Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Targeted Biomedical Treatment for Autism Spectrum Disorders

Author(s): Iliyana Pacheva* and Ivan Ivanov

Volume 25, Issue 41, 2019

Page: [4430 - 4453] Pages: 24

DOI: 10.2174/1381612825666191205091312

Price: $65

Abstract

Background: A diagnosis of autism spectrum disorders (ASD) represents presentations with impairment in communication and behaviour that vary considerably in their clinical manifestations and etiology as well as in their likely pathophysiology. A growing body of data indicates that the deleterious effect of oxidative stress, mitochondrial dysfunction, immune dysregulation and neuroinflammation, as well as their interconnections are important aspects of the pathophysiology of ASD. Glutathione deficiency decreases the mitochondrial protection against oxidants and tumor necrosis factor (TNF)-α; immune dysregulation and inflammation inhibit mitochondrial function through TNF-α; autoantibodies against the folate receptors underpin cerebral folate deficiency, resulting in disturbed methylation, and mitochondrial dysfunction. Such pathophysiological processes can arise from environmental and epigenetic factors as well as their combined interactions, such as environmental toxicant exposures in individuals with (epi)genetically impaired detoxification. The emerging evidence on biochemical alterations in ASD is forming the basis for treatments aimed to target its biological underpinnings, which is of some importance, given the uncertain and slow effects of the various educational interventions most commonly used.

Methods: Literature-based review of the biomedical treatment options for ASD that are derived from established pathophysiological processes.

Results: Most proposed biomedical treatments show significant clinical utility only in ASD subgroups, with specified pre-treatment biomarkers that are ameliorated by the specified treatment. For example, folinic acid supplementation has positive effects in ASD patients with identified folate receptor autoantibodies, whilst the clinical utility of methylcobalamine is apparent in ASD patients with impaired methylation capacity. Mitochondrial modulating cofactors should be considered when mitochondrial dysfunction is evident, although further research is required to identify the most appropriate single or combined treatment. Multivitamins/multiminerals formulas, as well as biotin, seem appropriate following the identification of metabolic abnormalities, with doses tapered to individual requirements. A promising area, requiring further investigations, is the utilization of antipurinergic therapies, such as low dose suramin.

Conclusion: The assessment and identification of relevant physiological alterations and targeted intervention are more likely to produce positive treatment outcomes. As such, current evidence indicates the utility of an approach based on personalized and evidence-based medicine, rather than treatment targeted to all that may not always be beneficial (primum non nocere).

Keywords: Autism spectrum disorders, mitochondria, pathophysiological mechanisms, biomedical treatment, biomarkers, personalized medicine.

[1]
American Psychiatric Association. Diagnostic and Statistical Manual of Mental Disorders. 5th ed. Arlington, VA: American Psychiatric Association 2013.
[2]
Baron-Cohen S, Scott FJ, Allison C, et al. Prevalence of autism-spectrum conditions: UK school-based population study. Br J Psychiatry 2009; 194(6): 500-9.
[http://dx.doi.org/10.1192/bjp.bp.108.059345] [PMID: 19478287]
[3]
Blumberg SJ, Bramlett MD, Kogan MD, Schieve LA, Jones JR, Lu MC. Changes in prevalence of parent-reported autism spectrum disorder in school-aged U.S. children: 2007 to 2011-2012. Natl Health Stat Rep 2013; (65): 1-11.
[PMID: 24988818]
[4]
Xu G, Strathearn L, Liu B, et al. Prevalence and treatment patterns of autism spectrum disorder in the United States, 2016. JAMA Pediatr 2019; 173(2): 153-9.
[http://dx.doi.org/10.1001/jamapediatrics.2018.4208] [PMID: 30508021]
[5]
Inui T, Kumagaya S, Myowa-Yamakoshi M. Neurodevelopmental hypothesis about the etiology of autism spectrum disorders. Front Hum Neurosci 2017; 11: 354.
[http://dx.doi.org/10.3389/fnhum.2017.00354] [PMID: 28744208]
[6]
Gabis LV, Pomeroy J. An etiologic classification of autism spectrum disorders. Isr Med Assoc J 2014; 16(5): 295-8.
[PMID: 24979834]
[7]
Betancur C, Coleman M. Etiological heterogeneity in autism spectrum disorders: role of rare variants. Oxford: Academic press 2013; pp. 113-44.
[http://dx.doi.org/10.1016/B978-0-12-391924-3.00008-9]
[8]
Bozzi Y, Provenzano G, Casarosa S. Neurobiological bases of autism-epilepsy comorbidity: a focus on excitation/inhibition imbalance. Eur J Neurosci 2018; 47(6): 534-48.
[http://dx.doi.org/10.1111/ejn.13595] [PMID: 28452083]
[9]
Jeste SS, Tuchman R. Autism spectrum disorder and epilepsy: two sides of the same coin? J Child Neurol 2015; 30(14): 1963-71.
[http://dx.doi.org/10.1177/0883073815601501] [PMID: 26374786]
[10]
Srivastava S, Sahin M. Autism spectrum disorder and epileptic encephalopathy: common causes, many questions. J Neurodev Disord 2017; 9: 23.
[http://dx.doi.org/10.1186/s11689-017-9202-0] [PMID: 28649286]
[11]
Robert C, Pasquier L, Cohen D, et al. Role of genetics in the etiology of autistic spectrum disorder: towards a hierarchical diagnostic strategy. Int J Mol Sci 2017; 18(3): 618.
[http://dx.doi.org/10.3390/ijms18030618] [PMID: 28287497]
[12]
van Eeghen AM, Pulsifer MB, Merker VL, et al. Understanding relationships between autism, intelligence, and epilepsy: a cross-disorder approach. Dev Med Child Neurol 2013; 55(2): 146-53.
[http://dx.doi.org/10.1111/dmcn.12044] [PMID: 23205844]
[13]
Dröge W. Free radicals in the physiological control of cell function. Physiol Rev 2002; 82(1): 47-95.
[http://dx.doi.org/10.1152/physrev.00018.2001] [PMID: 11773609]
[14]
Siddiqui MF, Elwell C, Johnson MH. Mitochondrial dysfunction in autism spectrum disorders Autism Open Access 2016; 6(5): 1000190
[http://dx.doi.org/10.4172/2165-7890.1000190]
[15]
Rossignol DA, Frye RE. Mitochondrial dysfunction in autism spectrum disorders: a systematic review and meta-analysis. Mol Psychiatry 2012; 17(3): 290-314.
[http://dx.doi.org/10.1038/mp.2010.136] [PMID: 21263444]
[16]
Chauhan A, Gu F, Essa MM, et al. Brain region-specific deficit in mitochondrial electron transport chain complexes in children with autism. J Neurochem 2011; 117(2): 209-20.
[http://dx.doi.org/10.1111/j.1471-4159.2011.07189.x] [PMID: 21250997]
[17]
Friedman SD, Shaw DW, Artru AA, et al. Regional brain chemical alterations in young children with autism spectrum disorder. Neurology 2003; 60(1): 100-7.
[http://dx.doi.org/10.1212/WNL.60.1.100] [PMID: 12525726]
[18]
Ipser JC, Syal S, Bentley J, Adnams CM, Steyn B, Stein DJ. 1H-MRS in autism spectrum disorders: a systematic meta-analysis. Metab Brain Dis 2012; 27(3): 275-87.
[http://dx.doi.org/10.1007/s11011-012-9293-y] [PMID: 22426803]
[19]
James SJ, Cutler P, Melnyk S, et al. Metabolic biomarkers of increased oxidative stress and impaired methylation capacity in children with autism. Am J Clin Nutr 2004; 80(6): 1611-7.
[http://dx.doi.org/10.1093/ajcn/80.6.1611] [PMID: 15585776]
[20]
Bennuri SC, Rose S, Frye RE. Mitochondrial dysfunction is inducible in lymphoblastoid cell lines from children with autism and may involve the TORC1 pathway. Front Psychiatry 2019; 10: 269.
[http://dx.doi.org/10.3389/fpsyt.2019.00269] [PMID: 31133888]
[21]
Rose S, Niyazov DM, Rossignol DA, Goldenthal M, Kahler SG, Frye RE. Clinical and molecular characteristics of mitochondrial dysfunction in autism spectrum disorder. Mol Diagn Ther 2018; 22(5): 571-93.
[http://dx.doi.org/10.1007/s40291-018-0352-x] [PMID: 30039193]
[22]
Naviaux RK. Antipurinergic therapy for autism-an in-depth review. Mitochondrion 2018; 43: 1-15.
[http://dx.doi.org/10.1016/j.mito.2017.12.007] [PMID: 29253638]
[23]
Sander LE, Garaude J. The mitochondrial respiratory chain: A metabolic rheostat of innate immune cell-mediated antibacterial responses. Mitochondrion 2018; 41: 28-36.
[http://dx.doi.org/10.1016/j.mito.2017.10.008] [PMID: 29054472]
[24]
Sipe GO, Lowery RL, Tremblay ME, Kelly EA, Lamantia CE, Majewska AK. Microglial P2Y12 is necessary for synaptic plasticity in mouse visual cortex. Nat Commun 2016; 7: 10905.
[http://dx.doi.org/10.1038/ncomms10905] [PMID: 26948129]
[25]
Rose S, Melnyk S, Pavliv O, et al. Evidence of oxidative damage and inflammation associated with low glutathione redox status in the autism brain. Transl Psychiatry 2012; 2 e134
[http://dx.doi.org/10.1038/tp.2012.61] [PMID: 22781167]
[26]
Tang G, Gutierrez Rios P, Kuo SH, et al. Mitochondrial abnormalities in temporal lobe of autistic brain. Neurobiol Dis 2013; 54: 349-61.
[http://dx.doi.org/10.1016/j.nbd.2013.01.006] [PMID: 23333625]
[27]
Palmieri L, Papaleo V, Porcelli V, et al. Altered calcium homeostasis in autism-spectrum disorders: evidence from biochemical and genetic studies of the mitochondrial aspartate/glutamate carrier AGC1. Mol Psychiatry 2010; 15(1): 38-52.
[http://dx.doi.org/10.1038/mp.2008.63] [PMID: 18607376]
[28]
Hardan AY, Fung LK, Frazier T, et al. A proton spectroscopy study of white matter in children with autism. Prog Neuropsychopharmacol Biol Psychiatry 2016; 66: 48-53.
[http://dx.doi.org/10.1016/j.pnpbp.2015.11.005] [PMID: 26593330]
[29]
James SJ, Melnyk S, Jernigan S, 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(8): 947-56.
[http://dx.doi.org/10.1002/ajmg.b.30366] [PMID: 16917939]
[30]
Rose S, Melnyk S, Trusty TA, et al. Intracellular and extracellular redox status and free radical generation in primary immune cells from children with autism. Autism Res Treat 2012; 2012 986519
[http://dx.doi.org/10.1155/2012/986519] [PMID: 22928106]
[31]
James SJ, Rose S, Melnyk S, et al. Cellular and mitochondrial glutathione redox imbalance in lymphoblastoid cells derived from children with autism. FASEB J 2009; 23(8): 2374-83.
[http://dx.doi.org/10.1096/fj.08-128926] [PMID: 19307255]
[32]
Rossignol DA, Frye RE. Evidence linking oxidative stress, mitochondrial dysfunction, and inflammation in the brain of individuals with autism. Front Physiol 2014; 5: 150.
[http://dx.doi.org/10.3389/fphys.2014.00150] [PMID: 24795645]
[33]
Yorbik O, Sayal A, Akay C, Akbiyik DI, Sohmen T. Investigation of antioxidant enzymes in children with autistic disorder. Prostaglandins Leukot Essent Fatty Acids 2002; 67(5): 341-3.
[http://dx.doi.org/10.1054/plef.2002.0439] [PMID: 12445495]
[34]
Gu F, Chauhan V, Chauhan A. Impaired synthesis and antioxidant defense of glutathione in the cerebellum of autistic subjects: alterations in the activities and protein expression of glutathione-related enzymes. Free Radic Biol Med 2013; 65: 488-96.
[http://dx.doi.org/10.1016/j.freeradbiomed.2013.07.021] [PMID: 23892356]
[35]
Griffiths KK, Levy RJ. Evidence of mitochondrial dysfunction in autism: biochemical links, genetic-based associations, and non-energy-related mechanisms. Oxid Med Cell Longev 2017; 2017 4314025
[http://dx.doi.org/10.1155/2017/4314025] [PMID: 28630658]
[36]
Ming X, Stein TP, Brimacombe M, Johnson WG, Lambert GH, Wagner GC. Increased excretion of a lipid peroxidation biomarker in autism. Prostaglandins Leukot Essent Fatty Acids 2005; 73(5): 379-84.
[http://dx.doi.org/10.1016/j.plefa.2005.06.002] [PMID: 16081262]
[37]
Frye RE, Slattery JC, Quadros EV. Folate metabolism abnormalities in autism: potential biomarkers. Biomarkers Med 2017; 11(8): 687-99.
[http://dx.doi.org/10.2217/bmm-2017-0109] [PMID: 28770615]
[38]
Hill BG, Higdon AN, Dranka BP, Darley-Usmar VM. Regulation of vascular smooth muscle cell bioenergetic function by protein glutathiolation. Biochim Biophys Acta 2010; 1797(2): 285-95.
[http://dx.doi.org/10.1016/j.bbabio.2009.11.005] [PMID: 19925774]
[39]
Dranka BP, Benavides GA, Diers AR, et al. Assessing bioenergetic function in response to oxidative stress by metabolic profiling. Free Radic Biol Med 2011; 51(9): 1621-35.
[http://dx.doi.org/10.1016/j.freeradbiomed.2011.08.005] [PMID: 21872656]
[40]
Frye RE, Melnyk S, Macfabe DF. Unique acyl-carnitine profiles are potential biomarkers for acquired mitochondrial disease in autism spectrum disorder. Transl Psychiatry 2013; 3 e220
[http://dx.doi.org/10.1038/tp.2012.143] [PMID: 23340503]
[41]
Weinberg SE, Sena LA, Chandel NS. Mitochondria in the regulation of innate and adaptive immunity. Immunity 2015; 42(3): 406-17.
[http://dx.doi.org/10.1016/j.immuni.2015.02.002] [PMID: 25786173]
[42]
Sacco R, Lenti C, Saccani M, et al. Cluster analysis of autistic patients based on principal pathogenetic components. Autism Res 2012; 5(2): 137-47.
[http://dx.doi.org/10.1002/aur.1226] [PMID: 22431251]
[43]
Hughes HK, Mills Ko E, Rose D, Ashwood P. Immune dysfunction and autoimmunity as pathological mechanisms in autism spectrum disorders. Front Cell Neurosci 2018; 12: 405.
[http://dx.doi.org/10.3389/fncel.2018.00405] [PMID: 30483058]
[44]
Zerbo O, Leong A, Barcellos L, Bernal P, Fireman B, Croen LA. Immune mediated conditions in autism spectrum disorders. Brain Behav Immun 2015; 46: 232-6.
[http://dx.doi.org/10.1016/j.bbi.2015.02.001] [PMID: 25681541]
[45]
Li X, Chauhan A, Sheikh AM, et al. Elevated immune response in the brain of autistic patients. J Neuroimmunol 2009; 207(1-2): 111-6.
[http://dx.doi.org/10.1016/j.jneuroim.2008.12.002] [PMID: 19157572]
[46]
Smith JA, Das A, Ray SK, Banik NL. Role of pro-inflammatory cytokines released from microglia in neurodegenerative diseases. Brain Res Bull 2012; 87(1): 10-20.
[http://dx.doi.org/10.1016/j.brainresbull.2011.10.004] [PMID: 22024597]
[47]
Wei H, Zou H, Sheikh AM, et al. IL-6 is increased in the cerebellum of autistic brain and alters neural cell adhesion, migration and synaptic formation. J Neuroinflammation 2011; 8: 52.
[http://dx.doi.org/10.1186/1742-2094-8-52] [PMID: 21595886]
[48]
Young AM, Campbell E, Lynch S, et al. Aberrant NF-kappa B expression in autism spectrum condition: a mechanism for neuroinflammation. Front Psychiatry 2011; 2: 27.
[49]
Rodriguez JI, Kern JK. Evidence of microglial activation in autism and its possible role in brain underconnectivity. Neuron Glia Biol 2011; 7(2-4): 205-13.
[http://dx.doi.org/10.1017/S1740925X12000142] [PMID: 22874006]
[50]
Morgan JT, Chana G, Pardo CA, et al. Microglial activation and increased microglial density observed in the dorsolateral prefrontal cortex in autism. Biol Psychiatry 2010; 68(4): 368-76.
[http://dx.doi.org/10.1016/j.biopsych.2010.05.024] [PMID: 20674603]
[51]
Wills S, Cabanlit M, Bennett J, Ashwood P, Amaral DG, Van de Water J. Detection of autoantibodies to neural cells of the cerebellum in the plasma of subjects with autism spectrum disorders. Brain Behav Immun 2009; 23(1): 64-74.
[http://dx.doi.org/10.1016/j.bbi.2008.07.007] [PMID: 18706993]
[52]
Wills S, Rossi CC, Bennett J, et al. Further characterization of autoantibodies to GABAergic neurons in the central nervous system produced by a subset of children with autism. Mol Autism 2011; 2: 5.
[http://dx.doi.org/10.1186/2040-2392-2-5] [PMID: 21521495]
[53]
Sweeten TL, Posey DJ, Shankar S, McDougle CJ. High nitric oxide production in autistic disorder: a possible role for interferon-gamma. Biol Psychiatry 2004; 55(4): 434-7.
[http://dx.doi.org/10.1016/j.biopsych.2003.09.001] [PMID: 14960298]
[54]
Marchezan J, Winkler Dos Santos EGA, Deckmann I, Riesgo RDS. Immunological dysfunction in autism spectrum disorder: a potential target for therapy. Neuroimmunomodulation 2018; 25(5-6): 300-19.
[http://dx.doi.org/10.1159/000492225] [PMID: 30184549]
[55]
Brenna JT, Carlson SE. Docosahexaenoic acid and human brain development: evidence that a dietary supply is needed for optimal development. J Hum Evol 2014; 77: 99-106.
[http://dx.doi.org/10.1016/j.jhevol.2014.02.017] [PMID: 24780861]
[56]
Hibbeln JR, Linnoila M, Umhau JC, Rawlings R, George DT, Salem N Jr. Essential fatty acids predict metabolites of serotonin and dopamine in cerebrospinal fluid among healthy control subjects, and early- and late-onset alcoholics. Biol Psychiatry 1998; 44(4): 235-42.
[http://dx.doi.org/10.1016/S0006-3223(98)00141-3] [PMID: 9715354]
[57]
Wurtman RJ. Synapse formation and cognitive brain development: effect of docosahexaenoic acid and other dietary constituents. Metabolism 2008; 57(Suppl. 2): S6-S10.
[http://dx.doi.org/10.1016/j.metabol.2008.07.007] [PMID: 18803968]
[58]
Brigandi SA, Shao H, Qian SY, Shen Y, Wu BL, Kang JX. Autistic children exhibit decreased levels of essential fatty acids in red blood cells. Int J Mol Sci 2015; 16(5): 10061-76.
[http://dx.doi.org/10.3390/ijms160510061] [PMID: 25946342]
[59]
Simopoulos AP. Evolutionary aspects of diet: the ω-6/ω-3 ratio and the brain. Mol Neurobiol 2011; 44(2): 203-15.
[http://dx.doi.org/10.1007/s12035-010-8162-0] [PMID: 21279554]
[60]
Parletta N, Niyonsenga T, Duff J. Omega-3 and Omega-6 polyunsaturated fatty acid levels and correlations with symptoms in children with attention deficit hyperactivity disorder, autistic spectrum disorder and typically developing controls. PLoS One 2016; 11(5) e0156432
[http://dx.doi.org/10.1371/journal.pone.0156432] [PMID: 27232999]
[61]
Mazahery H, Stonehouse W, Delshad M, et al. Relationship between long chain n-3 polyunsaturated fatty acids and autism spectrum disorder: systematic review and meta-analysis of case-control and randomised controlled trials. Nutrients 2017; 9(2) E155
[http://dx.doi.org/10.3390/nu9020155] [PMID: 28218722]
[62]
Howsmon DP, Adams JB, Kruger U, Geis E, Gehn E, Hahn J. Erythrocyte fatty acid profiles in children are not predictive of autism spectrum disorder status: a case control study. Biomark Res 2018; 6: 12.
[http://dx.doi.org/10.1186/s40364-018-0125-z] [PMID: 29568526]
[63]
Jiang X, Suenaga J, Pu H, et al. Post-stroke administration of omega-3 polyunsaturated fatty acids promotes neurovascular restoration after ischemic stroke in mice: efficacy declines with aging. Neurobiol Dis 2019; 126: 62-75.
[http://dx.doi.org/10.1016/j.nbd.2018.09.012] [PMID: 30218758]
[64]
Volpato M, Hull MA. Omega-3 polyunsaturated fatty acids as adjuvant therapy of colorectal cancer. Cancer Metastasis Rev 2018; 37(2-3): 545-55.
[http://dx.doi.org/10.1007/s10555-018-9744-y] [PMID: 29971573]
[65]
Yoshino J, Smith GI, Kelly SC, Julliand S, Reeds DN, Mittendorfer B. Effect of dietary n-3 PUFA supplementation on the muscle transcriptome in older adults. Physiol Rep 2016; 4(11) e12785
[http://dx.doi.org/10.14814/phy2.12785] [PMID: 27252251]
[66]
Mulder EJ, Anderson GM, Kemperman RFJ, Oosterloo-Duinkerken A, Minderaa RB, Kema IP. Urinary excretion of 5-hydroxyindoleacetic acid, serotonin and 6-sulphatoxymelatonin in normoserotonemic and hyperserotonemic autistic individuals. Neuropsychobiology 2010; 61(1): 27-32.
[http://dx.doi.org/10.1159/000258640] [PMID: 19923863]
[67]
Croonenberghs J, Delmeire L, Verkerk R, et al. Peripheral markers of serotonergic and noradrenergic function in post-pubertal, caucasian males with autistic disorder. Neuropsychopharmacology 2000; 22(3): 275-83.
[http://dx.doi.org/10.1016/S0893-133X(99)00131-1] [PMID: 10693155]
[68]
Uzunova G, Pallanti S, Hollander E. Excitatory/inhibitory imbalance in autism spectrum disorders: implications for interventions and therapeutics. World J Biol Psychiatry 2015; 1-13.
[PMID: 26469219]
[69]
El-Ansary A, Al Dera H. Biomarkers directed strategies to treat autism.In: Role of Biomarkers in Medicine. In Tech Publisher 2016; pp. 205-28.
[http://dx.doi.org/10.5772/62566]
[70]
Wei H, Ding C, Jin G, Yin H, Liu J, Hu F. Abnormal glutamate release in aged BTBR mouse model of autism. Int J Clin Exp Pathol 2015; 8(9): 10689-97.
[PMID: 26617779]
[71]
Tanaka K. [Role of glutamate transporters in the pathophysiology of major mental illnesses Nihon Shinkei Seishin Yakurigaku Zasshi 2009; 29(4): 161-4.
[PMID: 19764483]
[72]
Liu J, Zhang M, Kong X. Gut microbiome and autism: recent advances and future perspectives. North Am J Med Sci 2016; 9(3): 104-15.
[73]
Hsiao EY, McBride SW, Hsien S, et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell 2013; 155(7): 1451-63.
[http://dx.doi.org/10.1016/j.cell.2013.11.024] [PMID: 24315484]
[74]
Sandler RH, Finegold SM, Bolte ER, et al. Short-term benefit from oral vancomycin treatment of regressive-onset autism. J Child Neurol 2000; 15(7): 429-35.
[http://dx.doi.org/10.1177/088307380001500701] [PMID: 10921511]
[75]
Rodakis J. An n=1 case report of a child with autism improving on antibiotics and a father’s quest to understand what it may mean. Microb Ecol Health Dis 2015; 26: 26382.
[PMID: 25808801]
[76]
Xu M, Xu X, Li J, Li F. Association between Gut microbiota and autism spectrum disorder: a systematic review and meta-analysis. Front Psychiatry 2019; 10: 473.
[http://dx.doi.org/10.3389/fpsyt.2019.00473] [PMID: 31404299]
[77]
Song Y, Liu C, Finegold SM. Real-time PCR quantitation of clostridia in feces of autistic children. Appl Environ Microbiol 2004; 70(11): 6459-65.
[http://dx.doi.org/10.1128/AEM.70.11.6459-6465.2004] [PMID: 15528506]
[78]
Söderholm JD, Yang PC, Ceponis P, et al. Chronic stress induces mast cell-dependent bacterial adherence and initiates mucosal inflammation in rat intestine. Gastroenterology 2002; 123(4): 1099-108.
[http://dx.doi.org/10.1053/gast.2002.36019] [PMID: 12360472]
[79]
Guinane CM, Cotter PD. Role of the gut microbiota in health and chronic gastrointestinal disease: understanding a hidden metabolic organ. Therap Adv Gastroenterol 2013; 6(4): 295-308.
[http://dx.doi.org/10.1177/1756283X13482996] [PMID: 23814609]
[80]
Mayer EA, Savidge T, Shulman RJ. Brain-gut microbiome interactions and functional bowel disorders. Gastroenterology 2014; 146(6): 1500-12.
[http://dx.doi.org/10.1053/j.gastro.2014.02.037] [PMID: 24583088]
[81]
Shaw W. Increased urinary excretion of a 3-(3-hydroxyphenyl)-3-hydroxypropionic acid (HPHPA), an abnormal phenylalanine metabolite of Clostridia spp. in the gastrointestinal tract, in urine samples from patients with autism and schizophrenia. Nutr Neurosci 2010; 13(3): 135-43.
[http://dx.doi.org/10.1179/147683010X12611460763968] [PMID: 20423563]
[82]
Nankova BB, Agarwal R, MacFabe DF, et al. Enteric bacterial metabolites propionic and butyric acid modulate gene expression, including CREB-dependent catecholaminergic neurotransmission, in PC12 cells--possible relevance to autism spectrum disorders. PLoS One 2014; 9 e103740
[83]
Meyer-Lindenberg A, Domes G, Kirsch P, Heinrichs M. Oxytocin and vasopressin in the human brain: social neuropeptides for translational medicine. Nat Rev Neurosci 2011; 12(9): 524-38.
[http://dx.doi.org/10.1038/nrn3044] [PMID: 21852800]
[84]
Modahl C, Green L, Fein D, et al. Plasma oxytocin levels in autistic children. Biol Psychiatry 1998; 43(4): 270-7.
[http://dx.doi.org/10.1016/S0006-3223(97)00439-3] [PMID: 9513736]
[85]
Andari E, Duhamel J-R, Zalla T, Herbrecht E, Leboyer M, Sirigu A. Promoting social behavior with oxytocin in high-functioning autism spectrum disorders. Proc Natl Acad Sci USA 2010; 107(9): 4389-94.
[http://dx.doi.org/10.1073/pnas.0910249107] [PMID: 20160081]
[86]
Rossignol DA, Frye RE. Melatonin in autism spectrum disorders: a systematic review and meta-analysis. Dev Med Child Neurol 2011; 53(9): 783-92.
[http://dx.doi.org/10.1111/j.1469-8749.2011.03980.x] [PMID: 21518346]
[87]
Tordjman S, Anderson GM, Bellissant E, et al. Day and nighttime excretion of 6-sulphatoxymelatonin in adolescents and young adults with autistic disorder. Psychoneuroendocrinology 2012; 37(12): 1990-7.
[http://dx.doi.org/10.1016/j.psyneuen.2012.04.013] [PMID: 22613035]
[88]
Hardeland R, Pandi-Perumal SR. Melatonin, a potent agent in antioxidative defense: actions as a natural food constituent, gastrointestinal factor, drug and prodrug. Nutr Metab (Lond) 2005; 2: 22.
[http://dx.doi.org/10.1186/1743-7075-2-22] [PMID: 16153306]
[89]
Srinivasan V, Pandi-Perumal SR, Maestroni GJ, Esquifino AI, Hardeland R, Cardinali DP. Role of melatonin in neurodegenerative diseases. Neurotox Res 2005; 7(4): 293-318.
[http://dx.doi.org/10.1007/BF03033887] [PMID: 16179266]
[90]
Sarowar T, Chhabra R, Vilella A, Boeckers TM, Zoli M, Grabrucker AM. Activity and circadian rhythm influence synaptic Shank3 protein levels in mice. J Neurochem 2016; 138(6): 887-95.
[http://dx.doi.org/10.1111/jnc.13709] [PMID: 27329942]
[91]
Anderson G. Mitochondria and the Gut as crucial hubs for the interactions of melatonin with sirtuins, inflammation, butyrate, tryptophan metabolites, and alpha 7 nicotinic receptor across a host of medical conditions. Melatonin Res 2019; 2(2): 70-85.
[http://dx.doi.org/10.32794/mr11250022]
[92]
Delhey LM, Nur Kilinc E, Yin L, et al. The effect of mitochondrial supplements on mitochondrial activity in children with autism spectrum disorder. J Clin Med 2017; 6(2): 18.
[http://dx.doi.org/10.3390/jcm6020018] [PMID: 28208802]
[93]
Filipek PA, Juranek J, Nguyen MT, Cummings C, Gargus JJ. Relative carnitine deficiency in autism. J Autism Dev Disord 2004; 34(6): 615-23.
[http://dx.doi.org/10.1007/s10803-004-5283-1] [PMID: 15679182]
[94]
Lv QQ, You C, Zou XB, Deng HZ. Acyl-carnitine, C5DC, and C26 as potential biomarkers for diagnosis of autism spectrum disorder in children. Psychiatry Res 2018; 267: 277-80.
[http://dx.doi.org/10.1016/j.psychres.2018.06.027] [PMID: 29945069]
[95]
Geier DA, Kern JK, Davis G, et al. A prospective double-blind, randomized clinical trial of levocarnitine to treat autism spectrum disorders. Med Sci Monit 2011; 17(6): PI15-23.
[http://dx.doi.org/10.12659/MSM.881792] [PMID: 21629200]
[96]
Fahmy SF, El-Hamamsy M, Zaki O, Badary OA. Effect of L-carnitine on behavioral disorder in autistic children. Value Health 2013; 16(3): A15.
[http://dx.doi.org/10.1016/j.jval.2013.03.092]
[97]
Wawrzeńczyk A, Sacher A, Mac M, Nałecz MJ, Nałecz KA. Transport of L-carnitine in isolated cerebral cortex neurons. Eur J Biochem 2001; 268(7): 2091-8.
[http://dx.doi.org/10.1046/j.1432-1327.2001.02087.x] [PMID: 11277932]
[98]
Pastural E, Ritchie S, Lu Y, et al. Novel plasma phospholipid biomarkers of autism: mitochondrial dysfunction as a putative causative mechanism. Prostaglandins Leukot Essent Fatty Acids 2009; 81(4): 253-64.
[http://dx.doi.org/10.1016/j.plefa.2009.06.003] [PMID: 19608392]
[99]
Yubero D, Adin A, Montero R, et al. A statistical algorithm showing coenzyme Q10 and citrate synthase as biomarkers for mitochondrial respiratory chain enzyme activities. Sci Rep 2016; 6(1): 15.
[http://dx.doi.org/10.1038/s41598-016-0008-1] [PMID: 28442759]
[100]
Faust K, Gehrke S, Yang Y, et al. Neuroprotective effects of compounds with antioxidant and anti-inflammatory properties in a Drosophila model of Parkinson’s disease. BMC Neurosci 2009; 1(10): 109.
[http://dx.doi.org/10.1186/1471-2202-10-109]
[101]
Carmona MC, Lefebvre P, Lefebvre B, et al. Consortium of the french ministry of research and technology: ‘Molecules and new therapeutic targets’. Coadministration of coenzyme Q prevents rosiglitazone-induced adipogenesis in ob/ob mice. Int J Obes 2009; 33(2): 204-11.
[http://dx.doi.org/10.1038/ijo.2008.265] [PMID: 19125161]
[102]
Mousavinejad E, Ghaffari MA, Riahi F, Hajmohammadi M, Tiznobeyk Z, Mousavinejad M. Coenzyme Q10 supplementation reduces oxidative stress and decreases antioxidant enzyme activity in children with autism spectrum disorders. Psychiatry Res 2018; 265: 62-9.
[http://dx.doi.org/10.1016/j.psychres.2018.03.061] [PMID: 29684771]
[103]
Fan L, Feng Y, Chen GC, Qin LQ, Fu CL, Chen LH. Effects of coenzyme Q10 supplementation on inflammatory markers: a systematic review and meta-analysis of randomized controlled trials. Pharmacol Res 2017; 119: 128-36.
[http://dx.doi.org/10.1016/j.phrs.2017.01.032] [PMID: 28179205]
[104]
Gvozdjáková A, Kucharská J, Ostatníková D, Babinská K, Nakládal D, Crane FL. Ubiquinol improves symptoms in children with autism. Oxid Med Cell Longev 2014; 2014 798957
[http://dx.doi.org/10.1155/2014/798957] [PMID: 24707344]
[105]
Glover EI, Martin J, Maher A, Thornhill RE, Moran GR, Tarnopolsky MA. A randomized trial of coenzyme Q10 in mitochondrial disorders. Muscle Nerve 2010; 42(5): 739-48.
[http://dx.doi.org/10.1002/mus.21758] [PMID: 20886510]
[106]
Rodriguez MC, MacDonald JR, Mahoney DJ, Parise G, Beal MF, Tarnopolsky MA. Beneficial effects of creatine, CoQ10, and lipoic acid in mitochondrial disorders. Muscle Nerve 2007; 35(2): 235-42.
[http://dx.doi.org/10.1002/mus.20688] [PMID: 17080429]
[107]
Tarnopolsky MA, Raha S. Mitochondrial myopathies: diagnosis, exercise intolerance, and treatment options. Med Sci Sports Exerc 2005; 37(12): 2086-93.
[http://dx.doi.org/10.1249/01.mss.0000177341.89478.06] [PMID: 16331134]
[108]
Legido A, Goldenthal M, Garvin B, et al. Effect of a combination of carnitine, coenzyme Q10 and alpha-lipoic acid (MitoCocktail) on mitochondrial function and neurobehavioral performance in children with autism spectrum disorder (P3.313). Neurology 2018; 90(Suppl. 15).
[109]
James SJ. Autism and folate-dependent one-carbon metabolism: serendipity and critical branch-point decisions in science. Glob Adv Health Med 2013; 2(6): 48-51.
[http://dx.doi.org/10.7453/gahmj.2013.088] [PMID: 24416708]
[110]
Frye RE, James SJ. Metabolic pathology of autism in relation to redox metabolism. Biomarkers Med 2014; 8(3): 321-30.
[http://dx.doi.org/10.2217/bmm.13.158] [PMID: 24712422]
[111]
Frye RE, Sequeira JM, Quadros EV, James SJ, Rossignol DA. Cerebral folate receptor autoantibodies in autism spectrum disorder. Mol Psychiatry 2013; 18(3): 369-81.
[http://dx.doi.org/10.1038/mp.2011.175] [PMID: 22230883]
[112]
Frye RE, Slattery J, Delhey L, et al. Folinic acid improves verbal communication in children with autism and language impairment: a randomized double-blind placebo-controlled trial. Mol Psychiatry 2018; 23(2): 247-56.
[http://dx.doi.org/10.1038/mp.2016.168] [PMID: 27752075]
[113]
Ramaekers V, Sequeira JM, Quadros EV. Clinical recognition and aspects of the cerebral folate deficiency syndromes. Clin Chem Lab Med 2013; 51(3): 497-511.
[http://dx.doi.org/10.1515/cclm-2012-0543] [PMID: 23314536]
[114]
Gao Y, Sheng C, Xie R-H, et al. New perspective on impact of folic acid supplementation during pregnancy on neurodevelopment/autism in the offspring children - a systematic review. PLoS One 2016; 11(11) e0165626
[http://dx.doi.org/10.1371/journal.pone.0165626] [PMID: 27875541]
[115]
James SJ, Melnyk S, Fuchs G, et al. Efficacy of methylcobalamin and folinic acid treatment on glutathione redox status in children with autism. Am J Clin Nutr 2009; 89(1): 425-30.
[http://dx.doi.org/10.3945/ajcn.2008.26615] [PMID: 19056591]
[116]
Bottiglieri T. Folate, vitamin B12, and S-adenosylmethionine. Psychiatr Clin North Am 2013; 36(1): 1-13.
[http://dx.doi.org/10.1016/j.psc.2012.12.001] [PMID: 23538072]
[117]
Bertoglio K, Jill James S, Deprey L, Brule N, Hendren RL. Pilot study of the effect of methyl B12 treatment on behavioral and biomarker measures in children with autism. J Altern Complement Med 2010; 16(5): 555-60.
[http://dx.doi.org/10.1089/acm.2009.0177] [PMID: 20804367]
[118]
Hendren RL, James SJ, Widjaja F, Lawton B, Rosenblatt A, Bent S. Randomized, placebo-controlled trial of methyl B12 for children with autism. J Child Adolesc Psychopharmacol 2016; 26(9): 774-83.
[http://dx.doi.org/10.1089/cap.2015.0159] [PMID: 26889605]
[119]
Adams JB. Vitamin/mineral supplements for children and adults with autism. Vitam Miner 2015; 3: 127.
[http://dx.doi.org/10.4172/2376-1318.1000127]
[120]
Nakamura YK, Read MH, Elias JW, Omaye ST. Oxidation of serum low-density lipoprotein (LDL) and antioxidant status in young and elderly humans. Arch Gerontol Geriatr 2006; 42(3): 265-76.
[http://dx.doi.org/10.1016/j.archger.2005.08.002] [PMID: 16214244]
[121]
Mahmood LA, Al Saadi R, Matthews L. Dietary and antioxidant therapy for autistic children: does it really work? Arch Med Health Sci 2018; 6: 73-80.
[http://dx.doi.org/10.4103/amhs.amhs_82_17]
[122]
Adams JB, George F, Audhya T. Abnormally high plasma levels of vitamin B6 in children with autism not taking supplements compared to controls not taking supplements. J Altern Complement Med 2006; 12(1): 59-63.
[http://dx.doi.org/10.1089/acm.2006.12.59] [PMID: 16494569]
[123]
Adams JB, Audhya T, Geis E, et al. Comprehensive nutritional and dietary intervention for autism spectrum disorder-a randomized, controlled 12-month trial. Nutrients 2018; 10(3): 369.
[http://dx.doi.org/10.3390/nu10030369] [PMID: 29562612]
[124]
Frye R, Rossignol D. Treatments for mitochondrial dysfunction associated with autism spectrum disorders. J Pediatr Biochem 2012; 2(4): 241-9.
[125]
Adams JB, Audhya T, McDonough-Means S, et al. Effect of a vitamin/mineral supplement on children and adults with autism. BMC Pediatr 2011; 11: 111.
[http://dx.doi.org/10.1186/1471-2431-11-111] [PMID: 22151477]
[126]
Spilioti M, Evangeliou AE, Tramma 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.
[http://dx.doi.org/10.3389/fnhum.2013.00858] [PMID: 24399946]
[127]
Sghaier R, Zarrouk A, Nury T, et al. Biotin attenuation of oxidative stress, mitochondrial dysfunction, lipid metabolism alteration and 7β-hydroxycholesterol-induced cell death in 158N murine oligodendrocytes. Free Radic Res 2019; 53(5): 535-61.
[http://dx.doi.org/10.1080/10715762.2019.1612891] [PMID: 31039616]
[128]
Rimland B, Callaway E, Dreyfus P. The effect of high doses of vitamin B6 on autistic children: a double-blind crossover study. Am J Psychiatry 1978; 135(4): 472-5.
[http://dx.doi.org/10.1176/ajp.135.4.472] [PMID: 345827]
[129]
Lelord G, Muh JP, Barthelemy C, Martineau J, Garreau B, Callaway E. Effects of pyridoxine and magnesium on autistic symptoms--initial observations. J Autism Dev Disord 1981; 11(2): 219-30.
[http://dx.doi.org/10.1007/BF01531686] [PMID: 6765503]
[130]
Martineau J, Barthelemy C, Cheliakine C, Lelord G. Brief report: an open middle-term study of combined vitamin B6-magnesium in a subgroup of autistic children selected on their sensitivity to this treatment. J Autism Dev Disord 1988; 18(3): 435-47.
[http://dx.doi.org/10.1007/BF02212198] [PMID: 3170459]
[131]
Nye C, Brice A. Combined vitamin B6-magnesium treatment in autism spectrum disorder. Cochrane Database Syst Rev 2005; 4(4) CD003497
[http://dx.doi.org/10.1002/14651858.CD003497.pub2] [PMID: 16235322]
[132]
Findling RL, Maxwell K, Scotese-Wojtila L, Huang J, Yamashita T, Wiznitzer M. High-dose pyridoxine and magnesium administration in children with autistic disorder: an absence of salutary effects in a double-blind, placebo-controlled study. J Autism Dev Disord 1997; 27(4): 467-78.
[http://dx.doi.org/10.1023/A:1025861522935] [PMID: 9261669]
[133]
Obara T, Ishikuro M, Tamiya G, et al. Potential identification of vitamin B6 responsiveness in autism spectrum disorder utilizing phenotype variables and machine learning methods. Sci Rep 2018; 8(1): 14840.
[http://dx.doi.org/10.1038/s41598-018-33110-w] [PMID: 30287864]
[134]
Dolske MC, Spollen J, McKay S, Lancashire E, Tolbert L. A preliminary trial of ascorbic acid as supplemental therapy for autism. Prog Neuropsychopharmacol Biol Psychiatry 1993; 17(5): 765-74.
[http://dx.doi.org/10.1016/0278-5846(93)90058-Z] [PMID: 8255984]
[135]
Frustaci A, Neri M, Cesario A, et al. Oxidative stress-related biomarkers in autism: systematic review and meta-analyses. Free Radic Biol Med 2012; 52(10): 2128-41.
[http://dx.doi.org/10.1016/j.freeradbiomed.2012.03.011] [PMID: 22542447]
[136]
Frye RE, Huffman LC, Elliott GR. Tetrahydrobiopterin as a novel therapeutic intervention for autism. Neurotherapeutics 2010; 7(3): 241-9.
[http://dx.doi.org/10.1016/j.nurt.2010.05.004] [PMID: 20643376]
[137]
Danfors T, von Knorring AL, Hartvig P, et al. Tetrahydrobiopterin in the treatment of children with autistic disorder: a double-blind placebo-controlled crossover study. J Clin Psychopharmacol 2005; 25(5): 485-9.
[http://dx.doi.org/10.1097/01.jcp.0000177667.35016.e9] [PMID: 16160627]
[138]
Naruse H, Takesada M, Nakane Y, et al. Clinical evaluation of R-tetrahydrobiopterin (SUN 0588) on infantile autism: a double-blind comparative study using placebo as a control. Rinsho Iyaku 1990; 6: 1343-68.
[139]
Klaiman C, Huffman L, Masaki L, Elliott GR. Tetrahydrobiopterin as a treatment for autism spectrum disorders: a double-blind, placebo-controlled trial. J Child Adolesc Psychopharmacol 2013; 23(5): 320-8.
[http://dx.doi.org/10.1089/cap.2012.0127] [PMID: 23782126]
[140]
Frye RE, DeLatorre R, Taylor HB, et al. Metabolic effects of sapropterin treatment in autism spectrum disorder: a preliminary study. Transl Psychiatry 2013; 3 e237
[http://dx.doi.org/10.1038/tp.2013.14] [PMID: 23462988]
[141]
Delhey LM, Tippett M, Rose S, et al. Comparison of Treatment for metabolic disorders associated with autism:reanalysis of three clinical trials. Front Neurosci 2018; 12: 19.
[http://dx.doi.org/10.3389/fnins.2018.00019] [PMID: 29483858]
[142]
Vargason T, Kruger U, Roth E, et al. Comparison of three clinical trial treatments for autism spectrum disorder through multivariate analysis of changes in metabolic profiles and adaptive behavior. Front Cell Neurosci 2018; 12: 503.
[http://dx.doi.org/10.3389/fncel.2018.00503] [PMID: 30618645]
[143]
Deepmala D, Slattery J, Kumar N, et al. Clinical trials of N-acetylcysteine in psychiatry and neurology: a systematic review. Neurosci Biobehav Rev 2015; 55: 294-321.
[http://dx.doi.org/10.1016/j.neubiorev.2015.04.015] [PMID: 25957927]
[144]
Hardan AY, Fung LK, Libove RA, et al. A randomized controlled pilot trial of oral N-acetylcysteine in children with autism. Biol Psychiatry 2012; 71(11): 956-61.
[http://dx.doi.org/10.1016/j.biopsych.2012.01.014] [PMID: 22342106]
[145]
Dean OM, Gray KM, Villagonzalo KA, et al. A randomised, double blind, placebo-controlled trial of a fixed dose of N-acetyl cysteine in children with autistic disorder. Aust N Z J Psychiatry 2017; 51(3): 241-9.
[http://dx.doi.org/10.1177/0004867416652735] [PMID: 27316706]
[146]
Wink LK, Adams R, Wang Z, et al. A randomized placebo-controlled pilot study of N-acetylcysteine in youth with autism spectrum disorder. Mol Autism 2016; 7(7): 26.
[http://dx.doi.org/10.1186/s13229-016-0088-6] [PMID: 27103982]
[147]
Ghanizadeh A, Moghimi-Sarani E. A randomized double blind placebo controlled clinical trial of N-Acetylcysteine added to risperidone for treating autistic disorders. BMC Psychiatry 2013; 13: 196.
[http://dx.doi.org/10.1186/1471-244X-13-196] [PMID: 23886027]
[148]
Mazahery H, Stonehouse W, Delshad M, et al. Relationship between long chain n-3 polyunsaturated fatty acids and autism spectrum disorder: systematic review and meta- analysis of case-control and randomised controlled trials. Nutrients 2017; 9(2) E155
[http://dx.doi.org/10.3390/nu9020155] [PMID: 28218722]
[149]
Layé S, Nadjar A, Joffre C, Bazinet RP. Antiinflammatory effects of omega-3 fatty acids in the brain: physiological mechanisms and relevance to pharmacology. Pharmacol Rev 2018; 70(1): 12-38.
[http://dx.doi.org/10.1124/pr.117.014092] [PMID: 29217656]
[150]
Madore C, Leyrolle Q, Lacabanne C, et al. Neuroinflammation in autism: plausible role of maternal inflammation, dietary omega 3, and microbiota. Neural Plast 2016; 2016 3597209
[http://dx.doi.org/10.1155/2016/3597209] [PMID: 27840741]
[151]
Posar A, Visconti P. Omega-3 supplementation in autism spectrum disorders: a still open question? J Pediatr Neurosci 2016; 11(3): 225-7.
[http://dx.doi.org/10.4103/1817-1745.193363] [PMID: 27857792]
[152]
Farjadian S, Moghtaderi M, Kalani M, Gholami T, Hosseini Teshnizi S. Effects of omega-3 fatty acids on serum levels of T-helper cytokines in children with asthma. Cytokine 2016; 85: 61-6.
[http://dx.doi.org/10.1016/j.cyto.2016.06.002] [PMID: 27288633]
[153]
Brigandi SA, Shao H, Qian SY, et al. Autistic children exhibit decreased levels of essential fatty acids in red blood cells. Int J Mol Sci 2015; 16(5): 10061-76.
[http://dx.doi.org/10.3390/ijms160510061]
[154]
Amminger GP, Berger GE, Schäfer MR, Klier C, Friedrich MH, Feucht M. Omega-3 fatty acids supplementation in children with autism: a double-blind randomized, placebo-controlled pilot study. Biol Psychiatry 2007; 61(4): 551-3.
[http://dx.doi.org/10.1016/j.biopsych.2006.05.007] [PMID: 16920077]
[155]
Yui K, Koshiba M, Nakamura S, Kobayashi Y. Effects of large doses of arachidonic acid added to docosahexaenoic acid on social impairment in individuals with autism spectrum disorders: a double-blind, placebo-controlled, randomized trial. J Clin Psychopharmacol 2012; 32(2): 200-6.
[http://dx.doi.org/10.1097/JCP.0b013e3182485791] [PMID: 22370992]
[156]
Bent S, Bertoglio K, Ashwood P, Bostrom A, Hendren RL. A pilot randomized controlled trial of omega-3 fatty acids for autism spectrum disorder. J Autism Dev Disord 2011; 41(5): 545-54.
[http://dx.doi.org/10.1007/s10803-010-1078-8] [PMID: 20683766]
[157]
Bent S, Hendren RL, Zandi T, et al. Internet-based, randomized, controlled trial of omega-3 fatty acids for hyperactivity in autism. J Am Acad Child Adolesc Psychiatry 2014; 53(6): 658-66.
[http://dx.doi.org/10.1016/j.jaac.2014.01.018] [PMID: 24839884]
[158]
Voigt RG, Mellon MW, Katusic SK, et al. Dietary docosahexaenoic acid supplementation in children with autism. J Pediatr Gastroenterol Nutr 2014; 58(6): 715-22.
[PMID: 24345834]
[159]
Mankad D, Dupuis A, Smile S, et al. A randomized, placebo controlled trial of omega-3 fatty acids in the treatment of young children with autism. Mol Autism 2015; 6(1): 18.
[http://dx.doi.org/10.1186/s13229-015-0010-7] [PMID: 25798215]
[160]
Keim SA, Gracious B, Boone KM, et al. ω-3 and ω-6 fatty acid supplementation may reduce autism symptoms based on parent report in preterm toddlers. J Nutr 2018; 148(2): 227-35.
[http://dx.doi.org/10.1093/jn/nxx047] [PMID: 29490101]
[161]
Cheng YS, Tseng PT, Chen YW, et al. Supplementation of omega 3 fatty acids may improve hyperactivity, lethargy, and stereotypy in children with autism spectrum disorders: a meta-analysis of randomized controlled trials. Neuropsychiatr Dis Treat 2017; 13(13): 2531-43.
[http://dx.doi.org/10.2147/NDT.S147305] [PMID: 29042783]
[162]
Gogou M, Kolios G. The effect of dietary supplements on clinical aspects of autism spectrum disorder: a systematic review of the literature. Brain Dev 2017; 39(8): 656-64.
[http://dx.doi.org/10.1016/j.braindev.2017.03.029] [PMID: 28438367]
[163]
Li YJ, Ou JJ, Li YM, Xiang DX. Dietary supplement for core symptoms of autism spectrum disorder: where are we now and where should we go? Front Psychiatry 2017; 8: 155.
[http://dx.doi.org/10.3389/fpsyt.2017.00155] [PMID: 28878697]
[164]
Huang Y-N, Ho Y-J, Lai C-C, Chiu CT, Wang JY. 1,25-Dihydroxyvitamin D3 attenuates endotoxin-induced production of inflammatory mediators by inhibiting MAPK activation in primary cortical neuron-glia cultures. J Neuroinflammation 2015; 12(1): 147.
[http://dx.doi.org/10.1186/s12974-015-0370-0] [PMID: 26259787]
[165]
Patrick RP, Ames BN. Vitamin D hormone regulates serotonin synthesis. Part 1: relevance for autism. FASEB J 2014; 28(6): 2398-413.
[http://dx.doi.org/10.1096/fj.13-246546] [PMID: 24558199]
[166]
Schmidt RJ, Niu Q, Eyles DW, Hansen RL, Iosif AM. Neonatal vitamin D status in relation to autism spectrum disorder and developmental delay in the CHARGE case-control study. Autism Res 2019; 12(6): 976-88.
[http://dx.doi.org/10.1002/aur.2118] [PMID: 31094097]
[167]
Mazahery H, Conlon CA, Beck KL, et al. A randomised-controlled trial of vitamin D and omega-3 long chain polyunsaturated fatty acids in the treatment of core symptoms of autism spectrum disorder in children. J Autism Dev Disord 2019; 49(5): 1778-94.
[http://dx.doi.org/10.1007/s10803-018-3860-y] [PMID: 30607782]
[168]
Kerley CP, Power C, Gallagher L, Coghlan D. Lack of effect of vitamin D3 supplementation in autism: a 20-week, placebo-controlled RCT. Arch Dis Child 2017; 102(11): 1030-6.
[http://dx.doi.org/10.1136/archdischild-2017-312783] [PMID: 28626020]
[169]
Hajizadeh-Zaker R, Ghajar A, Mesgarpour B, Afarideh M, Mohammadi MR, Akhondzadeh S. L-carnosine as an adjunctive therapy to risperidone in children with autistic disorder: a randomized, double-blind, placebo- controlled trial. J Child Adolesc Psychopharmacol 2018; 28(1): 74-81.
[http://dx.doi.org/10.1089/cap.2017.0026] [PMID: 29027815]
[170]
Chez MG, Buchanan CP, Aimonovitch MC, et al. Double-blind, placebo-controlled study of L-carnosine supplementation in children with autistic spectrum disorders. J Child Neurol 2002; 17(11): 833-7.
[http://dx.doi.org/10.1177/08830738020170111501] [PMID: 12585724]
[171]
Kang DW, Adams JB, Coleman DM, et al. Long-term benefit of microbiota transfer therapy on autism symptoms and gut microbiota. Sci Rep 2019; 9(1): 5821.
[http://dx.doi.org/10.1038/s41598-019-42183-0] [PMID: 30967657]
[172]
Parker KJ, Oztan O, Libove RA, et al. Intranasal oxytocin treatment for social deficits and biomarkers of response in children with autism. Proc Natl Acad Sci USA 2017; 114(30): 8119-24.
[http://dx.doi.org/10.1073/pnas.1705521114] [PMID: 28696286]
[173]
Caldwell HK, Aulino EA, Freeman AR, Miller TV, Witchey SK. Oxytocin and behavior: lessons from knockout mice. Dev Neurobiol 2017; 77(2): 190-201.
[http://dx.doi.org/10.1002/dneu.22431] [PMID: 27513442]
[174]
Yatawara CJ, Einfeld SL, Hickie IB, Davenport TA, Guastella AJ. The effect of oxytocin nasal spray on social interaction deficits observed in young children with autism: a randomized clinical crossover trial. Mol Psychiatry 2016; 21(9): 1225-31.
[http://dx.doi.org/10.1038/mp.2015.162] [PMID: 26503762]
[175]
Anagnostou E, Soorya L, Chaplin W, et al. Intranasal oxytocin versus placebo in the treatment of adults with autism spectrum disorders: a randomized controlled trial. Mol Autism 2012; 3(1): 16.
[http://dx.doi.org/10.1186/2040-2392-3-16] [PMID: 23216716]
[176]
Yamasue H, Okada T, Munesue T, et al. Effect of intranasal oxytocin on the core social symptoms of autism spectrum disorder: a randomized clinical trial. Mol Psychiatry 2018.
[http://dx.doi.org/10.1038/s41380-018-0097-2] [PMID: 29955161]
[177]
Guastella AJ, Gray KM, Rinehart NJ, et al. The effects of a course of intranasal oxytocin on social behaviors in youth diagnosed with autism spectrum disorders: a randomized controlled trial. J Child Psychol Psychiatry 2015; 56(4): 444-52.
[http://dx.doi.org/10.1111/jcpp.12305] [PMID: 25087908]
[178]
Quintana DS, Westlye LT, Hope S, et al. Dose-dependent social-cognitive effects of intranasal oxytocin delivered with novel breath powered device in adults with autism spectrum disorder: a randomized placebo-controlled double-blind crossover trial. Transl Psychiatry 2017; 7(5) e1136
[http://dx.doi.org/10.1038/tp.2017.103] [PMID: 28534875]
[179]
Dadds MR, MacDonald E, Cauchi A, Williams K, Levy F, Brennan J. Nasal oxytocin for social deficits in childhood autism: a randomized controlled trial. J Autism Dev Disord 2014; 44(3): 521-31.
[http://dx.doi.org/10.1007/s10803-013-1899-3] [PMID: 23888359]
[180]
Cortesi F, Giannotti F, Sebastiani T, Panunzi S, Valente D. Controlled-release melatonin, singly and combined with cognitive behavioural therapy, for persistent insomnia in children with autism spectrum disorders: a randomized placebo-controlled trial. J Sleep Res 2012; 21(6): 700-9.
[http://dx.doi.org/10.1111/j.1365-2869.2012.01021.x] [PMID: 22616853]
[181]
Wright B, Sims D, Smart S, et al. Melatonin versus placebo in children with autism spectrum conditions and severe sleep problems not amenable to behaviour management strategies: a randomised controlled crossover trial. J Autism Dev Disord 2011; 41(2): 175-84.
[http://dx.doi.org/10.1007/s10803-010-1036-5] [PMID: 20535539]
[182]
Garstang J, Wallis M. Randomized controlled trial of melatonin for children with autistic spectrum disorders and sleep problems. Child Care Health Dev 2006; 32(5): 585-9.
[http://dx.doi.org/10.1111/j.1365-2214.2006.00616.x] [PMID: 16919138]
[183]
Wirojanan J, Jacquemont S, Diaz R, et al. The efficacy of melatonin for sleep problems in children with autism, fragile X syndrome, or autism and fragile X syndrome. J Clin Sleep Med 2009; 5(2): 145-50.
[PMID: 19968048]
[184]
Wasdell MB, Jan JE, Bomben MM, et al. A randomized, placebo-controlled trial of controlled release melatonin treatment of delayed sleep phase syndrome and impaired sleep maintenance in children with neurodevelopmental disabilities. J Pineal Res 2008; 44(1): 57-64.
[PMID: 18078449]
[185]
Gagnon K, Godbout R. Melatonin and comorbidities in children with autism spectrum disorder. Curr Dev Disord Rep 2018; 5(3): 197-206.
[http://dx.doi.org/10.1007/s40474-018-0147-0] [PMID: 30148039]
[186]
Gottfried C, Bambini-Junior V, Francis F, Riesgo R, Savino W. The impact of neuroimmune alterations in autism spectrum disorder. Front Psychiatry 2015; 6: 121.
[http://dx.doi.org/10.3389/fpsyt.2015.00121] [PMID: 26441683]
[187]
Meltzer A, Van de Water J. The role of the immune system in autism spectrum disorder. Neuropsychopharmacology 2017; 42(1): 284-98.
[http://dx.doi.org/10.1038/npp.2016.158] [PMID: 27534269]
[188]
Akhondzadeh S, Fallah J, Mohammadi M-R, et al. Double-blind placebo-controlled trial of pentoxifylline added to risperidone: effects on aberrant behavior in children with autism. Prog Neuropsychopharmacol Biol Psychiatry 2010; 34(1): 32-6.
[http://dx.doi.org/10.1016/j.pnpbp.2009.09.012] [PMID: 19772883]
[189]
Singh K, Connors SL, Macklin EA, et al. Sulforaphane treatment of autism spectrum disorder (ASD). Proc Natl Acad Sci USA 2014; 111(43): 15550-5.
[http://dx.doi.org/10.1073/pnas.1416940111] [PMID: 25313065]
[190]
Sedlak TW, Nucifora LG, Koga M, et al. Sulforaphane Augments glutathione and influences brain metabolites in human subjects: a clinical pilot study. Mol Neuropsychiatry 2018; 3(4): 214-22.
[http://dx.doi.org/10.1159/000487639] [PMID: 29888232]
[191]
Egner PA, Chen JG, Zarth AT, et al. Rapid and sustainable detoxication of airborne pollutants by broccoli sprout beverage: results of a randomized clinical trial in China. Cancer Prev Res (Phila) 2014; 7(8): 813-23.
[http://dx.doi.org/10.1158/1940-6207.CAPR-14-0103] [PMID: 24913818]
[192]
Zimmerman A, Diggins E, Connors S, Singh K. Sulforaphane treatment of children with autism spectrum disorder (ASD) - a progress report (N1.002). Neurology 2018; 90(Suppl. 15).
[193]
Sareddy GR, Geeviman K, Ramulu C, Babu PP. The nonsteroidal anti-inflammatory drug celecoxib suppresses the growth and induces apoptosis of human glioblastoma cells via the NF-κB pathway. J Neurooncol 2012; 106(1): 99-109.
[http://dx.doi.org/10.1007/s11060-011-0662-x] [PMID: 21847707]
[194]
Asadabadi M, Mohammadi M-R, Ghanizadeh A, et al. Celecoxib as adjunctive treatment to risperidone in children with autistic disorder: a randomized, double-blind, placebo-controlled trial. Psychopharmacology (Berl) 2013; 225(1): 51-9.
[http://dx.doi.org/10.1007/s00213-012-2796-8] [PMID: 22782459]
[195]
Ghaleiha A, Rasa SM, Nikoo M, Farokhnia M, Mohammadi MR, Akhondzadeh S. A pilot double-blind placebo-controlled trial of pioglitazone as adjunctive treatment to risperidone: effects on aberrant behavior in children with autism. Psychiatry Res 2015; 229(1-2): 181-7.
[http://dx.doi.org/10.1016/j.psychres.2015.07.043] [PMID: 26208985]
[196]
Ghaleiha A, Alikhani R, Kazemi M-R, et al. Minocycline as adjunctive treatment to risperidone in children with autistic disorder: a randomized, double-blind placebo-controlled trial. J Child Adolesc Psychopharmacol 2016; 26(9): 784-91.
[http://dx.doi.org/10.1089/cap.2015.0175] [PMID: 27128958]
[197]
Buitelaar JK, Dekker MEM, van Ree JM, van Engeland H. A controlled trial with ORG 2766, an ACTH-(4-9) analog, in 50 relatively able children with autism. Eur Neuropsychopharmacol 1996; 6(1): 13-9.
[http://dx.doi.org/10.1016/0924-977X(95)00049-U] [PMID: 8866933]
[198]
Niederhofer H, Staffen W, Mair A. Immunoglobulins as an alternative strategy of psychopharmacological treatment of children with autistic disorder. Neuropsychopharmacology 2003; 28(5): 1014-5.
[http://dx.doi.org/10.1038/sj.npp.1300130] [PMID: 12700706]
[199]
Boris M, Goldblatt A, Edelson S. Improvement in children with autism treated with intravenous gamma globulin. J Nutr Environ Med 2005; 15(4): 169-76.
[http://dx.doi.org/10.1080/13590840600681827]
[200]
Connery K, Tippett M, Delhey LM, et al. Intravenous immunoglobulin for the treatment of autoimmune encephalopathy in children with autism. Transl Psychiatry 2018; 8(1): 148.
[http://dx.doi.org/10.1038/s41398-018-0214-7] [PMID: 30097568]
[201]
Melamed IR, Heffron M, Testori A, Lipe K. A pilot study of high-dose intravenous immunoglobulin 5% for autism: impact on autism spectrum and markers of neuroinflammation. Autism Res 2018; 11(3): 421-33.
[http://dx.doi.org/10.1002/aur.1906] [PMID: 29427532]
[202]
Handen BL, Melmed RD, Hansen RL, et al. A double-blind, placebo-controlled trial of oral human immunoglobulin for gastrointestinal dysfunction in children with autistic disorder. J Autism Dev Disord 2009; 39(5): 796-805.
[http://dx.doi.org/10.1007/s10803-008-0687-y] [PMID: 19148734]
[203]
Naviaux RK, Zolkipli Z, Wang L, et al. Antipurinergic therapy corrects the autism-like features in the poly(IC) mouse model. PLoS One 2013; 8(3) e57380
[http://dx.doi.org/10.1371/journal.pone.0057380] [PMID: 23516405]
[204]
Ginsberg MR, Rubin RA, Falcone T, Ting AH, Natowicz MR. Brain transcriptional and epigenetic associations with autism. PLoS One 2012; 7(9) e44736
[http://dx.doi.org/10.1371/journal.pone.0044736] [PMID: 22984548]
[205]
Naviaux RK, Curtis B, Li K, et al. Low-dose suramin in autism spectrum disorder: a small, phase I/II, randomized clinical trial. Ann Clin Transl Neurol 2017; 4(7): 491-505.
[http://dx.doi.org/10.1002/acn3.424] [PMID: 28695149]
[206]
Gogou M, Evangeliou A. Is metabolic screening necessary in children with autism spectrum disorder? A mini review. J Pediatr Neurol 17(06): 199-205.
[http://dx.doi.org/10.1055/s-0038-1676633]
[207]
Smith AM, King JJ, West PR, et al. Amino acid dysregulation metabotypes: potential biomarkers for diagnosis and individualized treatment for subtypes of autism spectrum disorder. Biol Psychiatry 2019; 85(4): 345-54.
[http://dx.doi.org/10.1016/j.biopsych.2018.08.016] [PMID: 30446206]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy