methionine synthase due to its dependency on cobalamin and folate, hence impaired methylation, including dopamine-stimulated phospholipid methylation [53]. Reduced methylation capacity may also mediate epigenetic changes through modulating DNA and histone methylation in a sex-dependent manner [50]. These mechanisms are thought to induce changes in the neurotransmitter systems, such as γ-aminobutyric acid (GABA) and glutamate, serotonin, dopamine, melatonin, and acetylcholine [54], resulting in core symptoms and co-occurring behavioural and neurological symptoms.
Pathway enrichment analysis has demonstrated that microRNAs (miRNAs) differently expressed in ASD are involved in metabolic pathways, such as steroid biosynthesis, fatty acid metabolism, lysine degradation, and biotin metabolism [55]. Correspondingly ASD-associated IMDs can be mapped to disorders of the metabolism of amino acids, carbohydrates, fatty acids, sterol biosynthesis, ketone bodies, creatine, vitamins (B12, folate, and biotin), cofactors, nucleotide, mitochondrial metabolite repair, etc. Importantly, these genetic and metabolic disorders are associated with marked cognitive impairment and other clinical features, such as macrocephaly, extrapyramidal signs, motor developmental delay, dysmorphic features, failure to thrive, or hepatosplenomegaly, which are atypical for patients with ASD [17][56].
In the context of complex pathogenesis with multiple co-morbidities and the evidence of metabolic involvement and strong heritable component, although the pathophysiology is not well comprehended, the current disease trajectory is developing towards a better understanding of the metabolic component of ASD in order to advance future interventions to improve the overall quality of life for ASD individuals [57][58][59]. The future developments are mainly focused on improving early and accurate diagnostic algorithms in unravelling the metabolic and other components of ASD. The diagnostic yield of metabolic investigations in patients with isolated ASD and no clinical symptoms appears to be low [22][23].
Before the establishment of NBS, a large proportion of these individuals would have been brought to medical attention only after the development of autistic symptoms. However, with the NBS programs, especially in developed countries, many such individuals are brought to medical attention early, and necessary treatments are instigated; a good example is phenylalanine ketonuria, an IMD characterized by intellectual disability and ASD [60][61][62]. Therefore, the diagnostic approach to ASD should be equipped with a rational consideration of possible IMDs, as some are treatable [63]. A recently suggested approach to such investigation is untargeted metabolomic profiling, as many ASD patients demonstrate a wide range of metabolic abnormalities, from micronutrient deficiencies to severe metabolic derangements [64]. The basis for untargeted metabolic profiling is the emerging evidence of potential novel biomarkers of IMD associated with ASD, as found in many cohort studies conducted worldwide, further opening new avenues of treatments [24]. However, the additional healthcare costs need to be considered while expanding the investigation profile to weigh the medical benefits of further testing, and it may not be cost-effective in non-syndromic ASD [64]. Schiff et al. demonstrated that the prevalence of IMD among non-syndromic ASD individuals is not higher than the general population (<0.5%) by conducting a systematic metabolic work-up, highlighting the importance of a rational approach to metabolomics in ASD [22].

References

  1. American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders, 5th ed.; Text Revision; American psychiatric Association: Washington, DC, USA, 2022.
  2. Salari, N.; Rasoulpoor, S.; Rasoulpoor, S.; Shohaimi, S.; Jafarpour, S.; Abdoli, N.; Khaledi-Paveh, B.; Mohammadi, M. The global prevalence of autism spectrum disorder: A comprehensive systematic review and meta-analysis. Ital. J. Pediatr. 2022, 48, 112.
  3. Kilmer, M.; Boykin, A.A. Analysis of the 2000 to 2018 autism and developmental disabilities monitoring network surveillance reports: Implications for primary care clinicians. J. Pediatr. Nurs. 2022, 65, 55–68.
  4. Bhandari, R.; Paliwal, J.K.; Kuhad, A. Neuropsychopathology of Autism Spectrum Disorder: Complex Interplay of Genetic, Epigenetic, and Environmental Factors. Adv. Neurobiol. 2020, 24, 97–141.
  5. Srikantha, P.; Mohajeri, M.H. The Possible Role of the Microbiota-Gut-Brain-Axis in Autism Spectrum Disorder. Int. J. Mol. Sci. 2019, 20, 2115.
