Beta-defensin 1, aryl hydrocarbon receptor and plasma kynurenine in major depressive disorder: metabolomics-informed genomics

Duan Liu, Balmiki Ray, Drew R Neavin, Jiabin Zhang, Arjun P Athreya, Joanna M Biernacka, William V Bobo, Daniel K Hall-Flavin, Michelle K Skime, Hongjie Zhu, Gregory D Jenkins, Anthony Batzler, Krishna R Kalari, Felix Boakye-Agyeman, Wayne R Matson, Swati S Bhasin, Taisei Mushiroda, Yusuke Nakamura, Michiaki Kubo, Ravishankar K Iyer, Liewei Wang, Mark A Frye, Rima Kaddurah-Daouk, Richard M Weinshilboum, Duan Liu, Balmiki Ray, Drew R Neavin, Jiabin Zhang, Arjun P Athreya, Joanna M Biernacka, William V Bobo, Daniel K Hall-Flavin, Michelle K Skime, Hongjie Zhu, Gregory D Jenkins, Anthony Batzler, Krishna R Kalari, Felix Boakye-Agyeman, Wayne R Matson, Swati S Bhasin, Taisei Mushiroda, Yusuke Nakamura, Michiaki Kubo, Ravishankar K Iyer, Liewei Wang, Mark A Frye, Rima Kaddurah-Daouk, Richard M Weinshilboum

Abstract

Major depressive disorder (MDD) is a heterogeneous disease. Efforts to identify biomarkers for sub-classifying MDD and antidepressant therapy by genome-wide association studies (GWAS) alone have generally yielded disappointing results. We applied a metabolomics-informed genomic research strategy to study the contribution of genetic variation to MDD pathophysiology by assaying 31 metabolites, including compounds from the tryptophan, tyrosine, and purine pathways, in plasma samples from 290 MDD patients. Associations of metabolite concentrations with depressive symptoms were determined, followed by GWAS for selected metabolites and functional validation studies of the genes identified. Kynurenine (KYN), the baseline plasma metabolite that was most highly associated with depressive symptoms, was negatively correlated with severity of those symptoms. GWAS for baseline plasma KYN concentrations identified SNPs across the beta-defensin 1 (DEFB1) and aryl hydrocarbon receptor (AHR) genes that were cis-expression quantitative trait loci (eQTLs) for DEFB1 and AHR mRNA expression, respectively. Furthermore, the DEFB1 locus was associated with severity of MDD symptoms in a larger cohort of 803 MDD patients. Functional studies demonstrated that DEFB1 could neutralize lipopolysaccharide-stimulated expression of KYN-biosynthesizing enzymes in monocytic cells, resulting in altered KYN concentrations in the culture media. In addition, we demonstrated that AHR was involved in regulating the expression of enzymes in the KYN pathway and altered KYN biosynthesis in cell lines of hepatocyte and astrocyte origin. In conclusion, these studies identified SNPs that were cis-eQTLs for DEFB1 and AHR and, which were associated with variation in plasma KYN concentrations that were related to severity of MDD symptoms.

Trial registration: ClinicalTrials.gov NCT00613470.

Conflict of interest statement

Dr. Balmiki Ray is a salaried employee of Assurex Health Inc. Dr. Wayne Matxon and Dr. Swati Bhasin are salaried employees of Ixcela. Dr. Mark Frye receives grant support from AssureRx, Janssen Research & Development, Myriad, and Pfizer. He is also a consultant for Janssen Research & Development, LLC, Mitsubishi Tanabe Pharma Corporation, Myriad Genetics, Sunovion, Supernus Pharmaceuticals, Teva Pharmaceuticals, and Neuralstem, Inc. and he receives CME/Travel Support from the American Physician Institute and CME Outfitters. Drs. Wang and Weinshilboum are founders and stockholders in OneOme LLC.

