Unique acyl-carnitine profiles are potential biomarkers for acquired mitochondrial disease in autism spectrum disorder

R E Frye, S Melnyk, D F Macfabe, R E Frye, S Melnyk, D F Macfabe

Abstract

Autism spectrum disorder (ASD) has been associated with mitochondrial disease (MD). Interestingly, most individuals with ASD and MD do not have a specific genetic mutation to explain the MD, raising the possibility of that MD may be acquired, at least in a subgroup of children with ASD. Acquired MD has been demonstrated in a rodent ASD model in which propionic acid (PPA), an enteric bacterial fermentation product of ASD-associated gut bacteria, is infused intracerebroventricularly. This animal model shows validity as it demonstrates many behavioral, metabolic, neuropathologic and neurophysiologic abnormalities associated with ASD. This animal model also demonstrates a unique pattern of elevations in short-chain and long-chain acyl-carnitines suggesting abnormalities in fatty-acid metabolism. To determine if the same pattern of biomarkers of abnormal fatty-acid metabolism are present in children with ASD, the laboratory results from a large cohort of children with ASD (n=213) who underwent screening for metabolic disorders, including mitochondrial and fatty-acid oxidation disorders, in a medically based autism clinic were reviewed. Acyl-carnitine panels were determined to be abnormal if three or more individual acyl-carnitine species were abnormal in the panel and these abnormalities were verified by repeated testing. Overall, 17% of individuals with ASD demonstrated consistently abnormal acyl-carnitine panels. Next, it was determined if specific acyl-carnitine species were consistently elevated across the individuals with consistently abnormal acyl-carnitine panels. Significant elevations in short-chain and long-chain, but not medium-chain, acyl-carnitines were found in the ASD individuals with consistently abnormal acyl-carnitine panels-a pattern consistent with the PPA rodent ASD model. Examination of electron transport chain function in muscle and fibroblast culture, histological and electron microscopy examination of muscle and other biomarkers of mitochondrial metabolism revealed a pattern consistent with the notion that PPA could be interfering with mitochondrial metabolism at the level of the tricarboxylic-acid cycle (TCAC). The function of the fatty-acid oxidation pathway in fibroblast cultures and biomarkers for abnormalities in non-mitochondrial fatty-acid metabolism were not consistently abnormal across the subgroup of ASD children, consistent with the notion that the abnormalities in fatty-acid metabolism found in this subgroup of children with ASD were secondary to TCAC abnormalities. Glutathione metabolism was abnormal in the subset of ASD individuals with consistent acyl-carnitine panel abnormalities in a pattern similar to glutathione abnormalities found in the PPA rodent model of ASD. These data suggest that there are similar pathological processes between a subset of ASD children and an animal model of ASD with acquired mitochondrial dysfunction. Future studies need to identify additional parallels between the PPA rodent model of ASD and this subset of ASD individuals with this unique pattern of acyl-carnitine abnormalities. A better understanding of this animal model and subset of children with ASD should lead to better insight in mechanisms behind environmentally induced ASD pathophysiology and should provide guidance for developing preventive and symptomatic treatments.

Figures

Figure 1
Figure 1
Acyl-carnitine elevations in the brain of rats treated intracerebroventricularly with propionic acid. Notice that the majority of fatty-acid elevations were in short-chain (2–5 carbon length) and long-chain (13–18 carbon length) fatty-acids as compared with the medium-chain (6–12 carbon length) fatty-acids. This is adapted from Thomas et al. where it was presented as a table.
Figure 2
Figure 2
Algorithm for metabolic workup of autistic spectrum disease patients evaluated in the medically based autism clinic. Patients are screened with biomarkers of abnormal mitochondrial function in the fasting state. Abnormalities are verified with repeat fasting biomarker testing. For patients with biomarkers for a fatty-acid oxidation defect, other disorders of fatty-acid metabolism are ruled-out before further workup for a mitochondrial disorder. Patients with consistent biomarkers for mitochondrial dysfunction are first investigated for genetic causes of their mitochondrial disorder before considering a muscle and/or skin biopsy. mtDNA, mitochondrial deoxyribonucleic acid; RBC, red-blood cell.
Figure 3
Figure 3
Average acyl-carnitine values (with s.e. bars) from 20 patients with consistent abnormal elevations in multiple acyl-carnitines. Acyl-carnitine values are represented as percent upper limit of normal for each acyl-carnitine species. Notice that C4OH, C14 and C16:1 are significantly elevated as compared with the maximum upper limit of normal.
Figure 4
Figure 4
Gluthathione abnormalities in four children with consistent elevations in multiple acyl-carnitine species. Notice that the patients have lower total (tGSH, μM) and free (fGSH, μM) reduced gluthathione, as well as lower tGSH/fGSSG (free-oxidized gluthathione, μM) and fGSH/fGSSG ratios and higher fGSSG as compared with typically developing controls, suggesting both a reduction in the production of gluthathione and increase in gluthathione utilization by reactive species.
Figure 5
Figure 5
Electron transport chain (ETC) function of muscle (a, b) and fibroblast culture (c, d), as well as function of the fatty-acid oxidation pathway in fibroblast cultures (e, f). Graph values represent percent of normal ETC function, uncorrected (a, c) or corrected for citrate synthase (b, d). Muscle ETC results suggest a partial defect in complexes I/III and I/III rotenone sensitive (RS) while fibroblast culture ETC function suggests a partial defect in complex II/III activity. In fibroblast culture ETC studies complexes I/III RS and IV demonstrate considerable variability due to overactivity (>200% of the mean) in complex I/III RS in three patients and complex IV in one case. Fatty-acid oxidation values represent mean of specific acyl-carnitine species (higher is worse) uncorrected (e) and corrected for citrate synthase (f). Elevation in the short-chain fatty-acid D3-C4 was due to three patients demonstrating high D4-C4 values. The one patient with a significantly elevated D4-C4 value was found not to have a mutation in ;the short-chain acyl-CoA dehydrogenase gene suggesting that the abnormalities in fatty-acids in fibroblast culture were due to other mitochondrial metabolism abnormalities.
Figure 6
Figure 6
The tricarboxylic-acid cycle during (a) typical metabolism and (b) with high levels of propionic acid. Propionic acid is metabolized to propionyl-CoA, which inhibits the proximal portion of the tricarboxylic-acid cycle and enhances the distal portion of the tricarboxylic-acid cycle (see discussion for details). FADH2, flavin adenine dinucleotide; NADH, nicotinamide adenine dinucleotide.

References

    1. APA Diagnostic and statistical manual of mental disorders4th ednDC: American Psychiatric Association: Washington; 1994
    1. Frye RE, Rossignol DA. Mitochondrial dysfunction can connect the diverse medical symptoms associated with autism spectrum disorders. Pediatr Res. 2011;69 (5 Pt 2:41R–47RR.
    1. Rossignol D, Frye RE. Mitochondrial dysfunction in autism spectrum disorders: A systematic review and meta-analysis. Mol Psychiatry. 2012;17:290–314.
    1. Rossignol DA, Frye RE. A review of research trends in physiological abnormalities in autism spectrum disorders: immune dysregulation, inflammation, oxidative stress, mitochondrial dysfunction and environmental toxicant exposures. Mol Psychiatry. 2012;17:389–401.
    1. Weissman JR, Kelley RI, Bauman ML, Cohen BH, Murray KF, Mitchell RL, et al. Mitochondrial disease in autism spectrum disorder patients: a cohort analysis. PLoS ONE. 2008;3:e3815.
    1. Giulivi C, Zhang YF, Omanska-Klusek A, Ross-Inta C, Wong S, Hertz-Picciotto I, et al. Mitochondrial dysfunction in autism. JAMA. 2010;304:2389–2396.
    1. Hallmayer J, Cleveland S, Torres A, Phillips J, Cohen B, Torigoe T, et al. Genetic heritability and shared environmental factors among twin pairs with autism. Arch Gen Psychiatry. 2011;68:1095–1102.
    1. Herbert MR, Russo JP, Yang S, Roohi J, Blaxill M, Kahler SG, et al. Autism and environmental genomics. Neurotoxicology. 2006;27:671–684.
    1. Ashwood P, Anthony A, Torrente F, Wakefield AJ. Spontaneous mucosal lymphocyte cytokine profiles in children with autism and gastrointestinal symptoms: mucosal immune activation and reduced counter regulatory interleukin-10. J Clin Immunol. 2004;24:664 73.
    1. MacFabe DF, Rodríguez-Capote K, Hoffman JE. 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 Biotechnol. 2008;4:146–166.
    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. Shultz SR, MacFabe DF, Ossenkopp KP, Scratch S, Whelan J, Taylor R, et al. Intracerebroventricular injection of propionic acid, an enteric bacterial metabolic end-product, impairs social behavior in the rat: implications for an animal model of autism. Neuropharmacology. 2008;54:901–911.
    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 NE, Boon F, Ossenkopp KP, Cain DP. Effects of the enteric bacterial metabolic product propionic acid on object-directed behavior, social behavior, cognition, and neuroinflammation in adolescent rats: relevance to autism spectrum disorder. Behav Brain Res. 2011;217:47–54.
    1. Shultz SR, Macfabe DF, Martin S, Jackson J, Taylor R, Boon F, et al. Intracerebroventricular injections of the enteric bacterial metabolic product propionic acid impair cognition and sensorimotor ability in the Long-Evans rat: further development of a rodent model of autism. Behav Brain Res. 2009;200:33–41.
    1. Thomas RH, Meeking MM, Mepham JR, Tichenoff L, Possmayer F, Liu S, 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.
    1. MacFabe DF. Short-chain fatty acid fermentation products of the gut microbiome: implications in autism spectrum disorders. Microb Ecol Health Dis. 2012;23:19260.
    1. Finegold SM, Dowd SE, Gontcharova V, Liu C, Henley KE, Wolcott RD, et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe. 2010;16:444–453.
    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. Wajner M, Latini A, Wyse AT, Dutra-Filho CS. The role of oxidative damage in the neuropathology of organic acidurias: insights from animal studies. J Inherit Metab Dis. 2004;27:427–448.
    1. Coulter DL. Carnitine, valproate, and toxicity. J Child Neurol. 1991;6:7–14.
    1. Calabrese V, Rizza V. Formation of propionate after short-term ethanol treatment and its interaction with the carnitine pool in rat. Alcohol. 1999;19:169–176.
    1. Al-Owain M, Kaya N, Al-Shamrani H, Al-Bakheet A, Qari A, Al-Muaigl S, et al. Autism spectrum disorder in a child with propionic acidemia. JIMD Reports. 2012;7:63–66.
    1. Mostafa GA, El-Gamal HA, El-Wakkad ASE, El-Shorbagy OE, Hamza MM. Polyunsaturated fatty acids, carnitine and lactate as biological markers of brain energy in autistic children. Int J Child Neuropsychiatry. 2005;2:179–188.
    1. Haas RH, Parikh S, Falk MJ, Saneto RP, Wolf NI, Darin N, et al. The in-depth evaluation of suspected mitochondrial disease. Mol Genet Metab. 2008;94:16–37.
    1. Pastural E, Ritchie S, Lu Y, Jin W, Kavianpour A, Khine Su-Myat K, et al. Novel plasma phospholipid biomarkers of autism: mitochondrial dysfunction as a putative causative mechanism. Prostaglandins Leukot Essent Fatty Acids. 2009;81:253–264.
    1. Clark-Taylor T, Clark-Taylor BE. Is autism a disorder of fatty acid metabolism? possible dysfunction of mitochondrial beta-oxidation by long chain acyl-CoA dehydrogenase. Med Hypotheses. 2004;62:970–975.
    1. Frye RE. Biomarkers of abnormal energy metabolism in children with autism spectrum disorder. NAJ Med Sci. 2012;5:141–147.
    1. Munnich A, Rustin P. Clinical spectrum and diagnosis of mitochondrial disorders. Am J Med Genet. 2001;106:4–17.
    1. Mitochondrial Medicine Society's Committee on D. Haas RH, Parikh S, Falk MJ, Saneto RP, Wolf NI, Darin N, et al. The in-depth evaluation of suspected mitochondrial disease. Mol Genet Metab. 2008;94:16–37.
    1. Wong LJ. Pathogenic mitochondrial DNA mutations in protein-coding genes. Muscle Nerve. 2007;36:279–293.
    1. Wong LJ, Cobb BR, Chen TJ. Molecular analysis of mitochondrial DNA point mutations by polymerase chain reaction. Methods Mol Biol. 2006;336:135–143.
    1. Bai RK, Perng CL, Hsu CH, Wong LJ. Quantitative PCR analysis of mitochondrial DNA content in patients with mitochondrial disease. Ann N Y Acad Sci. 2004;1011:304–309.
    1. Bai RK, Wong LJ. Simultaneous detection and quantification of mitochondrial DNA deletion(s), depletion, and over-replication in patients with mitochondrial disease. J Mol Diagn. 2005;7:613–622.
    1. Kirby DM, Thorburn DR, Turnbull DM, Taylor RW. Biochemical assays of respiratory chain complex activity. Methods Cell Biol. 2007;80:93–119.
    1. Roe CR, Roe DS. Recent developments in the investigation of inherited metabolic disorders using cultured human cells. Mol Genet Metab. 1999;68:243–257.
    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. Melnyk S, Pogribna M, Pogribny I, Hine RJ, James SJ. A new HPLC method for the simultaneous determination of oxidized and reduced plasma aminothiols using coulometric electrochemical detection. J Nutr Biochem. 1999;10:490–497.
    1. Corydon MJ, Vockley J, Rinaldo P, Rhead WJ, Kjeldsen M, Winter V, et al. Role of common gene variations in the molecular pathogenesis of short-chain acyl-CoA dehydrogenase deficiency. Pediatr Res. 2001;49:18–23.
    1. Morava E, van den Heuvel L, Hol F, de Vries MC, Hogeveen M, Rodenburg RJ, et al. Mitochondrial disease criteria: diagnostic applications in children. Neurology. 2006;67:1823–1826.
    1. Graf WD, Marin-Garcia J, Gao HG, Pizzo S, Naviaux RK, Markusic D, et al. Autism associated with the mitochondrial DNA G8363A transfer RNA(Lys) mutation. J Child Neurol. 2000;15:357–361.
    1. Frye RE, Naviaux RK. Autistic disorder with complex IV overactivity: a new mitochondrial syndrome. J Ped Neurol. 2011;9:427–434.
    1. Gil Borlado MC, Moreno Lastres D, Gonzalez Hoyuela M, Moran M, Blazquez A, Pello R, et al. Impact of the mitochondrial genetic background in complex III deficiency. PloS one. 2010;5:e12801.
    1. Schwab MA, Sauer SW, Okun JG, Nijtmans LG, Rodenburg RJ, van den Heuvel LP, et al. Secondary mitochondrial dysfunction in propionic aciduria: a pathogenic role for endogenous mitochondrial toxins. Biochem J. 2006;398:107–112.
    1. Gil Borlado MC, Moreno Lastres D, Gonzalez Hoyuela M, Moran M, Blazquez A, Pello R, et al. Impact of the mitochondrial genetic background in complex III deficiency. PLoS ONE. 2010;5:e12801.
    1. Frye RE. Novel cytochrome b gene mutations causing mitochondrial disease in autism. J Ped Neurol. 2012;10:1–6.
    1. Ashwood P, Van de Water J. Is autism an autoimmune disease? autoimmunity reviews. 2004;3:557–562.
    1. Ashwood P, Van de Water J. A review of autism and the immune response. Clin Dev Immunol. 2004;11:165–174.
    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;141B:947–956.
    1. Chauhan A, Chauhan V. Oxidative stress in autism. Pathophysiology. 2006;13:171–181.
    1. Wegiel J, Kuchna I, Nowicki K, Imaki H, Wegiel J, Marchi E, et al. The neuropathology of autism: defects of neurogenesis and neuronal migration, and dysplastic changes. Acta Neuropathol. 2010;119:755–770.
    1. Faber S, Zinn GM, Kern JC, Kingston HM. The plasma zinc/serum copper ratio as a biomarker in children with autism spectrum disorders. Biomarkers. 2009;14:171–180.
    1. Yasuda H, Yoshida K, Yasuda Y, Tsutsui T. Infantile zinc deficiency: association with autism spectrum disorders. Scientific reports. 2011;1:129.
    1. Russo AJ, Devito R. Analysis of copper and zinc plasma concentration and the efficacy of zinc therapy in individuals with asperger's syndrome, pervasive developmental disorder not otherwise specified (PDD-NOS) and autism. Biomark Insights. 2011;6:127–133.
    1. Sikora SK, Spady D, Prosser C, El-Matary W. Trace elements and vitamins at diagnosis in pediatric-onset inflammatory bowel disease. Clin Pediatr. 2011;50:488–492.
    1. Iwaya H, Kashiwaya M, Shinoki A, Lee JS, Hayashi K, Hara H, et al. Marginal zinc deficiency exacerbates experimental colitis induced by dextran sulfate sodium in rats. J Nutr. 2011;141:1077–1082.
    1. Scrimgeour AG, Condlin ML. Zinc and micronutrient combinations to combat gastrointestinal inflammation. Curr Opin Clin Nutr Metab Care. 2009;12:653–660.
    1. Barollo M, Medici V, D'Inca R, Banerjee A, Ingravallo G, Scarpa M, et al. Antioxidative potential of a combined therapy of anti TNFalpha and Zn acetate in experimental colitis. World J Gastroenterol. 2011;17:4099–4103.
    1. Tran CD, Ball JM, Sundar S, Coyle P, Howarth GS. The role of zinc and metallothionein in the dextran sulfate sodium-induced colitis mouse model. Dig Dis Sci. 2007;52:2113–2121.
    1. Zhang Y, Okamura S, Kudo T, Masuo T, Mori M. Calcineurin inhibition by polaprezinc in rats with experimentally-induced colitis. Life Sci. 2011;88:432–439.
    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. Wang L, Christophersen CT, Sorich MJ, Gerber JP, Angley MT, Conlon MA. Elevated fecal short chain fatty acid and ammonia concentrations in children with autism spectrum disorder. Dig Dis Sci. 2012;57:2096–2102.
    1. Barton LL, Fauque GD. Biochemistry, physiology and biotechnology of sulfate-reducing bacteria. Adv Appl Microbiol. 2009;68:41–98.
    1. Lagoutte E, Mimoun S, Andriamihaja M, Chaumontet C, Blachier F, Bouillaud F. Oxidation of hydrogen sulfide remains a priority in mammalian cells and causes reverse electron transfer in colonocytes. Biochim Biophys Acta. 2010;1797:1500–1511.
    1. Fallon J. Could one of the most widely prescribed antibiotics amoxicillin/clavulanate "augmentin" be a risk factor for autism. Med Hypotheses. 2005;64:312–315.
    1. Sandler RH, Finegold SM, Bolte ER, Buchanan CP, Maxwell AP, Vaisanen ML, et al. Short-term benefit from oral vancomycin treatment of regressive-onset autism. J Child Neurol. 2000;15:429–435.
    1. Mellon AF, Deshpande SA, Mathers JC, Bartlett K. Effect of oral antibiotics on intestinal production of propionic acid. Arch Dis Child. 2000;82:169–172.
    1. Yorifuji T, Kawai M, Muroi J, Mamada M, Kurokawa K, Shigematsu Y, et al. Unexpectedly high prevalence of the mild form of propionic acidemia in Japan: presence of a common mutation and possible clinical implications. Hum Genet. 2002;111:161–165.
    1. Perez B, Desviat LR, Rodriguez-Pombo P, Clavero S, Navarrete R, Perez-Cerda C, et al. Propionic acidemia: identification of twenty-four novel mutations in Europe and North America. Mol Genet Metab. 2003;78:59–67.
    1. DeCastro M, Nankova BB, Shah P, Patel P, Mally PV, Mishra R, et al. Short chain fatty acids regulate tyrosine hydroxylase gene expression through a cAMP-dependent signaling pathway. Brain Res Mol Brain Res. 2005;142:28–38.
    1. Nakao S, Moriya Y, Furuyama S, Niederman R, Sugiya H. Propionic acid stimulates superoxide generation in human neutrophils. Cell Biol Int. 1998;22:331–337.
    1. Nguyen NH, Morland C, Gonzalez SV, Rise F, Storm-Mathisen J, Gundersen V, et al. Propionate increases neuronal histone acetylation, but is metabolized oxidatively by glia. Relevance for propionic acidemia. J Neurochem. 2007;101:806–814.
    1. Conn AR, Fell DI, Steele RD. Characterization of alpha-keto acid transport across blood-brain barrier in rats. Am J Physiol. 1983;245:E253–E260.
    1. Hara H, Haga S, Aoyama Y, Kiriyama S. Short-chain fatty acids suppress cholesterol synthesis in rat liver and intestine. J Nutr. 1999;129:942–948.
    1. Le Poul E, Loison C, Struyf S. Functional characterization of human receptors for short chain fatty acids and their role in polymorphonuclear cell activation. J Biol Chem. 2003;278:25481–25489.
    1. Rorig B, Klausa G, Sutor B. Intracellular acidification reduced gap junction coupling between immature rat neocortical pyramidal neurones. J Physiol. 1996;490 (Pt 1:31–49.
    1. Brass EP. Interaction of carnitine and propionate with pyruvate oxidation by hepatocytes from clofibrate-treated rats: importance of coenzyme A availability. J Nutr. 1992;122:234–240.
    1. Chandler RJ, Zerfas PM, Shanske S, Sloan J, Hoffmann V, DiMauro S, et al. Mitochondrial dysfunction in mut methylmalonic acidemia. The FASEB Journal. 2009;23:1252–1261.
    1. Pettenuzzo LF, Ferreira Gda C, Schmidt AL, Dutra-Filho CS, Wyse AT, Wajner M. Differential inhibitory effects of methylmalonic acid on respiratory chain complex activities in rat tissues. International J Dev Neurosci. 2006;24:45–52.
    1. Brusque AM, Borba Rosa R, Schuck PF, Dalcin KB, Ribeiro CA, Silva CG, et al. Inhibition of the mitochondrial respiratory chain complex activities in rat cerebral cortex by methylmalonic acid. Neurochem Int. 2002;40:593–601.
    1. Miecz D, Januszewicz E, Czeredys M, Hinton BT, Berezowski V, Cecchelli R, et al. Localization of organic cation/carnitine transporter (OCTN2) in cells forming the blood-brain barrier. J Neurochem. 2008;104:113–123.
    1. Celestino-Soper PB, Violante S, Crawford EL, Luo R, Lionel AC, Delaby E, et al. A common X-linked inborn error of carnitine biosynthesis may be a risk factor for nondysmorphic autism. Proc Nat Acad Sci USA. 2012;109:7974–7981.
    1. Scafidi S, Racz J, Hazelton J, McKenna MC, Fiskum G. Neuroprotection by acetyl-L-carnitine after traumatic injury to the immature rat brain. Dev Neurosci. 2010;32:480–487.
    1. Patel SP, Sullivan PG, Lyttle TS, Rabchevsky AG. Acetyl-ℒ-carnitine ameliorates mitochondrial dysfunction following contusion spinal cord injury. J Neurochem. 2010;114:291–301.
    1. Fortin G, Yurchenko K, Collette C, Rubio M, Villani AC, Bitton A, et al. L-carnitine, a diet component and organic cation transporter OCTN ligand, displays immunosuppressive properties and abrogates intestinal inflammation. Clin Exp Immunol. 2009;156:161–171.
    1. Geier DA, Kern JK, Davis G, King PG, Adams JB, Young JL, et al. A prospective double-blind, randomized clinical trial of levocarnitine to treat autism spectrum disorders. Med Sci Monit. 2011;17:PI15–PI23.
    1. Rossignol DA. Novel and emerging treatments for autism spectrum disorders: a systematic review. Ann Clin Psychiatr. 2009;21:213–236.

Source: PubMed

3
Sottoscrivi