Systematic Reviews
Copyright ©The Author(s) 2024.
World J Clin Pediatr. Jun 9, 2024; 13(2): 92737
Published online Jun 9, 2024. doi: 10.5409/wjcp.v13.i2.92737
Table 1 The changes observed in each amino acid metabolic pathway in individuals with autism spectrum disorder, along with relevant studies and their methods
Ref.
Study methods
Amino acid pathway
Changes in ASD
Boccuto et al[40]Blood or CSF sample analysisphenylalanine and glutamate Elevated phenylalanine and glutamate levels
Ormstad et al[41]Metabolomic profilingphenylalanine and glutamate Decreased glutathione and creatine levels
Almulla et al[42]Peripheral blood analysisTryptophan pathwayDisrupted tryptophan metabolism
Higazi et al[56]Genetic analysis of MAOA, HAAO and AADAT genes using real-time RT-qPCR.tryptophan pathwaysignificant decrease in the expression of the selected genes within ASD children relative to children with learning disabilities and healthy controls
Naushad et al[66]Genetic analysisTryptophan pathwayLower serotonin synthesis
Naushad et al[66]Enzyme activity measurementPhenylalanine and tyrosine metabolismReduced tyrosine synthesis
Arum et al[71]Dietary intake assessmentPhenylalanine and tyrosine metabolismAbnormal phenylalanine-to-tyrosine ratio
Geier et al[84]Peripheral blood analysisMethionine and sulfur metabolismreduced levels of serum glutathione, cysteine, methionine, cystathionine, and homocysteine in pre-pubertal children
Suh et al[85]Genetic analysisMethionine and sulfur metabolismReduced SAM levels
Guo et al[86]Meta-analysis of metabolomic profilingMethionine and sulfur metabolismImpaired methylation capacity
Haghiri et al[89]Genetic analysisMethionine metabolismmutations of methionine synthase in children with ASD
Table 2 The roles, impact, potential biomarkers, genetic associations, and therapeutic implications of both purine and pyrimidine metabolism in the context of autism spectrum disorder
Aspect
Purine metabolism
Pyrimidine metabolism
FunctionsBuilding blocks for nucleic acid synthesis; Metabolic signals; Provide energy; Regulate cell growth; Participate in coenzymes; Contribute to sugar transport; Donate phosphate groups in phosphorylation reactionsSynthesizing DNA and RNA; Energy metabolism; Neurotransmitter signaling
Specific rolesProvide energy for membrane-active pumps like Na+/K+ ATPase; Vital for sustaining synaptic transmission; Facilitate collaboration between neurons and glial cellsBiosynthesis of polysaccharides and phospholipids; Participate in detoxification processes; Contribute to protein and lipid glycosylation
Impact on brain functionAltered purine metabolism may impact brain function and contribute to ASD symptoms; Adenosine acts as a neuromodulator, inhibiting neurotransmitter release and regulating sleep-wake cyclesAbnormalities in pyrimidine metabolism may be linked to ASD and neurodevelopmental issues; Disturbances in uracil metabolism could contribute to mitochondrial dysfunction in ASD
Potential biomarkersElevated adenosine levels and altered ADA activity observed in ASD; Abnormal levels of purine metabolites such as uric acid reported in ASDAltered uracil levels and abnormal ratios of uracil to other pyrimidine bases reported in ASD; Abnormal levels of pyrimidine nucleotides observed in ASD
Genetic associationsMutations in genes encoding enzymes involved in purine metabolism found in individuals with ASDGenetic mutations in the gene encoding DPD identified in individuals with ASD
Therapeutic implicationsModulating adenosine signaling and targeting enzymes involved in purine metabolism could potentially improve neurochemical functioning in ASDSupplementation with pyrimidine precursors such as uridine was explored as a possible intervention to improve mitochondrial function and neurodevelopmental outcomes in ASD
Table 3 The prevalence, causes, impact, symptoms, diagnosis, management, challenges, and ongoing research related to mitochondrial metabolic disorders in individuals with autism spectrum disorder
Aspect
Mitochondrial metabolic disorders in ASD
OverviewMitochondria are cellular structures responsible for generating energy (ATP). High mitochondrial density in muscle and brain cells
Prevalence and causesIncreased prevalence of mitochondrial dysfunction in ASD compared to the general population. Can result from genetic mutations, environmental factors, or both
EvidenceAbout 80% of children with ASD show blood test indications of mitochondrial dysfunction and DNA abnormalities. Estimates of co-occurrence range from 5% to 80%
Contributing factorsGenetic mutations; Dietary deficiencies; Chemical and heavy metal exposure; Certain drugs; Bacterial and viral infections; Stressful conditions
Impact on ASDInsufficient ATP production can affect synaptic plasticity, neuronal development, signaling, and maintenance. Oxidative stress and damage to cellular components may occur. Disruption of metabolic processes can further impact ASD development
Symptoms and diagnosisSymptoms include delays in developmental milestones, impaired language and communication, motor difficulties, cognitive impairments, behavioral abnormalities, seizures, and gastrointestinal issues. Diagnosis involves comprehensive clinical assessments, biochemical analyses, genetic testing, and specific diagnostic criteria. Laboratory tests may include blood tests, urine tests, DNA analysis, brain imaging, and muscle biopsy
Management and treatmentTreatment strategies may include dietary interventions, nutritional supplements, antioxidants, and medications targeted at specific symptoms. A multidisciplinary approach involving healthcare professionals from various specialties is necessary for accurate diagnosis and management
Challenges and ongoing researchDiagnosis can be challenging due to overlapping symptoms and lack of specific criteria. The effectiveness of interventions in improving ASD symptoms associated with mitochondrial dysfunction is still under research
Table 4 The prevalence, contributing factors, impact, therapeutic implications, and research needs related to lipid metabolism abnormalities in individuals with autism spectrum disorder
Aspect
Lipid metabolism in ASD
OverviewLipid metabolism involves the synthesis, breakdown, and transportation of fats, which are crucial for cell membranes and energy
Dyslipidemia in ASDAbnormal lipid levels observed in children with ASD. Variations include elevated total cholesterol and LDL-C, reduced HDL-C, and increased triglycerides. Increased LDL-C to HDL-C ratio, a marker of cardiovascular risk
Factors contributing to abnormalitiesGenetic variations in lipid metabolism-related genes. Syndromes like Smith–Melli–Opitz syndrome linked to lipid metabolism and neurodevelopmental delay. Oxidative stress and chronic inflammation are common in ASD, affecting lipid metabolism. Gut microbiota alterations and dietary factors are also implicated
Impact on ASDAbnormal lipid metabolism can affect brain development, myelination, synaptogenesis, and neurotransmitter signaling in ASD. Disruptions may lead to oxidative stress, neuroinflammation, and neuronal damage. Potential implications for cellular energy production and utilization in the brain
Therapeutic implicationsInterest in lipid-based interventions for ASD, but effectiveness needs further research. Potential therapeutic targets to address lipid disorders and associated symptoms in ASD. Complex relationship between lipid disorders and ASD, influenced by genetic, environmental, and metabolic factors
Research needsFurther research is needed to understand the mechanisms underlying lipid disorders in ASD. Investigation required into the effectiveness of lipid-based interventions on ASD symptoms and lipid profiles. Recognition that not all individuals with ASD have lipid abnormalities, and vice versa