Mucosal Immunity to Gut Fungi in Health and Inflammatory Bowel Disease
Abstract
:1. Introduction
2. ASCA and Other Anti-Glycan Antibodies
3. The Mycobiome
4. Candida albicans Pathobiology
5. Key Virulence Factors: Hyphae, Adhesins, and Invasins
6. C. albicans Intra-Species Variation
7. Mucosal Immunology
8. C-Type Lectin Receptors and CARD9
9. Myeloid Cells
10. T Cells
11. Therapies
12. Conclusions
Funding
Conflicts of Interest
References
- Alatab, S.; Sepanlou, S.G.; Ikuta, K.; Vahedi, H.; Bisignano, C.; Safiri, S.; Sadeghi, A.; Nixon, M.R.; Abdoli, A.; Abolhassani, H.; et al. The global, regional, and national burden of inflammatory bowel disease in 195 countries and territories, 1990–2017: A systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol. Hepatol. 2020, 5, 17–30. [Google Scholar] [CrossRef]
- Brodin, P.; Davis, M.M. Human immune system variation. Nat. Rev. Immunol. 2017, 17, 21–29. [Google Scholar] [CrossRef] [PubMed]
- Malard, F.; Dore, J.; Gaugler, B.; Mohty, M. Introduction to host microbiome symbiosis in health and disease. Mucosal Immunol. 2021, 14, 547–554. [Google Scholar] [CrossRef]
- Chudnovskiy, A.; Mortha, A.; Kana, V.; Kennard, A.; Ramirez, J.D.; Rahman, A.; Remark, R.; Mogno, I.; Ng, R.; Gnjatic, S.; et al. Host-Protozoan Interactions Protect from Mucosal Infections through Activation of the Inflammasome. Cell 2016, 167, 444–456.e414. [Google Scholar] [CrossRef] [PubMed]
- Rangan, K.J.; Pedicord, V.A.; Wang, Y.C.; Kim, B.; Lu, Y.; Shaham, S.; Mucida, D.; Hang, H.C. A secreted bacterial peptidoglycan hydrolase enhances tolerance to enteric pathogens. Science 2016, 353, 1434–1437. [Google Scholar] [CrossRef] [PubMed]
- Levy, M.; Kolodziejczyk, A.A.; Thaiss, C.A.; Elinav, E. Dysbiosis and the immune system. Nat. Rev. Immunol. 2017, 17, 219–232. [Google Scholar] [CrossRef] [PubMed]
- Chehoud, C.; Albenberg, L.G.; Judge, C.; Hoffmann, C.; Grunberg, S.; Bittinger, K.; Baldassano, R.N.; Lewis, J.D.; Bushman, F.D.; Wu, G.D. Fungal Signature in the Gut Microbiota of Pediatric Patients With Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2015, 21, 1948–1956. [Google Scholar] [CrossRef]
- Liguori, G.; Lamas, B.; Richard, M.L.; Brandi, G.; da Costa, G.; Hoffmann, T.W.; Di Simone, M.P.; Calabrese, C.; Poggioli, G.; Langella, P.; et al. Fungal Dysbiosis in Mucosa-associated Microbiota of Crohn’s Disease Patients. J. Crohns Colitis 2016, 10, 296–305. [Google Scholar] [CrossRef]
- Hoarau, G.; Mukherjee, P.K.; Gower-Rousseau, C.; Hager, C.; Chandra, J.; Retuerto, M.A.; Neut, C.; Vermeire, S.; Clemente, J.; Colombel, J.F.; et al. Bacteriome and Mycobiome Interactions Underscore Microbial Dysbiosis in Familial Crohn’s Disease. mBio 2016, 7, e01250-16. [Google Scholar] [CrossRef]
- Wright, E.K.; Kamm, M.A.; Teo, S.M.; Inouye, M.; Wagner, J.; Kirkwood, C.D. Recent advances in characterizing the gastrointestinal microbiome in Crohn’s disease: A systematic review. Inflamm. Bowel Dis. 2015, 21, 1219–1228. [Google Scholar] [CrossRef]