  6. Cheng, N.; Rho, J.M.; Masino, S.A. Metabolic Dysfunction Underlying Autism Spectrum Disorder and Potential Treatment Approaches. Front. Mol. Neurosci. 2017, 10, 34.
  7. Zhang, J.; Li, X.; Shen, L.; Khan, N.U.; Zhang, X.; Chen, L.; Zhao, H.; Luo, P. Trace elements in children with autism spectrum disorder: A meta-analysis based on case-control studies. J. Trace Elem. Med. Biol. 2021, 67, 126782.
  8. Jensen, A.R.; Lane, A.L.; Werner, B.A.; McLees, S.E.; Fletcher, T.S.; Frye, R.E. Modern Biomarkers for Autism Spectrum Disorder: Future Directions. Mol. Diagn. Ther. 2022, 26, 483–495.
  9. Alharthi, A.; Alhazmi, S.; Alburae, N.; Bahieldin, A. The Human Gut Microbiome as a Potential Factor in Autism Spectrum Disorder. Int. J. Mol. Sci. 2022, 23, 1363.
  10. Sorboni, S.G.; Moghaddam, H.S.; Jafarzadeh-Esfehani, R.; Soleimanpour, S. A Comprehensive Review on the Role of the Gut Microbiome in Human Neurological Disorders. Clin. Microbiol. Rev. 2022, 35, e0033820.
  11. Ristori, M.V.; Quagliariello, A.; Reddel, S.; Ianiro, G.; Vicari, S.; Gasbarrini, A.; Putignani, L. Autism, Gastrointestinal Symptoms and Modulation of Gut Microbiota by Nutritional Interventions. Nutrients 2019, 11, 2812.
  12. Applegarth, D.A.; Toone, J.R.; Lowry, R.B. Incidence of inborn errors of metabolism in British Columbia, 1969–1996. Pediatrics 2000, 105, e10.
  13. Sanderson, S.; Green, A.; Preece, M.A.; Burton, H. The incidence of inherited metabolic disorders in the West Midlands, UK. Arch. Dis. Child. 2006, 91, 896–899.
  14. Pampols, T. Inherited metabolic rare disease. Adv. Exp. Med. Biol. 2010, 686, 397–431.
  15. Žigman, T.; Petković Ramadža, D.; Šimić, G.; Barić, I. Inborn Errors of Metabolism Associated With Autism Spectrum Disorders: Approaches to Intervention. Front. Neurosci. 2021, 15, 673600.
  16. Simons, A.; Eyskens, F.; Glazemakers, I.; Van West, D. Can psychiatric childhood disorders be due to inborn errors of metabolism? Eur. Child Adolesc. Psychiatry 2017, 26, 143–154.
  17. Frye, R.E.; Rossignol, D.A. Metabolic disorders and abnormalities associated with autism spectrum disorder. J. Pediatr. Biochem. 2012, 2, 181–191.
  18. Caglayan, A.O. Genetic causes of syndromic and non-syndromic autism. Dev. Med. Child. Neurol. 2010, 52, 130–138.
  19. Zafeiriou, D.I.; Ververi, A.; Dafoulis, V.; Kalyva, E.; Vargiami, E. Autism spectrum disorders: The quest for genetic syndromes. Am. J. Med. Genet. Part B Neuropsychiatr. Genet. 2013, 162, 327–366.
  20. Fernandez, B.A.; Scherer, S.W. Syndromic autism spectrum disorders: Moving from a clinically defined to a molecularly defined approach. Dialogues Clin. Neurosci. 2017, 19, 353.
  21. Genovese, A.; Butler, M.G. Clinical Assessment, Genetics, and Treatment Approaches in Autism Spectrum Disorder (ASD). Int. J. Mol. Sci. 2020, 21, 4726.
  22. Schiff, M.; Benoist, J.F.; Aïssaoui, S.; Boespflug-Tanguy, O.; Mouren, M.C.; de Baulny, H.O.; Delorme, R. Should metabolic diseases be systematically screened in nonsyndromic autism spectrum disorders? PLoS ONE 2011, 6, e21932.
