Cerebral folate receptor autoantibodies in autism spectrum disorder

R E Frye, J M Sequeira, E V Quadros, S J James, D A Rossignol, R E Frye, J M Sequeira, E V Quadros, S J James, D A Rossignol

Abstract

Cerebral folate deficiency (CFD) syndrome is a neurodevelopmental disorder typically caused by folate receptor autoantibodies (FRAs) that interfere with folate transport across the blood-brain barrier. Autism spectrum disorders (ASDs) and improvements in ASD symptoms with leucovorin (folinic acid) treatment have been reported in some children with CFD. In children with ASD, the prevalence of FRAs and the response to leucovorin in FRA-positive children has not been systematically investigated. In this study, serum FRA concentrations were measured in 93 children with ASD and a high prevalence (75.3%) of FRAs was found. In 16 children, the concentration of blocking FRA significantly correlated with cerebrospinal fluid 5-methyltetrahydrofolate concentrations, which were below the normative mean in every case. Children with FRAs were treated with oral leucovorin calcium (2 mg kg(-1) per day; maximum 50 mg per day). Treatment response was measured and compared with a wait-list control group. Compared with controls, significantly higher improvement ratings were observed in treated children over a mean period of 4 months in verbal communication, receptive and expressive language, attention and stereotypical behavior. Approximately one-third of treated children demonstrated moderate to much improvement. The incidence of adverse effects was low. This study suggests that FRAs may be important in ASD and that FRA-positive children with ASD may benefit from leucovorin calcium treatment. Given these results, empirical treatment with leucovorin calcium may be a reasonable and non-invasive approach in FRA-positive children with ASD. Additional studies of folate receptor autoimmunity and leucovorin calcium treatment in children with ASD are warranted.

Figures

Figure 1
Figure 1
The prevalence of blocking and binding folate receptor autoantibodies in ASD. (a) The prevalence of children with ASD for negative, low, medium and high titers of the folate receptor-blocking autoantibody. (b) The prevalence of children with ASD for negative, low, medium and high titers of the folate receptor-binding autoantibody. (c) The prevalence of being negative for both the binding and the blocking autoantibodies, being positive for only the blocking or the binding autoantibody and being positive for both the binding and the blocking autoantibodies. (d) The blocking folate receptor autoantibody titer was found to significantly decrease with age. ASD, autism spectrum disorder.
Figure 2
Figure 2
The relationship between cerebrospinal fluid 5-methyltetrahydrofolate concentrations and blocking folate receptor autoantibody titers. Lower cerebrospinal fluid 5-methyltetrahydrofolate concentrations are associated with higher blocking folate receptor autoantibody titers.
Figure 3
Figure 3
Improvement ratings for nine cognitive-behavioral dimensions for children treated with leucovorin calcium compared with the no-treatment group (a-i). Improvement is rated on a 7-point scale ranging from much worse (−3) to much better (+3). For each cognitive-behavioral dimension, we provide the score for each child treated with leucovorin on the right of each graph (filled circles), and the score for each control child who did not undergo treatment (unfilled circles). The median for each group is given by a thick line, and the mean for each group is provided by diamonds. The P-value for the Mann–Whitney U-test is provided at the middle bottom of each graph and the P-value for the t-test is provided at the middle top of each graph.
Figure 4
Figure 4
The relationship between improvement ratings in verbal communication (a, b) and expressive language (c, d) and age for children with (b, d) and without (a, c) binding folate receptor autoantibody. (a, c) For children without the binding autoantibody, improvements in verbal communication and expressive language are higher for older children than for younger children. (b, d) For children with the binding autoantibody, improvements in verbal communication and expressive language are higher for younger children than for older children. Correlation coefficients are provided for the relationship between improvement and age for each graph; hence, the P-value for these correlations are not provided.

References

    1. Ramaekers VT, Hausler M, Opladen T, Heimann G, Blau N. Psychomotor retardation, spastic paraplegia, cerebellar ataxia and dyskinesia associated with low 5-methyltetrahydrofolate in cerebrospinal fluid: a novel neurometabolic condition responding to folinic acid substitution. Neuropediatrics. 2002;33:301–308.
    1. Ramaekers VT, Blau N. Cerebral folate deficiency. Dev Med Child Neurol. 2004;46:843–851.
    1. Moretti P, Peters SU, Del Gaudio D, Sahoo T, Hyland K, Bottiglieri T, et al. Brief report: autistic symptoms, developmental regression, mental retardation, epilepsy, and dyskinesias in CNS folate deficiency. J Autism Dev Disord. 2008;38:1170–1177.
    1. Ramaekers VT, Blau N, Sequeira JM, Nassogne MC, Quadros EV. Folate receptor autoimmunity and cerebral folate deficiency in low-functioning autism with neurological deficits. Neuropediatrics. 2007;38:276–281.
    1. Ramaekers VT, Sequeira JM, Blau N, Quadros EV. A milk-free diet downregulates folate receptor autoimmunity in cerebral folate deficiency syndrome. Dev Med Child Neurol. 2008;50:346–352.
    1. Ramaekers VT, Rothenberg SP, Sequeira JM, Opladen T, Blau N, Quadros EV, et al. Autoantibodies to folate receptors in the cerebral folate deficiency syndrome. N Engl J Med. 2005;352:1985–1991.
    1. Moretti P, Sahoo T, Hyland K, Bottiglieri T, Peters S, del Gaudio D, et al. Cerebral folate deficiency with developmental delay, autism, and response to folinic acid. Neurology. 2005;64:1088–1090.
    1. James SJ, Cutler P, Melnyk S, Jernigan S, Janak L, Gaylor DW, et al. Metabolic biomarkers of increased oxidative stress and impaired methylation capacity in children with autism. Am J Clin Nutr. 2004;80:1611–1617.
    1. James SJ, Melnyk S, Jernigan S, Cleves MA, Halsted CH, Wong DH, et al. Metabolic endophenotype and related genotypes are associated with oxidative stress in children with autism. Am J Med Genet B Neuropsychiatr Genet. 2006;141:947–956.
    1. Melnyk S, Fuchs GJ, Schulz E, Lopez M, Kahler SG, Fussell JJ, et al. Metabolic imbalance associated with methylation dysregulation and oxidative damage in children with autism. J Autism Dev Disord. published online 26 April 2011.
    1. Pogribny IP, Karpf AR, James SR, Melnyk S, Han T, Tryndyak VP. Epigenetic alterations in the brains of Fisher 344 rats induced by long-term administration of folate/methyl-deficient diet. Brain Res. 2008;1237:25–34.
    1. Nagarajan RP, Hogart AR, Gwye Y, Martin MR, LaSalle JM. Reduced MeCP2 expression is frequent in autism frontal cortex and correlates with aberrant MECP2 promoter methylation. Epigenetics. 2006;1:e1–11.
    1. Sajdel-Sulkowska EM, Xu M, McGinnis W, Koibuchi N. Brain region-specific changes in oxidative stress and neurotrophin levels in autism spectrum disorders (ASD) Cerebellum. 2010;10:43–48.
    1. Ramaekers VT, Hansen SI, Holm J, Opladen T, Senderek J, Hausler M, et al. Reduced folate transport to the CNS in female Rett patients. Neurology. 2003;61:506–515.
    1. Chauhan A, Chauhan V. Oxidative stress in autism. Pathophysiology. 2006;13:171–181.
    1. Chauhan A, Gu F, Essa MM, Wegiel J, Kaur K, Ted Brown W, et al. Brain region-specific deficit in mitochondrial electron transport chain complexes in children with autism. J Neurochem. 2011;117:209–220.
    1. Duthie SJ, Hawdon A. DNA instability (strand breakage, uracil misincorporation, and defective repair) is increased by folic acid depletion in human lymphocytes in vitro. FASEB J. 1998;12:1491–1497.
