Medical & Scientific Literature

ADHD/ADD

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. 

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

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.

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.

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

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.

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.

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.      

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.

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.

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.

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).

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

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

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.

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.

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

Mossin, M.H., et al. Inverse associations 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.

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

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.

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.

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.

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.

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

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.

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.

Books

Bock, Kenneth. Healing the New Childhood Epidemics: Autism, ADHD, Asthma, and Allergies: The Groundbreaking Program for the 4-A Disorders. New York, NY. Ballantine Books, 2008.

Brandes, Bonnie. The Symphony of Reflexes: Interventions for Human Development, Autism, ADHD, CP, and Other Neurological Disorders, 2016.

Campbell-McBride, Natasha. Gut and Psychology Syndrome: Natural Treatment for Autism, Dyspraxia, A.D.D., Dyslexia, A.D.H.D., Depression, Schizophrenia, 2010.

Crook, William. Help for the Hyperactive Child: A Practical Guide Offering Parents of ADHD Children Alternatives to Ritalin. Square One, 2007

Giustra-Kozek Jennifer. Healing Without Hurting: Treating ADHD, Apraxia, and Autism Spectrum Disorders Naturally and Effectively Without Harmful Medication. Howard Beach, NY: Changing Lives Press, 2014.

Guyol, G. Who’s Crazy Here?: Steps for Recovery Without Drugs for: ADD/ADHD, Addiction & Eating Disorders, Anxiety & PTSD, Depression, Bipolar Disorder, Schizophrenia, Autism. Stonington, CT: Ajoite Pub., 2010.

Lambert, Beth, et al. Brain Under Attack: A Resource for Parents and Caregivers of Children with PANS, PANDAS, and Autoimmune Encephalitis. Answers Publications, 2018.

Mackarness, Richard. These Can Make You Ill: Not All in the Mind. BN Publishing, 2018.

Philpott, William H., et al. Brain Allergies: The Psychonutrient and Magnetic Connections. Los Angeles: Keats Publishing, 2000.

Allergies

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.

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

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.

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.

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.

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

Books

Bock, Kenneth. Healing the New Childhood Epidemics: Autism, ADHD, Asthma, and Allergies: The Groundbreaking Program for the 4-A Disorders. New York, NY. Ballantine Books, 2008.

Fraser, Heather. The Peanut Allergy Epidemic: What’s Causing It and How to Stop It. Skyhorse; Third Edition, 2017.

Jackson, Mark. Allergy: The History of a Modern Malady. London: Reaktion Books, 2007.

Walsh, William. Food Allergies: The Complete Guide to Understanding and Relieving Your Food Allergies. Hoboken, NJ: Wiley John and Sons Inc, 2000.

Asthma

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.

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.

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.

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.

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

Books

Bock, Kenneth. Healing the New Childhood Epidemics: Autism, ADHD, Asthma, and Allergies: The Groundbreaking Program for the 4-A Disorders. New York, NY. Ballantine Books, 2008.

Autism

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

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. 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.

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.

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.

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.

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.

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.

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.

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.

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.

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.

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.

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

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

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

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

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.

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.

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

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. Redox metabolism abnormalities in autistic children associated with mitochondrial disease. Transl Psychiatry. 2013;3:e273

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

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

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

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

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.

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.

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 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. 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.

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.

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.

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.

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).

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. 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.

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.

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).

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

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.

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

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.

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.

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, 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.

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.

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.

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

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

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

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

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.

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.

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.

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 (conference presentation). 2015.

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

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.

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.

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

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.

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

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.

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.

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.

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.

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.

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

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.

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.

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.

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.

 

Books

Bock, Kenneth. Healing the New Childhood Epidemics: Autism, ADHD, Asthma, and Allergies: The Groundbreaking Program for the 4-A Disorders. New York, NY. Ballantine Books, 2008.

Brandes, Bonnie. The Symphony of Reflexes: Interventions for Human Development, Autism, ADHD, CP, and Other Neurological Disorders, 2016.

Campbell-McBride, Natasha. Gut and Psychology Syndrome: Natural Treatment for Autism, Dyspraxia, A.D.D., Dyslexia, A.D.H.D., Depression, Schizophrenia, 2010.

Giustra-Kozek, Jennifer. Healing without hurting: treating ADHD, apraxia, and autism spectrum disorders naturally and effectively without harmful medication. Howard Beach, NY: Changing Lives Press, 2014.

Herbert, Martha, Weintraub Karen. The Autism Revolution: Whole-Body Strategies for Making Life All It Can Be. New York: Ballantine Books; 2012.

Hong, Maria Rickert. Almost Autism: Recovering Children from Sensory Processing Disorder, A Reference for Parents and Practitioners. 2014.

Kaufman, Raun K. Autism breakthrough: the groundbreaking method that has helped families all over the world. First edition. ed. New York: St. Martin’s Press; 2014. x, 353 p.p.