  23. Campistol, J.; Diez-Juan, M.; Callejon, L.; Fernandez-De Miguel, A.; Casado, M.; Garcia Cazorla, A.; Lozano, R.; Artuch, R. Inborn error metabolic screening in individuals with nonsyndromic autism spectrum disorders. Dev. Med. Child Neurol. 2016, 58, 842–847.
  24. Spilioti, M.; Evangeliou, A.E.; Tramma, D.; Theodoridou, Z.; Metaxas, S.; Michailidi, E.; Bonti, E.; Frysira, H.; Haidopoulou, A.; Asprangathou, D.; et al. Evidence for treatable inborn errors of metabolism in a cohort of 187 Greek patients with autism spectrum disorder (ASD). Front. Hum. Neurosci. 2013, 7, 858.
  25. Ghaziuddin, M.; Al-Owain, M. Autism spectrum disorders and inborn errors of metabolism: An update. Pediatr. Neurol. 2013, 49, 232–236.
  26. Saudubray, J.M.; Garcia-Cazorla, A. An overview of inborn errors of metabolism affecting the brain: From neurodevelopment to neurodegenerative disorders. Dialogues Clin. Neurosci. 2018, 20, 301–325.
  27. Celestino-Soper, P.B.; Violante, S.; Crawford, E.L.; Luo, R.; Lionel, A.C.; Delaby, E.; Cai, G.; Sadikovic, B.; Lee, K.; Lo, C.; et al. A common X-linked inborn error of carnitine biosynthesis may be a risk factor for nondysmorphic autism. Proc. Natl. Acad. Sci. USA 2012, 109, 7974–7981.
  28. Novarino, G.; El-Fishawy, P.; Kayserili, H.; Meguid, N.A.; Scott, E.M.; Schroth, J.; Silhavy, J.L.; Kara, M.; Khalil, R.O.; Ben-Omran, T.; et al. Mutations in BCKD-kinase lead to a potentially treatable form of autism with epilepsy. Science 2012, 338, 394–397.
  29. Satterstrom, F.K.; Kosmicki, J.A.; Wang, J.; Breen, M.S.; De Rubeis, S.; An, J.Y.; Peng, M.; Collins, R.; Grove, J.; Klei, L.; et al. Large-Scale Exome Sequencing Study Implicates Both Developmental and Functional Changes in the Neurobiology of Autism. Cell. 2020, 180, 568–584.e23.
  30. Trost, B.; Thiruvahindrapuram, B.; Chan, A.J.S.; Engchuan, W.; Higginbotham, E.J.; Howe, J.L.; Loureiro, L.O.; Reuter, M.S.; Roshandel, D.; Whitney, J.; et al. Genomic architecture of autism from comprehensive whole-genome sequence annotation. Cell 2022, 185, 4409–4427.e18.
  31. Srivastava, S.; Love-Nichols, J.A.; Dies, K.A.; Ledbetter, D.H.; Martin, C.L.; Chung, W.K.; Firth, H.V.; Frazier, T.; Hansen, R.L.; Prock, L.; et al. Meta-analysis and multidisciplinary consensus statement: Exomesequencing is a first-tier clinical diagnostic test for individuals with neurodevelopmental disorders. Genet. Med. 2019, 21, 2413–2421.
  32. Hyman, S.L.; Levy, S.E.; Myers, S.M.; Kuo, D.Z.; Apkon, S.; Davidson, L.F.; Ellerbeck, K.A.; Foster, J.E.A.; Noritz, G.H.; Leppert, M.O.; et al. Identification, Evaluation, and Management of Children With Autism Spectrum Disorder. Pediatrics 2020, 145, e20193447.
  33. Yu, T.W.; Chahrour, M.H.; Coulter, M.E.; Jiralerspong, S.; Okamura-Ikeda, K.; Ataman, B.; Schmitz-Abe, K.; Harmin, D.A.; Adli, M.; Malik, A.N.; et al. Using whole-exome sequencing to identify inherited causes of autism. Neuron 2013, 77, 259–273.
  34. Havdahl, A.; Niarchou, M.; Starnawska, A.; Uddin, M.; van der Merwe, C.; Warrier, V. Genetic contributions to autism spectrum disorder. Psychol. Med. 2021, 51, 2260–2273.
  35. Alonso-Gonzalez, A.; Rodriguez-Fontenla, C.; Carracedo, A. De novo Mutations (DNMs) in Autism Spectrum Disorder (ASD): Pathway and Network Analysis. Front. Genet. 2018, 9, 406.
  36. Mak, C.M.; Lee, H.C.; Chan, A.Y.; Lam, C.W. Inborn errors of metabolism and expanded newborn screening: Review and update. Crit. Rev. Clin. Lab. Sci. 2013, 50, 142–162.
  37. Therrell, B.L., Jr.; Padilla, C.D. Newborn screening in the developing countries. Curr. Opin. Pediatr. 2018, 30, 734–739.
  38. Delhey, L.M.; Tippett, M.; Rose, S.; Bennuri, S.C.; Slattery, J.C.; Melnyk, S.; James, S.J.; Frye, R.E. Comparison of Treatment for Metabolic Disorders Associated with Autism:Reanalysis of Three Clinical Trials. Front. Neurosci. 2018, 12, 19.
  39. Frye, R.E. Mitochondrial Dysfunction in Autism Spectrum Disorder: Unique Abnormalities and Targeted Treatments. Semin. Pediatr. Neurol. 2020, 35, 100829.
  40. Niyazov, D.M.; Kahler, S.G.; Frye, R.E. Primary Mitochondrial Disease and Secondary Mitochondrial Dysfunction: Importance of Distinction for Diagnosis and Treatment. Mol. Syndromol. 2016, 7, 122–137.
  41. Sun, J.M.; Kurtzberg, J. Cell therapy for diverse central nervous system disorders: Inherited metabolic diseases and autism. Pediatr. Res. 2018, 83, 364–371.
  42. Paprocka, J.; Kaminiów, K.; Kozak, S.; Sztuba, K.; Emich-Widera, E. Stem Cell Therapies for Cerebral Palsy and Autism Spectrum Disorder-A Systematic Review. Brain Sci. 2021, 11, 1606.
  43. Schaefer, G.B.; Mendelsohn, N.J. Clinical genetics evaluation in identifying the etiology of autism spectrum disorders: 2013 guideline revisions. Genet. Med. 2013, 15, 399–407.
  44. Rossignol, D.A.; Frye, R.E. Mitochondrial dysfunction in autism spectrum disorders: A systematic review and meta-analysis. Mol. Psychiatry 2012, 17, 290–314.
  45. Chen, L.; Shi, X.J.; Liu, H.; Mao, X.; Gui, L.N.; Wang, H.; Cheng, Y. Oxidative stress marker aberrations in children with autism spectrum disorder: A systematic review and meta-analysis of 87 studies (N = 9109). Transl. Psychiatry 2021, 11, 15.
  46. Guo, B.Q.; Ding, S.B.; Li, H.B. Blood biomarker levels of methylation capacity in autism spectrum disorder: A systematic review and meta-analysis. Acta Psychiatr. Scand. 2020, 141, 492–509.
  47. Indika, N.R.; Deutz, N.E.P.; Engelen, M.; Peiris, H.; Wijetunge, S.; Perera, R. Sulfur amino acid metabolism and related metabotypes of autism spectrum disorder: A review of biochemical evidence for a hypothesis. Biochimie 2021, 184, 143–157.
  48. Rossignol, D.A.; Frye, R.E. Evidence linking oxidative stress, mitochondrial dysfunction, and inflammation in the brain of individuals with autism. Front. Physiol. 2014, 5, 150.
  49. Pecorelli, A.; Ferrara, F.; Messano, N.; Cordone, V.; Schiavone, M.L.; Cervellati, F.; Woodby, B.; Cervellati, C.; Hayek, J.; Valacchi, G. Alterations of mitochondrial bioenergetics, dynamics, and morphology support the theory of oxidative damage involvement in autism spectrum disorder. FASEB J. 2020, 34, 6521–6538.
  50. Tisato, V.; Silva, J.A.; Longo, G.; Gallo, I.; Singh, A.V.; Milani, D.; Gemmati, D. Genetics and Epigenetics of One-Carbon Metabolism Pathway in Autism Spectrum Disorder: A Sex-Specific Brain Epigenome? Genes 2021, 12, 782.