    1. Crott JW, Mashiyama ST, Ames BN, Fenech M. The effect of folic acid deficiency and MTHFR C677T polymorphism on chromosome damage in human lymphocytes in vitro. Cancer Epidemiol Biomarkers Prev. 2001;10:1089–1096.
    1. Iskandar BJ, Rizk E, Meier B, Hariharan N, Bottiglieri T, Finnell RH, et al. Folate regulation of axonal regeneration in the rodent central nervous system through DNA methylation. J Clin Invest. 2010;120:1603–1616.
    1. Wollack JB, Makori B, Ahlawat S, Koneru R, Picinich SC, Smith A, et al. Characterization of folate uptake by choroid plexus epithelial cells in a rat primary culture model. J Neurochem. 2008;104:1494–1503.
    1. Solanky N, Requena Jimenez A, D'Souza SW, Sibley CP, Glazier JD. Expression of folate transporters in human placenta and implications for homocysteine metabolism. Placenta. 2010;31:134–143.
    1. Weitman SD, Weinberg AG, Coney LR, Zurawski VR, Jennings DS, Kamen BA. Cellular localization of the folate receptor: potential role in drug toxicity and folate homeostasis. Cancer Res. 1992;52:6708–6711.
    1. Molloy AM, Quadros EV, Sequeira JM, Troendle JF, Scott JM, Kirke PN, et al. Lack of association between folate-receptor autoantibodies and neural-tube defects. N Engl J Med. 2009;361:152–160.
    1. Berrocal-Zaragoza MI, Fernandez-Ballart JD, Murphy MM, Cavalle-Busquets P, Sequeira JM, Quadros EV. Association between blocking folate receptor autoantibodies and subfertility. Fertil Steril. 2009;91 (4 Suppl:1518–1521.
    1. Ramaekers VT, Weis J, Sequeira JM, Quadros EV, Blau N. Mitochondrial complex I encephalomyopathy and cerebral 5-methyltetrahydrofolate deficiency. Neuropediatrics. 2007;38:184–187.
    1. Garcia-Cazorla A, Quadros EV, Nascimento A, Garcia-Silva MT, Briones P, Montoya J, et al. Mitochondrial diseases associated with cerebral folate deficiency. Neurology. 2008;70:1360–1362.
    1. O'Byrne MR, Au KS, Morrison AC, Lin JI, Fletcher JM, Ostermaier KK, et al. Association of folate receptor (FOLR1, FOLR2, FOLR3) and reduced folate carrier (SLC19A1) genes with meningomyelocele. Birth Defects Res. 2010;88:689–694.
    1. Matherly LH, Hou Z, Deng Y. Human reduced folate carrier: translation of basic biology to cancer etiology and therapy. Cancer Metastasis Rev. 2007;26:111–128.
    1. Yuasa H, Inoue K, Hayashi Y. Molecular and functional characteristics of proton-coupled folate transporter. J Pharma Sci. 2009;98:1608–1616.
    1. Hou Z, Matherly LH. Oligomeric structure of the human reduced folate carrier: identification of homo-oligomers and dominant-negative effects on carrier expression and function. J Biol Chem. 2009;284:3285–3293.
    1. Hansen FJ, Blau N. Cerebral folate deficiency: life-changing supplementation with folinic acid. Mol Genet Metab. 2005;84:371–373.
    1. APA Diagnostic and Statistical Manual of Mental Disorders4th ednAmerican Psychiatric Association: Washington, DC; 1994
    1. Rossignol DA, Frye RE. Mitochondrial dysfunction in autism spectrum disorders: a systematic review and meta-analysis. Mol Psychiatry.
    1. Frye RE, Rossignol DA. Mitochondrial dysfunction can connect the diverse medical symptoms associated with autism spectrum disorders. Pediatric Res. 2011;69 (5 Pt 2:41R–47R.
