Generic placeholder image

Current Pharmaceutical Design

Editor-in-Chief

ISSN (Print): 1381-6128
ISSN (Online): 1873-4286

Review Article

Autism Spectrum Disorders: Role of Pre- and Post-Natal GammaDelta (γδ) T Cells and Immune Regulation

Author(s): George Anderson* and Susana R. Betancort Medina

Volume 25, Issue 41, 2019

Page: [4321 - 4330] Pages: 10

DOI: 10.2174/1381612825666191102170125

Price: $65

Abstract

Background: It is widely accepted that alterations in immune functioning are an important aspect of the pathoetiology and pathophysiology of autism spectrum disorders (ASD). A relatively under-explored aspect of these alterations is the role of gammaDelta (γδ) T cells, prenatally and in the postnatal gut, which seem important hubs in driving the course of ASD.

Methods: The present article describes the role of γδ T cells in ASD, including their interactions with other immune cells shown to be altered in this spectrum of conditions, including natural killer cells and mast cells.

Results: Other risk factors in ASD, such as decreased vitamins A & D, as well as toxin-associated activation of the aryl hydrocarbon receptor, may also be intimately linked to γδ T cells, and alterations in the regulation of these cells. A growing body of data has highlighted an important role for alterations in mitochondria functioning in the regulation of immune cells, including natural killer cells and mast cells. This is an area that requires investigation in γδ T cells and their putative subtypes.

Conclusion: It is also proposed that maternal stress may act through alterations in the maternal microbiome, leading to changes in how the balance of short-chain fatty acids, such as butyrate, which may act to regulate the placenta and foetal development. Following an overview of previous research on immune, especially γδ T cells, effects in ASD, the future research implications are discussed in detail.

Keywords: Autism spectrum disorders, vitamin A, vitamin D, gammaDelta T cells, aryl hydrocarbon receptor, gut, immune, mitochondria, melatonin, prenatal.