Figures

Fig. 1. Tryptophan metabolism, kynurenine pathway and…
Fig. 1. Tryptophan metabolism, kynurenine pathway and major depressive disorder
Tryptophan (TRP) is metabolized by two major pathways: the “kynurenine (KYN) pathway” and the “serotonin (5-HT)” pathways. 5-HT cannot pass the blood–brain barrier (BBB). The majority of TRP is metabolized to form KYN in the liver and is released into peripheral blood. The initial and rate-limiting step in the KYN pathway is catalyzed by indoleamine 2,3-dioxygenase 1 (IDO1), IDO2 and/or tryptophan 2,3-dioxygenase (TDO2)—depending on the tissue involved—to form N-formyl-kynurenine. N-formyl-kynurenine is metabolized by formamidase to produce KYN, which can cross the BBB. Approximately 60% of KYN in the CNS originates from the liver. In the periphery and in the CNS, KYN can be further metabolized by either kynurenine aminotransferases (KATs) or by kynurenine 3-monooxygenase (KMO) and kynureninase (KYNU), leading to the generation of the neuroactive metabolites kynurenic acid (KYNA) or quinolinic acid (QUIN), respectively. KYNA, which is neuroprotective, and QUIN, which is neuroactive in the CNS, have opposite effects on the NMDA receptor. Four KATs, encoded by AADAT, CCBL1, CCBL2, and GOT2, have been shown to catalyze the conversion of KYN to KYNA. The other branches of the KYN pathway involve KMO and KYNU, which catalyze the metabolism of kynurenine to form 3-hydroxykynurenine and 3-hydroxyanthranilic acid, respectively. KYNU can also metabolize KYN to form anthranilic acid, which can then be coverted to 3-hydroxyanthranilic acid by nonspecific hydroxylation. 3-Hydroxyanthranilic acid is metabolized further by 3-hydroxyanthranilic acid 3,4-dioxygenase (HAAO) to form either QUIN or, after a series of reactions, picolinic acid
Fig. 2. Plasma KYN concentrations GWAS
Fig. 2. Plasma KYN concentrations GWAS
a Manhattan plot for baseline plasma KYN concentrations. SNPs across the DEFB1 and AHR genes have been highlighted, with rs5743467 as the “top” DEFB1 SNP (P-value = 8.18E−07) and, rs17137566 as the “top” AHR SNP (P-value = 6.22E−06). Regional association plots (Locus Zooms) for the DEFB1 gene B and the AHR gene C are also shown. Circles and diamonds represent observed and imputed SNPs, respectively. The color of each SNP represents its’ linkage disequilibrium (LD) with the “top SNP”, which is colored purple
Fig. 3
Fig. 3
eQTL analysis for the top DEFB1A and AHRB SNPs based on the GTEx dataset. ADEFB1 mRNA expression was significantly decreased in human cerebellum (left), transverse colon (middle) and esophageal mucosa (right) from individuals with rs5743467 variant genotypes (G) when compared with WT genotype (C) (P < 0.05). BAHR mRNA expression was significantly decreased in human cerebellum (left), sigmoid colon (middle) and esophageal mucosa (right) from subjects with rs17137566 variant genotypes (C) when compared with those for subjects homozygous for the WT genotype (T) (P < 0.05). mRNA levels were determined by RNA sequencing data available in GTEx
Fig. 4. DEFB1 functional studies in THP-1…
Fig. 4. DEFB1 functional studies in THP-1 cells
A mRNA expression was determined by qRT-PCR after THP-1 cells were exposed to 10 and 100 ng/ml of LPS at different time points. Compared to vehicle treated cells, IDO1 mRNA levels were significantly increased after LPS treatment. mRNA levels for TNF, a pro-inflammatory cytokine used as a positive control for LPS effect, were also significantly increased after LPS treatment. B IDO1 protein expression was increased as analyzed by Western blot after LPS treatment. C KYN concentrations (left) in cell culture media were undetectable after 24 and 48 h of vehicle treatments, but were significantly increased after 10 ng/ml LPS treatment. At the same time, TRP concentrations (middle) were significantly decreased in cell culture media after LPS treatment and the K/T ratio was increased (right). After 