- Jin, J.; Yamamoto, R.; Takeuchi, T.; Cui, G.; Miyauchi, E.; Hojo, N.; Ikuta, K.; Ohno, H.; Shiroguchi, K. High-throughput identification and quantification of single bacterial cells in the microbiota. Nat. Commun. 2022, 13, 863. [Google Scholar] [CrossRef] [PubMed]
- Lagier, J.C.; Dubourg, G.; Million, M.; Cadoret, F.; Bilen, M.; Fenollar, F.; Levasseur, A.; Rolain, J.M.; Fournier, P.E.; Raoult, D. Culturing the human microbiota and culturomics. Nat. Rev. Microbiol. 2018, 16, 540–550. [Google Scholar] [CrossRef] [PubMed]
- Delavy, M.; Burdet, C.; Sertour, N.; Devente, S.; Docquier, J.D.; Grall, N.; Volant, S.; Ghozlane, A.; Duval, X.; Mentre, F.; et al. A Clinical Study Provides the First Direct Evidence That Interindividual Variations in Fecal beta-Lactamase Activity Affect the Gut Mycobiota Dynamics in Response to beta-Lactam Antibiotics. mBio 2022, 13, e0288022. [Google Scholar] [CrossRef] [PubMed]
- Standaert-Vitse, A.; Jouault, T.; Vandewalle, P.; Mille, C.; Seddik, M.; Sendid, B.; Mallet, J.M.; Colombel, J.F.; Poulain, D. Candida albicans is an immunogen for anti-Saccharomyces cerevisiae antibody markers of Crohn’s disease. Gastroenterology 2006, 130, 1764–1775. [Google Scholar] [CrossRef] [PubMed]
- Seow, C.H.; Stempak, J.M.; Xu, W.; Lan, H.; Griffiths, A.M.; Greenberg, G.R.; Steinhart, A.H.; Dotan, N.; Silverberg, M.S. Novel anti-glycan antibodies related to inflammatory bowel disease diagnosis and phenotype. Am. J. Gastroenterol. 2009, 104, 1426–1434. [Google Scholar] [CrossRef] [PubMed]
- Kaul, A.; Hutfless, S.; Liu, L.; Bayless, T.M.; Marohn, M.R.; Li, X. Serum anti-glycan antibody biomarkers for inflammatory bowel disease diagnosis and progression: A systematic review and meta-analysis. Inflamm. Bowel Dis. 2012, 18, 1872–1884. [Google Scholar] [CrossRef]
- Schaffer, T.; Muller, S.; Flogerzi, B.; Seibold-Schmid, B.; Schoepfer, A.M.; Seibold, F. Anti-Saccharomyces cerevisiae mannan antibodies (ASCA) of Crohn’s patients crossreact with mannan from other yeast strains, and murine ASCA IgM can be experimentally induced with Candida albicans. Inflamm. Bowel Dis. 2007, 13, 1339–1346. [Google Scholar] [CrossRef]
- Quinton, J.F.; Sendid, B.; Reumaux, D.; Duthilleul, P.; Cortot, A.; Grandbastien, B.; Charrier, G.; Targan, S.R.; Colombel, J.F.; Poulain, D. Anti-Saccharomyces cerevisiae mannan antibodies combined with antineutrophil cytoplasmic autoantibodies in inflammatory bowel disease: Prevalence and diagnostic role. Gut 1998, 42, 788–791. [Google Scholar] [CrossRef]
- Khan, K.; Schwarzenberg, S.J.; Sharp, H.; Greenwood, D.; Weisdorf-Schindele, S. Role of serology and routine laboratory tests in childhood inflammatory bowel disease. Inflamm. Bowel Dis. 2002, 8, 325–329. [Google Scholar] [CrossRef]
- Vasseur, F.; Sendid, B.; Jouault, T.; Standaert-Vitse, A.; Dubuquoy, L.; Francois, N.; Gower-Rousseau, C.; Desreumaux, P.; Broly, F.; Vermeire, S.; et al. Variants of NOD1 and NOD2 genes display opposite associations with familial risk of Crohn’s disease and anti-saccharomyces cerevisiae antibody levels. Inflamm. Bowel Dis. 2012, 18, 430–438. [Google Scholar] [CrossRef]
- Reese, G.E.; Constantinides, V.A.; Simillis, C.; Darzi, A.W.; Orchard, T.R.; Fazio, V.W.; Tekkis, P.P. Diagnostic precision of anti-Saccharomyces cerevisiae antibodies and perinuclear antineutrophil cytoplasmic antibodies in inflammatory bowel disease. Am. J. Gastroenterol. 2006, 101, 2410–2422. [Google Scholar] [CrossRef]
- Kansal, S.; Catto-Smith, A.G.; Boniface, K.; Thomas, S.; Cameron, D.J.; Oliver, M.; Alex, G.; Kirkwood, C.D.; Wagner, J. Variation of Gut Mucosal Microbiome With Anti-Saccharomyces cerevisiae Antibody Status in Pediatric Crohn Disease. J. Pediatr. Gastroenterol. Nutr. 2019, 69, 696–703. [Google Scholar] [CrossRef] [PubMed]
- Ruemmele, F.M.; Targan, S.R.; Levy, G.; Dubinsky, M.; Braun, J.; Seidman, E.G. Diagnostic accuracy of serological assays in pediatric inflammatory bowel disease. Gastroenterology 1998, 115, 822–829. [Google Scholar] [CrossRef] [PubMed]
- Forcione, D.G.; Rosen, M.J.; Kisiel, J.B.; Sands, B.E. Anti-Saccharomyces cerevisiae antibody (ASCA) positivity is associated with increased risk for early surgery in Crohn’s disease. Gut 2004, 53, 1117–1122. [Google Scholar] [CrossRef] [PubMed]
- Ryan, J.D.; Silverberg, M.S.; Xu, W.; Graff, L.A.; Targownik, L.E.; Walker, J.R.; Carr, R.; Clara, I.; Miller, N.; Rogala, L.; et al. Predicting complicated Crohn’s disease and surgery: Phenotypes, genetics, serology and psychological characteristics of a population-based cohort. Aliment. Pharmacol. Ther. 2013, 38, 274–283. [Google Scholar] [CrossRef]
- Israeli, E.; Grotto, I.; Gilburd, B.; Balicer, R.D.; Goldin, E.; Wiik, A.; Shoenfeld, Y. Anti-Saccharomyces cerevisiae and antineutrophil cytoplasmic antibodies as predictors of inflammatory bowel disease. Gut 2005, 54, 1232–1236. [Google Scholar] [CrossRef]
- van Schaik, F.D.; Oldenburg, B.; Hart, A.R.; Siersema, P.D.; Lindgren, S.; Grip, O.; Teucher, B.; Kaaks, R.; Bergmann, M.M.; Boeing, H.; et al. Serological markers predict inflammatory bowel disease years before the diagnosis. Gut 2013, 62, 683–688. [Google Scholar] [CrossRef]
- Bartunkova, J.; Kolarova, I.; Sediva, A.; Holzelova, E. Antineutrophil cytoplasmic antibodies, anti-Saccharomyces cerevisiae antibodies, and specific IgE to food allergens in children with inflammatory bowel diseases. Clin. Immunol. 2002, 102, 162–168. [Google Scholar] [CrossRef]
- Dubinsky, M.C.; Ofman, J.J.; Urman, M.; Targan, S.R.; Seidman, E.G. Clinical utility of serodiagnostic testing in suspected pediatric inflammatory bowel disease. Am. J. Gastroenterol. 2001, 96, 758–765. [Google Scholar] [CrossRef]
- Canani, R.B.; Romano, M.T.; Greco, L.; Terrin, G.; Sferlazzas, C.; Barabino, A.; Fontana, M.; Roggero, P.; Guariso, G.; De Angelis, G.; et al. Effects of disease activity on anti-Saccharomyces cerevisiae antibodies: Implications for diagnosis and follow-up of children with Crohn’s disease. Inflamm. Bowel Dis. 2004, 10, 234–239. [Google Scholar] [CrossRef]
- Standaert-Vitse, A.; Sendid, B.; Joossens, M.; François, N.; Vandewalle-El Khoury, P.; Branche, J.; Van Kruiningen, H.; Jouault, T.; Rutgeerts, P.; Gower-Rousseau, C.; et al. Candida albicans Colonization and ASCA in Familial Crohn’s Disease. Off. J. Am. Coll. Gastroenterol. ACG 2009, 104, 1745–1753. [Google Scholar] [CrossRef] [PubMed]
- Maaser, C.; Sturm, A.; Vavricka, S.R.; Kucharzik, T.; Fiorino, G.; Annese, V.; Calabrese, E.; Baumgart, D.C.; Bettenworth, D.; Borralho Nunes, P.; et al. ECCO-ESGAR Guideline for Diagnostic Assessment in IBD Part 1: Initial diagnosis, monitoring of known IBD, detection of complications. J. Crohns Colitis 2019, 13, 144–164. [Google Scholar] [CrossRef] [PubMed]
- Li, Q.; Wang, C.; Tang, C.; He, Q.; Li, N.; Li, J. Dysbiosis of gut fungal microbiota is associated with mucosal inflammation in Crohn’s disease. J. Clin. Gastroenterol. 2014, 48, 513–523. [Google Scholar] [CrossRef] [PubMed]
- Sokol, H.; Leducq, V.; Aschard, H.; Pham, H.P.; Jegou, S.; Landman, C.; Cohen, D.; Liguori, G.; Bourrier, A.; Nion-Larmurier, I.; et al. Fungal microbiota dysbiosis in IBD. Gut 2017, 66, 1039–1048. [Google Scholar] [CrossRef]
- Di Paola, M.; Rizzetto, L.; Stefanini, I.; Vitali, F.; Massi-Benedetti, C.; Tocci, N.; Romani, L.; Ramazzotti, M.; Lionetti, P.; De Filippo, C.; et al. Comparative immunophenotyping of Saccharomyces cerevisiae and Candida spp. strains from Crohn’s disease patients and their interactions with the gut microbiome. J. Transl. Autoimmun. 2020, 3, 100036. [Google Scholar] [CrossRef]
- Li, X.V.; Leonardi, I.; Putzel, G.G.; Semon, A.; Fiers, W.D.; Kusakabe, T.; Lin, W.Y.; Gao, I.H.; Doron, I.; Gutierrez-Guerrero, A.; et al. Immune regulation by fungal strain diversity in inflammatory bowel disease. Nature 2022, 603, 672–678. [Google Scholar] [CrossRef]
- Shah, J.; Dutta, U.; Rudramurthy, S.; Chakrabarti, A.; Sinha, S.K.; Sharma, V.; Mandavdhare, H.; Sharma, P.; Kalsi, D.; Popli, P.; et al. Colonic mucosa-associated candida assessed by biopsy culture is associated with disease severity in ulcerative colitis: A prospective study. J. Dig. Dis. 2019, 20, 642–648. [Google Scholar] [CrossRef]
- Hsia, K.; Zhao, N.; Chung, M.; Algarrahi, K.; Kouhsari, L.M.; Fu, M.; Chen, H.; Singh, S.; Michaud, D.S.; Jangi, S. Alterations in the Fungal Microbiome in Ulcerative Colitis. Inflamm. Bowel Dis. 2023, 29, 1613–1621. [Google Scholar] [CrossRef]
- Imai, T.; Inoue, R.; Kawada, Y.; Morita, Y.; Inatomi, O.; Nishida, A.; Bamba, S.; Kawahara, M.; Andoh, A. Characterization of fungal dysbiosis in Japanese patients with inflammatory bowel disease. J. Gastroenterol. 2019, 54, 149–159. [Google Scholar] [CrossRef]
- Mao, X.; Ma, J.; Jiao, C.; Tang, N.; Zhao, X.; Wang, D.; Zhang, Y.; Ye, Z.; Xu, C.; Jiang, J.; et al. Faecalibacterium prausnitzii Attenuates DSS-Induced Colitis by Inhibiting the Colonization and Pathogenicity of Candida albicans. Mol. Nutr. Food Res. 2021, 65, e2100433. [Google Scholar] [CrossRef]
- Bandara, H.M.; Cheung, B.P.; Watt, R.M.; Jin, L.J.; Samaranayake, L.P. Secretory products of Escherichia coli biofilm modulate Candida biofilm formation and hyphal development. J. Investig. Clin. Dent. 2013, 4, 186–199. [Google Scholar] [CrossRef] [PubMed]
- Savage, H.P.; Bays, D.J.; Gonzalez, M.A.F.; Bejarano, E.J.; Nguyen, H.; Masson, H.L.P.; Carvalho, T.P.; Santos, R.L.; Thompson, G.R.; Baumler, A.J. 5-ASA can functionally replace Clostridia to prevent a post-antibiotic bloom of Candida albicans by maintaining epithelial hypoxia. bioRxiv 2023. [Google Scholar] [CrossRef]
- Nash, A.K.; Auchtung, T.A.; Wong, M.C.; Smith, D.P.; Gesell, J.R.; Ross, M.C.; Stewart, C.J.; Metcalf, G.A.; Muzny, D.M.; Gibbs, R.A.; et al. The gut mycobiome of the Human Microbiome Project healthy cohort. Microbiome 2017, 5, 153. [Google Scholar] [CrossRef]
- Shao, T.Y.; Haslam, D.B.; Bennett, R.J.; Way, S.S. Friendly fungi: Symbiosis with commensal Candida albicans. Trends Immunol. 2022, 43, 706–717. [Google Scholar] [CrossRef] [PubMed]
- White, S.J.; Rosenbach, A.; Lephart, P.; Nguyen, D.; Benjamin, A.; Tzipori, S.; Whiteway, M.; Mecsas, J.; Kumamoto, C.A. Self-regulation of Candida albicans population size during GI colonization. PLoS Pathog. 2007, 3, e184. [Google Scholar] [CrossRef] [PubMed]
- Dalle, F.; Wachtler, B.; L’Ollivier, C.; Holland, G.; Bannert, N.; Wilson, D.; Labruere, C.; Bonnin, A.; Hube, B. Cellular interactions of Candida albicans with human oral epithelial cells and enterocytes. Cell Microbiol. 2010, 12, 248–271. [Google Scholar] [CrossRef]
- Moyes, D.L.; Wilson, D.; Richardson, J.P.; Mogavero, S.; Tang, S.X.; Wernecke, J.; Hofs, S.; Gratacap, R.L.; Robbins, J.; Runglall, M.; et al. Candidalysin is a fungal peptide toxin critical for mucosal infection. Nature 2016, 532, 64–68. [Google Scholar] [CrossRef]
- Lorenz, M.C.; Bender, J.A.; Fink, G.R. Transcriptional response of Candida albicans upon internalization by macrophages. Eukaryot. Cell 2004, 3, 1076–1087. [Google Scholar] [CrossRef]
- Sudbery, P.E. Growth of Candida albicans hyphae. Nat. Rev. Microbiol. 2011, 9, 737–748. [Google Scholar] [CrossRef]
- Leonardi, I.; Gao, I.H.; Lin, W.Y.; Allen, M.; Li, X.V.; Fiers, W.D.; De Celie, M.B.; Putzel, G.G.; Yantiss, R.K.; Johncilla, M.; et al. Mucosal fungi promote gut barrier function and social behavior via Type 17 immunity. Cell 2022, 185, 831–846.e814. [Google Scholar] [CrossRef]
- Koh, A.Y.; Kohler, J.R.; Coggshall, K.T.; Van Rooijen, N.; Pier, G.B. Mucosal damage and neutropenia are required for Candida albicans dissemination. PLoS Pathog. 2008, 4, e35. [Google Scholar] [CrossRef] [PubMed]
- Gregory, M.H.; Spec, A.; Stwalley, D.; Gremida, A.; Mejia-Chew, C.; Nickel, K.B.; Ciorba, M.A.; Rood, R.P.; Olsen, M.A.; Deepak, P. Corticosteroids Increase the Risk of Invasive Fungal Infections More Than Tumor Necrosis Factor-Alpha Inhibitors in Patients With Inflammatory Bowel Disease. Crohns Colitis 360 2023, 5, otad010. [Google Scholar] [CrossRef] [PubMed]
- World Health Organization. WHO Fungal Priority Pathogens List to Guide Research, Development and Public Health Action. Available online: https://www.who.int/publications/i/item/9789240060241 (accessed on 6 September 2023).
- de Groot, P.W.; