Carbon monoxide (CO) correlates with symptom severity, autoimmunity, and responses to probiotics treatment in a cohort of children with autism spectrum disorder (ASD): a post-hoc analysis of a randomized controlled trial

Hannah Tayla Sherman, Kevin Liu, Kenneth Kwong, Suk-Tak Chan, Alice Chukun Li, Xue-Jun Kong, Hannah Tayla Sherman, Kevin Liu, Kenneth Kwong, Suk-Tak Chan, Alice Chukun Li, Xue-Jun Kong

Abstract

Background: Inflammation, autoimmunity, and gut-brain axis have been implicated in the pathogenesis of autism spectrum disorder (ASD). Carboxyhemoglobin (SpCO) as a non-invasive measurement of inflammation has not been studied in individuals with ASD. We conducted this post-hoc study based on our published clinical trial to explore SpCO and its association with ASD severity, autoimmunity, and response to daily Lactobacillus plantarum probiotic supplementation.

Methods: In this study, we included 35 individuals with ASD aged 3-20 years from a previously published clinical trial of the probiotic Lactobacillus plantarum. Subjects were randomly assigned to receive daily Lactobacillus plantarum probiotic (6 × 1010 CFUs) or a placebo for 16 weeks. The outcomes in this analysis include Social Responsiveness Scale (SRS), Aberrant Behavior Checklist second edition (ABC-2), Clinical Global Impression (CGI) scale, SpCO measured by CO-oximetry, fecal microbiome by 16 s rRNA sequencing, blood serum inflammatory markers, autoantibodies, and oxytocin (OT) by ELISA. We performed Kendall's correlation to examine their interrelationships and used Wilcoxon rank-sum test to compare the means of all outcomes between the two groups at baseline and 16 weeks.

Results: Elevated levels of serum anti-tubulin, CaM kinase II, anti-dopamine receptor D1 (anti-D1), and SpCO were found in the majority of ASD subjects. ASD severity is correlated with SpCO (baseline, R = 0.38, p = 0.029), anti-lysoganglioside GM1 (R = 0.83, p = 0.022), anti-tubulin (R = 0.69, p = 0.042), and anti-D1 (R = 0.71, p = 0.045) in treatment group.

Conclusions: The findings of the present study suggests that the easily administered and non-invasive SpCO test offers a potentially promising autoimmunity and inflammatory biomarker to screen/subgroup ASD and monitor the treatment response to probiotics. Furthermore, we propose that the associations between autoantibodies, gut microbiome profile, serum OT level, GI symptom severity, and ASD core symptom severity scores are specific to the usage of probiotic treatment in our subject cohort. Taken together, these results warrant further studies to improve ASD early diagnosis and treatment outcomes.

Trial registration: ClinicalTrials.gov NCT03337035 , registered November 8, 2017.

Keywords: Autism spectrum disorder (ASD); Autoantibodies; Biomarker; Carboxyhemoglobin; Early screening; Inflammation; Microbiome; Probiotics.

Conflict of interest statement

The authors declare no conflicts of interest. The funding sources and free probiotics/control product provider had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
Timeline of study design, subject dropouts, and assessed variables
Fig. 2
Fig. 2
Overview of proportion of participants with detected positive SpCO and serum inflammatory markers at baseline. For SpCO, a measured percentage of ≥ 2 is denoted as “positive”; for titers of anti-Dopamine Receptor D1, D2L, anti-Lysoganglioside GM1, anti-Tubulin, and activity of CaM Kinase II of baseline, normal levels are denoted as “negative” and both borderline and elevated levels are denoted as “positive;” all remaining indices are denoted based on the absence (“negative”) or presence at detectable levels (“positive”). GFAP and OT were not included due to lack of reference for the determination of cutoff values
Fig. 3
Fig. 3
Correlations between SpCO, ASD socio-behavioral severity, and autoantibody titers at baseline and post-intervention with either probiotics (blue) or placebo (red). Shown correlations are based on Kendall’s correlation using a significance cutoff of α = 0.05. a SRS Total T-scores are positively correlated with SpCO for all subjects at baseline. b The absolute change in SpCO between weeks-0 and -16 is positively correlated with anti-lysoganglioside GM1 log-titers in those receiving the probiotic treatment
Fig. 4
Fig. 4
Correlations between serum autoantibody titers, inflammatory markers, and ASD severity scores, at baseline and post-intervention. Shown correlations are based on Kendall’s correlation using a significance cutoff of α = 0.05
Fig. 5
Fig. 5
GSI change and its correlations with inflammatory marker and ASD severity
Fig. 6
Fig. 6
Differentially enriched microbiota between placebo and probiotic groups. Analyses was conducted at (a) baseline and (b) post-treatment for both groups via LEfSe
Fig. 7
Fig. 7
Correlations between point measurements and change in Shannon index, OT, GFAP, ASD severity, and autoantibody titers at both baseline and post-intervention

References

    1. Maenner MJ, Shaw KA, Baio J, Washington A, Patrick M, et al. Prevalence of autism spectrum disorder among children aged 8 years — autism and developmental disabilities monitoring network, 11 Sites, United States, 2016. Mmwr Surveill Summ. 2020;69:1–12. doi: 10.15585/mmwr.ss6904a1.
    1. Alam R, Abdolmaleky HM, Zhou J. Microbiome, inflammation, epigenetic alterations, and mental diseases. Am J Med Genet B Neuropsychiatr Genet. 2017;174:651–660. doi: 10.1002/ajmg.b.32567.
    1. Braunschweig D, Krakowiak P, Duncanson P, Boyce R, Hansen RL, Ashwood P, et al. Autism-specific maternal autoantibodies recognize critical proteins in developing brain. Transl Psychiat. 2013;3:e277. doi: 10.1038/tp.2013.50.
    1. Cao X, Liu K, Liu J, Liu Y-W, Xu L, Wang H, et al. Dysbiotic gut microbiota and dysregulation of cytokine profile in children and teens with autism spectrum disorder. Front Neurosci-switz. 2021;15:635925. doi: 10.3389/fnins.2021.635925.
    1. Connery K, Tippett M, Delhey LM, Rose S, Slattery JC, Kahler SG, et al. Intravenous immunoglobulin for the treatment of autoimmune encephalopathy in children with autism. Transl Psychiat. 2018;8:148. doi: 10.1038/s41398-018-0214-7.
    1. Kong X-J, Liu J, Liu K, Koh M, Sherman H, Liu S, et al. Probiotic and oxytocin combination therapy in patients with autism spectrum disorder: a randomized, double-blinded. Nutrients. 2021;13:1552. doi: 10.3390/nu13051552.
    1. Melamed IR, Heffron M, Testori A, Lipe K. A pilot study of high-dose intravenous immunoglobulin 5% for autism: Impact on autism spectrum and markers of neuroinflammation. Autism Res. 2018;11:421–433. doi: 10.1002/aur.1906.
    1. Guloksuz SA, Abali O, Cetin EA, Gazioglu SB, Deniz G, Yildirim A, et al. Elevated plasma concentrations of S100 calcium-binding protein B and tumor necrosis factor alpha in children with autism spectrum disorders. Rev Bras Psiquiatr. 2017;39:195–200. doi: 10.1590/1516-4446-2015-1843.
    1. Masi A, Quintana DS, Glozier N, Lloyd AR, Hickie IB, Guastella AJ. Cytokine aberrations in autism spectrum disorder: a systematic review and meta-analysis. Mol Psychiatr. 2015;20:440–446. doi: 10.1038/mp.2014.59.
    1. Esnafoglu E, Ayyıldız SN, Cırrık S, Erturk EY, Erdil A, Daglı A, et al. Evaluation of serum Neuron-specific enolase, S100B, myelin basic protein and glial fibrilliary acidic protein as brain specific proteins in children with autism spectrum disorder. Int J Dev Neurosci. 2017;61:86–91. doi: 10.1016/j.ijdevneu.2017.06.011.
    1. Abou-Donia MB, Suliman HB, Siniscalco D, Antonucci N, ElKafrawy P, Brahmajothi MV. De novo blood biomarkers in autism: autoantibodies against neuronal and glial proteins. Behav Sci. 2019;9:47. doi: 10.3390/bs9050047.
    1. Gonzalez-Gronow M, Cuchacovich M, Francos R, Cuchacovich S, Blanco A, Sandoval R, et al. Catalytic autoantibodies against myelin basic protein (MBP) isolated from serum of autistic children impair in vitro models of synaptic plasticity in rat hippocampus. J Neuroimmunol. 2015;287:1–8. doi: 10.1016/j.jneuroim.2015.07.006.
    1. Kern JK, Geier DA, Sykes LK, Geier MR. Relevance of neuroinflammation and encephalitis in autism. Front Cell Neurosci. 2016;9:519. doi: 10.3389/fncel.2015.00519.
    1. Mostafa GA, Al-Ayadhi LY. A lack of association between hyperserotonemia and the increased frequency of serum anti-myelin basic protein auto-antibodies in autistic children. J Neuroinflamm. 2011;8:71–71. doi: 10.1186/1742-2094-8-71.
    1. Kealy J, Greene C, Campbell M. Blood-brain barrier regulation in psychiatric disorders. Neurosci Lett. 2018;726:133664. doi: 10.1016/j.neulet.2018.06.033.
    1. Zou T, Liu J, Zhang X, Tang H, Song Y, Kong X. Autoantibody and autism spectrum disorder: a systematic review. Res Autism Spect Dis. 2020;75:101568. doi: 10.1016/j.rasd.2020.101568.
    1. Connolly AM, Chez M, Streif EM, Keeling RM, Golumbek PT, Kwon JM, et al. Brain-derived neurotrophic factor and autoantibodies to neural antigens in sera of children with autistic spectrum disorders, landau-kleffner syndrome, and epilepsy. Biol Psychiat. 2006;59:354–363. doi: 10.1016/j.biopsych.2005.07.004.
    1. Connolly AM, Chez MG, Pestronk A, Arnold ST, Mehta S, Deuel RK. Serum autoantibodies to brain in Landau-Kleffner variant, autism, and other neurologic disorders. J Pediatrics. 1999;134:607–613. doi: 10.1016/S0022-3476(99)70248-9.
    1. Bashir S, Al-Ayadhi L. Endothelial antibody levels in the sera of children with autism spectrum disorders. J Chin Med Assoc. 2015;78:414–417. doi: 10.1016/j.jcma.2015.01.008.
    1. Sakurai Y. Autoimmune aspects of kawasaki disease. J Invest Allerg Clin. 2019;29:251–261. doi: 10.18176/jiaci.0300.
    1. Pavăl D. A dopamine hypothesis of autism spectrum disorder. Dev Neurosci-basel. 2017;39:355–360. doi: 10.1159/000478725.
    1. Cox CJ, Zuccolo AJ, Edwards EV, Mascaro-Blanco A, Alvarez K, Stoner J, et al. Antineuronal antibodies in a heterogeneous group of youth and young adults with tics and obsessive-compulsive disorder. J Child Adol Psychop. 2015;25:76–85. doi: 10.1089/cap.2014.0048.
    1. Kirvan CA, Swedo SE, Snider LA, Cunningham MW. Antibody-mediated neuronal cell signaling in behavior and movement disorders. J Neuroimmunol. 2006;179:173–179. doi: 10.1016/j.jneuroim.2006.06.017.
    1. Morita M, Wang Y, Sasaoka T, Okada K, Niwa M, Sawa A, et al. Dopamine D2L receptor is required for visual discrimination and reversal learning. Mol Neuropsychiatry. 2016;2:124–132.
    1. Breuss MW, Leca I, Gstrein T, Hansen AH, Keays DA. Tubulins and brain development – the origins of functional specification. Mol Cell Neurosci. 2017;84:58–67. doi: 10.1016/j.mcn.2017.03.002.
    1. Dale RC, Merheb V, Pillai S, Wang D, Cantrill L, Murphy TK, et al. Antibodies to surface dopamine-2 receptor in autoimmune movement and psychiatric disorders. Brain. 2012;135:3453–3468. doi: 10.1093/brain/aws256.
    1. Yang X, Liang S, Wang L, Han P, Jiang X, Wang J, et al. Sialic acid and anti-ganglioside antibody levels in children with autism spectrum disorders. Brain Res. 2018;1678:273–277. doi: 10.1016/j.brainres.2017.10.027.
    1. Ryter SW. Heme oxygenase-1/carbon monoxide as modulators of autophagy and inflammation. Arch Biochem Biophys. 2019;678:108186. doi: 10.1016/j.abb.2019.108186.
    1. Ryter SW, Choi AMK. Targeting heme oxygenase-1 and carbon monoxide for therapeutic modulation of inflammation. Transl Res. 2016;167:7–34. doi: 10.1016/j.trsl.2015.06.011.
    1. Kwong KK, Chan S. The role of carbon monoxide and heme oxygenase-1 in COVID-19. Toxicol Reports. 2020;7:1170–1171. doi: 10.1016/j.toxrep.2020.08.027.
    1. Wagener FADTG, Pickkers P, Peterson SJ, Immenschuh S, Abraham NG. Targeting the heme-heme oxygenase system to prevent severe complications following COVID-19 infections. Antioxidants. 2020;9(6):540. doi: 10.3390/antiox9060540.
    1. Kong X, Liu J, Cetinbas M, Sadreyev R, Koh M, Huang H, et al. New and preliminary evidence on altered oral and gut microbiota in individuals with Autism Spectrum Disorder (ASD): Implications for ASD diagnosis and subtyping based on microbial biomarkers. Nutrients. 2019;11:2128. doi: 10.3390/nu11092128.
    1. Kong X-J, Liu J, Li J, Kwong K, Koh M, Sukijthamapan P, et al. Probiotics and oxytocin nasal spray as neuro-social-behavioral interventions for patients with autism spectrum disorders: a pilot randomized controlled trial protocol. Pilot Feasibility Stud. 2020;6:20. doi: 10.1186/s40814-020-0557-8.
    1. Fattorusso A, Genova LD, Dell’Isola GB, Mencaroni E, Esposito S. Autism spectrum disorders and the gut microbiota. Nutrients. 2019;11:521. doi: 10.3390/nu11030521.
    1. Liu Y-W, Liong M-T, Chung Y-CE, Huang H-Y, Peng W-S, Cheng Y-F, et al. Effects of lactobacillus plantarum PS128 on children with autism spectrum disorder in Taiwan: a randomized, double-blind Placebo-Controlled Trial. Nutrients. 2019;11:820. doi: 10.3390/nu11040820.
    1. Liu W-H, Yang C-H, Lin C-T, Li S-W, Cheng W-S, Jiang Y-P, et al. Genome architecture of Lactobacillus plantarum PS128, a probiotic strain with potential immunomodulatory activity. Gut Pathog. 2015;7:22. doi: 10.1186/s13099-015-0068-y.
    1. Erdman SE, Poutahidis T. Chapter five microbes and oxytocin benefits for host physiology and behavior. Int Rev Neurobiol. 2016;131:91–126. doi: 10.1016/bs.irn.2016.07.004.
    1. Matsuura T, Motojima Y, Kawasaki M, Ohnishi H, Sakai A, Ueta Y. [Relationship between oxytocin and pain modulation and inflammation] J Uoeh. 2016;38:325–334. doi: 10.7888/juoeh.38.325.
    1. Light A, Grass C, Pursley D, Krause J. Carboxyhemoglobin levels in smokers vs non-smokers in a smoking environment. Respir Care. 2007;52(11):1576.
    1. Schimmel J, George N, Schwarz J, Yousif S, Suner S, Hack JB. Carboxyhemoglobin levels induced by cigarette smoking outdoors in smokers. J Medical Toxicol. 2018;14:68–73. doi: 10.1007/s13181-017-0645-1.
    1. Shimasaki C, Frye RE, Trifiletti R, Cooperstock M, Kaplan G, Melamed I, et al. Evaluation of the cunningham panel™ in pediatric autoimmune neuropsychiatric disorder associated with streptococcal infection (PANDAS) and pediatric acute-onset neuropsychiatric syndrome (PANS): changes in antineuronal antibody titers parallel changes in patient symptoms. J Neuroimmunol. 2020;339:577138. doi: 10.1016/j.jneuroim.2019.577138.
    1. Chain JL, Alvarez K, Mascaro-Blanco A, Reim S, Bentley R, Hommer R, et al. Autoantibody biomarkers for basal ganglia encephalitis in sydenham chorea and pediatric autoimmune neuropsychiatric disorder associated with streptococcal infections. Frontiers Psychiatry. 2020;11:564. doi: 10.3389/fpsyt.2020.00564.
    1. Lewis SJ, Heaton KW. Stool form scale as a useful guide to intestinal transit time. Scand J Gastroentero. 2009;32:920–924. doi: 10.3109/00365529709011203.
    1. Bölte S, Poustka F, Constantino JN. Assessing autistic traits: cross-cultural validation of the social responsiveness scale (SRS) Autism Res. 2008;1:354–363. doi: 10.1002/aur.49.
    1. Aman MG, Singh NN, Stewart AW, Field CJ. The aberrant behavior checklist: a behavior rating scale for the assessment of treatment effects. Am J Ment Defic. 1985;5:485–491.
    1. Busner J, Targum SD. The clinical global impressions scale: applying a research tool in clinical practice. Psychiatry (Edgmont). 2007;4:28–37.
    1. McIver LJ, Abu-Ali G, Franzosa EA, Schwager R, Morgan XC, Waldron L, et al. BioBakery: a meta’omic analysis environment. Bioinformatics. 2018;34:1235–1237. doi: 10.1093/bioinformatics/btx754.
    1. DeSantis TZ, Hugenholtz P, Larsen N, Rojas M, Brodie EL, Keller K, et al. Greengenes, a chimera-checked 16S rRNA gene database and workbench compatible with ARB. Appl Environ Microb. 2006;72:5069–5072. doi: 10.1128/AEM.03006-05.
    1. Segata N, Izard J, Waldron L, Gevers D, Miropolsky L, Garrett WS, et al. Metagenomic biomarker discovery and explanation. Genome Biol. 2011;12:R60–R60. doi: 10.1186/gb-2011-12-6-r60.
    1. DiStasio MM, Nagakura I, Nadler MJ, Anderson MP. T lymphocytes and cytotoxic astrocyte blebs correlate across autism brains. Ann Neurol. 2019;86:885–898. doi: 10.1002/ana.25610.
    1. Liu H, Wang J, He T, Becker S, Zhang G, Li D, et al. Butyrate: a double-edged sword for health? Adv Nutr. 2018;9:21–29. doi: 10.1093/advances/nmx009.
    1. Strati F, Cavalieri D, Albanese D, Felice CD, Donati C, Hayek J, et al. New evidences on the altered gut microbiota in autism spectrum disorders. Microbiome. 2017;5:24. doi: 10.1186/s40168-017-0242-1.
    1. Weisman O, Feldman R. Oxytocin administration affects the production of multiple hormones. Psychoneuroendocrino. 2013;38:626–627. doi: 10.1016/j.psyneuen.2013.03.004.

Source: PubMed

3
Se inscrever