[1]
Seo M, Anderson G. Gut-amygdala interactions in autism spectrum disorders: developmental roles via regulating mitochondria, exosomes, immunity and microRNAs. Curr Pharm Des 2019.
[http://dx.doi.org/dx.doi: 10.2174/1381612825666191105102545] [PMID: 31692435]
[2]
Hughes HK, Mills Ko E, Rose D, Ashwood P. Immune dysfunction and autoimmunity as pathological mechanisms in autism spectrum disorders. Front Cell Neurosci 2018; 12: 405.
[http://dx.doi.org/10.3389/fncel.2018.00405] [PMID: 30483058]
[3]
Furlano RI, Anthony A, Day R, et al. Colonic CD8 and gamma delta T-cell infiltration with epithelial damage in children with autism. J Pediatr 2001; 138(3): 366-72.
[http://dx.doi.org/10.1067/mpd.2001.111323] [PMID: 11241044]
[4]
Howie D, Spencer J, DeLord D, et al. Extrathymic T cell differentiation in the human intestine early in life. J Immunol 1998; 161(11): 5862-72.
[PMID: 9834065]
[5]
McVay LD, Carding SR. Extrathymic origin of human gamma delta T cells during fetal development. J Immunol 1996; 157(7): 2873-82.
[PMID: 8816392]
[6]
McVay LD, Jaswal SS, Kennedy C, Hayday A, Carding SR. The generation of human gammadelta T cell repertoires during fetal development. J Immunol 1998; 160(12): 5851-60.
[PMID: 9637496]
[7]
Dimova T, Brouwer M, Gosselin F, et al. Effector Vγ9Vδ2 T cells dominate the human fetal γδ T-cell repertoire. Proc Natl Acad Sci USA 2015; 112(6): E556-65.
[http://dx.doi.org/10.1073/pnas.1412058112] [PMID: 25617367]
[8]
Ribot JC, Ribeiro ST, Correia DV, Sousa AE, Silva-Santos B. Human γδ thymocytes are functionally immature and differentiate into cytotoxic type 1 effector T cells upon IL-2/IL-15 signaling. J Immunol 2014; 192(5): 2237-43.
[http://dx.doi.org/10.4049/jimmunol.1303119] [PMID: 24489097]
[9]
Parker CM, Groh V, Band H, et al. Evidence for extrathymic changes in the T cell receptor gamma/delta repertoire. J Exp Med 1990; 171(5): 1597-612.
[http://dx.doi.org/10.1084/jem.171.5.1597] [PMID: 2185330]
[10]
Davey MS, Willcox CR, Hunter S, et al. The human Vδ2+ T-cell compartment comprises distinct innate-like Vγ9+ and adaptive Vγ9- subsets. Nat Commun 2018; 9(1): 1760.
[http://dx.doi.org/10.1038/s41467-018-04076-0] [PMID: 29720665]
[11]
Willcox CR, Davey MS, Willcox BE. Development and selection of the human Vγ9Vδ2+ T-cell repertoire. Front Immunol 2018; 9: 1501.
[http://dx.doi.org/10.3389/fimmu.2018.01501] [PMID: 30013562]
[12]
Muschaweckh A, Petermann F, Korn T. IL-1β and IL-23 promote extrathymic commitment of CD27+CD122- γδ T Cells to γδT17 cells. J Immunol 2017; 199(8): 2668-79.
[http://dx.doi.org/10.4049/jimmunol.1700287] [PMID: 28855314]
[13]
Do JS, Visperas A, O’Brien RL, Min B. CD4 T cells play important roles in maintaining IL-17-producing γδ T-cell subsets in naive animals. Immunol Cell Biol 2012; 90(4): 396-403.
[http://dx.doi.org/10.1038/icb.2011.50] [PMID: 21647171]
[14]
Akitsu A, Iwakura Y. Interleukin-17-producing γδ T (γδ17) cells in inflammatory diseases. Immunology 2018; 155(4): 418-26.
[http://dx.doi.org/10.1111/imm.12993] [PMID: 30133701]
[15]
Al-Ayadhi LY, Mostafa GA. Elevated serum levels of interleukin-17A in children with autism. J Neuroinflammation 2012; 9: 158.
[http://dx.doi.org/10.1186/1742-2094-9-158] [PMID: 22748016]
[16]
Wong H, Hoeffer C. Maternal IL-17A in autism. Exp Neurol 2018; 299(Pt A): 228-40.
[17]
Stange J, Veldhoen M. The aryl hydrocarbon receptor in innate T cell immunity. Semin Immunopathol 2013; 35(6): 645-55.
[http://dx.doi.org/10.1007/s00281-013-0389-1] [PMID: 24030775]
[18]
Martin B, Hirota K, Cua DJ, Stockinger B, Veldhoen M. Interleukin-17-producing gammadelta T cells selectively expand in response to pathogen products and environmental signals. Immunity 2009; 31(2): 321-30.
[http://dx.doi.org/10.1016/j.immuni.2009.06.020] [PMID: 19682928]
[19]
Maes M, Anderson G, Medina SRB. Integrating autism spectrum disorder pathophysiology: mitochondria, vitamin A, CD38, oxytocin, serotonin and melatonergic alterations in the placenta and gut. Curr Pharm Des 2019.
[http://dx.doi.org/dx.doi: 10.2174/1381612825666191102165459] [PMID: 31682209]
[20]
Lai X, Wu X, Hou N, et al. Vitamin A deficiency induces autistic-like behaviors in rats by regulating the RARβ-CD38-oxytocin axis in the hypothalamus. Mol Nutr Food Res 2018; 62(5)
[http://dx.doi.org/10.1002/mnfr.201700754] [PMID: 29266770]
[21]
Zhou W, Li S. Decreased levels of serum retinoic acid in Chinese children with autism spectrum disorder. Psychiatry Res 2018; 269: 469-73.
[http://dx.doi.org/10.1016/j.psychres.2018.08.091] [PMID: 30195740]
[22]
Guo M, Zhu J, Yang T, et al. Vitamin A and vitamin D deficiencies exacerbate symptoms in children with autism spectrum disorders. Nutr Neurosci 2018; 22(9): 1-11.
[http://dx.doi.org/10.1080/1028415X.2018.1558762] [PMID: 29338670]
[23]
Hashimoto-Hill S, Friesen L, Kim M, Kim CH. Contraction of intestinal effector T cells by retinoic acid-induced purinergic receptor P2X7. Mucosal Immunol 2017; 10(4): 912-23.
[http://dx.doi.org/10.1038/mi.2016.109] [PMID: 27966552]
[24]
Frascoli M, Marcandalli J, Schenk U, Grassi F. Purinergic P2X7 receptor drives T cell lineage choice and shapes peripheral γδ cells. J Immunol 2012; 189(1): 174-80.
[http://dx.doi.org/10.4049/jimmunol.1101582] [PMID: 22649196]
[25]
Manohar M, Hirsh MI, Chen Y, Woehrle T, Karande AA, Junger WG. ATP release and autocrine signaling through P2X4 receptors regulate γδ T cell activation. J Leukoc Biol 2012; 92(4): 787-94.
[http://dx.doi.org/10.1189/jlb.0312121] [PMID: 22753954]
[26]
Gorodeski GI. Expression, regulation, and function of P2X(4) purinergic receptor in human cervical epithelial cells. Am J Physiol Cell Physiol 2002; 282(1): C84-93.
[http://dx.doi.org/10.1152/ajpcell.2002.282.1.C84] [PMID: 11742801]
[27]
Raverdeau M, Breen CJ, Misiak A, Mills KH. Retinoic acid suppresses IL-17 production and pathogenic activity of γδ T cells in CNS autoimmunity. Immunol Cell Biol 2016; 94(8): 763-73.
[http://dx.doi.org/10.1038/icb.2016.39] [PMID: 27089940]
[28]
Wu S, Wang W, Le Q. All-trans retinoic acid regulates the balance of Treg-Th17 cells through ERK and P38 signaling pathway. Iran J Immunol 2019; 16(1): 1-10.
[http://dx.doi.org/10.22034/IJI.2019.39402] [PMID: 30864551]
[29]
Priyanka SH, Syam Das S, Thushara AJ, Rauf AA, Indira M. All trans retinoic acid attenuates markers of neuroinflammation in rat brain by modulation of SIRT1 and NFκB. Neurochem Res 2018; 43(9): 1791-801.
[http://dx.doi.org/10.1007/s11064-018-2595-7] [PMID: 30022380]
[30]
Grahnert A, Grahnert A, Klein C, Schilling E, Wehrhahn J, Hauschildt S. Review: NAD +: a modulator of immune functions. Innate Immun 2011; 17(2): 212-33.
[http://dx.doi.org/10.1177/1753425910361989] [PMID: 20388721]
[31]
Bartz JA, McInnes LA. CD38 regulates oxytocin secretion and complex social behavior. BioEssays 2007; 29(9): 837-41.
[http://dx.doi.org/10.1002/bies.20623] [PMID: 17688286]
[32]
Higashida H, Yokoyama S, Huang JJ, et al. Social memory, amnesia, and autism: brain oxytocin secretion is regulated by NAD+ metabolites and single nucleotide polymorphisms of CD38. Neurochem Int 2012; 61(6): 828-38.
[http://dx.doi.org/10.1016/j.neuint.2012.01.030] [PMID: 22366648]
[33]
Alzghoul L, Al-Eitan LN, Aladawi M, Odeh M, Abu Hantash O. The association between serum vitamin D3 Levels and Autism among Jordanian Boys. J Autism Dev Disord 2019.
[http://dx.doi.org/10.1007/s10803-019-04017-w] [PMID: 30993503]
[34]
Sassi F, Tamone C, D’Amelio P. Vitamin D: nutrient, hormone, and immunomodulator. Nutrients 2018; 10(11) E1656
[http://dx.doi.org/10.3390/nu10111656] [PMID: 30400332]
[35]
García-Serna AM, Morales E. Neurodevelopmental effects of prenatal vitamin D in humans: systematic review and meta-analysis. Mol Psychiatry 2019.
[http://dx.doi.org/10.1038/s41380-019-0357-9] [PMID: 30696940]
[36]
Waterhouse M, Hope B, Krause L, et al. Vitamin D and the gut microbiome: a systematic review of in vivo studies. Eur J Nutr 2019; 58(7): 2895-910.
[http://dx.doi.org/10.1007/s00394-018-1842-7] [PMID: 30324342]
[37]
Chen L, Cencioni MT, Angelini DF, Borsellino G, Battistini L, Brosnan CF. Transcriptional profiling of gamma delta T cells identifies a role for vitamin D in the immunoregulation of the V gamma 9V delta 2 response to phosphate-containing ligands. J Immunol 2005; 174(10): 6144-52.
[http://dx.doi.org/10.4049/jimmunol.174.10.6144] [PMID: 15879110]
[38]
De Santis M, Cavaciocchi F, Ceribelli A, et al. Gamma-delta T lymphocytes and 25-hydroxy vitamin D levels as key factors in autoimmunity and inflammation: the case of zoledronic acid-induced acute phase reaction. Lupus 2015; 24(4-5): 442-7.
[http://dx.doi.org/10.1177/0961203314559633] [PMID: 25801887]
[39]
Liu S, Li E, Sun Z, et al. Altered gut microbiota and short chain fatty acids in Chinese children with autism spectrum disorder. Sci Rep 2019; 9(1): 287.
[http://dx.doi.org/10.1038/s41598-018-36430-z] [PMID: 30670726]
[40]
Vitali B, Ndagijimana M, Maccaferri S, Biagi E, Guerzoni ME, Brigidi P. An in vitro evaluation of the effect of probiotics and prebiotics on the metabolic profile of human microbiota. Anaerobe 2012; 18(4): 386-91.
[http://dx.doi.org/10.1016/j.anaerobe.2012.04.014] [PMID: 22579985]
[41]
Finamore A, Roselli M, Donini L, et al. Supplementation with Bifidobacterium longum Bar33 and Lactobacillus helveticus Bar13 mixture improves immunity in elderly humans (over 75 years) and aged mice. Nutrition 2019; 63-64: 184-92.
[http://dx.doi.org/10.1016/j.nut.2019.02.005] [PMID: 31029046]
[42]
Rossi LE, Avila DE, Spallanzani RG, et al. Histone deacetylase inhibitors impair NK cell viability and effector functions through inhibition of activation and receptor expression. J Leukoc Biol 2012; 91(2): 321-31.
[http://dx.doi.org/10.1189/jlb.0711339] [PMID: 22124136]
[43]
Fujisawa TX, Nishitani S, Iwanaga R, et al. Association of aryl hydrocarbon receptor-related gene variants with the severity of autism spectrum disorders. Front Psychiatry 2016; 7: 184.
[http://dx.doi.org/10.3389/fpsyt.2016.00184] [PMID: 27899901]
[44]
Beischlag TV, Anderson G, Mazzoccoli G. Glioma: tryptophan catabolite and melatoninergic pathways link microRNA, 14-3- 3, chromosome 4q35, epigenetic processes and other glioma biochemical changes. Curr Pharm Des 2016; 22(8): 1033-48.
[http://dx.doi.org/10.2174/1381612822666151214104941] [PMID: 26654773]
[45]
Bunaciu RP, Jensen HA, MacDonald RJ, LaTocha DH, Varner JD, Yen A. 6-formylindolo(3,2-b)carbazole (FICZ) modulates the signalsome responsible for RA-induced differentiation of HL-60 myeloblastic leukemia cells. PLoS One 2015; 10(8) e0135668
[http://dx.doi.org/10.1371/journal.pone.0135668] [PMID: 26287494]
[46]
Anderson G, Maes M. Interactions of tryptophan and its catabolites with melatonin and the alpha 7 nicotinic receptor in central nervous system and psychiatric disorders: role of the aryl hydrocarbon receptor and direct mitochondria regulation. Int J Tryptophan Res 2017; 10 1178646917691738
[http://dx.doi.org/10.1177/1178646917691738] [PMID: 28469467]
[47]
El-Ansary A, Cannell JJ, Bjørklund G, et al. In the search for reliable biomarkers for the early diagnosis of autism spectrum disorder: the role of vitamin D. Metab Brain Dis 2018; 33(3): 917-31.
[http://dx.doi.org/10.1007/s11011-018-0199-1] [PMID: 29497932]
[48]
Anderson G. Endometriosis pathoetiology and pathophysiology: roles of vitamin A, estrogen, immunity, adipocytes, gut microbiome and melatonergic pathway on mitochondria regulation. Biomol Concepts 2019; 10(1): 133-49.
[http://dx.doi.org/10.1515/bmc-2019-0017] [PMID: 31339848]
[49]
Anderson G. Breast cancer: occluded role of mitochondria N-acetylserotonin/melatonin ratio in co-ordinating pathophysiology. Biochem Pharmacol 2019; 168: 259-68.
[http://dx.doi.org/10.1016/j.bcp.2019.07.014] [PMID: 31310736]
[50]
Cai Y, Xue F, Qin H, et al. Differential roles of the mTOR-STAT3 signaling in dermal γδ T cell effector function in skin inflammation. Cell Rep 2019; 27(10): 3034-48.e5.
[http://dx.doi.org/10.1016/j.celrep.2019.05.019] [PMID: 31167146]
[51]
Jiang YZ, Wang K, Fang R, Zheng J. Expression of aryl hydrocarbon receptor in human placentas and fetal tissues. J Histochem Cytochem 2010; 58(8): 679-85.
[http://dx.doi.org/10.1369/jhc.2010.955955] [PMID: 20354149]
[52]
Weinberg SE, Singer BD, Steinert EM, et al. Mitochondrial complex III is essential for suppressive function of regulatory T cells. Nature 2019; 565(7740): 495-9.
[http://dx.doi.org/10.1038/s41586-018-0846-z] [PMID: 30626970]
[53]
Miranda D, Jara C, Mejias S, et al. Deficient mitochondrial biogenesis in IL-2 activated NK cells correlates with impaired PGC1-α upregulation in elderly humans. Exp Gerontol 2018; 110: 73-8.
[http://dx.doi.org/10.1016/j.exger.2018.05.014] [PMID: 29782967]
[54]
Kaufmann U, Kahlfuss S, Yang J, Ivanova E, Koralov SB, Feske S. Calcium signaling controls pathogenic Th17 cell-mediated inflammation by regulating mitochondrial function Cell Metab 2019; S1550-4131(19): 30019-1.
[http://dx.doi.org/10.1016/j.cmet.2019.01.019]
[55]
Oberkampf M, Guillerey C, Mouriès J, et al. Mitochondrial reactive oxygen species regulate the induction of CD8+ T cells by plasmacytoid dendritic cells. Nat Commun 2018; 9(1): 2241.
[http://dx.doi.org/10.1038/s41467-018-04686-8] [PMID: 29884826]
[56]
Jyonouchi H, Geng L, Rose S, Bennuri SC, Frye RE. Variations in mitochondrial respiration differ in IL-1ß/IL-10 ratio based subgroups in autism spectrum disorders. Front Psychiatry 2019; 10: 71.
[http://dx.doi.org/10.3389/fpsyt.2019.00071] [PMID: 30842746]
[57]
Chen W, Sandoval H, Kubiak JZ, Li XC, Ghobrial RM, Kloc M. The phenotype of peritoneal mouse macrophages depends on the mitochondria and ATP/ADP homeostasis. Cell Immunol 2018; 324: 1-7.
[http://dx.doi.org/10.1016/j.cellimm.2017.11.003] [PMID: 29129293]
[58]
Schwacha MG, Rani M, Zhang Q, Nunez-Cantu O, Cap AP. Mitochondrial damage-associated molecular patterns activate γδ T-cells. Innate Immun 2014; 20(3): 261-8.
[http://dx.doi.org/10.1177/1753425913488969] [PMID: 23757324]
[59]
Pagan C, Goubran-Botros H, Delorme R, et al. Disruption of melatonin synthesis is associated with impaired 14-3-3 and miR-451 levels in patients with autism spectrum disorders. Sci Rep 2017; 7(1): 2096.
[http://dx.doi.org/10.1038/s41598-017-02152-x] [PMID: 28522826]
[60]
Anderson G. Mitochondria and the gut as crucial hubs for the interactions of melatonin with sirtuins, inflammation, butyrate, tryptophan metabolites, and alpha 7 nicotinic receptor across a host of medical conditions. Melatonin Res 2019; 2(2): 70-85.
[http://dx.doi.org/10.32794/mr11250022]
[61]
Muxel SM, Pires-Lapa MA, Monteiro AW, et al. NF-κB drives the synthesis of melatonin in RAW 264.7 macrophages by inducing the transcription of the arylalkylamine-N-acetyltransferase (AA-NAT) gene. PLoS One 2012; 7(12) e52010
[http://dx.doi.org/10.1371/journal.pone.0052010] [PMID: 23284853]
[62]
Ahmad SF, Zoheir KMA, Ansari MA, et al. Dysregulation of Th1, Th2, Th17, and T regulatory cell-related transcription factor signaling in children with autism. Mol Neurobiol 2017; 54(6): 4390-400.
[http://dx.doi.org/10.1007/s12035-016-9977-0] [PMID: 27344332]
[63]
Bakheet SA, Alzahrani MZ, Ansari MA, et al. Resveratrol ameliorates dysregulation of Th1, Th2, Th17, and T regulatory cell-related transcription factor signaling in a BTBR T + tf/J mouse model of autism. Mol Neurobiol 2017; 54(7): 5201-12.
[http://dx.doi.org/10.1007/s12035-016-0066-1] [PMID: 27578011]
[64]
Safari MR, Ghafouri-Fard S, Noroozi R, et al. FOXP3 gene variations and susceptibility to autism: a case-control study. Gene 2017; 596: 119-22.
[http://dx.doi.org/10.1016/j.gene.2016.10.019] [PMID: 27751813]
[65]
Mostafa GA, Al-Ayadhi LY. Reduced serum concentrations of 25-hydroxy vitamin D in children with autism: relation to autoimmunity. J Neuroinflammation 2012; 9: 201.
[http://dx.doi.org/10.1186/1742-2094-9-201] [PMID: 22898564]
[66]
Park SG, Mathur R, Long M, et al. T regulatory cells maintain intestinal homeostasis by suppressing γδ T cells. Immunity 2010; 33(5): 791-803.
[http://dx.doi.org/10.1016/j.immuni.2010.10.014] [PMID: 21074460]
[67]
Yurchenko E, Levings MK, Piccirillo CA. CD4+ Foxp3+ regulatory T cells suppress γδ T-cell effector functions in a model of T-cell-induced mucosal inflammation. Eur J Immunol 2011; 41(12): 3455-66.
[http://dx.doi.org/10.1002/eji.201141814] [PMID: 21956668]
[68]
Kang N, Tang L, Li X, et al. Identification and characterization of Foxp3(+) gammadelta T cells in mouse and human. Immunol Lett 2009; 125(2): 105-13.
[http://dx.doi.org/10.1016/j.imlet.2009.06.005] [PMID: 19539651]
[69]
Casetti R, Agrati C, Wallace M, et al. Cutting edge: TGF-beta1 and IL-15 Induce FOXP3+ gammadelta regulatory T cells in the presence of antigen stimulation. J Immunol 2009; 183(6): 3574-7.
[http://dx.doi.org/10.4049/jimmunol.0901334] [PMID: 19710458]
[70]
Okeke EB, Uzonna JE. The pivotal role of regulatory T cells in the regulation of innate immune cells. Front Immunol 2019; 10: 680.
[http://dx.doi.org/10.3389/fimmu.2019.00680] [PMID: 31024539]
[71]
Bhat J, Dubin S, Dananberg A, et al. Histone deacetylase inhibitor modulates NKG2D receptor expression and memory phenotype of human gamma/delta T cells upon interaction with tumor cells. Front Immunol 2019; 10: 569.
[http://dx.doi.org/10.3389/fimmu.2019.00569] [PMID: 30972064]
[72]
Cao T, Zhang X, Chen D, Zhang P, Li Q, Muhammad A. The epigenetic modification during the induction of Foxp3 with sodium butyrate. Immunopharmacol Immunotoxicol 2018; 40(4): 309-18.
[http://dx.doi.org/10.1080/08923973.2018.1480631] [PMID: 30003817]
[73]
Göschl L, Scheinecker C, Bonelli M. Treg cells in autoimmunity: from identification to Treg-based therapies. Semin Immunopathol 2019; 41(3): 301-14.
[http://dx.doi.org/10.1007/s00281-019-00741-8] [PMID: 30953162]
[74]
Mostafa GA, Al Shehab A, Fouad NR. Frequency of CD4+CD25high regulatory T cells in the peripheral blood of Egyptian children with autism. J Child Neurol 2010; 25(3): 328-35.
[http://dx.doi.org/10.1177/0883073809339393] [PMID: 19713552]
[75]
Banoth B, Cassel SL. Mitochondria in innate immune signaling. Transl Res 2018; 202: 52-68.
[http://dx.doi.org/10.1016/j.trsl.2018.07.014] [PMID: 30165038]
[76]
Guerini FR, Bolognesi E, Chiappedi M, et al. Activating KIR molecules and their cognate ligands prevail in children with a diagnosis of ASD and in their mothers. Brain Behav Immun 2014; 36: 54-60.
[http://dx.doi.org/10.1016/j.bbi.2013.10.006] [PMID: 24120931]
[77]
Basheer S, Venkataswamy MM, Christopher R, et al. Immune aberrations in children with Autism Spectrum Disorder: a case-control study from a tertiary care neuropsychiatric hospital in India. Psychoneuroendocrinology 2018; 94: 162-7.
[http://dx.doi.org/10.1016/j.psyneuen.2018.05.002] [PMID: 29804052]
[78]
Torres AR, Westover JB, Gibbons C, Johnson RC, Ward DC. Activating killer-cell immunoglobulin-like receptors (KIR) and their cognate HLA ligands are significantly increased in autism. Brain Behav Immun 2012; 26(7): 1122-7.
[http://dx.doi.org/10.1016/j.bbi.2012.07.014] [PMID: 22884899]
[79]
Enstrom AM, Lit L, Onore CE, et al. Altered gene expression and function of peripheral blood natural killer cells in children with autism. Brain Behav Immun 2009; 23(1): 124-33.
[http://dx.doi.org/10.1016/j.bbi.2008.08.001] [PMID: 18762240]
[80]
Michelet X, Dyck L, Hogan A, et al. Metabolic reprogramming of natural killer cells in obesity limits antitumor responses. Nat Immunol 2018; 19(12): 1330-40.
[http://dx.doi.org/10.1038/s41590-018-0251-7] [PMID: 30420624]
[81]
Belenchia AM, Jones KL, Will M, et al. Maternal vitamin D deficiency during pregnancy affects expression of adipogenic-regulating genes peroxisome proliferator-activated receptor gamma (PPARγ) and vitamin D receptor (VDR) in lean male mice offspring. Eur J Nutr 2018; 57(2): 723-30.
[http://dx.doi.org/10.1007/s00394-016-1359-x] [PMID: 28004271]
[82]
Chiang HL, Liu CJ, Hu YW, et al. Risk of cancer in children, adolescents, and young adults with autistic disorder. J Pediatr 2015; 166(2): 418-23.e1.
[http://dx.doi.org/10.1016/j.jpeds.2014.10.029] [PMID: 25453246]
[83]
Calvo JR, González-Yanes C, Maldonado MD. The role of melatonin in the cells of the innate immunity: a review. J Pineal Res 2013; 55(2): 103-20.
[http://dx.doi.org/10.1111/jpi.12075] [PMID: 23889107]
[84]
Anderson G, Reiter RJ. Glioblastoma: role of mitochondria N-acetylserotonin/melatonin ratio in mediating effects of miR-451 and aryl hydrocarbon receptor and in coordinating wider biochemical changes. Int J Tryptophan Res 2019; 12 1178646919855942
[http://dx.doi.org/10.1177/1178646919855942] [PMID: 31244524]
[85]
Pozo D, García-Mauriño S, Guerrero JM, Calvo JR. mRNA expression of nuclear receptor RZR/RORalpha, melatonin membrane receptor MT, and hydroxindole-O-methyltransferase in different populations of human immune cells. J Pineal Res 2004; 37(1): 48-54.
[http://dx.doi.org/10.1111/j.1600-079X.2004.00135.x] [PMID: 15230868]
[86]
Chen HH, Huang WT, Yang LW, Lin CW. The PTEN-AKT-mTOR/RICTOR pathway in nasal natural killer cell lymphoma is activated by miR-494-3p via PTEN but inhibited by miR-142-3p via RICTOR. Am J Pathol 2015; 185(5): 1487-99.
[http://dx.doi.org/10.1016/j.ajpath.2015.01.025] [PMID: 25907832]
[87]
Tie G, Yan J, Khair L, et al. Hypercholesterolemia increases colorectal cancer incidence by reducing production of NKT and γδ T cells from hematopoietic stem cells. Cancer Res 2017; 77(9): 2351-62.
[http://dx.doi.org/10.1158/0008-5472.CAN-16-1916] [PMID: 28249902]
[88]
Maldonado MD, Mora-Santos M, Naji L, Carrascosa-Salmoral MP, Naranjo MC, Calvo JR. Evidence of melatonin synthesis and release by mast cells. Possible modulatory role on inflammation. Pharmacol Res 2010; 62(3): 282-7.
[http://dx.doi.org/10.1016/j.phrs.2009.11.014] [PMID: 19963060]
[89]
Theoharides TC, Stewart JM, Panagiotidou S, Melamed I. Mast cells, brain inflammation and autism. Eur J Pharmacol 2016; 778: 96-102.
[http://dx.doi.org/10.1016/j.ejphar.2015.03.086] [PMID: 25941080]
[90]
Chelombitko MA, Averina OA, Vasilyeva TV, et al. Mitochondria-targeted antioxidant SkQ1 (10-(6´-plastoquinonyl) decyltriphenylphosphonium bromide) inhibits mast cell degranulation in vivo and in vitro. Biochemistry (Mosc) 2017; 82(12): 1493-503.
[http://dx.doi.org/10.1134/S0006297917120082] [PMID: 29486699]
[91]
Albert-Bayo M, Paracuellos I, González-Castro AM, et al. Intestinal mucosal mast cells: key modulators of barrier function and homeostasis. Cells 2019; 8(2) E135
[http://dx.doi.org/10.3390/cells8020135] [PMID: 30744042]
[92]
Buhner S, Barki N, Greiter W, et al. Calcium imaging of nerve-mast cell signaling in the human intestine. Front Physiol 2017; 8: 971.
[http://dx.doi.org/10.3389/fphys.2017.00971] [PMID: 29238306]
[93]
Mantri CK, St John AL. Immune synapses between mast cells and γδ T cells limit viral infection. J Clin Invest 2019; 129(3): 1094-108.
[http://dx.doi.org/10.1172/JCI122530] [PMID: 30561384]
[94]
Ueshima C, Kataoka TR, Hirata M, et al. The killer cell Ig-like receptor 2DL4 expression in human mast cells and its potential role in breast cancer invasion. Cancer Immunol Res 2015; 3(8): 871-80.
[http://dx.doi.org/10.1158/2326-6066.CIR-14-0199] [PMID: 25735953]
[95]
Saadalla AM, Osman A, Gurish MF, et al. Mast cells promote small bowel cancer in a tumor stage-specific and cytokine-dependent manner. Proc Natl Acad Sci USA 2018; 115(7): 1588-92.
[http://dx.doi.org/10.1073/pnas.1716804115] [PMID: 29429965]
[96]
Benedé S, Berin MC. Mast cell heterogeneity underlies different manifestations of food allergy in mice. PLoS One 2018; 13(1) e0190453
[http://dx.doi.org/10.1371/journal.pone.0190453] [PMID: 29370173]
[97]
Ganda Mall JP, Casado-Bedmar M, Winberg ME, Brummer RJ, Schoultz I, Keita ÅV. A β-glucan-based dietary fiber reduces mast cell-induced hyperpermeability in ileum from patients with Crohn’s disease and control subjects. Inflamm Bowel Dis 2017; 24(1): 166-78.
[http://dx.doi.org/10.1093/ibd/izx002] [PMID: 29272475]
[98]
Conti P, Caraffa A, Ronconi G, et al. Impact of mast cells in mucosal immunity of intestinal inflammation: inhibitory effect of IL-37. Eur J Pharmacol 2018; 818: 294-9.
[http://dx.doi.org/10.1016/j.ejphar.2017.09.044] [PMID: 28970014]
[99]
Saresella M, Piancone F, Marventano I, et al. Multiple inflammasome complexes are activated in autistic spectrum disorders. Brain Behav Immun 2016; 57: 125-33.
[http://dx.doi.org/10.1016/j.bbi.2016.03.009] [PMID: 26979869]
[100]
Han YM, Cheung WK, Wong CK, et al. Distinct cytokine and chemokine profiles in autism spectrum disorders. Front Immunol 2017; 8: 11.
[http://dx.doi.org/10.3389/fimmu.2017.00011] [PMID: 28167942]
[101]
Skovgaard K, Cirera S, Vasby D, et al. Expression of innate immune genes, proteins and microRNAs in lung tissue of pigs infected experimentally with influenza virus (H1N2). Innate Immun 2013; 19(5): 531-44.
[http://dx.doi.org/10.1177/1753425912473668] [PMID: 23405029]
[102]
Park HJ, Kim HJ, Ra J, et al. Melatonin inhibits lipopolysaccharide-induced CC chemokine subfamily gene expression in human peripheral blood mononuclear cells in a microarray analysis. J Pineal Res 2007; 43(2): 121-9.
[http://dx.doi.org/10.1111/j.1600-079X.2007.00452.x] [PMID: 17645690]
[103]
Sakaguchi K, Itoh MT, Takahashi N, Tarumi W, Ishizuka B. The rat oocyte synthesises melatonin. Reprod Fertil Dev 2013; 25(4): 674-82.
[http://dx.doi.org/10.1071/RD12091] [PMID: 22951050]
[104]
Lansink MO, Patyk V, de Groot H, Effenberger-Neidnicht K. Melatonin reduces changes to small intestinal microvasculature during systemic inflammation. J Surg Res 2017; 211: 114-25.
[http://dx.doi.org/10.1016/j.jss.2016.11.055] [PMID: 28501107]
[105]
Wang CC, Wu H, Lin FH, et al. Sodium butyrate enhances intestinal integrity, inhibits mast cell activation, inflammatory mediator production and JNK signaling pathway in weaned pigs. Innate Immun 2018; 24(1): 40-6.
[http://dx.doi.org/10.1177/1753425917741970] [PMID: 29183244]
[106]
Zhang H, Du M, Yang Q, Zhu MJ. Butyrate suppresses murine mast cell proliferation and cytokine production through inhibiting histone deacetylase. J Nutr Biochem 2016; 27: 299-306.
[http://dx.doi.org/10.1016/j.jnutbio.2015.09.020] [PMID: 26601598]
[107]
Mezouar S, Ben Amara A, Vitte J, Mege JL. Isolation of human placental mast cells. Curr Protoc Cell Biol 2018; 80(1) e52
[http://dx.doi.org/10.1002/cpcb.52] [PMID: 30044540]
[108]
Muench MO, Beyer AI, Fomin ME, et al. The adult livers of immunodeficient mice support human hematopoiesis: evidence for a hepatic mast cell population that develops early in human ontogeny. PLoS One 2014; 9(5) e97312
[http://dx.doi.org/10.1371/journal.pone.0097312] [PMID: 24819392]
[109]
Faas MM, De Vos P. Innate immune cells in the placental bed in healthy pregnancy and preeclampsia. Placenta 2018; 69: 125-33.
[http://dx.doi.org/10.1016/j.placenta.2018.04.012] [PMID: 29748088]
[110]
Jin CJ, Engstler AJ, Sellmann C, et al. Sodium butyrate protects mice from the development of the early signs of non-alcoholic fatty liver disease: role of melatonin and lipid peroxidation. Br J Nutr 2016; 23: 1-12.
[http://dx.doi.org/10.1017/S0007114516004025] [PMID: 27876107]

Rights & Permissions Print Cite
© 2024 Bentham Science Publishers | Privacy Policy