    1. Guy W. ECDEU Assessment Manual for Psychopharmacology - Revised (DHEW Publ No ADM 76-338). Rockville, MD, U.S. Department of Health, Education, and Welfare, Public Health Service, Alcohol, Drug Abuse, and Mental Health Administration, NIMH Psychopharmacology Research Branch, Division of Extramural Research Programs. 1976. pp. 218–222.
    1. Hulley SB, Cummings SR, Browner WS, Grady DG, Newman TB.Designing Clinical Research3rd ednLippincott Williams & Wilkins: Philadephia; 2006
    1. Faul F, Erdfelder E, Lang AG, Buchner A. G*Power 3: a flexible statistical power analysis for the social, behavioral, and biomedical sciences. Behavior Res Methods. 2007;39:175–191.
    1. Berrocal-Zaragoza MI, Fernandez-Ballart JD, Murphy MM, Cavalle-Busquets P, Sequeira JM, Quadros EV. Association between blocking folate receptor autoantibodies and subfertility. Fertil Steril. 2009;91:1518–1521.
    1. Berrocal-Zaragoza MI, Murphy MM, Ceruelo S, Quadros EV, Sequeira JM, Fernandez-Ballart JD. High milk consumers have an increased risk of folate receptor blocking autoantibody production but this does not affect folate status in Spanish men and women. J Nutr. 2009;139:1037–1041.
    1. Koenig MK, Perez M, Rothenberg S, Butler IJ. Juvenile onset central nervous system folate deficiency and rheumatoid arthritis. J Child Neurol. 2008;23:106–107.
    1. Mohammad NS, Jain JM, Chintakindi KP, Singh RP, Naik U, Akella RR. Aberrations in folate metabolic pathway and altered susceptibility to autism. Psychiatr Genet. 2009;19:171–176.
    1. Boris M, Goldblatt A, Galanko J, James SJ. Association of MTHFR gene variants with autism. J Am Physicians Surgeons. 2004;9:106–108.
    1. Adams M, Lucock M, Stuart J, Fardell S, Baker K, Ng X. Preliminary evidence for involvement of the folate gene polymorphism 19bp deletion-DHFR in occurrence of autism. Neurosci Lett. 2007;422:24–29.
    1. Hyland K, Shoffner J, Heales SJ. Cerebral folate deficiency. J Inherit Metab Dis. 2010;33:563–570.
    1. Miller AL. The methylation, neurotransmitter, and antioxidant connections between folate and depression. Altern Med Rev. 2008;13:216–226.
    1. Neustadt J, Pieczenik SR. Medication-induced mitochondrial damage and disease. Mol Nutr Food Res. 2008;52:780–788.
    1. Ahn MS, Sims KB, Frazier JA. Risperidone-induced psychosis and depression in a child with a mitochondrial disorder. J Child Adolesc Psychopharmacol. 2005;15:520–525.
    1. Thomas RH, Foley KA, Mepham JR, Tichenoff LJ, Possmayer F, MacFabe DF. Altered brain phospholipid and acylcarnitine profiles in propionic acid infused rodents: further development of a potential model of autism spectrum disorders. J Neurochem. 2010;113:515–529.
    1. MacFabe DF, Cain DP, Rodriguez-Capote K, Franklin AE, Hoffman JE, Boon F, et al. Neurobiological effects of intraventricular propionic acid in rats: possible role of short chain fatty acids on the pathogenesis and characteristics of autism spectrum disorders. Behav Brain Res. 2007;176:149–169.
    1. Nikolov RN, Bearss KE, Lettinga J, Erickson C, Rodowski M, Aman MG, et al. Gastrointestinal symptoms in a sample of children with pervasive developmental disorders. J Autism Dev Disord. 2009;39:405–413.
    1. James SJ, Melnyk S, Fuchs G, Reid T, Jernigan S, Pavliv O, et al. Efficacy of methylcobalamin and folinic acid treatment on glutathione redox status in children with autism. Am J Clin Nutr. 2009;89:425–430.
    1. Croen LA, Braunschweig D, Haapanen L, Yoshida CK, Fireman B, Grether JK, et al. Maternal mid-pregnancy autoantibodies to fetal brain protein: the early markers for autism study. Biol Psychiatry. 2008;64:583–588.