Lambert, Beth, et al. Brain Under Attack: A Resource for Parents and Caregivers of Children with PANS, PANDAS, and Autoimmune Encephalitis. Answers Publications, 2018.

Lemer, Patricia S. Outsmarting Autism: The Ultimate Guide to Management, Healing and Prevention for Individuals with Autism Spectrum Disorders. Tarentum, PA, Word Association Publishers, 2014.

Romaniec, Mary. Victory Over Autism: Lessons on Raising an Autism-Free Child. New York, NY: Skyhorse Publishing, 2015.

Sears, Robert W. The Autism Book: What Every Parent Needs to Know about Early Detection, Treatment, Recovery, and Prevention. 1st ed. New York, NY: Little, Brown, 2010.

Seroussi, Karyn. Unraveling the Mystery of Autism and Pervasive Developmental Disorder: A Mother’s Story of Research and Recovery. New York: Simon & Schuster, 2000.

Breathing Issues

Brown, S.D., Baxter, K.M., Stephenson, S.T., Esper, A.M., Brown, L.A., Fitzpatrick, A.M. Airway TGFbeta1 and oxidant stress in children with severe asthma: association with airflow limitation. Allergy Clin Immunol. 2012;129(2):388-96, 96 e 1-8.

Chronic/Recurrent Infections

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

Books

Dorfman, K. Cure Your Child with Food. New York. Workman Publishing, 2013.

Oski, R. Don’t Drink Your Milk. Third edition. TEACH Services, 2013.

Colic

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.

Developmental Delays

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

Down Syndrome

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.

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.

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.

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.

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.

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.

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

Baker, S.M. A biochemical approach to the problem of dyslexia. Journal of Learning Disabilities, 1985 18(10): 581-584.

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.

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.

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.

Books

College of Optometrists in Vision Development. Vision and Dyslexia. 2008

Johnson, Kathy. Dyslexia: Recognizing, Screening and Treating. Albany, NY. Pyramid of Potential, 2014.

Lambert, Beth, et al. Brain Under Attack: A Resource for Parents and Caregivers of Children with PANS, PANDAS, and Autoimmune Encephalitis. Answers Publications, 2018.

Emotional/Behavioral/Mood Symptoms

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.

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.

Ceylan, M.F., et al. Lipid peroxidation markers in children with anxiety disorders and their diagnostic implications. Redox Rep. 2014;19(2):92-6.

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.

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.

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.

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.

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.

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.

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

Yang, B., et al. Effects of regulating intestinal micobiota on anxiety symptoms: A systematic review. General Psychiatry. 2019; 32: e100056.

Books

Brogan, Kelly. A Mind of Your Own. The Truth About Depression and How Women Can Heal Their Bodies to Reclaim Their Lives. New York, Harper, 2016.

Campbell-McBride, Natasha. Gut and Psychology Syndrome: Natural Treatment for Autism, Dyspraxia, A.D.D., Dyslexia, A.D.H.D., Depression, Schizophrenia. 2010.

Doman, Glenn. What To Do About Your Brain-Injured Child, or Your Brain-Damaged, Mentally Retarded, Mentally Deficient, Cerebral-Palsied, Emotionally Disturbed, Spastic, Flaccid, Rigid, Epileptic, Autistic, Athetoid, Hyperactive, Down’s Child. Garden City, New York. Doubleday; 1990.

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.

Hyman, Mark. The UltraMind Solution: Fix Your Broken Brain by Healing Your Body First: The Simple Way to Defeat Depression, Overcome Anxiety and Sharpen Your Mind. 1st Scribner hardcover ed. New York: Scribner; 2008.

Gastrointestinal Disorders

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.

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.

Buccigrossi, V., et al. Functions of intestinal microflora in children. Curr Opin Gastroenterol. 2013;29(1):31-8.

Carding, S., et al. Dysbiosis of the gut microbiota in disease. Microb Ecol Health Dis. 2015;26:26191.

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.

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.

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.

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.

Manichanh, C., et al. The gut microbiota in IBD. Nat Rev Gastroenterol Hepatol. 2012;9(10):599-608.

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., 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.

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.

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.

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

Books

Campbell-McBride, Natasha. Gut and Psychology Syndrome: Natural Treatment for Autism, Dyspraxia, A.D.D., Dyslexia, A.D.H.D., Depression, Schizophrenia. 2010

Chutkin, Robin. The Microbiome Solution: A Radical New Way to Heal Your Body from the Inside Out. Avery, 2016

Infections of the Ear, Nose and Throat

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

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

Books

Schmidt, Michael. Healing Childhood Ear Infections. 1996.

Learning Disabilities

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.

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.

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.

Books

College of Optometrists in Vision Development. Vision and Dyslexia. 2008

Johnson, Kathy. Dyslexia: Recognizing, Screening and Treating. Albany, NY. Pyramid of Potential, 2014.

Lambert, Beth, et al. Brain Under Attack: A Resource for Parents and Caregivers of Children with PANS, PANDAS, and Autoimmune Encephalitis. Answers Publications, 2018.

Lyme Disease

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. 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.

Books

Buhner, Stephen Harrod. Healing Lyme Disease Coinfections: Complementary and Holistic Treatments for Bartonella and Mycoplasma. Healing Arts Press, 2013.

Buhner, Stephen Harrod. Healing Lyme: Natural Healing of Lyme Borreliosis and the Coinfections Chlamydia and Spotted Fever Rickettsiosis, 2nd Edition. Raven Press, 2015.

Buhner, Stephen Harrod. Herbal Antivirals: Natural Remedies for Emerging & Resistant Viral Infections. Storey Publishing, 2013.

Buhner, Stephen Harrod. Herbal Antibiotics, 2nd Edition: Natural Alternatives for Treating Drug-resistant Bacteria. Storey Publishing, 2012.

Buhner, Stephen Harrod. Natural Treatments for Lyme Coinfections: Anaplasma, Babesia, and Ehrlichia. Healing Arts Press, 2015.

Chutkin, Robin. The Microbiome Solution: A Radical New Way to Heal Your Body from the Inside Out. Avery, 2016.

Lambert, Beth, et al. Brain Under Attack: A Resource for Parents and Caregivers of Children with PANS, PANDAS, and Autoimmune Encephalitis. Answers Publications, 2018.

Zhang, QingCai, et al. Lyme Disease and Modern Chinese Medicine. Sino-Med Research Institute, 2006.

Mitochondrial Dysfunction

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.

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.

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., 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 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.

Obesity/Diabetes

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.

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

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.

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

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.

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.

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

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.

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.

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.

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.

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.

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.

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.

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.

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.

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.

Sanchez, M., et al. Childhood obesity: a role for gut microbiota? Int J Environ Res Public Health. 2015;12(1):162-75.

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.

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.

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

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.

Books

Bernstein, Richard K. Dr. Bernstein’s Diabetes Solution: The Complete Guide to Achieving Normal Blood Sugars. Little, Brown Spark, 2011.

Bolin, K., Cawley, J.H. The economics of obesity. 1st ed. Amsterdam; Boston: Elsevier JAI; 2007.

Wahls, Terry. The Wahls Protocol: A Radical New Way to Treat All Chronic Autoimmune Conditions Using Paleo Principles. Avery, 2020.

Wahls, Terry. The Wahls Protocol Cooking for Life: The Revolutionary Modern Paleo Plan to Treat All Chronic Autoimmune Conditions. Avery, 2017.

 

PANS/PANDAS

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.

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.

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 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.

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, Hsiuying. Anti-NMDA Receptor Encephalitis. Int J Mol Sci. 2017 Jan 18;18(1). pii: E193..

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.

 

Books

Cahalan, Susannah. Brain on Fire: My Month of Madness. Simon & Schuster, 2013.

Lambert, Beth, et al. Brain Under Attack: A Resource for Parents and Caregivers of Children with PANS, PANDAS, and Autoimmune Encephalitis. Answers Publications, 2018.

Maloney, Alison Beth. Saving Sammy: A Mother’s Fight to Cure Her Son’s OCD. Broadway Books, 2010.

O’Hara, Nancy. Demystifying PANS/PANDAS: A Functional Medicine Desktop Reference on Basal Ganglia Encephalitis. DPWN Publishing, 2022.

Seizures

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.

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. Attentiondeficit/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.

Books

Blaylock, Russell, MD. Excitotoxins: The Taste That Kills. Health Press, 1996.

Haines, Caren, RN. Silently Seizing: Common, Unrecognized, and Frequently Missed Seizures and Their Potentially Damaging Impact on Individuals With Autism Spectrum Disorders. AAPC Publishing, 2012.

Herbert, Martha, MD PhD. The Autism Revolution: Whole Body Strategies for Making Life All That It Can Be. Ballantine Books, 2013.

Lambert, Beth, et al. Brain Under Attack: A Resource for Parents and Caregivers of Children with PANS, PANDAS and Autoimmune Encephalitis. Answers Publications, 2018.

Sensory Processing Disorder

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.

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.

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.

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.

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.

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.

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.

Books

Biel, Lindsey. Raising a Sensory Smart Child: The Definitive Handbook for Helping Your Child with Sensory Processing Issues. Revised Edition, 2009

Hong, Maria Rickert. Almost Autism: Recovering Children from Sensory Processing Disorder, A Reference for Parents and Practitioners. 2014.

Kranowitz, Carol. The Out-of-Sync Child: Recognizing and Coping with Sensory Processing Disorder. Penguin Random House, revised edition 2022.

Lambert, Beth, et al. Brain Under Attack: A Resource for Parents and Caregivers of Children with PANS, PANDAS, and Autoimmune Encephalitis. Answers Publications, 2018.

Tongue Tie

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.

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.

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.

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.

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.

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

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.

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.