  51. James, S.J.; Melnyk, S.; Jernigan, S.; Cleves, M.A.; Halsted, C.H.; Wong, D.H.; Cutler, P.; Bock, K.; Boris, M.; Bradstreet, J.J.; et al. Metabolic endophenotype and related genotypes are associated with oxidative stress in children with autism. Am. J. Med. Genet. B Neuropsychiatr. Genet. 2006, 141B, 947–956.
  52. Rossignol, D.A.; Frye, R.E. Cerebral Folate Deficiency, Folate Receptor Alpha Autoantibodies and Leucovorin (Folinic Acid) Treatment in Autism Spectrum Disorders: A Systematic Review and Meta-Analysis. J. Pers. Med. 2021, 11, 1141.
  53. Deth, R.; Muratore, C.; Benzecry, J.; Power-Charnitsky, V.A.; Waly, M. How environmental and genetic factors combine to cause autism: A redox/methylation hypothesis. Neurotoxicology 2008, 29, 190–201.
  54. Marotta, R.; Risoleo, M.C.; Messina, G.; Parisi, L.; Carotenuto, M.; Vetri, L.; Roccella, M. The Neurochemistry of Autism. Brain Sci. 2020, 10, 163.
  55. Huang, Z.X.; Chen, Y.; Guo, H.R.; Chen, G.F. Systematic Review and Bioinformatic Analysis of microRNA Expression in Autism Spectrum Disorder Identifies Pathways Associated With Cancer, Metabolism, Cell Signaling, and Cell Adhesion. Front. Psychiatry 2021, 12, 630876.
  56. Kiykim, E.; Zeybek, C.A.; Zubarioglu, T.; Cansever, S.; Yalcinkaya, C.; Soyucen, E.; Aydin, A. Inherited metabolic disorders in Turkish patients with autism spectrum disorders. Autism Res. 2016, 9, 217–223.
  57. Toscano, C.V.A.; Barros, L.; Lima, A.B.; Nunes, T.; Carvalho, H.M.; Gaspar, J.M. Neuroinflammation in autism spectrum disorders: Exercise as a “pharmacological” tool. Neurosci. Biobehav. Rev. 2021, 129, 63–74.
  58. McElhanon, B.O.; McCracken, C.; Karpen, S.; Sharp, W.G. Gastrointestinal symptoms in autism spectrum disorder: A meta-analysis. Pediatrics 2014, 133, 872–883.
  59. Granich, J.; Lin, A.; Hunt, A.; Wray, J.; Dass, A.; Whitehouse, A.J. Obesity and associated factors in youth with an autism spectrum disorder. Autism 2016, 20, 916–926.
  60. Witters, P.; Debbold, E.; Crivelly, K.; Vande Kerckhove, K.; Corthouts, K.; Debbold, B.; Andersson, H.; Vannieuwenborg, L.; Geuens, S.; Baumgartner, M.; et al. Autism in patients with propionic acidemia. Mol. Genet. Metab. 2016, 119, 317–321.
  61. Sikora, D.M.; Pettit-Kekel, K.; Penfield, J.; Merkens, L.S.; Steiner, R.D. The near universal presence of autism spectrum disorders in children with Smith-Lemli-Opitz syndrome. Am. J. Med. Genet. A 2006, 140, 1511–1518.
  62. Asato, M.R.; Goldstein, A.C.; Schiff, M. Autism and inborn errors of metabolism: How much is enough? Dev. Med. Child. Neurol. 2015, 57, 788–789.
  63. Márquez-Caraveo, M.E.; Ibarra-González, I.; Rodríguez-Valentín, R.; Ramírez-García, M.; Pérez-Barrón, V.; Lazcano-Ponce, E.; Vela-Amieva, M. Brief Report: Delayed Diagnosis of Treatable Inborn Errors of Metabolism in Children with Autism and Other Neurodevelopmental Disorders. J. Autism Dev. Disord. 2021, 51, 2124–2131.
  64. Glinton, K.E.; Elsea, S.H. Untargeted Metabolomics for Autism Spectrum Disorders: Current Status and Future Directions. Front. Psychiatry 2019, 10, 647.
More
Information
Subjects: Pediatrics
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , , , ,
View Times: 291
Revisions: 2 times (View History)
Update Date: 25 Apr 2023
1000/1000
Video Production Service