    1. Wills S, Cabanlit M, Bennett J, Ashwood P, Amaral D, Van de Water J. Autoantibodies in autism spectrum disorders (ASD) Ann NY Acad Sci. 2007;1107:79–91.
    1. Wills S, Rossi CC, Bennett J, Cerdeno VM, Ashwood P, Amaral DG, et al. Further characterization of autoantibodies to GABAergic neurons in the central nervous system produced by a subset of children with autism. Mol Autism. 2011;2:5.
    1. Wills S, Cabanlit M, Bennett J, Ashwood P, Amaral DG, Van de Water J. Detection of autoantibodies to neural cells of the cerebellum in the plasma of subjects with autism spectrum disorders. Brain Behav Immun. 2009;23:64–74.
    1. Braunschweig D, Ashwood P, Krakowiak P, Hertz-Picciotto I, Hansen R, Croen LA, et al. Autism: maternally derived antibodies specific for fetal brain proteins. Neurotoxicology. 2008;29:226–231.
    1. James SJ, Melnyk S, Jernigan S, Hubanks A, Rose S, Gaylor DW. Abnormal transmethylation/transsulfuration metabolism and DNA hypomethylation among parents of children with autism. J Autism Dev Disord. 2008;38:1966–1975.
    1. Goin-Kochel RP, Porter AE, Peters SU, Shinawi M, Sahoo T, Beaudet AL. The MTHFR 677C-->T polymorphism and behaviors in children with autism: exploratory genotype-phenotype correlations. Autism Res. 2009;2:98–108.
    1. Finegold SM, Molitoris D, Song Y, Liu C, Vaisanen ML, Bolte E, et al. Gastrointestinal microflora studies in late-onset autism. Clin Infect Dis. 2002;35 (Suppl 1:S6–S16.
    1. Finegold SM. State of the art; microbiology in health and disease intestinal bacterial flora in autism. Anaerobe.
    1. Williams BL, Hornig M, Buie T, Bauman ML, Cho Paik M, Wick I, et al. Impaired carbohydrate digestion and transport and mucosal dysbiosis in the intestines of children with autism and gastrointestinal disturbances. PLoS One. 2011;6:e24585.
    1. MacFabe DF, Thomas RH, Foley KA, Mepham JR, Tichenoff LJ, Possmayer F. Altered brain phospholipid and acylcarnitine profiles in propionic acid infused rodents: further development of a potential model of autism spectrum disorders. J Neurochem. 2010;113:515–529.
    1. MacFabe DF, Rodríguez-Capote K, Hoffman JE, Franklin AE, Mohammad-Asef Y, Taylor AR, et al. A novel rodent model of autism: intraventricular infusions of propionic acid increase locomotor activity and induce neuroinflammation and oxidative stress in discrete regions of adult rat brain. Am J Biochem Biotech. 2008;4:146–166.
    1. Sajdel-Sulkowska EM, Xu M, Koibuchi N. Increase in cerebellar neurotrophin-3 and oxidative stress markers in autism. Cerebellum. 2009;8:366–372.
    1. Filipek PA, Juranek J, Nguyen MT, Cummings C, Gargus JJ. Relative carnitine deficiency in autism. J Autism Dev Disord. 2004;34:615–623.
    1. Whiteley P, Haracopos D, Knivsberg AM, Reichelt KL, Parlar S, Jacobsen J, et al. The ScanBrit randomised, controlled, single-blind study of a gluten- and casein-free dietary intervention for children with autism spectrum disorders. Nutr Neurosci. 2010;13:87–100.
    1. Rice C. Prevalence of autism spectrum disorders –autism and developmental disabilities monitoring network, United States, 2006. MMWR Surveill Summ. 2009;58:1–20.
    1. Salojin KV, Cabrera RM, Sun W, Chang WC, Lin C, Duncan L, et al. A mouse model of hereditary folate malabsorption: deletion of the PCFT gene leads to systemic folate deficiency. Blood. 2011;117:4895–4904.

Source: PubMed

3
Suscribir