Abarenkov,, K., Henrik Nilsson,, R., Larsson,, K. H., Alexander,, I. J., Eberhardt,, U., Erland,, S., … Koljalg,, U. (2010). The UNITE database for molecular identification of fungi—Recent updates and future perspectives. New Phytologist, 186(2), 281–285. https://doi.org/10.1111/j.1469-8137.2009.03160.x
Adam,, B., Baillie,, G. S., & Douglas,, L. J. (2002). Mixed species biofilms of Candida albicans and Staphylococcus epidermidis. Journal of Medical Microbiology, 51(4), 344–349. https://doi.org/10.1099/0022-1317-51-4-344
Angebault,, C., Djossou,, F., Abelanet,, S., Permal,, E., Ben Soltana,, M., Diancourt,, L., … Bougnoux,, M. E. (2013). Candida albicans is not always the preferential yeast colonizing humans: A study in Wayampi Amerindians. Journal of Infectious Diseases, 208(10), 1705–1716. https://doi.org/10.1093/infdis/jit389
Arts,, R. J., Novakovic,, B., Ter Horst,, R., Carvalho,, A., Bekkering,, S., Lachmandas,, E., … Netea,, M. G. (2016). Glutaminolysis and fumarate accumulation integrate immunometabolic and epigenetic programs in trained immunity. Cell Metabolism, 24(6), 807–819. https://doi.org/10.1016/j.cmet.2016.10.008
Arumugam,, M., Raes,, J., Pelletier,, E., Le Paslier,, D., Yamada,, T., Mende,, D. R., … Bork,, P. (2011). Enterotypes of the human gut microbiome. Nature, 473(7346), 174–180. https://doi.org/10.1038/nature09944
Atarashi,, K., Tanoue,, T., Ando,, M., Kamada,, N., Nagano,, Y., Narushima,, S., … Honda,, K. (2015). Th17 cell induction by adhesion of microbes to intestinal epithelial cells. Cell, 163(2), 367–380. https://doi.org/10.1016/j.cell.2015.08.058
Atarashi,, K., Tanoue,, T., Oshima,, K., Suda,, W., Nagano,, Y., Nishikawa,, H., … Honda,, K. (2013). Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature, 500(7461), 232–236. https://doi.org/10.1038/nature12331
Atarashi,, K., Tanoue,, T., Shima,, T., Imaoka,, A., Kuwahara,, T., Momose,, Y., … Honda,, K. (2011). Induction of colonic regulatory T cells by indigenous Clostridium species. Science, 331(6015), 337–341. https://doi.org/10.1126/science.1198469
Auchtung,, T. A., Fofanova,, T. Y., Stewart,, C. J., Nash,, A. K., Wong,, M. C., Gesell,, J. R., … Petrosino,, J. F. (2018). Investigating colonization of the healthy adult gastrointestinal tract by fungi. mSphere, 3(2), e00092‐18. https://doi.org/10.1128/mSphere.00092-18
Bacher,, P., Kniemeyer,, O., Schonbrunn,, A., Sawitzki,, B., Assenmacher,, M., Rietschel,, E., … Scheffold,, A. (2014). Antigen‐specific expansion of human regulatory T cells as a major tolerance mechanism against mucosal fungi. Mucosal Immunology, 7(4), 916–928. https://doi.org/10.1038/mi.2013.107
Bajaj,, J. S., Liu,, E. J., Kheradman,, R., Fagan,, A., Heuman,, D. M., White,, M., … Gillevet,, P. M. (2018). Fungal dysbiosis in cirrhosis. Gut, 67(6), 1146–1154. https://doi.org/10.1136/gutjnl-2016-313170
Bak‐Romaniszyn,, L., Szala,, A., Sokolowska,, A., Mierzwa,, G., Czerwionka‐Szaflarska,, M., Swierzko,, A. S., … Cedzynski,, M. (2011). Mannan‐binding lectin deficiency in pediatric patients with inflammatory bowel disease. Scandinavian Journal of Gastroenterology, 46(10), 1275–1278. https://doi.org/10.3109/00365521.2011.594087
Bekkering,, S., Arts,, R. J. W., Novakovic,, B., Kourtzelis,, I., van der Heijden,, C., Li,, Y., … Netea,, M. G. (2018). Metabolic induction of trained immunity through the mevalonate pathway. Cell, 172(1–2), 135–146 e139. https://doi.org/10.1016/j.cell.2017.11.025
Belkaid,, Y., & Segre,, J. A. (2014). Dialogue between skin microbiota and immunity. Science, 346(6212), 954–959. https://doi.org/10.1126/science.1260144
Bellemain,, E., Carlsen,, T., Brochmann,, C., Coissac,, E., Taberlet,, P., & Kauserud,, H. (2010). ITS as an environmental DNA barcode for fungi: An in silico approach reveals potential PCR biases. BMC Microbiology, 10, 189. https://doi.org/10.1186/1471-2180-10-189
Bliss,, J. M., Basavegowda,, K. P., Watson,, W. J., Sheikh,, A. U., & Ryan,, R. M. (2008). Vertical and horizontal transmission of Candida albicans in very low birth weight infants using DNA fingerprinting techniques. Pediatric Infectious Disease Journal, 27(3), 231–235. https://doi.org/10.1097/INF.0b013e31815bb69d
Blostein,, F., Levin‐Sparenberg,, E., Wagner,, J., & Foxman,, B. (2017). Recurrent vulvovaginal candidiasis. Annals of Epidemiology, 27(9), 575–582 e573. https://doi.org/10.1016/j.annepidem.2017.08.010
Bohm,, L., Torsin,, S., Tint,, S. H., Eckstein,, M. T., Ludwig,, T., & Perez,, J. C. (2017). The yeast form of the fungus Candida albicans promotes persistence in the gut of gnotobiotic mice. PLoS Pathogens, 13(10), e1006699. https://doi.org/10.1371/journal.ppat.1006699
Boisson,, B., Wang,, C., Pedergnana,, V., Wu,, L., Cypowyj,, S., Rybojad,, M., … Casanova,, J. L. (2013). An ACT1 mutation selectively abolishes interleukin‐17 responses in humans with chronic mucocutaneous candidiasis. Immunity, 39(4), 676–686. https://doi.org/10.1016/j.immuni.2013.09.002
Bolnick,, D. I., Snowberg,, L. K., Hirsch,, P. E., Lauber,, C. L., Org,, E., Parks,, B., … Svanback,, R. (2014). Individual diet has sex‐dependent effects on vertebrate gut microbiota. Nature Communications, 5, 4500. https://doi.org/10.1038/ncomms5500
Boon,, C., Deng,, Y., Wang,, L. H., He,, Y., Xu,, J. L., Fan,, Y., … Zhang,, L. H. (2008). A novel DSF‐like signal from Burkholderia cenocepacia interferes with Candida albicans morphological transition. ISME Journal, 2(1), 27–36. https://doi.org/10.1038/ismej.2007.76
Brasch,, J., & Christophers,, E. (1993). Azelaic acid has antimycotic properties in vitro. Dermatology, 186(1), 55–58. https://doi.org/10.1159/000247303
Calderon,, E. J., Rivero,, L., Respaldiza,, N., Morilla,, R., Montes‐Cano,, M. A., Friaza,, V., … Horra Cde,, L. (2007). Systemic inflammation in patients with chronic obstructive pulmonary disease who are colonized with Pneumocystis jiroveci. Clinical Infectious Diseases, 45(2), e17–e19. https://doi.org/10.1086/518989
Carini,, P., Marsden,, P. J., Leff,, J. W., Morgan,, E. E., Strickland,, M. S., & Fierer,, N. (2016). Relic DNA is abundant in soil and obscures estimates of soil microbial diversity. Nature Microbiology, 2, 16242. https://doi.org/10.1038/nmicrobiol.2016.242
Carvalho,, A., Giovannini,, G., De Luca,, A., D`Angelo,, C., Casagrande,, A., Iannitti,, R. G., … Romani,, L. (2012). Dectin‐1 isoforms contribute to distinct Th1/Th17 cell activation in mucosal candidiasis. Cellular %26 Molecular Immunology, 9(3), 276–286. https://doi.org/10.1038/cmi.2012.1
Casadevall,, A., & Pirofski,, L. A. (1999). Host–pathogen interactions: Redefining the basic concepts of virulence and pathogenicity. Infection and Immunity, 67(8), 3703–3713.
Cella,, M., & Colonna,, M. (2015). Aryl hydrocarbon receptor: Linking environment to immunity. Seminars in Immunology, 27(5), 310–314. https://doi.org/10.1016/j.smim.2015.10.002
Chang,, P. V., Hao,, L., Offermanns,, S., & Medzhitov,, R. (2014). The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proceedings of the National Academy of Sciences of the United States of America, 111(6), 2247–2252. https://doi.org/10.1073/pnas.1322269111
Chaudhary,, N., Staab,, J. F., & Marr,, K. A. (2010). Healthy human T‐cell responses to Aspergillus fumigatus antigens. PLoS One, 5(2), e9036. https://doi.org/10.1371/journal.pone.0009036
Chehoud,, C., Albenberg,, L. G., Judge,, C., Hoffmann,, C., Grunberg,, S., Bittinger,, K., … Wu,, G. D. (2015). Fungal signature in the gut microbiota of pediatric patients with inflammatory bowel disease. Inflammatory Bowel Diseases, 21(8), 1948–1956. https://doi.org/10.1097/MIB.0000000000000454
Chen,, Y., Chen,, Z., Guo,, R., Chen,, N., Lu,, H., Huang,, S., … Li,, L. (2011). Correlation between gastrointestinal fungi and varying degrees of chronic hepatitis B virus infection. Diagnostic Microbiology and Infectious Disease, 70(4), 492–498. https://doi.org/10.1016/j.diagmicrobio.2010.04.005
Cheng,, S. C., Quintin,, J., Cramer,, R. A., Shepardson,, K. M., Saeed,, S., Kumar,, V., … Netea,, M. G. (2014). mTOR‐ and HIF‐1alpha‐mediated aerobic glycolysis as metabolic basis for trained immunity. Science, 345(6204), 1250684. https://doi.org/10.1126/science.1250684
Chiaro,, T. R., Soto,, R., Zac Stephens,, W., Kubinak,, J. L., Petersen,, C., Gogokhia,, L., … Round,, J. L. (2017). A member of the gut mycobiota modulates host purine metabolism exacerbating colitis in mice. Science Translational Medicine, 9(380), eaaf9044. https://doi.org/10.1126/scitranslmed.aaf9044
Cho,, I., & Blaser,, M. J. (2012). The human microbiome: At the interface of health and disease. Nature Reviews. Genetics, 13(4), 260–270. https://doi.org/10.1038/nrg3182
Choteau,, L., Parny,, M., Francois,, N., Bertin,, B., Fumery,, M., Dubuquoy,, L., … Jawhara,, S. (2016). Role of mannose‐binding lectin in intestinal homeostasis and fungal elimination. Mucosal Immunology, 9(3), 767–776. https://doi.org/10.1038/mi.2015.100
Christensen,, P. J., Preston,, A. M., Ling,, T., Du,, M., Fields,, W. B., Curtis,, J. L., & Beck,, J. M. (2008). Pneumocystis murina infection and cigarette smoke exposure interact to cause increased organism burden, development of airspace enlargement, and pulmonary inflammation in mice. Infection and Immunity, 76(8), 3481–3490. https://doi.org/10.1128/IAI.00165-08
Chung,, H., & Kasper,, D. L. (2010). Microbiota‐stimulated immune mechanisms to maintain gut homeostasis. Current Opinion in Immunology, 22(4), 455–460. https://doi.org/10.1016/j.coi.2010.06.008
Clarke,, T. B., Davis,, K. M., Lysenko,, E. S., Zhou,, A. Y., Yu,, Y., & Weiser,, J. N. (2010). Recognition of peptidoglycan from the microbiota by Nod1 enhances systemic innate immunity. Nature Medicine, 16(2), 228–231. https://doi.org/10.1038/nm.2087
Clemente,, J. C., Pehrsson,, E. C., Blaser,, M. J., Sandhu,, K., Gao,, Z., Wang,, B., … Dominguez‐Bello,, M. G. (2015). The microbiome of uncontacted Amerindians. Science Advances, 1(3), e1500183. https://doi.org/10.1126/sciadv.1500183
Collins,, S. M., Surette,, M., & Bercik,, P. (2012). The interplay between the intestinal microbiota and the brain. Nature Reviews. Microbiology, 10(11), 735–742. https://doi.org/10.1038/nrmicro2876
Cottier,, F., & Pavelka,, N. (2012). Complexity and dynamics of host–fungal interactions. Immunologic Research, 53(1–3), 127–135. https://doi.org/10.1007/s12026-012-8265-y
Cottier,, F., Srinivasan,, K. G., Yurieva,, M., Liao,, W., Poidinger,, M., Zolezzi,, F., & Pavelka,, N. (2018). Advantages of meta‐total RNA sequencing (MeTRS) over shotgun metagenomics and amplicon‐based sequencing in the profiling of complex microbial communities. NPJ Biofilms Microbiomes, 4, 2. https://doi.org/10.1038/s41522-017-0046-x
Cottier,, F., Tan,, A. S., Xu,, X., Wang,, Y., & Pavelka,, N. (2015). MIG1 regulates resistance of Candida albicans against the fungistatic effect of weak organic acids. Eukaryotic Cell, 14(10), 1054–1061. https://doi.org/10.1128/EC.00129-15
Cottier,, F., Tan,, A. S. M., Yurieva,, M., Liao,, W., Lum,, J., Poidinger,, M., … Pavelka,, N. (2017). The transcriptional response of Candida albicans to weak organic acids, carbon source, and MIG1 inactivation unveils a role for HGT16 in mediating the fungistatic effect of acetic acid. G3 (Bethesda), 7(11), 3597–3604. https://doi.org/10.1534/g3.117.300238
Cruz,, M. R., Graham,, C. E., Gagliano,, B. C., Lorenz,, M. C., & Garsin,, D. A. (2013). Enterococcus faecalis inhibits hyphal morphogenesis and virulence of Candida albicans. Infection and Immunity, 81(1), 189–200. https://doi.org/10.1128/IAI.00914-12
David,, L. A., Maurice,, C. F., Carmody,, R. N., Gootenberg,, D. B., Button,, J. E., Wolfe,, B. E., … Turnbaugh,, P. J. (2014). Diet rapidly and reproducibly alters the human gut microbiome. Nature, 505(7484), 559–563. https://doi.org/10.1038/nature12820
de Beaucoudrey,, L., Samarina,, A., Bustamante,, J., Cobat,, A., Boisson‐Dupuis,, S., Feinberg,, J., … Casanova,, J. L. (2010). Revisiting human IL‐12Rbeta1 deficiency: A survey of 141 patients from 30 countries. Medicine (Baltimore), 89(6), 381–402. https://doi.org/10.1097/MD.0b013e3181fdd832
De Filippo,, C., Cavalieri,, D., Di Paola,, M., Ramazzotti,, M., Poullet,, J. B., Massart,, S., … Lionetti,, P. (2010). Impact of diet in shaping gut microbiota revealed by a comparative study in children from Europe and rural Africa. Proceedings of the National Academy of Sciences of the United States of America, 107(33), 14691–14696. https://doi.org/10.1073/pnas.1005963107
Delhaes,, L., Monchy,, S., Frealle,, E., Hubans,, C., Salleron,, J., Leroy,, S., … Viscogliosi,, E. (2012). The airway microbiota in cystic fibrosis: A complex fungal and bacterial community—Implications for therapeutic management. PLoS One, 7(4), e36313. https://doi.org/10.1371/journal.pone.0036313
Dopico,, X. C., Evangelou,, M., Ferreira,, R. C., Guo,, H., Pekalski,, M. L., Smyth,, D. J., … Todd,, J. A. (2015). Widespread seasonal gene expression reveals annual differences in human immunity and physiology. Nature Communications, 6, 7000. https://doi.org/10.1038/ncomms8000
Drell,, T., Lillsaar,, T., Tummeleht,, L., Simm,, J., Aaspollu,, A., Vain,, E., … Metsis,, M. (2013). Characterization of the vaginal micro‐ and mycobiome in asymptomatic reproductive‐age Estonian women. PLoS One, 8(1), e54379. https://doi.org/10.1371/journal.pone.0054379
Drewniak,, A., Gazendam,, R. P., Tool,, A. T., van Houdt,, M., Jansen,, M. H., van Hamme,, J. L., … Kuijpers,, T. W. (2013). Invasive fungal infection and impaired neutrophil killing in human CARD9 deficiency. Blood, 121(13), 2385–2392. https://doi.org/10.1182/blood-2012-08-450551
Esser,, C., & Rannug,, A. (2015). The aryl hydrocarbon receptor in barrier organ physiology, immunology, and toxicology. Pharmacological Reviews, 67(2), 259–279. https://doi.org/10.1124/pr.114.009001
Fan,, D., Coughlin,, L. A., Neubauer,, M. M., Kim,, J., Kim,, M. S., Zhan,, X., … Koh,, A. Y. (2015). Activation of HIF‐1alpha and LL‐37 by commensal bacteria inhibits Candida albicans colonization. Nature Medicine, 21(7), 808–814. https://doi.org/10.1038/nm.3871
Fitzsimmons,, N., & Berry,, D. R. (1994). Inhibition of Candida albicans by Lactobacillus acidophilus: Evidence for the involvement of a peroxidase system. Microbios, 80(323), 125–133.
Foster,, K. R., Schluter,, J., Coyte,, K. Z., & Rakoff‐Nahoum,, S. (2017). The evolution of the host microbiome as an ecosystem on a leash. Nature, 548(7665), 43–51. https://doi.org/10.1038/nature23292
Franke,, A., McGovern,, D. P., Barrett,, J. C., Wang,, K., Radford‐Smith,, G. L., Ahmad,, T., … Parkes,, M. (2010). Genome‐wide meta‐analysis increases to 71 the number of confirmed Crohn`s disease susceptibility loci. Nature Genetics, 42(12), 1118–1125. https://doi.org/10.1038/ng.717
Friedman,, J., Higgins,, L. M., & Gore,, J. (2017). Community structure follows simple assembly rules in microbial microcosms. Nature Ecology and Evolution, 1(5), 109. https://doi.org/10.1038/s41559-017-0109
Fujimura,, K. E., Sitarik,, A. R., Havstad,, S., Lin,, D. L., Levan,, S., Fadrosh,, D., … Lynch,, S. V. (2016). Neonatal gut microbiota associates with childhood multisensitized atopy and T cell differentiation. Nature Medicine, 22(10), 1187–1191. https://doi.org/10.1038/nm.4176
Fung,, T. C., Olson,, C. A., & Hsiao,, E. Y. (2017). Interactions between the microbiota, immune and nervous systems in health and disease. Nature Neuroscience, 20(2), 145–155. https://doi.org/10.1038/nn.4476
Furue,, M., Takahara,, M., Nakahara,, T., & Uchi,, H. (2014). Role of AhR/ARNT system in skin homeostasis. Archives of Dermatological Research, 306(9), 769–779. https://doi.org/10.1007/s00403-014-1481-7
Gaffen,, S. L., Jain,, R., Garg,, A. V., & Cua,, D. J. (2014). The IL‐23‐IL‐17 immune axis: From mechanisms to therapeutic testing. Nature Reviews. Immunology, 14(9), 585–600. https://doi.org/10.1038/nri3707
Gaitanis,, G., Magiatis,, P., Stathopoulou,, K., Bassukas,, I. D., Alexopoulos,, E. C., Velegraki,, A., & Skaltsounis,, A. L. (2008). AhR ligands, malassezin, and indolo[3,2‐b]carbazole are selectively produced by Malassezia furfur strains isolated from seborrheic dermatitis. Journal of Investigative Dermatology, 128(7), 1620–1625. https://doi.org/10.1038/sj.jid.5701252
Gale,, D., & Sandoval,, B. (1957). Response of mice to the inoculations of both Candida albicans and Escherichia coli. I. The enhancement phenomenon. Journal of Bacteriology, 73(5), 616–624.
Ganal,, S. C., Sanos,, S. L., Kallfass,, C., Oberle,, K., Johner,, C., Kirschning,, C., … Diefenbach,, A. (2012). Priming of natural killer cells by nonmucosal mononuclear phagocytes requires instructive signals from commensal microbiota. Immunity, 37(1), 171–186. https://doi.org/10.1016/j.immuni.2012.05.020
George,, M. P., Kannass,, M., Huang,, L., Sciurba,, F. C., & Morris,, A. (2009). Respiratory symptoms and airway obstruction in HIV‐infected subjects in the HAART era. PLoS One, 4(7), e6328. https://doi.org/10.1371/journal.pone.0006328
Gevers,, D., Kugathasan,, S., Denson,, L. A., Vazquez‐Baeza,, Y., Van Treuren,, W., Ren,, B., … Xavier,, R. J. (2014). The treatment‐naive microbiome in new‐onset Crohn`s disease. Cell Host %26 Microbe, 15(3), 382–392. https://doi.org/10.1016/j.chom.2014.02.005
Ghannoum,, M. A., Jurevic,, R. J., Mukherjee,, P. K., Cui,, F., Sikaroodi,, M., Naqvi,, A., & Gillevet,, P. M. (2010). Characterization of the oral fungal microbiome (mycobiome) in healthy individuals. PLoS Pathogens, 6(1), e1000713. https://doi.org/10.1371/journal.ppat.1000713
Gibson,, J., Sood,, A., & Hogan,, D. A. (2009). Pseudomonas aeruginosa–Candida albicans interactions: Localization and fungal toxicity of a phenazine derivative. Applied and Environmental Microbiology, 75(2), 504–513. https://doi.org/10.1128/AEM.01037-08
Glocker,, E. O., Hennigs,, A., Nabavi,, M., Schaffer,, A. A., Woellner,, C., Salzer,, U., … Grimbacher,, B. (2009). A homozygous CARD9 mutation in a family with susceptibility to fungal infections. New England Journal of Medicine, 361(18), 1727–1735. https://doi.org/10.1056/NEJMoa0810719
Goodrich,, J. K., Davenport,, E. R., Waters,, J. L., Clark,, A. G., & Ley,, R. E. (2016). Cross‐species comparisons of host genetic associations with the microbiome. Science, 352(6285), 532–535. https://doi.org/10.1126/science.aad9379
Gore,, J., Youk,, H., & van Oudenaarden,, A. (2009). Snowdrift game dynamics and facultative cheating in yeast. Nature, 459(7244), 253–256. https://doi.org/10.1038/nature07921
Gour,, N., Lajoie,, S., Smole,, U., White,, M., Hu,, D., Goddard,, P., … Wills‐Karp,, M. (2018). Dysregulated invertebrate tropomyosin‐dectin‐1 interaction confers susceptibility to allergic diseases. Science Immunology, 3(20), eaam9841. https://doi.org/10.1126/sciimmunol.aam9841
Graham,, C. E., Cruz,, M. R., Garsin,, D. A., & Lorenz,, M. C. (2017). Enterococcus faecalis bacteriocin EntV inhibits hyphal morphogenesis, biofilm formation, and virulence of Candida albicans. Proceedings of the National Academy of Sciences of the United States of America, 114(17), 4507–4512. https://doi.org/10.1073/pnas.1620432114
Hall,, R. A., & Noverr,, M. C. (2017). Fungal interactions with the human host: Exploring the spectrum of symbiosis. Current Opinion in Microbiology, 40, 58–64. https://doi.org/10.1016/j.mib.2017.10.020
Hallen‐Adams,, H. E., Kachman,, S. D., Kim,, J., Legge,, R. M., & Martínez,, I. (2015). Fungi inhabiting the healthy human gastrointestinal tract: A diverse and dynamic community. Fungal Ecology, 15, 9–17. https://doi.org/10.1016/j.funeco.2015.01.006
Halwachs,, B., Madhusudhan,, N., Krause,, R., Nilsson,, R. H., Moissl‐Eichinger,, C., Hogenauer,, C., … Gorkiewicz,, G. (2017). Critical issues in mycobiota analysis. Frontiers in Microbiology, 8, 180. https://doi.org/10.3389/fmicb.2017.00180
Hamad,, I., Ranque,, S., Azhar,, E. I., Yasir,, M., Jiman‐Fatani,, A. A., Tissot‐Dupont,, H., … Bittar,, F. (2017). Culturomics and amplicon‐based metagenomic approaches for the study of fungal population in human gut microbiota. Scientific Reports, 7(1), 16788. https://doi.org/10.1038/s41598-017-17132-4
Hamad,, I., Raoult,, D., & Bittar,, F. (2016). Repertory of eukaryotes (eukaryome) in the human gastrointestinal tract: Taxonomy and detection methods. Parasite Immunology, 38(1), 12–36. https://doi.org/10.1111/pim.12284
Harada,, K., Saito,, M., Sugita,, T., & Tsuboi,, R. (2015). Malassezia species and their associated skin diseases. Journal of Dermatology, 42(3), 250–257. https://doi.org/10.1111/1346-8138.12700
Hebart,, H., Bollinger,, C., Fisch,, P., Sarfati,, J., Meisner,, C., Baur,, M., … Einsele,, H. (2002). Analysis of T‐cell responses to Aspergillus fumigatus antigens in healthy individuals and patients with hematologic malignancies. Blood, 100(13), 4521–4528. https://doi.org/10.1182/blood-2002-01-0265
Heisel,, T., Montassier,, E., Johnson,, A., Al‐Ghalith,, G., Lin,, Y. W., Wei,, L. N., … Gale,, C. A. (2017). High‐fat diet changes fungal microbiomes and interkingdom relationships in the murine gut. mSphere, 2(5), e00351‐17. https://doi.org/10.1128/mSphere.00351-17
Hibbett,, D. S., Binder,, M., Bischoff,, J. F., Blackwell,, M., Cannon,, P. F., Eriksson,, O. E., … Zhang,, N. (2007). A higher‐level phylogenetic classification of the fungi. Mycological Research, 111(Pt. 5), 509–547. https://doi.org/10.1016/j.mycres.2007.03.004
Hirota,, K., Yoshitomi,, H., Hashimoto,, M., Maeda,, S., Teradaira,, S., Sugimoto,, N., … Sakaguchi,, S. (2007). Preferential recruitment of CCR6‐expressing Th17 cells to inflamed joints via CCL20 in rheumatoid arthritis and its animal model. Journal of Experimental Medicine, 204(12), 2803–2812. https://doi.org/10.1084/jem.20071397
Hoarau,, G., Mukherjee,, P. K., Gower‐Rousseau,, C., Hager,, C., Chandra,, J., Retuerto,, M. A., … Ghannoum,, M. A. (2016). Bacteriome and mycobiome interactions underscore microbial dysbiosis in familial Crohn`s disease. mBio, 7(5), e01250‐16. https://doi.org/10.1128/mBio.01250-16
Hoffmann,, C., Dollive,, S., Grunberg,, S., Chen,, J., Li,, H., Wu,, G. D., … Bushman,, F. D. (2013). Archaea and fungi of the human gut microbiome: Correlations with diet and bacterial residents. PLoS One, 8(6), e66019. https://doi.org/10.1371/journal.pone.0066019
Hogan,, D. A., & Kolter,, R. (2002). Pseudomonas–Candida interactions: An ecological role for virulence factors. Science, 296(5576), 2229–2232. https://doi.org/10.1126/science.1070784
Hogan,, D. A., Vik,, A., & Kolter,, R. (2004). A Pseudomonas aeruginosa quorum‐sensing molecule influences Candida albicans morphology. Molecular Microbiology, 54(5), 1212–1223. https://doi.org/10.1111/j.1365-2958.2004.04349.x
Huffnagle,, G. B. (2010). The microbiota and allergies/asthma. PLoS Pathogens, 6(5), e1000549. https://doi.org/10.1371/journal.ppat.1000549
Human Microbiome Project Consortium. (2012). A framework for human microbiome research. Nature, 486(7402), 215–221. https://doi.org/10.1038/nature11209
Huseyin,, C. E., Rubio,, R. C., O`Sullivan,, O., Cotter,, P. D., & Scanlan,, P. D. (2017). The fungal frontier: A comparative analysis of methods used in the study of the human gut mycobiome. Frontiers in Microbiology, 8, 1432. https://doi.org/10.3389/fmicb.2017.01432
Iliev,, I. D., Funari,, V. A., Taylor,, K. D., Nguyen,, Q., Reyes,, C. N., Strom,, S. P., … Underhill,, D. M. (2012). Interactions between commensal fungi and the C‐type lectin receptor Dectin‐1 influence colitis. Science, 336(6086), 1314–1317. https://doi.org/10.1126/science.1221789
Iliev,, I. D., & Leonardi,, I. (2017). Fungal dysbiosis: Immunity and interactions at mucosal barriers. Nature Reviews. Immunology, 17(10), 635–646. https://doi.org/10.1038/nri.2017.55
Iovene,, M. R., Bombace,, F., Maresca,, R., Sapone,, A., Iardino,, P., Picardi,, A., … Bravaccio,, C. (2017). Intestinal dysbiosis and yeast isolation in stool of subjects with autism spectrum disorders. Mycopathologia, 182(3–4), 349–363. https://doi.org/10.1007/s11046-016-0068-6
Ivanov,, I. I., Atarashi,, K., Manel,, N., Brodie,, E. L., Shima,, T., Karaoz,, U., … Littman,, D. R. (2009). Induction of intestinal Th17 cells by segmented filamentous bacteria. Cell, 139(3), 485–498. https://doi.org/10.1016/j.cell.2009.09.033
Jaeger,, M., Carvalho,, A., Cunha,, C., Plantinga,, T. S., van de Veerdonk,, F., Puccetti,, M., … Netea,, M. G. (2016). Association of a variable number tandem repeat in the NLRP3 gene in women with susceptibility to RVVC. European Journal of Clinical Microbiology %26 Infectious Diseases, 35(5), 797–801. https://doi.org/10.1007/s10096-016-2600-5
Jarchum,, I., & Pamer,, E. G. (2011). Regulation of innate and adaptive immunity by the commensal microbiota. Current Opinion in Immunology, 23(3), 353–360. https://doi.org/10.1016/j.coi.2011.03.001
Jawhara,, S., Thuru,, X., Standaert‐Vitse,, A., Jouault,, T., Mordon,, S., Sendid,, B., … Poulain,, D. (2008). Colonization of mice by Candida albicans is promoted by chemically induced colitis and augments inflammatory responses through galectin‐3. Journal of Infectious Diseases, 197(7), 972–980. https://doi.org/10.1086/528990
Jiang,, T. T., Shao,, T. Y., Ang,, W. X. G., Kinder,, J. M., Turner,, L. H., Pham,, G., … Way,, S. S. (2017). Commensal fungi recapitulate the protective benefits of intestinal bacteria. Cell Host %26 Microbe, 22(6), 809–816 e804. https://doi.org/10.1016/j.chom.2017.10.013
Johnson,, N. C., Graham,, J.‐H., & Smith,, F. A. (1997). Functioning of mycorrhizal associations along the mutualism–parasitism continuum. New Phytologist, 135(4), 575–585. https://doi.org/10.1046/j.1469-8137.1997.00729.x
Jolink,, H., Meijssen,, I. C., Hagedoorn,, R. S., Arentshorst,, M., Drijfhout,, J. W., Mulder,, A., … Heemskerk,, M. H. (2013). Characterization of the T‐cell‐mediated immune response against the Aspergillus fumigatus proteins Crf1 and catalase 1 in healthy individuals. Journal of Infectious Diseases, 208(5), 847–856. https://doi.org/10.1093/infdis/jit237
Kashem,, S. W., Igyarto,, B. Z., Gerami‐Nejad,, M., Kumamoto,, Y., Mohammed,, J. A., Jarrett,, E., … Kaplan,, D. H. (2015). Candida albicans morphology and dendritic cell subsets determine T helper cell differentiation. Immunity, 42(2), 356–366. https://doi.org/10.1016/j.immuni.2015.01.008
Khosravi,, A., Yanez,, A., Price,, J. G., Chow,, A., Merad,, M., Goodridge,, H. S., & Mazmanian,, S. K. (2014). Gut microbiota promote hematopoiesis to control bacterial infection. Cell Host %26 Microbe, 15(3), 374–381. https://doi.org/10.1016/j.chom.2014.02.006
Kim,, H. J., Li,, H., Collins,, J. J., & Ingber,, D. E. (2016). Contributions of microbiome and mechanical deformation to intestinal bacterial overgrowth and inflammation in a human gut‐on‐a‐chip. Proceedings of the National Academy of Sciences of the United States of America, 113(1), E7–E15. https://doi.org/10.1073/pnas.1522193112
Kim,, S. H., Clark,, S. T., Surendra,, A., Copeland,, J. K., Wang,, P. W., Ammar,, R., … Cowen,, L. E. (2015). Global analysis of the fungal microbiome in cystic fibrosis patients reveals loss of function of the transcriptional repressor Nrg1 as a mechanism of pathogen adaptation. PLoS Pathogens, 11(11), e1005308. https://doi.org/10.1371/journal.ppat.1005308
Kim,, Y. G., Udayanga,, K. G., Totsuka,, N., Weinberg,, J. B., Nunez,, G., & Shibuya,, A. (2014). Gut dysbiosis promotes M2 macrophage polarization and allergic airway inflammation via fungi‐induced PGE(2). Cell Host %26 Microbe, 15(1), 95–102. https://doi.org/10.1016/j.chom.2013.12.010
Kisand,, K., Boe Wolff,, A. S., Podkrajsek,, K. T., Tserel,, L., Link,, M., Kisand,, K. V., … Meager,, A. (2010). Chronic mucocutaneous candidiasis in APECED or thymoma patients correlates with autoimmunity to Th17‐associated cytokines. Journal of Experimental Medicine, 207(2), 299–308. https://doi.org/10.1084/jem.20091669
Kiss,, L. (2012). Limits of nuclear ribosomal DNA internal transcribed spacer (ITS) sequences as species barcodes for fungi. Proceedings of the National Academy of Sciences of the United States of America, 109(27), E1811 ; author reply E1812. https://doi.org/10.1073/pnas.1207143109
Koenig,, J. E., Spor,, A., Scalfone,, N., Fricker,, A. D., Stombaugh,, J., Knight,, R., … Ley,, R. E. (2011). Succession of microbial consortia in the developing infant gut microbiome. Proceedings of the National Academy of Sciences of the United States of America, 108(Suppl. 1), 4578–4585. https://doi.org/10.1073/pnas.1000081107
Kurilshikov,, A., Wijmenga,, C., Fu,, J., & Zhernakova,, A. (2017). Host genetics and gut microbiome: Challenges and perspectives. Trends in Immunology, 38(9), 633–647. https://doi.org/10.1016/j.it.2017.06.003
Lamas,, B., Richard,, M. L., Leducq,, V., Pham,, H. P., Michel,, M. L., Da Costa,, G., … Sokol,, H. (2016). CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nature Medicine, 22(6), 598–605. https://doi.org/10.1038/nm.4102
Leegaard,, M. (1984). The incidence of Candida albicans in the vagina of “healthy young women”. How often do they have symptoms? Possible etiological factors. Acta Obstetricia et Gynecologica Scandinavica, 63(1), 85–89.
Leonardi,, I., Li,, X., Semon,, A., Li,, D., Doron,, I., Putzel,, G., … Iliev,, I. D. (2018). CX3CR1(+) mononuclear phagocytes control immunity to intestinal fungi. Science, 359(6372), 232–236. https://doi.org/10.1126/science.aao1503
Lev‐Sagie,, A., Prus,, D., Linhares,, I. M., Lavy,, Y., Ledger,, W. J., & Witkin,, S. S. (2009). Polymorphism in a gene coding for the inflammasome component NALP3 and recurrent vulvovaginal candidiasis in women with vulvar vestibulitis syndrome. American Journal of Obstetrics and Gynecology, 200(3), 303 e301–306. https://doi.org/10.1016/j.ajog.2008.10.039
Lewis,, J. D., Chen,, E. Z., Baldassano,, R. N., Otley,, A. R., Griffiths,, A. M., Lee,, D., … Bushman,, F. D. (2015). Inflammation, antibiotics, and diet as environmental stressors of the gut microbiome in pediatric Crohn`s disease. Cell Host %26 Microbe, 18(4), 489–500. https://doi.org/10.1016/j.chom.2015.09.008
Ley,, R. E., Backhed,, F., Turnbaugh,, P., Lozupone,, C. A., Knight,, R. D., & Gordon,, J. I. (2005). Obesity alters gut microbial ecology. Proceedings of the National Academy of Sciences of the United States of America, 102(31), 11070–11075. https://doi.org/10.1073/pnas.0504978102
Li,, Q., Wang,, C., Tang,, C., He,, Q., Li,, N., & Li,, J. (2014). Dysbiosis of gut fungal microbiota is associated with mucosal inflammation in Crohn`s disease. Journal of Clinical Gastroenterology, 48(6), 513–523. https://doi.org/10.1097/MCG.0000000000000035
Liguori,, G., Lamas,, B., Richard,, M. L., Brandi,, G., da Costa,, G., Hoffmann,, T. W., … Sokol,, H. (2016). Fungal dysbiosis in mucosa‐associated microbiota of Crohn`s disease patients. Journal of Crohn`s %26 Colitis, 10(3), 296–305. https://doi.org/10.1093/ecco-jcc/jjv209
Limon,, J. J., Skalski,, J. H., & Underhill,, D. M. (2017). Commensal fungi in health and disease. Cell Host %26 Microbe, 22(2), 156–165. https://doi.org/10.1016/j.chom.2017.07.002
Liu,, C. M., Kachur,, S., Dwan,, M. G., Abraham,, A. G., Aziz,, M., Hsueh,, P. R., … Price,, L. B. (2012). FungiQuant: A broad‐coverage fungal quantitative real‐time PCR assay. BMC Microbiology, 12, 255. https://doi.org/10.1186/1471-2180-12-255
Liu,, L., Okada,, S., Kong,, X. F., Kreins,, A. Y., Cypowyj,, S., Abhyankar,, A., … Casanova,, J. L. (2011). Gain‐of‐function human STAT1 mutations impair IL‐17 immunity and underlie chronic mucocutaneous candidiasis. Journal of Experimental Medicine, 208(8), 1635–1648. https://doi.org/10.1084/jem.20110958
Lozupone,, C. A., Stombaugh,, J. I., Gordon,, J. I., Jansson,, J. K., & Knight,, R. (2012). Diversity, stability and resilience of the human gut microbiota. Nature, 489(7415), 220–230. https://doi.org/10.1038/nature11550
Ma,, C. S., Chew,, G. Y., Simpson,, N., Priyadarshi,, A., Wong,, M., Grimbacher,, B., … Cook,, M. C. (2008). Deficiency of Th17 cells in hyper IgE syndrome due to mutations in STAT3. Journal of Experimental Medicine, 205(7), 1551–1557. https://doi.org/10.1084/jem.20080218
Magiatis,, P., Pappas,, P., Gaitanis,, G., Mexia,, N., Melliou,, E., Galanou,, M., … Bassukas,, I. D. (2013). Malassezia yeasts produce a collection of exceptionally potent activators of the Ah (dioxin) receptor detected in diseased human skin. Journal of Investigative Dermatology, 133(8), 2023–2030. https://doi.org/10.1038/jid.2013.92
Mar Rodriguez,, M., Perez,, D., Javier Chaves,, F., Esteve,, E., Marin‐Garcia,, P., Xifra,, G., … Fernandez Real,, J. M. (2015). Obesity changes the human gut mycobiome. Scientific Reports, 5, 14600. https://doi.org/10.1038/srep14600
Marchesi,, J. R., & Ravel,, J. (2015). The vocabulary of microbiome research: A proposal. Microbiome, 3, 31. https://doi.org/10.1186/s40168-015-0094-5
Markle,, J. G., Frank,, D. N., Mortin‐Toth,, S., Robertson,, C. E., Feazel,, L. M., Rolle‐Kampczyk,, U., … Danska,, J. S. (2013). Sex differences in the gut microbiome drive hormone‐dependent regulation of autoimmunity. Science, 339(6123), 1084–1088. https://doi.org/10.1126/science.1233521
Martinez,, I., Stegen,, J. C., Maldonado‐Gomez,, M. X., Eren,, A. M., Siba,, P. M., Greenhill,, A. R., & Walter,, J. (2015). The gut microbiota of rural Papua New Guineans: Composition, diversity patterns, and ecological processes. Cell Reports, 11(4), 527–538. https://doi.org/10.1016/j.celrep.2015.03.049
Mastrofrancesco,, A., Ottaviani,, M., Aspite,, N., Cardinali,, G., Izzo,, E., Graupe,, K., … Picardo,, M. (2010). Azelaic acid modulates the inflammatory response in normal human keratinocytes through PPARgamma activation. Experimental Dermatology, 19(9), 813–820. https://doi.org/10.1111/j.1600-0625.2010.01107.x
Mazmanian,, S. K., Round,, J. L., & Kasper,, D. L. (2008). A microbial symbiosis factor prevents intestinal inflammatory disease. Nature, 453(7195), 620–625. https://doi.org/10.1038/nature07008
McGovern,, D. P., Gardet,, A., Torkvist,, L., Goyette,, P., Essers,, J., Taylor,, K. D., … Seielstad,, M. (2010). Genome‐wide association identifies multiple ulcerative colitis susceptibility loci. Nature Genetics, 42(4), 332–337. https://doi.org/10.1038/ng.549
Mexia,, N., Gaitanis,, G., Velegraki,, A., Soshilov,, A., Denison,, M. S., & Magiatis,, P. (2015). Pityriazepin and other potent AhR ligands isolated from Malassezia furfur yeast. Archives of Biochemistry and Biophysics, 571, 16–20. https://doi.org/10.1016/j.abb.2015.02.023
Mitroulis,, I., Ruppova,, K., Wang,, B., Chen,, L. S., Grzybek,, M., Grinenko,, T., … Chavakis,, T. (2018). Modulation of myelopoiesis progenitors is an integral component of trained immunity. Cell, 172(1–2), 147–161 e112. https://doi.org/10.1016/j.cell.2017.11.034
Morris,, A., Sciurba,, F. C., Lebedeva,, I. P., Githaiga,, A., Elliott,, W. M., Hogg,, J. C., … Norris,, K. A. (2004). Association of chronic obstructive pulmonary disease severity and Pneumocystis colonization. American Journal of Respiratory and Critical Care Medicine, 170(4), 408–413. https://doi.org/10.1164/rccm.200401-094OC
Mortensen,, P. B., & Clausen,, M. R. (1996). Short‐chain fatty acids in the human colon: Relation to gastrointestinal health and disease. Scandinavian Journal of Gastroenterology. Supplement, 216, 132–148.
Mowat,, E., Rajendran,, R., Williams,, C., McCulloch,, E., Jones,, B., Lang,, S., & Ramage,, G. (2010). Pseudomonas aeruginosa and their small diffusible extracellular molecules inhibit Aspergillus fumigatus biofilm formation. FEMS Microbiology Letters, 313(2), 96–102. https://doi.org/10.1111/j.1574-6968.2010.02130.x
Murdock,, B. J., Shreiner,, A. B., McDonald,, R. A., Osterholzer,, J. J., White,, E. S., Toews,, G. B., & Huffnagle,, G. B. (2011). Coevolution of TH1, TH2, and TH17 responses during repeated pulmonary exposure to Aspergillus fumigatus conidia. Infection and Immunity, 79(1), 125–135. https://doi.org/10.1128/IAI.00508-10
Nagata,, R., Nagano,, H., Ogishima,, D., Nakamura,, Y., Hiruma,, M., & Sugita,, T. (2012). Transmission of the major skin microbiota, Malassezia, from mother to neonate. Pediatrics International, 54(3), 350–355. https://doi.org/10.1111/j.1442-200X.2012.03563.x
Naik,, S., Bouladoux,, N., Linehan,, J. L., Han,, S. J., Harrison,, O. J., Wilhelm,, C., … Belkaid,, Y. (2015). Commensal‐dendritic‐cell interaction specifies a unique protective skin immune signature. Nature, 520(7545), 104–108. https://doi.org/10.1038/nature14052
Nash,, A. K., Auchtung,, T. A., Wong,, M. C., Smith,, D. P., Gesell,, J. R., Ross,, M. C., … Petrosino,, J. F. (2017). The gut mycobiome of the Human Microbiome Project healthy cohort. Microbiome, 5(1), 153. https://doi.org/10.1186/s40168-017-0373-4
Nedovic,, B., Posteraro,, B., Leoncini,, E., Ruggeri,, A., Amore,, R., Sanguinetti,, M., … Boccia,, S. (2014). Mannose‐binding lectin codon 54 gene polymorphism and vulvovaginal candidiasis: A systematic review and meta‐analysis. BioMed Research International, 2014, 1–7. https://doi.org/10.1155/2014/738298
Neely,, A. N., Law,, E. J., & Holder,, I. A. (1986). Increased susceptibility to lethal Candida infections in burned mice preinfected with Pseudomonas aeruginosa or pretreated with proteolytic enzymes. Infection and Immunity, 52(1), 200–204.
Netea,, M. G., Joosten,, L. A., Li,, Y., Kumar,, V., Oosting,, M., Smeekens,, S., … Wijmenga,, C. (2016). Understanding human immune function using the resources from the Human Functional Genomics Project. Nature Medicine, 22(8), 831–833. https://doi.org/10.1038/nm.4140
Netea,, M. G., Quintin,, J., & van der Meer,, J. W. (2011). Trained immunity: A memory for innate host defense. Cell Host %26 Microbe, 9(5), 355–361. https://doi.org/10.1016/j.chom.2011.04.006
Noverr,, M. C., Noggle,, R. M., Toews,, G. B., & Huffnagle,, G. B. (2004). Role of antibiotics and fungal microbiota in driving pulmonary allergic responses. Infection and Immunity, 72(9), 4996–5003. https://doi.org/10.1128/IAI.72.9.4996-5003.2004
Noverr,, M. C., Toews,, G. B., & Huffnagle,, G. B. (2002). Production of prostaglandins and leukotrienes by pathogenic fungi. Infection and Immunity, 70(1), 400–402.
O`Donnell,, K., & Cigelnik,, E. (1997). Two divergent intragenomic rDNA ITS2 types within a monophyletic lineage of the fungus Fusarium are nonorthologous. Molecular Phylogenetics and Evolution, 7(1), 103–116. https://doi.org/10.1006/mpev.1996.0376
Oh,, J., Freeman,, A. F., Program,, N. C. S., Park,, M., Sokolic,, R., Candotti,, F., … Kong,, H. H. (2013). The altered landscape of the human skin microbiome in patients with primary immunodeficiencies. Genome Research, 23(12), 2103–2114. https://doi.org/10.1101/gr.159467.113
Okada,, S., Markle,, J. G., Deenick,, E. K., Mele,, F., Averbuch,, D., Lagos,, M., … Casanova,, J. L. (2015). IMMUNODEFICIENCIES. Impairment of immunity to Candida and Mycobacterium in humans with bi‐allelic RORC mutations. Science, 349(6248), 606–613. https://doi.org/10.1126/science.aaa4282
Ott,, S. J., Kuhbacher,, T., Musfeldt,, M., Rosenstiel,, P., Hellmig,, S., Rehman,, A., … Schreiber,, S. (2008). Fungi and inflammatory bowel diseases: Alterations of composition and diversity. Scandinavian Journal of Gastroenterology, 43(7), 831–841. https://doi.org/10.1080/00365520801935434
Ouederni,, M., Sanal,, O., Ikinciogullari,, A., Tezcan,, I., Dogu,, F., Sologuren,, I., … Rodriguez‐Gallego,, C. (2014). Clinical features of candidiasis in patients with inherited interleukin 12 receptor beta1 deficiency. Clinical Infectious Diseases, 58(2), 204–213. https://doi.org/10.1093/cid/cit722
Patel,, D. D., & Kuchroo,, V. K. (2015). Th17 cell pathway in human immunity: Lessons from genetics and therapeutic interventions. Immunity, 43(6), 1040–1051. https://doi.org/10.1016/j.immuni.2015.12.003
Peleg,, A. Y., Hogan,, D. A., & Mylonakis,, E. (2010). Medically important bacterial–fungal interactions. Nature Reviews. Microbiology, 8(5), 340–349. https://doi.org/10.1038/nrmicro2313
Plantinga,, T. S., van der Velden,, W. J., Ferwerda,, B., van Spriel,, A. B., Adema,, G., Feuth,, T., … Netea,, M. G. (2009). Early stop polymorphism in human DECTIN‐1 is associated with increased Candida colonization in hematopoietic stem cell transplant recipients. Clinical Infectious Diseases, 49(5), 724–732. https://doi.org/10.1086/604714
Prando,, C., Samarina,, A., Bustamante,, J., Boisson‐Dupuis,, S., Cobat,, A., Picard,, C., … Casanova,, J. L. (2013). Inherited IL‐12p40 deficiency: Genetic, immunologic, and clinical features of 49 patients from 30 kindreds. Medicine (Baltimore), 92(2), 109–122. https://doi.org/10.1097/MD.0b013e31828a01f9
Prohic,, A., Jovovic Sadikovic,, T., Krupalija‐Fazlic,, M., & Kuskunovic‐Vlahovljak,, S. (2016). Malassezia species in healthy skin and in dermatological conditions. International Journal of Dermatology, 55(5), 494–504. https://doi.org/10.1111/ijd.13116
Pruesse,, E., Quast,, C., Knittel,, K., Fuchs,, B. M., Ludwig,, W., Peplies,, J., & Glockner,, F. O. (2007). SILVA: A comprehensive online resource for quality checked and aligned ribosomal RNA sequence data compatible with ARB. Nucleic Acids Research, 35(21), 7188–7196. https://doi.org/10.1093/nar/gkm864
Puel,, A., Cypowyj,, S., Bustamante,, J., Wright,, J. F., Liu,, L., Lim,, H. K., … Casanova,, J. L. (2011). Chronic mucocutaneous candidiasis in humans with inborn errors of interleukin‐17 immunity. Science, 332(6025), 65–68. https://doi.org/10.1126/science.1200439
Puel,, A., Doffinger,, R., Natividad,, A., Chrabieh,, M., Barcenas‐Morales,, G., Picard,, C., … Casanova,, J. L. (2010). Autoantibodies against IL‐17A, IL‐17F, and IL‐22 in patients with chronic mucocutaneous candidiasis and autoimmune polyendocrine syndrome type I. Journal of Experimental Medicine, 207(2), 291–297. https://doi.org/10.1084/jem.20091983
Qin,, J., Li,, R., Raes,, J., Arumugam,, M., Burgdorf,, K. S., Manichanh,, C., … Wang,, J. (2010). A human gut microbial gene catalogue established by metagenomic sequencing. Nature, 464(7285), 59–65. https://doi.org/10.1038/nature08821
Qiu,, X., Ma,, J., Jiao,, C., Mao,, X., Zhao,, X., Lu,, M., … Zhang,, H. (2017). Alterations in the mucosa‐associated fungal microbiota in patients with ulcerative colitis. Oncotarget, 8(64), 107577–107588. https://doi.org/10.18632/oncotarget.22534
Qiu,, X., Zhang,, F., Yang,, X., Wu,, N., Jiang,, W., Li,, X., … Liu,, Y. (2015). Changes in the composition of intestinal fungi and their role in mice with dextran sulfate sodium‐induced colitis. Scientific Reports, 5, 10416. https://doi.org/10.1038/srep10416
Quintana,, F. J., & Sherr,, D. H. (2013). Aryl hydrocarbon receptor control of adaptive immunity. Pharmacological Reviews, 65(4), 1148–1161. https://doi.org/10.1124/pr.113.007823
Quintin,, J., Saeed,, S., Martens,, J. H. A., Giamarellos‐Bourboulis,, E. J., Ifrim,, D. C., Logie,, C., … Netea,, M. G. (2012). Candida albicans infection affords protection against reinfection via functional reprogramming of monocytes. Cell Host %26 Microbe, 12(2), 223–232. https://doi.org/10.1016/j.chom.2012.06.006
Ramirez Granillo,, A., Canales,, M. G., Espindola,, M. E., Martinez Rivera,, M. A., de Lucio,, V. M., & Tovar,, A. V. (2015). Antibiosis interaction of Staphylococccus aureus on Aspergillus fumigatus assessed in vitro by mixed biofilm formation. BMC Microbiology, 15, 33. https://doi.org/10.1186/s12866-015-0363-2
Ravel,, J., Gajer,, P., Abdo,, Z., Schneider,, G. M., Koenig,, S. S., McCulle,, S. L., … Forney,, L. J. (2011). Vaginal microbiome of reproductive‐age women. Proceedings of the National Academy of Sciences of the United States of America, 108(Suppl. 1), 4680–4687. https://doi.org/10.1073/pnas.1002611107
Rick,, E. M., Woolnough,, K., Pashley,, C. H., & Wardlaw,, A. J. (2016). Allergic fungal airway disease. Journal of Investigational Allergology %26 Clinical Immunology, 26(6), 344–354. https://doi.org/10.18176/jiaci.0122
Ridaura,, V. K., Faith,, J. J., Rey,, F. E., Cheng,, J., Duncan,, A. E., Kau,, A. L., … Gordon,, J. I. (2013). Gut microbiota from twins discordant for obesity modulate metabolism in mice. Science, 341(6150), 1241214. https://doi.org/10.1126/science.1241214
Rivas,, M. A., Beaudoin,, M., Gardet,, A., Stevens,, C., Sharma,, Y., Zhang,, C. K., … Daly,, M. J. (2011). Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nature Genetics, 43(11), 1066–1073. https://doi.org/10.1038/ng.952
Rizzetto,, L., Buschow,, S. I., Beltrame,, L., Figdor,, C. G., Schierer,, S., Schuler,, G., & Cavalieri,, D. (2012). The modular nature of dendritic cell responses to commensal and pathogenic fungi. PLoS One, 7(8), e42430. https://doi.org/10.1371/journal.pone.0042430
Rizzetto,, L., De Filippo,, C., & Cavalieri,, D. (2014). Richness and diversity of mammalian fungal communities shape innate and adaptive immunity in health and disease. European Journal of Immunology, 44(11), 3166–3181. https://doi.org/10.1002/eji.201344403
Rizzetto,, L., Giovannini,, G., Bromley,, M., Bowyer,, P., Romani,, L., & Cavalieri,, D. (2013). Strain dependent variation of immune responses to A. fumigatus: Definition of pathogenic species. PLoS One, 8(2), e56651. https://doi.org/10.1371/journal.pone.0056651
Rizzetto,, L., Ifrim,, D. C., Moretti,, S., Tocci,, N., Cheng,, S. C., Quintin,, J., … Cavalieri,, D. (2016). Fungal chitin induces trained immunity in human monocytes during cross‐talk of the host with Saccharomyces cerevisiae. Journal of Biological Chemistry, 291(15), 7961–7972. https://doi.org/10.1074/jbc.M115.699645
Rizzetto,, L., Kuka,, M., De Filippo,, C., Cambi,, A., Netea,, M. G., Beltrame,, L., … Cavalieri,, D. (2010). Differential IL‐17 production and mannan recognition contribute to fungal pathogenicity and commensalism. Journal of Immunology, 184(8), 4258–4268. https://doi.org/10.4049/jimmunol.0902972
Sampson,, T. R., Debelius,, J. W., Thron,, T., Janssen,, S., Shastri,, G. G., Ilhan,, Z. E., … Mazmanian,, S. K. (2016). Gut microbiota regulate motor deficits and neuroinflammation in a model of Parkinson`s disease. Cell, 167(6), 1469–1480 e1412. https://doi.org/10.1016/j.cell.2016.11.018
Scher,, J. U., Sczesnak,, A., Longman,, R. S., Segata,, N., Ubeda,, C., Bielski,, C., … Littman,, D. R. (2013). Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis. eLife, 2, e01202. https://doi.org/10.7554/eLife.01202
Schirmer,, M., Smeekens,, S. P., Vlamakis,, H., Jaeger,, M., Oosting,, M., Franzosa,, E. A., … Xavier,, R. J. (2016). Linking the human gut microbiome to inflammatory cytokine production capacity. Cell, 167(7), 1897. https://doi.org/10.1016/j.cell.2016.11.046
Schlapbach,, C., Gehad,, A., Yang,, C., Watanabe,, R., Guenova,, E., Teague,, J. E., … Clark,, R. A. (2014). Human TH9 cells are skin‐tropic and have autocrine and paracrine proinflammatory capacity. Science Translational Medicine, 6(219), 219ra8. https://doi.org/10.1126/scitranslmed.3007828
Schoch,, C. L., Seifert,, K. A., Huhndorf,, S., Robert,, V., Spouge,, J. L., Levesque,, C. A., … Fungal Barcoding Consortium. (2012). Nuclear ribosomal internal transcribed spacer (ITS) region as a universal DNA barcode marker for fungi. Proceedings of the National Academy of Sciences of the United States of America, 109(16), 6241–6246. https://doi.org/10.1073/pnas.1117018109
Schubert,, K., Groenewald,, J. Z., Braun,, U., Dijksterhuis,, J., Starink,, M., Hill,, C. F., … Crous,, P. W. (2007). Biodiversity in the Cladosporium herbarum complex (Davidiellaceae, Capnodiales), with standardisation of methods for Cladosporium taxonomy and diagnostics. Studies in Mycology, 58, 105–156. https://doi.org/10.3114/sim.2007.58.05
Schulte,, B. C., Wu,, W., & Rosen,, T. (2015). Azelaic acid: Evidence‐based update on mechanism of action and clinical application. Journal of Drugs in Dermatology, 14(9), 964–968.
Shapiro,, H., Thaiss,, C. A., Levy,, M., & Elinav,, E. (2014). The cross talk between microbiota and the immune system: Metabolites take center stage. Current Opinion in Immunology, 30, 54–62. https://doi.org/10.1016/j.coi.2014.07.003
Shipley,, T. W., Kling,, H. M., Morris,, A., Patil,, S., Kristoff,, J., Guyach,, S. E., … Norris,, K. A. (2010). Persistent pneumocystis colonization leads to the development of chronic obstructive pulmonary disease in a nonhuman primate model of AIDS. Journal of Infectious Diseases, 202(2), 302–312. https://doi.org/10.1086/653485
Skouboe,, P., Frisvad,, J. C., Taylor,, J. W., Lauritsen,, D., Boysen,, M., & Rossen,, L. (1999). Phylogenetic analysis of nucleotide sequences from the ITS region of terverticillate Penicillium species. Mycological Research, 103(7), 873–881. https://doi.org/10.1017/S0953756298007904
Smits,, S. A., Leach,, J., Sonnenburg,, E. D., Gonzalez,, C. G., Lichtman,, J. S., Reid,, G., … Sonnenburg,, J. L. (2017). Seasonal cycling in the gut microbiome of the Hadza hunter–gatherers of Tanzania. Science, 357(6353), 802–806. https://doi.org/10.1126/science.aan4834
Sokol,, H., Conway,, K. L., Zhang,, M., Choi,, M., Morin,, B., Cao,, Z., … Xavier,, R. J. (2013). Card9 mediates intestinal epithelial cell restitution, T‐helper 17 responses, and control of bacterial infection in mice. Gastroenterology, 145(3), 591–601 e593. https://doi.org/10.1053/j.gastro.2013.05.047
Sokol,, H., Leducq,, V., Aschard,, H., Pham,, H. P., Jegou,, S., Landman,, C., … Beaugerie,, L. (2017). Fungal microbiota dysbiosis in IBD. Gut, 66(6), 1039–1048. https://doi.org/10.1136/gutjnl-2015-310746
Sonnenburg,, J. L., & Backhed,, F. (2016). Diet–microbiota interactions as moderators of human metabolism. Nature, 535(7610), 56–64. https://doi.org/10.1038/nature18846
Spinillo,, A., Capuzzo,, E., Acciano,, S., De Santolo,, A., & Zara,, F. (1999). Effect of antibiotic use on the prevalence of symptomatic vulvovaginal candidiasis. American Journal of Obstetrics and Gynecology, 180(1 Pt. 1), 14–17.
Strati,, F., Calabro,, A., Donati,, C., De Felice,, C., Hayek,, J., Jousson,, O., … Cavalieri,, D. (2018). Intestinal Candida parapsilosis isolates from Rett syndrome subjects bear potential virulent traits and capacity to persist within the host. BMC Gastroenterology, 18(1), 57. https://doi.org/10.1186/s12876-018-0785-z
Strati,, F., Cavalieri,, D., Albanese,, D., De Felice,, C., Donati,, C., Hayek,, J., … De Filippo,, C. (2016). Altered gut microbiota in Rett syndrome. Microbiome, 4(1), 41. https://doi.org/10.1186/s40168-016-0185-y
Strati,, F., Cavalieri,, D., Albanese,, D., De Felice,, C., Donati,, C., Hayek,, J., … De Filippo,, C. (2017). New evidences on the altered gut microbiota in autism spectrum disorders. Microbiome, 5(1), 24. https://doi.org/10.1186/s40168-017-0242-1
Strati,, F., Di Paola,, M., Stefanini,, I., Albanese,, D., Rizzetto,, L., Lionetti,, P., … De Filippo,, C. (2016). Age and gender affect the composition of fungal population of the human gastrointestinal tract. Frontiers in Microbiology, 7, 1227. https://doi.org/10.3389/fmicb.2016.01227
Surana,, N. K., & Kasper,, D. L. (2017). Moving beyond microbiome‐wide associations to causal microbe identification. Nature, 552(7684), 244–247. https://doi.org/10.1038/nature25019
Tamboli,, C. P., Neut,, C., Desreumaux,, P., & Colombel,, J. F. (2004). Dysbiosis as a prerequisite for IBD. Gut, 53(7), 1057.
Tang,, J., Iliev,, I. D., Brown,, J., Underhill,, D. M., & Funari,, V. A. (2015). Mycobiome: Approaches to analysis of intestinal fungi. Journal of Immunological Methods, 421, 112–121. https://doi.org/10.1016/j.jim.2015.04.004
Ter Horst,, R., Jaeger,, M., Smeekens,, S. P., Oosting,, M., Swertz,, M. A., Li,, Y., … Netea,, M. G. (2016). Host and environmental factors influencing individual human cytokine responses. Cell, 167(4), 1111–1124 e1113. https://doi.org/10.1016/j.cell.2016.10.018
Trompette,, A., Gollwitzer,, E. S., Pattaroni,, C., Lopez‐Mejia,, I. C., Riva,, E., Pernot,, J., … Marsland,, B. J. (2018). Dietary fiber confers protection against flu by shaping Ly6c(−) patrolling monocyte hematopoiesis and CD8(+) T cell metabolism. Immunity, 48(5), 992–1005 e1008. https://doi.org/10.1016/j.immuni.2018.04.022
Turnbaugh,, P. J., Ley,, R. E., Mahowald,, M. A., Magrini,, V., Mardis,, E. R., & Gordon,, J. I. (2006). An obesity‐associated gut microbiome with increased capacity for energy harvest. Nature, 444(7122), 1027–1031. https://doi.org/10.1038/nature05414
Tyc,, K. M., Herwald,, S. E., Hogan,, J. A., Pierce,, J. V., Klipp,, E., & Kumamoto,, C. A. (2016). The game theory of Candida albicans colonization dynamics reveals host status‐responsive gene expression. BMC Systems Biology, 10, 20. https://doi.org/10.1186/s12918-016-0268-1
van de Pavert,, S. A., Ferreira,, M., Domingues,, R. G., Ribeiro,, H., Molenaar,, R., Moreira‐Santos,, L., … Veiga‐Fernandes,, H. (2014). Maternal retinoids control type 3 innate lymphoid cells and set the offspring immunity. Nature, 508(7494), 123–127. https://doi.org/10.1038/nature13158
van de Veerdonk,, F. L., Plantinga,, T. S., Hoischen,, A., Smeekens,, S. P., Joosten,, L. A., Gilissen,, C., … Netea,, M. G. (2011). STAT1 mutations in autosomal dominant chronic mucocutaneous candidiasis. New England Journal of Medicine, 365(1), 54–61. https://doi.org/10.1056/NEJMoa1100102
van Woerden,, H. C., Gregory,, C., Brown,, R., Marchesi,, J. R., Hoogendoorn,, B., & Matthews,, I. P. (2013). Differences in fungi present in induced sputum samples from asthma patients and non‐atopic controls: A community based case control study. BMC Infectious Diseases, 13, 69. https://doi.org/10.1186/1471-2334-13-69
VanDussen,, K. L., Marinshaw,, J. M., Shaikh,, N., Miyoshi,, H., Moon,, C., Tarr,, P. I., … Stappenbeck,, T. S. (2015). Development of an enhanced human gastrointestinal epithelial culture system to facilitate patient‐based assays. Gut, 64(6), 911–920. https://doi.org/10.1136/gutjnl-2013-306651
Velegraki,, A., Cafarchia,, C., Gaitanis,, G., Iatta,, R., & Boekhout,, T. (2015). Malassezia infections in humans and animals: Pathophysiology, detection, and treatment. PLoS Pathogens, 11(1), e1004523. https://doi.org/10.1371/journal.ppat.1004523
Velraeds,, M. M., van de Belt‐Gritter,, B., van der Mei,, H. C., Reid,, G., & Busscher,, H. J. (1998). Interference in initial adhesion of uropathogenic bacteria and yeasts to silicone rubber by a Lactobacillus acidophilus biosurfactant. Journal of Medical Microbiology, 47(12), 1081–1085. https://doi.org/10.1099/00222615-47-12-1081
Vesty,, A., Biswas,, K., Taylor,, M. W., Gear,, K., & Douglas,, R. G. (2017). Evaluating the impact of DNA extraction method on the representation of human oral bacterial and fungal communities. PLoS One, 12(1), e0169877. https://doi.org/10.1371/journal.pone.0169877
Virgin,, H. W. (2014). The virome in mammalian physiology and disease. Cell, 157(1), 142–150. https://doi.org/10.1016/j.cell.2014.02.032
Vlachos,, C., Schulte,, B. M., Magiatis,, P., Adema,, G. J., & Gaitanis,, G. (2012). Malassezia‐derived indoles activate the aryl hydrocarbon receptor and inhibit toll‐like receptor‐induced maturation in monocyte‐derived dendritic cells. British Journal of Dermatology, 167(3), 496–505. https://doi.org/10.1111/j.1365-2133.2012.11014.x
Wagener,, J., MacCallum,, D. M., Brown,, G. D., & Gow,, N. A. (2017). Candida albicans chitin increases Arginase‐1 activity in human macrophages, with an impact on macrophage antimicrobial functions. mBio, 8(1), e01820‐16. https://doi.org/10.1128/mBio.01820-16
Waggoner‐Fountain,, L. A., Walker,, M. W., Hollis,, R. J., Pfaller,, M. A., Ferguson,, J. E., 2nd, Wenzel,, R. P., & Donowitz,, L. G. (1996). Vertical and horizontal transmission of unique Candida species to premature newborns. Clinical Infectious Diseases, 22(5), 803–808.
Wang,, T., Pan,, D., Zhou,, Z., You,, Y., Jiang,, C., Zhao,, X., & Lin,, X. (2016). Dectin‐3 deficiency promotes colitis development due to impaired antifungal innate immune responses in the gut. PLoS Pathogens, 12(6), e1005662. https://doi.org/10.1371/journal.ppat.1005662
Wheeler,, M. L., Limon,, J. J., Bar,, A. S., Leal,, C. A., Gargus,, M., Tang,, J., … Iliev,, I. D. (2016). Immunological consequences of intestinal fungal dysbiosis. Cell Host %26 Microbe, 19(6), 865–873. https://doi.org/10.1016/j.chom.2016.05.003
White,, T. C., Findley,, K., Dawson,, T. L., Jr., Scheynius,, A., Boekhout,, T., Cuomo,, C. A., … Saunders,, C. W. (2014). Fungi on the skin: Dermatophytes and Malassezia. Cold Spring Harbor Perspectives in Medicine, 4(8), a019802. https://doi.org/10.1101/cshperspect.a019802
Wisecaver,, J. H., Slot,, J. C., & Rokas,, A. (2014). The evolution of fungal metabolic pathways. PLoS Genetics, 10(12), e1004816. https://doi.org/10.1371/journal.pgen.1004816
Wolfe,, B. E., Button,, J. E., Santarelli,, M., & Dutton,, R. J. (2014). Cheese rind communities provide tractable systems for in situ and in vitro studies of microbial diversity. Cell, 158(2), 422–433. https://doi.org/10.1016/j.cell.2014.05.041
Xin,, L., Jiang,, T. T., Chaturvedi,, V., Kinder,, J. M., Ertelt,, J. M., Rowe,, J. H., … Way,, S. S. (2014). Commensal microbes drive intestinal inflammation by IL‐17‐producing CD4+ T cells through ICOSL and OX40L costimulation in the absence of B7‐1 and B7‐2. Proceedings of the National Academy of Sciences of the United States of America, 111(29), 10672–10677. https://doi.org/10.1073/pnas.1402336111
Xu,, J., Schwartz,, K., Bartoces,, M., Monsur,, J., Severson,, R. K., & Sobel,, J. D. (2008). Effect of antibiotics on vulvovaginal candidiasis: A MetroNet study. Journal of American Board of Family Medicine, 21(4), 261–268. https://doi.org/10.3122/jabfm.2008.04.070169
Xu,, X. L., Lee,, R. T., Fang,, H. M., Wang,, Y. M., Li,, R., Zou,, H., … Wang,, Y. (2008). Bacterial peptidoglycan triggers Candida albicans hyphal growth by directly activating the adenylyl cyclase Cyr1p. Cell Host %26 Microbe, 4(1), 28–39. https://doi.org/10.1016/j.chom.2008.05.014
Yang,, A. M., Inamine,, T., Hochrath,, K., Chen,, P., Wang,, L., Llorente,, C., … Schnabl,, B. (2017). Intestinal fungi contribute to development of alcoholic liver disease. Journal of Clinical Investigation, 127(7), 2829–2841. https://doi.org/10.1172/JCI90562
Yatsunenko,, T., Rey,, F. E., Manary,, M. J., Trehan,, I., Dominguez‐Bello,, M. G., Contreras,, M., … Gordon,, J. I. (2012). Human gut microbiome viewed across age and geography. Nature, 486(7402), 222–227. https://doi.org/10.1038/nature11053
Yissachar,, N., Zhou,, Y., Ung,, L., Lai,, N. Y., Mohan,, J. F., Ehrlicher,, A., … Benoist,, C. (2017). An intestinal organ culture system uncovers a role for the nervous system in microbe‐immune crosstalk. Cell, 168(6), 1135–1148 e1112. https://doi.org/10.1016/j.cell.2017.02.009
Yoshitomi,, H., Sakaguchi,, N., Kobayashi,, K., Brown,, G. D., Tagami,, T., Sakihama,, T., … Sakaguchi,, S. (2005). A role for fungal β‐glucans and their receptor Dectin‐1 in the induction of autoimmune arthritis in genetically susceptible mice. Journal of Experimental Medicine, 201(6), 949–960. https://doi.org/10.1084/jem.20041758
Zelante,, T., Iannitti,, R. G., Cunha,, C., De Luca,, A., Giovannini,, G., Pieraccini,, G., … Romani,, L. (2013). Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin‐22. Immunity, 39(2), 372–385. https://doi.org/10.1016/j.immuni.2013.08.003
Zhang,, I., Pletcher,, S. D., Goldberg,, A. N., Barker,, B. M., & Cope,, E. K. (2017). Fungal microbiota in chronic airway inflammatory disease and emerging relationships with the host immune response. Frontiers in Microbiology, 8, 2477. https://doi.org/10.3389/fmicb.2017.02477
Zhang,, Y., Kastman,, E. K., Guasto,, J. S., & Wolfe,, B. E. (2018). Fungal networks shape dynamics of bacterial dispersal and community assembly in cheese rind microbiomes. Nature Communications, 9(1), 336. https://doi.org/10.1038/s41467-017-02522-z
Zhang,, Z., Li,, J., Zheng,, W., Zhao,, G., Zhang,, H., Wang,, X., … Shi,, Y. (2016). Peripheral lymphoid volume expansion and maintenance are controlled by gut microbiota via RALDH+ dendritic cells. Immunity, 44(2), 330–342. https://doi.org/10.1016/j.immuni.2016.01.004
Zielinski,, C. E., Mele,, F., Aschenbrenner,, D., Jarrossay,, D., Ronchi,, F., Gattorno,, M., … Sallusto,, F. (2012). Pathogen‐induced human TH17 cells produce IFN‐gamma or IL‐10 and are regulated by IL‐1beta. Nature, 484(7395), 514–518. https://doi.org/10.1038/nature10957
Zwolinska‐Wcislo,, M., Brzozowski,, T., Budak,, A., Kwiecien,, S., Sliwowski,, Z., Drozdowicz,, D., … Pawlik,, W. W. (2009). Effect of Candida colonization on human ulcerative colitis and the healing of inflammatory changes of the colon in the experimental model of colitis ulcerosa. Journal of Physiology and Pharmacology, 60(1), 107–118.