3, 6, or 12 h of LPS treatment, KYN concentrations were undetectable. D mRNA levels for IDO1 and TNF were significantly increased after 10 ng/ml LPS treatment, but recombinant human DEFB1 co-incubation with LPS significantly decreased mRNA levels for IDO1 and TNF when compared with LPS treatment alone. E When DEFB1 was co-incubated with LPS as compared to LPS alone, KYN concentrations (left) in cell culture media were significantly decreased, TRP concentrations were increased (middle) and K/T ratios (right) were decreased after DEFB1 was pre-incubated with LPS when compared with results for cells treated with LPS alone. N ≥ 3 for all the experiments. Data = mean ± SEM, with statistical significance determined by two-tailed t-test denoted as *P < 0.05, **P < 0.01, and ***P < 0.001
Fig. 5. AHR functional studies in HepaRG…
Fig. 5. AHR functional studies in HepaRG and U-87 MG cells
A mRNA expression determined by qRT-PCR after AHR KD in HepaRG cells. TDO2, KMO, and KYNU expression was significantly increased and AHRR and CYP1A1 expression was significantly decreased following AHR KD. B Protein expression analysis for AHR, TDO2, KMO, and KYNU by Western blot analysis after AHR KD in HepaRG cells. C mRNA expression in HepaRG cells determined by qRT-PCR after 24-h treatment with 1 µM 3-MC, an AHR agonist. TDO2, KMO, and KYNU expression was significantly decreased. AHRR, CYP1A1 (in HepaRG), was  induced by 3-MC, indicating that AHR was activated by the treatment. D KYN concentrations in HepaRG cell culture media after AHR and AHR plus KMO or KYNU KD. ETDO2, KMO, and KYNU expression was significantly increased while AHRR and CYP1B1 expression was significantly decreased following AHR KD. F AHR, TDO2, KMO, and KYNU protein concentrations were significantly altered in U-87 MG cells after AHR KD. G mRNA expression in U-87 MG cells after 1 µM 3-MC treatment showed significant decreases in TDO2, KMO, and KYNU expression and significant increases in AHRR and CYP1B1 expression. H KYN concentrations in U-87 MG cell culture media were significantly decreased after AHR KD but were less decreased after AHR KD together with KMO or KYNU KD. N ≥ 3 for all experiments. Data = mean ± SEM, with statistical significance determined by two-tailed t-test denoted as *P < 0.05, **P < 0.01 and ***P < 0.001 when compared with control. ns = not significant

References

    1. Martinowich K, Jimenez DV, Zarate CA, Jr., Manji HK. Rapid antidepressant effects: moving right along. Mol Psychiatry. 2013;18:856–863. doi: 10.1038/mp.2013.55.
    1. Kessler RC, et al. Prevalence and effects of mood disorders on work performance in a nationally representative sample of U.S. workers. Am J Psychiatry. 2006;163:1561–1568. doi: 10.1176/ajp.2006.163.9.1561.
    1. Trivedi MH, et al. Evaluation of outcomes with citalopram for depression using measurement-based care in STAR*D: implications for clinical practice. Am J Psychiatry. 2006;163:28–40. doi: 10.1176/appi.ajp.163.1.28.
    1. Sullivan PF, et al. Genome-wide association for major depressive disorder: a possible role for the presynaptic protein piccolo. Mol Psychiatry. 2009;14:359–375. doi: 10.1038/mp.2008.125.
    1. Wray NR, et al. Genome-wide association study of major depressive disorder: new results, meta-analysis, and lessons learned. Mol Psychiatry. 2012;17:36–48. doi: 10.1038/mp.2010.109.
    1. Ripke S, et al. A mega-analysis of genome-wide association studies for major depressive disorder. Mol Psychiatry. 2013;18:497–511. doi: 10.1038/mp.2012.21.
    1. Ising M, et al. A genomewide association study points to multiple loci that predict antidepressant drug treatment outcome in depression. Arch. Gen. Psychiatry. 2009;66:966–975. doi: 10.1001/archgenpsychiatry.2009.95.
    1. Garriock HA, et al. A genomewide association study of citalopram response in major depressive disorder. Biol. Psychiatry. 2010;67:133–138. doi: 10.1016/j.biopsych.2009.08.029.
    1. Uher R, et al. Genome-wide pharmacogenetics of antidepressant response in the GENDEP project. Am. J. Psychiatry. 2010;167:555–564. doi: 10.1176/appi.ajp.2009.09070932.
    1. Ji Y, et al. Pharmacogenomics of selective serotonin reuptake inhibitor treatment for major depressive disorder: genome-wide associations and functional genomics. Pharmacogenom. J. 2013;13:456–463. doi: 10.1038/tpj.2012.32.
    1. Biernacka JM, et al. The International SSRI Pharmacogenomics Consortium (ISPC): a genome-wide association study of antidepressant treatment response. Transl. Psychiatry. 2015;5:e553. doi: 10.1038/tp.2015.47.
    1. Tansey KE, et al. Contribution of common genetic variants to antidepressant response. Biol. Psychiatry. 2013;73:679–682. doi: 10.1016/j.biopsych.2012.10.030.
    1. Krishnan V, Nestler EJ. The molecular neurobiology of depression. Nature. 2008;455:894–902. doi: 10.1038/nature07455.
    1. Mrazek DA, et al. Treatment outcomes of depression: the pharmacogenomic research network antidepressant medication pharmacogenomic study. J. Clin. Psychopharmacol. 2014;34:313–317. doi: 10.1097/JCP.0000000000000099.
    1. Ji Y, et al. Citalopram and escitalopram plasma drug and metabolite concentrations: genome-wide associations. Br. J. Clin. Pharmacol. 2014;78:373–383. doi: 10.1111/bcp.12348.
    1. Kaddurah-Daouk R, Weinshilboum RM. Pharmacometabolomics: implications for clinical pharmacology and systems pharmacology. Clin. Pharmacol. Ther. 2014;95:154–167. doi: 10.1038/clpt.2013.217.
    1. Neavin D, Kaddurah-Daouk R, Weinshilboum R. Pharmacometabolomics informs pharmacogenomics. Metabolomics. 2016;12:121. doi: 10.1007/s11306-016-1066-x.
    1. Kaddurah-Daouk R, et al. Pretreatment metabotype as a predictor of response to sertraline or placebo in depressed outpatients: a proof of concept. Transl. Psychiatry. 2011;1:e26. doi: 10.1038/tp.2011.22.
    1. Kaddurah-Daouk R, et al. Pharmacometabolomic mapping of early biochemical changes induced by sertraline and placebo. Transl. Psychiatry. 2013;3:e223. doi: 10.1038/tp.2012.142.
    1. Zhu H, et al. Pharmacometabolomics of response to sertraline and to placebo in major depressive disorder—possible role for methoxyindole pathway. PLoS One. 2013;8:e68283. doi: 10.1371/journal.pone.0068283.
    1. Gupta M, et al. TSPAN5, ERICH3 and selective serotonin reuptake inhibitors in major depressive disorder: pharmacometabolomics-informed pharmacogenomics. Mol. Psychiatry. 2016;21:1717–1725. doi: 10.1038/mp.2016.6.
    1. Miller AH, Raison CL. The role of inflammation in depression: from evolutionary imperative to modern treatment target. Nat. Rev. Immunol. 2016;16:22–34. doi: 10.1038/nri.2015.5.
    1. Raison CL, Capuron L, Miller AH. Cytokines sing the blues: inflammation and the pathogenesis of depression. Trends Immunol. 2006;27:24–31. doi: 10.1016/j.it.2005.11.006.
    1. Miller AH, Maletic V, Raison CL. Inflammation and its discontents: the role of cytokines in the pathophysiology of major depression. Biol. Psychiatry. 2009;65:732–741. doi: 10.1016/j.biopsych.2008.11.029.
    1. Thomas AJ, et al. Increase in interleukin-1beta in late-life depression. Am. J. Psychiatry. 2005;162:175–177. doi: 10.1176/appi.ajp.162.1.175.
    1. Alesci S, et al. Major depression is associated with significant diurnal elevations in plasma interleukin-6 levels, a shift of its circadian rhythm, and loss of physiological complexity in its secretion: clinical implications. J. Clin. Endocrinol. Metab. 2005;90:2522–2530. doi: 10.1210/jc.2004-1667.
    1. Iwata M, et al. Psychological stress activates the inflammasome via release of adenosine triphosphate and stimulation of the purinergic type 2X7 receptor. Biol. Psychiatry. 2016;80:12–22. doi: 10.1016/j.biopsych.2015.11.026.
    1. Rogers GB, et al. From gut dysbiosis to altered brain function and mental illness: mechanisms and pathways. Mol. Psychiatry. 2016;21:738–748. doi: 10.1038/mp.2016.50.
    1. Foster JA, McVey Neufeld KA. Gut-brain axis: how the microbiome influences anxiety and depression. Trends Neurosci. 2013;36:305–312. doi: 10.1016/j.tins.2013.01.005.
    1. Maes M, Kubera M, Leunis JC. The gut-brain barrier in major depression: intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression. Neuro Endocrinol. Lett. 2008;29:117–124.
    1. Raison CL, et al. A randomized controlled trial of the tumor necrosis factor antagonist infliximab for treatment-resistant depression: the role of baseline inflammatory biomarkers. JAMA Psychiatry. 2013;70:31–41. doi: 10.1001/2013.jamapsychiatry.4.
    1. Kohler O, et al. Effect of anti-inflammatory treatment on depression, depressive symptoms, and adverse effects: a systematic review and meta-analysis of randomized clinical trials. JAMA Psychiatry. 2014;71:1381–1391. doi: 10.1001/jamapsychiatry.2014.1611.
    1. Kappelmann, N., Lewis, G., Dantzer, R., Jones, P. B., Khandaker, G. M. Antidepressant activity of anti-cytokine treatment: a systematic review and meta-analysis of clinical trials of chronic inflammatory conditions. Mol. Psychiatry advance online publication, 18 October 2016; doi:10.1038/mp.2016. 167 (2016).
    1. Lapin IP, Oxenkrug GF. Intensification of the central serotoninergic processes as a possible determinant of the thymoleptic effect. Lancet. 1969;1:132–136. doi: 10.1016/S0140-6736(69)91140-4.
    1. Oxenkrug GF. Tryptophan kynurenine metabolism as a common mediator of genetic and environmental impacts in major depressive disorder: the serotonin hypothesis revisited 40 years later. Isr. J. Psychiatry Relat. Sci. 2010;47:56–63.
    1. Schwarcz R, Bruno JP, Muchowski PJ, Wu HQ. Kynurenines in the mammalian brain: when physiology meets pathology. Nat. Rev. Neurosci. 2012;13:465–477. doi: 10.1038/nrn3257.
    1. O’Connor JC, et al. Lipopolysaccharide-induced depressive-like behavior is mediated by indoleamine 2,3-dioxygenase activation in mice. Mol. Psychiatry. 2009;14:511–522. doi: 10.1038/sj.mp.4002148.
    1. O’Connor JC, et al. Interferon-gamma and tumor necrosis factor-alpha mediate the upregulation of indoleamine 2,3-dioxygenase and the induction of depressive-like behavior in mice in response to bacillus Calmette-Guerin. J. Neurosci. 2009;29:4200–4209. doi: 10.1523/JNEUROSCI.5032-08.2009.
    1. Raison CL, et al. CSF concentrations of brain tryptophan and kynurenines during immune stimulation with IFN-alpha: relationship to CNS immune responses and depression. Mol. Psychiatry. 2010;15:393–403. doi: 10.1038/mp.2009.116.
    1. Agudelo LZ, et al. Skeletal muscle PGC-1alpha1 modulates kynurenine metabolism and mediates resilience to stress-induced depression. Cell. 2014;159:33–45. doi: 10.1016/j.cell.2014.07.051.
    1. Ganz T. Defensins: antimicrobial peptides of innate immunity. Nat. Rev. Immunol. 2003;3:710–720. doi: 10.1038/nri1180.
    1. Schroeder BO, et al. Reduction of disulphide bonds unmasks potent antimicrobial activity of human beta-defensin 1. Nature. 2011;469:419–423. doi: 10.1038/nature09674.
    1. Ostaff MJ, Stange EF, Wehkamp J. Antimicrobial peptides and gut microbiota in homeostasis and pathology. EMBO Mol. Med. 2013;5:1465–1483. doi: 10.1002/emmm.201201773.
    1. Murray IA, Patterson AD, Perdew GH. Aryl hydrocarbon receptor ligands in cancer: friend and foe. Nat. Rev. Cancer. 2014;14:801–814. doi: 10.1038/nrc3846.
    1. Denison MS, Nagy SR. Activation of the aryl hydrocarbon receptor by structurally diverse exogenous and endogenous chemicals. Annu. Rev. Pharmacol. Toxicol. 2003;43:309–334. doi: 10.1146/annurev.pharmtox.43.100901.135828.
    1. Vogel CF, Goth SR, Dong B, Pessah IN, Matsumura F. Aryl hydrocarbon receptor signaling mediates expression of indoleamine 2,3-dioxygenase. Biochem. Biophys. Res. Commun. 2008;375:331–335. doi: 10.1016/j.bbrc.2008.07.156.
    1. Jux B, Kadow S, Esser C. Langerhans cell maturation and contact hypersensitivity are impaired in aryl hydrocarbon receptor-null mice. J. Immunol. 2009;182:6709–6717. doi: 10.4049/jimmunol.0713344.
    1. Nguyen NT, et al. Aryl hydrocarbon receptor negatively regulates dendritic cell immunogenicity via a kynurenine-dependent mechanism. Proc. Natl. Acad. Sci. USA. 2010;107:19961–19966. doi: 10.1073/pnas.1014465107.
    1. Opitz CA, et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature. 2011;478:197–203. doi: 10.1038/nature10491.
    1. Bessede A, et al. Aryl hydrocarbon receptor control of a disease tolerance defence pathway. Nature. 2014;511:184–190. doi: 10.1038/nature13323.
    1. GTExConsortium. The Genotype-Tissue Expression (GTEx) pilot analysis: multitissue gene regulation in humans. Science. 2015;348:648–660. doi: 10.1126/science.1262110.
    1. Ramasamy A, et al. Genetic variability in the regulation of gene expression in ten regions of the human brain. Nat. Neurosci. 2014;17:1418–1428. doi: 10.1038/nn.3801.
    1. Niu N, et al. Radiation pharmacogenomics: a genome-wide association approach to identify radiation response biomarkers using human lymphoblastoid cell lines. Genome Res. 2010;20:1482–1492. doi: 10.1101/gr.107672.110.
    1. Liu D, et al. Breast cancer chemoprevention pharmacogenomics: Deep sequencing and functional genomics of the ZNF423 and CTSO genes. npj Breast Cancer. 2017;3:30. doi: 10.1038/s41523-017-0036-4.
    1. Lee SH, Jun HK, Lee HR, Chung CP, Choi BK. Antibacterial and lipopolysaccharide (LPS)-neutralising activity of human cationic antimicrobial peptides against periodontopathogens. Int. J. Antimicrob. Agents. 2010;35:138–145. doi: 10.1016/j.ijantimicag.2009.09.024.
    1. Takashiba S, et al. Differentiation of monocytes to macrophages primes cells for lipopolysaccharide stimulation via accumulation of cytoplasmic nuclear factor kappaB. Infect. Immun. 1999;67:5573–5578.
    1. Park EK, et al. Optimized THP-1 differentiation is required for the detection of responses to weak stimuli. Inflamm. Res. 2007;56:45–50. doi: 10.1007/s00011-007-6115-5.
    1. Daigneault M, Preston JA, Marriott HM, Whyte MK, Dockrell DH. The identification of markers of macrophage differentiation in PMA-stimulated THP-1 cells and monocyte-derived macrophages. PLoS One. 2010;5:e8668. doi: 10.1371/journal.pone.0008668.
    1. Cerec V, et al. Transdifferentiation of hepatocyte-like cells from the human hepatoma HepaRG cell line through bipotent progenitor. Hepatology. 2007;45:957–967. doi: 10.1002/hep.21536.
    1. Denison MS, Whitlock JP., Jr. Xenobiotic-inducible transcription of cytochrome P450 genes. J. Biol. Chem. 1995;270:18175–18178. doi: 10.1074/jbc.270.31.18175.
    1. Bersten DC, Sullivan AE, Peet DJ, Whitelaw ML. bHLH-PAS proteins in cancer. Nat. Rev. Cancer. 2013;13:827–841. doi: 10.1038/nrc3621.
    1. Setoyama D, et al. Plasma metabolites predict severity of depression and suicidal ideation in psychiatric patients—a multicenter pilot analysis. PLoS One. 2016;11:e0165267. doi: 10.1371/journal.pone.0165267.
    1. Orlikov AB, Prakhye IB, Ryzov IV. Kynurenine in blood plasma and DST in patients with endogenous anxiety and endogenous depression. Biol. Psychiatry. 1994;36:97–102. doi: 10.1016/0006-3223(94)91189-4.
    1. O’Neil DA, et al. Expression and regulation of the human beta-defensins hBD-1 and hBD-2 in intestinal epithelium. J. Immunol. 1999;163:6718–6724.
    1. Kocsis AK, et al. Association of beta-defensin 1 single nucleotide polymorphisms with Crohn’s disease. Scand. J. Gastroenterol. 2008;43:299–307. doi: 10.1080/00365520701682615.
    1. Chen QX, et al. Genomic variations within DEFB1 are associated with the susceptibility to and the fatal outcome of severe sepsis in Chinese Han population. Genes Immun. 2007;8:439–443. doi: 10.1038/sj.gene.6364401.
    1. Jurevic RJ, Bai M, Chadwick RB, White TC, Dale BA. Single-nucleotide polymorphisms (SNPs) in human beta-defensin 1: high-throughput SNP assays and association with Candida carriage in type I diabetics and nondiabetic controls. J. Clin. Microbiol. 2003;41:90–96. doi: 10.1128/JCM.41.1.90-96.2003.
    1. Kalus AA, et al. Association of a genetic polymorphism (-44 C/G SNP) in the human DEFB1 gene with expression and inducibility of multiple beta-defensins in gingival keratinocytes. BMC Oral Health. 2009;9:21. doi: 10.1186/1472-6831-9-21.
    1. Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 2016;16:341–352. doi: 10.1038/nri.2016.42.
    1. Sekirov I, Russell SL, Antunes LC, Finlay BB. Gut microbiota in health and disease. Physiol. Rev. 2010;90:859–904. doi: 10.1152/physrev.00045.2009.
    1. Zhang Y, et al. Crystal structure and mechanism of tryptophan 2,3-dioxygenase, a heme enzyme involved in tryptophan catabolism and in quinolinate biosynthesis. Biochemistry. 2007;46:145–155. doi: 10.1021/bi0620095.
    1. Wonodi I, et al. Downregulated kynurenine 3-monooxygenase gene expression and enzyme activity in schizophrenia and genetic association with schizophrenia endophenotypes. Arch. Gen. Psychiatry. 2011;68:665–674. doi: 10.1001/archgenpsychiatry.2011.71.
    1. Lavebratt C, et al. The KMO allele encoding Arg452 is associated with psychotic features in bipolar disorder type 1, and with increased CSF KYNA level and reduced KMO expression. Mol. Psychiatry. 2014;19:334–341. doi: 10.1038/mp.2013.11.
    1. Zwilling D, et al. Kynurenine 3-monooxygenase inhibition in blood ameliorates neurodegeneration. Cell. 2011;145:863–874. doi: 10.1016/j.cell.2011.05.020.
    1. Myint AM, et al. Kynurenine pathway in major depression: evidence of impaired neuroprotection. J. Affect. Disord. 2007;98:143–151. doi: 10.1016/j.jad.2006.07.013.
    1. Muller N, Schwarz MJ. The immune-mediated alteration of serotonin and glutamate: towards an integrated view of depression. Mol. Psychiatry. 2007;12:988–1000. doi: 10.1038/sj.mp.4002006.

Source: PubMed

3
Abonnieren