Medical & Scientific Literature

Acupuncture

Sources & References

Bower, W.F., et al. Acupuncture as a treatment for nocturnal enuresis. Auton Neurosci. 2010 Oct 28;157(1-2):63-7.

Jindal, V., et al. Safety and efficacy of acupuncture in children: a review of the evidence. J Pediatr Hematol Oncol. 2008 Jun;30(6):431-42.

Libonate, J., et al. Efficacy of acupuncture for health conditions in children: a review. ScientificWorldJournal. 2008 Jul 13;8:670-82.

Lin, Y.C., et al. Acupuncture for pediatric pain: The trend of evidence-based research. J Tradit Complement Med. 2019 Aug 17;10(4):315-319.

Yang, C., et al. Efficacy and safety of acupuncture in children: an overview of systematic reviews. Pediatr Res. 2015 Aug;78(2):112-9.

ADD/ADHD

Sources & References

Adams, L.J., et al. Infant feeding method and special educational need in 191,745 Scottish schoolchildren: A national, population cohort study. PLoS Med. 2023 Apr 6;20(4):e1004191.

Akinbami, L.J., et al. Attention deficit hyperactivity disorder among children aged 5-17 years in the United States, 1998-2009. NCHS Data Brief. 2011(70):1-8. 

Alemany, S., et al. Prenatal and postnatal exposure to acetaminophen in relation to autism spectrum and attention-deficit and hyperactivity symptoms in childhood: Meta-analysis in six European population-based cohorts. Eur J Epidemiol. 2021 Oct;36(10):993-1004.

Archer, T., et al. Physical exercise alleviates ADHD symptoms: regional deficits and development trajectory. Neurotox Res. 2012;21(2):195-209.

Arnold, L.E., et al. Does hair zinc predict amphetamine improvement of ADD/hyperactivity? Int J Neurosci. 1990;50:103-107.

Avella-Garcia, C.B., et al. Acetaminophen use in pregnancy and neurodevelopment: attention function and autism spectrum symptoms. Int J Epidemiol. 2016 Dec 1;45(6):1987-1996.

Baker, B.H., et al. Association of Prenatal Acetaminophen Exposure Measured in Meconium With Risk of Attention-Deficit/Hyperactivity Disorder Mediated by Frontoparietal Network Brain Connectivity. JAMA Pediatrics. 2020 Sep 28;e203080.

Bartscherer, et al. Interactive metronome training for a 9-year-old boy with attention and motor coordination difficulties. Physiother Theory Pract. Oct-Dec 2005;21(4):257-69.

Bateman, B., et al. The effects of a double blind, placebo controlled, artificial food colourings and benzoate preservative challenge on hyperactivity in a general population sample of preschool children. Arch Dis Child. 2004 Jun;89(6):506-11.

Bauer, A.Z., et al. Prenatal paracetamol exposure and child neurodevelopment: A review. Horm Behav. 2018 May:101:125-147.

Bauer, J., et al. Hyperactivity and impulsivity in adult attention-deficit/hyperactivity disorder is related to glutamatergic dysfunction in the anterior cingulate cortex. World J Biol Psychiatry. 2018 Oct;19(7):538-546.

Bekaroğlu, M., et al. Relationships between serum free fatty acids and zinc, and attention deficit hyperactivity disorder: a research note. J Child Psychol Psychiatry. 1996;37:225-227.

Bennings, M.A., et al. Colonic transit times and behaviour profiles in children with defecation disorders. Archives of the Diseases of Childhood. 2004 Jan;89(1):13-6.

Bernfort, L., et al. ADHD from a socio-economic perspective. Acta Paediatr. 2008;97(2):239-45

Bertelsen, E.N., et al. Childhood Epilepsy, Febrile Seizures, and Subsequent Risk of ADHD. Pediatrics. 2016 Aug;138(2). pii: e20154654.

Blondis, T.A. Motor disorders and attention-deficit/hyperactivity disorder. Pediatr Clin North Am. 1999 Oct;46(5):899-913, vi-vii.

Blum, K., et al. Attention-deficit-hyperactivity disorder and reward deficiency syndrome. Neuropsychiatric Disease and Treatment. 2008 Oct;4(5):893-918.

Boat, T.F., et al. Prevalence of Learning Disabilities. Mental Disorders and Disabilities Among Low-Income Children. Washington (DC): National Academies Press (US); 2015 Oct 28. 16.

Bondopadhyay, U., et al. The Role of the Circadian System in Attention Deficit Hyperactivity Disorder. Adv Exp Med Biol. 2021:1344:113-127.

Boris, M.J., et al. Pollen Exposure as a Cause for the Deterioration of Neurobehavioral Function in Children with Autism and Attention Deficit Hyperactive Disorder. J of Nutritional and Environmental Medicine. Mar 2004;14(1):47 – 54.

Borre, Y.E., et al. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med. 2014 Sep;20(9):509-18.

Bradstreet, J.J., et al. Biomarker-guided interventions of clinically relevant conditions associated with autism spectrum disorders and attention deficit hyperactivity disorder. Altern Med Rev. 2010 Mar;15(1):15-32.

Braun, J.M., et al. Association of Environmental Toxicants and Conduct Disorder in U.S. Children: NHANES 2001-2004. Environ Health Perspect. 2008 Jul;116(7):956-62.

Braun, J.M., et al. Exposures to environmental toxicants and attention deficit hyperactivity disorder in U.S. children. Environ Health Project. Dec 2006;114(12):1904 – 1909.

Centers for Disease Control and Prevention. Data and Statistics on Children's Mental Health. Accessed 2 Sep 2020.

Ceylan, M.F., et al. Changes in oxidative stress and cellular immunity serum markers in attention-deficit/hyperactivity disorder. Psychiatry Clin Neurosci. 2012;66(3):220-6.

Coogan, A.N., et al. A systematic review of circadian function, chronotype and chronotherapy in attention deficit hyperactivity disorder. Atten Defic Hyperact Disord. 2017 Sep;9(3):129-147.

Cosper, S.M., et al. Interactive Metronome training in children with attention deficit and developmental coordination disorders. Int J Rehabil Res. 2009 Dec;32(4):331-6.

Cortese, S., et al. Association between attention deficit hyperactivity disorder and asthma: a systematic review and meta-analysis and a Swedish population-based study. Lancet Psychiatry. 2018 Sep;5(9):717-726.

Cortese, S., et al. Attention-deficit/hyperactivity disorder, iron deficiency, and obesity: is there a link? Postgrad Med. 2014;126(4):155-70.

Curran, E.A., et al. Research review: Birth by caesarean section and development of autism spectrum disorder and attention-deficit/hyperactivity disorder: a systematic review and meta-analysis. J Child Psychol Psychiatry. 2015 May;56(5):500-8.

Danielson, M.L., et al. ADHD Prevalence Among U.S. Children and Adolescents in 2022: Diagnosis, Severity, Co-Occurring Disorders, and Treatment. J Clin Child Adolesc Psychol. 2024 May 22:1-18.

Darling, A.L., et al. Association between maternal vitamin D status in pregnancy and neurodevelopmental outcomes in childhood: results from the Avon Longitudinal Study of Parents and Children (ALSPAC). Br J Nutr 2017 Jun;117(12):1682-1692.

Ende, G., et al. Impulsivity and Aggression in Female BPD and ADHD Patients: Association with ACC Glutamate and GABA Concentrations. Neuropsychopharmacology. 2016 Jan;41(2):410-8.

Eppright, T.D., et al. Attention deficit hyperactivity disorder, infantile autism, and elevated blood-lead: a possible relationship. Mo Med. 1996;93(3):136-8).

Erskine, H.E., et al. The global burden of conduct disorder and attention-deficit/hyperactivity disorder in 2010. J Child Psychol Psychiatry. 2014;55(4):328-36.

Egset, K., et al. Magno App: Exploring Visual Processing in Adults with High and Low Reading Competence. Scandinavian Journal of Educational Research. 07 Jan 2020.

Fargason, R.E., et al. Correcting delayed circadian phase with bright light therapy predicts improvement in ADHD symptoms: A pilot study. J Psychiatr Res. 2017 Aug:91:105-110.

Fletcher, J., National Bureau of Economic Research. The effects of childhood ADHD on adult labor market outcomes. Cambridge, MA: National Bureau of Economic Research; 2013. 25 p.

Goulardins, J.B., et al. Attention deficit hyperactivity disorder and developmental coordination disorder: Two separate disorders or do they share a common etiology. Behav Brain Res. 2015 Oct 1;292:484-92.

Goulardins, J.B., et al. Attention Deficit Hyperactivity Disorder and Motor Impairment. Percept Mot Skills. 2017 Apr;124(2):425-440.

Greenblatt, J. Nutritional supplements in ADHD. J Am Acad Child Adolesc Psychiatry;38(10):1209-1210

Grigg. T.M., et al. Primitive Reflex Integration and Reading Achievement in the Classroom. Journal of Neurology and Experimental Neuroscience. 2023; 9(1), 18-26.

Joinson, C., et al. Psychological Difference Between Children With and Without Soiling Problems. Pediatrics. 2006 May;117(5):1575-84.

Hertz-Picciotto, I., et al. Organophosphate exposures during pregnancy and child neurodevelopment: Recommendations for essential policy reforms. PLoS Med. 2018 Oct 24;15(10):e1002671.

Hodgkins, P., et al. Risk of injury associated with attentiondeficit/ hyperactivity disorder in adults enrolled in employer-sponsored health plans: a retrospective analysis. Prim Care Companion CNS Disord. 2011;13(2).

Ibilola, O., et al. Environmental (Perinatal) risk factors of ADHD in a sibling control design study. Open Access Journal of Behavioural Science & Psychology. 2021 Feb 05;4(1).

Ji, Y. Association of Cord Plasma Biomarkers of In Utero Acetaminophen Exposure With Risk of Attention-Deficit/Hyperactivity Disorder and Autism Spectrum Disorder in Childhood. JAMA Psychiatry. 2020 Feb 1;77(2):180-189.

Karatekin, C., et al. A preliminary study of motor problems in children with attention-deficit/hyperactivity disorder. Percept Mot Skills. 2003 Dec;97(3 Pt 2):1267-80.

Kawatani, M., et al. Evaluation of oxidative stress status in children with pervasive developmental disorder and attention deficit hyperactivity disorder using urinaryspecific biomarkers. Redox Rep. 2011;16(1):45-6.

Konofal, E., et al. Effects of iron supplementation on attention deficit hyperactivity disorder in children. Pediatr Neurol. Jan 2008;38(1):20-6.

Korman, M., et al. ADHD 24/7: Circadian clock genes, chronotherapy and sleep/wake cycle insufficiencies in ADHD. World J Biol Psychiatry. 2020 Mar;21(3):156-171.

Kozielec, T., et al. Assessment of magnesium levels in children with attention deficit hyperactivity disorder (ADHD). Magnes Res. 1997;10(2):143-8.

Kurth, L., et al. Perinatal Pitocin as an early ADHD biomarker: neurodevelopmental risk? J Atten Disord. 2011 Jul;15(5):423-31.

Liao, T.C., et al. Comorbidity of Atopic Disorders with Autism Spectrum Disorder and Attention Deficit/Hyperactivity Disorder.  J Pediatr. 2016 Apr;171:248-55.

Liew, Z., et al. Use of Negative Control Exposure Analysis to Evaluate Confounding: An Example of Acetaminophen Exposure and Attention-Deficit/Hyperactivity Disorder in Nurses’ Health Study II. Am J Epidemiol. 2019 Apr 1;188(4):768-775.

Lyon, M.R., et al. Effect of the herbal extract combination Panax quinquefolium and Ginkgo biloba on attention-deficit hyperactivity disorder: a pilot study. J Psychiatry Neurosci. 2001;26(3):221-228.

Mahmoud, M.M., et al. Zinc, ferritin, magnesium and copper in a group of Egyptian children with attention deficit hyperactivity disorder. Ital J Pediatr. 2011;37:60.

Malin, A.J., et al. Maternal Urinary Fluoride and Child Neurobehavior at Age 36 Months. JAMA Netw Open. 2024 May 1;7(5):e2411987.

McLeod, K.R., et al. Functional connectivity of neural motor networks is disrupted in children with developmental coordination disorder and attention-deficit/hyperactivity disorder. Neuroimage Clin. 2014 Mar 26;4:566-75.

Melillo, R., et al. Persistent Childhood Primitive Reflex Reduction Effects on Cognitive, Sensorimotor, and Academic Performance in ADHD. Front Public Health. 2020 Nov 17;8:431835.

Mercer, J.S., et al. The Effects of Delayed Cord Clamping on 12-Month Brain Myelin Content and Neurodevelopment: A Randomized Controlled Trial. Am J Perinatol. 2022 Jan;39(1):37-44.

Millichap, J.G., et al. The diet factor in attention-deficit/hyperactivity disorder. Pediatrics.2012 Feb;129(2):330-7.

Mitchell, E.A., et al. Clinical characteristics and serum essential fatty acid levels in hyperactive children. Clin Pediatr (Phila). 1987;26:406-411.

Morris, C.R., et al. Syndrome of allergy, apraxia, and malabsorption: characterization of a neurodevelopmental phenotype that responds to omega 3 and vitamin E supplementation. Alternative Therapies in Health and Medicine. Jul-Aug 2009;15(4):34-43.

Mossin, M.H., et al. Inverseassociations between cord vitamin D and attention deficit hyperactivity disorder symptoms: A child cohort study. Aust N Z J Psychiatry. 2017 Jul;51(7):703-710.

Naganuma, F., et al. Histamine N-methyltransferase regulates aggression and the sleep-wake cycle. Sci Rep. 2017 Nov 21;7(1):15899.

Niederhofer, H., et al. A preliminary investigation of ADHD symptoms in persons with celiac disease. J Atten Disorder. 2006 Nov;10(2):200-4.

Ogundele, M.O., et al. Management of sleep disorders among children and adolescents with neurodevelopmental disorders: A practical guide for clinicians. World J Clin Pediatr. 2022 Mar 15;11(3):239-252.

Parker, W., et al. The Dangers of Acetaminophen for Neurodevelopment Outweigh Scant Evidence for Long-Term Benefits. Children (Basel). 2023 Dec 29;11(1):44.

Pelsser, L.M., et al. Effects of a restricted elimination diet on the behaviour of children with attention-deficit hyperactivity disorder (INCA study): a randomised controlled trial. Lancet. 2011 Feb 5;377(9764):494-503.

Peltier, M.R., et al. Maternal Hypothyroidism Increases the Risk of Attention-Deficit Hyperactivity Disorder in the Offspring. Am J Perinat. 2020 Oct 21.

Phillipot, G., et al. Paracetamol (Acetaminophen) and its Effect on the Developing Mouse Brain. Front Toxicol. 2022 Mar 22:4:867748.

Physician’s Postgraduate Press. Managing ADHD in Children, Adolescents, and Adults With Comorbid Anxiety in Primary Care. The Primary Care Companion to The Journal of Clinical Psychiatry. 2007;9(2):129-38.

Piek, J.P., et al. Motor coordination and kinaesthesis in boys with attention deficit-hyperactivity disorder. Dev Med Child Neurol. 1999 Mar;41(3):159-65.

Richardson, A.J., et al. Omega-3 fatty acids in ADHD and related neurodevelopmental disorders. Int Rev Psychiatry. 2006;18(2):155-172).

Rifas-Shiman, S.L., et al. Associations of prenatal or infant exposure to acetaminophen or ibuprofen with mid-childhood executive function and behaviour. Paediatr Perinat Epidemiol. 2020 May;34(3):287-298.

Robinette, L.M., et al. Fruit and vegetable intake is inversely associated with severity of inattention in a pediatric population with ADHD symptoms: the MADDY Study. Nutr Neurosci. 2023 Jun;26(6):572-581.

Sabuncuoglu, O. Understanding the relationships between breastfeeding, malocclusion, ADHD, sleep-disordered breathing and traumatic dental injuries. Med Hypotheses. 2013;80(3):315-20

Scassellati, C., et al. Biomarkers and attention deficit/hyperactivity disorder: a systematic review and meta-analyses. J Am Acad Child Adolesc Psychiatry. 2012;51(10):1003-19 e20.

Schab, D.W., et al. Do artificial food colors promote hyperactivity in children with hyperactive syndromes? A meta-analysis of double-blind placebo-controlled trials. J Dev Behav Pediatr. 2004 Dec;25(6):423-34.

Schmitt, J., et al. Atopic eczema and attention-deficit/hyperactivity disorder in a population-based sample of children and adolescents. JAMA. 2009 Feb 18;301(7):724-6.

Shaffer, R.J., et al. Effect of interactive metronome training on children with ADHD. Am J Occup Ther. Mar-Apr 2001;55(2):155-62.

Slater, J.L., et al. Timing Deficits in ADHD: Insights From the Neuroscience of Musical Rhythm. Front Comput Neurosci. 2018 Jul 6;12:51.

Smith, A., et al. Evidence for a pure time perception deficit in children with ADHD. J Child Psychol Psychiatry. 2002 May;43(4):529-42.

Sonuga-Barke, E., et al. Beyond the dual pathway model: evidence for the dissociation of timing, inhibitory, and delay-related impairments in attention-deficit/hyperactivity disorder. J Am Acad Child Adolesc Psychiatry. 2010 Apr;49(4):345-55.

Sonuga-Barke, E.J.S., et al. Nonpharmacological interventions for ADHD: systematic review and meta-analyses of randomized controlled trials of dietary and psychological treatments. Am J Psychiatry. 2013 Mar;170(3):275-89.

Sucksdorff, M., et al. Maternal Vitamin D Levels and the Risk of Offspring Attention-Deficit/Hyperactivity Disorder. J Am Acad Child Adolesc Psychiatry. 2019 Dec 18.

Swanson, J.M., et al. Etiologic subtypes of attention-deficit/hyperactivity disorder: brain imaging, molecular genetic and environmental factors and the dopamine hypothesis. Neuropsychology Review. 2007 Mar;17(1):39-59.

Taige, N.M., et al. Gestational Age at Term, Delivery Circumstance, and Their Association with Childhood Attention Deficit Hyperactivity Disorder Symptoms. Paediatr Perinat Epidemiol. 2016 Mar;30(2):171-80.

Teicher, M.H. Final White Paper: Effects of Brain Balance Exercises and Interactive Metronome on Children with Attention Deficit Hyperactivity Disorderare Similar to the Effects of Stimulant Medication. Harvard Medical School, Department of Psychiatry. 2020.

Telford, C., et al. Estimating the costs of ongoing care for adolescents with attention-deficit hyperactivity disorder. Soc Psychiatry Psychiatr Epidemiol. 2013;48(2):337-44.

Toplak, M.E., et al. Time perception: modality and duration effects in attention-deficit/hyperactivity disorder (ADHD). J Abnorm Child Psychol. 2005 Oct;33(5):639-54.

van Andel, W., et al. Effects of chronotherapy on circadian rhythm and ADHD symptoms in adults with attention-deficit/hyperactivity disorder and delayed sleep phase syndrome: a randomized clinical trial. Chronobiol Int. 2021 Feb;38(2):260-269.

Verlaet, A.A., et al. Nutrition, immunological mechanisms and dietary immunomodulation in ADHD. Eur Child Adolesc Psychiatry. 2014;23(7):519-29.

Wang, H.L., et al. Case-Control Study of Blood Lead Levels and Attention Deficit Hyperactivity Disorder in Chinese Children. Environmental Health Perspectives. 2008 Oct;116(10):1401-6.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Weiss, M.D., et al. Effectiveness outcomes in attention-deficit/hyperactivity disorder. J Clin Psychiatry. 2006;67 Suppl 8:38-45.

Woodbury, M.L., et al. The relationship of prenatal acetaminophen exposure and attention-related behavior in early childhood. Neurotoxicol Teratol. 2024 Jan-Feb:101:107319.

Yang, B., et al. Time perception deficit in children with ADHD. Brain Res. 2007 Sep 19;1170:90-6.

Zablotsky, B., et al. Prevalence and Trends of Developmental Disabilities among Children in the United States: 2009-2017. Pediatrics. 2019 Oct;144(4):e20190811.

Zaigham, M., et al. Prelabour caesarean section and neurodevelopmental outcome at 4 and 12 months of age: an observational study. BMC Pregnancy and Childbirth. 2020 (20)564.

Zhao, L., et al. Acetaminophen causes neurodevelopmental injury in susceptible babies and children: no valid rationale for controversy. Clin Exp Pediatr. 2024 Mar;67(3):126-139.

Airway Orthodontics

Sources & References

Amdo, T., et al. Somatic syndromes, insomnia, anxiety, and stress among sleep disordered breathing patients. Sleep Breath. 2016 May;20(2):759-68.

Bussi, M.T., et al. Is ankyloglossia associated with obstructive sleep apnea? Braz J Otorhinolaryngol. 2021 Nov 5;S1808-8694(21)00181-6.

de Godoy, L.B., et al. Upper Airway Resistance Syndrome Patients Have Worse Sleep Quality Compared to Mild Obstructive Sleep Apnea. PLoS One. 2016; 11(5): e0156244.

Dieltjens, M., et al. Treatment of obstructive sleep apnea using a custom-made titratable duobloc oral appliance: a prospective clinical study. Sleep Breath. 2013 May;17(2):565-72.

Doff, M.H., et al. Oral Appliance Versus Continuous Positive Airway Pressure in Obstructive Sleep Apnea Syndrome: A 2-year Follow-Up. Sleep. 2013 Sep 1;36(9):1289-96.

Harari, D., et al. The effect of mouth breathing versus nasal breathing on dentofacial and craniofacial development in orthodontic patients. Laryngoscope. 2010 Oct;120(10):2089-93.

Hesselbacher, S., et al. A Study to Assess the Relationship between Attention Deficit Hyperactivity Disorder and Obstructive Sleep Apnea in Adults. Cureus. 2019 Oct 24;11(10):e5979.

Hsu, B., et al. Effects of respiratory muscle therapy on obstructive sleep apnea: a systematic review and meta-analysis. J Clin Sleep Med. 2020 May 15;16(5):785-801.

Huang, Y.S., et al. Attention-deficit/hyperactivity disorder with obstructive sleep apnea: a treatment outcome study. Sleep Med. 2007 Jan;8(1):18-30.

Huang, Y.S., et al. Short Lingual Frenulum and Obstructive Sleep Apnea in Children. Intl Journal of Clinical Pediatrics. 2015,1(1):1-4.

Hvolby, A. Associations of sleep disturbance with ADHD: implications for treatment. Atten Defic Hyperact Disord. 2015 Mar;7(1):1-18.

Kamata, S., et al. Assessment of Obstructive Apnea by Using Polysomnography and Surgical Treatment in Patients With Beckwith-Wiedemann Syndrome. J Pediatr Surg. 2005 Mar;40(3):E17-9.

Martel, J., et al. Could nasal nitric oxide help to mitigate the severity of COVID-19? Microbes Infect. 2020 May-June; 22(4): 168–171.

Felippe, N.L.O., et al. Relationship between rapid maxillary expansion and nasal cavity size and airway resistance: short- and long-term effects. Am J Orthod Dentofacial Orthop. 2008 Sep;134(3):370-82.

Reyes-Zúñiga, M., et al. Anxiety and depression symptoms in patients with sleep-disordered breathing. Open Respir Med J. 2012;6:97-103.

Sedky, K., et al. Attention deficit hyperactivity disorder and sleep disordered breathing in pediatric populations: a meta-analysis. Sleep Med Rev. 2014 Aug;18(4):349-56.

Urbano, G.L., et al. The Link between Pediatric Obstructive Sleep Apnea (OSA) and Attention Deficit Hyperactivity Disorder (ADHD). Children (Basel). 2021 Sep 19;8(9):824.

White, David P., et al. Mandibular Advancement Device vs. CPAP in the Treatment of Obstructive Sleep Apnea: Are They Equally Effective in Short Term Health Outcomes? J Clin Sleep Med. 2013 Sep 15;9(9):971-2.

Xiangming, L., et al. The relationship between inflammation and neurocognitive dysfunction in obstructive sleep apnea syndrome. J Neuroinflammation. 2020 Aug 1;17(1):229.

Yeghiazarians, Y., et al. Obstructive Sleep Apnea and Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation. 2021 Jul 20;144(3):e56-e67.

Youssef, N.A., et al. Is obstructive sleep apnea associated with ADHD? Ann Clin Psychiatry. 2011 Aug;23(3):213-24.

Allergies

Sources & References

Gastrointestinal permeability in food-allergic children. Nutr Rev. 1985 Aug;43(8):233-5.

Allen, K.J., et al. Food Allergy in Childhood. Medical Journal of Australia. 2006 Oct 2;185(7):394-400.

Bunyavanich, S., et al. Peanut allergy prevalence among school-age children in a US cohort not selected for any disease. J Allergy Clin Immunol. 2014;134(3):753-5

Campbell, et al. Mechanisms of Allergic Disease – Environmental and genetic determinants for the development of allergy. Clin Exp Allergy. 2015

Della Giustina, A., et al.. Vitamin D, allergies and asthma: focus on pediatric patients. World Allergy Organ J. 2014;7(1):27

Feehley, T., et al. Healthy infants harbor intestinal bacteria that protect against food allergy. Nature Medicine. 2019 Jan 14.

Gupta, R.S., et al. The Public Health Impact of Parent-Reported Childhood Food Allergies in the United States. Pediatrics. 2018 Dec;142(6). pii: e20181235.

Heuer, L., et al. Reduced levels of immunoglobulin in children with autism correlates with behavioral symptoms. Autism Res, Oct 2008, 1:5, 275–83.

Hoskin-Parr, L., et al. Antibiotic exposure in the first two years of life and development of asthma and other allergic diseases by 7.5 yr: A dose-dependent relationship. Pediatr Allergy Immunol. 2013 Dec; 24(8): 762–771.

Isolauri, E., et al. Food allergy in irritable bowel syndrome: new facts and old fallacies. Gut. 2004 Oct;53(10):1391-3.

Jackson, P.G., et al. Intestinal permeability in patients with eczema and food allergy. Lancet. 1981 Jun 13;1(8233):1285-6.

Jyonouchi, H., et al. Dysregulated innate immune responses in young children with autism spectrum disorders: their relationship to gastrointestinal symptoms and dietary intervention. Neuropsychobiology. 2005;51(2):77-85.

Kim-Lee, C., et al. Gastrointestinal disease in Sjogren's syndrome: related to food hypersensitivities. Springerplus. 2015 Dec 12;4:766.

Ly, N.P., et al. Gut microbiota, probiotics, and vitamin D: interrelated exposures influencing allergy, asthma, and obesity? J Allergy Clin Immunol. 2011;127(5):1087-94; quiz 95-6.

Maksimova, O.V., et al. [Intestine microbiota and allergic diseases]. Zh Mikrobiol Epidemiol Immunobiol. 2014(3):49-60.

Mitre, E., et al. Association Between Use of Acid-Suppressive Medications and Antibiotics During Infancy and Allergic Diseases in Early Childhood. JAMA Pediatr. 2018 Jun 4;172(6):e180315.

Möller, C., et al. Intestinal permeability as assessed with polyethyleneglycols in birch pollen allergic children undergoing oral immunotherapy. Allergy. 1986 May;41(4):280-5.

Morris, C.R., et al. Syndrome of allergy, apraxia, and malabsorption: characterization of a neurodevelopmental phenotype that responds to omega 3 and vitamin E supplementation. Alternative Therapies in Health and Medicine. Jul-Aug 2009;15(4):34-43.

Peters, R.L., et al. Infant food allergy phenotypes and association with lung function deficits and asthma at age 6 years: a population-based, prospective cohort study in Australia. Lancet Child Adolesc Health. 2023 Jul 24;S2352-4642(23)00133-5.

Prescott, S.L. Early-life environmental determinants of allergic diseases and the wider pandemic of inflammatory noncommunicable diseases. J Allergy Clin Immunol. 2013;131(1):23-30.

Rueter, K., et al. In "High-Risk" Infants with Sufficient Vitamin D Status at Birth, Infant Vitamin D Supplementation Had No Effect on Allergy Outcomes: A Randomized Controlled Trial. Nutrients. 2020 Jun 11;12(6):1747.

Severance, E.G., et al. IgG dynamics of dietary antigens point to cerebrospinal fluid barrier or flow dysfunction in first-episode schizophrenia. Brain Behav Immun. 2015 Feb;44:148-58.

Stinson, L.F., et al. Human Milk From Atopic Mothers Has Lower Levels of Short Chain Fatty Acids. Front Immunol. 2020 Jul 21:11:1427.

Suen, R.M., et al. The Clinical Relevance of IgG Food Allergy Testing Through ELISA. Townsend Letter for Doctors & Patients, Jan 2004, 61–66.

Taylor-Black, S.A., et al. Prevalence of food allergy in New York City school children. Ann Allergy Asthma Immunol. 2014;112(6):554-6 e1.

Tsabouri, S., et al. Modulation of gut microbiota downregulates the development of food allergy in infancy. Allergol Immunopathol (Madr). 2014;42(1):69-77.

Uhde, M., et al. Intestinal cell damage and systemic immune activation in individuals reporting sensitivity to wheat in the absence of coeliac disease. Gut. 2016 Dec;65(12):1930-1937.

Vael, C., et al. Early intestinal Bacteroides fragilis colonization and development of asthma. BMC Pulmonary Medicine. 2008 Sep 26;8:19.

Asthma

Sources & References

American Lung Association. The Impact of Asthma. Accessed 2 Sep 2020.

Azad, Meghan B., et al. Perinatal Programming of Asthma: The Role of Gut Microbiota. Clin Dev Immunol. 2012; 2012: 932072.

Brown, S.D., et al. Airway TGFbeta1 and oxidant stress in children with severe asthma: association with airflow limitation. Allergy Clin Immunol. 2012;129(2):388-96, 96 e1-8.

Bunyavanich, S., et al. Systems biology of asthma and allergic diseases: a multiscale approach. J Allergy Clin Immunol. 2015;135(1):31-42.

Chauhan, B.F., et al. Intermittent versus daily inhaled corticosteroids for persistent asthma in children and adults. Cochrane Database Syst Rev. 2013;2:CD009611.

Checkley, W., et al. 25-hydroxy vitamin D levels are associated with childhood asthma in a population-based study in Peru. Clin Exp Allergy. 2015 Jan;45(1):273–82.

Cortese, S., et al. Association between attention deficit hyperactivity disorder and asthma: a systematic review and meta-analysis and a Swedish population-based study. Lancet Psychiatry. 2018 Sep;5(9):717-726.

D’Auria, E., et al. Omega-3 fatty acids and asthma in children. Allergy Asthma Proc. 2014;35(3):233-40.

Della Giustina, A., et al. Vitamin D, allergies and asthma: focus on pediatric patients. World Allergy Organ J. 2014;7(1):27.

Desai, J.R., et al. Diabetes and asthma case identification, validation, and representativeness when using electronic health data to construct registries for comparative effectiveness and epidemiologic research. Med Care. 2012;50 Suppl:S30-5.

Fabian E., et al. Nutritional supplements and plasma antioxidants in childhood asthma. Wien Klin Wochenschr. 2013;125(11-12):309-15.

Frieri, M. Asthma linked with rhinosinusitis: An extensive review. Allergy Rhinol (Providence). 2014;5(1):41-9

HGuo, C.H., et al. Nutritional supplement therapy improves oxidative stress, immune response, pulmonary function, and quality of life in allergic asthma patients: an open-label pilot study. Altern Med Rev. 2012;17(1):42-56

Hansel, T.T., et al. Microbes and mucosal immune responses in asthma. Lancet. 2013;381(9869):861-73.

Hijazi, N., et al. Diet and childhood asthma in a society in transition: A study in urban and rural Saudi Arabia. Thorax. 2000 Sep;55(9):775-9.

Hoskin-Parr, L., et al. Antibiotic exposure in the first two years of life and development of asthma and other allergic diseases by 7.5 yr: A dose-dependent relationship. Pediatr Allergy Immunol. 2013 Dec; 24(8): 762–771.

Jolliffe, D.A., et al. “Vitamin D supplementation to prevent asthma exacerbations: a systematic review and meta-analysis of individual participant data.Lancet Respir Med. 2017 Nov;5(11):881-890.

Kreiner-Møller, E., et al. Infant acetaminophen use associates with early asthmatic symptoms independently of respiratory tract infections: the Copenhagen Prospective Study on Asthma in Childhood 2000 (COPSAC(2000)) cohort. J Allergy Clin Immunol. 2012 Dec;130(6):1434-6.

Lang, J.E., et al. Role of biomarkers in understanding and treating children with asthma: towards personalized care. Pharmgenomics Pers Med. 2013;6:73-84.

Lessa, N., et al. Asthma and suicide-related adverse events: a review of observational studies. Eur Respir Rev. 2011;20(122):287-92.

Litonjua, A.A. Childhood asthma may be a consequence of vitamin D deficiency. Curr Opin Allergy Clin Immunol. 2009;9(3):202-7.

Ly, N.P., et al. Gut microbiota, probiotics, and vitamin D: interrelated exposures influencing allergy, asthma, and obesity? J Allergy Clin Immunol. 2011;127(5):1087-94; quiz 95-6.

Mabalirajan, U., et al. Effects of vitamin E on mitochondrial and asthma features in an experimental allergic murine model. J Appl Physiol. 2009 Oct;107(4):1285-92.

Martineau, A.R., et al. Vitamin D for the management of asthma. Cochrane Library, 2016 DOI: 10.1002/14651858.CD011511.pub2.

McCloud, E., et al. A medical nutrition therapy primer for childhood asthma: current and emerging perspectives. J Am Diet Assoc. 2011 Jul;111(7):1052–64.

McCormack, et al. Indoor particulate matter increases asthma morbidity in children with non-atopic and atopic asthma. Ann Allergy Asthma Immunol. 2011;106(4):308-15.

Norton, R.L., et al. Selenium and asthma. Mol Aspects Med. 2012;33(1):98-106.

Orivuori, L., et al. High level of fecal calprotectin at age 2 months as a marker of intestinal inflammation predicts atopic dermatitis and asthma by age 6. Clin Exp Allergy. 2015.

Peters, R.L., et al. Infant food allergy phenotypes and association with lung function deficits and asthma at age 6 years: a population-based, prospective cohort study in Australia. Lancet Child Adolesc Health. 2023 Jul 24;S2352-4642(23)00133-5.

Pfeffer, P.E., et al. Vitamin D influences asthmatic pathology through its action on diverse immunological pathways. Ann Am Thorac Soc. 2014;11 Suppl 5:S314-21.

Propp, P., Becker, A. Prevention of asthma: where are we in the 21st century? Expert Rev Clin Immunol. 2013;9(12):1267-78.

Schwalfenberg, G.K. The Importance of Magnesium in Clinical Healthcare. Scientifica (Cairo). 2017:2017:4179326.

Searing, D.A., et al. Decreased serum vitamin D levels in children with asthma are associated with increased corticosteroid use. J Allergy Clin Immunol. 2010;125(5):995-1000.

Tollånes, M.C., et al. Cesarean section and risk of severe childhood asthma: a population-based cohort study. J Pediatr. 2008 Jul;153(1):112-6.

Vael, C., et al. Early intestinal Bacteroides fragilis colonization and development of asthma. BMC Pulmonary Medicine. 2008 Sep 26;8:19.

Wlasiuk, G., et al. The farm effect, or: when, what and how a farming environment protects from asthma and allergic disease. Curr Opin Allergy Clin Immunol. 2012;12(5):461-6

Auditory Therapy

Sources & References

Autism

Sources & References

Ābele, S., et al. Specific Carbohydrate Diet (SCD/GAPS) and Dietary Supplements for Children with Autistic Spectrum Disorder. Proc. Latv. Acad. Sci. 2021;75:417–425.

Accardo, P.J., et al. Toe walking in autism: further observations. J Child Neurol. 2015 Apr;30(5):606-9.

Adam, G., et al. The influence of choline treatment on behavioral and neurochemical autistic-like phenotype in Mthfr-deficient mice. Transl Psychiatry. 2020 Sep 18;10(1):316.

Adams, J.B., et al. Comprehensive Nutritional and Dietary Intervention for Autism Spectrum Disorder-A Randomized, Controlled 12-Month Trial. Nutrients. 2018 Mar 17;10(3).

Adams, J.B., et al. Effect of a vitamin/mineral supplement on children and adults with autism. BMC Pediatr. 2011;11:111.

Adams, J.B., et al. Gastrointestinal Flora and Gastrointestinal Status in Children with Autism—Comparisons to Typical Children and Correlation with Autism Severity. BMC Gastroenterol. 2011;11:22.

Adams, J.B., et al. Mercury in first-cut baby hair of children with autism versus typically-developing children. Toxicological & Environmental Chemistry. 2007 Jun;70(12):1046-51.

Adams, J.B., et al. Mercury, Lead, and Zinc in Baby Teeth of Children with Autism Versus Controls. Journal of Toxicology and Environmental Health. 2007 Jun;70(12):1046-51.

Adams, J.B., et al. Nutritional and metabolic status of children with autism vs. neurotypical children, and the association with autism severity. Nutr Metab (Lond) 2011 Jun 8;8(1):34.

Adams, J.B., et al. Vitamin/mineral/micronutrient supplement for autism spectrum disorders: a research survey. BMC Pediatr. 2022 Oct 13;22(1):590.

Adams, L.J., et al. Infant feeding method and special educational need in 191,745 Scottish schoolchildren: A national, population cohort study. PLoS Med. 2023 Apr 6;20(4):e1004191.

Aguilera, M., et al. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice. Gut Microbes. 2015;6(1):10-23.

Ahn, R.R., et al. Prevalence of parents’ perceptions of sensory processing disorders among kindergarten children. Am J Occup Ther. May-Jun 2004;58(3):287-93.

Alabdali, A., et al. A key role for an impaired detoxification mechanism in the etiology and severity of autism spectrum disorders. Behav Brain Funct. 2014;10:14.

Alabdali, A., et al. Association of social and cognitive impairment and biomarkers in autism spectrum disorders. J Neuroinflammation. 2014;11:4.

Alampi, J.D., et al. Gestational Exposure to Toxicants and Autistic Behaviors using Bayesian Quantile Regression. Am J Epedemiol. 2021 Sep 1;190(9):1803-1813.

Alberti, A., et al. Sulphation Deficit in “Low-Functioning” Autistic Children: A Pilot Study. Biol. Psychiatry. 1999;46:420–424.

Aldad, T.S., et al. Fetal Radiofrequency Radiation Exposure From 800-1900 Mhz-Rated Cellular Telephones Affects Neurodevelopment and Behavior in Mice. Sci Rep. 2012;2:312.

Amminger, G.P., et al. Omega-3 fatty acids supplementation in children with autism: a double-blind randomized, placebo-controlled pilot study. Biol Psychiatry. 2007 Feb 15;61(4):551-3.

Ashraghi, R.S., et al. Early Disruption of the Microbiome Leading to Decreased Antioxidant Capacity and Epigenetic Changes: Implications for the Rise in Autism. Front. Cell. Neurosci., 15 Aug 2018.

Ashwood, P., et al. Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav Immun. 2011 Jan;25(1):40-5.

Ashwood, P., et al. The immune response in autism: a new frontier for autism research. Journal of Leukocyte Biology. 2006 Jul;80(1):1-15.

Atladóttir, H.Ó., et al. Association of family history of autoimmune diseases and autism spectrum disorders. Pediatrics. 2009 Aug;124(2):687-94.

Atladóttir, H.Ó., et al. Autism after infection, febrile episodes, and antibiotic use during pregnancy: an exploratory study. Pediatrics. 2012 Dec;130(6):e1447-54.

Baker, S. Canaries and Miners. Alternative Therapies in Health and Medicine. Nov-Dec 2008;14(6):24-6.

Baker, S., et al. Case Study: Rapid Complete Recovery From An Autism Spectrum Disorder After Treatment of Aspergillus With The Antifungal Drugs Itraconazole And Sporanox. Integr Med (Encinitas). 2020 Aug;19(4):20-27.

Barnhill, K., et al. Brief Report: Implementation of a Specific Carbohydrate Diet for a Child with Autism Spectrum Disorder and Fragile X Syndrome. J. Autism Dev. Disord. 2020;50:1800–1808. 

Barrett, B. Substantial lifelong cost of autism spectrum disorder. J Pediatr. 2014;165(5):1068-9.

Bateman, C. Autism–mitigating a global epidemic. S Afr Med J. 2013;103(5):276-7.

Ben-Sasson, A., et al. Sensory over-responsivity in elementary school: prevalence and social-emotional correlates. J Abnorm Child Psychol. 2009 Jul;37(5):705-16.

Berding, K., et al. Diet Can Impact Microbiota Composition in Children With Autism Spectrum Disorder. Front. Neurosci. 2018;12:515.

Bernard, S., et al. Autism: a novel form of mercury poisoning. Med Hypotheses. 2001 Apr;56(4):462-71.

Binder, D.K., et al. Brain-derived neurotrophic factor. Growth Factors. 2004 Sep;22(3):123-31.

Bittker, S.S., et al. Postnatal Acetaminophen and Potential Risk of Autism Spectrum Disorder among Males. Behav Sci (Basel). 2020 Jan 1;10(1):26.

Bitsika, V., et al. Hypothalamus-pituitary-adrenal axis daily fluctuation, anxiety and age interact to predict cortisol concentrations in boys with an autism spectrum disorder. Physiol Behav. 2015;138:200-7.

Bjørklund, G., et al. Gastrointestinal alterations in autism spectrum disorder: What do we know? Neurosci Biobehav Rev. 2020 Nov:118:111-120.

Blaxill, Mark, et al. Autism Tsunami: the Impact of Rising Prevalence on the Societal Cost of Autism in the United States. J Autism Dev Disord. 2022 Jun;52(6):2627-2643.

Blaylock, R.L. A possible central mechanism in autism spectrum disorders, part 1. Altern Ther Health Med. 2008 Nov-Dec;14(6):46-53.

Blaylock, R.L. A possible central mechanism in autism spectrum disorders, part 2: immunoexcitotoxicity. Altern Ther Health Med. 2009 Jan-Feb;15(1):60-7.

Blaylock, R.L. A possible central mechanism in autism spectrum disorders, part 3: the role of excitotoxin food additives and the synergistic effects of other environmental toxins. Altern Ther Health Med. 2009 Mar-Apr;15(2):56-60.

Blaylock, R.L., et al. Immune-glutamatergic dysfunction as a central mechanism of the autism spectrum disorders. Curr Med Chem. 2009;16(2):157-70.

Boat, T.F., et al. Prevalence of Learning Disabilities. Mental Disorders and Disabilities Among Low-Income Children. Washington (DC): National Academies Press (US); 2015 Oct 28. 16.

Boris, M.J., et al. Pollen Exposure as a Cause for the Deterioration of Neurobehavioral Function in Children with Autism and Attention Deficit Hyperactive Disorder. J of Nutritional and Environmental Medicine. Mar 2004;14(1):47 – 54.

Borre, Y.E., et al. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med. 2014 Sep;20(9):509-18.

Bouder, et al. Brief report: Quantifying the impact of autism coverage on private insurance premiums. J Autism Dev Disord. 2009;39(6):953-7.

Bradstreet, et al. Biomarker-guided interventions of clinically relevant conditions associated with autism spectrum disorders and attention deficit hyperactivity disorder. Altern Med Rev. 2010;15(1):15-32.

Bransfield, R.C., et al. The association between tick-borne infections, Lyme borreliosis and autism spectrum disorders. Medical Hypotheses. 2008;70(5):967-74.

Breitenkamp, A.F., et al. Voltage-gated Calcium Channels and Autism Spectrum Disorders. Curr Mol Pharmacol. 2015;8(2):123-32.

Brown, et al. Observable essential fatty acid deficiency markers and autism spectrum disorder. Breastfeed Rev. 2014;22(2):21-6.

Buescher, et al. Costs of autism spectrum disorders in the United Kingdom and the United States. JAMA Pediatr. 2014;168(8):721-8.

Buie, T., et al. Evaluation, diagnosis, and treatment of gastrointestinal disorders in individuals with ASDs: a consensus report. Pediatrics. 2010 Jan;125 Suppl 1:S1-18.

Buie, T., et al. Recommendations for evaluation and treatment of common gastrointestinal problems in children with ASDs. Pediatrics. 2010 Jan;125 Suppl 1:S19-29.

Bull, G., et al. Indolyl-3-acryloylglycine (IAG) is a putative diagnostic urinary marker for autism spectrum disorders. Med Sci Monit. 2003;9(10):CR422-5.

Büsselberg, D. Calcium channels as target sites of heavy metals. Toxicol Lett. 1995 Dec:82-83:255-61.

Camilleri, M. Serotonin in the gastrointestinal tract. Curr Opin Endrocrinol Diabetes Obes. 2009 Feb;16(1):53-9.

Cardinal, D.N., et al. The Maturing of Facilitated Communication: A Means Toward Independent Communication. Res. Pract. Persons Severe Disabil. 2014; (39)189–194.

Carlo, G.L., et al. Wireless radiation in the aetiology and treatment of autism: clinical observations and mechanisms. Journal of the Australasian College of Nutritional and Environmental Medicine, 26(2), 3–7.

Cekici, H., et al. Current Nutritional Approaches in Managing Autism Spectrum Disorder: A Review. Nutr. Neurosci. 2019;22:145–155.

Centers for Disease Control and Prevention. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years — Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2020. Accessed 24 Mar 2023.

Cheng, N., et al. Metabolic Dysfunction Underlying Autism Spectrum Disorder and Potential Treatment Approaches. Front Mol Neurosci. 2017 Feb 21:10:34.

Chistol, L.T., et al. Sensory Sensitivity and Food Selectivity in Children with Autism Spectrum Disorder. J. Autism Dev. Disord. 2018;48:583–591.

Clarke, E.B., et al. Understanding profound autism: implications for stigma and supports. Front Psychiatry. 2024 Jan 22:15:1287096.

Cleary, D.B., et al. A Parent-Mediated Intervention for Newborns at Familial Likelihood of Autism: Initial Feasibility Study in the General Population. Adv Neurodev Disord. 2022;6(4):494-505.

Connolly, A.M., et al. Serum autoantibodies to brain in Landau-Kleffner variant, autism, and other neurologic disorders. The Journal of Pediatrics. 1999 May;134(5):607-13.

Courchesne, V., et al. Autistic children at risk of being underestimated: school-based pilot study of a strength-informed assessment. Molecular Autism. 2015 Mar 6;6:12.

Critchfield, et al. The potential role of probiotics in the management of childhood autism spectrum disorders. Gastroenterol Res Pract. 2011;2011:161358.

Cubala-Kucharska M. The review of most frequently occurring medical disorders related to aetiology of autism and the methods of treatment. Acta Neurobiol Exp (Wars). 2010;70(2):141-6.

Curran, E.A., et al. Research review: Birth by caesarean section and development of autism spectrum disorder and attention-deficit/hyperactivity disorder: a systematic review and meta-analysis. J Child Psychol Psychiatry. 2015 May;56(5):500-8.

Currenti, S.A. Understanding and determining the etiology of autism. Cell Mol Neurobiol. 2010 Mar;30(2):161-71.

D'Adamo, C.R., et al. Reversal of Autism Symptoms among Dizygotic Twins through a Personalized Lifestyle and Environmental Modification Approach: A Case Report and Review of the Literature. J Pers Med. 2024, 14(6), 641.

Dale, R.C., et al. Encephalitis lethargica syndrome: 20 new cases and evidence of basal ganglia autoimmunity. Brain. 2004 Jan;127(Pt 1):21-33.

Darling, A.L., et al. Association between maternal vitamin D status in pregnancy and neurodevelopmental outcomes in childhood: results from the Avon Longitudinal Study of Parents and Children (ALSPAC). Br J Nutr 2017 Jun;117(12):1682-1692.

Dasdaq, S., et al. Effects of 2.4 GHz radiofrequency radiation emitted from Wi-Fi equipment on microRNA expression in brain tissue. Int J Radiat Biol. 2015 Jul;91(7):555-61.

Dave, D., et al. The effect of an increase in autism prevalence on the demand for auxiliary healthcare workers : evidence from California. Cambridge, MA: National Bureau of Economic Research; 2012. 37 p.p.

D’Eufemia, P., et al. Abnormal intestinal permeability in children with autism. Acta Paediatr. 1996 Sep;85(9):1076-9.

Deisher, T.A., et al. Impact of environmental factors on the prevalence of autistic disorder after 1979. J Public Health and Epidemiology. Sep 2014;6(9):271-286.

de Magistris, et al. Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J Pediatr Gastroenterol Nutr. 2010;51(4):418-24.

De Magistris. L., et al. Antibodies against Food Antigens in Patients with Autistic Spectrum Disorders. BioMed Res. Int. 2013;2013:729349.

Deth, R., et al. How environmental and genetic factors combine to cause autism: A redox/methylation hypothesis. Neurotoxicology. 2008;29(1):190-201.

Doreswamy, S., et al. Effects of Diet, Nutrition, and Exercise in Children With Autism and Autism Spectrum Disorder: A Literature Review. Cureus. 2020;12:e12222.

Dyńka, D., et al. The Role of Ketogenic Diet in the Treatment of Neurological Diseases. Nutrients. 2022 Nov 24;14(23):5003.

Egset, K., et al. Magno App: Exploring Visual Processing in Adults with High and Low Reading Competence. Scandinavian Journal of Educational Research. 07 Jan 2020.

Elamin, N.E., et al. Brain autoantibodies in autism spectrum disorder. Biomark Med. 2014;8(3):345-52.

El-Ansary, A., et al. Neuroinflammation in autism spectrum disorders. J Neuroinflammation. 2012;9:265.

El-Ansary, A., et al. Lipid mediators in plasma of autism spectrum disorders. Lipids Health Dis. 2012;11:160.

El-Rashidy, O., et al. Ketogenic Diet versus Gluten Free Casein Free Diet in Autistic Children: A Case-Control Study. Metab. Brain Dis. 2017;32:1935–1941.

Eppright, T.D., et al. Attention deficit hyperactivity disorder, infantile autism, and elevated blood-lead: a possible relationship. Mo Med. 1996;93(3):136-8).

Erickson, C.A., et al. Gastrointestinal Factors in Autistic Disorder: A Critical Review. Journal of Autism and Developmental Disorders. 2005 Dec;35(6):713-27.

Faber, S., et al. A cleanroom sleeping environment’s impact on markers of oxidative stress, immune dysregulation, and behavior in children with autism spectrum disorders. BMC Complement Altern Med. 2015;15:71.

Fattorusso, A., et al. Autism Spectrum Disorders and the Gut Microbiota. Nutrients. 2019 Feb 28;11(3):521.

Fein, D., et al. Optimal outcome in individuals with a history of autism. J Child Psychol Psychiatry. 2013 Feb;54(2):195-205.

Feingold, B.F. Hyperkinesis and Learning Disabilities Linked to Artificial Food Flavors and Colors. Am. J. Nurs. 1975;75:797–803.

Feng, P., et al. A review of probiotics in the treatment of autism spectrum disorders: Perspectives from the gut-brain axis. Front Microbiol. 2023 Mar 16:14:1123462.

Fezer, G.F., et al. Perinatal Features of Children with Autism Spectrum Disorder. Rev Paul Pediatr. 2017 Apr-Jun;35(2):130-135.

Frisch, M., et al. Ritual circumcision and risk of autism spectrum disorder in 0- to 9-year-old boys: national cohort study in Denmark. J R Soc Med. 2015 Jul;108(7):266-79.

Frustaci, A., et al. Oxidative stress-related biomarkers in autism: systematic review and meta-analyses. Free Radic Biol Med. 2012;52(10):2128-41.

Frye, R.E., et al. Blocking and Binding Folate Receptor Alpha Autoantibodies Identify Novel Autism Spectrum Disorder Subgroups. Front Neurosci. 2016 Mar 9;10:80.

Frye, R.E., et al. Cerebral folate receptor autoantibodies in autism spectrum disorder. Mol Psychiatry. 2013 Mar;18(3):369-81.

Frye, R.E., et al. Metabolic pathology of autism in relation to redox metabolism. Biomark Med. 2014;8(3):321-30.

Frye, R.E., et al. Redox metabolism abnormalities in autistic children associated with mitochondrial disease. Transl Psychiatry. 2013;3:e273.

Gabis, L.V., et al. Improvement of Language in Children with Autism with Combined Donepezil and Choline Treatment. J Mol Neurosci. 2019 Oct;69(2):224-234.

Gabriele, S., et al. Blood serotonin levels in autism spectrum disorder: a systematic review and meta-analysis. Eur Neuropsychopharmacol. 2014;24(6):919-29.

Gadow, K.D., et al. Association of COMT (Val158Met) and BDNF (Val66Met) gene polymorphisms with anxiety, ADHD and tics in children with autism spectrum disorder. J Autism Dev Disord. 2009;39(11):1542-51.

Gadow, K.D., et al. Association of DRD4 polymorphism with severity of oppositional defiant disorder, separation anxiety disorder and repetitive behaviors in children with autism spectrum disorder. Eur J Neurosci. 2010;32(6):1058-65.

Gardender, H., et al. Perinatal and neonatal risk factors for autism: a comprehensive meta-analysis. Pediatrics. 2011 Aug;128(2):344-55.

Gebril, O.H., et al. HFE gene polymorphisms and the risk for autism in Egyptian children and impact on the effect of oxidative stress. Dis Markers. 2011;31(5):289-94.

Geier, D.A., et al. The biological basis of autism spectrum disorders: Understanding causation and treatment by clinical geneticists. Acta Neurobiol Exp (Wars). 2010;70(2):209-26.

Geier, M.R., et al. The potential importance of steroids in the treatment of autistic spectrum disorders and other disorders involving mercury toxicity. Med Hypotheses. 2005;64(5):946-54.

Ghalichi, F., et al. Effect of Gluten Free Diet on Gastrointestinal and Behavioral Indices for Children with Autism Spectrum Disorders: A Randomized Clinical Trial. World J. Pediatr. 2016;12:436–442.

Ghanizadeh, A. Increased glutamate and homocysteine and decreased glutamine levels in autism: a review and strategies for future studies of amino acids in autism. Dis Markers. 2013;35(5):281-6.

Glasson, E.J., et al. Perinatal factors and the development of autism: a population study. Arch Gen Psychiatry. 2004 Jun;61(6):618-27.

Goldani, A.A., et al. Biomarkers in autism. Front Psychiatry. 2014;5:100

Goncalves, M.V.M., et al. Pediatric acute-onset neuropsychiatric syndrome (PANS) misdiagnosed as autism spectrum disorder. Immunol Lett. 2018 Nov;203:52-53.

Gough, S., et al. Neuroprotection by the Ketogenic Diet: Evidence and Controversies. Front Nutr. 2021 Nov 23:8:782657.

Grandjean, P., et al. Developmental neurotoxicity of industrial chemicals. Lancet. 2006 Dec 16;368(9553):2167-78.

Grimaldi, R., et al. A prebiotic intervention study in children with autism spectrum disorders (ASDs). Microbiome. 2018 Aug 2;6(1):133.

Guilford, T., et al. Deficient Glutathione in the Pathophysiology of Mycotoxin-Related Illness. Toxins (Basel). 2014 Feb 10;6(2):608-23.

Guyol, G. Who’s crazy here?: Steps for recovery without drugs for: ADD/ADHD, addiction & eating disorders, anxiety & PTSD, depression, bipolar disorder, schizophrenia, autism. 1st U.S. ed. Stonington, CT: Ajoite Pub.; 2010. 123 p.p.

Hacohen, Y., et al. N‐methyl‐d‐aspartate (NMDA) receptor antibodies encephalitis mimicking an autistic regression. Dev Med Child Neurol. 2016 Oct;58(10):1092-4.

Hadjivassiliou, M., et al. Gluten sensitivity: from gut to brain. Lancet Neurol. 2010 Mar;9(3):318-30.

Hallmayer, J., et al. Genetic heritability and shared environmental factors among twin pairs with autism. Arch Gen Psychiatry. 2011 Nov;68(11):1095-102.

Hamad, A.F., et al. Prenatal antibiotics exposure and the risk of autism spectrum disorders: A population-based cohort study. PLoS One. 2019 Aug 29;14(8):e0221921.

Hamlin, J.C., et al. Dietary intake and plasma levels of choline and betaine in children with autism spectrum disorders. Autism Res Treat. 2013;2013:578429.

Hartman, R.E., et al. Dietary Approaches to the Management of Autism Spectrum Disorders. Adv. Neurobiol. 2020;24:547–571.

Hauser, P., et al. Resistance to thyroid hormone: implications for neurodevelopmental research on the effects of thyroid hormone disruptors.Toxicol Ind Health. 1998 Jan-Apr;14(1-2):85-101.

Hejitz, R.D., et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011 Feb 15;108(7):3047-52.

Herbert, M.R., et al. Autism and Dietary Therapy: Case Report and Review of the Literature. J. Child. Neurol. 2013;28:975–982.

Herbert, M.R., et al. Autism and environmental genomics. Neurotoxicology. 2006;27(5):671-84.

Herbert, M.R, et al. Autism and EMF? Plausibility of a pathophysiological link–Part I. Pathophysiology 20.3 (2013): 191-209.

Herbert, M.R., et al. Autism and EMF? Plausibility of a pathophysiological link Part II. Pathophysiology 20.3 (2013): 211-234.

Herbert, M.R. Contributions of the environment and environmentally vulnerable physiology to autism spectrum disorders. Curr Opin Neurol. 2010 Apr;23(2):103-10.

Hertz-Picciotto, I., et al. The rise in autism and the role of age at diagnosis. Epidemiology. 2009 Jan;20(1):84-90.

Hertz-Picciotto, I., et al. Organophosphate exposures during pregnancy and child neurodevelopment: Recommendations for essential policy reforms. PLoS Med. 2018 Oct 24;15(10):e1002671.

Hertz-Picciotto, I., et al. Polybrominated diphenyl ethers in relation to autism and developmental delay: a case-control study. Environ Health. 2011 Jan 5;10(1):1.

Hertz-Picciotto, I., et al. Prenatal exposures to persistent and non-persistent organic compounds and effects on immune system development. Basic Clin Pharmacol Toxicol. 2008 Feb;102(2):146-54.

Heyer, N.J., et al. Disordered porphyrin metabolism: a potential biological marker for autism risk assessment. Autism Res. 2012;5(2):84-92.

Holmes, A., et al. Reduced Levels of Mercury in First Baby Haircuts of Autistic Children. International Journal of Toxicology. Jul-Aug 2003;22(4):277-85.

Hopf, K.P., et al. Use and Perceived Effectiveness of Complementary and Alternative Medicine to Treat and Manage the Symptoms of Autism in Children: A Survey of Parents in a Community Population. J. Altern. Complement. Med. 2016;22:25–32. 

Horn, J., et al. Role of Diet and Its Effects on the Gut Microbiome in the Pathophysiology of Mental Disorders. Transl. Psychiatry. 2022;12:164.

Horvath, K., et al. Autism and gastrointestinal symptoms. Curr Gastroenterol Rep. 2002 Jun;4(3):251-8.

Horvath, K., et al. Autistic disorder and gastrointestinal disease. Current Opinion in Pediatrics. 2002 Oct;14(5):583-7.

Horvath, K., et al. Gastrointestinal abnormalities in children with autistic disorder. Journal of Pediatrics. 1999 Nov;135(5):559-63.

Howsmon, D. P., et al. Multivariate techniques enable a biochemical classification of children with autism spectrum disorder versus typically‐developing peers: A comparison and validation study. Bioengineering & Translational Medicine. 2018. doi:10.1002/btm2.10095.

Hsu, C.-L., et al. The Effects of a Gluten and Casein-Free Diet in Children with Autism: A Case Report. Chang Gung Med. J. 2009;32:459–465.

Hyman, M. Autism: Is It All in the Head? Alternative Therapies in Health and Medicine. Nov-Dec 2008;14(6):12-5.

Isaksson, J., et al. Brief Report: Association Between Autism Spectrum Disorder, Gastrointestinal Problems and Perinatal Risk Factors Within Sibling Pairs. J Autism Dev Disord. 2017 Aug;47(8):2621-2627.

Ivanovski, I., et al. Aluminum in brain tissue in autism. J Trace Elem Med Biol. 2019 Jan;51:138-140.

Jadavji, N.M., et al. B-vitamin and choline supplementation increases neuroplasticity and recovery after stroke. Neurobiol Dis. 2017 Jul;103:89-100.

Jafari, M.H., et al. The Relationship Between the Level of Copper, Lead, Mercury and Autism Disorders: A Meta-Analysis. Pediatric Health, Medicine and Therapeutics. 21 Sep 2020(11):369—378.

Jafari, T., et al. The association between mercury levels and autism spectrum disorders: A systematic review and meta-analysis. J Trace Elem Med Biol. 2017 Dec;44:289-297.

James, S.J., 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.

Jarusiewicz, B. Efficacy of Neurofeedback for Children in the Autistic Spectrum: A Pilot Study. Journal of Neurotherapy. 2002;6(4).

Jaswal, V.K., et al. Eye-tracking reveals agency in assisted autistic communication. Scientific Reports. 2020 May 12;10(1):7882.

Jyonouchi, H., et al. Dysregulated innate immune responses in young children with autism spectrum disorders: their relationship to gastrointestinal symptoms and dietary intervention. Neuropsychobiology. 2005;51(2):77-85.

Jyonouchi, H., et al. Evaluation of an Association between Gastrointestinal Symptoms and Cytokine Production against Common Dietary Proteins in Children with Autism Spectrum Disorders. J. Pediatr. 2005;146:605–610.

Jyonouchi, H., et al. Impact of innate immunity in a subset of children with autism spectrum disorders: a case control study. Journal of Neuroinflammation. 2008 Nov 21;5:52.

Jyonouchi, H., et al. Innate Immunity Associated with Inflammatory Responses and Cytokine Production against Common Dietary Proteins in Patients with Autism Spectrum Disorder. Neuropsychobiology. 2002;46:76–84.

Kaluzna-Czaplinska, J., et al. Identification of organic acids as potential biomarkers in the urine of autistic children using gas chromatography/mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 2014;966:70-6.

Kałużna-Czaplińska, J., et al. Nutritional Strategies and Personalized Diet in Autism-Choice or Necessity? Trends Food Sci. Technol. 2016;49:45–50.

Kane, R.C. A possible association between fetal/neonatal exposure to radiofrequency electromagnetic radiation and the increased incidence of autism spectrum disorders (ASD). Med Hypotheses. 2004;62(2):195-7.

Kang, D.W., et al. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome. 2017 Jan 23;5(1):10.

Kang, D.W., et al.  Long-term benefit of Microbiota Transfer Therapy on autism symptoms and gut microbiota. Scientific Reports. 9, 5821 (2019).

Karagözlü, S., et al. The Relationship of Severity of Autism with Gastrointestinal Symptoms and Serum Zonulin Levels in Autistic Children. J. Autism Dev. Disord. 2022;52:623–629.

Karhu, E., et al. Nutritional interventions for autism spectrum disorder. Nutr Rev. 2020 Jul 1;78(7):515-531.

Kawicka, A., et al. How Nutritional Status, Diet and Dietary Supplements Can Affect Autism. A Review. Rocz. Panstw. Zakl. Hig. 2013;64:1–12.

Kern, J.K., et al. A biomarker of mercury body-burden correlated with diagnostic domain specific clinical symptoms of autism spectrum disorder. Biometals. 2010;23(6):1043-51.

Khan, Z., et al. Slow CCL2-dependent translocation of biopersistent particles from muscle to brain. BMC Med. 2013 Apr 4;11:99.

Knivsberg, A.M., et al. A Randomised, Controlled Study of Dietary Intervention in Autistic Syndromes. Nutr. Neurosci. 2002;5:251–261.

Kobliner, V., et al. Reduction in Obsessive Compulsive Disorder and Self-Injurious Behavior With Saccharomyces boulardii in a Child with Autism: A Case Report. Integr Med (Encinitas). 2018 Dec;17(6):38-41.

Kohen-Raz, R., et al. Postural control in children with autism. J Autism Dev Disord. 1992 Sep;22(3):419-32.

Konkel, L. Phthalates and Autistic Traits: Exploring the Association between Prenatal Exposures and Child Behavior. Environ Health Perspec. 2020 Oct;128(10):104001.

Konstantareas, M.M., et al. Ear infections in autistic and normal children. Journal of Autism and Developmental Disorders. 1987 Dec;17(4):585-94.

Konstantynowicz, J., et al. A Potential Pathogenic Role of Oxalate in Autism. Eur. J. Paediatr. Neurol. 2012;16:485–491.

Kordulewska, N.K., et al. Serum cytokine levels in children with spectrum autism disorder: Differences in pro- and anti-inflammatory balance. J Neuroimmunol. 2019 Dec 15;337:577066.

Kushak, R.I., et al. Intestinal Disaccharidase Activity in Patients with Autism: Effect of Age, Gender, and Intestinal Inflammation. Autism. 2011;15:285–294.

Kuwabara, H., et al. Altered metabolites in the plasma of autism spectrum disorder: a capillary electrophoresis time-of-flight mass spectroscopy study. PLoS One. 2013;8(9):e73814.

Lai, C.C.W., et al. The association between gut-health promoting diet and depression: A mediation analysis. J Affect Disord. 2023 Mar 1:324:136-142.

Langley, E.A., et al. High maternal choline consumption during pregnancy and nursing alleviates deficits in social interaction and improves anxiety-like behaviors in the BTBR T+Itpr3tf/J mouse model of autism. Behav Brain Res. 2015 Feb 1;278:210-20.

Lathe, R. Environmental factors and limbic vulnerability in childhood autism; Clinical report. American Journal of Biochemistry and Biotechnology. 4 (2): 183-197, 2008.

Lathe, R. Microwave Electromagnetic Radiation and Autism. E-Journal of Applied Psychology. June 2009;5(1):11-30.

Lavelle, T.A., et al. Economic burden of childhood autism spectrum disorders. Pediatrics. 2014;133(3):e520-9.

Lee, K., et al. Autism-associated Shank3 mutations alter mGluR expression and mGluR-dependent but not NMDA receptor-dependent long-term depression. Synapse. 2019 Aug;73(8):e22097.

Lee, R.W.Y., et al. A modified ketogenic gluten-free diet with MCT improves behavior in children with autism spectrum disorder. Physiol Behav. 2018 May 1:188:205-211.

Li, C., et al. Study on Aberrant Eating Behaviors, Food Intolerance, and Stereotyped Behaviors in Autism Spectrum Disorder. Front. Psychiatry. 2020;11:493695.

Li, Q., et al. A Ketogenic Diet and the Treatment of Autism Spectrum Disorder. Front Pediatr. 2021 May 11:9:650624.

Li, Q., et al. Prevalence of Autism Spectrum Disorder Among Children and Adolescents in the United States From 2019 to 2020. JAMA Pediatr. 2022 Sep 1;176(9):943-945.

Li, S.O., et al. Serum copper and zinc levels in individuals with autism spectrum disorders. Neuroreport. 2014;25(15):1216-20.

Li, Y., et al. Association between MTHFR C677T/A1298C and susceptibility to autism spectrum disorders: a meta-analysis. BMC Pediatrics. 2020(20)449.

Liao, T.C., et al. Comorbidity of Atopic Disorders with Autism Spectrum Disorder and Attention Deficit/Hyperactivity Disorder.  J Pediatr. 2016 Apr;171:248-55.

Lionetti, E., et al. Gluten Psychosis: Confirmation of a New Clinical Entity. Nutrients. 2015 Jul 8;7(7):5532-9.

López-Aranda, M.F., et al. Postnatal immune activation causes social deficits in a mouse model of tuberous sclerosis: Role of microglia and clinical implications. Sci Adv.2021 Sep 17;7(38):eabf2073.

Lucarelli, S., et al. Food Allergy and Infantile Autism. Panminerva Med. 1995;37:137–141.

Lyall, K., et al. Prenatal Serum Concentrations of Brominated Flame Retardants and Autism Spectrum Disorder and Intellectual Disability in the Early Markers of Autism Study: A Population-Based Case-Control Study in California. Environ Health Perspect. 2017 Aug 30;125(8):087023.

Mader, S., et al. The Role of Brain-Reactive Autoantibodies in Brain Pathology and Cognitive Impairment. Front Immunol. 2017; 8: 1101.

Madra, M., et al. Gastrointestinal Issues and Autism Spectrum Disorder. Psychiatr Clin North Am. 2021 Mar; 44(1): 69–81.

Maenner, M.J., et al. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years – Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2018. MMWR Surveill Summ. 2021 Dec 3;70(11):1-16.

Mahapatra, S., et al. Autism Treatment Evaluation Checklist (ATEC) Norms: A "Growth Chart" for ATEC Score Changes as a Function of Age. Children (Basel). 2018 Feb 16;5(2):25.

Maher, P. Methylglyoxal, advanced glycation end products and autism: is there a connection? Med Hypotheses. 2012;78(4):548-52.

Mandecka, A., et al. The importance of nutritional management and education in the treatment of autism. Rocz. Panstw. Zakl. Hig. 2022;73:247–258. [

Marí-Bauset, S., et al. Nutritional Impact of a Gluten-Free Casein-Free Diet in Children with Autism Spectrum Disorder. J. Autism Dev. Disord. 2016;46:673–684.

Matthews, J.S., et al. Ratings of the Effectiveness of 13 Therapeutic Diets for Autism Spectrum Disorder: Results of a National Survey. J Pers Med. 2023 Sep 29;13(10):1448.

McCann, D., et al. Food Additives and Hyperactive Behaviour in 3-Year-Old and 8/9-Year-Old Children in the Community: A Randomised, Double-Blinded, Placebo-Controlled Trial. Lancet. 2007;370:1560–1567.

Melke, J., et al. Abnormal melatonin synthesis in autism spectrum disorders. Mol Psychiatry. 2008 Jan;13(1):90-8.

Mercer, J.S., et al. The Effects of Delayed Cord Clamping on 12-Month Brain Myelin Content and Neurodevelopment: A Randomized Controlled Trial. Am J Perinatol. 2022 Jan;39(1):37-44.

Mesleh, A.G., et al. Paving the Way toward Personalized Medicine: Current Advances and Challenges in Multi-OMICS Approach in Autism Spectrum Disorder for Biomarkers Discovery and Patient Stratification. J. Pers. Med. 2021;11:41.

Messer, A. Mini-review: polybrominated diphenyl ether (PBDE) flame retardants as potential autism risk factors. Physiol Behav. 2010 Jun 1;100(3):245-9.

Mierau, S.B., et al. Metabolic interventions in Autism Spectrum Disorder. Neurobiol Dis. 2019 Dec:132:104544.

Modabbernia, A., et al. Apgar score and risk of autism. Eur J Epidemiol. 2018 Oct 5.

Mold, M., et al. Aluminium in brain tissue in autism. J Trace Elem Med Biol. 2018 Mar;46:76-82.

Molina-López, J., et al. Food Selectivity, Nutritional Inadequacies, and Mealtime Behavioral Problems in Children with Autism Spectrum Disorder Compared to Neurotypical Children. Int. J. Eat. Disord. 2021;54:2155–2166.

Momeni, N., et al. A novel bloodbased biomarker for detection of autism spectrum disorders. Transl Psychiatry. 2012;2:e91.

Mu, C., et al. Metabolic Framework for the Improvement of Autism Spectrum Disorders by a Modified Ketogenic Diet: A Pilot Study. J Proteome Res. 2020 Jan 3;19(1):382-390.

Nankova, B.B., 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(8):e103740.

Naviaux, R.K. Metabolic features of the cell danger response. Mitochondrion. 2014 May:16:7-17.

Nemecheck, P., et al. Autism Spectrum Disorder Symptoms Improve with Combination Therapy Directed at Improving Gut Microbiota and Reducing Inflammation. Applied Psychiatry. 2020 Jul; (1)1.

Nevison, C.D., et al. Diagnostic Substitution for Intellectual Disability: A Flawed Explanation for the Rise in Autism. J Autism Dev Disord. 2017 Sep;47(9):2733-2742.

Ngounou Wetie, A.G., et al. A pilot proteomic study of protein markers in autism spectrum disorder. Electrophoresis. 2014;35(14):2046-54.

Nicolson, G.L., et al. Chronic Mycoplasmal Infections in Autism Patients. Proc. Intern. Mind of a Child Conference, Sydney, Australia 2002.

Nicolson, G.L., et al. Evidence for Mycoplasma ssp., Chlamydia pneunomiae, and human herpes virus-6 coinfections in the blood of patients with autistic spectrum disorders. J Neurosci Res. 2007 Apr;85(5):1143-8.

Nikolova, V.L., et al. Acceptability, Tolerability, and Estimates of Putative Treatment Effects of Probiotics as Adjunctive Treatment in Patients With Depression. JAMA Psychiatry. 2023 Aug 1;80(8):842-847.

Nirmalkar, K., et al. Shotgun Metagenomics Study Suggests Alteration in Sulfur Metabolism and Oxidative Stress in Children with Autism and Improvement after Microbiota Transfer Therapy. Int J Mol Sci. 2022 Nov 3;23(21):13481.

Noto, A., et al. The urinary metabolomics profile of an Italian autistic children population and their unaffected siblings. J Matern Fetal Neonatal Med. 2014;27 Suppl 2:46-52.

O'Hara, N.H., et al. The recovery of a child with autism spectrum disorder through biomedical interventions. Altern Ther Health Med. 2008 Nov-Dec;14(6):42-4.

Oliveira, G., et al. Mitochondrial dysfunction in autism spectrum disorders: a population-based study. Dev Med Child Neurol. 2005 Mar;47(3):185-9.

Olivito, I., et al. Ketogenic diet ameliorates autism spectrum disorders-like behaviors via reduced inflammatory factors and microbiota remodeling in BTBR T+ Itpr3tf/J mice. Exp Neurol. 2023 Aug:366:114432.

Ozonoff, S., et al. Onset patterns in autism: Variation across informants, methods, and timing. Autism Res. 2018 Mar 10.

Pall, M.L. Microwave frequency electromagnetic fields (EMFs) produce widespread neuropsychiatric effects including depression. J Chem Neuroanat. 2016 Sep;75(Pt B):43-51.

Pall, M.L. The Autism Epidemic Is Caused by EMFs, Acting via Calcium Channels and Chemicals Acting via NMDA-Rs: Downstream Effects Cause Autism, Autism One, Chicago, Illinois, USA, 2015.

Pall, M.L. Wi-Fi is an important threat to human health. Environ Res. 2018 Jul;164:405-416.

Palmer, et al. Environmental mercury release, special education rates, and autism disorder: an ecological study of Texas. Health Place. 2006(12):203-209.

Palmer, R.F., et al. Proximity to point sources of environmental mercury release as a predictor of autism prevalence. Health and Place. 2009 Mar;15(1):18-24.

Palmieri, L., et al. Altered calcium homeostasis in autism-spectrum disorders: evidence from biochemical and genetic studies of the mitochondrial aspartate/glutamate carrier AGC. Mol Psychiatry. 2010 Jan;15(1):38-52.

Palmieri, L., et al. Mitochondrial dysfunction in autism spectrum disorders: Cause or effect? Biochim Biophys Acta. 2010 June – July;1797(6-7):1130-1137.

Pastural, E., et al. Novel plasma phospholipid biomarkers of autism: mitochondrial dysfunction as a putative causative mechanism. Prostaglandins Leukot Essent Fatty Acids. 2009 Oct;81(4):253-64.

Patil, A.A., et al. An Ayurvedic perspective about Autism - Literary Review. Journal of Ayurveda and Integrated Medical Sciences. 2023 Feb;8(2).

Patrick, R.P., et al. Vitamin D hormone regulates serotonin synthesis. Part 1: relevance for autism. FASEB J. 2014;28(6):2398-413.

Patterson, P.H. Immune involvement in schizophrenia and autism: etiology, pathology and animal models. Behav Brain Res. 2009 Dec 7;204(2):313-21.

Peterson, B.S., et al. Brain lactate as a potential biomarker for comorbid anxiety disorder in autism spectrum disorder-reply. JAMA Psychiatry. 2015;72(2):190-1.

Pietrzak, D., et al. The Therapeutic Role of Ketogenic Diet in Neurological Disorders. Nutrients. 2022 May 6;14(9):1952.

Pino-López, M., et al. [Parental occupational exposures and autism spectrum disorder in children]. Rev Esp Salud Publica. 2013 Jan-Feb;87(1):73-85.

Piwowarczyk, A., et al. Gluten-Free Diet in Children with Autism Spectrum Disorders: A Randomized, Controlled, Single-Blinded Trial. J. Autism Dev. Disord. 2020;50:482–490.

Quan, L., et al. A Systematic Review and Meta-Analysis of the Benefits of a Gluten-Free Diet and/or Casein-Free Diet for Children with Autism Spectrum Disorder. Nutr. Rev. 2022;80:1237–1246.

Qiu, C., et al. Association Between Epidural Analgesia During Labor and Risk of Autism Spectrum Disorders in Offspring. JAMA Pediatr. 2020 Oct 12.

Qureshi, F., et al. Multivariate Analysis of Metabolomic and Nutritional Profiles among Children with Autism Spectrum Disorder. J. Pers. Med. 2022;12:923.

Ranjbar, A., et al. Comparison of urinary oxidative biomarkers in Iranian children with autism. Res Dev Disabil. 2014;35(11):2751-5.

Ratajczak, H.V. Theoretical aspects of autism: biomarkers–a review. J Immunotoxicol. 2011;8(1):80-94.

Resseguie, M.E., et al. Aberrant estrogen regulation of PEMT results in choline deficiency-associated liver dysfunction. J Biol Chem. 2011 Jan 14;286(2):1649-58.

Reynolds, A., et al. Iron status in children with autism spectrum disorder. Pediatrics. 2012;130 Suppl 2:S154-9.

Rimland, B., et al. Parent Ratings of Behavioral Effects of Biomedical Interventions. Autism Research Institute Newsletter. Volume 34 ARI Publication; San Diego, CA, USA: 2009.

Ristori, M.V., et al. Autism, Gastrointestinal Symptoms and Modulation of Gut Microbiota by Nutritional Interventions. Nutrients. 2019;11:2812.

Rook, G.A., et al. Microbiota, immunoregulatory old friends and psychiatric disorders. Adv Exp Med Biol. 2014:817:319-56.

Rossignol, D.A., et al. 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 Nov 3;11(11):1141.

Rossignol, D. Diagnosis Autism: Now What? A Simplified Biomedical Approach. The Autism File. 2009(3).

Rutter, M. Changing concepts and findings on autism. J Autism Dev Disord. 2013;43(8):1749-57.

Ruggeri, B., et al. Biomarkers in autism spectrum disorder: the old and the new. Psychopharmacology (Berl). 2014;231(6):1201-16.

Sakurai, T., et al. Slc25a12 disruption alters myelination and neurofilaments: a model for a hypomyelination syndrome and childhood neurodevelopmental disorders. Biol Psychiatry. 2010 May 1;67(9):887-94.

Shenouda, J., et al. Prevalence and Disparities in the Detection of Autism Without Intellectual Disability. Pediatrics. 2023 Feb 1;151(2):e2022056594.

Simpson, C.A., et al. The gut microbiota in anxiety and depression - A systematic review. Clin Psychol Rev. 2021 Feb:83:101943.

Singh, V.K., et al. Circulating autoantibodies to neuronal and glial filament proteins in autism. Pediatr Neurol. 1997 Jul;17(1):88-90.

Singh, V.K., et al. Elevated levels of measles antibodies in children with autism. Pediatr Neurol. 2003 Apr;28(4):292-4.

Skripuletz, T., et al. The choline pathway as a strategy to promote central nervous system (CNS) remyelination. Neural Regen Res. 2015 Sep;10(9):1369-70.

Smith, J., et al. Ketogenic diet restores aberrant cortical motor maps and excitation-to-inhibition imbalance in the BTBR mouse model of autism spectrum disorder. Behav Brain Res. 2016 May 1:304:67-70.

Spilioti, M., 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.

Stafstrom, C.E., et al. The ketogenic diet as a treatment paradigm for diverse neurological disorders. Front Pharmacol. 2012 Apr 9:3:59.

Strunecka, A., et al. Immunoexcitotoxicity as the central mechanism of etiopathology and treatment of autism spectrum disorders: A possible role of fluoride and aluminum. Surg Neurol Int. 2018 Apr 9;9:74.

Swann., O.G., et al. Dietary fiber and its associations with depression and inflammation. Nutr Rev. 2020 May 1;78(5):394-411.

Tan S., et al. The Association between Sugar-Sweetened Beverages and Milk Intake with Emotional and Behavioral Problems in Children with Autism Spectrum Disorder. Front. Nutr. 2022;9:927212.

Tan, Y., et al. Review of research progress on intestinal microbiota based on metabolism and inflammation for depression. Arch Microbiol. 2024 Mar 10;206(4):146.

Taurines, R., et al. Expression analyses of the mitochondrial complex I 75-kDa subunit in early onset schizophrenia and autism spectrum disorder: increased levels as a potential biomarker for early onset schizophrenia. Eur Child Adolesc Psychiatry. 2010 May;19(5):441-8.

Teitelbaum, P., et al. Movement analysis in infancy may be useful for early diagnosis of autism. Proc Natl Acad Sci U S A. 1998 Nov 10;95(23):13982-7.

Teitelbaum, P., et al. Reflexes gone astray in autism in infancy. Journal of Developmental and Learning Disorders, 2003;6, 15-22.

Theoharides, T.C. Is a subtype of autism an allergy of the brain? Clin Ther. 2013; 35(5):584-91.

Thomas, R.H., et al. The enteric bacterial metabolite propionic acid alters brain and plasma phospholipid molecular species: further development of a rodent model of autism spectrum disorders. J Neuroinflammation. 2012;9:153.

Thornton, I.M. Out of time: a possible link between mirror neurons, autism and electromagnetic radiation. Med Hypotheses. 2006;67(2):378-82.

Tomcheck, S.D., et al. Sensory Processing in Children with and without Autism: A Comparative Study Using the Short Sensory Profile. American Journal of Occupational Therapy. 2007. 61, 190-200.

Tomljenovic, L., et al. Do aluminum adjuvants contribute to the rising prevalence of autism? J Inorg Biochem. 2011 Nov;105(11):1489-99.

Tomova, A., et al. The Influence of Food Intake Specificity in Children with Autism on Gut Microbiota. Int. J. Mol. Sci. 2020;21:2797.

Treffert, D.A. Epidemiology of Infantile Autism. Arch Gen Psychiatry. 1970;22(5):431-438.

Tzang, R.F., et al. Autism Associated With Anti-NMDAR Encephalitis: Glutamate-Related Therapy. Front Psychiatry. 2019 Jun 21:10:440.

Vargas, D.D., et al. Effectiveness of nutritional interventions on behavioral symptomatology of autism spectrum disorder: a systematic review. Nutr Hosp. 2022 Dec 20;39(6):1378-1388.

Vargas, D.L., et al. Neuroglial activation and neuroinflammation in the brain of patients with autism. Annals of Neurology. 2005 Jan;57(1):67-81.

Verena, L., et al. Elimination diets’ efficacy and mechanisms in attention deficit hyperactivity disorder and autism spectrum disorder. Eur Child Adolesc Psychiatry. 2017; 26(9): 1067–1079.

Vita, A.A., et al. Associations between Food-Specific IgG Antibodies and Intestinal Permeability Biomarkers. Front. Nutr. 2022;9:962093.

Vojdani, A., et al. A Gut Feeling for Immune Dysregulation & Neuroinflammation in Autism. The Autism File. 2009(31).

Vuillermot, S., et al. Vitamin D treatment during pregnancy prevents autism-related phenotypes in a mouse model of maternal immune activation. Mol Autism. 2017 Mar 7;8:9.

Wang, J., et al. Global Prevalence of Autism Spectrum Disorder and Its Gastrointestinal Symptoms: A Systematic Review and Meta-Analysis. Front. Psychiatry. 2022;13:963102.

Wang, L., et al. Gastrointestinal microbiota and metabolite biomarkers in children with autism spectrum disorders. Biomark Med. 2014;8(3):331-44.

Wang, X., et al. Association between Dietary Quality and Executive Functions in School-Aged Children with Autism Spectrum Disorder. Front. Nutr. 2022;9:940246.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Wasilewska, J., et al. Gastrointestinal symptoms and autism spectrum disorder: links and risks – a possible new overlap syndrome. Pediatric Health Med Ther. 2015; 6: 153–166.

Waterhouse, L. Autism Overflows: Increasing Prevalence and Proliferating Theories. Neuropsychology Review. 2008 Dec;18(4):273-86.

Wen, Y., et al. Autism Pathway Network Analyses Identify Overlaps with Other Disease Groups, Involve Many Functions and Converge upon MAPK and Calcium Signaling. Conference: International Meeting for Autism Research, Baltimore Convention Center Baltimore, MD, May 2016.

Wen, Y., et al. Connecting the dots: Overlaps between autism and cancer suggest possible common mechanisms regarding signaling pathways related to metabolic alterations. Med Hypotheses. 2017 Jun:103:118-123.

Westmark, C.J. Soy Infant Formula and Seizures in Children with Autism: A Retrospective Study. PLoS ONE. 2014;9:e80488.

Whiteley, P., et al. The ScanBrit Randomised, Controlled, Single-Blind Study of a Gluten- and Casein-Free Dietary Intervention for Children with Autism Spectrum Disorders. Nutr. Neurosci. 2010;13:87–100.

Williams, B.L., et al. Impaired Carbohydrate Digestion and Transport and Mucosal Dysbiosis in the Intestines of Children with Autism and Gastrointestinal Disturbances. PLoS ONE. 2011;6:e24585.

Williams, E.L., et al. Potential teratogenic effects of ultrasound on corticogenesis: implications for autism. Med Hypotheses. 2010 Jul;75(1):53-8.

Willfors, C., et al. Medical history of discordant twins and environmental etiologies of autism. Transl Psychiatry. 2017 Jan; 7(1): e1014.

Wilson, S., et al. Role of the NLRP3 Inflammasome in Responses. J Allergy Clin Immunol. 2012 Feb 1; 129(2):Supplement AB162.

Windham, G.C., et al. Autism Spectrum Disorders in Relation to Distribution of Hazardous Air Pollutants in the San Francisco Bay Area. Environmental Health Perspectives. 2006 Sep;114(9):1438-44.

Wong, S., et al. Autism, Mitochondria and Polybrominated Diphenyl Ether Exposure. CNS Neurol Disord Drug Targets. 2016;15(5):614-23.

Woodman, A.C., et al. Change in autism symptoms and maladaptive behaviors in adolescence and adulthood: the role of positive family processes. J Autism Dev Disord. 2015;45(1):111-26.

Wu, D.M., et al. Relationship Between Neonatal Vitamin D at Birth and Risk of Autism Spectrum Disorders: the NBSIB Study. J Bone Miner Res. 2018 Mar;33(3):458-466.

Yu, Y., et al. Efficacy and Safety of Diet Therapies in Children With Autism Spectrum Disorder: A Systematic Literature Review and Meta-Analysis. Front. Neurol. 2022;13:844117.

Zablotsky, B., et al. Estimated Prevalence of Children With Diagnosed Developmental Disabilities in the United States, 2014–2016. National Center for Health Statistics. NCHS Data Brief, No. 291, Nov 2017.

Zablotsky, B., et al. Prevalence and Trends of Developmental Disabilities among Children in the United States: 2009-2017. Pediatrics. 2019 Oct;144(4):e20190811.

Zaigham, M., et al. Prelabour caesarean section and neurodevelopmental outcome at 4 and 12 months of age: an observational study. BMC Pregnancy and Childbirth. 2020 (20)564.

Zeisel, S.H. Choline: Critical Role During Fetal Development and Dietary Requirements in Adults. Annu Rev Nutr. 2006;26:229-50.

Zengeler, K.E., et al. SSRI treatment modifies the effects of maternal inflammation on in utero physiology and offspring neurobiology. Brain Behav Immun. 2023 Feb:108:80-97.

Autoimmune Disorders

Sources & References

Bahr, L.S., et al. Ketogenic diet and fasting diet as Nutritional Approaches in Multiple Sclerosis (NAMS): protocol of a randomized controlled study. Trials. 2020 Jan 2;21(1):3.

Bisht, B., et al. Multimodal intervention improves fatigue and quality of life in subjects with progressive multiple sclerosis: a pilot study. Degener Neurol Neuromuscul Dis. 2015;5:19-35.

Bourre, J.M. Effects of nutrients (in food) on the structure and function of the nervous system: update on dietary requirements for brain. Part 1: micronutrients. J Nutr Health Aging. 2006 Sep-Oct;10(5):377-85.

Bourre, J.M. Effects of nutrients (in food) on the structure and function of the nervous system: update on dietary requirements for brain. Part 2 : macronutrients. J Nutr Health Aging. 2006 Sep-Oct;10(5):386-99.

Brenton, J.N., et al. Phase II study of ketogenic diets in relapsing multiple sclerosis: safety, tolerability and potential clinical benefits. J Neurol Neurosurg Psychiatry. 2022 Jun;93(6):637-644.

Brenton, J.N., et al. Pilot study of a ketogenic diet in relapsing-remitting MS. Neurol Neuroimmunol Neuroinflamm. 2019 Apr 12;6(4):e565.

Camara-Lemarroy, C.R., et al. Focus on the gut-brain axis: Multiple sclerosis, the intestinal barrier and the microbiome. World J Gastroenterol. 2018 Oct 7;24(37):4217-4223.

Chenard, C.A., et al. Nutrient Composition Comparison between a Modified Paleolithic Diet for Multiple Sclerosis and the Recommended Healthy U.S.-Style Eating Pattern. Nutrients. 2019 Mar 1;11(3):537.

Correale, J., et al. The role of the gut microbiota in multiple sclerosis. Nat Rev Neurol. 2022 Sep;18(9):544-558.

de Goffau, M.C., et al. Fecal microbiota composition differs between children with beta-cell autoimmunity and those without. Diabetes. 2013;62(4):1238-44.

Di Majo, D., et al. Ketogenic and Modified Mediterranean Diet as a Tool to Counteract Neuroinflammation in Multiple Sclerosis: Nutritional Suggestions. Nutrients. 2022 Jun 8;14(12):2384.

Dinse, G.E., et al. Increasing Prevalence of Antinuclear Antibodies in the United States. Arthritis Rheumatol. 2022 Dec;74(12):2032-2041.

Engelenburg, H.J., et al. Multiple sclerosis and the microbiota: Progress in understanding the contribution of the gut microbiome to disease. Evol Med Public Health. 2022 Jun 13;10(1):277-294.

Fan, Y., et al. Dietary Modulation of Intestinal Microbiota: Future Opportunities in Experimental Autoimmune Encephalomyelitis and Multiple Sclerosis. Front Microbiol. 2019 Apr 16;10:740.

Fettig, N.M., et al. Direct and indirect effects of microbiota-derived metabolites on neuroinflammation in multiple sclerosis. Microbes Infect. 2021 Jul-Aug;23(6-7):104814.

Irish, A.K., et al. Randomized control trial evaluation of a modified Paleolithic dietary intervention in the treatment of relapsing-remitting multiple sclerosis: a pilot study. Degener Neurol Neuromuscul Dis. 2017 Jan 4;7:1-18.

Jayasinghe, M., et al. The Role of Diet and Gut Microbiome in Multiple Sclerosis. Cureus. 2022 Sep 9;14(9):e28975.

Kim-Lee, C., et al. Gastrointestinal disease in Sjogren's syndrome: related to food hypersensitivities. Springerplus. 2015 Dec 12;4:766.

Martinelli, V., et al. Gut-oriented interventions in patients with multiple sclerosis: fact or fiction? Eur Rev Med Pharmacol Sci. 2022 Feb;26(3):935-946.

Melbye, P., et al. Short-chain fatty acids and gut microbiota in multiple sclerosis. Acta Neurol Scand. 2019 Mar;139(3):208-219.

Pitt, D., et al. Glutamate excitotoxicity in a model of multiple sclerosis. Nat Med. 2000 Jan;6(1):67-70.

Sanchez, A., et al. Role of sugars in human neutrophilic phagocytosis. Am J Clin Nutr. 1973 Nov;26(11):1180-4.

Severance, E.G., et al. IgG dynamics of dietary antigens point to cerebrospinal fluid barrier or flow dysfunction in first-episode schizophrenia. Brain Behav Immun. 2015 Feb;44:148-58.

Stagi, S., et al. Determinants of vitamin D levels in children, adolescents, and young adults with juvenile idiopathic arthritis. J Rheumatol. 2014;41(9):1884-92.

Titcomb, T.J., et al. Change in Micronutrient Intake among People with Relapsing-Remitting Multiple Sclerosis Adapting the Swank and Wahls Diets: An Analysis of Weighed Food Records. Nutrients. 2021 Oct 5;13(10):3507.

Titcomb, T.J., et al. Eating Pattern and Nutritional Risks among People with Multiple Sclerosis Following a Modified Paleolithic Diet. Nutrients. 2020 Jun 20;12(6):1844.

Wahls, T.L. Dietary Approaches to Treating Multiple Sclerosis-Related Symptoms. Phys Med Rehabil Clin N Am. 2022 Aug;33(3):605-620.

Wahls, T.L., et al. Dietary approaches to treat MS-related fatigue: comparing the modified Paleolithic (Wahls Elimination) and low saturated fat (Swank) diets on perceived fatigue in persons with relapsing-remitting multiple sclerosis: study protocol for a randomized controlled trial.Trials. 2018 Jun 4;19(1):309.

Wahls, T.L., et al. Impact of the Swank and Wahls elimination dietary interventions on fatigue and quality of life in relapsing-remitting multiple sclerosis: The WAVES randomized parallel-arm clinical trial. Mult Scler J Exp Transl Clin. 2021 Jul 31;7(3):20552173211035399.

Wahls, T.L., et al. Review of Two Popular Eating Plans within the Multiple Sclerosis Community: Low Saturated Fat and Modified Paleolithic. Nutrients. 2019 Feb 7;11(2):352.

Wang, X., et al. Role of Gut Microbiota in Multiple Sclerosis and Potential Therapeutic Implications. Curr Neuropharmacol. 2022;20(7):1413-1426.

Watt, T., et al. Is Thyroid Autoimmunity per se a Determinant of Quality of Life in Patients with Autoimmune Hypothyroidism? Eur Thyroid J. 2012;1(3):186-92.

Biomagnetism Therapy

Sources & References

Damayanov, C. IT’S TIME TO CHANGE THE CONCEPT OF CANCER TREATMENT. SOME NEW PERSPECTIVES. ResearchGate. 2021 Jul.

Frank, B.L. Biomagnetic Pair Therapy and Typhoid Fever: A Pilot Study. Med Acupunct. 2017 Oct 1;29(5):308-312.

Birth Trauma

Sources & References

Ang, E.S.B.C., Jr., et al. Prenatal exposure to ultrasound waves impacts neuronal migration in mice. Proc Natl Acad Sci U S A. 2006 Aug 22;103(34):12903-10.

Chaparro, C.M., et al. Effect of timing of umbilical cord clamping on iron status in Mexican infants: a randomised controlled trial. Lancet. 2006 Jun 17;367(9527):1997-2004.

Curran, E.A., et al. Research review: Birth by caesarean section and development of autism spectrum disorder and attention-deficit/hyperactivity disorder: a systematic review and meta-analysis. J Child Psychol Psychiatry. 2015 May;56(5):500-8.

Fezer, G.F., et al. Perinatal Features of Children with Autism Spectrum Disorder. Rev Paul Pediatr. 2017 Apr-Jun;35(2):130-135.

Frisch, M., et al. Ritual circumcision and risk of autism spectrum disorder in 0- to 9-year-old boys: national cohort study in Denmark. J R Soc Med. 2015 Jul;108(7):266-79.

Gardender, H., et al. Perinatal and neonatal risk factors for autism: a comprehensive meta-analysis. Pediatrics. 2011 Aug;128(2):344-55.

Glasson, E.J., et al. Perinatal factors and the development of autism: a population study. Arch Gen Psychiatry. 2004 Jun;61(6):618-27.

González-Valenzuela, M.J., et al. Exposure to synthetic oxytocin during delivery and its effect on psychomotor development. Dev Psychobiol. 2015 Dec;57(8):908-20.

Hutton, E.K., et al. Late vs early clamping of the umbilical cord in full-term neonates: systematic review and meta-analysis of controlled trials. JAMA. 2007 Mar 21;297(11):1241-52.

Korpela, K., et al. Maternal Fecal Microbiota Transplantation in Cesarean-Born Infants Rapidly Restores Normal Gut Microbial Development: A Proof-of-Concept Study. Cell, 2020.

Kurth, L., et al. Perinatal Pitocin as an early ADHD biomarker: neurodevelopmental risk? J Atten Disord. 2011 Jul;15(5):423-31.

Mercer, J.S., et al. The Effects of Delayed Cord Clamping on 12-Month Brain Myelin Content and Neurodevelopment: A Randomized Controlled Trial. Am J Perinatol. 2022 Jan;39(1):37-44.

Modabbernia, A., et al. Apgar score and risk of autism. Eur J Epidemiol. 2018 Oct 5.

Qiu, C., et al. Association Between Epidural Analgesia During Labor and Risk of Autism Spectrum Disorders in Offspring. JAMA Pediatr. 2020 Oct 12.

Rosman, N.P., et al. Association of Prenatal Ultrasonography and Autism Spectrum Disorder. JAMA Pediatr. 2018 Apr 1;172(4):336-344.

Taddio, A., et al. Effect of neonatal circumcision on pain responses during vaccination in boys. Lancet. 1995 Feb 4;345(8945):291-2.

Taige, N.M., et al. Gestational Age at Term, Delivery Circumstance, and Their Association with Childhood Attention Deficit Hyperactivity Disorder Symptoms. Paediatr Perinat Epidemiol. 2016 Mar;30(2):171-80.

Torres, G., et al. Perspectives of Pitocin administration on behavioral outcomes in the pediatric population: recent insights and future implications. Heliyon. 2020 May 29;6(5):e04047.

Tollånes, M.C., et al. Cesarean section and risk of severe childhood asthma: a population-based cohort study. J Pediatr. 2008 Jul;153(1):112-6.

Watai,, K., et al. Epidemiological association between multiple chemical sensitivity and birth by caesarean section: a nationwide case-control study. Environ Health. 2018 Dec 14;17(1):89

Willfors, C., et al. Medical history of discordant twins and environmental etiologies of autism. Transl Psychiatry. 2017 Jan; 7(1): e1014.

Williams, E.L., et al. Potential teratogenic effects of ultrasound on corticogenesis: implications for autism. Med Hypotheses. 2010 Jul;75(1):53-8.

Zaigham, M., et al. Prelabour caesarean section and neurodevelopmental outcome at 4 and 12 months of age: an observational study. BMC Pregnancy and Childbirth. 2020 (20)564.

Brain Harmony

Sources & References

Amos, P. Rhythm and timing in autism: learning to dance. Front Integr Neurosci. 2013 Apr 19;7:27.

Barnhill, E. Neural connectivity, music, and movement: a response to Pat Amos. Front Integr Neurosci. 2013 Apr 24;7:29.

Bartscherer, et al. Interactive metronome training for a 9-year-old boy with attention and motor coordination difficulties. Physiother Theory Pract. Oct-Dec 2005;21(4):257-69.

Blondis, T.A. Motor disorders and attention-deficit/hyperactivity disorder. Pediatr Clin North Am. 1999 Oct;46(5):899-913, vi-vii.

Cho, H., et al. Effects of Action Observation Training with Auditory Stimulation on Static and Dynamic Balance in Chronic Stroke Patients. J Stroke Cerebrovasc Dis. 2020 May;29(5):104775.

Cosper, S.M., et al. Interactive Metronome training in children with attention deficit and developmental coordination disorders. Int J Rehabil Res. 2009 Dec;32(4):331-6.

Goulardins, J.B., et al. Attention deficit hyperactivity disorder and developmental coordination disorder: Two separate disorders or do they share a common etiology. Behav Brain Res. 2015 Oct 1;292:484-92.

Goulardins, J.B., et al. Attention Deficit Hyperactivity Disorder and Motor Impairment. Percept Mot Skills. 2017 Apr;124(2):425-440.

Grigg, T.M., et al. Retained primitive reflexes: Perceptions of parents who have used Rhythmic Movement Training with their children. J Child Health Care. 2018 Sep;22(3):406-418.

Grzywniak, C. Integration exercise programme for children with learning difficulties who have preserved vestigial primitive reflexes. Acta Neuropsychologica. 2017;15(3).

Hardy, M.W., et al. Rhythm, movement, and autism: using rhythmic rehabilitation research as a model for autism. Front Integr Neurosci. 2013 Mar 28;7:19.

Herbert, J., et al. Crawling is associated with more flexible memory retrieval by 9-month-old infants. Dev Sci. 2007 Mar;10(2):183-9.

Hong, H.J., et al. Effect of Rhythmic Movement Program to Improve Walking Ability for Elderly Patients with Stroke. Indian Journal of Science and Technology. 2016 Jul;9(26).

Iverson, J.M. Developing language in a developing body: the relationship between motor development and language development. J Child Lang. 2010 Mar;37(2):229-61.

Jordan-Black, J. The effects of the Primary Movement programme on the academic performance of children attending ordinary primary school. Journal of Research in Special Educational Needs. 2005 Nov;5(3):101 - 111.

Kadivar, Z., et al. Effect of step training and rhythmic auditory stimulation on functional performance in Parkinson patients. Neurorehabil Neural Repair. 2011 Sep;25(7):626-35.

Karatekin, C., et al. A preliminary study of motor problems in children with attention-deficit/hyperactivity disorder. Percept Mot Skills. 2003 Dec;97(3 Pt 2):1267-80.

Koomar, J., et al. Theoretical and clinical perspectives on the Interactive Metronome: a view from occupational therapy practice. Am J Occup Ther. Mar-Apr 2001;55(2):163-6.

Ladányi, K. et al. Is atypical rhythm a risk factor for developmental speech and language disorders? Wiley Interdiscip Rev Cogn Sci. 2020 Sep;11(5):e1528.

Lakatos, P., et al. A New Unifying Account of the Roles of Neuronal Entrainment. Curr Biol. 2019 Sep 23;29(18):R890-R905.

McLeod, K.R., et al. Functional connectivity of neural motor networks is disrupted in children with developmental coordination disorder and attention-deficit/hyperactivity disorder. Neuroimage Clin. 2014 Mar 26;4:566-75.

McWhirter, K., et al. The association between learning disorders, motor function, and primitive reflexes in pre-school children: A systematic review. J Child Health Care. 2022 Jul 13;13674935221114187.

Melillo, R., et al. Persistent Childhood Primitive Reflex Reduction Effects on Cognitive, Sensorimotor, and Academic Performance in ADHD. Front Public Health. 2020 Nov 17;8:431835.

Piek, J.P., et al. Motor coordination and kinaesthesis in boys with attention deficit-hyperactivity disorder. Dev Med Child Neurol. 1999 Mar;41(3):159-65.

Shaffer, R.J., et al. Effect of interactive metronome training on children with ADHD. Am J Occup Ther. Mar-Apr 2001;55(2):155-62.

Slater, J.L., et al. Timing Deficits in ADHD: Insights From the Neuroscience of Musical Rhythm. Front Comput Neurosci. 2018 Jul 6;12:51.

Smith, A., et al. Evidence for a pure time perception deficit in children with ADHD. J Child Psychol Psychiatry. 2002 May;43(4):529-42.

Sonuga-Barke, E., et al. Beyond the dual pathway model: evidence for the dissociation of timing, inhibitory, and delay-related impairments in attention-deficit/hyperactivity disorder. J Am Acad Child Adolesc Psychiatry. 2010 Apr;49(4):345-55.

Suh, J.H., et al. Effect of rhythmic auditory stimulation on gait and balance in hemiplegic stroke patients. NeuroRehabilitation. 2014;34(1):193-9.

Teicher, M.H. Final White Paper: Effects of Brain Balance Exercisesand Interactive Metronome on Children with Attention Deficit Hyperactivity Disorderare Similar to the Effects of Stimulant Medication. Harvard Medical School, Department of Psychiatry. 2020.

Toplak, M.E., et al. Time perception: modality and duration effects in attention-deficit/hyperactivity disorder (ADHD). J Abnorm Child Psychol. 2005 Oct;33(5):639-54.

Van Hirtum, T., et al. Is atypical rhythm a riskfactor for developmental speech and language disorders? J Assoc Res Otolaryngol. 2021 Jul;22(4):465-480.

Winkler, I., et al. Newborn infants detect the beat in music. Proc Natl Acad Sci U S A. 2009 Feb 17;106(7):2468-71.

Yang, B., et al. Time perception deficit in children with ADHD. Brain Res. 2007 Sep 19;1170:90-6.

Zaigham, M., et al. Prelabour caesarean section and neurodevelopmental outcome at 4 and 12 months of age: an observational study. BMC Pregnancy and Childbirth. 2020 (20)564.

Zentner, M., et al. Rhythmic engagement with music in infancy. Proc Natl Acad Sci U S A. 2010 Mar 30;107(13):5768-73.

Breastfeeding

Cancer

Sources & References

Accurso, A., et al. Dietary carbohydrate restriction in type 2 diabetes mellitus and metabolic syndrome: time for a critical appraisal. Nutr Metab (Lond). 2008 Apr 8:5:9.

Aggarwal, B.B., et al. Molecular targets of dietary agents for prevention and therapy of cancer. Biochem Pharmacol. 2006 May 14;71(10):1397-421.

Allen, B.G., et al. Ketogenic diets as an adjuvant cancer therapy: History and potential mechanism. Redox Biol. 2014:2:963-70.

Anand, P., et al. Cancer is a preventable disease that requires major lifestyle changes. Pharm Res. 2008 Sep;25(9):2097-116.

Anemoulis, M., et al. Intermittent Fasting in Breast Cancer: A Systematic Review and Critical Update of Available Studies. Nutrients. 2023 Feb; 15(3): 532.

Bahr, L.S., et al. Ketogenic diet and fasting diet as Nutritional Approaches in Multiple Sclerosis (NAMS): protocol of a randomized controlled study. Trials. 2020 Jan 2;21(1):3.

Coller, H.A. Is cancer a metabolic disease? Am J Pathol. 2014 Jan;184(1):4-17.

Duan, W., et al. Hyperglycemia, a neglected factor during cancer progression. Biomed Res Int. 2014:2014:461917.

Giovannucci, E., et al. Diabetes and cancer: a consensus report. Diabetes Care. 2010 Jul;33(7):1674-85.

Gunatilake, S., et al. Glyphosate’s Synergistic Toxicity in Combination with Other Factors as a Cause of Chronic Kidney Disease of Unknown Origin. Int J Environ Res Public Health. 2019 Jul 31;16(15):2734.

Kundi, M., et al. Mobile phone specific radiation disturbs cytokinesis and causes cell death but not acute chromosomal damage in buccal cells: Results of a controlled human intervention study. Environ Res. 2024 Jun 15;251(Pt 1):118634.

Labbé, D.P., et al. Role of diet in prostate cancer: the epigenetic link. Oncogene. 2015 Sep 3;34(36):4683-91.

Liu, H., et al. Refined fructose and cancer. Expert Opin Ther Targets. 2011 Sep;15(9):1049-59.

Lombardi, C., et al. Residential proximity to pesticide application as a risk factor for childhood central nervous system tumors. Environ Res. 2021 Jun;197:111078.

Lorenzi, M., et al. High glucose induces DNA damage in cultured human endothelial cells. J Clin Invest. 1986 Jan;77(1):322-5.

Mathew, A. Allostatic Load in Cancer: A Systematic Review and Mini Meta-Analysis. Biol Res Nurs. 2021 Jul;23(3):341-361.

Morley, W.A., et al. Diminished brain resilience syndrome: A modern day neurological pathology of increased susceptibility to mild brain trauma, concussion, and downstream neurodegeneration. Surg Neurol Int. 2014 Jun 18;5:97.

Onodera, Y., et al. Increased sugar uptake promotes oncogenesis via EPAC/RAP1 and O-GlcNAc pathways. J Clin Invest. 2014 Jan;124(1):367-84.

Pedretti, L., et al. Role of Nutrition in Pediatric Patients with Cancer. Nutrients. 2023 Feb; 15(3): 710.

Samsel, A, et al. Glyphosate’s suppression of cytochrome P450 enzymes and amino acid biosynthesis by the gut microbiome: Pathways to modern diseases. Entropy. 2013;15:1416–1463.

Seyfried, T.N. Cancer as a metabolic disease. Nutr Metab (Lond). 2010 Jan 27:7:7.

Seyfried, T.N. Cancer as a metabolic disease: implications for novel therapeutics. Carcinogenesis. 2014 Mar;35(3):515-27.

Seyfried, T.N., et al. Consideration of Ketogenic Metabolic Therapy as a Complementary or Alternative Approach for Managing Breast Cancer. Front Nutr. 2020 Mar 11:7:21.

Seyfried, T.N., et al. Is the restricted ketogenic diet a viable alternative to the standard of care for managing malignant brain cancer? Epilepsy Res. 2012 Jul;100(3):310-26.

Seyfried, T.N., et al. Metabolic management of brain cancer. Biochim Biophys Acta. 2011 Jun;1807(6):577-94.

Takano, T., et al. Glutamate release promotes growth of malignant gliomas. Nat Med. 2001 Sep;7(9):1010-5.

Tinkum, K.L., et al. Fasting protects mice from lethal DNA damage by promoting small intestinal epithelial stem cell survival. Proc Natl Acad Sci U S A. 2015 Dec 22;112(51):E7148-54.

Chronic/Recurrent Infections

Sources & References

Nsouli, T.M. The role of food allergy in serious otitis media. Ann Allergy. 1991; 66:91

Pfotenhauer, K.M., et al. Vitamin D Deficiency, Its Role in Health and Disease, and Current Supplementation Recommendations. J Am Osteopath Assoc. 2017 May 1;117(5):301-305.

Colic

Sources & References

Aguilera, M., Cerda-Cuellar, M., Martinez, V. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice. Gut Microbes. 2015;6(1):10-23.

Aroniadis, O.C., Brandt, L.J. Fecal microbiota transplantation: past, present and future. Curr Opin Gastroenterol. 2013;29(1):79-84.

Assa, A., Vong, L., Pinnell, L.J., Avitzur, N., Johnson-Henry, K.C., Sherman, P.M. Vitamin D deficiency promotes epithelial barrier dysfunction and intestinal inflammation. J Infect Dis. 2014;210(8):1296-305.

Azad, Meghan B., et al. Perinatal Programming of Asthma: The Role of Gut Microbiota. Clin Dev Immunol. 2012; 2012: 932072.

Bag, Ozelem, et al. Is it infant colic or early symptom of autistic spectrum disorder? Pediatr Int 23 Mar 2018.

Buccigrossi, V., Nicastro, E., Guarino, A. Functions of intestinal microflora in children. Curr Opin Gastroenterol. 2013;29(1):31-8.

Carding, S., Verbeke, K., Vipond, D.T., Corfe, B.M., Owen, L.J. Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis. 2015;26:26191.

Cucchiara, S., Stronati, L., Aloi, M. Interactions between intestinal microbiota and innate immune system in pediatric inflammatory bowel disease. J Clin Gastroenterol. 2012;46 Suppl:S64-6.

Fukuda, K., Fujita, Y. Determination of the discriminant score of intestinal microbiota as a biomarker of disease activity in patients with ulcerative colitis. BMC Gastroenterol. 2014;14:49.

Galland, L. The gut microbiome and the brain. J Med Food 2014; 17(12): 1261-72.

Guandalini, S. Are probiotics or prebiotics useful in pediatric irritable bowel syndrome or inflammatory bowel disease? Front Med (Lausanne). 2014;1:23.

Guandalini, S., Cernat, E., Moscoso, D. Prebiotics and probiotics in irritable bowel syndrome and inflammatory bowel disease in children. Benef Microbes. 2015;6(2):209-17.

Jakobsen, C., Paerregaard, A., Munkholm, P., Wewer, V. Environmental factors and risk of developing paediatric inflammatory bowel disease — a population based study. 2007-2009. J Crohns Colitis. 2013;7(1):79-88.

Manichanh, C., Borruel, N., Casellas, F., Guarner, F. The gut microbiota in IBD. Nat Rev Gastroenterol Hepatol. 2012;9(10):599-608.

Nocerino, R., et al. The therapeutic efficacy of Bifidobacterium animalis subsp. lactis BB‐12® in infant colic: A randomised, double blind, placebo‐controlled trial. Alimentary Pharmacology & Therapeutics. Dec 2019.

Savino, F., et al. Lactobacillus reuteri (American Type Culture Collection Strain 55730) versus simethicone in the treatment of infantile colic: a prospective randomized study. Pediatrics. 2007 Jan;119(1):e124-30.

Sela, D.A., Mills, D.A. The marriage of nutrigenomics with the microbiome: the case of infant associated bifidobacteria and milk. Am J Clin Nutr. 2014;99(3):697S-703S.

Stiemsma, Leah T., et al. The Role of the Microbiome in the Developmental Origins of Health and Disease. Pediatrics. Mar 2018.

West, C.E., Renz, H., Jenmalm, M.C., Kozyrskyj, A.L., Allen, K.J., Vuillermin, P., et al. The gut microbiota and inflammatory noncommunicable diseases: associations and potentials for gut microbiota therapies. J Allergy Clin Immunol. 2015;135(1):3-13; quiz 4.

Common Genetic Mutations

Sources & References

Arnold, P.A., et al. Glutamate transporter gene SLC1A1 associated with obsessive-compulsive disorder. Arch Gen Psychiatry. 2006 Jul;63(7):769-76.

Bidwell, L.C., et al. Genetic influences on ADHD symptom dimensions: Examination of a priori candidates, gene-based tests, genome-wide variation, and SNP heritability. Am J Med Genet B Neuropsychiatr Genet. 2017 Jun;174(4):458-466.

Bowers, K., et al. Glutathione pathway gene variation and risk of autism spectrum disorders. J Neurodev Disord. 2011 Jun;3(2):132-43.

Esmaiel, N.N., et al. The potential impact of COMT gene variants on dopamine regulation and phenotypic traits of ASD patients. Behav Brain Res. 2020 Jan 27;378:112272.

Hausman-Cohen, S., et al. Utilizing Genomically Targeted Molecular Data to Improve Patient-Specific Outcomes in Autism Spectrum Disorder. Int J Mol Sci. 2022 Feb 16;23(4):2167.

Hausman-Cohen, S.R., et al. Genomics of Detoxification: How Genomics can be Used for Targeting Potential Intervention and Prevention Strategies Including Nutrition for Environmentally Acquired Illness. J Am Coll Nutr. 2020 Feb;39(2):94-102.

Hwang, I.W., et al. Association of Monoamine Oxidase A (MAOA) Gene uVNTR and rs6323 Polymorphisms with Attention Deficit and Hyperactivity Disorder in Korean Children. Medicina (Kaunas). 2018 May 18;54(3):32.

Li, Y., et al. Association between MTHFR C677T/A1298C and susceptibility to autism spectrum disorders: a meta-analysis. BMC Pediatrics. 2020(20)449.

Meng, X., et al. Association between MTHFR (677C>T and 1298A>C) polymorphisms and psychiatric disorder: A meta-analysis. PLoS One. 2022 Jul 14;17(7):e0271170.

Rahbar, M.H., et al. Detoxification Role of Metabolic Glutathione S-Transferase (GST) Genes in Blood Lead Concentrations of Jamaican Children with and without Autism Spectrum Disorder. Genes (Basel). 2022 May 29;13(6):975.

Sadeghiyeh. T., et al. Association of MTHFR 677C > T and 1298A > C polymorphisms with susceptibility to attention deficit and hyperactivity disorder. Fetal Pediatr Pathol. 2020 Oct;39(5):422-429.

Way, H., et al. Genomics as a Clinical Decision Support Tool: Successful Proof of Concept for Improved ASD Outcomes. J Pers Med. 2021 Jun 24;11(7):596.

Constipation and Diarrhea

Sources & References

Coregulation

Sources & References

Ashhad, S., et al. Breathing Rhythm and Pattern and Their Influence on Emotion. Annu Rev Neurosci. 2022 Jul 8:45:223-247.

Chevalier, G., et al. Earthing (grounding) the human body reduces blood viscosity-a major factor in cardiovascular disease. J Altern Complement Med. 2013 Feb;19(2):102-10.

Chevalier, G., et al. Earthing: health implications of reconnecting the human body to the Earth's surface electrons. J Environ Public Health. 2012;2012:291541.

Cramer, H., et al. Yoga for anxiety: A systematic review and meta-analysis of randomized controlled trials. Depress Anxiety. 2018 Sep;35(9):830-843.

do Valle, S., et al. Acupuncture Treatment for Generalized Anxiety Disorder by Activating the Vagus Nerve and Improving Heart-Rate Variability and Heart-Rhythm Coherence, A Case-Series Study. Med Acupunct. 2024 Feb 1;36(1):21-26.

Koniver L. Practical applications of grounding to support health. Biomed J. 2023;46(1):41–47.

Lemay, V., et al. Impact of a Yoga and Meditation Intervention on Students' Stress and Anxiety Levels. Am J Pharm Educ. 2019 Jun;83(5):7001.

Martel, J. Is a return to nature a piece of the health puzzle? Biomed J. 2023 Feb;46(1):8-10.

Oschman, J.L., et al. The effects of grounding (earthing) on inflammation, the immune response, wound healing, and prevention and treatment of chronic inflammatory and autoimmune diseases. J Inflamm Res. 2015 Mar 24;8:83-96.

Oschman, J.L. Illnesses in technologically advanced societies due to lack of grounding (earthing). Biomed J. 2023 Feb;46(1):17-29.

Saeed, S.A., et al. Depression and Anxiety Disorders: Benefits of Exercise, Yoga, and Meditation. Am Fam Physician. 2019 May 15;99(10):620-627.

Sinatra, S.T., et al. Electric Nutrition: The Surprising Health and Healing Benefits of Biological Grounding (Earthing). Altern Ther Health Med. 2017 Sep;23(5):8-16.

Developmental Delays

Sources & References

Adams, L.J., et al. Infant feeding method and special educational need in 191,745 Scottish schoolchildren: A national, population cohort study. PLoS Med. 2023 Apr 6;20(4):e1004191.

Aguilera, M., et al. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice. Gut Microbes. 2015;6(1):10-23.

Darling, A.L., et al. Association between maternal vitamin D status in pregnancy and neurodevelopmental outcomes in childhood: results from the Avon Longitudinal Study of Parents and Children (ALSPAC). Br J Nutr 2017 Jun;117(12):1682-1692.

Egset, K., et al. Magno App: Exploring Visual Processing in Adults with High and Low Reading Competence. Scandinavian Journal of Educational Research. 07 Jan 2020.

Elliott, C., et al. Early Moves: A protocol for a population-based prospective cohort study to establish General Movements as an early biomarker of cognitive impairment in infants. BMJ Open. 2021 Apr 9;11(4):e041695.

Malin, A.J., et al. Maternal Urinary Fluoride and Child Neurobehavior at Age 36 Months. JAMA Netw Open. 2024 May 1;7(5):e2411987.

Mercer, J.S., et al. The Effects of Delayed Cord Clamping on 12-Month Brain Myelin Content and Neurodevelopment: A Randomized Controlled Trial. Am J Perinatol. 2022 Jan;39(1):37-44.

Morris, C.R., et al. Syndrome of allergy, apraxia, and malabsorption: characterization of a neurodevelopmental phenotype that responds to omega 3 and vitamin E supplementation. Alternative Therapies in Health and Medicine. Jul-Aug 2009;15(4):34-43.

Ozonoff, S., et al. Onset patterns in autism: Variation across informants, methods, and timing. Autism Res. 2018 Mar 10.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Zablotsky, B., et al. Prevalence and Trends of Developmental Disabilities among Children in the United States: 2009-2017. Pediatrics. 2019 Oct;144(4):e20190811.

Zaigham, M., et al. Prelabour caesarean section and neurodevelopmental outcome at 4 and 12 months of age: an observational study. BMC Pregnancy and Childbirth. 2020 (20)564.

Diabetes/Obesity

Sources & References

Accurso, A., et al. Dietary carbohydrate restriction in type 2 diabetes mellitus and metabolic syndrome: time for a critical appraisal. Nutr Metab (Lond). 2008 Apr 8:5:9.

Acuña-Catalán, D., et al. Ketogenic diet administration later in life improves memory by modifying the synaptic cortical proteome via the PKA signaling pathway in aging mice. Cell Rep Med. 2024 Jun 18;5(6):101593.

Adebayo, O., et al. The changing face of diabetes in America. Emerg Med Clin North Am. 2014;32(2):319-27.

Aggarwal, B.B., et al. Molecular targets of dietary agents for prevention and therapy of cancer. Biochem Pharmacol. 2006 May 14;71(10):1397-421.

Allen, B.G., et al. Ketogenic diets as an adjuvant cancer therapy: History and potential mechanism. Redox Biol. 2014:2:963-70.

Anand, P., et al. Cancer is a preventable disease that requires major lifestyle changes. Pharm Res. 2008 Sep;25(9):2097-116.

Aucoin, M., et al. Generalized anxiety disorder and hypoglycemia symptoms improved with diet modificationCase Rep Psychiatry. 2016;2016.

Bădescu, S.V., et al. The association between Diabetes mellitus and Depression. J Med Life. 2016 Apr-Jun; 9(2): 120–125.

Bahr, L.S., et al. Ketogenic diet and fasting diet as Nutritional Approaches in Multiple Sclerosis (NAMS): protocol of a randomized controlled study. Trials. 2020 Jan 2;21(1):3.

Banerjee, S., et al. Ayurveda in changing scenario of diabetes management for developing safe and effective treatment choices for the future. J Complement Integr Med. 2015.

Basic, M., et al. Obesity: genome and environment interactions. Arh Hig Rada Toksikol. 2012;63(3):395-405.

Bernstein, R.K. Blood glucose self-monitoring by diabetic patients: refinements of procedural technique. Diabetes Care. 1979 Mar-Apr;2(2):233-6.

Bernstein, R.K. Virtually continuous euglycemia for 5 yr in a labile juvenile-onset diabetic patient under noninvasive closed-loop control. Diabetes Care. 1980 Jan-Feb;3(1):140-3.

Bipartisan Policy Center. Lots to lose how America’s health and obesity crisis threatens our economic future. Washington, D.C.: Bipartisan Policy Center,; 2012.

Boison, D. New insights into the mechanisms of the ketogenic diet. Curr Opin Neurol. 2017 Apr;30(2):187-192.

Bostock, E.C.S., et al. The Current Status of the Ketogenic Diet in Psychiatry. Front Psychiatry. 2017 Mar 20:8:43.

Bradford, B.L., et al. Mitochondrial Dysfunction and Type 2 Diabetes. Science. 2005 Jan 21;307(5708):384-7.

Brenton, J.N., et al. Phase II study of ketogenic diets in relapsing multiple sclerosis: safety, tolerability and potential clinical benefits. J Neurol Neurosurg Psychiatry. 2022 Jun;93(6):637-644.

Brenton, J.N., et al. Pilot study of a ketogenic diet in relapsing-remitting MS. Neurol Neuroimmunol Neuroinflamm. 2019 Apr 12;6(4):e565.

Carlson, J.A., et al. Dietary-related and physical activity-related predictors of obesity in children: a 2-year prospective study. Child Obes. 2012;8(2):110-5.

Centers for Disease Control and Prevention. All About Your A1C.

Centers for Disease Control and Prevention. Diabetes Tests.

Centers for Disease Control and Prevention. National Diabetes Statistics Report, 2020: Estimates of Diabetes and Its Burden in the United States.

Cheng, N., et al. Metabolic Dysfunction Underlying Autism Spectrum Disorder and Potential Treatment Approaches. Front Mol Neurosci. 2017 Feb 21:10:34.

Choquet, H., et al. Genomic insights into early-onset obesity. Genome Med. 2010;2(6):36.

Classen, J.B. Review of evidence that epidemics of type 1 diabetes and type 2 diabetes/metabolic syndrome are polar opposite responses to iatrogenic inflammation. Curr Diabetes Rev. 2012;8(6):413-8.

Coller, H.A. Is cancer a metabolic disease? Am J Pathol. 2014 Jan;184(1):4-17.

Cortese, S., et al. Attention-deficit/hyperactivity disorder, iron deficiency, and obesity: is there a link? Postgrad Med. 2014;126(4):155-70.

Cui, J., et al. Butyrate-Producing Bacteria and Insulin Homeostasis: The Microbiome and Insulin Longitudinal Evaluation Study (MILES). Diabetes. 2022 Nov 1;71(11):2438-2446.

Danan, K., et al. The Ketogenic Diet for Refractory Mental Illness: A Retrospective Analysis of 31 Inpatients. Front Psychiatry. 2022 Jul 6:13:951376.

deCampo, D.M., et al. Ketogenic dietary therapies for epilepsy and beyond. Curr Opin Clin Nutr Metab Care. 2019 Jul;22(4):264-268.

de la Rubia Orti, J.E., et al. Can Ketogenic Diet Improve Alzheimer’s Disease? Association With Anxiety, Depression, and Glutamate System. Front Nutr. 2021 Oct 27:8:744398.

de la Rubia Orti, J.E., et al. Exploring the impact of ketogenic diet on multiple sclerosis: obesity, anxiety, depression, and the glutamate system. Front Nutr. 2023 Aug 25:10:1227431.

Desai, J.R., et al. Diabetes and asthma case identification, validation, and representativeness when using electronic health data to construct registries for comparative effectiveness and epidemiologic research. Med Care. 2012;50 Suppl:S30-5.

Dietch, D.M., et al. Efficacy of low carbohydrate and ketogenic diets in treating mood and anxiety disorders: systematic review and implications for clinical practice. BJPsych Open. 2023 Apr 17;9(3):e70.

Duan, W., et al. Hyperglycemia, a neglected factor during cancer progression. Biomed Res Int. 2014:2014:461917.

Dyńka, D., et al. The Role of Ketogenic Diet in the Treatment of Neurological Diseases. Nutrients. 2022 Nov 24;14(23):5003.

Feinman, R.K., et al. Dietary carbohydrate restriction as the first approach in diabetes management: critical review and evidence base. Nutrition. 2015 Jan;31(1):1-13.

Giovannucci, E., et al. Diabetes and cancer: a consensus report. Diabetes Care. 2010 Jul;33(7):1674-85.

Gough, S., et al. Neuroprotection by the Ketogenic Diet: Evidence and Controversies. Front Nutr. 2021 Nov 23:8:782657.

Grammes, J., et al. Fear of hypoglycemia in patients with type 2 diabetes: The role of interoceptive accuracy and prior episodes of hypoglycemia. J Psychosom Res. 2018;105:58-63.

Grochowska, K., et al. The Effect of the Ketogenic Diet on the Therapy of Neurodegenerative Diseases and Its Impact on Improving Cognitive Functions. Dement Geriatr Cogn Dis Extra. 2022 May 30;12(2):100-106.

Han, H., et al. Gut Microbiota and Type 1 Diabetes. Int J Mol Sci. 2018 Mar 27;19(4):995.

Hao, J., et al. Mitochondrial nutrients improve immune dysfunction in the type 2 diabetic Goto-Kakizaki rats. J Cell Mol Med. 2009 Apr;13(4):701-11.

He, C., et al. Targeting gut microbiota as a possible therapy for diabetes. Nutr Res. 2015.

Hinzmann, R., et al. What do we need beyond hemoglobin A1c to get the complete picture of glycemia in people with diabetes? Int J Med Sci. 2012;9(8):665-81.

Irish, A.K., et al. Randomized control trial evaluation of a modified Paleolithic dietary intervention in the treatment of relapsing-remitting multiple sclerosis: a pilot study. Degener Neurol Neuromuscul Dis. 2017 Jan 4;7:1-18.

Jiang, Z., et al. Effects of Ketogenic Diet on Neuroinflammation in Neurodegenerative Diseases. Aging Dis. 2022 Jul 11;13(4):1146-1165.

Kamba, A., et al. Association between higher serum cortisol levels and decreased insulin secretion in a general populationPLOS ONE. 2016;11(11):e0166077.

Kapoor, D., et al. Emerging Role of the Ketogenic Dietary Therapies beyond Epilepsy in Child Neurology. Ann Indian Acad Neurol. 2021 Jul-Aug;24(4):470-480.

Karkafi, R.E., et al. Ketogenic Diet and Inflammation: Implications for Mood and Anxiety Disorders. Adv Exp Med Biol. 2023:1411:537-554.

Kogut, S.J., et al. Evaluation of a program to improve diabetes care through intensified care management activities and diabetes medication copayment reduction. J Manag Care Pharm. 2012;18(4):297-310.

Kong., A.P, et al. Diabetes and its comorbidities–where East meets West. Nat Rev Endocrinol. 2013;9(9):537-47.

Labbé, D.P., et al. Role of diet in prostate cancer: the epigenetic link. Oncogene. 2015 Sep 3;34(36):4683-91.

Laron, Z. Interplay between heredity and environment in the recent explosion of type 1 childhood diabetes mellitus. Am J Med Genet. 2002;115(1):4-7.

Lee, C., et al. Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index. Cancer Res. 2010 Feb 15;70(4):1564-72.

Lee, R.W.Y., et al. A modified ketogenic gluten-free diet with MCT improves behavior in children with autism spectrum disorder. Physiol Behav. 2018 May 1:188:205-211.

Lefevre, F., et al. Ketogenic diet for the treatment of refractory epilepsy in children: A systematic review of efficacy. Pediatrics. 2000 Apr;105(4):E46.

Lennerz, B.S., et al. Management of Type 1 Diabetes With a Very Low-Carbohydrate Diet.
Pediatrics. 2018 Jun;141(6):e20173349.

Levi, J., et al. F as in fat how obesity threatens America’s future : 2012. Washington, D.C.: Trust for America’s Health; 2012.

Li, Q., et al. A Ketogenic Diet and the Treatment of Autism Spectrum Disorder. Front Pediatr. 2021 May 11:9:650624.

Liu, H., et al. Refined fructose and cancer. Expert Opin Ther Targets. 2011 Sep;15(9):1049-59.

Lorenzi, M., et al. High glucose induces DNA damage in cultured human endothelial cells. J Clin Invest. 1986 Jan;77(1):322-5.

Ly, N.P., et al. Gut microbiota, probiotics, and vitamin D: interrelated exposures influencing allergy, asthma, and obesity? J Allergy Clin Immunol. 2011;127(5):1087-94; quiz 95-6.

Magrone, T., et al. Childhood obesity: immune response and nutritional approaches. Front Immunol. 2015;6:76.

Mentzelou, M., et al. The Relationship of Ketogenic Diet with Neurodegenerative and Psychiatric Diseases: A Scoping Review from Basic Research to Clinical Practice. Nutrients. 2023 May 11;15(10):2270.

Mierau, S.B., et al. Metabolic interventions in Autism Spectrum Disorder. Neurobiol Dis. 2019 Dec:132:104544.

Mu, C., et al. Metabolic Framework for the Improvement of Autism Spectrum Disorders by a Modified Ketogenic Diet: A Pilot Study. J Proteome Res. 2020 Jan 3;19(1):382-390.

Noland, R.C., et al. Carnitine insufficiency caused by aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem. 2009 Aug 21;284(34):22840-52.

Olivito, I., et al. Ketogenic diet ameliorates autism spectrum disorders-like behaviors via reduced inflammatory factors and microbiota remodeling in BTBR T+ Itpr3tf/J mice. Exp Neurol. 2023 Aug:366:114432.

Onodera, Y., et al. Increased sugar uptake promotes oncogenesis via EPAC/RAP1 and O-GlcNAc pathways. J Clin Invest. 2014 Jan;124(1):367-84.

Operta, F.F., et al. The Ketogenic Diet for the Treatment of Mood Disorders in Comorbidity With Epilepsy in Children and Adolescents. Front Pharmacol. 2020 Nov 24:11:578396.

Pacal, L., et al. Parameters of oxidative stress, DNA damage and DNA repair in type 1 and type 2 diabetes mellitus. Arch Physiol Biochem. 2011;117(4):222-30.

Pietrzak, D., et al. The Therapeutic Role of Ketogenic Diet in Neurological Disorders. Nutrients. 2022 May 6;14(9):1952.

Power, R.A., et al. Carnitine revisited: potential use as adjunctive treatment in diabetes. Diabetologia. 2007 Apr;50(4):824-32.

Pulgaron, E.R. Childhood obesity: a review of increased risk for physical and psychological comorbidities. Clin Ther. 2013;35(1):A18-32.

Pulsifer, M.B., et al. Effects of ketogenic diet on development and behavior: preliminary report of a prospective study. Dev Med Child Neurol. 2001 May;43(5):301-6.

Rector, R.S., et al. Mitochondrial dysfunction precedes insulin resistance and hepatic steatosis and contributes to the natural history of non-alcoholic fatty liver disease in an obese rodent model. J Hepatol. 2010 May;52(5):727-36.

Renteria, I., et al. [Factors affecting oxidative damage in obese children: an exploratory study]. Nutr Hosp. 2015;31(4):1499-503.

Rosa, J.S., et al. Altered inflammatory, oxidative, and metabolic responses to exercise in pediatric obesity and type 1 diabetes. Pediatr Diabetes. 2011;12(5):464-72.

Salzer, H.M. Relative hypoglycemia as a cause of neuropsychiatric illness. J Natl Med Assoc. 1966;58(1):12-17.

Sanchez, A., et al. Role of sugars in human neutrophilic phagocytosis. Am J Clin Nutr. 1973 Nov;26(11):1180-4.

Sanchez, M., et al. Childhood obesity: a role for gut microbiota? Int J Environ Res Public Health. 2015;12(1):162-75. J Integr Complement Med. 2023 Feb;29(2):69-79.

Sanago, F., et al. Mind- and Body-Based Interventions Improve Glycemic Control in Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis. J Integr Complement Med. 2023 Feb;29(2):69-79.

Schwalfenberg, G.K. The Importance of Magnesium in Clinical Healthcare. Scientifica (Cairo). 2017:2017:4179326.

Seyfried, T.N. Cancer as a metabolic disease. Nutr Metab (Lond). 2010 Jan 27:7:7.

Seyfried, T.N. Cancer as a metabolic disease: implications for novel therapeutics. Carcinogenesis. 2014 Mar;35(3):515-27.

Seyfried, T.N., et al. Consideration of Ketogenic Metabolic Therapy as a Complementary or Alternative Approach for Managing Breast Cancer. Front Nutr. 2020 Mar 11:7:21.

Seyfried, T.N., et al. Is the restricted ketogenic diet a viable alternative to the standard of care for managing malignant brain cancer? Epilepsy Res. 2012 Jul;100(3):310-26.

Seyfried, T.N., et al. Metabolic management of brain cancer. Biochim Biophys Acta. 2011 Jun;1807(6):577-94.

Shen, W., et al. Protective effects of R-alpha-lipoic acid and acetyl-L-carnitine in MIN6 and isolated rat islet cells chronically exposed to oleic acid. J Cell Biochem. 2008 Jul 1;104(4):1232-43.

Shlomowitz, A., et al. Anxiety associated with self monitoring of capillary blood glucoseBritish Journal of Diabetes. 2014;14(2):60-63.

Smith, J., et al. Ketogenic diet restores aberrant cortical motor maps and excitation-to-inhibition imbalance in the BTBR mouse model of autism spectrum disorder. Behav Brain Res. 2016 May 1:304:67-70.

Smith, K.J., et al. Association of diabetes with anxiety: a systematic review and meta-analysisJ Psychosom Res. 2013;74(2):89-99.

Sreekumar, R., et al. Skeletal muscle mitochondrial dysfunction & diabetes. Indian J Med Res. 2007 Mar;125(3):399-410.

Stafstrom, C.E., et al. The ketogenic diet as a treatment paradigm for diverse neurological disorders. Front Pharmacol. 2012 Apr 9:3:59.

Tao, Y., et al. Ketogenic Diet: An Effective Treatment Approach for Neurodegenerative Diseases. Curr Neuropharmacol. 2022 Nov 15;20(12):2303-2319.

Tidman, M.M., et al. Effects of an low carbohydrate/healthy fat/ketogenic diet on biomarkers of health and symptoms, anxiety and depression in Parkinson's disease: a pilot study. Neurodegener Dis Manag. 2022 Apr;12(2):57-66.

Tinkum, K.L., et al. Fasting protects mice from lethal DNA damage by promoting small intestinal epithelial stem cell survival. Proc Natl Acad Sci U S A. 2015 Dec 22;112(51):E7148-54.

Tuomi, T., et al. The many faces of diabetes: a disease with increasing heterogeneity. Lancet. 2014;383(9922):1084-94.

Vaarala, O. Is the origin of type 1 diabetes in the gut? Immunol Cell Biol. 2012;90(3):271-6.

Vaarala, O. Gut microbiota and type 1 diabetes. Rev Diabet Stud. 2012;9(4):251-9.

Vargas, D.D., et al. Effectiveness of nutritional interventions on behavioral symptomatology of autism spectrum disorder: a systematic review. Nutr Hosp. 2022 Dec 20;39(6):1378-1388.

Verberne, A.J.M., et al. Adrenaline: insights into its metabolic roles in hypoglycaemia and diabetesBr J Pharmacol. 2016;173(9):1425-1437.

Witkowski, M., et al. The artificial sweetener erythritol and cardiovascular event risk. Nat Med. 2023 Mar;29(3):710-718.

Yuan, J., et al. Regular use of proton pump inhibitors and risk of type 2 diabetes: results from three prospective cohort studies. Gut. 28 Sep 2020.

Zheng, P., et al. Gut microbiome in type 1 diabetes: A comprehensive review. Diabetes Metab Res Rev. 2018 Oct;34(7):e3043.

Zhou, H., et al. Evaluating the Causal Role of Gut Microbiota in Type 1 Diabetes and Its Possible Pathogenic Mechanisms. Front Endocrinol (Lausanne). 2020 Mar 24:11:125.

Down Syndrome

Sources & References

Al-Gazali, L.I., et al. Abnormal folate metabolism and genetic polymorphism of the folate pathway in a child with Down syndrome and neural tube defect. Am J Med Genet. 2001 Oct 1;103(2):128-32.

Amorim, M.R., et al. MTHFR 677C–>T and 1298A–>C polymorphisms in children with Down syndrome and acute myeloid leukemia in Brazil. Pediatr Hematol Oncol. 2008 Dec;25(8):744-50.

Bianchi, P., et al. Lithium restores neurogenesis in the subventricular zone of the Ts65Dn mouse, a model for Down syndrome. Brain Pathol. 2010 Jan;20(1):106-18.

Brandalize, A.P.C., et al. Evaluation of C677T and A1298C polymorphisms of the MTHFR gene as maternal risk factors for Down syndrome and congenital heart defects. Am J Med Genet. 2009 Oct;149A(10):2080-7.

Cantor, D.S., et al. A report on phosphatidylcholine therapy in a Down syndrome child. Psychological Reports. 1986, 58, 207-217. 

Chung, S.Y., et al. Administration of phosphatidylcholine increases brain acetylcholine concentration and improves memory in mice with dementia. J Nutr. 1995 Jun;125(6):1484-9.

Contestabile, A., et al. Lithium rescues synaptic plasticity and memory in Down syndrome mice. J Clin Invest. 2013 Jan;123(1):348-61.

Cyril, C., et al. MTHFR Gene variants C677T, A1298C and association with Down syndrome: A Case-control study from South India. Indian J Hum Genet. 2009 May;15(2):60-4.

De la Torre, R., et al. Epigallocatechin-3-gallate, a DYRK1A inhibitor, rescues cognitive deficits in Down syndrome mouse models and in humans. Mol Nutr Food Res. 2014 Feb;58(2):278-88.

DiBaise, J.K. Nutritional consequences of small intestinal bacterial overgrowth. Practical Gastroenterology. 2008. 32(12), 15-28.

Dutta, S., et al. Risk of Down syndrome conferred by MTHFR C677T polymorphism: Ethnic variations. Indian J Hum Genet. 2007 May-Aug; 13(2): 76–77.

Fodale, V., et al. The cholinergic system in Down's syndrome. J Intellect Disabil. 2006 Sep;10(3):261-74.

Hobbs, C.A., et al. Polymorphisms in Genes Involved in Folate Metabolism as Maternal Risk Factors for Down Syndrome. Am J Med Genet. 2000 Sep; 67(3): 623–630.

Hung, M.C., et al. Learning behaviour and cerebral protein kinase C, antioxidant status, lipid composition in senescence-accelerated mouse: influence of a phosphatidylcholine-vitamin B12 diet. Br J Nutr. 2001 Aug;86(2):163-71.

Izzo, A., et al. Mitochondrial dysfunction in down syndrome: molecular mechanisms and therapeutic targets. Mol Med. 2018;24(1):2. Published 2018 Mar 15.

Jovanovic, S.V., et al. Biomarkers of oxidative stress are significantly elevated in Down syndrome. Free Medic Biol Med. 1998 Dec;25(9):1044-8.

Karmiloff-Smith, A., et al. The importance of understanding individual differences in Down syndrome. F1000Res. 2016 Mar 23;5:F1000 Faculty Rev-389.

Kokotas, H., et al. Investigating the impact of the Down syndrome related common MTHFR 677C>T polymorphism in the Danish population. Dis Markers. 2009;27(6):279-85.

Labudova, O., et al. Impaired brain glucose metabolism in patients with Down syndrome. J Neural Transm Suppl. 1999;57:247-56.

Lima, A.S., et al. Nutritional status of zinc in children with Down syndrome. Biol Trace Elem Res. 2010 Jan;133(1):20-8.

Liu, F., et al. Overexpression of Dyrk1A contributes to neurofibrillary degeneration in Down syndrome. FASEB J. 2008 Sep; 22(9): 3224–3233.

Lockrow, J., et al. Cholinergic degeneration and memory loss delayed by vitamin E in a Down syndrome mouse model. Exp Neurol. 2009 Apr;216(2):278-89.

Martínez-Frías, M.L., et al. Maternal polymorphisms 677C-T and 1298A-C of MTHFR, and 66A-G MTRR genes: is there any relationship between polymorphisms of the folate pathway, maternal homocysteine levels, and the risk for having a child with Down syndrome? Am J Med Genet. 2006 May 1;140(9):987-97.

Meguid, N.A., et al. MTHFR genetic polymorphism as a risk factor in Egyptian mothers with Down syndrome children. Dis Markers. 2008;24(1):19-26.

Moon, J., et al. Perinatal choline supplementation improves cognitive functioning and emotion regulation in the Ts65Dn mouse model of Down syndrome. Behav Neurosci. 2010 Jun;124(3):346-61.

Nachvak, S.M. α-Tocopherol supplementation reduces biomarkers of oxidative stress in children with Down syndrome: a randomized controlled trial. Eur J Clin Nutr. 2014 Oct;68(10):1119-23.

Napolitano, G., et al. Is zinc deficiency a cause of subclinical hypothyroidism in Down syndrome? Ann Genet. 1990;33(1):9-15.

Oka, A., et al. The up-regulation of metabotropic glutamate receptor 5 (mGluR5) in Down’s syndrome brains. Acta Neuropathol. 1999 Mar;97(3):275-8.

O’Leary, V.B., et al. MTRR and MTHFR polymorphism: link to Down syndrome? Am J Med Genet. 2002 Jan 15;107(2):151-5.

Parisotto, E.B., et al. Antioxidant intervention attenuates oxidative stress in children and teenagers with Down syndrome. Res Dev Disabil. 2014 Jun;35(6):1228-36.

Pietrini, P., et al. Low glucose metabolism during brain stimulation in older Down's syndrome subjects at risk for
Alzheimer's disease prior to dementia. Am J Psychiatry. 1997 Aug;154(8):1063-9.

Rabinowitz, S.S., et al. Pediatric Beriberi Clinical Presentation. Medscape. 2014 Mar 17.

Reutter, H., et al. MTHFR 677 TT genotype in a mother and her child with Down syndrome, atrioventricular canal and exstrophy of the bladder: implications of a mutual genetic risk factor? Eur J Pediatr. 2006 Aug;165(8):566-8.

Starbuck, J.M., et al. Green tea extracts containing epigallocatechin-3-gallate modulate facial development in Down syndrome. Sci Rep. 2021 Feb 25;11(1):4715.

Stringer, M., et al. Epigallocatechin-3-gallate (EGCG) consumption in the Ts65Dn model of Down syndrome fails to improve behavioral deficits and is detrimental to skeletal phenotypes. Physiol Behav. 2017 Aug 1;177:230-241.

Takeda, A., et al. Release of glutamate and GABA in the hippocampus under zinc deficiency. J Neurosci Res. 2003 May 15;72(4):537-42.

Tanzi, R.E., et al. Neuropathology in the Down’s syndrome brain. Nature Medicine. 1996; (2)3132.

Vacca, R.S., et al. Green tea EGCG plus fish oil omega-3 dietary supplements rescue mitochondrial dysfunctions and are safe in a Down’s syndrome child. Clinical Nutrition. 2015 1-2.

Valenti, D., et al. Epigallocatechin-3-gallate prevents oxidative phosphorylation deficit and promotes mitochondrial biogenesis in human cells from subjects with Down’s syndrome. Biochim Biophys Acta. 2013 Apr;1832(4):542-52.

Wang, S.S., et al. Polymorphisms in genes involved in folate metabolism as maternal risk factors for Down syndrome in China*. J Zhejiang Univ Sci B. 2008 Feb; 9(2): 93–99.

Dyslexia

Sources & References

Adams, L.J., et al. Infant feeding method and special educational need in 191,745 Scottish schoolchildren: A national, population cohort study. PLoS Med. 2023 Apr 6;20(4):e1004191.

Baker, S.M. A biochemical approach to the problem of dyslexia. Journal of Learning Disabilities. 1985 18(10): 581-584.

Boat, T.F., et al. Prevalence of Learning Disabilities. Mental Disorders and Disabilities Among Low-Income Children. Washington (DC): National Academies Press (US); 2015 Oct 28. 16.

Borre, Y.E., et al. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med. 2014 Sep;20(9):509-18.

Darling, A.L., et al. Association between maternal vitamin D status in pregnancy and neurodevelopmental outcomes in childhood: results from the Avon Longitudinal Study of Parents and Children (ALSPAC). Br J Nutr 2017 Jun;117(12):1682-1692.

Edwards, E.S., et al. Dyslexia on a continuum: A complex network approach. PLOS ONE, 2018; 13 (12): e0208923.

Egset, K., et al. Magno App: Exploring Visual Processing in Adults with High and Low Reading Competence. Scandinavian Journal of Educational Research. 07 Jan 2020.

Hertz-Picciotto, I., et al. Organophosphate exposures during pregnancy and child neurodevelopment: Recommendations for essential policy reforms. PLoS Med. 2018 Oct 24;15(10):e1002671.

Mercer, J.S., et al. The Effects of Delayed Cord Clamping on 12-Month Brain Myelin Content and Neurodevelopment: A Randomized Controlled Trial. Am J Perinatol. 2022 Jan;39(1):37-44.

Morris, C.R., et al. Syndrome of allergy, apraxia, and malabsorption: characterization of a neurodevelopmental phenotype that responds to omega 3 and vitamin E supplementation. Alternative Therapies in Health and Medicine. Jul-Aug 2009;15(4):34-43.

Rifas-Shiman, S.L., et al. Associations of prenatal or infant exposure to acetaminophen or ibuprofen with mid-childhood executive function and behaviour. Paediatr Perinat Epidemiol. 2020 May;34(3):287-298.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Zablotsky, B., et al. Prevalence and Trends of Developmental Disabilities among Children in the United States: 2009-2017. Pediatrics. 2019 Oct;144(4):e20190811.

Zaigham, M., et al. Prelabour caesarean section and neurodevelopmental outcome at 4 and 12 months of age: an observational study. BMC Pregnancy and Childbirth. 2020 (20)564.

Dyspraxia and Apraxia

Sources & References

Bartscherer, et al. Interactive metronome training for a 9-year-old boy with attention and motor coordination difficulties. Physiother Theory Pract. Oct-Dec 2005;21(4):257-69.

Blondis, T.A. Motor disorders and attention-deficit/hyperactivity disorder. Pediatr Clin North Am. 1999 Oct;46(5):899-913, vi-vii.

Borre, Y.E., et al. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med. 2014 Sep;20(9):509-18.

Cosper, S.M., et al. Interactive Metronome training in children with attention deficit and developmental coordination disorders. Int J Rehabil Res. 2009 Dec;32(4):331-6.

Goulardins, J.B., et al. Attention deficit hyperactivity disorder and developmental coordination disorder: Two separate disorders or do they share a common etiology. Behav Brain Res. 2015 Oct 1;292:484-92.

Goulardins, J.B., et al. Attention Deficit Hyperactivity Disorder and Motor Impairment. Percept Mot Skills. 2017 Apr;124(2):425-440.

Karatekin, C., et al. A preliminary study of motor problems in children with attention-deficit/hyperactivity disorder. Percept Mot Skills. 2003 Dec;97(3 Pt 2):1267-80.

McLeod, K.R., et al. Functional connectivity of neural motor networks is disrupted in children with developmental coordination disorder and attention-deficit/hyperactivity disorder. Neuroimage Clin. 2014 Mar 26;4:566-75.

Morris, C.R., et al. Syndrome of Allergy, Apraxia, and Malabsorption: Characterization of the Neurodevelopmental Phenotype that Responds to Omega 3 and Vitamin E Supplement. Altern Ther Health Med. 2009 Jul-Aug;15(4):34-43.

Piek, J.P., et al. Motor coordination and kinaesthesis in boys with attention deficit-hyperactivity disorder. Dev Med Child Neurol. 1999 Mar;41(3):159-65.

Richardson, A.J. Dyslexia, Dyspraxia and ADHD – Can Nutrition Help?

Stordy, B.J. Dark adaptation, motor skills, docosahexaenoic acid, and dyslexia. Am J Clin Nutr. 2000 Jan;71(1 Suppl):323S-6S.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Zaigham, M., et al. Prelabour caesarean section and neurodevelopmental outcome at 4 and 12 months of age: an observational study. BMC Pregnancy and Childbirth. 2020 (20)564.

Ear, Nose & Throat Infections

Sources & References

Adams, J.D., et al. Otitis Media and Related Complications Among Children with Autism Spectrum Disorders. J Autism Dev Disord. 2016 May;46(5):1636-42.

Bell, I.R., et al. Homeopathic medications as clinical alternatives for symptomatic care of acute otitis media and upper respiratory infections in children. Global Adv Health Med. 2013 Jan;2(1):32-43.

Frieri, M. Asthma linked with rhinosinusitis: An extensive review. Allergy Rhinol (Providence). 2014;5(1):41-9.

Jacobs, J., et al. Homeopathic treatment of acute otitis media in children: a preliminary randomized placebo-controlled trial. Pediatric Infect Dis J. 2001 Feb;20(2):177-83.

Juntti, H., et al. Cow’s milk allergy is associated with recurrent otitis media during childhood. Acta Otolaryngol. 1999;119:867–873.

Konstantareas, M.M., et al. Ear infections in autistic and normal children. Journal of Autism and Developmental Disorders. 1987 Dec;17(4):585-94.

Nsouli, T.M., et al. The role of food allergy in serious otitis media. Ann Allergy. 1994 Sep; 66:91.

Ramakrishnan, J.B.. The role of food allergy in otolaryngology disorders. Curr Opin Otolaryngol Head Neck Surg. 2010;18:195–199.

Taylor, J.A., et al. Homeopathic Ear Drops as an Adjunct in Reducing Antibiotic Usage in Children With Acute Otitis Media. Global Pediatr Health. 2014 Nov 21;1:2333794X14559395.

Zemotti, E.M., et al. Otitis media with effusion and atopy: is there a causal relationship? World Allergy Organ J. 2017; 10(1): 37.

Eat a Clean Diet

Sources & References

Bateman, B., et al. The effects of a double blind, placebo controlled, artificial food colourings and benzoate preservative challenge on hyperactivity in a general population sample of preschool children. Arch Dis Child. 2004 Jun;89(6):506-11.

Bauer, J., et al. Hyperactivity and impulsivity in adult attention-deficit/hyperactivity disorder is related to glutamatergic dysfunction in the anterior cingulate cortex. World J Biol Psychiatry. 2018 Oct;19(7):538-546.

Boris, M., et al. Foods and additives are common causes of the attention deficit hyperactive disorder in children. Ann Allergy. 1994 May;72(5):462-8.

Chhabra, R., et al. Organically grown food provides health benefits to Drosophila melanogaster. PLoS One. 2013;8(1):e52988.

Crinnion, W.J. Organic foods contain higher levels of certain nutrients, lower levels of pesticides, and may provide health benefits for the consumer. Altern Med Rev. 2010 Apr;15(1):4-12.

Davalli, D.M., et al. The potential role of glutamate in the current diabetes epidemic. Acta Diabetol. 2012 Jun;49(3):167-83.

DeHavenon, A., et al. The Secret “Spice”: An Undetectable Toxic Cause of Seizure. Neurohospitalist. 2011 Oct; 1(4): 182–186.

Gunatilake, S., et al. Glyphosate’s Synergistic Toxicity in Combination with Other Factors as a Cause of Chronic Kidney Disease of Unknown Origin. Int J Environ Res Public Health. 2019 Jul 31;16(15):2734.

Haroon, E., et al. Inflammation, Glutamate, and Glia: A Trio of Trouble in Mood Disorders. Neuropsychopharmacology. 2017 Jan;42(1):193-215.

Hertz-Picciotto, I., et al. Organophosphate exposures during pregnancy and child neurodevelopment: Recommendations for essential policy reforms. PLoS Med. 2018 Oct 24;15(10):e1002671.

Kanarek, R.B. Artificial food dyes and attention deficit hyperactivity disorder. Nutr Rev. 2011 Jul;69(7):385-91.

Khoshnoud, M.J., et al. Effects of sodium benzoate, a commonly used food preservative, on learning, memory, and oxidative stress in brain of mice. J Biochem Mol Toxicol. 2018 Feb;32(2).

McCann, D., et al. Food additives and hyperactive behaviour in 3-year-old and 8/9-year-old children in the community: a randomised, double-blinded, placebo-controlled trial. Lancet. 2007 Nov 3;370(9598):1560-7.

McNally, L., et al. Inflammation, glutamate, and glia in depression: a literature review. CNS Spectr. 2008 Jun;13(6):501-10.

Millichap, J.G., et al. The diet factor in attention-deficit/hyperactivity disorder. Pediatrics.2012 Feb;129(2):330-7.

Nigg, J.T., et al. Meta-analysis of attention-deficit/hyperactivity disorder or attention-deficit/hyperactivity disorder symptoms, restriction diet, and synthetic food color additives. J Am Acad Child Adolesc Psychiatry. 2012 Jan;51(1):86-97.e8.

Pitt, D., et al. Glutamate excitotoxicity in a model of multiple sclerosis. Nat Med. 2000 Jan;6(1):67-70.

Robinette, L.M., et al. Fruit and vegetable intake is inversely associated with severity of inattention in a pediatric population with ADHD symptoms: the MADDY Study. Nutr Neurosci. 2023 Jun;26(6):572-581.

Samsel, A., et al. Glyphosate, pathways to modern diseases II: Celiac sprue and gluten intolerance. Interdiscip Toxicol. 2013 Dec;6(4):159-84.

Samsel, A., et al. Glyphosate, pathways to modern diseases III: Manganese, neurological diseases, and associated pathologiesSurg Neurol Int. 2015 Mar 24;6:45.

Samsel, A, et al. Glyphosate’s suppression of cytochrome P450 enzymes and amino acid biosynthesis by the gut microbiome: Pathways to modern diseases. Entropy. 2013;15:1416–1463.

Samuels, A. The toxicity/safety of processed free glutamic acid (MSG): a study in suppression of information. Account Res. 1999;6(4):259-310.

Schab, D.W., et al. Do artificial food colors promote hyperactivity in children with hyperactive syndromes? A meta-analysis of double-blind placebo-controlled trials. J Dev Behav Pediatr. 2004 Dec;25(6):423-34.

Silfverdal, S.A., et al. [Food additives can increase hyperactivity in children. Results from a British study confirm the connection]. Lakartidningen. 2008 Feb 6-12;105(6):354-5.

Tobacman, J.K. Review of Harmful Gastrointestinal Effects of Carrageenan in Animal Experiments. Environmental Health Perspectives. 2001 Oct;109(10):983-94.

Vigar, V., et al. A Systematic Review of Organic Versus Conventional Food Consumption: Is There a Measurable Benefit on Human Health? Nutrients. 2019 Dec 18;12(1):7.

Wallinga, D., et al. Not So Sweet: Missing Mercury and High Fructose Corn Syrup. Institute for Agriculture and Trade Policy, Minneapolis, Minnesota. Jan 2009.

Watkins, J.C., et al. The glutamate story. Br J Pharmacol. 2006 Jan; 147(Suppl 1): S100–S108.

Yu, C.J., et al. Sugar-Sweetened Beverage Consumption Is Adversely Associated with Childhood Attention Deficit/Hyperactivity Disorder. Int J Environ Res Public Health. 2016 Jul 4;13(7).

Zhang, Z., et al. Blood Glutamate Levels in Autism Spectrum Disorder: A Systematic Review and Meta-Analysis. PLoS One. 2016 Jul 8;11(7):e0158688.

Eczema and Other Skin Problems

Sources & References

da Costa Baptista, I.P., et al. Effect of the use of probiotics in the treatment of children with atopic dermatitis; a literature review. Nutr Hosp. 2013;28(1):16-26.

Drago, L., et al. Changing of fecal flora and clinical effect of L. salivarius LS01 in adults with atopic dermatitis. J Clin Gastroenterol. 2012;46 Suppl:S56-63.

Eichenfield, L.F., et al. Guidelines of care for the management of atopic dermatitis: section 1. Diagnosis and assessment of atopic dermatitis. J Am Acad Dermatol. 2014;70(2):338-51.

Gamez, C., et al. Lower Cord Blood IL-17 and IL-25, but Not Other Epithelial Cell-Derived Cytokines Are Associated with Atopic Dermatitis in Infancy. Int Arch Allergy Immunol. 2021;182(6):474-478.

Liao, T.C., et al. Comorbidity of Atopic Disorders with Autism Spectrum Disorder and Attention Deficit/Hyperactivity Disorder. J Pediatr. 2016 Apr;171:248-55.

Oh, S.Y., et al. Antioxidant nutrient intakes and corresponding biomarkers associated with the risk of atopic dermatitis in young children. Eur J Clin Nutr. 2010;64(3):245-52.

Orivuori, L., et al. High level of fecal calprotectin at age 2 months as a marker of intestinal inflammation predicts atopic dermatitis and asthma by age 6. Clin Exp Allergy. 2015.

Schmitt, J., et al. Atopic eczema and attention-deficit/hyperactivity disorder in a population-based sample of children and adolescents. JAMA. 2009 Feb 18;301(7):724-6.

Stinson, L.F., et al. Human Milk From Atopic Mothers Has Lower Levels of Short Chain Fatty Acids. Front Immunol. 2020 Jul 21:11:1427.

ElectroMagnetic Frequencies (EMFs)

Sources & References

Abramson, M.J., et al. Mobile telephone use is associated with changes in cognitive function in young adolescents. Bioelectromagnetics. 2009 Dec;30(8):678-86.

Aldad, T.S., et al. Fetal Radiofrequency Radiation Exposure From 800-1900 Mhz-Rated Cellular Telephones Affects Neurodevelopment and Behavior in Mice. Sci Rep. 2012;2:312.

Breitenkamp, A.F., et al. Voltage-gated Calcium Channels and Autism Spectrum Disorders. Curr Mol Pharmacol. 2015;8(2):123-32.

Büsselberg, D. Calcium channels as target sites of heavy metals. Toxicol Lett. 1995 Dec:82-83:255-61.

Carlo, G.L., et al. Wireless radiation in the aetiology and treatment of autism: clinical observations and mechanisms. Journal of the Australasian College of Nutritional and Environmental Medicine, 26(2), 3–7.

Currenti, S.A. Understanding and determining the etiology of autism. Cell Mol Neurobiol. 2010 Mar;30(2):161-71.

Dasdaq, S., et al. Effects of 2.4 GHz radiofrequency radiation emitted from Wi-Fi equipment on microRNA expression in brain tissue. Int J Radiat Biol. 2015 Jul;91(7):555-61.

Davis, D., et al. Wireless technologies, non-ionizing electromagnetic fields and children: Identifying and reducing health risk. Curr Probl Pediatr Adolesc Health Care. 2023 Feb;53(2):101374.

Faber, S., et al. A cleanroom sleeping environment’s impact on markers of oxidative stress, immune dysregulation, and behavior in children with autism spectrum disorders. BMC Complement Altern Med. 2015 Mar 19:15:71.

Havas, M. Dirty electricity elevates blood sugar among electrically sensitive diabetics and may explain brittle diabetes. Electromagn Biol Med. 2008;27(2):135-46.

Havas, M. Radiation from wireless technology affects the blood, the heart, and the autonomic nervous system. Rev Environ Health. 2013;28(2-3):75-84.

Herbert, M.R, et al. Autism and EMF? Plausibility of a pathophysiological link–Part I. Pathophysiology 20.3 (2013): 191-209.

Herbert, M.R., et al. Autism and EMF? Plausibility of a pathophysiological link Part II. Pathophysiology 20.3 (2013): 211-234.

Hosseinabadi, M.B., et al. The effect of chronic exposure to extremely low-frequency electromagnetic fields on sleep quality, stress, depression and anxiety. Electromagn Biol Med. 2019;38(1):96-101.

Johansson, O. Disturbance of the immune system by electromagnetic fields—A potentially underlying cause for cellular damage and tissue repair reduction which could lead to disease and impairment. Pathophysiology. 2009 Aug;16(2-3):157-177.

Kane, R.C. A possible association between fetal/neonatal exposure to radiofrequency electromagnetic radiation and the increased incidence of autism spectrum disorders (ASD). Med Hypotheses. 2004;62(2):195-7.

Koyu, A., et al. Effects of 900 MHz electromagnetic field on TSH and thyroid hormones in rats. Toxicol Lett. 2005 Jul 4;157(3):257-62.

Kundi, M., et al. Mobile phone specific radiation disturbs cytokinesis and causes cell death but not acute chromosomal damage in buccal cells: Results of a controlled human intervention study. Environ Res. 2024 Jun 15;251(Pt 1):118634.

Lathe, R. Microwave Electromagnetic Radiation and Autism. E-Journal of Applied Psychology. June 2009;5(1):11-30.

Li, D.K., et al. A population-based prospective cohort study of personal exposure to magnetic fields during pregnancy and the risk of miscarriage. Epidemiology. 2002 Jan;13(1):9-20.

Milham, S. Historical evidence that electrification caused the 20th century epidemic of "diseases of civilization". Med Hypotheses. 2010 Feb;74(2):337-45.

Pall, M.L., Electromagnetic fields act via activation of voltage‐gated calcium channels to produce beneficial or adverse effects. Journal of Cellular and Molecular Medicine 17.8 (2013): 958-965.

Pall, M.L. Microwave frequency electromagnetic fields (EMFs) produce widespread neuropsychiatric effects including depression. J Chem Neuroanat. 2016 Sep;75(Pt B):43-51.

Pall, M.L. The Autism Epidemic Is Caused by EMFs, Acting via Calcium Channels and Chemicals Acting via NMDA-Rs: Downstream Effects Cause Autism, Autism One, Chicago, Illinois, USA, 2015.

Pall, M.L. Wi-Fi is an important threat to human health. Environ Res. 2018 Jul;164:405-416.

Panagopolous, D.J., et al. Cell death induced by GSM 900-MHz and DCS 1800-MHz mobile telephony radiation. Mutat Res. 2007 Jan 10;626(1-2):69-78.

Pino-López, M., et al. [Parental occupational exposures and autism spectrum disorder in children]. Rev Esp Salud Publica. 2013 Jan-Feb;87(1):73-85.

Reiter, R.J. Melatonin suppression by static and extremely low frequency electromagnetic fields: relationship to the reported increased incidence of cancer. Rev Environ Health. 1994 Jul-Dec;10(3-4):171-86.

Tang, J., et al. Exposure to 900 MHz electromagnetic fields activates the mkp-1/ERK pathway and causes blood-brain barrier damage and cognitive impairment in rats. Brain Res. 2015 Mar 19;1601:92-101.

Thornton, I.M. Out of time: a possible link between mirror neurons, autism and electromagnetic radiation. Med Hypotheses. 2006;67(2):378-82.

Wu, J., et al. Structure of the voltage-gated calcium channel Cav1.1 complex. Science. 2015 Dec 18;350(6267):aad2395.

Emotional/Behavioral/Mood Symptoms

Sources & References

Andreazza, A.C., et al. Mitochondrial complex I activity and oxidative damage to mitochondrial proteins in the prefrontal cortex of patients with bipolar disorder. Arch Gen Psychiatry. 2010 Apr;67(4):360-8.

Appleton, J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr Med (Encinitas). 2018 Aug;17(4):28-32.

Atladottir, H.O., et al. The increasing prevalence of reported diagnoses of childhood psychiatric disorders: a descriptive multinational comparison. Eur Child Adolesc Psychiatry. 2015;24(2):173-83.

Aucoin, M., et al. Major Depressive Disorder and Food Hypersensitivity: A Case Report. Neuropsychobiology. 2019 Oct 10:1-7.

Baughman, N., et al. The Prevention of Anxiety and Depression in Early Childhood. Front Psychol. 2020 Sep 30:11:517896.

Bayer, J.K., et al. The Cool Little Kids randomised controlled trial: population-level early prevention for anxiety disorders. BMC Public Health. 2011;11:11.

Berk, M., et al. So depression is an inflammatory disease, but where does the inflammation come from? BMC Med. 2013;11:200.

Berry, E.A., et al. National estimates of the inpatient burden of pediatric bipolar disorder in the United States. J Ment Health Policy Econ. 2011;14(3):115-23.

Bitsko, R.H., et al. Epidemiology and Impact of Health Care Provider-Diagnosed Anxiety and Depression Among US Children. J Dev Behav Pediatr 2018 Apr 24.

Bonaccio, M., et al. Mediterranean-type diet is associated with higher psychological resilience in a general adult population: findings from the Moli-sani study. Eur J Clin Nutr. 2018 Jan;72(1):154-160.

Bonnot, O., et al. Children and adolescents with severe mental illness need vitamin D supplementation regardless of disease or treatment. J Child Adolesc Psychopharmacol. 2011;21(2):157-61.

Burrows, T., et al. Effectiveness of dietary interventions in mental health treatment: A rapid review of reviews. Nutr Diet. 2022 Jul;79(3):279-290.

Camilleri, M. Serotonin in the gastrointestinal tract. Curr Opin Endrocrinol Diabetes Obes. 2009 Feb;16(1):53-9.

Ceylan, M.F., et al. Lipid peroxidation markers in children with anxiety disorders and their diagnostic implications. Redox Rep. 2014;19(2):92-6.

Chen, Y., et al. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients. 2021 Jun 19;13(6):2099.

Cohen-Cline, H., et al. Access to green space, physical activity and mental health: a twin study. J Epidemiol Community Health. 2015 Jun;69(6):523-9.

Costello, E.J., et al. 10-year research update review: the epidemiology of child and adolescent psychiatric disorders: II. Developmental epidemiology. J Am Acad Child Adolesc Psychiatry. 2006 Jan;45(1):8–25.

Dusetzina, S.B., et al. Treatment use and costs among privately insured youths with diagnoses of bipolar disorder. Psychiatr Serv. 2012;63(10):1019-25.

Guney, E., et al. Oxidative stress in children and adolescents with anxiety disorders. J Affect Disord. 2014;156:62-6.

Fernandes, A.C., et al. Development and evaluation of a de-identification procedure for a case register sourced from mental health electronic records. BMC Med Inform Decis Mak. 2013;13:71.

Haroon, E., et al. Inflammation, Glutamate, and Glia: A Trio of Trouble in Mood Disorders. Neuropsychopharmacology. 2017 Jan;42(1):193-215.

Hepgul, N., et al. Depression pathogenesis and treatment: what can we learn from blood mRNA expression? BMC Med. 2013;11:28.

Heuer, L., et al. Reduced levels of immunoglobulin in children with autism correlates with behavioral symptoms. Autism Res, Oct 2008, 1:5, 275–83.

Hyman, M.A. Is the Cure for Brain Disorders Outside the Brain? Alternative Therapies in Health and Medicine. Nov-Dec 2007;13(6):10-5.

Karakula, H., et al. [Does diet affect our mood? The significance of folic acid and homocysteine]. Pol Merkur Lekarski. 2009;26(152):136-41.

Kato, T. The role of mitochondrial dysfunction in bipolar disorder. Drug News Perspect. 2006 Dec;19(10):597-602.

Konradi, C., et al. Molecular evidence for mitochondrial dysfunction in bipolar disorder. Arch Gen Psychiatry. 2004 Mar;61(3):300-8.

Lai, C.C.W., et al. The association between gut-health promoting diet and depression: A mediation analysis. J Affect Disord. 2023 Mar 1:324:136-142.

Lavebratt, C., et al. Early exposure to antibiotic drugs and risk for psychiatric disorders: a population-based study. Transl Psychiatry. 2019 Nov 26;9(1):317.

Leipold, B., et al. Physical activity and nutrition in relation to resilience: a cross-sectional study. Sci Rep. 2024 Jan 27;14(1):2272.

Leuchter, A.F., et al. Intermediate phenotypes and biomarkers of treatment outcome in major depressive disorder. Dialogues Clin Neurosci. 2014;16(4):525-37.

Manigault, A.W., et al. Psychosocial Resilience to Inflammation-Associated Depression: A Prospective Study of Breast-Cancer Survivors. Psychol Sci. 2022 Aug;33(8):1328-1339.

Marazziti, D., et al. Psychiatric disorders and mitochondrial dysfunctions. Eur Rev Med Pharmacol Sci. 2012;16(2):270-5.

McNally, L., et al. Inflammation, glutamate, and glia in depression: a literature review. CNS Spectr. 2008 Jun;13(6):501-10.

Memon, A., et al. Association between naturally occurring lithium in drinking water and suicide rates: systematic review and meta-analysis of ecological studies. British Journal of Psychiatry. 2020 Dec; 217(6): 667-678.

Mitchell, E.S., et al. B vitamin polymorphisms and behavior: evidence of associations with neurodevelopment, depression, schizophrenia,bipolar disorder and cognitive decline. Neurosci Biobehav Rev. 2014;47:307-20.

Netz, K. Is the Comparison between Exercise and Pharmacologic Treatment of Depression in the Clinical Practice Guideline of the American College of Physicians Evidence-Based? Front Pharmacol. 2017 May 15:8:257.

Nikolova, V.L., et al. Acceptability, Tolerability, and Estimates of Putative Treatment Effects of Probiotics as Adjunctive Treatment in Patients With Depression. JAMA Psychiatry. 2023 Aug 1;80(8):842-847.

Onaolapo, A.Y., et al. Glutamate and depression: Reflecting a deepening knowledge of the gut and brain effects of a ubiquitous molecule. World J Psychiatry. 2021 Jul 19;11(7):297-315.

Ostiguy, C.S., et al. Sensitivity to stress among the offspring of parents with bipolar disorder: a study of daytime cortisol levels. Psychol Med. 2011;41(11):2447-57.

Rifas-Shiman, S.L., et al. Associations of prenatal or infant exposure to acetaminophen or ibuprofen with mid-childhood executive function and behaviour. Paediatr Perinat Epidemiol. 2020 May;34(3):287-298.

Rollins, B., et al. Mitochondrial variants in schizophrenia, bipolar disorder, and major depressive disorder. PLoS One. 2009;4(3):e4913.

Rook, G.A., et al. Microbiota, immunoregulatory old friends and psychiatric disorders. Adv Exp Med Biol. 2014;817:319-56.

Schwalfenberg, G.K. The Importance of Magnesium in Clinical Healthcare. Scientifica (Cairo). 2017:2017:4179326.

Simpson, C.A., et al. The gut microbiota in anxiety and depression - A systematic review. Clin Psychol Rev. 2021 Feb:83:101943.

Strandwitz, P. Neurotransmitter Modulation by the Gut Microbiota. Brain Res. 2018 Aug 15;1693(Pt B):128-133.

Swann., O.G., et al. Dietary fiber and its associations with depression and inflammation. Nutr Rev. 2020 May 1;78(5):394-411.

Tan, Y., et al. Review of research progress on intestinal microbiota based on metabolism and inflammation for depression. Arch Microbiol. 2024 Mar 10;206(4):146.

Taurines, R., et al. Expression analyses of the mitochondrial complex I 75-kDa subunit in early onset schizophrenia and autism spectrum disorder: increased levels as a potential biomarker for early onset schizophrenia. Eur Child Adolesc Psychiatry. 2010 May;19(5):441-8.

Thompson, L., et al. What have birth cohort studies asked about genetic, pre- and perinatal exposures and child and adolescent onset mental health outcomes? A systematic review. Eur Child Adolesc Psychiatry. 2010;19(1):1-15.

Van Meter, A.R., et al. What goes up must come down: the burden of bipolar depression in youth. J Affect Disord. 2013;150(3):1048-54.

Verena, L., et al. Elimination diets’ efficacy and mechanisms in attention deficit hyperactivity disorder and autism spectrum disorder. Eur Child Adolesc Psychiatry. 2017; 26(9): 1067–1079.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Whatnall, M.C., et al. Are Psychological Distress and Resilience Associated with Dietary Intake Among Australian University Students? Int J Environ Res Public Health. 2019 Oct 24;16(21):4099.

Yang, B., et al. Effects of regulating intestinal micobiota on anxiety symptoms: A systematic review. General Psychiatry. 2019; 32: e100056.

Yin, Z., et al. Dietary Diversity Was Positively Associated with Psychological Resilience among Elders: A Population-Based Study. Nutrients. 2019 Mar 18;11(3):650.

Environmental Toxicity

Sources & References

Adams, J.B., et al. Mercury in first-cut baby hair of children with autism versus typically-developing children. Toxicological & Environmental Chemistry. 2007 Jun;70(12):1046-51.

Adams, J.B., et al. Mercury, Lead, and Zinc in Baby Teeth of Children with Autism Versus Controls. Journal of Toxicology and Environmental Health. 2007 Jun;70(12):1046-51.

Alampi, J.D., et al. Gestational Exposure to Toxicants and Autistic Behaviors using Bayesian Quantile Regression. Am J Epedemiol. 2021 Sep 1;190(9):1803-1813.

Aldad, T.S., et al. Fetal Radiofrequency Radiation Exposure From 800-1900 Mhz-Rated Cellular Telephones Affects Neurodevelopment and Behavior in Mice. Sci Rep. 2012;2:312.

Banks, W.A., et al. Aluminum-induced neurotoxicity: alterations in membrane function at the blood-brain barrier. Neurosci Biobehav Rev. 1989 Spring;13(1):47-53.

Bernard, S., et al. Autism: a novel form of mercury poisoning. Med Hypotheses. 2001 Apr;56(4):462-71.

Bethell, C., et al. A National and State Profile of Leading Health Problems and Health Care Quality for US Children: Key Insurance Disparities and Across-State Variations. Academic Pediatrics, May–June 2011, Volume 11, Issue 3, Supplement, p. S22–S33.

Björvang, R.D., et al. Mixtures of persistent organic pollutants are found in vital organs of late gestation human fetuses. Chemosphere. 2021 Nov;283:131125.

Braun, J.M., et al. Association of Environmental Toxicants and Conduct Disorder in U.S. Children: NHANES 2001-2004. Environ Health Perspect. 2008 Jul;116(7):956-62.

Braun, J.M., et al. Exposures to environmental toxicants and attention deficit hyperactivity disorder in U.S. children. Environ Health Project. Dec 2006;114(12):1904-1909.

Choi, A.L., et al. Developmental Fluoride Neurotoxicity: A Systematic Review and Meta-Analysis. Environ Health Perspect. 2012;120(10).

Cohen, Patricia. Roundup Maker to Pay $10 Billion to Settle Cancer Suits. The New York Times. 24 Jun 2020.

Deisher, T.A., et al. Impact of environmental factors on the prevalence of autistic disorder after 1979. J Public Health and Epidemiology. Sep 2014;6(9):271-286.

Deth, R., et al. How environmental and genetic factors combine to cause autism: A redox/methylation hypothesis. Neurotoxicology. 2008;29(1):190-201.

Environmental Working Group. Body Burden: The Pollution in Newborns. 14 Jul 2005.

Geier, M.R., et al. The potential importance of steroids in the treatment of autistic spectrum disorders and other disorders involving mercury toxicity. Med Hypotheses. 2005;64(5):946-54.

Grandjean, P., et al. Developmental neurotoxicity of industrial chemicals. Lancet. 2006 Dec 16;368(9553):2167-78.

Gunatilake, S., et al. Glyphosate’s Synergistic Toxicity in Combination with Other Factors as a Cause of Chronic Kidney Disease of Unknown Origin. Int J Environ Res Public Health. 2019 Jul 31;16(15):2734.

Hansen, J.B., et al. Prenatal exposure to bisphenol A and autistic- and ADHD-related symptoms in children aged 2 and5 years from the Odense Child Cohort. Environ Health. 2021 Mar 12;20(1):24.

Hanson, D.R., et al. Theories of schizophrenia: a genetic-inflammatory-vascular synthesis. BMC Medical Genetics. 2005 Feb 11;6:7.

Harley, K.G., et al. Changes in Latina Women’s Exposure to Cleaning Chemicals Associated with Switching from Conventional to “Green” Household Cleaning Products: The LUCIR Intervention Study. Environ Health Perspect. 2021 Sep;129(9):97001.

Hauser, P., et al. Resistance to thyroid hormone: implications for neurodevelopmental research on the effects of thyroid hormone disruptors. Toxicol Ind Health. 1998 Jan-Apr;14(1-2):85-101.

Herbert, M.R., et al. Autism and environmental genomics. Neurotoxicology. 2006;27(5):671-84.

Herbert, M.R., et al. Autism and EMF? Plausibility of a pathophysiological link part I. Pathophysiology. 2013 Jul;1-19.

Herbert, M.R., et al. Autism and EMF? Plausibility of a pathophysiological link part II. Pathophysiology. 2013 Jun;20(3):211-34.

Hertz-Picciotto, I., et al. Organophosphate exposures during pregnancy and child neurodevelopment: Recommendations for essential policy reforms. PLoS Med. 2018 Oct 24;15(10):e1002671.

Hertz-Picciotto, I., et al. Polybrominated diphenyl ethers in relation to autism and developmental delay: a case-control study. Environ Health. 2011 Jan 5;10(1):1.

Hertz-Picciotto, I., et al. Prenatal exposures to persistent and non-persistent organic compounds and effects on immune system development. Basic Clin Pharmacol Toxicol. 2008 Feb;102(2):146-54.

Hinhumpatch, P., et al. Oxidative DNA damage and repair in children exposed to low levels of arsenic in utero and during early childhood: application of salivary and urinary biomarkers. Toxicol Appl Pharmacol. 2013;273(3):569-79.

Holmes, A., et al. Reduced Levels of Mercury in First Baby Haircuts of Autistic Children. International Journal of Toxicology. Jul-Aug 2003;22(4):277-85.

Jafari, M.H., et al. The Relationship Between the Level of Copper, Lead, Mercury and Autism Disorders: A Meta-Analysis. Pediatric Health, Medicine and Therapeutics. 21 Sep 2020(11):369—378.

Jafari, T., et al. The association between mercury levels and autism spectrum disorders: A systematic review and meta-analysis. J Trace Elem Med Biol. 2017 Dec;44:289-297.

Jedrychowski, W., et al. COGNITIVE FUNCTION OF 6-YEAR OLD CHILDREN EXPOSED TO MOLD-CONTAMINATED HOMES IN EARLY POSTNATAL PERIOD. PROSPECTIVE BIRTH COHORT STUDY IN POLAND. Physiol Behav. 2011 Oct 24; 104(5): 989–995.

Jett, D.A. Chemical toxins that cause seizures. Neurotoxicology. 2012 Dec;33(6):1473-5.

Johansson, O., et al. Exacerbation of demyelinating syndrome after exposure to wireless modem with public hotspot. Electromagn Biol Med. 2016;35(4):393-7.

Julvez, J., et al. Early life multiple exposures and child cognitive function: A multi-centric birth cohort study in six European countries. Environ Pollut. 2021 Sep 1;284:117404.

Kern, J.K., et al. A biomarker of mercury body-burden correlated with diagnostic domain specific clinical symptoms of autism spectrum disorder. Biometals. 2010;23(6):1043-51.

Konkel, L. Phthalates and Autistic Traits: Exploring the Association between Prenatal Exposures and Child Behavior. Environ Health Perspec. 2020 Oct;128(10):104001.

Landrigan, P.J., et al. Children’s vulnerability to toxic chemicals: a challenge and opportunity to strengthen health and environmental policy. Health Aff. (Millwood). 2011 May;30(5):842-50.

Lanphear, B.P., et al. Low-level environmental lead exposure and children's intellectual function: an international pooled analysis. Environ Health Perspect. 2005;113(7).

Lathe, R. Environmental factors and limbic vulnerability in childhood autism; Clinical report. American Journal of Biochemistry and Biotechnology. 4 (2): 183-197, 2008.

Lombardi, C., et al. Residential proximity to pesticide application as a risk factor for childhood central nervous system tumors. Environ Res. 2021 Jun;197:111078.

Lyall, K., et al. Prenatal Serum Concentrations of Brominated Flame Retardants and Autism Spectrum Disorder and Intellectual Disability in the Early Markers of Autism Study: A Population-Based Case-Control Study in California. Environ Health Perspect. 2017 Aug 30;125(8):087023.

Malin, A.J., et al. Maternal Urinary Fluoride and Child Neurobehavior at Age 36 Months. JAMA Netw Open. 2024 May 1;7(5):e2411987.

Messer, A. Mini-review: polybrominated diphenyl ether (PBDE) flame retardants as potential autism risk factors. Physiol Behav. 2010 Jun 1;100(3):245-9.

Napoli, E., et al. Toxicity of the flame-retardant BDE-49 on brain mitochondria and neuronal progenitor striatal cells enhanced by a PTEN-deficient background. Toxicol Sci. 2013 Mar;132(1):196-210.

Palmer, R.F., et al. Environmental mercury release, special education rates, and autism disorder: an ecological study of Texas. Health Place. 2006(12):203-209.

Palmer, R.F., et al. Proximity to point sources of environmental mercury release as a predictor of autism prevalence. Health and Place. 2009 Mar;15(1):18-24.

Rauh, V.A., et al. Impact of prenatal chlorpyrifos exposure on neurodevelopment in the first 3 years of life among inner-city children. Pediatrics. 2006;118;e1845-1859.

Rowland, I.R., et al. Effects of diet on mercury metabolism and excretion in mice given methylmercury: role of gut flora. Archives of Environmental Health. Nov-Dec 1984;39(6):401-8.

Samsel, A., et al. Glyphosate, pathways to modern diseases II: Celiac sprue and gluten intolerance. Interdiscip Toxicol. 2013 Dec;6(4):159-84.

Samsel, A., et al. Glyphosate, pathways to modern diseases III: Manganese, neurological diseases, and associated pathologiesSurg Neurol Int. 2015 Mar 24;6:45.

Samsel, A, et al. Glyphosate’s suppression of cytochrome P450 enzymes and amino acid biosynthesis by the gut microbiome: Pathways to modern diseases. Entropy. 2013;15:1416–1463.

Song, Y., et al. Effects of acute exposure to aluminum on blood-brain barrier and the protection of zinc. Neurosci Lett. 2008 Nov 7;445(1):42-6.

Tang, J., et al. Exposure to 900 MHz electromagnetic fields activates the mkp-1/ERK pathway and causes blood-brain barrier damage and cognitive impairment in rats. Brain Res. 2015 Mar 19;1601:92-101.

Thompson, L., et al. What have birth cohort studies asked about genetic, pre- and perinatal exposures and child and adolescent onset mental health outcomes? A systematic review. Eur Child Adolesc Psychiatry. 2010;19(1):1-15.

U.S. Food and Drug Administration. FDA Issues Recommendations for Certain High-Risk Groups Regarding Mercury-Containing Dental Amalgam. 24 Sep 2020.

Wallinga, D., et al. Not So Sweet: Missing Mercury and High Fructose Corn Syrup. Institute for Agriculture and Trade Policy, Minneapolis, Minnesota. Jan 2009.

Wang, H.L., et al. Case-Control Study of Blood Lead Levels and Attention Deficit Hyperactivity Disorder in Chinese Children. Environmental Health Perspectives. 2008 Oct;116(10):1401-6.

Windham, G.C., et al. Autism Spectrum Disorders in Relation to Distribution of Hazardous Air Pollutants in the San Francisco Bay Area. Environmental Health Perspectives. 2006 Sep;114(9):1438-44.

Wong, S., et al. Autism, Mitochondria and Polybrominated Diphenyl Ether Exposure. CNS Neurol Disord Drug Targets. 2016;15(5):614-23.

Essential Oils

Sources & References

Beyliklioğlu, A., et al. Effect of Lavender Oil on the Anxiety of Patients Before Breast Surgery. J Perianesth Nurs. 2019 Jan 16. pii: S1089-9472(18)30356-3.

Feng, J., et al. Identification of Essential Oils with Strong Activity against Stationary Phase Borrelia burgdorferi. Antibiotics (Basel). 2018 Oct 16;7(4):89.

Feng, J., et al. Selective Essential Oils from Spice or Culinary Herbs Have High Activity against Stationary Phase and Biofilm Borrelia burgdorferi. Front Med (Lausanne). 2017 Oct 11;4:169.

Perna, S., et al. Efficacy of bergamot: From anti-inflammatory and anti-oxidative mechanisms to clinical applications as preventive agent for cardiovascular morbidity, skin diseases, and mood alterations. Food Sci Nutr. 2019 Jan 25;7(2):369-384.

Sánchez-Vidaña, D.I., et al. Lavender essential oil ameliorates depression-like behavior and increases neurogenesis and dendritic complexity in rats. Neurosci Lett. 2019 May 14;701:180-192.

Seyyed-Rasooli, A., et al. Comparing the effects of aromatherapy massage and inhalation aromatherapy on anxiety and pain in burn patients: A single-blind randomized clinical trial. Burns. 2016 Dec;42(8):1774-1780.

Trambert, R., et al. A Randomized Controlled Trial Provides Evidence to Support Aromatherapy to Minimize Anxiety in Women Undergoing Breast Biopsy. Worldviews Evid Based Nurs. 2017 Oct;14(5):394-402.

Watson, K., et al. A randomised controlled trial of Lavender (Lavandula Angustifolia) and Lemon Balm (Melissa Officinalis) essential oils for the treatment of agitated behaviour in older people with and without dementia. Complement Ther Med. 2019 Feb;42:366-373.

Feingold Diet

Sources & References

Adams, J.B., et al. Comprehensive Nutritional and Dietary Intervention for Autism Spectrum Disorder-A Randomized, Controlled 12-Month Trial. Nutrients. 2018 Mar 17;10(3).

Alberti, A., et al. Sulphation Deficit in “Low-Functioning” Autistic Children: A Pilot Study. Biol. Psychiatry. 1999;46:420–424.

Bateman, B., et al. The effects of a double blind, placebo controlled, artificial food colourings and benzoate preservative challenge on hyperactivity in a general population sample of preschool children. Arch Dis Child. 2004 Jun;89(6):506-11.

Boris, M., et al. Foods and additives are common causes of the attention deficit hyperactive disorder in children. Ann Allergy. 1994 May;72(5):462-8.

Kanarek, R.B. Artificial food dyes and attention deficit hyperactivity disorder. Nutr Rev. 2011 Jul;69(7):385-91.

Khoshnoud, M.J., et al. Effects of sodium benzoate, a commonly used food preservative, on learning, memory, and oxidative stress in brain of mice. J Biochem Mol Toxicol. 2018 Feb;32(2).

Konikowska, K., et al.  The influence of components of diet on the symptoms of ADHD in children. Rocz Panstw Zakl Hig. 2012;63(2):127-34.

Matthews, J.S., et al. Ratings of the Effectiveness of 13 Therapeutic Diets for Autism Spectrum Disorder: Results of a National Survey. J Pers Med. 2023 Sep 29;13(10):1448.

McCann, D., et al. Food additives and hyperactive behaviour in 3-year-old and 8/9-year-old children in the community: a randomised, double-blinded, placebo-controlled trial. Lancet. 2007 Nov 3;370(9598):1560-7.

Millichap, J.G., et al. The diet factor in attention-deficit/hyperactivity disorder. Pediatrics. 2012 Feb;129(2):330-7.

Moss, M., et al. The Plasma Cysteine/Sulphate Ratio: A Possible Clinical Biomarker. Journal of Nutritional & Environmental Medicine. 2009 Jul;13(4):215-229.

Naganuma, F., et al. Histamine N-methyltransferase regulates aggression and the sleep-wake cycle. Sci Rep. 2017 Nov 21;7(1):15899.

Nigg, J.T., et al. Meta-analysis of attention-deficit/hyperactivity disorder or attention-deficit/hyperactivity disorder symptoms, restriction diet, and synthetic food color additives. J Am Acad Child Adolesc Psychiatry. 2012 Jan;51(1):86-97.e8.

Schab, D.W., et al. Do artificial food colors promote hyperactivity in children with hyperactive syndromes? A meta-analysis of double-blind placebo-controlled trials. J Dev Behav Pediatr. 2004 Dec;25(6):423-34.

Silfverdal, S.A., et al. [Food additives can increase hyperactivity in children. Results from a British study confirm the connection]. Lakartidningen. 2008 Feb 6-12;105(6):354-5.

Sonuga-Barke, E.J.S., et al. Nonpharmacological interventions for ADHD: systematic review and meta-analyses of randomized controlled trials of dietary and psychological treatments. Am J Psychiatry. 2013 Mar;170(3):275-89.

Stare, F.J., et al. Diet and hyperactivity: is there a relationship. Pediatrics. 1980 Oct;66(4):521-5.

Stevens, L.J., et  al. Dietary sensitivities and ADHD symptoms: thirty-five years of research. Clin Pediatr (Phila). 2011 Apr;50(4):279-93.

Stevenson, J. The role of histamine degradation gene polymorphisms in moderating the effects of food additives on children's ADHD symptoms. Am J Psychiatry. 2010 Sep;167(9):1108-15.

Yu, C.J., et al. Sugar-Sweetened Beverage Consumption Is Adversely Associated with Childhood Attention Deficit/Hyperactivity Disorder. Int J Environ Res Public Health. 2016 Jul 4;13(7).

Food Sensitivities and Intolerances

Sources & References

Adams, J.B., et al. Comprehensive Nutritional and Dietary Intervention for Autism Spectrum Disorder—A Randomized, Controlled 12-Month Trial. Nutrients. 2018, 10, 369.

Aucoin, M., et al. Major Depressive Disorder and Food Hypersensitivity: A Case Report. Neuropsychobiology. 2019 Oct 10:1-7.

Chistol, L.T., et al. Sensory Sensitivity and Food Selectivity in Children with Autism Spectrum Disorder. J. Autism Dev. Disord. 2018;48:583–591.

De Magistris. L., et al. Antibodies against Food Antigens in Patients with Autistic Spectrum Disorders. BioMed Res. Int. 2013;2013:729349.

Ghalichi, F., et al. Effect of Gluten Free Diet on Gastrointestinal and Behavioral Indices for Children with Autism Spectrum Disorders: A Randomized Clinical Trial. World J. Pediatr. 2016;12:436–442.

Heuer, L., et al. Reduced levels of immunoglobulin in children with autism correlates with behavioral symptoms. Autism Res, Oct 2008, 1:5, 275–83.

Hsu, C.-L., et al. The Effects of a Gluten and Casein-Free Diet in Children with Autism: A Case Report. Chang Gung Med. J. 2009;32:459–465.

Jyonouchi, H., et al. Evaluation of an Association between Gastrointestinal Symptoms and Cytokine Production against Common Dietary Proteins in Children with Autism Spectrum Disorders. J. Pediatr. 2005;146:605–610.

Jyonouchi, H., et al. Impact of innate immunity in a subset of children with autism spectrum disorders: a case control study. Journal of Neuroinflammation. 2008 Nov 21;5:52.

Jyonouchi, H., et al. Innate Immunity Associated with Inflammatory Responses and Cytokine Production against Common Dietary Proteins in Patients with Autism Spectrum Disorder. Neuropsychobiology. 2002;46:76–84. 

Karakula-Juchnowicz, H., et al. The Food-Specific Serum IgG Reactivity in Major Depressive Disorder Patients, Irritable Bowel Syndrome Patients and Healthy Controls. Nutrients. 2018 Apr 28;10(5). pii: E548.

Karakula-Juchnowicz, H., et al. The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders. Nutr Neurosci. 2017 Feb;20(2):110-118.

Kim-Lee, C., et al. Gastrointestinal disease in Sjogren's syndrome: related to food hypersensitivities. Springerplus. 2015 Dec 12;4:766.

Lucarelli, S., et al. Food Allergy and Infantile Autism. Panminerva Med. 1995;37:137–141. 

Martin, V.T., et al. Diet and Headache: Part 1. Headache. 2016 Oct;56(9):1543-1552.

Pelsser, L.M., et al. Effects of a restricted elimination diet on the behaviour of children with attention-deficit hyperactivity disorder (INCA study): a randomised controlled trial. Lancet. 2011 Feb 5;377(9764):494-503.

Peters, R.L., et al. Infant food allergy phenotypes and association with lung function deficits and asthma at age 6 years: a population-based, prospective cohort study in Australia. Lancet Child Adolesc Health. 2023 Jul 24;S2352-4642(23)00133-5.

Philpott, H., et al. Allergy tests do not predict food triggers in adult patients with eosinophilic oesophagitis. A comprehensive prospective study using five modalities. Aliment Pharmacol Ther. 2016 Aug;44(3):223-33.

Severance, E.G., et al. IgG dynamics of dietary antigens point to cerebrospinal fluid barrier or flow dysfunction in first-episode schizophrenia. Brain Behav Immun. 2015 Feb;44:148-58.

Shakoor, Z., et al. Prevalence of IgG-mediated food intolerance among patients with allergic symptoms. Ann Saudi Med. 2016 Nov-Dec;36(6):386-390.

Sloper, K.S., et al. Children with atopic eczema. II: Immunological findings associated with dietary manipulations. Q J Med. 1991 Aug;80(292):695-705.

Stockton, S., et al. The Impact of a Food Elimination Diet on Collegiate Athletes’ 300-meter Run Time and Concentration. Glob Adv Health Med. 2014 Nov;3(6):25-40.

Suen, R.M., et al. The Clinical Relevance of IgG Food Allergy Testing Through ELISA. Townsend Letter for Doctors & Patients, Jan 2004, 61–66.

Uhde, M., et al. Intestinal cell damage and systemic immune activation in individuals reporting sensitivity to wheat in the absence of coeliac disease. Gut. 2016 Dec;65(12):1930-1937.

Verena, L., et al. Elimination diets’ efficacy and mechanisms in attention deficit hyperactivity disorder and autism spectrum disorder. Eur Child Adolesc Psychiatry. 2017; 26(9): 1067–1079.

Virdee, K., et al. Food-specific IgG Antibody-guided Elimination Diets Followed by Resolution of Asthma Symptoms and Reduction in Pharmacological Interventions in Two Patients: A Case Report. Glob Adv Health Med. 2015 Jan;4(1):62-6.

Vita, A.A., et al. Associations between Food-Specific IgG Antibodies and Intestinal Permeability Biomarkers. Front. Nutr. 2022;9:962093.

Westmark, C.J. Soy Infant Formula and Seizures in Children with Autism: A Retrospective Study. PLoS ONE. 2014;9:e80488.

Xie, Y., et al. Effects of Diet Based on IgG Elimination Combined with Probiotics on Migraine Plus Irritable Bowel Syndrome. Pain Res Manag. 2019 Aug 21;2019:7890461.

Gastrointestinal Disorders

Sources & References

Adams, J.B., et al. Gastrointestinal Flora and Gastrointestinal Status in Children with Autism—Comparisons to Typical Children and Correlation with Autism Severity. BMC Gastroenterol. 2011;11:22.

Aguilera, M., et al. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice. Gut Microbes. 2015;6(1):10-23.

Appleton, J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr Med (Encinitas). 2018 Aug;17(4):28-32.

Aroniadis, O.C., et al. Fecal microbiota transplantation: past, present and future. Curr Opin Gastroenterol. 2013;29(1):79-84.

Assa, A., et al. Vitamin D deficiency promotes epithelial barrier dysfunction and intestinal inflammation. J Infect Dis. 2014;210(8):1296-305.

Atladóttir, H.Ó., et al. Autism after infection, febrile episodes, and antibiotic use during pregnancy: an exploratory study. Pediatrics. 2012 Dec;130(6):e1447-54.

Aversa, Z., et al. Association of Infant Antibiotic Exposure With Childhood Health Outcomes. Mayo Clin Proc. 2020 Nov 6;S0025-6196(20)30785-0.

Bäckhed, F., et al. Host-Bacterial Mutualism in the Human Intestine. Science. 2005 Mar 25;307(5717):1915-20.

Bennings, M.A., et al. Colonic transit times and behaviour profiles in children with defecation disorders. Archives of the Diseases of Childhood. 2004 Jan;89(1):13-6.

Berding, K., et al. Diet Can Impact Microbiota Composition in Children With Autism Spectrum Disorder. Front. Neurosci. 2018;12:515.

Bjørklund, G., et al. Gastrointestinal alterations in autism spectrum disorder: What do we know? Neurosci Biobehav Rev. 2020 Nov:118:111-120.

Bora, S.A., et al. Regulation of vitamin D metabolism following disruption of the microbiota using broad spectrum antibiotics. J Nutr Biochem. 2018 Jun;56:65-73.

Borchers, A.T., et al. Probiotics and immunity. Journal of Gastroenterology. 44 (2009): 26-46.

Borre, Y.E., et al. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med. 2014 Sep;20(9):509-18.

Buccigrossi, V., et al. Functions of intestinal microflora in children. Curr Opin Gastroenterol. 2013;29(1):31-8.

Buie, T., et al. Evaluation, diagnosis, and treatment of gastrointestinal disorders in individuals with ASDs: a consensus report. Pediatrics. 2010 Jan;125 Suppl 1:S1-18.

Camilleri, M. Serotonin in the gastrointestinal tract. Curr Opin Endrocrinol Diabetes Obes. 2009 Feb;16(1):53-9.

Carding, S., et al. Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis. 2015;26:26191.

Chen, C.Q., et al. Distribution, function and physiological role of melatonin in the lower gut. World J Gastroenterol. 2011 Sep 14;17(34):3888-98.

Chen, Y., et al. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients. 2021 Jun 19;13(6):2099.

Clark, J.A., et al. Intestinal crosstalk—a new paradigm for understanding the gut as the ‘motor’ of critical illness. Shock. 2007 Oct;28(4):384-93.

Cohen-Mekelburg, S., et al. Morning light treatment for inflammatory bowel disease: a clinical trial. BMC Gastroenterol. 2024 May 22;24(1):179.

Correale, J., et al. The role of the gut microbiota in multiple sclerosis. Nat Rev Neurol. 2022 Sep;18(9):544-558.

Critchfield, et al. The potential role of probiotics in the management of childhood autism spectrum disorders. Gastroenterol Res Pract. 2011;2011:161358.

Cucchiara, S., et al. Interactions between intestinal microbiota and innate immune system in pediatric inflammatory bowel disease. J Clin Gastroenterol. 2012;46 Suppl:S64-6.

Cui, J., et al. Butyrate-Producing Bacteria and Insulin Homeostasis: The Microbiome and Insulin Longitudinal Evaluation Study (MILES). Diabetes. 2022 Nov 1;71(11):2438-2446.

de Goffau, et al. Fecal microbiota composition differs between children with beta-cell autoimmunity and those without. Diabetes. 2013;62(4):1238-44.

D’Eufemia, P., et al. Abnormal intestinal permeability in children with autism. Acta Paediatr. 1996 Sep;85(9):1076-9.

de Magistris, et al. Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J Pediatr Gastroenterol Nutr. 2010;51(4):418-24.

Derrien, M., et al. The Gut Microbiota in the First Decade of Life. Trends Microbiol. 2019 Dec;27(12):997-1010.

Dinan, T.G., et al. The Microbiome-Gut-Brain Axis in Health and Disease. Gastroenterol Clin North Am. 2017 Mar;46(1):77-89.

Dogra, S.K., et al. Gut Microbiota Resilience: Definition, Link to Health and Strategies for Intervention. Front Microbiol. 2020 Sep 15:11:572921.

Edwards, C.A., et al. Intestinal flora during the first months of life: new perspectives. British Journal of Nutrition. 88 (2002): S11-S18.

Erickson, C.A., et al. Gastrointestinal Factors in Autistic Disorder: A Critical Review. Journal of Autism and Developmental Disorders. 2005 Dec;35(6):713-27.

Fattorusso, A., et al. Autism Spectrum Disorders and the Gut Microbiota. Nutrients. 2019 Feb 28;11(3):521.

Feng, P., et al. A review of probiotics in the treatment of autism spectrum disorders: Perspectives from the gut-brain axis. Front Microbiol. 2023 Mar 16:14:1123462.

Fukuda, K., et al. Determination of the discriminant score of intestinal microbiota as a biomarker of disease activity in patients with ulcerative colitis. BMC Gastroenterol. 2014;14:49.

Galland, L. The gut microbiome and the brain. J Med Food 2014; 17(12): 1261-72.

Ganal-Vonarburg, S.C., et al. Microbial-host molecular exchange and its functional consequences in early mammalian life. Science. 2020 May 8;368(6491):604-607.

Gastrointestinal permeability in food-allergic children. Nutr Rev. 1985 Aug;43(8):233-5.

Grizotte-Lake, M., et al. Commensals Suppress Intestinal Epithelial Cell Retinoic Acid Synthesis to Regulate Interleukin-22 Activity and Prevent Microbial Dysbiosis. Immunity. 2018 Dec 18;49(6):1103-1115.e6.

Guandalini, S. Are probiotics or prebiotics useful in pediatric irritable bowel syndrome or inflammatory bowel disease? Front Med (Lausanne). 2014;1:23.

Guandalini, S., et al. Prebiotics and probiotics in irritable bowel syndrome and inflammatory bowel disease in children. Benef Microbes. 2015;6(2):209-17.

Hamad, A.F., et al. Prenatal antibiotics exposure and the risk of autism spectrum disorders: A population-based cohort study. PLoS One. 2019 Aug 29;14(8):e0221921.

Han, H., et al. Gut Microbiota and Type 1 Diabetes. Int J Mol Sci. 2018 Mar 27;19(4):995.

Hanaway, P. Balance of Flora, GALT, and Mucosal Integrity. Alternative Therapies in Health and Medicine. Sep-Oct 2006;12(5):52-60; quiz 61-2.

Hejitz, R.D., et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011 Feb 15;108(7):3047-52.

Horn, J., et al. Role of Diet and Its Effects on the Gut Microbiome in the Pathophysiology of Mental Disorders. Transl. Psychiatry. 2022;12:164.

Horvath, K., et al. Autism and gastrointestinal symptoms. Curr Gastroenterol Rep. 2002 Jun;4(3):251-8.

Horvath, K., et al. Autistic disorder and gastrointestinal disease. Current Opinion in Pediatrics. 2002 Oct;14(5):583-7.

Horvath, K., et al. Gastrointestinal abnormalities in children with autistic disorder. Journal of Pediatrics. 1999 Nov;135(5):559-63.

Hrncir, T., et al. Gut microbiota and lipopolysaccharide content of the diet influence development of regulatory T cells: studies in germ-free mice. BMC Immunology. 9 (2008): 65.

Hyman, M.A. Is the Cure for Brain Disorders Outside the Brain? Alternative Therapies in Health and Medicine. Nov-Dec 2007;13(6):10-5.

Isaksson, J., et al. Brief Report: Association Between Autism Spectrum Disorder, Gastrointestinal Problems and Perinatal Risk Factors Within Sibling Pairs. J Autism Dev Disord. 2017 Aug;47(8):2621-2627.

Jackson, P.G., et al. Intestinal permeability in patients with eczema and food allergy. Lancet. 1981 Jun 13;1(8233):1285-6.

Jakobsen, C., et al. Environmental factors and risk of developing paediatric inflammatory bowel disease — a population based study. 2007-2009. J Crohns Colitis. 2013;7(1):79-88.

Jyonouchi, H., et al. Dysregulated innate immune responses in young children with autism spectrum disorders: their relationship to gastrointestinal symptoms and dietary intervention. Neuropsychobiology. 2005;51(2):77-85.

Jyonouchi, H., et al. Evaluation of an Association between Gastrointestinal Symptoms and Cytokine Production against Common Dietary Proteins in Children with Autism Spectrum Disorders. J. Pediatr. 2005;146:605–610.

Kang, D.W., et al. Long-term benefit of Microbiota Transfer Therapy on autism symptoms and gut microbiota. Scientific Reports. 9, 5821 (2019).

Kang, D.W., et al. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome.2017 Jan 23;5(1):10.

Karagözlü, S., et al. The Relationship of Severity of Autism with Gastrointestinal Symptoms and Serum Zonulin Levels in Autistic Children. J. Autism Dev. Disord. 2022;52:623–629.

Kelly, J.R., et al. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disordersFront Cell Neurosci. 2015 Oct 14;9:392.

Kim-Lee, C., et al. Gastrointestinal disease in Sjogren's syndrome: related to food hypersensitivities. Springerplus. 2015 Dec 12;4:766.

Kitano, H., et al. Robustness trade-offs and host-microbial symbiosis in the immune system. Molecular Systems Biology. 2 (2006).

Kobliner, V., et al. Reduction in Obsessive Compulsive Disorder and Self-Injurious Behavior With Saccharomyces boulardii in a Child with Autism: A Case Report. Integr Med (Encinitas). 2018 Dec;17(6):38-41.

Korpela, K., et al. Maternal Fecal Microbiota Transplantation in Cesarean-Born Infants Rapidly Restores Normal Gut Microbial Development: A Proof-of-Concept Study. Cell, 2020.

Lai, C.C.W., et al. The association between gut-health promoting diet and depression: A mediation analysis. J Affect Disord. 2023 Mar 1:324:136-142.

Lavebratt, C., et al. Early exposure to antibiotic drugs and risk for psychiatric disorders: a population-based study. Transl Psychiatry. 2019 Nov 26;9(1):317.

Liu, Z., et al. Tight junctions, leaky intestines, and pediatric diseases. Acta Paediatricia. 94 (2005): 386-393.

Madra, M., et al. Gastrointestinal Issues and Autism Spectrum Disorder. Psychiatr Clin North Am. 2021 Mar; 44(1): 69–81.

Manichanh, C., et al. The gut microbiota in IBD. Nat Rev Gastroenterol Hepatol. 2012;9(10):599-608.

Mine, Y., et al. Surfactants Enhance the Tight-Junction Permeability of Food Allergens in Human Intestinal Epithelial Caco-2 Cells. International Archives of Allergy and Immunology. 2003 Feb;130(2):135-42.

Mitre, E., et al. Association Between Use of Acid-Suppressive Medications and Antibiotics During Infancy and Allergic Diseases in Early Childhood. JAMA Pediatr. 2018 Jun 4;172(6):e180315.

Mohan, R., et al. Effects of Bifidobacterium lactis Bb12 Supplementation on Intestinal Microbiota of Preterm Infants: A Double-Blind, Placebo-Controlled, Randomized Study. Journal of Clinical Microbiology. 2006 Nov;44(11):4025-31.

Möller, C., et al. Intestinal permeability as assessed with polyethyleneglycols in birch pollen allergic children undergoing oral immunotherapy. Allergy. 1986 May;41(4):280-5.

Montalto, M., et al. Fecal Calprotectin Concentrations in Patients with Small Intestinal Bacterial Overgrowth. Digestive Diseases. 2008;26(2):183-6.

Morris, C.R., et al. Syndrome of allergy, apraxia, and malabsorption: characterization of a neurodevelopmental phenotype that responds to omega 3 and vitamin E supplementation. Alternative Therapies in Health and Medicine. Jul-Aug 2009;15(4):34-43.

Moser, L.A. Astrovirus Increases Epithelial Barrier Permeability Independently of Viral Replication. Journal of Virology. 2007 Nov;81(21):11937-45.

Nankova, B.B., 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(8):e103740.

Nankova, B.B. Nicotinic Induction of Preproenkephalin and Tyrosine Hydroxylase Gene Expression in Butyrate-Differentiated Rat PC12 Cells: A Model for Adaptation to Gut-Derived Environmental Signals. Pediatric Research. 2003 Jan;53(1):113-8.

Nemechek, P., et al. Autism Spectrum Disorder Symptoms Improve with Combination Therapy Directed at Improving Gut Microbiota and Reducing Inflammation. Applied Psychiatry. 2020 Jul; (1)1.

Niederhofer, H., et al. A preliminary investigation of ADHD symptoms in persons with celiac disease. J Atten Disorder. 2006 Nov;10(2):200-4.

Nirmalkar, K., et al. Shotgun Metagenomics Study Suggests Alteration in Sulfur Metabolism and Oxidative Stress in Children with Autism and Improvement after Microbiota Transfer Therapy. Int J Mol Sci. 2022 Nov 3;23(21):13481.

Nikolova, V.L., et al. Acceptability, Tolerability, and Estimates of Putative Treatment Effects of Probiotics as Adjunctive Treatment in Patients With Depression. JAMA Psychiatry. 2023 Aug 1;80(8):842-847.

Nirmalkar, K., et al. Shotgun Metagenomics Study Suggests Alteration in Sulfur Metabolism and Oxidative Stress in Children with Autism and Improvement after Microbiota Transfer Therapy. Int J Mol Sci. 2022 Nov 3;23(21):13481.

O’Hara, A.M., et al. The gut flora as a forgotten organ. European Molecular Biology Organization Report 7, no 7 (July 2006): 688-693.

Oliva-Hemker, M., et al. Fecal Microbiota Transplantation: Information for the Pediatrician. Pediatrics. 2023 Dec 1;152(6):e2023062922.

Ozkul, C., et al. A single early-in-life antibiotic course increases susceptibility to DSS-induced colitis. Genome Med. 2020;12(1):65.

Parkin, K.,, et al. Risk Factors for Gut Dysbiosis in Early Life. Microorganisms. 2021 Sep 30;9(10):2066.

Pearson, A.D., et al. Intestinal permeability in children with Crohn's disease and coeliac disease. Br Med J (Clin Res Ed). 1982 Jul 3;285(6334):20-1.

Rautava, S., et al. The Development of Gut Immune Responses and Gut Microbiota: Effects of Probiotics in Prevention and Treatment of Allergic Disease. Current Issues in Intestinal Microbiology. 2002 Mar;3(1):15-22.

Rediers, H., et al. Unraveling the Secret Lives of Bacteria: Use of In Vivo Expression Technology and Differential Fluorescence Induction Promoter Traps as Tools for Exploring Niche-Specific Gene Expression. Microbiology and Molecular Biology Reviews. 2005 Jun;69(2):217-61.

Relman, D.A. The human microbiome: ecosystem resilience and health. Nutr Rev. 2012 Aug;70 Suppl 1(Suppl 1):S2-9.

Ristori, M.V., et al. Autism, Gastrointestinal Symptoms and Modulation of Gut Microbiota by Nutritional Interventions. Nutrients. 2019;11:2812.

Rook, G.A., et al. Microbiota, immunoregulatory old friends and psychiatric disorders. Adv Exp Med Biol. 2014;817:319-56.

Rowland, I.R., et al. Effects of diet on mercury metabolism and excretion in mice given methylmercury: role of gut flora. Archives of Environmental Health. Nov-Dec 1984;39(6):401-8.

Sanchez, A., et al. Role of sugars in human neutrophilic phagocytosis. Am J Clin Nutr. 1973 Nov;26(11):1180-4.

Savino, F., et al. Lactobacillus reuteri (American Type Culture Collection Strain 55730) versus simethicone in the treatment of infantile colic: a prospective randomized study. Pediatrics. 2007 Jan;119(1):e124-30.

Scirocco, A., et al. Exposure of Toll-like receptors 4 to bacterial lipopolysaccharide (LPS) impairs human colonic smooth muscle cell function. J Cell Physiol. 2010 May;223(2):442-50.

Sela, D.A., et al. The marriage of nutrigenomics with the microbiome: the case of infant associated bifidobacteria and milk. Am J Clin Nutr. 2014;99(3):697S-703S.

Severance, E.G., et al. IgG dynamics of dietary antigens point to cerebrospinal fluid barrier or flow dysfunction in first-episode schizophrenia. Brain Behav Immun. 2015 Feb;44:148-58.

Shekhawat, P.S., et al. Spontaneous development of intestinal and colonic atrophy and inflammation in the carnitine-deficient jvs (OCTN2(-/-)) mice. Mol Genet Metab. 2007 Dec;92(4):315-24.

Sifroni, K.G., et al. Mitochondrial respiratory chain in the colonic mucosal of patients with ulcerative colitis. Mol Cell Biochem. 2010 Sep;342(1-2):111-5.

Simpson, C.A., et al. The gut microbiota in anxiety and depression - A systematic review. Clin Psychol Rev. 2021 Feb:83:101943.

Stewart, C.J., et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature. 2018 Oct;562(7728):583-588.

Strauch, U.G., et al. Influence of intestinal bacteria on induction of regulatory T cells: lessons from a transfer model of colitis. Gut 54 (2005):1546-1552.

Strandwitz, P. Neurotransmitter Modulation by the Gut Microbiota. Brain Res. 2018 Aug 15;1693(Pt B):128-133.

Tobacman, J.K. Review of Harmful Gastrointestinal Effects of Carrageenan in Animal Experiments. Environmental Health Perspectives. 2001 Oct;109(10):983-94.

Uhde, M., et al. Intestinal cell damage and systemic immune activation in individuals reporting sensitivity to wheat in the absence of coeliac disease. Gut. 2016 Dec;65(12):1930-1937.

Vael, C., et al. Early intestinal Bacteroides fragilis colonization and development of asthma. BMC Pulmonary Medicine. 2008 Sep 26;8:19.

van Nood, E., et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013 Jan 31;368(5):407-15.

Voigt, R.M., et al. The intestinal microbiota: determinants of resiliency? Lancet Healthy Longev. 2021 Jan;2(1):e2-e3.

Vojdani, A., et al. A Gut Feeling for Immune Dysregulation & Neuroinflammation in Autism. The Autism File. 2009(31).

Wang, J., et al. Global Prevalence of Autism Spectrum Disorder and Its Gastrointestinal Symptoms: A Systematic Review and Meta-Analysis. Front. Psychiatry. 2022;13:963102.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Wasilewska, J., et al. Gastrointestinal symptoms and autism spectrum disorder: links and risks – a possible new overlap syndrome. Pediatric Health Med Ther. 2015; 6: 153–166.

West, C.E., et al. The gut microbiota and inflammatory noncommunicable diseases: associations and potentials for gut microbiota therapies. J Allergy Clin Immunol. 2015;135(1):3-13; quiz 4.

Wexler, H. Bacteroides: the Good, the Bad, and the Nitty-Gritty. Clinical Microbiology Reviews 20, no. 4 (October 2007): 593-621.

White, E., et al. The Effect of Nutritional Therapy for Yeast Infection (Candidiasis) in Cases of Chronic Fatigue Syndrome. Journal of Orthomolecular Medicine. 2005;20(3).

Williams, B.L., et al. Impaired Carbohydrate Digestion and Transport and Mucosal Dysbiosis in the Intestines of Children with Autism and Gastrointestinal Disturbances. PLoS ONE. 2011;6:e24585.

Williams, R.E.O., et al. The influence of intestinal bacteria on the absorption and metabolism of foreign compounds. Journal of Clinical Pathology.1971; 5: 125–129.

Wilmanski, T., et al. Gut microbiome pattern reflects healthy ageing and predicts survival in humans. Nat Metab. 2021 Feb;3(2):274-286.

Yang, B., et al. Effects of regulating intestinal micobiota on anxiety symptoms: A systematic review. General Psychiatry. 2019; 32: e100056.

Zheng, P., et al. Gut microbiome in type 1 diabetes: A comprehensive review. Diabetes Metab Res Rev. 2018 Oct;34(7):e3043.

Zhou, H., et al. Evaluating the Causal Role of Gut Microbiota in Type 1 Diabetes and Its Possible Pathogenic Mechanisms. Front Endocrinol (Lausanne). 2020 Mar 24:11:125.

Zhu, B., et al. Human gut microbiome: the second genome of human body. Protein Cell. 2010 Aug;1(8):718-25.

Gluten-Free/Casein-Free Diet

Sources & References

Adams, J.B., et al. Comprehensive Nutritional and Dietary Intervention for Autism Spectrum Disorder-A Randomized, Controlled 12-Month Trial. Nutrients. 2018 Mar 17;10(3).

Cekici, H., et al. Current Nutritional Approaches in Managing Autism Spectrum Disorder: A Review. Nutr. Neurosci. 2019;22:145–155.

El-Rashidy, O., et al. Ketogenic Diet versus Gluten Free Casein Free Diet in Autistic Children: A Case-Control Study. Metab. Brain Dis. 2017;32:1935–1941.

Epstein, S.S. Unlabeled milk from cows treated with biosynthetic growth hormones: a case of regulatory abdication. Int J Health Serv. 1996;26(1):173-85.

Ghalichi, F., et al. Effect of Gluten Free Diet on Gastrointestinal and Behavioral Indices for Children with Autism Spectrum Disorders: A Randomized Clinical Trial. World J. Pediatr. 2016;12:436–442.

Hadjivassiliou, M., et al. Gluten sensitivity: from gut to brain. Lancet Neurol. 2010 Mar;9(3):318-30.

Hartman, R.E., et al. Dietary Approaches to the Management of Autism Spectrum Disorders. Adv. Neurobiol. 2020;24:547–571.

Herbert, M.R., et al. Autism and Dietary Therapy: Case Report and Review of the Literature. J. Child. Neurol. 2013;28:975–982.

Hsu, C.-L., et al. The Effects of a Gluten and Casein-Free Diet in Children with Autism: A Case Report. Chang Gung Med. J. 2009;32:459–465.

Julian, T., et al. Gluten sensitivity and epilepsy: a systematic review. J Neurol. 2018 Aug 23.

Juntti, H., et al. Cow’s milk allergy is associated with recurrent otitis media during childhood. Acta Otolaryngol. 1999;119:867–873.

Karakula-Juchnowicz, H., et al. The Food-Specific Serum IgG Reactivity in Major Depressive Disorder Patients, Irritable Bowel Syndrome Patients and Healthy Controls. Nutrients. 2018 Apr 28;10(5). pii: E548.

Karakula-Juchnowicz, H., et al. The role of IgG hypersensitivity in the pathogenesis and therapy of depressive disorders. Nutr Neurosci. 2017 Feb;20(2):110-118.

Knivsberg, A.M., et al. A Randomised, Controlled Study of Dietary Intervention in Autistic Syndromes. Nutr. Neurosci. 2002;5:251–261.

Lee, R.W.Y., et al. A Modified Ketogenic Gluten-Free Diet with MCT Improves Behavior in Children with Autism Spectrum Disorder. Physiol. Behav. 2018;188:205–211.

Lionetti, E., et al. Gluten Psychosis: Confirmation of a New Clinical Entity. Nutrients. 2015 Jul 8;7(7):5532-9.

Marí-Bauset, S., et al. Nutritional Impact of a Gluten-Free Casein-Free Diet in Children with Autism Spectrum Disorder. J. Autism Dev. Disord. 2016;46:673–684.

Martin, V.T., et al. Diet and Headache: Part 1. Headache. 2016 Oct;56(9):1543-1552.

Matthews, J.S., et al. Ratings of the Effectiveness of 13 Therapeutic Diets for Autism Spectrum Disorder: Results of a National Survey. J Pers Med. 2023 Sep 29;13(10):1448.

Nsouli, T.M., et al. The role of food allergy in serious otitis media. Ann Allergy. 1994 Sep; 66:91.

Pelsser, L.M., et al. Effects of a restricted elimination diet on the behaviour of children with attention-deficit hyperactivity disorder (INCA study): a randomised controlled trial. Lancet. 2011 Feb 5;377(9764):494-503.

Piwowarczyk, A., et al. Gluten-Free Diet in Children with Autism Spectrum Disorders: A Randomized, Controlled, Single-Blinded Trial. J. Autism Dev. Disord. 2020;50:482–490.

Quan, L., et al. A Systematic Review and Meta-Analysis of the Benefits of a Gluten-Free Diet and/or Casein-Free Diet for Children with Autism Spectrum Disorder. Nutr. Rev. 2022;80:1237–1246.

Ramakrishnan, J.B.. The role of food allergy in otolaryngology disorders. Curr Opin Otolaryngol Head Neck Surg. 2010;18:195–199.

Rizwan, M.Z., et al. Dietary wheat gluten induces astro- and microgliosis in the hypothalamus of male mice. J Neuroendocrinol. 2023 Jul 17;e13326.

Shakoor, Z., et al. Prevalence of IgG-mediated food intolerance among patients with allergic symptoms. Ann Saudi Med. 2016 Nov-Dec;36(6):386-390.

Sloper, K.S., et al. Children with atopic eczema. II: Immunological findings associated with dietary manipulations. Q J Med. 1991 Aug;80(292):695-705.

Stockton, S., et al. The Impact of a Food Elimination Diet on Collegiate Athletes’ 300-meter Run Time and Concentration. Glob Adv Health Med. 2014 Nov;3(6):25-40.

Verena, L., et al. Elimination diets’ efficacy and mechanisms in attention deficit hyperactivity disorder and autism spectrum disorder. Eur Child Adolesc Psychiatry. 2017; 26(9): 1067–1079.

Virdee, K., et al. Food-specific IgG Antibody-guided Elimination Diets Followed by Resolution of Asthma Symptoms and Reduction in Pharmacological Interventions in Two Patients: A Case Report. Glob Adv Health Med. 2015 Jan;4(1):62-6.

Whiteley, P., et al. The ScanBrit Randomised, Controlled, Single-Blind Study of a Gluten- and Casein-Free Dietary Intervention for Children with Autism Spectrum Disorders. Nutr. Neurosci. 2010;13:87–100.

Xie, Y., et al. Effects of Diet Based on IgG Elimination Combined with Probiotics on Migraine Plus Irritable Bowel Syndrome. Pain Res Manag. 2019 Aug 21;2019:7890461.

Yu, Y., et al. Efficacy and Safety of Diet Therapies in Children With Autism Spectrum Disorder: A Systematic Literature Review and Meta-Analysis. Front. Neurol. 2022;13:844117.

Grounding Energy

Hyperbaric Oxygen Therapy (HBOT)

Sources & References

Hypotonia and Hypertonia

Sources & References

Dehley, Leanna M., et al. The Effect of Mitochondrial Supplements on Mitochondrial Activity in Children with Autism Spectrum Disorder. J Clin Med. 2017 Feb; 6(2): 18.

Filipek, P.A., et al. Relative carnitine deficiency in autism. J Autism Dev Disord. 2004 Dec;34(6):615-23.

Hao, J., et al. Mitochondrial nutrients improve immune dysfunction in the type 2 diabetic Goto-Kakizaki rats. J Cell Mol Med. 2009 Apr;13(4):701-11.

Liu, J. The effects and mechanisms of mitochondrial nutrient alpha-lipoic acid on improving age-associated mitochondrial and cognitive dysfunction: an overview. Neurochem Res. 2008 Jan;33(1):194-203.

Long, J., et al. Mitochondrial decay in the of old rats: ameliorating effect of alpha-lipoic acid and acetyl-L-carnitine. Neurochem Res. 2009 Apr;34(4):755-63.

Mabalirajan, U., et al. Effects of vitamin E on mitochondrial and asthma features in an experimental allergic murine model. J Appl Physiol. 2009 Oct;107(4):1285-92.

Maes, M., et al. Coenzyme Q10 deficiency in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is related to fatigue, autonomic and neurocognitive symptoms and is another risk factor explaining the early mortality in ME/CFS due to cardiovascular disorder. Neuro Endocrinol Lett. 2009;30(4):470-6.

Maes, M., et al. Lower plasma Coenzyme Q10 in depression: a marker for treatment resistance and chronic fatigue in depression and a risk factor to cardiovascular disorder in that illness. Neuro Endocrinol Lett. 2009;30(4):462-9.

Noland, R.C., et al. Carnitine insufficiency caused by aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem. 2009 Aug 21;284(34):22840-52.

Power, R.A., et al. Carnitine revisited: potential use as adjunctive treatment in diabetes. Diabetologia. 2007 Apr;50(4):824-32.

Shen, W., et al. Protective effects of R-alpha-lipoic acid and acetyl-L-carnitine in MIN6 and isolated rat islet cells chronically exposed to oleic acid. J Cell Biochem. 2008 Jul 1;104(4):1232-43.

Shekhawat, P.S., et al. Spontaneous development of intestinal and colonic atrophy and inflammation in the carnitine-deficient jvs (OCTN2(-/-)) mice. Mol Genet Metab. 2007 Dec;92(4):315-24.

Spindler, M., et al. Coenzyme Q10 effects in neurodegenerative disease. Neuropsychiatr Dis Treat. 2009;5:597-610.

Zhang, H., et al. Combined Ralpha-lipoic acid and acetyl-L-carnitine exerts efficient preventative effects in a cellular model of Parkinson’s disease. J Cell Mol Med. 2010 Jan;14(1-2):215-25.

Interactive Metronome

Sources & References

Bartscherer, et al. Interactive metronome training for a 9-year-old boy with attention and motor coordination difficulties. Physiother Theory Pract. Oct-Dec 2005;21(4):257-69.

Blondis, T.A. Motor disorders and attention-deficit/hyperactivity disorder. Pediatr Clin North Am. 1999 Oct;46(5):899-913, vi-vii.

Cosper, S.M., et al. Interactive Metronome training in children with attention deficit and developmental coordination disorders. Int J Rehabil Res. 2009 Dec;32(4):331-6.

Goulardins, J.B., et al. Attention deficit hyperactivity disorder and developmental coordination disorder: Two separate disorders or do they share a common etiology. Behav Brain Res. 2015 Oct 1;292:484-92.

Goulardins, J.B., et al. Attention Deficit Hyperactivity Disorder and Motor Impairment. Percept Mot Skills. 2017 Apr;124(2):425-440.

Karatekin, C., et al. A preliminary study of motor problems in children with attention-deficit/hyperactivity disorder. Percept Mot Skills. 2003 Dec;97(3 Pt 2):1267-80.

Koomar, J., et al. Theoretical and clinical perspectives on the Interactive Metronome: a view from occupational therapy practice. Am J Occup Ther. Mar-Apr 2001;55(2):163-6.

McLeod, K.R., et al. Functional connectivity of neural motor networks is disrupted in children with developmental coordination disorder and attention-deficit/hyperactivity disorder. Neuroimage Clin. 2014 Mar 26;4:566-75.

Piek, J.P., et al. Motor coordination and kinaesthesis in boys with attention deficit-hyperactivity disorder. Dev Med Child Neurol. 1999 Mar;41(3):159-65.

Shaffer, R.J., et al. Effect of interactive metronome training on children with ADHD. Am J Occup Ther. Mar-Apr 2001;55(2):155-62.

Slater, J.L., et al. Timing Deficits in ADHD: Insights From the Neuroscience of Musical Rhythm. Front Comput Neurosci. 2018 Jul 6;12:51.

Smith, A., et al. Evidence for a pure time perception deficit in children with ADHD. J Child Psychol Psychiatry. 2002 May;43(4):529-42.

Sonuga-Barke, E., et al. Beyond the dual pathway model: evidence for the dissociation of timing, inhibitory, and delay-related impairments in attention-deficit/hyperactivity disorder. J Am Acad Child Adolesc Psychiatry. 2010 Apr;49(4):345-55.

Teicher, M.H. Final White Paper: Effects of Brain Balance Exercises and Interactive Metronome on Children with Attention Deficit Hyperactivity Disorder are Similar to the Effects of Stimulant Medication. Harvard Medical School, Department of Psychiatry. 2020.

Toplak, M.E., et al. Time perception: modality and duration effects in attention-deficit/hyperactivity disorder (ADHD). J Abnorm Child Psychol. 2005 Oct;33(5):639-54.

Yang, B., et al. Time perception deficit in children with ADHD. Brain Res. 2007 Sep 19;1170:90-6.

Ketogenic Diet

Sources & References

Accurso, A., et al. Dietary carbohydrate restriction in type 2 diabetes mellitus and metabolic syndrome: time for a critical appraisal. Nutr Metab (Lond). 2008 Apr 8:5:9.

Acuña-Catalán, D., et al. Ketogenic diet administration later in life improves memory by modifying the synaptic cortical proteome via the PKA signaling pathway in aging mice. Cell Rep Med. 2024 Jun 18;5(6):101593.

Adebayo, O., et al. The changing face of diabetes in America. Emerg Med Clin North Am. 2014;32(2):319-27.

Aggarwal, B.B., et al. Molecular targets of dietary agents for prevention and therapy of cancer. Biochem Pharmacol. 2006 May 14;71(10):1397-421.

Allen, B.G., et al. Ketogenic diets as an adjuvant cancer therapy: History and potential mechanism. Redox Biol. 2014:2:963-70.

Anand, P., et al. Cancer is a preventable disease that requires major lifestyle changes. Pharm Res. 2008 Sep;25(9):2097-116.

Aucoin, M., et al. Generalized anxiety disorder and hypoglycemia symptoms improved with diet modificationCase Rep Psychiatry. 2016;2016.

Bădescu, S.V., et al. The association between Diabetes mellitus and Depression. J Med Life. 2016 Apr-Jun; 9(2): 120–125.

Bahr, L.S., et al. Ketogenic diet and fasting diet as Nutritional Approaches in Multiple Sclerosis (NAMS): protocol of a randomized controlled study. Trials. 2020 Jan 2;21(1):3.

Banerjee, S., et al. Ayurveda in changing scenario of diabetes management for developing safe and effective treatment choices for the future. J Complement Integr Med. 2015.

Basic, M., et al. Obesity: genome and environment interactions. Arh Hig Rada Toksikol. 2012;63(3):395-405.

Bernstein, R.K. Blood glucose self-monitoring by diabetic patients: refinements of procedural technique. Diabetes Care. 1979 Mar-Apr;2(2):233-6.

Bernstein, R.K. Virtually continuous euglycemia for 5 yr in a labile juvenile-onset diabetic patient under noninvasive closed-loop control. Diabetes Care. 1980 Jan-Feb;3(1):140-3.

Bipartisan Policy Center. Lots to lose how America’s health and obesity crisis threatens our economic future. Washington, D.C.: Bipartisan Policy Center,; 2012.

Boison, D. New insights into the mechanisms of the ketogenic diet. Curr Opin Neurol. 2017 Apr;30(2):187-192.

Bostock, E.C.S., et al. The Current Status of the Ketogenic Diet in Psychiatry. Front Psychiatry. 2017 Mar 20:8:43.

Bradford, B.L., et al. Mitochondrial Dysfunction and Type 2 Diabetes. Science. 2005 Jan 21;307(5708):384-7.

Brenton, J.N., et al. Phase II study of ketogenic diets in relapsing multiple sclerosis: safety, tolerability and potential clinical benefits. J Neurol Neurosurg Psychiatry. 2022 Jun;93(6):637-644.

Brenton, J.N., et al. Pilot study of a ketogenic diet in relapsing-remitting MS. Neurol Neuroimmunol Neuroinflamm. 2019 Apr 12;6(4):e565.

Carlson, J.A., et al. Dietary-related and physical activity-related predictors of obesity in children: a 2-year prospective study. Child Obes. 2012;8(2):110-5.

Centers for Disease Control and Prevention. All About Your A1C.

Centers for Disease Control and Prevention. Diabetes Tests.

Centers for Disease Control and Prevention. National Diabetes Statistics Report, 2020: Estimates of Diabetes and Its Burden in the United States.

Cheng, N., et al. Metabolic Dysfunction Underlying Autism Spectrum Disorder and Potential Treatment Approaches. Front Mol Neurosci. 2017 Feb 21:10:34.

Choquet, H., et al. Genomic insights into early-onset obesity. Genome Med. 2010;2(6):36.

Classen, J.B. Review of evidence that epidemics of type 1 diabetes and type 2 diabetes/metabolic syndrome are polar opposite responses to iatrogenic inflammation. Curr Diabetes Rev. 2012;8(6):413-8.

Coller, H.A. Is cancer a metabolic disease? Am J Pathol. 2014 Jan;184(1):4-17.

Cortese, S., et al. Attention-deficit/hyperactivity disorder, iron deficiency, and obesity: is there a link? Postgrad Med. 2014;126(4):155-70.

Cui, J., et al. Butyrate-Producing Bacteria and Insulin Homeostasis: The Microbiome and Insulin Longitudinal Evaluation Study (MILES). Diabetes. 2022 Nov 1;71(11):2438-2446.

Danan, K., et al. The Ketogenic Diet for Refractory Mental Illness: A Retrospective Analysis of 31 Inpatients. Front Psychiatry. 2022 Jul 6:13:951376.

deCampo, D.M., et al. Ketogenic dietary therapies for epilepsy and beyond. Curr Opin Clin Nutr Metab Care. 2019 Jul;22(4):264-268.

de la Rubia Orti, J.E., et al. Can Ketogenic Diet Improve Alzheimer’s Disease? Association With Anxiety, Depression, and Glutamate System. Front Nutr. 2021 Oct 27:8:744398.

de la Rubia Orti, J.E., et al. Exploring the impact of ketogenic diet on multiple sclerosis: obesity, anxiety, depression, and the glutamate system. Front Nutr. 2023 Aug 25:10:1227431.

Desai, J.R., et al. Diabetes and asthma case identification, validation, and representativeness when using electronic health data to construct registries for comparative effectiveness and epidemiologic research. Med Care. 2012;50 Suppl:S30-5.

Dietch, D.M., et al. Efficacy of low carbohydrate and ketogenic diets in treating mood and anxiety disorders: systematic review and implications for clinical practice. BJPsych Open. 2023 Apr 17;9(3):e70.

Duan, W., et al. Hyperglycemia, a neglected factor during cancer progression. Biomed Res Int. 2014:2014:461917.

Dyńka, D., et al. The Role of Ketogenic Diet in the Treatment of Neurological Diseases. Nutrients. 2022 Nov 24;14(23):5003.

Feinman, R.K., et al. Dietary carbohydrate restriction as the first approach in diabetes management: critical review and evidence base. Nutrition. 2015 Jan;31(1):1-13.

Giovannucci, E., et al. Diabetes and cancer: a consensus report. Diabetes Care. 2010 Jul;33(7):1674-85.

Gough, S., et al. Neuroprotection by the Ketogenic Diet: Evidence and Controversies. Front Nutr. 2021 Nov 23:8:782657.

Grammes, J., et al. Fear of hypoglycemia in patients with type 2 diabetes: The role of interoceptive accuracy and prior episodes of hypoglycemia. J Psychosom Res. 2018;105:58-63.

Grochowska, K., et al. The Effect of the Ketogenic Diet on the Therapy of Neurodegenerative Diseases and Its Impact on Improving Cognitive Functions. Dement Geriatr Cogn Dis Extra. 2022 May 30;12(2):100-106.

Han, H., et al. Gut Microbiota and Type 1 Diabetes. Int J Mol Sci. 2018 Mar 27;19(4):995.

Hao, J., et al. Mitochondrial nutrients improve immune dysfunction in the type 2 diabetic Goto-Kakizaki rats. J Cell Mol Med. 2009 Apr;13(4):701-11.

He, C., et al. Targeting gut microbiota as a possible therapy for diabetes. Nutr Res. 2015.

Hinzmann, R., et al. What do we need beyond hemoglobin A1c to get the complete picture of glycemia in people with diabetes? Int J Med Sci. 2012;9(8):665-81.

Irish, A.K., et al. Randomized control trial evaluation of a modified Paleolithic dietary intervention in the treatment of relapsing-remitting multiple sclerosis: a pilot study. Degener Neurol Neuromuscul Dis. 2017 Jan 4;7:1-18.

Jiang, Z., et al. Effects of Ketogenic Diet on Neuroinflammation in Neurodegenerative Diseases. Aging Dis. 2022 Jul 11;13(4):1146-1165.

Kamba, A., et al. Association between higher serum cortisol levels and decreased insulin secretion in a general populationPLOS ONE. 2016;11(11):e0166077.

Kapoor, D., et al. Emerging Role of the Ketogenic Dietary Therapies beyond Epilepsy in Child Neurology. Ann Indian Acad Neurol. 2021 Jul-Aug;24(4):470-480.

Karkafi, R.E., et al. Ketogenic Diet and Inflammation: Implications for Mood and Anxiety Disorders. Adv Exp Med Biol. 2023:1411:537-554.

Kogut, S.J., et al. Evaluation of a program to improve diabetes care through intensified care management activities and diabetes medication copayment reduction. J Manag Care Pharm. 2012;18(4):297-310.

Kong., A.P, et al. Diabetes and its comorbidities–where East meets West. Nat Rev Endocrinol. 2013;9(9):537-47.

Labbé, D.P., et al. Role of diet in prostate cancer: the epigenetic link. Oncogene. 2015 Sep 3;34(36):4683-91.

Laron, Z. Interplay between heredity and environment in the recent explosion of type 1 childhood diabetes mellitus. Am J Med Genet. 2002;115(1):4-7.

Lee, C., et al. Reduced levels of IGF-I mediate differential protection of normal and cancer cells in response to fasting and improve chemotherapeutic index. Cancer Res. 2010 Feb 15;70(4):1564-72.

Lee, R.W.Y., et al. A modified ketogenic gluten-free diet with MCT improves behavior in children with autism spectrum disorder. Physiol Behav. 2018 May 1:188:205-211.

Lefevre, F., et al. Ketogenic diet for the treatment of refractory epilepsy in children: A systematic review of efficacy. Pediatrics. 2000 Apr;105(4):E46.

Lennerz, B.S., et al. Management of Type 1 Diabetes With a Very Low-Carbohydrate Diet.
Pediatrics. 2018 Jun;141(6):e20173349.

Levi, J., et al. F as in fat how obesity threatens America’s future : 2012. Washington, D.C.: Trust for America’s Health; 2012.

Li, Q., et al. A Ketogenic Diet and the Treatment of Autism Spectrum Disorder. Front Pediatr. 2021 May 11:9:650624.

Liu, H., et al. Refined fructose and cancer. Expert Opin Ther Targets. 2011 Sep;15(9):1049-59.

Lorenzi, M., et al. High glucose induces DNA damage in cultured human endothelial cells. J Clin Invest. 1986 Jan;77(1):322-5.

Ly, N.P., et al. Gut microbiota, probiotics, and vitamin D: interrelated exposures influencing allergy, asthma, and obesity? J Allergy Clin Immunol. 2011;127(5):1087-94; quiz 95-6.

Magrone, T., et al. Childhood obesity: immune response and nutritional approaches. Front Immunol. 2015;6:76.

Mentzelou, M., et al. The Relationship of Ketogenic Diet with Neurodegenerative and Psychiatric Diseases: A Scoping Review from Basic Research to Clinical Practice. Nutrients. 2023 May 11;15(10):2270.

Mierau, S.B., et al. Metabolic interventions in Autism Spectrum Disorder. Neurobiol Dis. 2019 Dec:132:104544.

Mu, C., et al. Metabolic Framework for the Improvement of Autism Spectrum Disorders by a Modified Ketogenic Diet: A Pilot Study. J Proteome Res. 2020 Jan 3;19(1):382-390.

Noland, R.C., et al. Carnitine insufficiency caused by aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem. 2009 Aug 21;284(34):22840-52.

Olivito, I., et al. Ketogenic diet ameliorates autism spectrum disorders-like behaviors via reduced inflammatory factors and microbiota remodeling in BTBR T+ Itpr3tf/J mice. Exp Neurol. 2023 Aug:366:114432.

Onodera, Y., et al. Increased sugar uptake promotes oncogenesis via EPAC/RAP1 and O-GlcNAc pathways. J Clin Invest. 2014 Jan;124(1):367-84.

Operta, F.F., et al. The Ketogenic Diet for the Treatment of Mood Disorders in Comorbidity With Epilepsy in Children and Adolescents. Front Pharmacol. 2020 Nov 24:11:578396.

Pacal, L., et al. Parameters of oxidative stress, DNA damage and DNA repair in type 1 and type 2 diabetes mellitus. Arch Physiol Biochem. 2011;117(4):222-30.

Pietrzak, D., et al. The Therapeutic Role of Ketogenic Diet in Neurological Disorders. Nutrients. 2022 May 6;14(9):1952.

Power, R.A., et al. Carnitine revisited: potential use as adjunctive treatment in diabetes. Diabetologia. 2007 Apr;50(4):824-32.

Pulgaron, E.R. Childhood obesity: a review of increased risk for physical and psychological comorbidities. Clin Ther. 2013;35(1):A18-32.

Pulsifer, M.B., et al. Effects of ketogenic diet on development and behavior: preliminary report of a prospective study. Dev Med Child Neurol. 2001 May;43(5):301-6.

Rector, R.S., et al. Mitochondrial dysfunction precedes insulin resistance and hepatic steatosis and contributes to the natural history of non-alcoholic fatty liver disease in an obese rodent model. J Hepatol. 2010 May;52(5):727-36.

Renteria, I., et al. [Factors affecting oxidative damage in obese children: an exploratory study]. Nutr Hosp. 2015;31(4):1499-503.

Rosa, J.S., et al. Altered inflammatory, oxidative, and metabolic responses to exercise in pediatric obesity and type 1 diabetes. Pediatr Diabetes. 2011;12(5):464-72.

Salzer, H.M. Relative hypoglycemia as a cause of neuropsychiatric illness. J Natl Med Assoc. 1966;58(1):12-17.

Sanchez, A., et al. Role of sugars in human neutrophilic phagocytosis. Am J Clin Nutr. 1973 Nov;26(11):1180-4.

Sanchez, M., et al. Childhood obesity: a role for gut microbiota? Int J Environ Res Public Health. 2015;12(1):162-75. J Integr Complement Med. 2023 Feb;29(2):69-79.

Sanago, F., et al. Mind- and Body-Based Interventions Improve Glycemic Control in Patients with Type 2 Diabetes: A Systematic Review and Meta-Analysis. J Integr Complement Med. 2023 Feb;29(2):69-79.

Schwalfenberg, G.K. The Importance of Magnesium in Clinical Healthcare. Scientifica (Cairo). 2017:2017:4179326.

Seyfried, T.N. Cancer as a metabolic disease. Nutr Metab (Lond). 2010 Jan 27:7:7.

Seyfried, T.N. Cancer as a metabolic disease: implications for novel therapeutics. Carcinogenesis. 2014 Mar;35(3):515-27.

Seyfried, T.N., et al. Consideration of Ketogenic Metabolic Therapy as a Complementary or Alternative Approach for Managing Breast Cancer. Front Nutr. 2020 Mar 11:7:21.

Seyfried, T.N., et al. Is the restricted ketogenic diet a viable alternative to the standard of care for managing malignant brain cancer? Epilepsy Res. 2012 Jul;100(3):310-26.

Seyfried, T.N., et al. Metabolic management of brain cancer. Biochim Biophys Acta. 2011 Jun;1807(6):577-94.

Shen, W., et al. Protective effects of R-alpha-lipoic acid and acetyl-L-carnitine in MIN6 and isolated rat islet cells chronically exposed to oleic acid. J Cell Biochem. 2008 Jul 1;104(4):1232-43.

Shlomowitz, A., et al. Anxiety associated with self monitoring of capillary blood glucoseBritish Journal of Diabetes. 2014;14(2):60-63.

Smith, J., et al. Ketogenic diet restores aberrant cortical motor maps and excitation-to-inhibition imbalance in the BTBR mouse model of autism spectrum disorder. Behav Brain Res. 2016 May 1:304:67-70.

Smith, K.J., et al. Association of diabetes with anxiety: a systematic review and meta-analysisJ Psychosom Res. 2013;74(2):89-99.

Sreekumar, R., et al. Skeletal muscle mitochondrial dysfunction & diabetes. Indian J Med Res. 2007 Mar;125(3):399-410.

Stafstrom, C.E., et al. The ketogenic diet as a treatment paradigm for diverse neurological disorders. Front Pharmacol. 2012 Apr 9:3:59.

Tao, Y., et al. Ketogenic Diet: An Effective Treatment Approach for Neurodegenerative Diseases. Curr Neuropharmacol. 2022 Nov 15;20(12):2303-2319.

Tidman, M.M., et al. Effects of an low carbohydrate/healthy fat/ketogenic diet on biomarkers of health and symptoms, anxiety and depression in Parkinson's disease: a pilot study. Neurodegener Dis Manag. 2022 Apr;12(2):57-66.

Tinkum, K.L., et al. Fasting protects mice from lethal DNA damage by promoting small intestinal epithelial stem cell survival. Proc Natl Acad Sci U S A. 2015 Dec 22;112(51):E7148-54.

Tuomi, T., et al. The many faces of diabetes: a disease with increasing heterogeneity. Lancet. 2014;383(9922):1084-94.

Vaarala, O. Is the origin of type 1 diabetes in the gut? Immunol Cell Biol. 2012;90(3):271-6.

Vaarala, O. Gut microbiota and type 1 diabetes. Rev Diabet Stud. 2012;9(4):251-9.

Vargas, D.D., et al. Effectiveness of nutritional interventions on behavioral symptomatology of autism spectrum disorder: a systematic review. Nutr Hosp. 2022 Dec 20;39(6):1378-1388.

Verberne, A.J.M., et al. Adrenaline: insights into its metabolic roles in hypoglycaemia and diabetesBr J Pharmacol. 2016;173(9):1425-1437.

Witkowski, M., et al. The artificial sweetener erythritol and cardiovascular event risk. Nat Med. 2023 Mar;29(3):710-718.

Yuan, J., et al. Regular use of proton pump inhibitors and risk of type 2 diabetes: results from three prospective cohort studies. Gut. 28 Sep 2020.

Zheng, P., et al. Gut microbiome in type 1 diabetes: A comprehensive review. Diabetes Metab Res Rev. 2018 Oct;34(7):e3043.

Zhou, H., et al. Evaluating the Causal Role of Gut Microbiota in Type 1 Diabetes and Its Possible Pathogenic Mechanisms. Front Endocrinol (Lausanne). 2020 Mar 24:11:125.

Kryptopyrroluria (Pyrrole Disorder / Pyroluria)

Sources & References

Learning Disabilities

Sources & References

Adams, L.J., et al. Infant feeding method and special educational need in 191,745 Scottish schoolchildren: A national, population cohort study. PLoS Med. 2023 Apr 6;20(4):e1004191.

Baker, S.M. A biochemical approach to the problem of dyslexia. Journal of Learning Disabilities. 1985 18(10): 581-584.

Boat, T.F., et al. Prevalence of Learning Disabilities. Mental Disorders and Disabilities Among Low-Income Children. Washington (DC): National Academies Press (US); 2015 Oct 28. 16.

Borre, Y.E., et al. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med. 2014 Sep;20(9):509-18.

Darling, A.L., et al. Association between maternal vitamin D status in pregnancy and neurodevelopmental outcomes in childhood: results from the Avon Longitudinal Study of Parents and Children (ALSPAC). Br J Nutr 2017 Jun;117(12):1682-1692.

Edwards, E.S., et al. Dyslexia on a continuum: A complex network approach. PLOS ONE, 2018; 13 (12): e0208923.

Egset, K., et al. Magno App: Exploring Visual Processing in Adults with High and Low Reading Competence. Scandinavian Journal of Educational Research. 07 Jan 2020.

Hertz-Picciotto, I., et al. Organophosphate exposures during pregnancy and child neurodevelopment: Recommendations for essential policy reforms. PLoS Med. 2018 Oct 24;15(10):e1002671.

Mercer, J.S., et al. The Effects of Delayed Cord Clamping on 12-Month Brain Myelin Content and Neurodevelopment: A Randomized Controlled Trial. Am J Perinatol. 2022 Jan;39(1):37-44.

Morris, C.R., et al. Syndrome of allergy, apraxia, and malabsorption: characterization of a neurodevelopmental phenotype that responds to omega 3 and vitamin E supplementation. Alternative Therapies in Health and Medicine. Jul-Aug 2009;15(4):34-43.

Rifas-Shiman, S.L., et al. Associations of prenatal or infant exposure to acetaminophen or ibuprofen with mid-childhood executive function and behaviour. Paediatr Perinat Epidemiol. 2020 May;34(3):287-298.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Zablotsky, B., et al. Prevalence and Trends of Developmental Disabilities among Children in the United States: 2009-2017. Pediatrics. 2019 Oct;144(4):e20190811.

Zaigham, M., et al. Prelabour caesarean section and neurodevelopmental outcome at 4 and 12 months of age: an observational study. BMC Pregnancy and Childbirth. 2020 (20)564.

Lithium Orotate

Sources & References

Benson, D. Conversation with William Shaw, PhD-Integrative Medicine for Mental Health Conference. Integr Med (Encinitas). 2021 Jun;20(3):16-18.

Eugene, A.R., et al. Isolating the Norepinephrine Pathway Comparing Lithium in Bipolar Patients to SSRIs in Depressive Patients. Brain (Bacau). 2014 Dec;5(1-4):5-15.

Hamstra, S.I., et al. Beyond its Psychiatric Use: The Benefits of Low-dose Lithium Supplementation. Curr Neuropharmacol. 2023;21(4):891-910.

Karlović, D., et al. EIGHTY YEARS OF ELECTROCONVULSIVE THERAPY IN CROATIA AND IN SESTRE MILOSRDNICE UNIVERSITY HOSPITAL CENTRE. Acta Clin Croat. 2020 Sep;59(3):489-495.

Kuffler, D.P. Can lithium enhance the extent of axon regeneration and neurological recovery following peripheral nerve trauma? Neural Regen Res. 2022 May;17(5):948-952.

Leeds, P.R., et al. A new avenue for lithium: intervention in traumatic brain injury. ACS Chem Neurosci. 2014 Jun 18;5(6):422-33.

Lundberg, M., et al. Lithium and the Interplay Between Telomeres and Mitochondria in Bipolar Disorder. Front Psychiatry. 2020 Sep 29:11:586083.

Marshall, T. Lithium as a Nutrient. J Am Phys and Surg. 2015;20(4): 104-109.

Matsunaga, S., et al. Lithium as a Treatment for Alzheimer's Disease: A Systematic Review and Meta-Analysis. J Alzheimers Dis. 2015;48(2):403-10.

Memon, A., et al. Association between naturally occurring lithium in drinking water and suicide rates: systematic review and meta-analysis of ecological studies. Br J Psychiatry. 2020 Dec;217(6):667-678.

Mintz, M., et al. Revisiting Lithium: Utility for Behavioral Stabilization in Adolescents and Adults with Autism Spectrum Disorder. Psychopharmacol Bull. 2019 Jun 20;49(2):28-40.

Pacholko, A.G., et al. Lithium orotate: A superior option for lithium therapy? Brain Behav. 2021 Aug;11(8):e2262.

Palmos, A.B., et al. Lithium treatment and human hippocampal neurogenesis. Transl Psychiatry. 2021 Oct 30;11(1):555.

Rijal, S., et al. Lithium Enhances the GABAergic Synaptic Activities on the Hypothalamic Preoptic Area (hPOA) Neurons. Int J Mol Sci. 2021 Apr 9;22(8):3908.

Ruffalo, M.L. A Brief History of Lithium Treatment in Psychiatry. Prim Care Companion CNS Disord. 2017 Oct 12;19(5):17br02140.

Schou, M., et al. The treatment of manic psychoses by the administration of lithium salts. J Neurol Neurosurg Psychiatry. 1954 Nov;17(4):250-60.

Schrauzer, G.N., et al. Lithium in drinking water and the incidences of crimes, suicides, and arrests related to drug addictions. Biol Trace Elem Res. 1990 May;25(2):105-13.

Shorter, E. The history of lithium therapy. Bipolar Disord. 2009 Jun;11 Suppl 2(Suppl 2):4-9.

A Treatise on the Nature and Cure of Gout and Rheumatism, Including General Considerations on Morbid States of the Digestive Organs; Some Remarks on Regimen; and Practical Observations on Gravel. Med Chir J. 1820 Jan 1;2(7):367–92.

Young, W. Review of lithium effects on brain and blood. Cell Transplant. 2009;18(9):951-75.

Low-Glutamate Diet

Sources & References

Bauer, J., et al. Hyperactivity and impulsivity in adult attention-deficit/hyperactivity disorder is related to glutamatergic dysfunction in the anterior cingulate cortex. World J Biol Psychiatry. 2018 Oct;19(7):538-546.

Blaylock, R.L., et al. Natural plant products and extracts that reduce immunoexcitotoxicity-associated neurodegeneration and promote repair within the central nervous system. Surg Neurol Int. 2012;3:19

Blaylock, R.L. A possible central mechanism in autism spectrum disorders, part 1. Altern Ther Health Med. 2008 Nov-Dec;14(6):46-53.

Blaylock, R.L. A possible central mechanism in autism spectrum disorders, part 2: immunoexcitotoxicity. Altern Ther Health Med. 2009 Jan-Feb;15(1):60-7.

Blaylock, R.L. A possible central mechanism in autism spectrum disorders, part 3: the role of excitotoxin food additives and the synergistic effects of other environmental toxins. Altern Ther Health Med. 2009 Mar-Apr;15(2):56-60.

Blaylock, R.L., et al. Immune-glutamatergic dysfunction as a central mechanism of the autism spectrum disorders. Curr Med Chem. 2009;16(2):157-70.

Bravo, J.A., et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A. 2011 Sep 20;108(38):16050-5.

Breitenkamp, A.F., et al. Voltage-gated Calcium Channels and Autism Spectrum Disorders. Curr Mol Pharmacol. 2015;8(2):123-32.

Brown, M.S., et al. Increased glutamate concentration in the auditory cortex of persons with autism and first-degreer elatives: A(1)H-MR study. Autism Res. 2013;6(1):1–10.

Cartmell, J., et al. Regulation of neurotransmitter release by metabotropic glutamate receptors. J Neurochem. 2000 Sep;75(3):889-907.

Chapman, G. Glutamate and epilepsy. J Nutr. 2000 Apr;130(4S Suppl):1043S-5S.

Choudhury, P.R., et al. Glutamate mediated signaling in the pathophysiology of autism spectrum disorders. Pharmacol Biochem Behav 100(2012)841–849

Cocito, L., et al. GABA and phosphatidylserine in human photosensitivity: a pilot study. Epilepsy Res. 1994 Jan;17(1):49-53.

Coghlan, S., et al. GABA System Dysfunction in Autism and Related Disorders: From Synapse to Symptoms. Neurosci Biobehav Rev. 2012;36(9):2044–2055.

Davalli, D.M., et al. The potential role of glutamate in the current diabetes epidemic. Acta Diabetol. 2012 Jun;49(3):167-83.

DeHavenon, A., et al. The Secret “Spice”: An Undetectable Toxic Cause of Seizure. Neurohospitalist. 2011 Oct; 1(4): 182–186.

Ding, H., et al. Molecular Pathogenesis of Anti-NMDAR Encephalitis. Biomed Res Int. 2015;2015:643409.

D’Souza, C.E., et al. GAD65 antibody-associated autoimmune epilepsy with unique independent bitemporal-onset ictal asystole. Epileptic Disord. 2018 Jun 1;20(3):204-208.

Eimerl, S., et al. Acute glutamate toxicity and its potentiation by serum albumin are determined by the Ca2+ concentration. Neurosci Lett. 130 (1991) 125–127.

El-Ansary, A. GABA and Glutamate Imbalance in Autism and Their Reversal as Novel Hypothesis for Effective Treatment Strategy. Autism and Developmental Disorders. 18, no. 3 (2020): 46-63.

Ende, G., et al. Impulsivity and Aggression in Female BPD and ADHD Patients: Association with ACC Glutamate and GABA Concentrations. Neuropsychopharmacology. 2016 Jan;41(2):410-8.

Essa, M.M., et al. Excitotoxicity in the pathogenesis of autism. Neurotox Res. 2013 May;23(4):393-400.

Feng, Z.M., et al. Monosodium L-Glutamate and Dietary Fat Differently Modify the Composition of the Intestinal Microbiota in Growing Pigs. Obes Facts. 2015;8(2):87-100.

Fernell, E. Further studies of GABA and Glutamate imbalances in autism are important challenges for future research. Acta Paediatr. 2019 Feb;108(2):200-201.

Ford, T.C., et al. Psychosocial deficits across autism and schizotypal spectra are interactively modulated by excitatory and inhibitory neurotransmission. Autism. 2019 Jul 24:1362361319866030.

Gilliam, L.K., et al. Multiplicity of the antibody response to GAD65 in Type I diabetes. Clin Exp Immunol. 2004;138(2):337–341.

Hacohen, Y., et al. N‐methyl‐d‐aspartate (NMDA) receptor antibodies encephalitis mimicking an autistic regression. Dev Med Child Neurol. 2016 Oct;58(10):1092-4.

Haroon, E., et al. Inflammation, Glutamate, and Glia: A Trio of Trouble in Mood Disorders. Neuropsychopharmacology. 2017 Jan;42(1):193-215.

Hassel, B., et al. Brain infection with Staphylococcus aureus leads to high extracellular levels of glutamate, aspartate, γ-aminobutyric acid, and zinc. J Neurosci Res. 2014 Dec;92(12):1792-800.

Herbert, M.R, et al. Autism and EMF? Plausibility of a pathophysiological link–Part I. Pathophysiology 20.3 (2013): 191-209.

Herbert, M.R., et al. Autism and EMF? Plausibility of a pathophysiological link Part II. Pathophysiology 20.3 (2013): 211-234.

Huang, Y., et al. Pyridoxine Supplementation Improves the Activity of Recombinant Glutamate Decarboxylase and the Enzymatic Production of Gama-Aminobutyric Acid. PLoS One. 2016 Jul 20;11(7):e0157466.

Hughes, R., et al. Protein degradation in the large intestine: relevance to colorectal cancer. Curr Issues Intest Microbiol. 2000 Sep;1(2):51-8.

Humphries, P., et al. Direct and indirect cellular effects of aspartame on the brain. Eur J Clin Nutr. 2008 Apr;62(4):451-62.

Kash, S.F., et al. Epilepsy in mice deficient in the 65-kDa isoform of glutamic acid decarboxylase. Proc Natl Acad Sci U S A. 1997;94(25):14060–14065.

Khalifa, D., et al. Serum glutamate was elevated in children aged 3-10 years with autism spectrum disorders when they were compared with controls. Acta Paediatr. 2019 Feb;108(2):295-299.

Luo, P., et al. The role of glutamate receptors in traumatic brain injury: implications for postsynaptic density in pathophysiology. Brain Res Bull. 2011 Jul 15;85(6):313-20.

Magalhães, J.Z., et al. Prenatal exposure to fipronil disturbs maternal aggressive behavior in rats. Neurotoxicol Teratol. Nov-Dec 2015;52(Pt A):11-6.

Manev, H., et al. Delayed increase of Ca2+ influx elicited by glutamate: role in neuronal death. Mol Pharmacol. 1989 Jul;36(1):106-12.

McNally, L., et al. Inflammation, glutamate, and glia in depression: a literature review. CNS Spectr. 2008 Jun;13(6):501-10.

Möhler, H. The GABA system in anxiety and depression and its therapeutic potential. Neuropharmacology. 2012 Jan;62(1):42-53.

Naaijen, J., et al. Fronto-Striatal Glutamate in Autism Spectrum Disorder and Obsessive Compulsive Disorder. Neuropsychopharmacology. 2017 Nov;42(12):2456-2465.

Napolini, V., et al. The mitochondrial aspartate/glutamate carrier AGC1 and calcium homeostasis: Physiological links and abnormalities in autism. Mol Neurobiol. 44 (2011) 83–92.

Nuss, P. Anxiety disorders and GABA neurotransmission: a disturbance of modulation. Neuropsychiatr Dis Treat. 2015; 11: 165–175.

Onaolapo, A.Y., et al. Glutamate and depression: Reflecting a deepening knowledge of the gut and brain effects of a ubiquitous molecule. World J Psychiatry. 2021 Jul 19;11(7):297-315.

Ou, D., et al. Cross-reactive rubella virus and glutamic acid decarboxylase (65 and 67) protein determinants recognised by T cells of patients with Type I diabetes mellitus. Diabetologia. 2000 Jun;43(6):750-62.

Pall, M.L. The Autism Epidemic Is Caused by EMFs, Acting via Calcium Channels and Chemicals Acting via NMDA-Rs: Downstream Effects Cause Autism, Autism One, Chicago, Illinois, USA, 2015.

Pall, M.L., Electromagnetic fields act via activation of voltage‐gated calcium channels to produce beneficial or adverse effects. Journal of Cellular and Molecular Medicine 17.8 (2013): 958-965.

Palmieri, L., et al. Altered calcium homeostasis in autism-spectrum disorders: evidence from biochemical and genetic studies of the mitochondrial aspartate/glutamate carrier AGC. Mol Psychiatry. 2010 Jan;15(1):38-52.

Pelsser, L.M., et al. Effects of a restricted elimination diet on the behaviour of children with attention-deficit hyperactivity disorder (INCA study): a randomised controlled trial. Lancet. 2011 Feb 5;377(9764):494-503.

Pitt, D., et al. Glutamate excitotoxicity in a model of multiple sclerosis. Nat Med. 2000 Jan;6(1):67-70.

Qi, J., et al. Enhanced susceptibility to stress and seizures in GAD65 deficient mice. PLoS One. 2018 Jan 29;13(1):e0191794.

Rojas, D.C. The role of glutamate and its receptors in autism and the use of glutamate receptor antagonists in treatment. J Neural Transm (Vienna). 2014;121(8):891–905.

Rout, U.K., et al. Presence of GAD65 autoantibodies in the serum of children with autism or ADHD. Eur Child Adolesc Psychiatry. 2012 Mar;21(3):141-7.

Rowley, N.M., et al. Glutamate and GABA synthesis, release, transport and metabolism as targets for seizure control. Neurochem Int. 2012 Sep;61(4):546-58.

Sai, Y., et al. Clinical diagnosis and treatment of pediatric anti-N-methyl-D-aspartate receptor encephalitis: A single center retrospective study. Exp Ther Med. 2018 Aug;16(2):1442-1448.

Samuels, A. The toxicity/safety of processed free glutamic acid (MSG): a study in suppression of information. Account Res. 1999;6(4):259-310.

Schauwecker, P.A. The effects of glycemic control on seizures and seizure-induced excitotoxic cell death. BMC Neurosci. 2012 Aug 6;13:94.

Silvestrin, R.B., et al. Animal model of autism induced by prenatal exposure to valproate: altered glutamate metabolism in the hippocampus. BrainRes. 1495 (2013) 52–60.

Takano, T., et al. Glutamate release promotes growth of malignant gliomas. Nat Med. 2001 Sep;7(9):1010-5.

Tang, J., et al. Exposure to 900 MHz electromagnetic fields activates the mkp-1/ERK pathway and causes blood-brain barrier damage and cognitive impairment in rats. Brain Res. 2015 Mar 19;1601:92-101.

Terpstra, M., et al. Changes in human brain glutamate concentration during hypoglycemia: insights into cerebral adaptations in hypoglycemia-associated autonomic failure in type 1 diabetes. J Cereb Blood Flow Metab. 2014;34(5):876–882.

Tzang, R.F., et al. Autism Associated With Anti-NMDAR Encephalitis: Glutamate-Related Therapy. Front Psychiatry. 2019 Jun 21;10:440.

Walls, A.B., et al. GAD65 is essential for synthesis of GABA destined for tonic inhibition regulating epileptiform activity. J Neurochem. 2010 Dec;115(6):1398-408.

Wang, Hsiuying. Anti-NMDA Receptor Encephalitis. Int J Mol Sci. 2017 Jan 18;18(1). pii: E193.

Watkins, J.C., et al. The glutamate story. Br J Pharmacol. 2006 Jan; 147(Suppl 1): S100–S108.

Xu, C.L., et al. Anti-N-methyl-D-aspartate receptor encephalitis with serum anti-thyroid antibodies and IgM antibodies against Epstein-Barr virus viral capsid antigen: a case report and one year follow-up. BMC Neurol. 2011 Nov 29;11:149.

Yang, Y., et al. Role of metabotropic glutamate receptor 7 in autism spectrum disorders: A pilot study. Life Sci. 92 (2013) 149–153.

Zhang, Z., et al. Blood Glutamate Levels in Autism Spectrum Disorder: A Systematic Review and Meta-Analysis. PLoS One. 2016 Jul 8;11(7):e0158688.

Low-Histamine Diet

Sources & References

Afrin, L.B., et al. Often seen, rarely recognized: mast cell activation disease–a guide to diagnosis and therapeutic options. Ann Med. 2016;48(3):190–201. doi:10.3109/07853890.2016.1161231.

Afrin, L.B., et al. Mast cell activation disease: An underappreciated cause of neurologic and psychiatric symptoms and diseases. Brain Behav Immun. 2015;50:314–321. doi:10.1016/j.bbi.2015.07.002.

Campbell, et al. Mechanisms of Allergic Disease – Environmental and genetic determinants for the development of allergy. Clin Exp Allergy. 2015

Della Giustina, A., et al.. Vitamin D, allergies and asthma: focus on pediatric patients. World Allergy Organ J. 2014;7(1):27

Feehley, T., et al. Healthy infants harbor intestinal bacteria that protect against food allergy. Nature Medicine. 2019 Jan 14.

Frieri, M. Mast Cell Activation Syndrome. Clin Rev Allergy Immunol. 2018;54(3):353–365. doi:10.1007/s12016-015-8487-6.

Frieri, M., et al. Mast cell activation syndrome: a review. Curr Allergy Asthma Rep. 2013;13(1):27–32. doi:10.1007/s11882-012-0322-z.

Gupta, R.S., et al. The Public Health Impact of Parent-Reported Childhood Food Allergies in the United States. Pediatrics. 2018 Dec;142(6). pii: e20181235.

Hamilton, M.J. Nonclonal Mast Cell Activation Syndrome: A Growing Body of Evidence. Immunol Allergy Clin North Am. 2018;38(3):469–481. doi:10.1016/j.iac.2018.04.002.

Ly, N.P., et al. Gut microbiota, probiotics, and vitamin D: interrelated exposures influencing allergy, asthma, and obesity? J Allergy Clin Immunol. 2011;127(5):1087-94; quiz 95-6

Maksimova, O.V., et al. [Intestine microbiota and allergic diseases]. Zh Mikrobiol Epidemiol Immunobiol. 2014(3):49-60.

Naganuma, F., et al. Histamine N-methyltransferase regulates aggression and the sleep-wake cycle. Sci Rep. 2017 Nov 21;7(1):15899.

Petra, A.I., et al. Spectrum of mast cell activation disorders. Expert Rev Clin Immunol. 2014;10(6):729–739. doi:10.1586/1744666X.2014.906302

Prescott, S.L. Early-life environmental determinants of allergic diseases and the wider pandemic of inflammatory noncommunicable diseases. J Allergy Clin Immunol. 2013;131(1):23-30.

Sánchez-Pérez, S. et al. Intestinal Dysbiosis in Patients with Histamine Intolerance.
Nutrients. 2022 Apr 23;14(9):1774.

Sánchez-Pérez, S. et al. The dietary treatment of histamine intolerance reduces the abundance of some histamine-secreting bacteria of the gut microbiota in histamine intolerant women. A pilot study. Front Nutr. 2022 Oct 21;9:1018463.

Schink, M., et al. Microbial patterns in patients with histamine intolerance. J Physiol Pharmacol. 2018 Aug;69(4).

Schnedl, W.J., et al. Histamine Intolerance Originates in the Gut. Nutrients. 2021 Apr 12;13(4):1262.

Son, J.H., et al. A Histamine-Free Diet Is Helpful for Treatment of Adult Patients with Chronic Spontaneous Urticaria. Ann Dermatol. 2018 Apr;30(2):164-172.

Terwee, C.B. Succesful treatment of food allergy with Nambudripad's Allergy Elimination Techniques (NAET) in a 3-year old: A case report. Cases J. 2008 Sep 19;1(1):166.

Tsabouri, S., et al. Modulation of gut microbiota downregulates the development of food allergy in infancy. Allergol Immunopathol (Madr). 2014;42(1):69-77.

Vassilopoulou, E., et al. The Impact of Food Histamine Intake on Asthma Activity: A Pilot Study. Nutrients. 2020 Nov 5;12(11):3402.

Yang, M.T., et al. Attention-deficit/hyperactivity disorder-related symptoms improved with allergic rhinitis treatment in children. Am J Rhinol Allergy. 2016 May;30(3):209-14.

Low-Oxolate Diet

Sources & References

Konstantynowicz, J., et al. A Potential Pathogenic Role of Oxalate in Autism. Eur. J. Paediatr. Neurol. 2012;16:485–491.

Stewart, C.S. Oxalobacter formigenes and its role in oxalate metabolism in the human gut. FEMS Microbiol Lett. 2004 Jan 15;230(1):1-7.

Lyme Disease

Sources & References

Bransfield, R.C., et al. The association between tick-borne infections, Lyme borreliosis and autism spectrum disorders. Medical Hypotheses. 2008;70(5):967-74.

Brorson, O., et al. Grapefruit seed extract is a powerful in vitro agent against motile and cystic forms of Borrelia burgdorferi sensu lato. Infection. 2007 Jun;35(3):206-8.

Cantorna, M.T., et al. 1,25-Dihydroxycholecalciferol inhibits the progression of arthritis in murine models of human arthritis. J Nutr. 1998 Jan;128(1):68-72.

Cantorna, M.T., et al. Vitamin A deficiency exacerbates murine Lyme arthritis. J Infect Dis. 1996 Oct;174(4):747-51.

Feng, J., et al. Evaluation of Natural and Botanical Medicines for Activity Against Growing and Non-growing Forms of B. burgdorferi. Front. Med., 21 Feb 2020.

Feng, J., et al. Identification of Essential Oils with Strong Activity against Stationary Phase Borrelia burgdorferi. Antibiotics (Basel). 2018 Oct 16;7(4):89.

Feng, J., et al. Selective Essential Oils from Spice or Culinary Herbs Have High Activity against Stationary Phase and Biofilm Borrelia burgdorferi. Front Med (Lausanne). 2017 Oct 11;4:169.

Ferreira Maya, M., et al. Plant-based insect repellents: a review of their efficacy, development and testing. Malar J. 2011; 10(Suppl 1): S11. Published online 2011 Mar 15.

Hutschenreuther, A., et al. Growth inhibiting activity of volatile oil from Cistus creticus L. against Borrelia burgdorferi s.s. in vitro. Pharmazie. 2010 Apr;65(4):290-5.

Javid, A., et al. Hyperglycemia Impairs Neutrophil-Mediated Bacterial Clearance in Mice Infected with the Lyme Disease Pathogen. PLoS One. 2016 Jun 24;11(6):e0158019.

Kepka, A., et al. Serum carnitine concentration is decreased in patients with Lyme borreliosis. Postepy Hig Med Dosw (Online). 2016 Mar 4;70:180-5.

Lovegrove, M.C., et al. US Emergency Department Visits for Adverse Drug Events From Antibiotics in Children, 2011–2015. Journal of the Pediatric Infectious Diseases Society, piy066. 2018 Aug 23.
Lubke, L.L., et al. The antimicrobial agent melittin exhibits powerful in vitro inhibitory effects on the Lyme disease spirochete. Clin Infect Dis. 1997 Jul;25 Suppl 1:S48-51.

Socarra, K.M., et al. Antimicrobial Activity of Bee Venom and Melittin against Borrelia burgdorferi. Antibiotics (Basel). 2017 Nov 29;6(4). pii: E31.

Theophilus, P.A., et al. Effectiveness of Stevia Rebaudiana Whole Leaf Extract Against the Various Morphological Forms of Borrelia Burgdorferi in Vitro. Eur J Microbiol Immunol (Bp). 2015 Nov 12;5(4):268-80.

Troxell, B., et al. Manganese and zinc regulate virulence determinants in Borrelia burgdorferi. Infect Immun. 2013 Aug;81(8):2743-52.

Magnesium

Sources & References

Arab, A., et al. The Role of Magnesium in Sleep Health: a Systematic Review of Available Literature. Biol Trace Elem Res. 2023 Jan;201(1):121-128.

Boyle, N.B., et al. The Effects of Magnesium Supplementation on Subjective Anxiety and Stress-A Systematic Review. Nutrients. 2017 Apr 26;9(5):429.

Młyniec, K., et al. Essential elements in depression and anxiety. Part I. Pharmacol Rep. 2014 Aug;66(4):534-44.

Sartori, S.B., et al. Magnesium deficiency induces anxiety and HPA axis dysregulation: modulation by therapeutic drug treatment. Neuropharmacology. 2012 Jan;62(1):304-12.

Schwalfenberg, G.K. The Importance of Magnesium in Clinical Healthcare. Scientifica (Cairo). 2017:2017:4179326.

Mast Cell Activation Syndrome (MCAS)

Sources & References

Afrin, L.B., et al. Often seen, rarely recognized: mast cell activation disease–a guide to diagnosis and therapeutic options. Ann Med. 2016;48(3):190–201. doi:10.3109/07853890.2016.1161231.

Afrin, L.B., et al. Mast cell activation disease: An underappreciated cause of neurologic and psychiatric symptoms and diseases. Brain Behav Immun. 2015;50:314–321. doi:10.1016/j.bbi.2015.07.002.

Campbell, et al. Mechanisms of Allergic Disease – Environmental and genetic determinants for the development of allergy. Clin Exp Allergy. 2015

Della Giustina, A., et al.. Vitamin D, allergies and asthma: focus on pediatric patients. World Allergy Organ J. 2014;7(1):27

Feehley, T., et al. Healthy infants harbor intestinal bacteria that protect against food allergy. Nature Medicine. 2019 Jan 14.

Frieri, M. Mast Cell Activation Syndrome. Clin Rev Allergy Immunol. 2018;54(3):353–365. doi:10.1007/s12016-015-8487-6.

Frieri, M., et al. Mast cell activation syndrome: a review. Curr Allergy Asthma Rep. 2013;13(1):27–32. doi:10.1007/s11882-012-0322-z.

Gupta, R.S., et al. The Public Health Impact of Parent-Reported Childhood Food Allergies in the United States. Pediatrics. 2018 Dec;142(6). pii: e20181235.

Hamilton, M.J. Nonclonal Mast Cell Activation Syndrome: A Growing Body of Evidence. Immunol Allergy Clin North Am. 2018;38(3):469–481. doi:10.1016/j.iac.2018.04.002.

Ly, N.P., et al. Gut microbiota, probiotics, and vitamin D: interrelated exposures influencing allergy, asthma, and obesity? J Allergy Clin Immunol. 2011;127(5):1087-94; quiz 95-6

Maksimova, O.V., et al. [Intestine microbiota and allergic diseases]. Zh Mikrobiol Epidemiol Immunobiol. 2014(3):49-60.

Petra, A.I., et al. Spectrum of mast cell activation disorders. Expert Rev Clin Immunol. 2014;10(6):729–739. doi:10.1586/1744666X.2014.906302

Prescott, S.L. Early-life environmental determinants of allergic diseases and the wider pandemic of inflammatory noncommunicable diseases. J Allergy Clin Immunol. 2013;131(1):23-30.

Sánchez-Pérez, S. et al. Intestinal Dysbiosis in Patients with Histamine Intolerance.
Nutrients. 2022 Apr 23;14(9):1774.

Sánchez-Pérez, S. et al. The dietary treatment of histamine intolerance reduces the abundance of some histamine-secreting bacteria of the gut microbiota in histamine intolerant women. A pilot study. Front Nutr. 2022 Oct 21;9:1018463.

Schink, M., et al. Microbial patterns in patients with histamine intolerance. J Physiol Pharmacol. 2018 Aug;69(4).

Schnedl, W.J., et al. Histamine Intolerance Originates in the Gut. Nutrients. 2021 Apr 12;13(4):1262.

Son, J.H., et al. A Histamine-Free Diet Is Helpful for Treatment of Adult Patients with Chronic Spontaneous Urticaria. Ann Dermatol. 2018 Apr;30(2):164-172.

Terwee, C.B. Succesful treatment of food allergy with Nambudripad's Allergy Elimination Techniques (NAET) in a 3-year old: A case report. Cases J. 2008 Sep 19;1(1):166.

Tsabouri, S., et al. Modulation of gut microbiota downregulates the development of food allergy in infancy. Allergol Immunopathol (Madr). 2014;42(1):69-77.

Vassilopoulou, E., et al. The Impact of Food Histamine Intake on Asthma Activity: A Pilot Study. Nutrients. 2020 Nov 5;12(11):3402.

Yang, M.T., et al. Attention-deficit/hyperactivity disorder-related symptoms improved with allergic rhinitis treatment in children. Am J Rhinol Allergy. 2016 May;30(3):209-14.

Microbiome

Sources & References

Adams, J.B., et al. Gastrointestinal Flora and Gastrointestinal Status in Children with Autism—Comparisons to Typical Children and Correlation with Autism Severity. BMC Gastroenterol. 2011;11:22.

Aguilera, M., et al. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice. Gut Microbes. 2015;6(1):10-23.

Appleton, J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr Med (Encinitas). 2018 Aug;17(4):28-32.

Aroniadis, O.C., et al. Fecal microbiota transplantation: past, present and future. Curr Opin Gastroenterol. 2013;29(1):79-84.

Assa, A., et al. Vitamin D deficiency promotes epithelial barrier dysfunction and intestinal inflammation. J Infect Dis. 2014;210(8):1296-305.

Atladóttir, H.Ó., et al. Autism after infection, febrile episodes, and antibiotic use during pregnancy: an exploratory study. Pediatrics. 2012 Dec;130(6):e1447-54.

Aversa, Z., et al. Association of Infant Antibiotic Exposure With Childhood Health Outcomes. Mayo Clin Proc. 2020 Nov 6;S0025-6196(20)30785-0.

Bäckhed, F., et al. Host-Bacterial Mutualism in the Human Intestine. Science. 2005 Mar 25;307(5717):1915-20.

Bennings, M.A., et al. Colonic transit times and behaviour profiles in children with defecation disorders. Archives of the Diseases of Childhood. 2004 Jan;89(1):13-6.

Berding, K., et al. Diet Can Impact Microbiota Composition in Children With Autism Spectrum Disorder. Front. Neurosci. 2018;12:515.

Bjørklund, G., et al. Gastrointestinal alterations in autism spectrum disorder: What do we know? Neurosci Biobehav Rev. 2020 Nov:118:111-120.

Bora, S.A., et al. Regulation of vitamin D metabolism following disruption of the microbiota using broad spectrum antibiotics. J Nutr Biochem. 2018 Jun;56:65-73.

Borchers, A.T., et al. Probiotics and immunity. Journal of Gastroenterology. 44 (2009): 26-46.

Borre, Y.E., et al. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med. 2014 Sep;20(9):509-18.

Buccigrossi, V., et al. Functions of intestinal microflora in children. Curr Opin Gastroenterol. 2013;29(1):31-8.

Buie, T., et al. Evaluation, diagnosis, and treatment of gastrointestinal disorders in individuals with ASDs: a consensus report. Pediatrics. 2010 Jan;125 Suppl 1:S1-18.

Camilleri, M. Serotonin in the gastrointestinal tract. Curr Opin Endrocrinol Diabetes Obes. 2009 Feb;16(1):53-9.

Carding, S., et al. Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis. 2015;26:26191.

Chen, C.Q., et al. Distribution, function and physiological role of melatonin in the lower gut. World J Gastroenterol. 2011 Sep 14;17(34):3888-98.

Chen, Y., et al. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients. 2021 Jun 19;13(6):2099.

Clark, J.A., et al. Intestinal crosstalk—a new paradigm for understanding the gut as the ‘motor’ of critical illness. Shock. 2007 Oct;28(4):384-93.

Cohen-Mekelburg, S., et al. Morning light treatment for inflammatory bowel disease: a clinical trial. BMC Gastroenterol. 2024 May 22;24(1):179.

Correale, J., et al. The role of the gut microbiota in multiple sclerosis. Nat Rev Neurol. 2022 Sep;18(9):544-558.

Critchfield, et al. The potential role of probiotics in the management of childhood autism spectrum disorders. Gastroenterol Res Pract. 2011;2011:161358.

Cucchiara, S., et al. Interactions between intestinal microbiota and innate immune system in pediatric inflammatory bowel disease. J Clin Gastroenterol. 2012;46 Suppl:S64-6.

Cui, J., et al. Butyrate-Producing Bacteria and Insulin Homeostasis: The Microbiome and Insulin Longitudinal Evaluation Study (MILES). Diabetes. 2022 Nov 1;71(11):2438-2446.

de Goffau, et al. Fecal microbiota composition differs between children with beta-cell autoimmunity and those without. Diabetes. 2013;62(4):1238-44.

D’Eufemia, P., et al. Abnormal intestinal permeability in children with autism. Acta Paediatr. 1996 Sep;85(9):1076-9.

de Magistris, et al. Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J Pediatr Gastroenterol Nutr. 2010;51(4):418-24.

Derrien, M., et al. The Gut Microbiota in the First Decade of Life. Trends Microbiol. 2019 Dec;27(12):997-1010.

Dinan, T.G., et al. The Microbiome-Gut-Brain Axis in Health and Disease. Gastroenterol Clin North Am. 2017 Mar;46(1):77-89.

Dogra, S.K., et al. Gut Microbiota Resilience: Definition, Link to Health and Strategies for Intervention. Front Microbiol. 2020 Sep 15:11:572921.

Edwards, C.A., et al. Intestinal flora during the first months of life: new perspectives. British Journal of Nutrition. 88 (2002): S11-S18.

Erickson, C.A., et al. Gastrointestinal Factors in Autistic Disorder: A Critical Review. Journal of Autism and Developmental Disorders. 2005 Dec;35(6):713-27.

Fattorusso, A., et al. Autism Spectrum Disorders and the Gut Microbiota. Nutrients. 2019 Feb 28;11(3):521.

Feng, P., et al. A review of probiotics in the treatment of autism spectrum disorders: Perspectives from the gut-brain axis. Front Microbiol. 2023 Mar 16:14:1123462.

Fukuda, K., et al. Determination of the discriminant score of intestinal microbiota as a biomarker of disease activity in patients with ulcerative colitis. BMC Gastroenterol. 2014;14:49.

Galland, L. The gut microbiome and the brain. J Med Food 2014; 17(12): 1261-72.

Ganal-Vonarburg, S.C., et al. Microbial-host molecular exchange and its functional consequences in early mammalian life. Science. 2020 May 8;368(6491):604-607.

Gastrointestinal permeability in food-allergic children. Nutr Rev. 1985 Aug;43(8):233-5.

Grizotte-Lake, M., et al. Commensals Suppress Intestinal Epithelial Cell Retinoic Acid Synthesis to Regulate Interleukin-22 Activity and Prevent Microbial Dysbiosis. Immunity. 2018 Dec 18;49(6):1103-1115.e6.

Guandalini, S. Are probiotics or prebiotics useful in pediatric irritable bowel syndrome or inflammatory bowel disease? Front Med (Lausanne). 2014;1:23.

Guandalini, S., et al. Prebiotics and probiotics in irritable bowel syndrome and inflammatory bowel disease in children. Benef Microbes. 2015;6(2):209-17.

Hamad, A.F., et al. Prenatal antibiotics exposure and the risk of autism spectrum disorders: A population-based cohort study. PLoS One. 2019 Aug 29;14(8):e0221921.

Han, H., et al. Gut Microbiota and Type 1 Diabetes. Int J Mol Sci. 2018 Mar 27;19(4):995.

Hanaway, P. Balance of Flora, GALT, and Mucosal Integrity. Alternative Therapies in Health and Medicine. Sep-Oct 2006;12(5):52-60; quiz 61-2.

Hejitz, R.D., et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011 Feb 15;108(7):3047-52.

Horn, J., et al. Role of Diet and Its Effects on the Gut Microbiome in the Pathophysiology of Mental Disorders. Transl. Psychiatry. 2022;12:164.

Horvath, K., et al. Autism and gastrointestinal symptoms. Curr Gastroenterol Rep. 2002 Jun;4(3):251-8.

Horvath, K., et al. Autistic disorder and gastrointestinal disease. Current Opinion in Pediatrics. 2002 Oct;14(5):583-7.

Horvath, K., et al. Gastrointestinal abnormalities in children with autistic disorder. Journal of Pediatrics. 1999 Nov;135(5):559-63.

Hrncir, T., et al. Gut microbiota and lipopolysaccharide content of the diet influence development of regulatory T cells: studies in germ-free mice. BMC Immunology. 9 (2008): 65.

Hyman, M.A. Is the Cure for Brain Disorders Outside the Brain? Alternative Therapies in Health and Medicine. Nov-Dec 2007;13(6):10-5.

Isaksson, J., et al. Brief Report: Association Between Autism Spectrum Disorder, Gastrointestinal Problems and Perinatal Risk Factors Within Sibling Pairs. J Autism Dev Disord. 2017 Aug;47(8):2621-2627.

Jackson, P.G., et al. Intestinal permeability in patients with eczema and food allergy. Lancet. 1981 Jun 13;1(8233):1285-6.

Jakobsen, C., et al. Environmental factors and risk of developing paediatric inflammatory bowel disease — a population based study. 2007-2009. J Crohns Colitis. 2013;7(1):79-88.

Jyonouchi, H., et al. Dysregulated innate immune responses in young children with autism spectrum disorders: their relationship to gastrointestinal symptoms and dietary intervention. Neuropsychobiology. 2005;51(2):77-85.

Jyonouchi, H., et al. Evaluation of an Association between Gastrointestinal Symptoms and Cytokine Production against Common Dietary Proteins in Children with Autism Spectrum Disorders. J. Pediatr. 2005;146:605–610.

Kang, D.W., et al. Long-term benefit of Microbiota Transfer Therapy on autism symptoms and gut microbiota. Scientific Reports. 9, 5821 (2019).

Kang, D.W., et al. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome.2017 Jan 23;5(1):10.

Karagözlü, S., et al. The Relationship of Severity of Autism with Gastrointestinal Symptoms and Serum Zonulin Levels in Autistic Children. J. Autism Dev. Disord. 2022;52:623–629.

Kelly, J.R., et al. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disordersFront Cell Neurosci. 2015 Oct 14;9:392.

Kim-Lee, C., et al. Gastrointestinal disease in Sjogren's syndrome: related to food hypersensitivities. Springerplus. 2015 Dec 12;4:766.

Kitano, H., et al. Robustness trade-offs and host-microbial symbiosis in the immune system. Molecular Systems Biology. 2 (2006).

Kobliner, V., et al. Reduction in Obsessive Compulsive Disorder and Self-Injurious Behavior With Saccharomyces boulardii in a Child with Autism: A Case Report. Integr Med (Encinitas). 2018 Dec;17(6):38-41.

Korpela, K., et al. Maternal Fecal Microbiota Transplantation in Cesarean-Born Infants Rapidly Restores Normal Gut Microbial Development: A Proof-of-Concept Study. Cell, 2020.

Lai, C.C.W., et al. The association between gut-health promoting diet and depression: A mediation analysis. J Affect Disord. 2023 Mar 1:324:136-142.

Lavebratt, C., et al. Early exposure to antibiotic drugs and risk for psychiatric disorders: a population-based study. Transl Psychiatry. 2019 Nov 26;9(1):317.

Liu, Z., et al. Tight junctions, leaky intestines, and pediatric diseases. Acta Paediatricia. 94 (2005): 386-393.

Madra, M., et al. Gastrointestinal Issues and Autism Spectrum Disorder. Psychiatr Clin North Am. 2021 Mar; 44(1): 69–81.

Manichanh, C., et al. The gut microbiota in IBD. Nat Rev Gastroenterol Hepatol. 2012;9(10):599-608.

Mine, Y., et al. Surfactants Enhance the Tight-Junction Permeability of Food Allergens in Human Intestinal Epithelial Caco-2 Cells. International Archives of Allergy and Immunology. 2003 Feb;130(2):135-42.

Mitre, E., et al. Association Between Use of Acid-Suppressive Medications and Antibiotics During Infancy and Allergic Diseases in Early Childhood. JAMA Pediatr. 2018 Jun 4;172(6):e180315.

Mohan, R., et al. Effects of Bifidobacterium lactis Bb12 Supplementation on Intestinal Microbiota of Preterm Infants: A Double-Blind, Placebo-Controlled, Randomized Study. Journal of Clinical Microbiology. 2006 Nov;44(11):4025-31.

Möller, C., et al. Intestinal permeability as assessed with polyethyleneglycols in birch pollen allergic children undergoing oral immunotherapy. Allergy. 1986 May;41(4):280-5.

Montalto, M., et al. Fecal Calprotectin Concentrations in Patients with Small Intestinal Bacterial Overgrowth. Digestive Diseases. 2008;26(2):183-6.

Morris, C.R., et al. Syndrome of allergy, apraxia, and malabsorption: characterization of a neurodevelopmental phenotype that responds to omega 3 and vitamin E supplementation. Alternative Therapies in Health and Medicine. Jul-Aug 2009;15(4):34-43.

Moser, L.A. Astrovirus Increases Epithelial Barrier Permeability Independently of Viral Replication. Journal of Virology. 2007 Nov;81(21):11937-45.

Nankova, B.B., 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(8):e103740.

Nankova, B.B. Nicotinic Induction of Preproenkephalin and Tyrosine Hydroxylase Gene Expression in Butyrate-Differentiated Rat PC12 Cells: A Model for Adaptation to Gut-Derived Environmental Signals. Pediatric Research. 2003 Jan;53(1):113-8.

Nemechek, P., et al. Autism Spectrum Disorder Symptoms Improve with Combination Therapy Directed at Improving Gut Microbiota and Reducing Inflammation. Applied Psychiatry. 2020 Jul; (1)1.

Niederhofer, H., et al. A preliminary investigation of ADHD symptoms in persons with celiac disease. J Atten Disorder. 2006 Nov;10(2):200-4.

Nirmalkar, K., et al. Shotgun Metagenomics Study Suggests Alteration in Sulfur Metabolism and Oxidative Stress in Children with Autism and Improvement after Microbiota Transfer Therapy. Int J Mol Sci. 2022 Nov 3;23(21):13481.

Nikolova, V.L., et al. Acceptability, Tolerability, and Estimates of Putative Treatment Effects of Probiotics as Adjunctive Treatment in Patients With Depression. JAMA Psychiatry. 2023 Aug 1;80(8):842-847.

Nirmalkar, K., et al. Shotgun Metagenomics Study Suggests Alteration in Sulfur Metabolism and Oxidative Stress in Children with Autism and Improvement after Microbiota Transfer Therapy. Int J Mol Sci. 2022 Nov 3;23(21):13481.

O’Hara, A.M., et al. The gut flora as a forgotten organ. European Molecular Biology Organization Report 7, no 7 (July 2006): 688-693.

Oliva-Hemker, M., et al. Fecal Microbiota Transplantation: Information for the Pediatrician. Pediatrics. 2023 Dec 1;152(6):e2023062922.

Ozkul, C., et al. A single early-in-life antibiotic course increases susceptibility to DSS-induced colitis. Genome Med. 2020;12(1):65.

Parkin, K.,, et al. Risk Factors for Gut Dysbiosis in Early Life. Microorganisms. 2021 Sep 30;9(10):2066.

Pearson, A.D., et al. Intestinal permeability in children with Crohn's disease and coeliac disease. Br Med J (Clin Res Ed). 1982 Jul 3;285(6334):20-1.

Rautava, S., et al. The Development of Gut Immune Responses and Gut Microbiota: Effects of Probiotics in Prevention and Treatment of Allergic Disease. Current Issues in Intestinal Microbiology. 2002 Mar;3(1):15-22.

Rediers, H., et al. Unraveling the Secret Lives of Bacteria: Use of In Vivo Expression Technology and Differential Fluorescence Induction Promoter Traps as Tools for Exploring Niche-Specific Gene Expression. Microbiology and Molecular Biology Reviews. 2005 Jun;69(2):217-61.

Relman, D.A. The human microbiome: ecosystem resilience and health. Nutr Rev. 2012 Aug;70 Suppl 1(Suppl 1):S2-9.

Ristori, M.V., et al. Autism, Gastrointestinal Symptoms and Modulation of Gut Microbiota by Nutritional Interventions. Nutrients. 2019;11:2812.

Rook, G.A., et al. Microbiota, immunoregulatory old friends and psychiatric disorders. Adv Exp Med Biol. 2014;817:319-56.

Rowland, I.R., et al. Effects of diet on mercury metabolism and excretion in mice given methylmercury: role of gut flora. Archives of Environmental Health. Nov-Dec 1984;39(6):401-8.

Sanchez, A., et al. Role of sugars in human neutrophilic phagocytosis. Am J Clin Nutr. 1973 Nov;26(11):1180-4.

Savino, F., et al. Lactobacillus reuteri (American Type Culture Collection Strain 55730) versus simethicone in the treatment of infantile colic: a prospective randomized study. Pediatrics. 2007 Jan;119(1):e124-30.

Scirocco, A., et al. Exposure of Toll-like receptors 4 to bacterial lipopolysaccharide (LPS) impairs human colonic smooth muscle cell function. J Cell Physiol. 2010 May;223(2):442-50.

Sela, D.A., et al. The marriage of nutrigenomics with the microbiome: the case of infant associated bifidobacteria and milk. Am J Clin Nutr. 2014;99(3):697S-703S.

Severance, E.G., et al. IgG dynamics of dietary antigens point to cerebrospinal fluid barrier or flow dysfunction in first-episode schizophrenia. Brain Behav Immun. 2015 Feb;44:148-58.

Shekhawat, P.S., et al. Spontaneous development of intestinal and colonic atrophy and inflammation in the carnitine-deficient jvs (OCTN2(-/-)) mice. Mol Genet Metab. 2007 Dec;92(4):315-24.

Sifroni, K.G., et al. Mitochondrial respiratory chain in the colonic mucosal of patients with ulcerative colitis. Mol Cell Biochem. 2010 Sep;342(1-2):111-5.

Simpson, C.A., et al. The gut microbiota in anxiety and depression - A systematic review. Clin Psychol Rev. 2021 Feb:83:101943.

Stewart, C.J., et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature. 2018 Oct;562(7728):583-588.

Strauch, U.G., et al. Influence of intestinal bacteria on induction of regulatory T cells: lessons from a transfer model of colitis. Gut 54 (2005):1546-1552.

Strandwitz, P. Neurotransmitter Modulation by the Gut Microbiota. Brain Res. 2018 Aug 15;1693(Pt B):128-133.

Tobacman, J.K. Review of Harmful Gastrointestinal Effects of Carrageenan in Animal Experiments. Environmental Health Perspectives. 2001 Oct;109(10):983-94.

Uhde, M., et al. Intestinal cell damage and systemic immune activation in individuals reporting sensitivity to wheat in the absence of coeliac disease. Gut. 2016 Dec;65(12):1930-1937.

Vael, C., et al. Early intestinal Bacteroides fragilis colonization and development of asthma. BMC Pulmonary Medicine. 2008 Sep 26;8:19.

van Nood, E., et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N Engl J Med. 2013 Jan 31;368(5):407-15.

Voigt, R.M., et al. The intestinal microbiota: determinants of resiliency? Lancet Healthy Longev. 2021 Jan;2(1):e2-e3.

Vojdani, A., et al. A Gut Feeling for Immune Dysregulation & Neuroinflammation in Autism. The Autism File. 2009(31).

Wang, J., et al. Global Prevalence of Autism Spectrum Disorder and Its Gastrointestinal Symptoms: A Systematic Review and Meta-Analysis. Front. Psychiatry. 2022;13:963102.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Wasilewska, J., et al. Gastrointestinal symptoms and autism spectrum disorder: links and risks – a possible new overlap syndrome. Pediatric Health Med Ther. 2015; 6: 153–166.

West, C.E., et al. The gut microbiota and inflammatory noncommunicable diseases: associations and potentials for gut microbiota therapies. J Allergy Clin Immunol. 2015;135(1):3-13; quiz 4.

Wexler, H. Bacteroides: the Good, the Bad, and the Nitty-Gritty. Clinical Microbiology Reviews 20, no. 4 (October 2007): 593-621.

White, E., et al. The Effect of Nutritional Therapy for Yeast Infection (Candidiasis) in Cases of Chronic Fatigue Syndrome. Journal of Orthomolecular Medicine. 2005;20(3).

Williams, B.L., et al. Impaired Carbohydrate Digestion and Transport and Mucosal Dysbiosis in the Intestines of Children with Autism and Gastrointestinal Disturbances. PLoS ONE. 2011;6:e24585.

Williams, R.E.O., et al. The influence of intestinal bacteria on the absorption and metabolism of foreign compounds. Journal of Clinical Pathology.1971; 5: 125–129.

Wilmanski, T., et al. Gut microbiome pattern reflects healthy ageing and predicts survival in humans. Nat Metab. 2021 Feb;3(2):274-286.

Yang, B., et al. Effects of regulating intestinal micobiota on anxiety symptoms: A systematic review. General Psychiatry. 2019; 32: e100056.

Zheng, P., et al. Gut microbiome in type 1 diabetes: A comprehensive review. Diabetes Metab Res Rev. 2018 Oct;34(7):e3043.

Zhou, H., et al. Evaluating the Causal Role of Gut Microbiota in Type 1 Diabetes and Its Possible Pathogenic Mechanisms. Front Endocrinol (Lausanne). 2020 Mar 24:11:125.

Zhu, B., et al. Human gut microbiome: the second genome of human body. Protein Cell. 2010 Aug;1(8):718-25.

Mitochondrial Dysfunction

Sources & References

Andreazza, A.C., et al. Mitochondrial complex I activity and oxidative damage to mitochondrial proteins in the prefrontal cortex of patients with bipolar disorder. Arch Gen Psychiatry. 2010 Apr;67(4):360-8.

Balcells, Cristy. Autism & Mitochondrial Disorders: How Much Do We Really Know?. MitoAction.org. 29 Jan 2009

Beal, M.F., et al. Therapeutic approaches to mitochondrial dysfunction in Parkinson's disease. Parkinsonism Relat Disord. 2009 Dec;15 Suppl 3:S189-94.

Bradford, B.L., et al. Mitochondrial Dysfunction and Type 2 Diabetes. Science. 2005 Jan 21;307(5708):384-7.

Bradstreet, J.J., et al. Biomarker-guided interventions of clinically relevant conditions associated with autism spectrum disorders and attention deficit hyperactivity disorder. Altern Med Rev. 2010 Apr;15(1):15-32.

Burchell, V.S., et al. Targeting mitochondrial dysfunction in neurodegenerative disease: Part I. Expert Opin Ther Targets. 2010 Apr;14(4):369-85.

Burchell, V.S., et al. Targeting mitochondrial dysfunction in neurodegenerative disease: Part II. Expert Opin Ther Targets. 2010 May;14(5):497-511.

Davi, Alyssa. Has Your Child with Autistic Symptoms Been Properly Screened for a Subset of Mitochondrial Disease Known as OXPHOS?...Probably Not. Autism File. 2010; 36.

Dehley, Leanna M., et al. The Effect of Mitochondrial Supplements on Mitochondrial Activity in Children with Autism Spectrum Disorder. J Clin Med. 2017 Feb; 6(2): 18.

Ferrer, et al. Early involvement of the cerebral cortex in Parkinson's disease: convergence of multiple metabolic defects. Prog Neurobiol. 2009 Jun;88(2):89-103.

Filipek, P.A., et al. Relative carnitine deficiency in autism. J Autism Dev Disord. 2004 Dec;34(6):615-23.

Haas, R.H., et al. Mitochondrial disease: a practical approach for primary care physicians. Pediatrics. 2007 Dec;120(6):1326-33.

Hao, J., et al. Mitochondrial nutrients improve immune dysfunction in the type 2 diabetic Goto-Kakizaki rats. J Cell Mol Med. 2009 Apr;13(4):701-11.

Herbert, M.R. Contributions of the environment and environmentally vulnerable physiology to autism spectrum disorders. Curr Opin Neurol. 2010 Apr;23(2):103-10

James, S.J., et al. Cellular and mitochondrial glutathione redox imbalance in lymphoblastoid cells derived from children with autism. FASEB J. 2009 Aug;23(8):2374-83.

Kato, T. The role of mitochondrial dysfunction in bipolar disorder. Drug News Perspect. 2006 Dec;19(10):597-602.

Kelley, R.I. Evaluation and Treatment of Patients with Autism and Mitochondrial Disease. Kennedy Krieger Institute, Division of Metabolism.

Klehm, M., et al. Clinician's Guide to the Management of Mitochondrial Disease: A Manual for Primary Care Providers. MitiAction.org. 2014.

Konradi, C., et al. Molecular evidence for mitochondrial dysfunction in bipolar disorder. Arch Gen Psychiatry. 2004 Mar;61(3):300-8.

Korson, M. Mitochondrial Disease and Patient Challenges. MitoAction. 2008.

Leib, S., et al. Mitochondrial Oxidative Phosphorylation (OXPHOS) Dysfunction: A Newly Emerging Category of Autistic Spectrum Disorder Information for Primary Care Physicians. MitoAction.org. 2010.

Liu, J. The effects and mechanisms of mitochondrial nutrient alpha-lipoic acid on improving age-associated mitochondrial and cognitive dysfunction: an overview. Neurochem Res. 2008 Jan;33(1):194-203.

Long, J., et al. Mitochondrial decay in the of old rats: ameliorating effect of alpha-lipoic acid and acetyl-L-carnitine. Neurochem Res. 2009 Apr;34(4):755-63.

Mabalirajan, U., et al. Effects of vitamin E on mitochondrial and asthma features in an experimental allergic murine model. J Appl Physiol. 2009 Oct;107(4):1285-92.

Maes, M., et al. Coenzyme Q10 deficiency in myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is related to fatigue, autonomic and neurocognitive symptoms and is another risk factor explaining the early mortality in ME/CFS due to cardiovascular disorder. Neuro Endocrinol Lett. 2009;30(4):470-6.

Maes, M., et al. Lower plasma Coenzyme Q10 in depression: a marker for treatment resistance and chronic fatigue in depression and a risk factor to cardiovascular disorder in that illness. Neuro Endocrinol Lett. 2009;30(4):462-9.

Morino, K., et al. Molecular mechanisms of insulin resistance in humans and their potential links with mitochondrial dysfunction. Diabetes. 2006 Dec;55 Suppl 2:S9-S15.

Napoli, E., et al. Toxicity of the flame-retardant BDE-49 on brain mitochondria and neuronal progenitor striatal cells enhanced by a PTEN-deficient background. Toxicol Sci. 2013 Mar;132(1):196-210.

Noland, R.C., et al. Carnitine insufficiency caused by aging and overnutrition compromises mitochondrial performance and metabolic control. J Biol Chem. 2009 Aug 21;284(34):22840-52.

Oliveira, G., et al. Mitochondrial dysfunction in autism spectrum disorders: a population-based study. Dev Med Child Neurol. 2005 Mar;47(3):185-9.

Palmieri, L., et al. Mitochondrial dysfunction in autism spectrum disorders: cause or effect? Biochim Biophys Acta. 2010 Jun-Jul;1797(6-7):1130-7.

Parikh, S. The neurologic manifestations of mitochondrial disease. Dev Disabil Res Rev. 2010;16(2):120-8.

Parikh, S., et al. A Modern Approach to the Treatment of Mitochondrial Disease. Current Treatment Options in Neurology. 2009 Nov;11(6):414-30.

Pastural, E., et al. Novel plasma phospholipid biomarkers of autism: mitochondrial dysfunction as a putative causative mechanism. Prostaglandins Leukot Essent Fatty Acids. 2009 Oct;81(4):253-64.

Power, R.A., et al. Carnitine revisited: potential use as adjunctive treatment in diabetes. Diabetologia. 2007 Apr;50(4):824-32.

Rector, R.S., et al. Mitochondrial dysfunction precedes insulin resistance and hepatic steatosis and contributes to the natural history of non-alcoholic fatty liver disease in an obese rodent model. J Hepatol. 2010 May;52(5):727-36.

Schmidt, Charles W. Mito-Conundrum: Unraveling Environmental Effects on Mitochondria. Environmental Health Perspectives. 2010 July; 118(7).

Scirocco, A., et al. Exposure of Toll-like receptors 4 to bacterial lipopolysaccharide (LPS) impairs human colonic smooth muscle cell function. J Cell Physiol. 2010 May;223(2):442-50.

Shen, W., et al. Protective effects of R-alpha-lipoic acid and acetyl-L-carnitine in MIN6 and isolated rat islet cells chronically exposed to oleic acid. J Cell Biochem. 2008 Jul 1;104(4):1232-43.

Shekhawat, P.S., et al. Spontaneous development of intestinal and colonic atrophy and inflammation in the carnitine-deficient jvs (OCTN2(-/-)) mice. Mol Genet Metab. 2007 Dec;92(4):315-24.

Shoffner, J., et al. Fever Plus Mitochondrial Disease Could Be Risk Factors for Autistic Regression. J Child Neurol. 2010 Apr;25(4):429-34.

Shokolenko, I., et al. Oxidative stress induces degradation of mitochondrial DNA. Nucleic Acids Res. 2009 May; 37(8): 2539–2548.

Sifroni, K.G., et al. Mitochondrial respiratory chain in the colonic mucosal of patients with ulcerative colitis. Mol Cell Biochem. 2010 Sep;342(1-2):111-5.

Spindler, M., et al. Coenzyme Q10 effects in neurodegenerative disease. Neuropsychiatr Dis Treat. 2009;5:597-610.

Sreekumar, R., et al. Skeletal muscle mitochondrial dysfunction & diabetes. Indian J Med Res. 2007 Mar;125(3):399-410.

Taurines, R., et al. Expression analyses of the mitochondrial complex I 75-kDa subunit in early onset schizophrenia and autism spectrum disorder: increased levels as a potential biomarker for early onset schizophrenia. Eur Child Adolesc Psychiatry. 2010 May;19(5):441-8.

Weissman, J.R., et al. Mitochondrial Disease in Autism Spectrum Disorder Patients: A Cohort Analysis. PLoS One. 2008;3(11):e3815.

Zhang, H., et al. Combined R-alpha-lipoic acid and acetyl-L-carnitine exerts efficient preventative effects in a cellular model of Parkinson's disease. J Cell Mol Med. 2010 Jan;14(1-2):215-25.

Mold

Sources & References

Baker, S., et al. Case Study: Rapid Complete Recovery From An Autism Spectrum Disorder After Treatment of Aspergillus With The Antifungal Drugs Itraconazole And Sporanox. Integr Med (Encenitas). 2020 Aug;19(4):20-27.

Bennett, J.W., et al. Mycotoxins. Clinical Microbiology Reviews. Jul 2003: 497-516.

Berndtson, K., et al. Medically sound investigation and remediation of water-damaged buildings in cases of CIRS-WDB. Center for Research on Biotoxin Associated Illness. 2016 Jan 19.

Berry, Y. A Physician’s Guide to Understanding and Treating Biotoxin Illness. 2014.

de Allori, M.C.G., et al. Antimicrobial resistance and production of biofilms in clinical isolates of coagulase-negative Staphylococcus strains. Biol Pharm Bull. 2006 Aug;29(8):1592-6.

De Santis, B., et al. Role of mycotoxins in the pathobiology of autism: A first evidence. Nutr Neurosci. 2019 Feb;22(2):132-144.

De Santis, B., et al. Study on the Association among Mycotoxins and other Variables in Children with Autism. Toxins (Basel). 2017 Jun 29;9(7):203.

Hope, J. A review of the mechanism of injury and treatment approaches for illness resulting from exposure to water-damaged buildings, mold, and mycotoxins. ScientificWorldJournal. 2013 Apr 18;2013:767482.

Hudnell, H.K. Chronic biotoxin-associated illness: multiple-system symptoms, a vision deficit, and effective treatment. Neurotoxicol Teratol. Sep-Oct 2005;27(5):733-43.

Institute of Medicine (US) Committee on Damp Indoor Spaces and Health. Damp Indoor Spaces and Health. Washington (DC): National Academies Press (US); 2004.

Kali, A. Antibiotics and bioactive natural products in treatment of methicillin resistant Staphylococcus aureus: A brief review. Pharmacogn Rev. Jan-Jun 2015;9(17):29-34.

Kontoyiannis, D.P., et al. Treatment principles for the management of mold infections. Cold Spring Harb Perspect Med. 2014 Nov 6;5(4):a019737.

Oliva, A., et al. High Potency of Melaleuca alternifolia Essential Oil against Multi-Drug Resistant Gram-Negative Bacteria and Methicillin-Resistant Staphylococcus aureus. Molecules. 2018 Oct 9;23(10):2584.

Ratnaseelen, A.M., et al. Effects of Mycotoxins on Neuropsychiatric Symptoms and Immune Processes. Clin Ther. 2018 Jun;40(6):903-917.

Salari, M.H., et al. Antibacterial effects of Eucalyptus globulus leaf extract on pathogenic bacteria isolated from specimens of patients with respiratory tract disorders. Clin Microbiol Infect. 2006 Feb;12(2):194-6.

Shoemaker, R.C., et al. Sick building syndrome (SBS) and exposure to water-damaged buildings: time series study, clinical trial and mechanisms. Neurotoxicol Teratol. Sep-Oct 2006;28(5):573-88.

Shoemaker, R.C., et al. Structural brain abnormalities in patients with inflammatory illness acquired following exposure to water-damaged buildings: a volumetric MRI study using NeuroQuant®. Neurotoxicol Teratol. Sep-Oct 2014;45:18-26.

Shoemaker, R.C., et al. A time-series study of sick building syndrome: chronic, biotoxin-associated illness from exposure to water-damaged buildings. Neurotoxicol Teratol. Jan-Feb 2005;27(1):29-46.

Music Therapy

Sources & References

Amos, P. Rhythm and timing in autism: learning to dance. Front Integr Neurosci. 2013 Apr 19;7:27.

Barnhill, E. Neural connectivity, music, and movement: a response to Pat Amos. Front Integr Neurosci. 2013 Apr 24;7:29.

Hardy, M.W., et al. Rhythm, movement, and autism: using rhythmic rehabilitation research as a model for autism. Front Integr Neurosci. 2013 Mar 28;7:19.

Hong, H.J., et al. Effect of Rhythmic Movement Program to Improve Walking Ability for Elderly Patients with Stroke. Indian Journal of Science and Technology. 2016 Jul;9(26).

Kadivar, Z., et al. Effect of step training and rhythmic auditory stimulation on functional performance in Parkinson patients. Neurorehabil Neural Repair. 2011 Sep;25(7):626-35.

Ladányi, K. et al. Is atypical rhythm a risk factor for developmental speech and language disorders? Wiley Interdiscip Rev Cogn Sci. 2020 Sep;11(5):e1528.

Lakatos, P., et al. A New Unifying Account of the Roles of Neuronal Entrainment. Curr Biol. 2019 Sep 23;29(18):R890-R905.

Suh, J.H., et al. Effect of rhythmic auditory stimulation on gait and balance in hemiplegic stroke patients. NeuroRehabilitation. 2014;34(1):193-9.

Van Hirtum, T., et al. Is atypical rhythm a riskfactor for developmental speech and language disorders? J Assoc Res Otolaryngol. 2021 Jul;22(4):465-480.

Winkler, I., et al. Newborn infants detect the beat in music. Proc Natl Acad Sci U S A. 2009 Feb 17;106(7):2468-71.

Zentner, M., et al. Rhythmic engagement with music in infancy. Proc Natl Acad Sci U S A. 2010 Mar 30;107(13):5768-73.

Myofunctional Therapy

Sources & References

Baxter, R., et al. Speech and Feeding Improvements in Children After Posterior Tongue-Tie Release: A Case Series. Intl Journal of Clinical Pediatrics. 2018 Sep,7(3):29-35.

Berry, J., et al. A double-blind, randomized, controlled trial of tongue-tie division and its immediate effect on breastfeeding. Breastfeed Med. 2012 Jun;7(3):189-93.

Bussi, M.T., et al. Is ankyloglossia associated with obstructive sleep apnea? Braz J Otorhinolaryngol. 2021 Nov 5;S1808-8694(21)00181-6.

Diaféria, G. et al. Myofunctional therapy improves adherence to continuous positive airway pressure treatment. Sleep Breath. 2017 May;21(2):387-395.

Edmond, A., et al. Randomised controlled trial of early frenotomy in breastfed infants with mild-moderate tongue-tie. Arch Dis Child Fetal Neonatal Ed. 2014 May;99(3):F189-95.

Erturk, N., et al. The effectiveness of oropharyngeal exercises compared to inspiratory muscle training in obstructive sleep apnea: A randomized controlled trial. Heart Lung. 2020 Nov-Dec;49(6):940-948.

Ghaheri, B.A., et al. Breastfeeding improvement following tongue-tie and lip-tie release: A prospective cohort study. Laryngoscope. 2017 May;127(5):1217-1223.

Ghaheri, B.A., et al. Revision Lingual Frenotomy Improves Patient-Reported Breastfeeding Outcomes: A Prospective Cohort Study. J Hum Lact. 2018 Aug;34(3):566-574.

Guimarães, K.C., et al. Effects of oropharyngeal exercises on patients with moderate obstructive sleep apnea syndrome. Am J Respir Crit Care Med. 2009 May 15;179(10):962-6.

Harari, D., et al. The effect of mouth breathing versus nasal breathing on dentofacial and craniofacial development in orthodontic patients. Laryngoscope. 2010 Oct;120(10):2089-93.

Hesselbacher, S., et al. A Study to Assess the Relationship between Attention Deficit Hyperactivity Disorder and Obstructive Sleep Apnea in Adults. Cureus. 2019 Oct 24;11(10):e5979.

Hogan, M., et al. Randomized, controlled trial of division of tongue-tie in infants with feeding problems. J Paediatr Child Health. May-Jun 2005;41(5-6):246-50.

Hsu, B., et al. Effects of respiratory muscle therapy on obstructive sleep apnea: a systematic review and meta-analysis. J Clin Sleep Med. 2020 May 15;16(5):785-801.

Huang, Y.S., et al. Attention-deficit/hyperactivity disorder with obstructive sleep apnea: a treatment outcome study. Sleep Med. 2007 Jan;8(1):18-30.

Huang, Y.S., et al. Short Lingual Frenulum and Obstructive Sleep Apnea in Children. Intl Journal of Clinical Pediatrics. 2015,1(1):1-4.

Hvolby, A. Associations of sleep disturbance with ADHD: implications for treatment. Atten Defic Hyperact Disord. 2015 Mar;7(1):1-18.

Ieto, V., et al. Effects of Oropharyngeal Exercises on Snoring: A Randomized Trial. Chest. 2015 Sep;148(3):683-691.

Ito, Y., et al. Effectiveness of tongue-tie division for speech disorder in children. Pediatr Int. 2015 Apr;57(2):222-6.

Messner, A.H., et al. The effect of ankyloglossia on speech in children. Otolaryngol Head Neck Surg. 2002 Dec;127(6):539-45.

O’Callahan, C., et al. The effects of office-based frenotomy for anterior and posterior ankyloglossia on breastfeeding. Int J Pediatr Otorhinolaryngol. 2013 May;77(5):827-32.

Rueda, J.R., et al. Myofunctional therapy (oropharyngeal exercises) for obstructive sleep apnoea. Cochrane Database Syst Rev. 2020 Nov 3;11(11):CD013449.

Sabuncuoglu, O. Understanding the relationships between breastfeeding, malocclusion, ADHD, sleep-disordered breathing and traumatic dental injuries. Med Hypotheses. 2013 Mar;80(3):315-20.

Siegel, S. Aerophagia Induced Reflux in Breastfeeding Infants With Ankyloglossia and Shortened Maxillary Labial Frenula (Tongue and Lip Tie). Int J Clin Pediatr. 2016;5(1):6-8.

Villa, M.P., et al. Oropharyngeal exercises to reduce symptoms of OSA after AT. Sleep Breath. 2015 Mar;19(1):281-9.

Neurofeedback

Sources & References

American Academy of Pediatrics. Biofeedback now a “Level 1 — Best Support” Intervention for Attention & Hyperactivity Behaviors. Evidence-based Child and Adolescent Psycho-social Interventions. 2012 Oct 5.

Arns, M., et al. Efficacy of neurofeedback treatment in ADHD: the effects on inattention, impulsivity and hyperactivity: a meta-analysis. Clinical EEG and Neuroscience, 40(3), 180-189.

Beauregard, M., et al. Functional magnetic resonance imaging investigation of the effects of neurofeedback training on neural bases of selective attention and response inhibition in children with attention-deficit/hyperactivity disorder. Applied Psychophysiology and Biofeedback, 2006 Mar;31(1):3-20.

Chiu, H.J., et al. Surface electroencephalographic neurofeedback improves sustained attention in ADHD: a meta-analysis of randomized controlled trials. Child Adolesc Psychiatry Ment Health. 2022 Dec 19;16(1):104.

Dudek, E., et al. The efficacy of real-time functional magnetic resonance imaging neurofeedback for psychiatric illness: A meta-analysis of brain and behavioral outcomes.
Neurosci Biobehav Rev. 2021 Feb;121:291-306.

Duric, N.S., et al. Neurofeedback for the treatment of children and adolescents with ADHD: A randomized and controlled clinical trial using parental reports. BMC Psychiatry, 2012 Aug 10;12:107.

Gani, C., et al. Long term effects after feedback of slow cortical potentials and of theta-beta-amplitudes in children with attention-deficit/hyperactivity disorder. International Journal of Bioelectromagnetism, 2008; 10, 4, 209 -232.

Gevensleben, H., et al. (2009). Is neurofeedback an efficacious treatment for ADHD?: A randomized controlled clinical trial. Journal of Child Psychology and Psychiatry, 2009 Jul;50(7):780-9.

Jarusiewicz, B. Efficacy of Neurofeedback for Children in the Autistic Spectrum: A Pilot Study. Journal of Neurotherapy. 2002;6(4).

Leins, U., et al. Neurofeedback for children with ADHD: A comparison of SCP and Theta/Beta protocols. Appl Psychophysiol Biofeedback, 2007 Jun;32(2):73-88.

Levesque, J., et al. Effect of neurofeedback training on the neural substrates of selective attention in children with attention deficit/hyperactivity disorder: A functional magnetic resonance imaging study. Neuroscience Letters, 2006 Feb 20;394(3):216-21.

Linhartová, P., et al. fMRI neurofeedback in emotion regulation: A literature review.
Neuroimage. 2019 Jun;193:75-92.

Monastra, V.J., et al. The effects of stimulant therapy, EEG biofeedback, and parenting style on the primary symptoms of attention-deficit/hyperactivity disorder. Applied Psychophysiology and Biofeedback, 2002 Dec;27(4):231-49.

Monastra, V.J., et al. Electroencephalographic biofeedback (neurotherapy) as a treatment for attention deficit hyperactivity disorder: rationale and empirical foundation. Child Adolesc Psychiatric Clin N Am, 2005 Jan;14(1):55-82, vi.

Moreno-García, I., et al. Results of Neurofeedback in Treatment of Children with ADHD: A Systematic Review of Randomized Controlled Trials. Appl Psychophysiol Biofeedback. 2022 Sep;47(3):145-181.

Neurofeedback Collaborative Group, et al.
Neurofeedback for Attention-Deficit/Hyperactivity Disorder: 25-Month Follow-up of Double-Blind Randomized Controlled Trial.
J Am Acad Child Adolesc Psychiatry. 2022 Dec 8;S0890-8567(22)01971-2.

Nicholson, A.A., et al. The neurobiology of emotion regulation in posttraumatic stress disorder: Amygdala downregulation via real-time fMRI neurofeedback. Hum Brain Mapp. 2017 Jan;38(1):541-560.

Pindi, P., et al. Real-time fMRI neurofeedback as a new treatment for psychiatric disorders: A meta-analysis. Prog Neuropsychopharmacol Biol Psychiatry. 2022 Dec 20;119:110605.

Rothenberger, A., et al. Informing the ADHD Debate. Scientific American Special Edition. 2004 Dec; 14(5):50-55.

Roy, S., et al. Effectiveness of neurofeedback training, behaviour management including attention enhancement training and medication in children with attention-deficit/hyperactivity disorder - A comparative follow up study. Asian J Psychiatr. 2022 Oct;76:103133.

Neurotransmitter Imbalances

Sources & References

Appleton, J. The Gut-Brain Axis: Influence of Microbiota on Mood and Mental Health. Integr Med (Encinitas). 2018 Aug;17(4):28-32.

Atladottir, H.O., et al. The increasing prevalence of reported diagnoses of childhood psychiatric disorders: a descriptive multinational comparison. Eur Child Adolesc Psychiatry. 2015;24(2):173-83.

Berk, M., et al. So depression is an inflammatory disease, but where does the inflammation come from? BMC Med. 2013;11:200.

Berry, E.A., et al. National estimates of the inpatient burden of pediatric bipolar disorder in the United States. J Ment Health Policy Econ. 2011;14(3):115-23.

Bayer, J.K., et al. The Cool Little Kids randomised controlled trial: population-level early prevention for anxiety disorders. BMC Public Health. 2011;11:11.

Bitsko, R.H., et al. Epidemiology and Impact of Health Care Provider-Diagnosed Anxiety and Depression Among US Children. J Dev Behav Pediatr 2018 Apr 24.

Bonnot, O., et al. Children and adolescents with severe mental illness need vitamin D supplementation regardless of disease or treatment. J Child Adolesc Psychopharmacol. 2011;21(2):157-61.

Camilleri, M. Serotonin in the gastrointestinal tract. Curr Opin Endrocrinol Diabetes Obes. 2009 Feb;16(1):53-9.

Ceylan, M.F., et al. Lipid peroxidation markers in children with anxiety disorders and their diagnostic implications. Redox Rep. 2014;19(2):92-6.

Chen, Y., et al. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients. 2021 Jun 19;13(6):2099.

Cohen-Cline, H., et al. Access to green space, physical activity and mental health: a twin study. J Epidemiol Community Health. 2015 Jun;69(6):523-9.

Costello, E.J., et al. 10-year research update review: the epidemiology of child and adolescent psychiatric disorders: II. Developmental epidemiology. J Am Acad Child Adolesc Psychiatry. 2006 Jan;45(1):8–25.

Dusetzina, S.B., et al. Treatment use and costs among privately insured youths with diagnoses of bipolar disorder. Psychiatr Serv. 2012;63(10):1019-25.

Guney, E., et al. Oxidative stress in children and adolescents with anxiety disorders. J Affect Disord. 2014;156:62-6.

Fernandes, A.C., et al. Development and evaluation of a de-identification procedure for a case register sourced from mental health electronic records. BMC Med Inform Decis Mak. 2013;13:71.

Hepgul, N., et al. Depression pathogenesis and treatment: what can we learn from blood mRNA expression? BMC Med. 2013;11:28.

Karakula, H., et al. [Does diet affect our mood? The significance of folic acid and homocysteine]. Pol Merkur Lekarski. 2009;26(152):136-41.

Leuchter, A.F., et al. Intermediate phenotypes and biomarkers of treatment outcome in major depressive disorder. Dialogues Clin Neurosci. 2014;16(4):525-37.

Marazziti, D., et al. Psychiatric disorders and mitochondrial dysfunctions. Eur Rev Med Pharmacol Sci. 2012;16(2):270-5.

Mitchell, E.S., et al. B vitamin polymorphisms and behavior: evidence of associations with neurodevelopment, depression, schizophrenia,bipolar disorder and cognitive decline. Neurosci Biobehav Rev. 2014;47:307-20.

Ostiguy, C.S., et al. Sensitivity to stress among the offspring of parents with bipolar disorder: a study of daytime cortisol levels. Psychol Med. 2011;41(11):2447-57.

Rollins, B., et al. Mitochondrial variants in schizophrenia, bipolar disorder, and major depressive disorder. PLoS One. 2009;4(3):e4913.

Rook, G.A., et al. Microbiota, immunoregulatory old friends and psychiatric disorders. Adv Exp Med Biol. 2014;817:319-56.

Strandwitz, P. Neurotransmitter Modulation by the Gut Microbiota. Brain Res. 2018 Aug 15;1693(Pt B):128-133.

Thompson, L., et al. What have birth cohort studies asked about genetic, pre- and perinatal exposures and child and adolescent onset mental health outcomes? A systematic review. Eur Child Adolesc Psychiatry. 2010;19(1):1-15.

Van Meter, A.R., et al. What goes up must come down: the burden of bipolar depression in youth. J Affect Disord. 2013;150(3):1048-54.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Winter, C., et al. Dopamine and serotonin levels following prenatal viral infection in mouse—implications for psychiatric disorders such as schizophrenia and autism. European Neuropsychopharmacology. 2008 Oct;18(10):712-6.

Yang, B., et al. Effects of regulating intestinal micobiota on anxiety symptoms: A systematic review. General Psychiatry. 2019; 32: e100056.

Obstructive Sleep Apnea

Sources & References

Amdo, T., et al. Somatic syndromes, insomnia, anxiety, and stress among sleep disordered breathing patients. Sleep Breath. 2016 May;20(2):759-68.

Bussi, M.T., et al. Is ankyloglossia associated with obstructive sleep apnea? Braz J Otorhinolaryngol. 2021 Nov 5;S1808-8694(21)00181-6.

de Godoy, L.B., et al. Upper Airway Resistance Syndrome Patients Have Worse Sleep Quality Compared to Mild Obstructive Sleep Apnea. PLoS One. 2016; 11(5): e0156244.

Dieltjens, M., et al. Treatment of obstructive sleep apnea using a custom-made titratable duobloc oral appliance: a prospective clinical study. Sleep Breath. 2013 May;17(2):565-72.

Doff, M.H., et al. Oral Appliance Versus Continuous Positive Airway Pressure in Obstructive Sleep Apnea Syndrome: A 2-year Follow-Up. Sleep. 2013 Sep 1;36(9):1289-96.

Harari, D., et al. The effect of mouth breathing versus nasal breathing on dentofacial and craniofacial development in orthodontic patients. Laryngoscope. 2010 Oct;120(10):2089-93.

Hesselbacher, S., et al. A Study to Assess the Relationship between Attention Deficit Hyperactivity Disorder and Obstructive Sleep Apnea in Adults. Cureus. 2019 Oct 24;11(10):e5979.

Hsu, B., et al. Effects of respiratory muscle therapy on obstructive sleep apnea: a systematic review and meta-analysis. J Clin Sleep Med. 2020 May 15;16(5):785-801.

Huang, Y.S., et al. Attention-deficit/hyperactivity disorder with obstructive sleep apnea: a treatment outcome study. Sleep Med. 2007 Jan;8(1):18-30.

Huang, Y.S., et al. Short Lingual Frenulum and Obstructive Sleep Apnea in Children. Intl Journal of Clinical Pediatrics. 2015,1(1):1-4.

Hvolby, A. Associations of sleep disturbance with ADHD: implications for treatment. Atten Defic Hyperact Disord. 2015 Mar;7(1):1-18.

Kamata, S., et al. Assessment of Obstructive Apnea by Using Polysomnography and Surgical Treatment in Patients With Beckwith-Wiedemann Syndrome. J Pediatr Surg. 2005 Mar;40(3):E17-9.

Martel, J., et al. Could nasal nitric oxide help to mitigate the severity of COVID-19? Microbes Infect. 2020 May-June; 22(4): 168–171.

Felippe, N.L.O., et al. Relationship between rapid maxillary expansion and nasal cavity size and airway resistance: short- and long-term effects. Am J Orthod Dentofacial Orthop. 2008 Sep;134(3):370-82.

Reyes-Zúñiga, M., et al. Anxiety and depression symptoms in patients with sleep-disordered breathing. Open Respir Med J. 2012;6:97-103.

Sabuncuoglu, O. Understanding the relationships between breastfeeding, malocclusion, ADHD, sleep-disordered breathing and traumatic dental injuries. Med Hypotheses. 2013 Mar;80(3):315-20.

Sedky, K., et al. Attention deficit hyperactivity disorder and sleep disordered breathing in pediatric populations: a meta-analysis. Sleep Med Rev. 2014 Aug;18(4):349-56.

Urbano, G.L., et al. The Link between Pediatric Obstructive Sleep Apnea (OSA) and Attention Deficit Hyperactivity Disorder (ADHD). Children (Basel). 2021 Sep 19;8(9):824.

White, David P., et al. Mandibular Advancement Device vs. CPAP in the Treatment of Obstructive Sleep Apnea: Are They Equally Effective in Short Term Health Outcomes? J Clin Sleep Med. 2013 Sep 15;9(9):971-2.

Xiangming, L., et al. The relationship between inflammation and neurocognitive dysfunction in obstructive sleep apnea syndrome. J Neuroinflammation. 2020 Aug 1;17(1):229.

Yeghiazarians, Y., et al. Obstructive Sleep Apnea and Cardiovascular Disease: A Scientific Statement From the American Heart Association. Circulation. 2021 Jul 20;144(3):e56-e67.

Youssef, N.A., et al. Is obstructive sleep apnea associated with ADHD? Ann Clin Psychiatry. 2011 Aug;23(3):213-24.

Oral/Dental Health

Sources & References

Afeke, I., et al. Acinetobacter baumannii-induced infective endocarditis: new insights into pathophysiology and antibiotic resistance mechanisms. Future Microbiol. 2022 Nov;17:1335-1344.

Andriankaja, O., et al. Association between periodontal pathogens and risk of nonfatal myocardial infarction. Community Dent Oral Epidemiol. 2011 Apr;39(2):177-85.

Beydoun, M.A., et al. Clinical and Bacterial Markers of Periodontitis and Their Association with Incident All-Cause and Alzheimer’s Disease Dementia in a Large National Survey. J Alzheimers Dis. 2020;75(1):157-172.

Bonasoni, M.P., et al. Fulminant Sepsis and Perinatal Death at 23 Weeks Due to Fusobacterium nucleatum. Fetal Pediatr Pathol. 2023 Jun;42(3):456-463.

Carelli, M., et al. Oral Microbiota in Children and Adolescents with Type 1 Diabetes Mellitus: Novel Insights into the Pathogenesis of Dental and Periodontal Disease. Microorganisms. 2023 Mar 6;11(3):668.

Cerajewska, T.L. Periodontitis: a potential risk factor for Alzheimer’s disease. Br Dent J. 2015 Jan;218(1):29-34.

Chang, Y.R., et al. Links between oral microbiome and insulin resistance: Involvement of MAP kinase signaling pathway. Biochimie. 2023 Jul 12;214(Pt B):134-144.

Cho, H., et al. Selenomonas sputigena acts as a pathobiont mediating spatial structure and biofilm virulence in early childhood caries.
Nat Commun. 2023 May 22;14(1):2919.

Chopra, A., et al. Are Inflamed Periodontal Tissues Endogenous Source of Advanced Glycation End-Products (AGEs) in Individuals with and without Diabetes Mellitus? A Systematic Review. Biomolecules. 2022 Apr 27;12(5):642.

Choudhury, P., et al. Microorganisms of maternal periodontitis cause adverse pregnancy outcomes in gestational diabetic individuals: a preliminary observational report. Quintessence Int. 2022 Oct 21;53(10):850-857.

Cobb, C.M., et al. The oral microbiome and adverse pregnancy outcomes. Int J Womens Health. 2017 Aug 8;9:551-559.

Czerniuk, M.R., et al. Unexpected Relationships: Periodontal Diseases: Atherosclerosis-Plaque Destabilization? From the Teeth to a Coronary Event. Biology (Basel). 2022 Feb 9;11(2):272.

Fan, T., et al. Study on the effect of periodontitis on renal tissue in atherosclerotic mice. J Periodontal Res. 2023 Jun;58(3):655-667.

Jia, S., et al. Host insulin resistance caused by Porphyromonas gingivalis-review of recent progresses. Front Cell Infect Microbiol. 2023 Jul 13;13:1209381.

Joshi, C., et al. Myocardial infarction risk is increased by periodontal pathobionts: a cross-sectional study. Sci Rep. 2022 Nov 3;12(1):18608.

Nagasawa, Y., et al. Relationship between IgA Nephropathy and Porphyromonas gingivalis; Red Complex of Periodontopathic Bacterial Species. Int J Mol Sci. 2021 Dec 1;22(23):13022.

Olsen, I., et al. Porphyromonas gingivalis infection may contribute to systemic and intracerebral amyloid-beta: implications for Alzheimer’s disease onset. Expert Rev Anti Infect Ther. 2020 Nov;18(11):1063-1066.

Pavlic, V., et al. Identification of Periopathogens in Atheromatous Plaques Obtained from Carotid and Coronary Arteries. Biomed Res Int. 2021 Jun 17;2021:9986375.

Rahamat-Langendoen, J.C., et al. Brain abscess associated with Aggregatibacter actinomycetemcomitans: case report and review of literature. J Clin Periodontol. 2011 Aug;38(8):702-6.

Ricciardi, R.M., et al. Risk Factors and Immunoinflammatory Mechanisms Leading to Atherosclerosis: Focus on the Role of Oral Microbiota Dysbiosis. Microorganisms. 2023 Jun 1;11(6):1479.

Ripari, F., et al. The Role of Coconut Oil in Treating Patients Affected by Plaque-Induced Gingivitis: A Pilot Study. Eur J Dent. 2020 Oct;14(4):558-565.

Schulz, S., et al. Periodontal pathogens and their role in cardiovascular outcome. J Clin Periodontol. 2020 Feb;47(2):173-181.

Sedghi, L.M., et al. Periodontal Disease: The Good, The Bad, and The Unknown. Front Cell Infect Microbiol. 2021 Dec 7;11:766944.

Seoane, T., et al. Periodontitis and Other Risk Factors Related to Myocardial Infarction and Its Follow-Up. J Clin Med. 2022 May 6;11(9):2618.

Shapira, L., et al. Effects of Porphyromonas gingivalis on the central nervous system: activation of glial cells and exacerbation of experimental autoimmune encephalomyelitis. J Periodontol. 2002 May;73(5):511-6.

Svärd, A., et al. Antibodies against Porphyromonas gingivalis in serum and saliva and their association with rheumatoid arthritis and periodontitis. Data from two rheumatoid arthritis cohorts in Sweden. Front Immunol. 2023 May 30;14:1183194.

Tanneeru, S., et al. Evaluation of Microflora (Viral and Bacterial) in Subgingival and Placental Samples of Pregnant Women with Preeclampsia with and without Periodontal Disease: A Cross-Sectional Study. J Int Soc Prev Community Dent. 2020 Mar 10;10(2):171-176.

Woolley, J., et al. The effect of oil pulling with coconut oil to improve dental hygiene and oral health: A systematic review. Heliyon. 2020 Aug 27;6(8):e04789.

Wu, H., et al. The Periodontal Pathogen Fusobacterium nucleatum Exacerbates Alzheimer’s Pathogenesis via Specific Pathways. Front Aging Neurosci. 2022 Jun 23;14:912709.

Yamaguchi, S., et al. Associations of Dental Health With the Progression of Hippocampal Atrophy in Community-Dwelling Individuals: The Ohasama Study. Neurology. 2023 Jul 5;10.1212/WNL.0000000000207579.

ORIGINS Project

Sources & References

Ashwin, D., et al. The impact a Mediterranean Diet in the third trimester of pregnancy has on neonatal body fat percentage. J Dev Orig Health Dis. 2022 Aug;13(4):500-507.

Azimi, S., et al. Experience of primary caregivers in utilising an mHealth application for remote dental screening in preschool children. Aust Health Rev. 2023 Oct;47(5):545-552.

Azimi, S., et al. The feasibility of a digital health approach to facilitate remote dental screening among preschool children during COVID-19 and social restrictions. Int J Paediatr Dent. 2023 May;33(3):234-245.

Batty, G.D., et al. Adverse childhood experiences and adult health: The need for stronger study designs to evaluate impact. J Epidemiol Community Health. 2021 Jan 25:jech-2020-215870.

Baughman, N., et al. The Prevention of Anxiety and Depression in Early Childhood. Front Psychol. 2020 Sep 30:11:517896.

Carboni, L. Active Folate Versus Folic Acid: The Role of 5-MTHF (Methylfolate) in Human Health. Integr Med (Encinitas). 2022 Jul;21(3):36-41.

Cleary, D.B., et al. A Parent-Mediated Intervention for Newborns at Familial Likelihood of Autism: Initial Feasibility Study in the General Population. Adv Neurodev Disord. 2022;6(4):494-505.

Davis, J.A., et al. Can Positive Mindsets Be Protective Against Stress and Isolation Experienced during the COVID-19 Pandemic? A Mixed Methods Approach to Understanding Emotional Health and Wellbeing Needs of Perinatal Women. Int J Environ Res Public Health. 2021 Jun 29;18(13):6958.

Davis. J.A., et al. Perinatal Women's Perspectives of, and Engagement in, Digital Emotional Well-Being Training: Mixed Methods Study. J Med Internet Res. 2023 Oct 17:25:e46852.

Davis. J.A., et al. Time-out for well-being: A mixed methods evaluation of attitudes and likelihood to engage in different types of online emotional well-being programmes in the perinatal period. Womens Health (Lond). 2023 Jan-Dec:19:17455057231184507.

Divarka, N., et al. Effect of maternal prebiotic supplementation on human milk immunological composition: Insights from the SYMBA study.
Pediatr Allergy Immunol. 2024 Sep;35(9):e14226.

D'Vaz, N., et al. The ORIGINS Project Biobank: A Collaborative Bio Resource for Investigating the Developmental Origins of Health and Disease. Int J Environ Res Public Health. 2023 Jul 4;20(13):6297.

Elliott, C., et al. Early Moves: A protocol for a population-based prospective cohort study to establish General Movements as an early biomarker of cognitive impairment in infants. BMJ Open. 2021 Apr 9;11(4):e041695.

Finlay-Jones, A.J., et al. Comparing Web-Based Mindfulness With Loving-Kindness and Compassion Training for Promoting Well-Being in Pregnancy: Protocol for a Three-Arm Pilot Randomized Controlled Trial. JMIR Res Protoc. 2020 Oct 14;9(10):e19803.

Gamez, C., et al. Lower Cord Blood IL-17 and IL-25, but Not Other Epithelial Cell-Derived Cytokines Are Associated with Atopic Dermatitis in Infancy. Int Arch Allergy Immunol. 2021;182(6):474-478.

Gibson, L.Y., et al. Comparison of Experiences in Two Birth Cohorts Comprising Young Families with Children under Four Years during the Initial COVID-19 Lockdown in Australia and the UK: A Qualitative Study. Int J Environ Res Public Health. 2021 Aug 29;18(17):9119.

Hadlow, N.C., et al. Anti-Müllerian hormone concentration is associated with central adiposity and reproductive hormones in expectant fathers. Clin Endocrinol (Oxf). 2022 Nov;97(5):634-642.

Hagemann, E., et al. Developmental Origins of Health and Disease (DOHaD): The importance of life-course and transgenerational approaches. Paediatr Respir Rev. 2021 Dec:40:3-9.

Hood, R., et al. "Coronavirus Changed the Rules on Everything": Parent Perspectives on How the COVID-19 Pandemic Influenced Family Routines, Relationships and Technology Use in Families with Infants. Int J Environ Res Public Health. 2021 Dec 6;18(23):12865.

Hood, R., et al. "It helps and it doesn't help": maternal perspectives on how the use of smartphones and tablet computers influences parent-infant attachment. Ergonomics. 2024 Feb;67(2):148-167.

Hood, R., et al. The association of mobile touch screen device use with parent-child attachment: a systematic review. Ergonomics. 2021 Dec;64(12):1606-1622.

Hood, R., et al. 'There's good and bad': parent perspectives on the influence of mobile touch screen device use on prenatal attachment. Ergonomics. 2022 Dec;65(12):1593-1608.

Huang, R.C., et al. Feasibility of conducting an early pregnancy diet and lifestyle e-health intervention: the Pregnancy Lifestyle Activity Nutrition (PLAN) project. J Dev Orig Health Dis. 2020 Feb;11(1):58-70.

Ibilola, O., et al. Environmental (Perinatal) risk factors of ADHD in a sibling control design study. Open Access Journal of Behavioural Science & Psychology. 2021 Feb 05;4(1).

Johnson, B.Z., et al. Pediatric Burn Survivors Have Long-Term Immune Dysfunction With Diminished Vaccine Response. Front Immunol. 2020 Jul 21:11:1481.

Jois, R.S., et al. Do probiotics in pregnancy reduce the risk of group B streptococcal colonisation? J Paediatr Child Health. 2020 Sep;56(9):1468-1472.

Lai, C.L., et al. Longitudinal egg-specific regulatory T- and B-cell development: Insights from primary prevention clinical trials examining the timing of egg introduction. Allergy. 2021 May;76(5):1385-1397.

Lozinsky, A.C., et al. Study protocol of a multicentre, randomised, controlled trial evaluating the effectiveness of probiotic and peanut oral immunotherapy (PPOIT) in inducing desensitisation or tolerance in children with peanut allergy compared with oral immunotherapy (OIT) alone and with placebo (the PPOIT-003 study). BMJ Open. 2020 Sep 9;10(9):e035871.

Meldrum, S.J., et al. Do infants of breast-feeding mothers benefit from additional long-chain PUFA from fish oil? A 6-year follow-up. Br J Nutr. 2020 Oct 14;124(7):701-708.

Nguyen, L.D.., et al. Women's holistic self-care behaviors during pregnancy and associations with psychological well-being: implications for maternal care facilities. BMC Pregnancy Childbirth. 2022 Aug 9;22(1):631.

Palmer, D.J., et al. Feasibility and safety of introducing cashew nut spread in infant diets-A randomized trial. Pediatr Allergy Immunol. 2023 Jun;34(6):e13969.

Palmer, D.J., et al. Maternal peanut and egg consumption during breastfeeding randomized pilot trial. Pediatr Allergy Immunol. 2022 Sep;33(9):e13845.

Parkin, K.,, et al. Risk Factors for Gut Dysbiosis in Early Life. Microorganisms. 2021 Sep 30;9(10):2066.

Pedrick-Case, R., et al. Built Environments And Child Health in WalEs and AuStralia (BEACHES): a study protocol. BMJ Open. 2022 Oct 25;12(10):e061978.

Pettigrew, S., et al. Health-related behaviours and weight status of expectant fathers. Aust N Z J Public Health. 2022 Jun;46(3):275-280.

Prescott, S.L. Allergy as a sentinel measure of planetary health and biodiversity loss. Allergy. 2020 Sep;75(9):2358-2360.

Rowley, C.E., et al. Altered dietary behaviour during pregnancy impacts systemic metabolic phenotypes. Front Nutr. 2023 Dec 4:10:1230480.

Rueter, K., et al. In "High-Risk" Infants with Sufficient Vitamin D Status at Birth, Infant Vitamin D Supplementation Had No Effect on Allergy Outcomes: A Randomized Controlled Trial. Nutrients. 2020 Jun 11;12(6):1747.

Sakalidis, V.S., et al. Wellbeing of breastfeeding women in Australia and New Zealand during the COVID-19 Pandemic: A cross-sectional study. Nutrients. 2021 May 27;13(6):1831.

Saw, C., et al. Service evaluation and retrospective audit of electronic HEEADSSS (e-HEEADSSS) screening device in paediatric inpatient service in Western Australia. Int J Adolesc Med Health. 2020 Sep 22;34(6):401-409.

Silva, D.T., et al. Introducing the ORIGINS project: a community-based interventional birth cohort. Rev Environ Health. 2020 Sep 25;35(3):281-293.

Stinson, L.F., et al. Human Milk From Atopic Mothers Has Lower Levels of Short Chain Fatty Acids. Front Immunol. 2020 Jul 21:11:1427.

Wabnitz, K.J., et al. A pledge for planetary health to unite health professionals in the Anthropocene. Lancet. 2020 Nov 7;396(10261):1471-1473.

Wilcox, J.C., et al. Evaluating Engagement in a Digital and Dietetic Intervention Promoting Healthy Weight Gain in Pregnancy: Mixed Methods Study. J Med Internet Res. 2020 Jun 26;22(6):e17845.

Paleo/Primal Diet

Sources & References

Abbott, R.D., et al. Efficacy of the Autoimmune Protocol Diet as Part of a Multi-disciplinary, Supported Lifestyle Intervention for Hashimoto's Thyroiditis. Cureus. 2019 Apr 27;11(4):e4556.

Bisht, B., et al. Multimodal intervention improves fatigue and quality of life in subjects with progressive multiple sclerosis: a pilot study. Degener Neurol Neuromuscul Dis. 2015;5:19-35.

Bourre, J.M. Effects of nutrients (in food) on the structure and function of the nervous system: update on dietary requirements for brain. Part 1: micronutrients. J Nutr Health Aging. 2006 Sep-Oct;10(5):377-85.

Bourre, J.M. Effects of nutrients (in food) on the structure and function of the nervous system: update on dietary requirements for brain. Part 2 : macronutrients. J Nutr Health Aging. 2006 Sep-Oct;10(5):386-99.

Chenard, C.A., et al. Nutrient Composition Comparison between a Modified Paleolithic Diet for Multiple Sclerosis and the Recommended Healthy U.S.-Style Eating Pattern. Nutrients. 2019 Mar 1;11(3):537.

Clemens, Z., et al. Childhood absence epilepsy successfully treated with the paleolithic ketogenic diet. Neurol Ther. 2013 Sep 21;2(1-2):71-6.

Cordian, L. Cereal grains: humanity's double-edged sword. World Rev Nutr Diet. 1999;84:19-73.

Eaton, S.B., et al. Paleolithic nutrition. A consideration of its nature and current implications. N Engl J Med. 1985 Jan 31;312(5):283-9.

Haskey, N., et al. An Examination of Diet for the Maintenance of Remission in Inflammatory Bowel Disease. Nutrients. 2017 Mar 10;9(3):259.

Hsieh, M., et al. Nutritional and dietary strategy in the clinical care of inflammatory bowel disease. J Formos Med Assoc. 2020 Dec;119(12):1742-1749.

Irish, A.K., et al. Randomized control trial evaluation of a modified Paleolithic dietary intervention in the treatment of relapsing-remitting multiple sclerosis: a pilot study. Degener Neurol Neuromuscul Dis. 2017 Jan 4;7:1-18.

Konijeti, G.G., et al. Efficacy of the Autoimmune Protocol Diet for Inflammatory Bowel Disease. Inflamm Bowel Dis. 2017 Nov; 23(11): 2054–2060.

Lavie, M., et al. Paleolithic diet during pregnancy-A potential beneficial effect on metabolic indices and birth weight. Eur J Obstet Gynecol Reprod Biol. 2019 Nov;242:7-11.

Masharani, U., et al. Metabolic and physiologic effects from consuming a hunter-gatherer (Paleolithic)-type diet in type 2 diabetes. Eur J Clin Nutr. 2015 Aug;69(8):944-8.

Ogle, K.A., et al. Children with allergic rhinitis and/or bronchial asthma treated with elimination diet: a five-year follow-up. Ann Allergy. 1980 May;44(5):273.

Otten, J., et al. Benefits of a Paleolithic diet with and without supervised exercise on fat mass, insulin sensitivity, and glycemic control: a randomized controlled trial in individuals with type 2 diabetes. Diabetes Metab Res Rev. 2017 Jan;33(1):10.1002/dmrr.2828.

Sohouli, M.H., et al. The effect of paleolithic diet on glucose metabolism and lipid profile among patients with metabolic disorders: a systematic review and meta-analysis of randomized controlled trials. Crit Rev Food Sci Nutr. 2022;62(17):4551-4562.

Tóth, C., et al. Type 1 diabetes mellitus successfully managed with the paleolithic ketogenic diet. Int J Case Rep Images. 2014;5(10):699–703.

Wahls, T.L. Dietary Approaches to Treating Multiple Sclerosis-Related Symptoms. Phys Med Rehabil Clin N Am. 2022 Aug;33(3):605-620.

Wahls, T.L., et al. Dietary approaches to treat MS-related fatigue: comparing the modified Paleolithic (Wahls Elimination) and low saturated fat (Swank) diets on perceived fatigue in persons with relapsing-remitting multiple sclerosis: study protocol for a randomized controlled trial.Trials. 2018 Jun 4;19(1):309.

Whalen, K.A., et al. Paleolithic and Mediterranean Diet Pattern Scores Are Inversely Associated with All-Cause and Cause-Specific Mortality in Adults. J Nutr. 2017 Apr;147(4):612-620.

PANS/PANDAS

Sources & References

Aldad, T.S., et al. Fetal Radiofrequency Radiation Exposure From 800-1900 Mhz-Rated Cellular Telephones Affects Neurodevelopment and Behavior in Mice. Sci Rep. 2012;2:312.

Allen, A.J., et al. Case study: a new infection-triggered, autoimmune subtype of pediatric OCD and Tourette's syndrome. J Am Acad Child Adolesc Psychiatry. 1995 Mar;34(3):307-11.

Arnold, P.D., et al. Is obsessive-compulsive disorder an autoimmune disease? Canadian Medical Association Journal. 2001 Nov 13;165(10):1353-8.

Baj, J., et al. Alterations in the Nervous System and Gut Microbiota after β-Hemolytic Streptococcus Group A Infection-Characteristics and Diagnostic Criteria of PANDAS Recognition. Int J Mol Sci. 2020 Feb 21;21(4):1476.

Banks, W.A., et al. Aluminum-induced neurotoxicity: alterations in membrane function at the blood-brain barrier. Neurosci Biobehav Rev. 1989 Spring;13(1):47-53.

Baytunca, M.B. [Evaluation of a Neuropsychiatric Disorder: From PANDAS to PANS and CANS]. Turk Psikiyatri Derg. 2016 Summer;27(2):0.

Bitnun, A., et al. Acute Childhood Encephalitis and Mycoplasma pneumoniae. Clin Infect Dis. 2001 Jun 15;32(12):1674-84.

Bjarnason, I., et al. Mechanisms of Damage to the Gastrointestinal Tract From Nonsteroidal Anti-Inflammatory Drugs. Gastroenterology. 2018 Feb;154(3):500-514.

Bloch, M.H., et al. N-Acetylcysteine in the Treatment of Pediatric Tourette Syndrome: Randomized, Double-Blind, Placebo-Controlled Add-On Trial. J Child Adolesc Psychopharmacol. 2016 May;26(4):327-34.

Boileau, B. A review of obsessive-compulsive disorder in children and adolescents. Dialogues Clin Neurosci. 2011;13(4):401-11.

Bora, S.A., et al. Regulation of vitamin D metabolism following disruption of the microbiota using broad spectrum antibiotics. J Nutr Biochem. 2018 Jun;56:65-73.

Boris, M.J., et al. Pollen Exposure as a Cause for the Deterioration of Neurobehavioral Function in Children with Autism and Attention Deficit Hyperactive Disorder. J of Nutritional and Environmental Medicine. Mar 2004;14(1):47 – 54.

Braniste, V., et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci Transl Med. 2014 Nov 19;6(263):263ra158.

Bransfield, R.C., et al. The association between tick-borne infections, Lyme borreliosis and autism spectrum disorders. Medical Hypotheses. 2008;70(5):967-74.

Bravo, J.A., et al. Ingestion of Lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A. 2011 Sep 20;108(38):16050-5.

Brown, K., et al. Pediatric Acute-Onset Neuropsychiatric Syndrome Response to Oral Corticosteroid Bursts: An Observational Study of Patients in an Academic Community-Based PANS Clinic. J Child Adolesc Psychopharmacol. 2017 Jul 17.

Brown, K.D., et al. Effect of Early and Prophylactic Nonsteroidal Anti-Inflammatory Drugs on Flare Duration in Pediatric Acute-Onset Neuropsychiatric Syndrome: An Observational Study of Patients Followed by an Academic Community-Based Pediatric Acute-Onset Neuropsychiatric Syndrome Clinic. J Child Adolesc Psychopharmacol. 2017 Jul 11.

Butel, M.J., et al. The developing gut microbiota and its consequences for health. J Dev Orig Health Dis. 2018 Mar 22:1-8.

Burkett, P.R., et al. Pouring fuel on the fire: Th17 cells, the environment, and autoimmunity. J Clin Invest. 2015 Jun;125(6):2211-9.

Calaprice, D., et al. A Survey of Pediatric Acute-Onset Neuropsychiatric Syndrome Characteristics and Course. J Child Adolesc Psychopharmacol. 2017 Jan 31.

Carpentier, A., et al. Clinical trial of blood-brain barrier disruption by pulsed ultrasound. Sci Transl Med. 2016 Jun 15;8(343):343re2.

Chang, K., et al. Clinical evaluation of youth with pediatric acute–onset neuropsychiatric syndrome (PANS): recommendations from the 2013 PANS Consensus Conference. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):3-13.

Chiarello, F., et al. An expert opinion on PANDAS/PANS: highlights and controversies. Int J Psychiatry Clin Pract. 2017 Jun;21(2):91-98.

Connolly, A.M., et al. Serum autoantibodies to brain in Landau-Kleffner variant, autism, and other neurologic disorders. The Journal of Pediatrics. 1999 May;134(5):607-13.

Cooperstock, M., et al. Clinical Management of Pediatric Acute-Onset Neuropsychiatric Syndrome: Part III—Treatment and Prevention of Infections. J Child Adolesc Psychopharmacol. 2017 Jul, ahead of print.

Cosford, Robyn. Occult Infections, Streptococcus, Biofilms, PANDAS and MINDDD in Autism. Defeat Autism Now! Conference, San Diego, CA, Oct 2008.

Cox, C.J., et al. Antineuronal antibodies in a heterogeneous group of youth and young adults with tics and obsessive-compulsive disorder. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):76-85.

Dahm, T., et al. Neuroinvasion and Inflammation in Viral Central Nervous System Infections. Mediators Inflamm. 2016;2016:8562805.

Dale, R.C., et al. Encephalitis lethargica syndrome: 20 new cases and evidence of basal ganglia autoimmunity. Brain. 2004 Jan;127(Pt 1):21-33.

Dale, R.C. Immune-mediated extrapyramidal movement disorders, including Sydenham chorea. Handb Clin Neurol. 2013;112:1235-41.

Dallasta, L.M., et al. Blood-Brain Barrier Tight Junction Disruption in Human Immunodeficiency Virus-1 Encephalitis. Am J Pathol. 1999 Dec;155(6):1915-27.

Das, T., et al. Influence of Calcium in Extracellular DNA Mediated Bacterial Aggregation and Biofilm Formation. PLoS One. 2014 Mar 20;9(3):e91935.

Dasdaq, S., et al. Effects of 2.4 GHz radiofrequency radiation emitted from Wi-Fi equipment on microRNA expression in brain tissue. Int J Radiat Biol. 2015 Jul;91(7):555-61.

DeLong, G.R., et al. Acquired reversible autistic syndrome in acute encephalopathic illness in children. Arch Neurol. 1981 Mar;38(3):191-4.

Ding, H., et al. Molecular Pathogenesis of Anti-NMDAR Encephalitis. Biomed Res Int. 2015;2015:643409.

Ercan, T.E., et al. Mycoplasma pneumoniae infection and obsessive-compulsive disease: a case report. J Child Neurol. 2008 Mar;23(3):338-40.

Endres, D., et al. Immunological causes of obsessive-compulsive disorder: is it time for the concept of an “autoimmune OCD” subtype? Translational Psychiatry. 12, Article number: 5 (2022).

Egset, K., et al. Magno App: Exploring Visual Processing in Adults with High and Low Reading Competence. Scandinavian Journal of Educational Research. 07 Jan 2020.

Feng, J., et al. Evaluation of Natural and Botanical Medicines for Activity Against Growing and Non-growing Forms of B. burgdorferi. Front. Med., 21 Feb 2020.

Feng, P., et al. A review of probiotics in the treatment of autism spectrum disorders: Perspectives from the gut-brain axis. Front Microbiol. 2023 Mar 16:14:1123462.

Ferretti, J.W., et al. Post-Streptococcal Autoimmune Sequelae: Rheumatic Fever and Beyond. Streptococcus pyogenes : Basic Biology to Clinical Manifestations [Internet]. 2016 Feb 10. Oklahoma City (OK): University of Oklahoma Health Sciences Center.

Fiorentino, M., et al. Blood–brain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Molecular Autism. 2016 Nov 29;7:49.

Frankovich, J., et al. Clinical Management of Pediatric Acute-Onset Neuropsychiatric Syndrome: Part II—Use of Immunomodulatory Therapies. J Child Adolesc Psychopharmacol. 2017 Jul

Frankovich, J., et al. Five youth with pediatric acute-onset neuropsychiatric syndrome of differing etiologies. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):31-7.

Frankovich, J., et al. Multidisciplinary clinic dedicated to treating youth with pediatric acute-onset neuropsychiatric syndrome: presenting characteristics of the first 47 consecutive patients. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):38-47.

Gabbay, V., et al. A Double-Blind, Placebo-Controlled Trial of Omega-3 Fatty Acids in Tourette’s Disorder. Pediatrics. 2012 Jun; 129(6): e1493–e1500.

Gaughan, T., et al. Rapid Eye Movement Sleep Abnormalities in Children with Pediatric Acute-Onset Neuropsychiatric Syndrome (PANS). J Clin Sleep Med. 2016 Jul 15;12(7):1027-32.

Gerardi, D.M., et al. PANDAS and comorbid Kleine-Levin syndrome. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):93-8.

Gerentes, M., et al. Obsessive-Compulsive Disorder: Autoimmunity and Neuroinflammation. Curr Psychiatry Rep. 2019 Aug 1;21(8):78.

Ghaziuddin, M., et al. Brief report: autism and herpes simplex encephalitis. J Autism Dev Disord. 1992 Mar;22(1):107-13.

Gillberg, C. Onset at age 14 of a typical autistic syndrome. A case report of a girl with herpes simplex encephalitis. J Autism Dev Disord. 1986 Sep;16(3):369-75.

Goncalves, M.V.M., et al. Pediatric acute-onset neuropsychiatric syndrome (PANS) misdiagnosed as autism spectrum disorder. Immunol Lett. 2018 Nov;203:52-53.

Greer, M.K., et al. A case study of the cognitive and behavioral deficits of temporal lobe damage in herpes simplex encephalitis. J Autism Dev Disord. 1989 Jun;19(2):317-26.

Gulati, G., et al. Anti-NR2 antibodies, blood-brain barrier, and cognitive dysfunction. Clin Rheumatol. 2016 Dec;35(12):2989-2997.

Hacohen, Y., et al. N‐methyl‐d‐aspartate (NMDA) receptor antibodies encephalitis mimicking an autistic regression. Dev Med Child Neurol. 2016 Oct;58(10):1092-4.

Hadjivassiliou, M., et al. Gluten sensitivity: from gut to brain. Lancet Neurol. 2010 Mar;9(3):318-30.

Havas, M. Radiation from wireless technology affects the blood, the heart, and the autonomic nervous system. Rev Environ Health. 2013;28(2-3):75-84.

Hazlett, H.C., et al. Early brain development in infants at high risk for autism spectrum disorder. Nature. 2017 Feb 16;542:348–351.

Herbert, M.R., et al. Autism and EMF? Plausibility of a pathophysiological link part I. Pathophysiology. 2013 Jul;1-19.

Herbert, M.R., et al. Autism and EMF? Plausibility of a pathophysiological link part II. Pathophysiology. 2013 Jun;20(3):211-34.

Hirai, N., et al. A new infectious encephalopathy syndrome, clinically mild encephalopathy associated with excitotoxicity (MEEX). J Neurol Sci. 2017 Sep 15;380:27-30.

Hou, J., et al. Viral interactions with the blood-brain barrier: old dog, new tricks. Tissue Barriers. 2016 Jan 28;4(1):e1142492.

Hsu, C.J., et al. Immunological Dysfunction in Tourette Syndrome and Related Disorders. Int J Mol Sci. 2021 Jan 16;22(2):853.

Ivanovski, I., et al. Aluminum in brain tissue in autism. J Trace Elem Med Biol. 2019 Jan;51:138-140.

Jain, R., et al. The effect of medical treatments on the bacterial microbiome in patients with chronic rhinosinusitis: a pilot study. Int Forum Allergy Rhinol. 2018 Mar 8.

Jaspers-Fayer, F., et al. Prevalence of Acute-Onset Subtypes in Pediatric Obsessive-Compulsive Disorder. J Child Adolesc Psychopharmacol. 2017 May;27(4):332-341.

Johansson, O. Disturbance of the immune system by electromagnetic fields—A potentially underlying cause for cellular damage and tissue repair reduction which could lead to disease and impairment. Pathophysiology. 2009 Aug;16(2-3):157-177.

Jyonouchi, H., et al. Cytokine profiles by peripheral blood monocytes are associated with changes in behavioral symptoms following immune insults in a subset of ASD subjects: an inflammatory subtype? J Neuroinflammation. 2014 Oct 27;11:187.

Kantarcioglu, A.S., et al. Microbiota-Gut-Brain Axis: Yeast Species Isolated from Stool Samples of Children with Suspected or Diagnosed Autism Spectrum Disorders and In Vitro Susceptibility Against Nystatin and Fluconazole. Mycopathologia. 2016 Feb;181(1-2):1-7.

Kelly, J.R., et al. Breaking down the barriers: the gut microbiome, intestinal permeability and stress-related psychiatric disorders. Front Cell Neurosci. 2015 Oct 14;9:392.

Khan, Z., et al. Slow CCL2-dependent translocation of biopersistent particles from muscle to brain. BMC Med. 2013 Apr 4;11:99.

Kim, C., et al. Strep and Urinary Frequency: Is There a Connection? New England Section of the American Urological Association, 75th Annual Meeting. 2006 Sep 28-30.

Kim., Y., et al. Obsessive-Compulsive Disorder Related to Mycoplasma-Associated Autoimmune Encephalopathy with Basal Ganglia Involvement. Child Adolesc Psychopharmacol. 2016 May;26(4):400-2.

King, L., et al. Intuition: a critical review of the research and rhetoric. J Adv Nurs. 1997 Jul;26(1):194-202.

Kinoshita, M., et al. Targeted delivery of antibodies through the blood-brain barrier by MRI-guided focused ultrasound. Biochem Biophys Res Commun. 2006 Feb 24;340(4):1085-90.

Kirvan, C.A., et al. Streptococcal mimicry and antibody-mediated cell signaling in the pathogenesis of Sydenham's chorea. Autoimmunity. 2006 Feb;39(1):21-9.

Kobliner, V., et al. Reduction in Obsessive Compulsive Disorder and Self-Injurious Behavior With Saccharomyces boulardii in a Child with Autism: A Case Report. Integr Med (Encinitas). 2018 Dec;17(6):38-41.

Koffie, R.M., et al. Nanoparticles enhance brain delivery of blood–brain barrier-impermeable probes for in vivo optical and magnetic resonance imaging. Proc Natl Acad Sci U S A. 2011 Nov 15;108(46):18837-42.

Kovacevic, M., et al. Use of Intravenous Immunoglobulin in the Treatment of Twelve Youths with Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal infections. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):65-9.

Kurlan, R. Tourette's syndrome and 'PANDAS': will the relation bear out? Pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection. Neurology. 1998 Jun;50(6):1530-4.

Lancaster, E. The Diagnosis and Treatment of Autoimmune Encephalitis. J Clin Neurol. 2016 Jan;12(1):1-13.

Latimer, M.E., et al. Therapeutic plasma apheresis as a treatment for 35 severely ill children and adolescents with pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):70-5.

Leclercq, S., et al. Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior. Nature Communications. 2017; 8.

Lionetti, E., et al. Gluten Psychosis: Confirmation of a New Clinical Entity. Nutrients. 2015 Jul 8;7(7):5532-9.

López-Aranda, M.F., et al. Postnatal immune activation causes social deficits in a mouse model of tuberous sclerosis: Role of microglia and clinical implications. Sci Adv. 2021 Sep 17;7(38):eabf2073.

Ludlow, A.K., et al. Understanding the impact of diet and nutrition on symptoms of Tourette syndrome: A scoping review. J Child Health Care. 2018 Mar;22(1):68-83.

Mahony, T., et al. Improvement of psychiatric symptoms in youth following resolution of sinusitis. Int J Pediatr Otorhinolaryngol. 2017 Jan;92:38-44.

Mahony, T., et al. Palatal Petechiae in the Absence of Group A Streptococcus in Pediatric Patients with Acute-Onset Neuropsychiatric Deterioration: A Cohort Study. J Child Adolesc Psychopharmacol. 2017 Apr 7.

Marcello, A., et al. Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections (PANDAS): An Evolving Concept. Tremor Other Hyperkinet Mov (N Y). 2013 Sep 25;3. pii: tre-03-167-4158-7.

Marquet, F., et al. Noninvasive, Transient and Selective Blood-Brain Barrier Opening in Non-Human Primates In Vivo. PLoS One. 2011;6(7):e22598.

Masand, P.S., et al. Prevalence of irritable bowel syndrome in obsessive-compulsive disorder. CNS Spectr. 2006 Jan;11(1):21-5.

Mason, R. Expanding Diagnostic Vision with Medical Intuition. Alternative and Complementary Therapies. 2009 Mar 19; 6(6).

Mink, J., et al. Acute postinfectious movement and psychiatric disorders in children and adolescents. J Child Neurol. 2011 Feb;26(2):214-7.

Moretti, G., et al. What every psychiatrist should know about PANDAS: a review. Clinical Practice and Epidemiology in Mental Health. 2008 May 21;4:13.

Morris, C.R., et al. Syndrome of allergy, apraxia, and malabsorption: characterization of a neurodevelopmental phenotype that responds to omega 3 and vitamin E supplementation. Alternative Therapies in Health and Medicine. Jul-Aug 2009;15(4):34-43.

Müller-Vahl, K.R., et al. The influence of different food and drink on tics in Tourette syndrome. Acta Paediatr. 2008 Apr;97(4):442-6.

Muir, K.E., et al. A case report of obsessive-compulsive disorder following acute disseminated encephalomyelitis. Pediatrics. 2013 Sep;132(3):e771-4.

Murphy, T.K., et al. Cefdinir for recent-onset pediatric neuropsychiatric disorders: a pilot randomized trial. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):57-64.

Murphy, T.K., et al. Characterization of the pediatric acute-onset neuropsychiatric syndrome phenotype. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):14-25

Murphy, T.K., et al. A Double-Blind Randomized Placebo-Controlled Pilot Study of Azithromycin in Youth with Acute-Onset Obsessive-Compulsive Disorder. J Child Adolesc Psychopharmacol. 2017 Mar 30.

Murphy, T.K., et al. Pediatric acute-onset neuropsychiatric syndrome. Psychiatr Clin North Am. 2014 Sep;37(3):353-74.

Nadeau, J.M., et al. A pilot trial of cognitive-behavioral therapy augmentation of antibiotic treatment in youth with pediatric acute-onset neuropsychiatric syndrome-related obsessive-compulsive disorder. J Child Adolesc Psychopharmacol. 2015 May;25(4):337-43.

Nicolson, G.L. Chronic Bacterial and Viral Infections in Neurodegenerative and Neurobehavioral Diseases. Lab Medicine. 2008;39(5):291-299.

Nicolson, G.L., et al. Chronic Mycoplasmal Infections in Autism Patients. Proc. Intern. Mind of a Child Conference, Sydney, Australia 2002.

Nicolson, G.L., et al. Evidence for Mycoplasma ssp., Chlamydia pneunomiae, and human herpes virus-6 coinfections in the blood of patients with autistic spectrum disorders. J Neurosci Res. 2007 Apr;85(5):1143-8.

Onalapo, A.Y., et al. Dietary glutamate and the brain: In the footprints of a Jekyll and Hyde molecule. Neurotoxicology. 2020 Sep;80:93-104.

Orefici, G., et al. Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections (PANDAS). Streptococcus pyogenes: Basic Biology to Clinical Manifestations [Internet]. Oklahoma City (OK): University of Oklahoma Health Sciences Center; 2016-. 2016 Feb 10.

Orlovska, S., et al. Association of Streptococcal Throat Infection With Mental Disorders: Testing Key Aspects of the PANDAS Hypothesis in a Nationwide Study. JAMA Psychiatry. 2017 Jul 1;74(7):740-746.

Pall, M.L. Microwave frequency electromagnetic fields (EMFs) produce widespread neuropsychiatric effects including depression. J Chem Neuroanat. 2016 Sep;75(Pt B):43-51.

Parker-Athill, E.C., et al. Cytokine correlations in youth with tic disorders. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):86-92.

Pavone, P., et al. Anti-brain antibodies in PANDAS versus uncomplicated streptococcal infection. Pediatr Neurol. 2004 Feb;30(2):107-10.

Pearlman, D.M., et al. Anti-basal ganglia antibodies in primary obsessive-compulsive disorder: systematic review and meta-analysis. Br J Psychiatry. 2014 Jul;205(1):8-16.

Perlmutter, S.J., et al. Therapeutic plasma exchange and intravenous immunoglobulin for obsessive-compulsive disorder and tic disorders in childhood. Lancet. 1999 Oct 2;354(9185):1153-8.

Pihtili, A., et al. Evidence for the Efficacy of a Bioresonance Method in Smoking Cessation: A Pilot Study. Forsch Komplementmed. 2014;21(4):239-45.

Plioplys, A.V., et al. Anti-CNS antibodies in childhood neurologic diseases. Neuropediatrics. 1989 May;20(2):93-102.

Rawls, W.E., et al. Encephalitis associated with herpes simplex virus. Ann Intern Med. 1966 Jan;64(1):104-15.

Rodrigo, L., et al. Tourette Syndrome and Non-Coeliac Gluten Sensitivity. Clinical Remission with a Gluten-Free Diet: A Description Case. Journal of Sleep Disorders and Therapy. 2015.

Sahyouni, R., et al. Effects of concussion on the blood–brain barrier in humans and rodents. Journal of Concussion. 2017 Jan 1; 10.1177/2059700216684518

Sai, Y., et al. Clinical diagnosis and treatment of pediatric anti-N-methyl-D-aspartate receptor encephalitis: A single center retrospective study. Exp Ther Med. 2018 Aug;16(2):1442-1448.

Samsel, A., et al. Glyphosate, pathways to modern diseases III: Manganese, neurological diseases, and associated pathologies. Surg Neurol Int. 2015 Mar 24;6:45.

Sifra, S., et al. Treatment of PANDAS and PANS: a systematic review. Neuroscience & Biobehavioral Reviews. 2018 Mar;86:51-65.

Singer, H.S., et al. Moving from PANDAS to CANS. The Journal of Pediatrics. 2012 May; 160(5):725-731.

Song, Y., et al. Effects of acute exposure to aluminum on blood-brain barrier and the protection of zinc. Neurosci Lett. 2008 Nov 7;445(1):42-6.

Spartz, E.J., et al. Course of Neuropsychiatric Symptoms After Introduction and Removal of Nonsteroidal Anti-Inflammatory Drugs: A Pediatric Observational Study. J Child Adolesc Psychopharmacol. 2017 Jul 11.

Spindler, K.R., et al. Viral disruption of the blood-brain barrier. Trends Microbiol. 2012 Jun;20(6):282-90.

Swedo, S.E., et al. Clinical Presentation of Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal infections in Research and Community Settings. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):26-30

Swedo, S.E., et al. Identification of children with pediatric autoimmune neuropsychiatric disorders associated with streptococcal infections by a marker associated with rheumatic fever. Am J Psychiatry. 1997 Jan;154(1):110-2.

Swedo, S.E., et al. Overview of Treatment of Pediatric Acute-Onset Neuropsychiatric Syndrome. J Child Adolesc Psychopharmacol. 2017 Jul 19.

Tanaka, S., et al. Autoantibodies against four kinds of neurotransmitter receptors in psychiatric disorders. J Neuroimmunol. 2003 Aug;141(1-2):155-64.

Tang, J., et al. Exposure to 900 MHz electromagnetic fields activates the mkp-1/ERK pathway and causes blood-brain barrier damage and cognitive impairment in rats. Brain Res. 2015 Mar 19;1601:92-101.

Theoharides, T.C., et al. Neuro-inflammation, blood-brain barrier, seizures and autism. J Neuroinflammation. 2011 Nov 30;8:168.

Thienemann M., et al. Clinical Management of Pediatric Acute-Onset Neuropsychiatric Syndrome: Part I-Psychiatric and Behavioral Interventions. J Child Adolesc Psychopharmacol. 2017 Jul 19.

Tohidpour, A., et al. Neuroinflammation and Infection: Molecular Mechanisms Associated with Dysfunction of Neurovascular Unit. Front Cell Infect Microbiol. 2017 Jun 20;7:276.

Tona, J.T., et al. Impact of PANS and PANDAS Exacerbations on Occupational Performance: A Mixed-Methods Study. Am J Occup Ther. 2017 May/Jun;71(3):7103220020P1-7103220020P9.

Toufexis, M.D., et al. Disordered eating and food restrictions in children with PANDAS/PANS. J Child Adolesc Psychopharmacol. 2015 Feb;25(1):48-56.

Tuchscherr, L., et al. Staphylococcus aureus phenotype switching: an effective bacterial strategy to escape host immune response and establish a chronic infection. EMBO Mol Med. 2011 Mar;3(3):129-41.

Venâncio, P., et al. Anti-N-methyl-D-aspartate receptor encephalitis with positive serum antithyroid antibodies, IgM antibodies against mycoplasma pneumoniae and human herpesvirus 7 PCR in the CSF. Pediatr Infect Dis J. 2014 Aug;33(8):882-3.

Vitaliti, G., et al. A new clinical feature associated with familial early-onset of dystonic-guttural tics: An unusual diagnosis of PANDAS. J Pediatr Neurosci. 2014 Jan;9(1):79-81.

Warner, B.B. The contribution of the gut microbiome to neurodevelopment and neuropsychiatric disorders. Pediatr Res. 2019 Jan;85(2):216-224.

Xu, C.L., et al. Anti-N-methyl-D-aspartate receptor encephalitis with serum anti-thyroid antibodies and IgM antibodies against Epstein-Barr virus viral capsid antigen: a case report and one year follow-up. BMC Neurol. 2011 Nov 29;11:149.

Xu, J., et al. Antibodies From Children With PANDAS Bind Specifically to Striatal Cholinergic Interneurons and Alter Their Activity. Am J Psychiatry. 2021 Jan 1;178(1):48-64.

Waites, K.B., et al. Mycoplasma pneumoniae and its role as a human pathogen. Clin Microbiol Rev. 2004 Oct;17(4):697-728.

Wang, H. Anti-NMDA Receptor Encephalitis. Int J Mol Sci. 2017 Jan 18;18(1). pii: E193.

Wells, L., et al. Folate Receptor Alpha Autoantibodies in the Pediatric Acute-Onset Neuropsychiatric Syndrome (PANS) and Pediatric Autoimmune Neuropsychiatric Disorders Associated with Streptococcal Infections (PANDAS) Population. J Pers Med. 2024 Jan 31;14(2):166.

Yaddanapudi, K., et al. Passive transfer of streptococcus-induced antibodies reproduces behavioral disturbances in a mouse model of pediatric autoimmune neuropsychiatric disorders associated with streptococcal infection. Mol Psychiatry. 2010 Jul;15(7):712-26.

Zong, S., et al. Neuronal Surface Autoantibodies in Neuropsychiatric Disorders: Are There Implications for Depression? Front Immunol. 2017 Jul 5;8:752.

Prenatal & Perinatal Health

Sources & References

Alemany, S., et al. Prenatal and postnatal exposure to acetaminophen in relation to autism spectrum and attention-deficit and hyperactivity symptoms in childhood: Meta-analysis in six European population-based cohorts. Eur J Epidemiol. 2021 Oct;36(10):993-1004.

Ang, E.S.B.C., Jr., et al. Prenatal exposure to ultrasound waves impacts neuronal migration in mice. Proc Natl Acad Sci U S A. 2006 Aug 22;103(34):12903-10.

Ashwin, D., et al. The impact a Mediterranean Diet in the third trimester of pregnancy has on neonatal body fat percentage. J Dev Orig Health Dis. 2022 Aug;13(4):500-507.

Atladóttir, H.Ó., et al. Association of family history of autoimmune diseases and autism spectrum disorders. Pediatrics. 2009 Aug;124(2):687-94.

Atladóttir, H.Ó., et al. Autism after infection, febrile episodes, and antibiotic use during pregnancy: an exploratory study. Pediatrics. 2012 Dec;130(6):e1447-54.

Avella-Garcia, C.B., et al. Acetaminophen use in pregnancy and neurodevelopment: attention function and autism spectrum symptoms. Int J Epidemiol. 2016 Dec 1;45(6):1987-1996.

Baker, B.H., et al. Association of Prenatal Acetaminophen Exposure Measured in Meconium With Risk of Attention-Deficit/Hyperactivity Disorder Mediated by Frontoparietal Network Brain Connectivity. JAMA Pediatrics. 2020 Sep 28;e203080.

Bauer, A.Z., et al. Prenatal paracetamol exposure and child neurodevelopment: A review. Horm Behav. 2018 May:101:125-147.

Brandalize, A.P.C., et al. Evaluation of C677T and A1298C polymorphisms of the MTHFR gene as maternal risk factors for Down syndrome and congenital heart defects. Am J Med Genet. 2009 Oct;149A(10):2080-7.

Chaparro, C.M., et al. Effect of timing of umbilical cord clamping on iron status in Mexican infants: a randomised controlled trial. Lancet. 2006 Jun 17;367(9527):1997-2004.

Choudhury, P., et al. Microorganisms of maternal periodontitis cause adverse pregnancy outcomes in gestational diabetic individuals: a preliminary observational report. Quintessence Int. 2022 Oct 21;53(10):850-857.

Cobb, C.M., et al. The oral microbiome and adverse pregnancy outcomes. Int J Womens Health. 2017 Aug 8;9:551-559.

Curran, E.A., et al. Research review: Birth by caesarean section and development of autism spectrum disorder and attentiondeficit/hyperactivity disorder: a systematic review and meta-analysis. J Child Psychol Psychiatry. 2015 May;56(5):500-8.

Darling, A.L., et al. Association between maternal vitamin D status in pregnancy and neurodevelopmental outcomes in childhood: results from the Avon Longitudinal Study of Parents and Children (ALSPAC). Br J Nutr 2017 Jun;117(12):1682-1692.

Davis, J.A., et al. Can Positive Mindsets Be Protective Against Stress and Isolation Experienced during the COVID-19 Pandemic? A Mixed Methods Approach to Understanding Emotional Health and Wellbeing Needs of Perinatal Women. Int J Environ Res Public Health. 2021 Jun 29;18(13):6958.

Davis. J.A., et al. Perinatal Women's Perspectives of, and Engagement in, Digital Emotional Well-Being Training: Mixed Methods Study. J Med Internet Res. 2023 Oct 17:25:e46852.

Davis. J.A., et al. Time-out for well-being: A mixed methods evaluation of attitudes and likelihood to engage in different types of online emotional well-being programmes in the perinatal period. Womens Health (Lond). 2023 Jan-Dec:19:17455057231184507.

Divarka, N., et al. Effect of maternal prebiotic supplementation on human milk immunological composition: Insights from the SYMBA study.
Pediatr Allergy Immunol. 2024 Sep;35(9):e14226.

D'Vaz, N., et al. The ORIGINS Project Biobank: A Collaborative Bio Resource for Investigating the Developmental Origins of Health and Disease. Int J Environ Res Public Health. 2023 Jul 4;20(13):6297.

Fezer, G.F., et al. Perinatal Features of Children with Autism Spectrum Disorder. Rev Paul Pediatr. 2017 Apr-Jun;35(2):130-135.

Finlay-Jones, A.J., et al. Comparing Web-Based Mindfulness With Loving-Kindness and Compassion Training for Promoting Well-Being in Pregnancy: Protocol for a Three-Arm Pilot Randomized Controlled Trial. JMIR Res Protoc. 2020 Oct 14;9(10):e19803.

Frisch, M., et al. Ritual circumcision and risk of autism spectrum disorder in 0- to 9-year-old boys: national cohort study in Denmark. J R Soc Med. 2015 Jul;108(7):266-79.

Gamez, C., et al. Lower Cord Blood IL-17 and IL-25, but Not Other Epithelial Cell-Derived Cytokines Are Associated with Atopic Dermatitis in Infancy. Int Arch Allergy Immunol. 2021;182(6):474-478.

Gardender, H., et al. Perinatal and neonatal risk factors for autism: a comprehensive meta-analysis. Pediatrics. 2011 Aug;128(2):344-55.

Glasson, E.J., et al. Perinatal factors and the development of autism: a population study. Arch Gen Psychiatry. 2004 Jun;61(6):618-27.

González-Valenzuela, M.J., et al. Exposure to synthetic oxytocin during delivery and its effect on psychomotor development. Dev Psychobiol. 2015 Dec;57(8):908-20.

Hamad, A.F., et al. Prenatal antibiotics exposure and the risk of autism spectrum disorders: A population-based cohort study. PLoS One. 2019 Aug 29;14(8):e0221921.

Hansen, J.B., et al. Prenatal exposure to bisphenol A and autistic- and ADHD-related symptoms in children aged 2 and5 years from the Odense Child Cohort. Environ Health. 2021 Mar 12;20(1):24.

Hantsoo, L., et al. Childhood adversity impact on gut microbiota and inflammatory response to stress during pregnancy. Brain Behav Immun. 2019 Jan:75:240-250.

Hertz-Picciotto, I., et al. Organophosphate exposures during pregnancy and child neurodevelopment: Recommendations for essential policy reforms. PLoS Med. 2018 Oct 24;15(10):e1002671.

Hertz-Picciotto, I., et al. Prenatal exposures to persistent and non-persistent organic compounds and effects on immune system development. Basic Clin Pharmacol Toxicol. 2008 Feb;102(2):146-54.

Hobbs, C.A., et al. Polymorphisms in Genes Involved in Folate Metabolism as Maternal Risk Factors for Down Syndrome. Am J Med Genet. 2000 Sep; 67(3): 623–630.

Hood, R., et al. "It helps and it doesn't help": maternal perspectives on how the use of smartphones and tablet computers influences parent-infant attachment. Ergonomics. 2024 Feb;67(2):148-167.

Hood, R., et al. The association of mobile touch screen device use with parent-child attachment: a systematic review. Ergonomics. 2021 Dec;64(12):1606-1622.

Hood, R., et al. 'There's good and bad': parent perspectives on the influence of mobile touch screen device use on prenatal attachment. Ergonomics. 2022 Dec;65(12):1593-1608.

Huang, R.C., et al. Feasibility of conducting an early pregnancy diet and lifestyle e-health intervention: the Pregnancy Lifestyle Activity Nutrition (PLAN) project. J Dev Orig Health Dis. 2020 Feb;11(1):58-70.

Hutton, E.K., et al. Late vs early clamping of the umbilical cord in full-term neonates: systematic review and meta-analysis of controlled trials. JAMA. 2007 Mar 21;297(11):1241-52.

Ibilola, O., et al. Environmental (Perinatal) risk factors of ADHD in a sibling control design study. Open Access Journal of Behavioural Science & Psychology. 2021 Feb 05;4(1).

Jois, R.S., et al. Do probiotics in pregnancy reduce the risk of group B streptococcal colonisation? J Paediatr Child Health. 2020 Sep;56(9):1468-1472.

Konkel, L. Phthalates and Autistic Traits: Exploring the Association between Prenatal Exposures and Child Behavior. Environ Health Perspec. 2020 Oct;128(10):104001.

Korpela, K., et al. Maternal Fecal Microbiota Transplantation in Cesarean-Born Infants Rapidly Restores Normal Gut Microbial Development: A Proof-of-Concept Study. Cell, 2020.

Kurth, L., et al. Perinatal Pitocin as an early ADHD biomarker: neurodevelopmental risk? J Atten Disord. 2011 Jul;15(5):423-31.

Langley, E.A., et al. High maternal choline consumption during pregnancy and nursing alleviates deficits in social interaction and improves anxiety-like behaviors in the BTBR T+Itpr3tf/J mouse model of autism. Behav Brain Res. 2015 Feb 1;278:210-20.

Lavie, M., et al. Paleolithic diet during pregnancy-A potential beneficial effect on metabolic indices and birth weight. Eur J Obstet Gynecol Reprod Biol. 2019 Nov;242:7-11.

Li, D.K., et al. A population-based prospective cohort study of personal exposure to magnetic fields during pregnancy and the risk of miscarriage. Epidemiology. 2002 Jan;13(1):9-20.

Lu, X., et al. Azithromycin exposure during pregnancy disturbs the fetal development and its characteristic of multi-organ toxicity. Life Sci. 2023 Sep 15:329:121985.

Lueth, A.J. Allostatic Load and Adverse Pregnancy Outcomes. Obstet Gynecol. 2022 Dec 1;140(6):974-982.

Lyall, K., et al. Prenatal Serum Concentrations of Brominated Flame Retardants and Autism Spectrum Disorder and Intellectual Disability in the Early Markers of Autism Study: A Population-Based Case-Control Study in California. Environ Health Perspect. 2017 Aug 30;125(8):087023.

Magalhães, J.Z., et al. Prenatal exposure to fipronil disturbs maternal aggressive behavior in rats. Neurotoxicol Teratol. Nov-Dec 2015;52(Pt A):11-6.

Malin, A.J., et al. Maternal Urinary Fluoride and Child Neurobehavior at Age 36 Months. JAMA Netw Open. 2024 May 1;7(5):e2411987.

Martínez-Frías, M.L., et al. Maternal polymorphisms 677C-T and 1298A-C of MTHFR, and 66A-G MTRR genes: is there any relationship between polymorphisms of the folate pathway, maternal homocysteine levels, and the risk for having a child with Down syndrome? Am J Med Genet. 2006 May 1;140(9):987-97.

Mercer, J.S., et al. The Effects of Delayed Cord Clamping on 12-Month Brain Myelin Content and Neurodevelopment: A Randomized Controlled Trial. Am J Perinatol. 2022 Jan;39(1):37-44.

Muanda, F.T., et al. Use of antibiotics during pregnancy and the risk of major congenital malformations: a population based cohort study. Br J Clin Pharmacol. 2017 Nov;83(11):2557-2571.

Modabbernia, A., et al. Apgar score and risk of autism. Eur J Epidemiol. 2018 Oct 5.

Mulder, E.J.H., et al. Prenatal maternal stress: effects on pregnancy and the (unborn) child. Early Hum Dev. 2002 Dec;70(1-2):3-14.

Nguyen, L.D.., et al. Women's holistic self-care behaviors during pregnancy and associations with psychological well-being: implications for maternal care facilities. BMC Pregnancy Childbirth. 2022 Aug 9;22(1):631.

Palmer, D.J., et al. Maternal peanut and egg consumption during breastfeeding randomized pilot trial. Pediatr Allergy Immunol. 2022 Sep;33(9):e13845.

Parkin, K.,, et al. Risk Factors for Gut Dysbiosis in Early Life. Microorganisms. 2021 Sep 30;9(10):2066.

Peltier, M.R., et al. Maternal Hypothyroidism Increases the Risk of Attention-Deficit Hyperactivity Disorder in the Offspring. Am J Perinat. 2020 Oct 21.

Premji, S.S., et al. Prenatal allostatic load and preterm birth: A systematic review. Front Psychol. 2022 Oct 4:13:1004073.

Qiu, C., et al. Association Between Epidural Analgesia During Labor and Risk of Autism Spectrum Disorders in Offspring. JAMA Pediatr. 2020 Oct 12.

Rauh, V.A., et al. Impact of prenatal chlorpyrifos exposure on neurodevelopment in the first 3 years of life among inner-city children. Pediatrics. 2006;118;e1845-1859.

Rifas-Shiman, S.L., et al. Associations of prenatal or infant exposure to acetaminophen or ibuprofen with mid-childhood executive function and behaviour. Paediatr Perinat Epidemiol. 2020 May;34(3):287-298.

Rosman, N.P., et al. Association of Prenatal Ultrasonography and Autism Spectrum Disorder. JAMA Pediatr. 2018 Apr 1;172(4):336-344.

Rowley, C.E., et al. Altered dietary behaviour during pregnancy impacts systemic metabolic phenotypes. Front Nutr. 2023 Dec 4:10:1230480.

Silva, D.T., et al. Introducing the ORIGINS project: a community-based interventional birth cohort. Rev Environ Health. 2020 Sep 25;35(3):281-293.

Silvestrin, R.B., et al. Animal model of autism induced by prenatal exposure to valproate: altered glutamate metabolism in the hippocampus. BrainRes. 1495 (2013) 52–60.

Sucksdorff, M., et al. Maternal Vitamin D Levels and the Risk of Offspring Attention-Deficit/Hyperactivity Disorder. J Am Acad Child Adolesc Psychiatry. 2019 Dec 18.

Taddio, A., et al. Effect of neonatal circumcision on pain responses during vaccination in boys. Lancet. 1995 Feb 4;345(8945):291-2.

Taige, N.M., et al. Gestational Age at Term, Delivery Circumstance, and Their Association with Childhood Attention Deficit Hyperactivity Disorder Symptoms. Paediatr Perinat Epidemiol. 2016 Mar;30(2):171-80.

Tanneeru, S., et al. Evaluation of Microflora (Viral and Bacterial) in Subgingival and Placental Samples of Pregnant Women with Preeclampsia with and without Periodontal Disease: A Cross-Sectional Study. J Int Soc Prev Community Dent. 2020 Mar 10;10(2):171-176.

Tollånes, M.C., et al. Cesarean section and risk of severe childhood asthma: a population-based cohort study. J Pediatr. 2008 Jul;153(1):112-6.

Torres, G., et al. Perspectives of Pitocin administration on behavioral outcomes in the pediatric population: recent insights and future implications. Heliyon. 2020 May 29;6(5):e04047.

Vuillermot, S., et al. Vitamin D treatment during pregnancy prevents autism-related phenotypes in a mouse model of maternal immune activation. Mol Autism. 2017 Mar 7;8:9.

Wilcox, J.C., et al. Evaluating Engagement in a Digital and Dietetic Intervention Promoting Healthy Weight Gain in Pregnancy: Mixed Methods Study. J Med Internet Res. 2020 Jun 26;22(6):e17845.

Wang, A., et al. Association of Prenatal Exposure to Organophosphate, Pyrethroid, and Neonicotinoid Insecticides with Child Neurodevelopment at 2 Years of Age: A Prospective Cohort Study. Environ Health Perspect. 2023 Oct;131(10):107011.

Wang, S.S., et al. Polymorphisms in genes involved in folate metabolism as maternal risk factors for Down syndrome in China*. J Zhejiang Univ Sci B. 2008 Feb; 9(2): 93–99.

Watai,, K., et al. Epidemiological association between multiple chemical sensitivity and birth by caesarean section: a nationwide case-control study. Environ Health. 2018 Dec 14;17(1):89.

Willfors, C., et al. Medical history of discordant twins and environmental etiologies of autism. Transl Psychiatry. 2017 Jan; 7(1): e1014.

Williams, E.L., et al. Potential teratogenic effects of ultrasound on corticogenesis: implications for autism. Med Hypotheses. 2010 Jul;75(1):53-8.

Winter, C., et al. Dopamine and serotonin levels following prenatal viral infection in mouse—implications for psychiatric disorders such as schizophrenia and autism. European Neuropsychopharmacology. 2008 Oct;18(10):712-6.

Woodbury, M.L., et al. Examining the relationship of acetaminophen use during pregnancy with early language development in children. Pediatr Res. 2023 Dec 11.

Zaigham, M., et al. Prelabour caesarean section and neurodevelopmental outcome at 4 and 12 months of age: an observational study. BMC Pregnancy and Childbirth. 2020 (20)564.

Zengeler, K.E., et al. SSRI treatment modifies the effects of maternal inflammation on in utero physiology and offspring neurobiology. Brain Behav Immun. 2023 Feb:108:80-97.

Resilience

Sources & References

Ahuja, S.K., et al. Immune resilience despite inflammatory stress promotes longevity and favorable health outcomes including resistance to infection. Nat Commun. 2023 Jun 13;14(1):3286.

Bonaccio, M., et al. Mediterranean-type diet is associated with higher psychological resilience in a general adult population: findings from the Moli-sani study. Eur J Clin Nutr. 2018 Jan;72(1):154-160.

Burrows, T., et al. Effectiveness of dietary interventions in mental health treatment: A rapid review of reviews. Nutr Diet. 2022 Jul;79(3):279-290.

da Cunha, L.L., et al. Effects of exercise training on inflammatory, neurotrophic and immunological markers and neurotransmitters in people with depression: A systematic review and meta-analysis. J Affect Disord. 2023 Apr 1:326:73-82.

Dantzer, R., et al. Resilience and immunity. Brain Behav Immun. 2018 Nov:74:28-42.

Dinan, T.G., et al. The Microbiome-Gut-Brain Axis in Health and Disease. Gastroenterol Clin North Am. 2017 Mar;46(1):77-89.

Dogra, S.K., et al. Gut Microbiota Resilience: Definition, Link to Health and Strategies for Intervention. Front Microbiol. 2020 Sep 15:11:572921.

Grossman, P., et al. Mindfulness-based stress reduction and health benefits. A meta-analysis. J Psychosom Res. 2004 Jul;57(1):35-43.

Hantsoo, L., et al. Childhood adversity impact on gut microbiota and inflammatory response to stress during pregnancy. Brain Behav Immun. 2019 Jan:75:240-250.

Jacka, F.N., et al. A randomised controlled trial of dietary improvement for adults with major depression (the 'SMILES' trial). BMC Med. 2017 Jan 30;15(1):23.

Leipold, B., et al. Physical activity and nutrition in relation to resilience: a cross-sectional study. Sci Rep. 2024 Jan 27;14(1):2272.

Manigault, A.W., et al. Psychosocial Resilience to Inflammation-Associated Depression: A Prospective Study of Breast-Cancer Survivors. Psychol Sci. 2022 Aug;33(8):1328-1339.

McDonald, D., et al. American Gut: an Open Platform for Citizen Science Microbiome Research. mSystems. 2018 May 15;3(3):e00031-18.

Relman, D.A. The human microbiome: ecosystem resilience and health. Nutr Rev. 2012 Aug;70 Suppl 1(Suppl 1):S2-9.

Voigt, R.M., et al. The intestinal microbiota: determinants of resiliency? Lancet Healthy Longev. 2021 Jan;2(1):e2-e3.

Wastyk, H.C., et al. Gut-microbiota-targeted diets modulate human immune status. Cell. 2021 Aug 5;184(16):4137-4153.e14.

Whatnall, M.C., et al. Are Psychological Distress and Resilience Associated with Dietary Intake Among Australian University Students? Int J Environ Res Public Health. 2019 Oct 24;16(21):4099.

Yin, Z., et al. Dietary Diversity Was Positively Associated with Psychological Resilience among Elders: A Population-Based Study. Nutrients. 2019 Mar 18;11(3):650.

Retained Reflexes

Sources & References

Amos, P. Rhythm and timing in autism: learning to dance. Front Integr Neurosci. 2013 Apr 19;7:27.

Akhmatova, N.K., et al. Influence of MNRI on the Immune Status of Children with Down Syndrome. Journal of Clinical & Cellular Immunology. 8 (2017): 1-8.

Barnhill, E. Neural connectivity, music, and movement: a response to Pat Amos. Front Integr Neurosci. 2013 Apr 24;7:29.

Bell, C., et al. The Effect of the MNRI Method on Neurotransmitter Biomarkers of Individuals with Neurodevelopmental Disorders. Neuroscience and Medicine. 2019.

Cho, H., et al. Effects of Action Observation Training with Auditory Stimulation on Static and Dynamic Balance in Chronic Stroke Patients. J Stroke Cerebrovasc Dis. 2020 May;29(5):104775.

Deiss, T., et al. Physiological Markers and Reflex Pattern Progression in Individuals with Neurodevelopmental Deficits Utilizing the MNRI Method. Neuroscience and Medicine. 2019.

Grigg. T.M., et al. Primitive Reflex Integration and Reading Achievement in the Classroom. Journal of Neurology and Experimental Neuroscience. 2023; 9(1), 18-26.

Grigg, T.M., et al. Retained primitive reflexes: Perceptions of parents who have used Rhythmic Movement Training with their children. J Child Health Care. 2018 Sep;22(3):406-418.

Grzywniak, C. Integration exercise programme for children with learning difficulties who have preserved vestigial primitive reflexes. Acta Neuropsychologica. 2017;15(3).

Hardy, M.W., et al. Rhythm, movement, and autism: using rhythmic rehabilitation research as a model for autism. Front Integr Neurosci. 2013 Mar 28;7:19.

Herbert, J., et al. Crawling is associated with more flexible memory retrieval by 9-month-old infants. Dev Sci. 2007 Mar;10(2):183-9.

Hong, H.J., et al. Effect of Rhythmic Movement Program to Improve Walking Ability for Elderly Patients with Stroke. Indian Journal of Science and Technology. 2016 Jul;9(26).

Iverson, J.M. Developing language in a developing body: the relationship between motor development and language development. J Child Lang. 2010 Mar;37(2):229-61.

Jordan-Black, J. The effects of the Primary Movement programme on the academic performance of children attending ordinary primary school. Journal of Research in Special Educational Needs. 2005 Nov;5(3):101 – 111.

Kadivar, Z., et al. Effect of step training and rhythmic auditory stimulation on functional performance in Parkinson patients. Neurorehabil Neural Repair. 2011 Sep;25(7):626-35.

Ladányi, K. et al. Is atypical rhythm a risk factor for developmental speech and language disorders? Wiley Interdiscip Rev Cogn Sci. 2020 Sep;11(5):e1528.

Lakatos, P., et al. A New Unifying Account of the Roles of Neuronal Entrainment. Curr Biol. 2019 Sep 23;29(18):R890-R905.

Masgutova, S., et al. Effect of the MNRI Reflex Neuromodulation on the QEEG and Neurotransmitters of Children Diagnosed with Cerebral Palsy. 2020.

Masgutova, S., et al. Effects of MNRI Visual Reflex Neuro-Training on Visual and Academic Skills of Children with Autism. International Journal of School and Cognitive Psychology 05 (2018): 1-15.

Masgutova, S. Post-Trauma Recovery in Children of Newtown, CT using MNRI Reflex Integration. Journal of
Traumatic Stress Disorders & Treatment. 5(5) 2017.

Masgutova, S., et al. Reflex Profile of Children with Down Syndrome Improvement of Neurosensorimotor Development Using the MNRI® Reflex Integration Program. International Journal of Neurorehabilitation. 3 (2016): 1-9.

Masgutova, S., et al. Progress with Neurosensorimotor Reflex Integration for Children with Autism Spectrum Disorder. 2016.

Masgutova, S. Reflex Integration Disorder as a New Treatment Paradigm for Children with Autism. 2015.

McWhirter, K., et al. The association between learning disorders, motor function, and primitive reflexes in pre-school children: A systematic review. J Child Health Care. 2022 Jul 13;13674935221114187.

Melillo, R., et al. Persistent Childhood Primitive Reflex Reduction Effects on Cognitive, Sensorimotor, and Academic Performance in ADHD. Front Public Health. 2020 Nov 17;8:431835.

Mohamed, M.A., et al. Sensory integration versus Masgutova neuro-sensorimotor reflex integration program on controlling primitive reflexes and gross motor abilities in children with diplegic cerebral palsy. Physiother Res Int. 2023 Jun 29;e2030.

Nowak, K., et al. Evaluation of the Therapeutic Effect of MNRI Reflex Neuromodulation on Children Diagnosed with Autism Based on Reflex Assessments, QEEG Analysis and ATEC Questionnaire. 2020.

Pilecki, W., et al. The impact of rehabilitation carried out using the Masgutova Neurosensorimotor Reflex Integration method in children with cerebral palsy on the results of brain stem auditory potential examinations. Adv Clin Exp Med. 2012 May-Jun;21(3):363-71.

Schmidt, N.B., et al. Exploring human freeze responses to a threat stressor. J Behav Ther Exp Psychiatry. 2008 Sep;39(3):292-304.

Suh, J.H., et al. Effect of rhythmic auditory stimulation on gait and balance in hemiplegic stroke patients. NeuroRehabilitation. 2014;34(1):193-9.

Tatarinova, T.V., et al. The Impact of MNRI Therapy on the Levels of Neurotransmitters Associated with Inflammatory Processes. Int J Mol Sci. 2020 Feb 18;21(4):1358.

Teitelbaum, P., et al. Movement analysis in infancy may be useful for early diagnosis of autism. Proc Natl Acad Sci U S A. 1998 Nov 10;95(23):13982-7.

Teitelbaum, P., et al. Reflexes gone astray in autism in infancy. Journal of Developmental and Learning Disorders, 2003;6, 15-22.

Van Hirtum, T., et al. Is atypical rhythm a riskfactor for developmental speech and language disorders? J Assoc Res Otolaryngol. 2021 Jul;22(4):465-480.

Winkler, I., et al. Newborn infants detect the beat in music. Proc Natl Acad Sci U S A. 2009 Feb 17;106(7):2468-71.

Zaigham, M., et al. Prelabour caesarean section and neurodevelopmental outcome at 4 and 12 months of age: an observational study. BMC Pregnancy and Childbirth. 2020 (20)564.

Zentner, M., et al. Rhythmic engagement with music in infancy. Proc Natl Acad Sci U S A. 2010 Mar 30;107(13):5768-73.

Seizures

Sources & References

American Academy of Neurology. Cannabis-based medicine may cut seizures in half for those with tough-to-treat epilepsy. 2017 Apr 18.

Ari, C., et al. Delaying latency to hyperbaric oxygen-induced CNS oxygen toxicity seizures by combinations of exogenous ketone supplements. Physiol Rep. 2019 Jan;7(1):e13961.

Bertelsen, E.N., et al. Childhood Epilepsy, Febrile Seizures, and Subsequent Risk of ADHD. Pediatrics. 2016 Aug;138(2). pii: e20154654.

Bigi, S., et al. The growing spectrum of antibody-associated inflammatory brain diseases in children. Neurol Neuroimmunol Neuroinflamm. 2015 Apr 2;2(3):e92.

Bochynska, A., et al. The effect of vitamin B supplementation on homocysteine metabolism and clinical state of patients with chronic epilepsy treated with carbamazepine and valproic acid. Seizure. 2012 May;21(4):276-81.

Chakravarthi, N., et al. A switch in G protein coupling for type 1 corticotropin-releasing factor receptors promotes excitability in epileptic brains. Sci Signal. 2016 Jun 14;9(432):ra60.

Chang, P., et al. Seizure control by ketogenic diet-associated medium chain fatty acids. Neuropharmacology. 2013 Jun;69:105-14.

Chapman, G. Glutamate and epilepsy. J Nutr. 2000 Apr;130(4S Suppl):1043S-5S.

Cocito, L., et al. GABA and phosphatidylserine in human photosensitivity: a pilot study. Epilepsy Res. 1994 Jan;17(1):49-53.

DeHavenon, A., et al. The Secret “Spice”: An Undetectable Toxic Cause of Seizure. Neurohospitalist. 2011 Oct; 1(4): 182–186.

Devinsky, O., et al. Trial of Cannabidiol for Drug-Resistant Seizures in the Dravet Syndrome. N Engl J Med. 2017 Aug 17;377(7):699-700.

Dick, F.D. Solvent neurotoxicity. Occup Environ Med. 2006 Mar; 63(3): 221–226.

D'Souza, C.E., et al. GAD65 antibody-associated autoimmune epilepsy with unique independent bitemporal-onset ictal asystole. Epileptic Disord. 2018 Jun 1;20(3):204-208.

Erturk, C.O., et al. EEG abnormalities and long term seizure outcome in high functioning autism. Acta Neurol Belg. 2017 Sep;117(3):729-732.

Ferreira, P., et al. Late diagnosis of cerebral folate deficiency: Fewer seizures with folinic acid in adult siblings. Neurol Genet. 2015 Dec 22;2(1):e38.

Fregni, F., et al. A randomized clinical trial of repetitive transcranial magnetic stimulation in patients with refractory epilepsy. Ann Neurol. 2006 Oct;60(4):447-55.

Frye, R., et al. A Review of Traditional and Novel Treatments for Seizures in ADD: Findings from a Systematic Review and Expert Panel. Front Public Health. 2013 Sep 13;1:31.

Frye, R.E., et al. Seizures in Autism Spectrum Disorder: Symptoms, Assessment, and Treatment. Autism File. 2016 Feb-Mar, Issue 66.

Fukahori, M., et al. Effects of dietary zinc status on seizure susceptibility and hippocampal zinc content in the El (epilepsy) mouse. Brain Res. 1990 Oct 8;529(1-2):16-22.

Geier, M.R., et al. The potential importance of steroids in the treatment of autistic spectrum disorders and other disorders involving mercury toxicity. Med Hypotheses. 2005;64(5):946-54.

Hassel, B., et al. Brain infection with Staphylococcus aureus leads to high extracellular levels of glutamate, aspartate, γ-aminobutyric acid, and zinc. J Neurosci Res. 2014 Dec;92(12):1792-800.

Hausman-Kedem, M., et al. Efficacy of CBD-enriched medical cannabis for treatment of refractory epilepsy in children and adolescents - An observational, longitudinal study. Brain Dev. 2018 Aug;40(7):544-551.

Herbert, M.R., et al. Autism and EMF? Plausibility of a pathophysiological link - Part I. Pathophysiology. 2013 Jun;20(3):191-209.

Hollo, A., et al. Correction of vitamin D deficiency improves seizure control in epilepsy: a pilot study. Epilepsy Behav. 2012 May;24(1):131-3.

Huang, Y., et al. Pyridoxine Supplementation Improves the Activity of Recombinant Glutamate Decarboxylase and the Enzymatic Production of Gama-Aminobutyric Acid. PLoS One. 2016 Jul 20;11(7):e0157466.

Humphries, P., et al. Direct and indirect cellular effects of aspartame on the brain. Eur J Clin Nutr. 2008 Apr;62(4):451-62.

Ibrahim, F.A.S., et al. The differential effects of eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) on seizure frequency in patients with drug-resistant epilepsy - A randomized, double-blind, placebo-controlled trial. Epilepsy Behav. 2018 Aug 28;87:32-38.

Irevall, T., et al. B12 deficiency is common in infants and is accompanied by serious neurological symptoms. Acta Paediatr. 2017 Jan;106(1):101-104.

Jang, D.H., et al. Status epilepticus and wide-complex tachycardia secondary to diphenhydramine overdose. Clin Toxicol (Phila). 2010 Nov;48(9):945-8.

Jett, D.A. Chemical toxins that cause seizures. Neurotoxicology. 2012 Dec;33(6):1473-5.

Johansson, O., et al. Exacerbation of demyelinating syndrome after exposure to wireless modem with public hotspot. Electromagn Biol Med. 2016;35(4):393-7.

Johnson, A.C., et al. Magnesium sulfate treatment reverses seizure susceptibility and decreases neuroinflammation in a rat model of severe preeclampsia. PLoS One. 2014 Nov 19;9(11):e113670

Julian, T., et al. Gluten sensitivity and epilepsy: a systematic review. J Neurol. 2018 Aug 23.

Klein, P., et al. Dietary treatment in adults with refractory epilepsy: a review. Neurology. 2014 Nov 18;83(21):1978-85.

Kossoff, E.H., et al. Optimal clinical management of children receiving dietary therapies for epilepsy: Updated recommendations of the International Ketogenic Diet Study Group. Epilepsia Open. 2018 Jun; 3(2): 175–192.

Koutroumanidou, E., et al. Increased seizure latency and decreased severity of pentylenetetrazol-induced seizures in mice after essential oil administration. Epilepsy Res Treat. 2013;2013:532657.

Kr, Pandy Shashi, et al. Herbal and synthetic approaches for the treatment of epilepsy. Intl Journal of Nutrition, Pharmacology and Herbal Medicine. 2014;4(1):43-52.

Kubik, P., et al. [Neurofeedback therapy influence on clinical status and some EEG parameters in children with localized epilepsy]. Przegl Lek. 2016;73(3):157-60.

Luo, P., et al. The role of glutamate receptors in traumatic brain injury: implications for postsynaptic density in pathophysiology. Brain Res Bull. 2011 Jul 15;85(6):313-20.

Manev, H., et al. Delayed increase of Ca2+ influx elicited by glutamate: role in neuronal death. Mol Pharmacol. 1989 Jul;36(1):106-12.

Masdaghinia, A., et al. Anticonvulsant effects of thiamine on pentylenetetrazole-induced seizure in mice. Nutr Neurosci. 2017 Aug 2:1-9.

McCue, L.M., et al. Prevalence of non-febrile seizures in children with idiopathic autism spectrum disorder and their unaffected siblings: a retrospective cohort study. BMC Neurol. 2016 Nov 28;16(1):245.

Menon, B., et al. The effect of anti epileptic drug therapy on serum 25-hydroxyvitamin D and parameters of calcium and bone metabolism--a longitudinal studySeizure. 2010 Apr;19(3):153-8.

Napoli, E., et al. Toxicity of the flame-retardant BDE-49 on brain mitochondria and neuronal progenitor striatal cells enhanced by a PTEN-deficient background. Toxicol Sci. 2013 Mar;132(1):196-210.

Nei, M., et al. Ketogenic diet in adolescents and adults with epilepsy. Seizure. 2014 Jun;23(6):439-42.

Ogunmekan, A.O. Vitamin E deficiency and seizures in animals and man. Can J Neurol Sci. 1979 Feb;6(1):43-5.

Olson, C.A., et al. The Gut Microbiota Mediates the Anti-Seizure Effects of the Ketogenic Diet. Cell. 2018 Jul 12;174(2):497.

Ozkale, Y., et al. Serum vitamin B12, folic acid, and homocysteine levels in children with febrile seizure. Turk J Pediatr. 2015 Jul-Aug;57(4):345-52.

Park, E.G., et al. Use of the Modified Atkins Diet in Intractable Pediatric Epilepsy. J Epilepsy Res. 2018 Jun 30;8(1):20-26.

Pickett, J., et al. Mortality in Individuals With Autism, With and Without Epilepsy. J Child Neurol. 2011 Aug;26(8):932-9.

Qi, J., et al. Enhanced susceptibility to stress and seizures in GAD65 deficient mice. PLoS One. 2018 Jan 29;13(1):e0191794.

Quadros, E.V., et al. Folate receptor autoantibodies are prevalent in children diagnosed with autism spectrum disorder, their normal siblings and parents. Autism Res. 2018 May;11(5):707-712.

Reda, D.M., et al. Fish Oil Intake and Seizure Control in Children with Medically Resistant Epilepsy. N Am J Med Sci. 2015 Jul;7(7):317-21.

Rout, U.K., et al. Presence of GAD65 autoantibodies in the serum of children with autism or ADHD. Eur Child Adolesc Psychiatry. 2012 Mar;21(3):141-7.

Rowley, N.M., et al. Glutamate and GABA synthesis, release, transport and metabolism as targets for seizure control. Neurochem Int. 2012 Sep;61(4):546-58.

Sai, Y., et al. Clinical diagnosis and treatment of pediatric anti-N-methyl-D-aspartate receptor encephalitis: A single center retrospective study. Exp Ther Med. 2018 Aug;16(2):1442-1448.

Schauwecker, P.A. The effects of glycemic control on seizures and seizure-induced excitotoxic cell death. BMC Neurosci. 2012 Aug 6;13:94.

Schugar, R.C., et al. Low-carbohydrate ketogenic diets, glucose homeostasis, and nonalcoholic fatty liver disease. Curr Opin Clin Nutr Metab Care. 2012 Jul; 15(4): 374–380.

Schwartzkroin, P.A. (Ed.) Encyclopedia of basic epilepsy research. UK, Academic Press/Elsevier (2009), pp. 519-522.

Spatola, M., et al. Seizures and risk of epilepsy in autoimmune and other inflammatory encephalitis. Curr Opin Neurol. 2017 Jun;30(3):345-353.

Stockings, E., et al. Evidence for cannabis and cannabinoids for epilepsy: a systematic review of controlled and observational evidence. J Neurol Neurosurg Psychiatry. 2018 Jul;89(7):741-753.

Strehl, U., et al. Sustained reduction of seizures in patients with intractable epilepsy after self-regulation training of slow cortical potentials - 10 years after. Front Hum Neurosci. 2014 Aug 8;8:604.

Suvasini, S., et al. The Modified Atkins Diet in Refractory Epilepsy. Epilepsy Res Treat. 2014; 2014: 404202.

Tan, G., et al. Meta-analysis of EEG biofeedback in treating epilepsy. Clin EEG Neurosci. 2009 Jul;40(3):173-9.

Taub, K.S., et al. Risk of seizure recurrence after achieving initial seizure freedom on the ketogenic diet. Epilepsia. 2014 Apr;55(4):579-83.

Taubøll, E., et al. Interactions between hormones and epilepsy. Seizure. 2015 May;28:3-11.

Thomas, S., et al. Brief Report: Prevalence of Co-occurring Epilepsy and Autism Spectrum Disorder: The U.S. National Survey of Children's Health 2011-2012. J Autism Dev Disord. 2017 Jan;47(1):224-229.

Tu, Y.F., et al. Postnatal Steroids and Febrile Seizure Susceptibility in Preterm Children. Pediatrics. 2016 Apr;137(4). pii: e20153404.

van Koert, R.R., et al. Caffeine and seizures: A systematic review and quantitative analysis. Epilepsy Behav. 2018 Mar;80:37-47.

Walker, J.E., et al. Neurofeedback treatment of epilepsy. Child Adolesc Psychiatr Clin N Am. 2005 Jan;14(1):163-76, viii.

Walls, A.B., et al. GAD65 is essential for synthesis of GABA destined for tonic inhibition regulating epileptiform activity. J Neurochem. 2010 Dec;115(6):1398-408.

Watkins, J.C., et al. The glutamate story. Br J Pharmacol. 2006 Jan; 147(Suppl 1): S100–S108.

Wei, H., et al. Auricular Acupuncture May Suppress Epileptic Seizures via Activating the Parasympathetic Nervous System: A Hypothesis Based on Innovative Methods. Evid Based Complement Alternat Med. 2012; 2012: 615476.

West, R.W., et al. The effect of color on light-induced seizures: a case report. Optom Vis Sci. 1996 Feb;73(2):109-13.

Westmark, C.J. Soy infant formula and seizures in children with autism: a retrospective study. PLoS One. 2014 Mar 12;9(3):e80488.

Wiggs, K.K., et al. Attention-deficit/hyperactivity disorder medication and seizures. Neurology. 2018 Mar 27;90(13):e1104-e1110.

Wilkins, A.J., et al. Treatment of photosensitive epilepsy using coloured glasses. Seizure. 1999 Dec;8(8):444-9.

Zifkin, E.G. , et al. Reflex epilepsy and reflex seizures of the visual system: a clinical review. Epileptic Disord. 2000 Sep;2(3):129-36.

Sensory Processing Disorder

Sources & References

Ābele, S., et al. Specific Carbohydrate Diet (SCD/GAPS) and Dietary Supplements for Children with Autistic Spectrum Disorder. Proc. Latv. Acad. Sci. 2021;75:417–425.

Accardo, P.J., et al. Toe walking in autism: further observations. J Child Neurol. 2015 Apr;30(5):606-9.

Adam, G., et al. The influence of choline treatment on behavioral and neurochemical autistic-like phenotype in Mthfr-deficient mice. Transl Psychiatry. 2020 Sep 18;10(1):316.

Adams, J.B., et al. Comprehensive Nutritional and Dietary Intervention for Autism Spectrum Disorder-A Randomized, Controlled 12-Month Trial. Nutrients. 2018 Mar 17;10(3).

Adams, J.B., et al. Effect of a vitamin/mineral supplement on children and adults with autism. BMC Pediatr. 2011;11:111.

Adams, J.B., et al. Gastrointestinal Flora and Gastrointestinal Status in Children with Autism—Comparisons to Typical Children and Correlation with Autism Severity. BMC Gastroenterol. 2011;11:22.

Adams, J.B., et al. Mercury in first-cut baby hair of children with autism versus typically-developing children. Toxicological & Environmental Chemistry. 2007 Jun;70(12):1046-51.

Adams, J.B., et al. Mercury, Lead, and Zinc in Baby Teeth of Children with Autism Versus Controls. Journal of Toxicology and Environmental Health. 2007 Jun;70(12):1046-51.

Adams, J.B., et al. Nutritional and metabolic status of children with autism vs. neurotypical children, and the association with autism severity. Nutr Metab (Lond) 2011 Jun 8;8(1):34.

Adams, J.B., et al. Vitamin/mineral/micronutrient supplement for autism spectrum disorders: a research survey. BMC Pediatr. 2022 Oct 13;22(1):590.

Adams, L.J., et al. Infant feeding method and special educational need in 191,745 Scottish schoolchildren: A national, population cohort study. PLoS Med. 2023 Apr 6;20(4):e1004191.

Aguilera, M., et al. Antibiotic-induced dysbiosis alters host-bacterial interactions and leads to colonic sensory and motor changes in mice. Gut Microbes. 2015;6(1):10-23.

Ahn, R.R., et al. Prevalence of parents’ perceptions of sensory processing disorders among kindergarten children. Am J Occup Ther. May-Jun 2004;58(3):287-93.

Alabdali, A., et al. A key role for an impaired detoxification mechanism in the etiology and severity of autism spectrum disorders. Behav Brain Funct. 2014;10:14.

Alabdali, A., et al. Association of social and cognitive impairment and biomarkers in autism spectrum disorders. J Neuroinflammation. 2014;11:4.

Alampi, J.D., et al. Gestational Exposure to Toxicants and Autistic Behaviors using Bayesian Quantile Regression. Am J Epedemiol. 2021 Sep 1;190(9):1803-1813.

Alberti, A., et al. Sulphation Deficit in “Low-Functioning” Autistic Children: A Pilot Study. Biol. Psychiatry. 1999;46:420–424.

Aldad, T.S., et al. Fetal Radiofrequency Radiation Exposure From 800-1900 Mhz-Rated Cellular Telephones Affects Neurodevelopment and Behavior in Mice. Sci Rep. 2012;2:312.

Amminger, G.P., et al. Omega-3 fatty acids supplementation in children with autism: a double-blind randomized, placebo-controlled pilot study. Biol Psychiatry. 2007 Feb 15;61(4):551-3.

Ashraghi, R.S., et al. Early Disruption of the Microbiome Leading to Decreased Antioxidant Capacity and Epigenetic Changes: Implications for the Rise in Autism. Front. Cell. Neurosci., 15 Aug 2018.

Ashwood, P., et al. Elevated plasma cytokines in autism spectrum disorders provide evidence of immune dysfunction and are associated with impaired behavioral outcome. Brain Behav Immun. 2011 Jan;25(1):40-5.

Ashwood, P., et al. The immune response in autism: a new frontier for autism research. Journal of Leukocyte Biology. 2006 Jul;80(1):1-15.

Atladóttir, H.Ó., et al. Association of family history of autoimmune diseases and autism spectrum disorders. Pediatrics. 2009 Aug;124(2):687-94.

Atladóttir, H.Ó., et al. Autism after infection, febrile episodes, and antibiotic use during pregnancy: an exploratory study. Pediatrics. 2012 Dec;130(6):e1447-54.

Baker, S. Canaries and Miners. Alternative Therapies in Health and Medicine. Nov-Dec 2008;14(6):24-6.

Baker, S., et al. Case Study: Rapid Complete Recovery From An Autism Spectrum Disorder After Treatment of Aspergillus With The Antifungal Drugs Itraconazole And Sporanox. Integr Med (Encinitas). 2020 Aug;19(4):20-27.

Barnhill, K., et al. Brief Report: Implementation of a Specific Carbohydrate Diet for a Child with Autism Spectrum Disorder and Fragile X Syndrome. J. Autism Dev. Disord. 2020;50:1800–1808. 

Barrett, B. Substantial lifelong cost of autism spectrum disorder. J Pediatr. 2014;165(5):1068-9.

Bateman, C. Autism–mitigating a global epidemic. S Afr Med J. 2013;103(5):276-7.

Ben-Sasson, A., et al. Sensory over-responsivity in elementary school: prevalence and social-emotional correlates. J Abnorm Child Psychol. 2009 Jul;37(5):705-16.

Berding, K., et al. Diet Can Impact Microbiota Composition in Children With Autism Spectrum Disorder. Front. Neurosci. 2018;12:515.

Bernard, S., et al. Autism: a novel form of mercury poisoning. Med Hypotheses. 2001 Apr;56(4):462-71.

Binder, D.K., et al. Brain-derived neurotrophic factor. Growth Factors. 2004 Sep;22(3):123-31.

Bittker, S.S., et al. Postnatal Acetaminophen and Potential Risk of Autism Spectrum Disorder among Males. Behav Sci (Basel). 2020 Jan 1;10(1):26.

Bitsika, V., et al. Hypothalamus-pituitary-adrenal axis daily fluctuation, anxiety and age interact to predict cortisol concentrations in boys with an autism spectrum disorder. Physiol Behav. 2015;138:200-7.

Bjørklund, G., et al. Gastrointestinal alterations in autism spectrum disorder: What do we know? Neurosci Biobehav Rev. 2020 Nov:118:111-120.

Blaxill, Mark, et al. Autism Tsunami: the Impact of Rising Prevalence on the Societal Cost of Autism in the United States. J Autism Dev Disord. 2022 Jun;52(6):2627-2643.

Blaylock, R.L. A possible central mechanism in autism spectrum disorders, part 1. Altern Ther Health Med. 2008 Nov-Dec;14(6):46-53.

Blaylock, R.L. A possible central mechanism in autism spectrum disorders, part 2: immunoexcitotoxicity. Altern Ther Health Med. 2009 Jan-Feb;15(1):60-7.

Blaylock, R.L. A possible central mechanism in autism spectrum disorders, part 3: the role of excitotoxin food additives and the synergistic effects of other environmental toxins. Altern Ther Health Med. 2009 Mar-Apr;15(2):56-60.

Blaylock, R.L., et al. Immune-glutamatergic dysfunction as a central mechanism of the autism spectrum disorders. Curr Med Chem. 2009;16(2):157-70.

Boat, T.F., et al. Prevalence of Learning Disabilities. Mental Disorders and Disabilities Among Low-Income Children. Washington (DC): National Academies Press (US); 2015 Oct 28. 16.

Boris, M.J., et al. Pollen Exposure as a Cause for the Deterioration of Neurobehavioral Function in Children with Autism and Attention Deficit Hyperactive Disorder. J of Nutritional and Environmental Medicine. Mar 2004;14(1):47 – 54.

Borre, Y.E., et al. Microbiota and neurodevelopmental windows: implications for brain disorders. Trends Mol Med. 2014 Sep;20(9):509-18.

Bouder, et al. Brief report: Quantifying the impact of autism coverage on private insurance premiums. J Autism Dev Disord. 2009;39(6):953-7.

Bradstreet, et al. Biomarker-guided interventions of clinically relevant conditions associated with autism spectrum disorders and attention deficit hyperactivity disorder. Altern Med Rev. 2010;15(1):15-32.

Bransfield, R.C., et al. The association between tick-borne infections, Lyme borreliosis and autism spectrum disorders. Medical Hypotheses. 2008;70(5):967-74.

Breitenkamp, A.F., et al. Voltage-gated Calcium Channels and Autism Spectrum Disorders. Curr Mol Pharmacol. 2015;8(2):123-32.

Brown, et al. Observable essential fatty acid deficiency markers and autism spectrum disorder. Breastfeed Rev. 2014;22(2):21-6.

Buescher, et al. Costs of autism spectrum disorders in the United Kingdom and the United States. JAMA Pediatr. 2014;168(8):721-8.

Buie, T., et al. Evaluation, diagnosis, and treatment of gastrointestinal disorders in individuals with ASDs: a consensus report. Pediatrics. 2010 Jan;125 Suppl 1:S1-18.

Buie, T., et al. Recommendations for evaluation and treatment of common gastrointestinal problems in children with ASDs. Pediatrics. 2010 Jan;125 Suppl 1:S19-29.

Bull, G., et al. Indolyl-3-acryloylglycine (IAG) is a putative diagnostic urinary marker for autism spectrum disorders. Med Sci Monit. 2003;9(10):CR422-5.

Büsselberg, D. Calcium channels as target sites of heavy metals. Toxicol Lett. 1995 Dec:82-83:255-61.

Camilleri, M. Serotonin in the gastrointestinal tract. Curr Opin Endrocrinol Diabetes Obes. 2009 Feb;16(1):53-9.

Cardinal, D.N., et al. The Maturing of Facilitated Communication: A Means Toward Independent Communication. Res. Pract. Persons Severe Disabil. 2014; (39)189–194.

Carlo, G.L., et al. Wireless radiation in the aetiology and treatment of autism: clinical observations and mechanisms. Journal of the Australasian College of Nutritional and Environmental Medicine, 26(2), 3–7.

Cekici, H., et al. Current Nutritional Approaches in Managing Autism Spectrum Disorder: A Review. Nutr. Neurosci. 2019;22:145–155.

Centers for Disease Control and Prevention. Prevalence and Characteristics of Autism Spectrum Disorder Among Children Aged 8 Years — Autism and Developmental Disabilities Monitoring Network, 11 Sites, United States, 2020. Accessed 24 Mar 2023.

Cheng, N., et al. Metabolic Dysfunction Underlying Autism Spectrum Disorder and Potential Treatment Approaches. Front Mol Neurosci. 2017 Feb 21:10:34.

Chistol, L.T., et al. Sensory Sensitivity and Food Selectivity in Children with Autism Spectrum Disorder. J. Autism Dev. Disord. 2018;48:583–591.

Clarke, E.B., et al. Understanding profound autism: implications for stigma and supports. Front Psychiatry. 2024 Jan 22:15:1287096.

Cleary, D.B., et al. A Parent-Mediated Intervention for Newborns at Familial Likelihood of Autism: Initial Feasibility Study in the General Population. Adv Neurodev Disord. 2022;6(4):494-505.

Connolly, A.M., et al. Serum autoantibodies to brain in Landau-Kleffner variant, autism, and other neurologic disorders. The Journal of Pediatrics. 1999 May;134(5):607-13.

Courchesne, V., et al. Autistic children at risk of being underestimated: school-based pilot study of a strength-informed assessment. Molecular Autism. 2015 Mar 6;6:12.

Critchfield, et al. The potential role of probiotics in the management of childhood autism spectrum disorders. Gastroenterol Res Pract. 2011;2011:161358.

Cubala-Kucharska M. The review of most frequently occurring medical disorders related to aetiology of autism and the methods of treatment. Acta Neurobiol Exp (Wars). 2010;70(2):141-6.

Curran, E.A., et al. Research review: Birth by caesarean section and development of autism spectrum disorder and attention-deficit/hyperactivity disorder: a systematic review and meta-analysis. J Child Psychol Psychiatry. 2015 May;56(5):500-8.

Currenti, S.A. Understanding and determining the etiology of autism. Cell Mol Neurobiol. 2010 Mar;30(2):161-71.

D'Adamo, C.R., et al. Reversal of Autism Symptoms among Dizygotic Twins through a Personalized Lifestyle and Environmental Modification Approach: A Case Report and Review of the Literature. J Pers Med. 2024, 14(6), 641.

Dale, R.C., et al. Encephalitis lethargica syndrome: 20 new cases and evidence of basal ganglia autoimmunity. Brain. 2004 Jan;127(Pt 1):21-33.

Darling, A.L., et al. Association between maternal vitamin D status in pregnancy and neurodevelopmental outcomes in childhood: results from the Avon Longitudinal Study of Parents and Children (ALSPAC). Br J Nutr 2017 Jun;117(12):1682-1692.

Dasdaq, S., et al. Effects of 2.4 GHz radiofrequency radiation emitted from Wi-Fi equipment on microRNA expression in brain tissue. Int J Radiat Biol. 2015 Jul;91(7):555-61.

Dave, D., et al. The effect of an increase in autism prevalence on the demand for auxiliary healthcare workers : evidence from California. Cambridge, MA: National Bureau of Economic Research; 2012. 37 p.p.

D’Eufemia, P., et al. Abnormal intestinal permeability in children with autism. Acta Paediatr. 1996 Sep;85(9):1076-9.

Deisher, T.A., et al. Impact of environmental factors on the prevalence of autistic disorder after 1979. J Public Health and Epidemiology. Sep 2014;6(9):271-286.

de Magistris, et al. Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J Pediatr Gastroenterol Nutr. 2010;51(4):418-24.

De Magistris. L., et al. Antibodies against Food Antigens in Patients with Autistic Spectrum Disorders. BioMed Res. Int. 2013;2013:729349.

Deth, R., et al. How environmental and genetic factors combine to cause autism: A redox/methylation hypothesis. Neurotoxicology. 2008;29(1):190-201.

Doreswamy, S., et al. Effects of Diet, Nutrition, and Exercise in Children With Autism and Autism Spectrum Disorder: A Literature Review. Cureus. 2020;12:e12222.

Dyńka, D., et al. The Role of Ketogenic Diet in the Treatment of Neurological Diseases. Nutrients. 2022 Nov 24;14(23):5003.

Egset, K., et al. Magno App: Exploring Visual Processing in Adults with High and Low Reading Competence. Scandinavian Journal of Educational Research. 07 Jan 2020.

Elamin, N.E., et al. Brain autoantibodies in autism spectrum disorder. Biomark Med. 2014;8(3):345-52.

El-Ansary, A., et al. Neuroinflammation in autism spectrum disorders. J Neuroinflammation. 2012;9:265.

El-Ansary, A., et al. Lipid mediators in plasma of autism spectrum disorders. Lipids Health Dis. 2012;11:160.

El-Rashidy, O., et al. Ketogenic Diet versus Gluten Free Casein Free Diet in Autistic Children: A Case-Control Study. Metab. Brain Dis. 2017;32:1935–1941.

Eppright, T.D., et al. Attention deficit hyperactivity disorder, infantile autism, and elevated blood-lead: a possible relationship. Mo Med. 1996;93(3):136-8).

Erickson, C.A., et al. Gastrointestinal Factors in Autistic Disorder: A Critical Review. Journal of Autism and Developmental Disorders. 2005 Dec;35(6):713-27.

Faber, S., et al. A cleanroom sleeping environment’s impact on markers of oxidative stress, immune dysregulation, and behavior in children with autism spectrum disorders. BMC Complement Altern Med. 2015;15:71.

Fattorusso, A., et al. Autism Spectrum Disorders and the Gut Microbiota. Nutrients. 2019 Feb 28;11(3):521.

Fein, D., et al. Optimal outcome in individuals with a history of autism. J Child Psychol Psychiatry. 2013 Feb;54(2):195-205.

Feingold, B.F. Hyperkinesis and Learning Disabilities Linked to Artificial Food Flavors and Colors. Am. J. Nurs. 1975;75:797–803.

Feng, P., et al. A review of probiotics in the treatment of autism spectrum disorders: Perspectives from the gut-brain axis. Front Microbiol. 2023 Mar 16:14:1123462.

Fezer, G.F., et al. Perinatal Features of Children with Autism Spectrum Disorder. Rev Paul Pediatr. 2017 Apr-Jun;35(2):130-135.

Frisch, M., et al. Ritual circumcision and risk of autism spectrum disorder in 0- to 9-year-old boys: national cohort study in Denmark. J R Soc Med. 2015 Jul;108(7):266-79.

Frustaci, A., et al. Oxidative stress-related biomarkers in autism: systematic review and meta-analyses. Free Radic Biol Med. 2012;52(10):2128-41.

Frye, R.E., et al. Blocking and Binding Folate Receptor Alpha Autoantibodies Identify Novel Autism Spectrum Disorder Subgroups. Front Neurosci. 2016 Mar 9;10:80.

Frye, R.E., et al. Cerebral folate receptor autoantibodies in autism spectrum disorder. Mol Psychiatry. 2013 Mar;18(3):369-81.

Frye, R.E., et al. Metabolic pathology of autism in relation to redox metabolism. Biomark Med. 2014;8(3):321-30.

Frye, R.E., et al. Redox metabolism abnormalities in autistic children associated with mitochondrial disease. Transl Psychiatry. 2013;3:e273.

Gabis, L.V., et al. Improvement of Language in Children with Autism with Combined Donepezil and Choline Treatment. J Mol Neurosci. 2019 Oct;69(2):224-234.

Gabriele, S., et al. Blood serotonin levels in autism spectrum disorder: a systematic review and meta-analysis. Eur Neuropsychopharmacol. 2014;24(6):919-29.

Gadow, K.D., et al. Association of COMT (Val158Met) and BDNF (Val66Met) gene polymorphisms with anxiety, ADHD and tics in children with autism spectrum disorder. J Autism Dev Disord. 2009;39(11):1542-51.

Gadow, K.D., et al. Association of DRD4 polymorphism with severity of oppositional defiant disorder, separation anxiety disorder and repetitive behaviors in children with autism spectrum disorder. Eur J Neurosci. 2010;32(6):1058-65.

Gardender, H., et al. Perinatal and neonatal risk factors for autism: a comprehensive meta-analysis. Pediatrics. 2011 Aug;128(2):344-55.

Gebril, O.H., et al. HFE gene polymorphisms and the risk for autism in Egyptian children and impact on the effect of oxidative stress. Dis Markers. 2011;31(5):289-94.

Geier, D.A., et al. The biological basis of autism spectrum disorders: Understanding causation and treatment by clinical geneticists. Acta Neurobiol Exp (Wars). 2010;70(2):209-26.

Geier, M.R., et al. The potential importance of steroids in the treatment of autistic spectrum disorders and other disorders involving mercury toxicity. Med Hypotheses. 2005;64(5):946-54.

Ghalichi, F., et al. Effect of Gluten Free Diet on Gastrointestinal and Behavioral Indices for Children with Autism Spectrum Disorders: A Randomized Clinical Trial. World J. Pediatr. 2016;12:436–442.

Ghanizadeh, A. Increased glutamate and homocysteine and decreased glutamine levels in autism: a review and strategies for future studies of amino acids in autism. Dis Markers. 2013;35(5):281-6.

Glasson, E.J., et al. Perinatal factors and the development of autism: a population study. Arch Gen Psychiatry. 2004 Jun;61(6):618-27.

Goldani, A.A., et al. Biomarkers in autism. Front Psychiatry. 2014;5:100

Goncalves, M.V.M., et al. Pediatric acute-onset neuropsychiatric syndrome (PANS) misdiagnosed as autism spectrum disorder. Immunol Lett. 2018 Nov;203:52-53.

Gough, S., et al. Neuroprotection by the Ketogenic Diet: Evidence and Controversies. Front Nutr. 2021 Nov 23:8:782657.

Grandjean, P., et al. Developmental neurotoxicity of industrial chemicals. Lancet. 2006 Dec 16;368(9553):2167-78.

Grimaldi, R., et al. A prebiotic intervention study in children with autism spectrum disorders (ASDs). Microbiome. 2018 Aug 2;6(1):133.

Guilford, T., et al. Deficient Glutathione in the Pathophysiology of Mycotoxin-Related Illness. Toxins (Basel). 2014 Feb 10;6(2):608-23.

Guyol, G. Who’s crazy here?: Steps for recovery without drugs for: ADD/ADHD, addiction & eating disorders, anxiety & PTSD, depression, bipolar disorder, schizophrenia, autism. 1st U.S. ed. Stonington, CT: Ajoite Pub.; 2010. 123 p.p.

Hacohen, Y., et al. N‐methyl‐d‐aspartate (NMDA) receptor antibodies encephalitis mimicking an autistic regression. Dev Med Child Neurol. 2016 Oct;58(10):1092-4.

Hadjivassiliou, M., et al. Gluten sensitivity: from gut to brain. Lancet Neurol. 2010 Mar;9(3):318-30.

Hallmayer, J., et al. Genetic heritability and shared environmental factors among twin pairs with autism. Arch Gen Psychiatry. 2011 Nov;68(11):1095-102.

Hamad, A.F., et al. Prenatal antibiotics exposure and the risk of autism spectrum disorders: A population-based cohort study. PLoS One. 2019 Aug 29;14(8):e0221921.

Hamlin, J.C., et al. Dietary intake and plasma levels of choline and betaine in children with autism spectrum disorders. Autism Res Treat. 2013;2013:578429.

Hartman, R.E., et al. Dietary Approaches to the Management of Autism Spectrum Disorders. Adv. Neurobiol. 2020;24:547–571.

Hauser, P., et al. Resistance to thyroid hormone: implications for neurodevelopmental research on the effects of thyroid hormone disruptors.Toxicol Ind Health. 1998 Jan-Apr;14(1-2):85-101.

Hejitz, R.D., et al. Normal gut microbiota modulates brain development and behavior. Proc Natl Acad Sci U S A. 2011 Feb 15;108(7):3047-52.

Herbert, M.R., et al. Autism and Dietary Therapy: Case Report and Review of the Literature. J. Child. Neurol. 2013;28:975–982.

Herbert, M.R., et al. Autism and environmental genomics. Neurotoxicology. 2006;27(5):671-84.

Herbert, M.R, et al. Autism and EMF? Plausibility of a pathophysiological link–Part I. Pathophysiology 20.3 (2013): 191-209.

Herbert, M.R., et al. Autism and EMF? Plausibility of a pathophysiological link Part II. Pathophysiology 20.3 (2013): 211-234.

Herbert, M.R. Contributions of the environment and environmentally vulnerable physiology to autism spectrum disorders. Curr Opin Neurol. 2010 Apr;23(2):103-10.

Hertz-Picciotto, I., et al. The rise in autism and the role of age at diagnosis. Epidemiology. 2009 Jan;20(1):84-90.

Hertz-Picciotto, I., et al. Organophosphate exposures during pregnancy and child neurodevelopment: Recommendations for essential policy reforms. PLoS Med. 2018 Oct 24;15(10):e1002671.

Hertz-Picciotto, I., et al. Polybrominated diphenyl ethers in relation to autism and developmental delay: a case-control study. Environ Health. 2011 Jan 5;10(1):1.

Hertz-Picciotto, I., et al. Prenatal exposures to persistent and non-persistent organic compounds and effects on immune system development. Basic Clin Pharmacol Toxicol. 2008 Feb;102(2):146-54.

Heyer, N.J., et al. Disordered porphyrin metabolism: a potential biological marker for autism risk assessment. Autism Res. 2012;5(2):84-92.

Holmes, A., et al. Reduced Levels of Mercury in First Baby Haircuts of Autistic Children. International Journal of Toxicology. Jul-Aug 2003;22(4):277-85.

Hopf, K.P., et al. Use and Perceived Effectiveness of Complementary and Alternative Medicine to Treat and Manage the Symptoms of Autism in Children: A Survey of Parents in a Community Population. J. Altern. Complement. Med. 2016;22:25–32. 

Horn, J., et al. Role of Diet and Its Effects on the Gut Microbiome in the Pathophysiology of Mental Disorders. Transl. Psychiatry. 2022;12:164.

Horvath, K., et al. Autism and gastrointestinal symptoms. Curr Gastroenterol Rep. 2002 Jun;4(3):251-8.

Horvath, K., et al. Autistic disorder and gastrointestinal disease. Current Opinion in Pediatrics. 2002 Oct;14(5):583-7.

Horvath, K., et al. Gastrointestinal abnormalities in children with autistic disorder. Journal of Pediatrics. 1999 Nov;135(5):559-63.

Howsmon, D. P., et al. Multivariate techniques enable a biochemical classification of children with autism spectrum disorder versus typically‐developing peers: A comparison and validation study. Bioengineering & Translational Medicine. 2018. doi:10.1002/btm2.10095.

Hsu, C.-L., et al. The Effects of a Gluten and Casein-Free Diet in Children with Autism: A Case Report. Chang Gung Med. J. 2009;32:459–465.

Hyman, M. Autism: Is It All in the Head? Alternative Therapies in Health and Medicine. Nov-Dec 2008;14(6):12-5.

Isaksson, J., et al. Brief Report: Association Between Autism Spectrum Disorder, Gastrointestinal Problems and Perinatal Risk Factors Within Sibling Pairs. J Autism Dev Disord. 2017 Aug;47(8):2621-2627.

Ivanovski, I., et al. Aluminum in brain tissue in autism. J Trace Elem Med Biol. 2019 Jan;51:138-140.

Jadavji, N.M., et al. B-vitamin and choline supplementation increases neuroplasticity and recovery after stroke. Neurobiol Dis. 2017 Jul;103:89-100.

Jafari, M.H., et al. The Relationship Between the Level of Copper, Lead, Mercury and Autism Disorders: A Meta-Analysis. Pediatric Health, Medicine and Therapeutics. 21 Sep 2020(11):369—378.

Jafari, T., et al. The association between mercury levels and autism spectrum disorders: A systematic review and meta-analysis. J Trace Elem Med Biol. 2017 Dec;44:289-297.

James, S.J., 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.

Jarusiewicz, B. Efficacy of Neurofeedback for Children in the Autistic Spectrum: A Pilot Study. Journal of Neurotherapy. 2002;6(4).

Jaswal, V.K., et al. Eye-tracking reveals agency in assisted autistic communication. Scientific Reports. 2020 May 12;10(1):7882.

Jyonouchi, H., et al. Dysregulated innate immune responses in young children with autism spectrum disorders: their relationship to gastrointestinal symptoms and dietary intervention. Neuropsychobiology. 2005;51(2):77-85.

Jyonouchi, H., et al. Evaluation of an Association between Gastrointestinal Symptoms and Cytokine Production against Common Dietary Proteins in Children with Autism Spectrum Disorders. J. Pediatr. 2005;146:605–610.

Jyonouchi, H., et al. Impact of innate immunity in a subset of children with autism spectrum disorders: a case control study. Journal of Neuroinflammation. 2008 Nov 21;5:52.

Jyonouchi, H., et al. Innate Immunity Associated with Inflammatory Responses and Cytokine Production against Common Dietary Proteins in Patients with Autism Spectrum Disorder. Neuropsychobiology. 2002;46:76–84.

Kaluzna-Czaplinska, J., et al. Identification of organic acids as potential biomarkers in the urine of autistic children using gas chromatography/mass spectrometry. J Chromatogr B Analyt Technol Biomed Life Sci. 2014;966:70-6.

Kałużna-Czaplińska, J., et al. Nutritional Strategies and Personalized Diet in Autism-Choice or Necessity? Trends Food Sci. Technol. 2016;49:45–50.

Kane, R.C. A possible association between fetal/neonatal exposure to radiofrequency electromagnetic radiation and the increased incidence of autism spectrum disorders (ASD). Med Hypotheses. 2004;62(2):195-7.

Kang, D.W., et al. Microbiota Transfer Therapy alters gut ecosystem and improves gastrointestinal and autism symptoms: an open-label study. Microbiome. 2017 Jan 23;5(1):10.

Kang, D.W., et al.  Long-term benefit of Microbiota Transfer Therapy on autism symptoms and gut microbiota. Scientific Reports. 9, 5821 (2019).

Karagözlü, S., et al. The Relationship of Severity of Autism with Gastrointestinal Symptoms and Serum Zonulin Levels in Autistic Children. J. Autism Dev. Disord. 2022;52:623–629.

Karhu, E., et al. Nutritional interventions for autism spectrum disorder. Nutr Rev. 2020 Jul 1;78(7):515-531.

Kawicka, A., et al. How Nutritional Status, Diet and Dietary Supplements Can Affect Autism. A Review. Rocz. Panstw. Zakl. Hig. 2013;64:1–12.

Kern, J.K., et al. A biomarker of mercury body-burden correlated with diagnostic domain specific clinical symptoms of autism spectrum disorder. Biometals. 2010;23(6):1043-51.

Khan, Z., et al. Slow CCL2-dependent translocation of biopersistent particles from muscle to brain. BMC Med. 2013 Apr 4;11:99.

Knivsberg, A.M., et al. A Randomised, Controlled Study of Dietary Intervention in Autistic Syndromes. Nutr. Neurosci. 2002;5:251–261.

Kobliner, V., et al. Reduction in Obsessive Compulsive Disorder and Self-Injurious Behavior With Saccharomyces boulardii in a Child with Autism: A Case Report. Integr Med (Encinitas). 2018 Dec;17(6):38-41.

Kohen-Raz, R., et al. Postural control in children with autism. J Autism Dev Disord. 1992 Sep;22(3):419-32.

Konkel, L. Phthalates and Autistic Traits: Exploring the Association between Prenatal Exposures and Child Behavior. Environ Health Perspec. 2020 Oct;128(10):104001.

Konstantareas, M.M., et al. Ear infections in autistic and normal children. Journal of Autism and Developmental Disorders. 1987 Dec;17(4):585-94.

Konstantynowicz, J., et al. A Potential Pathogenic Role of Oxalate in Autism. Eur. J. Paediatr. Neurol. 2012;16:485–491.

Kordulewska, N.K., et al. Serum cytokine levels in children with spectrum autism disorder: Differences in pro- and anti-i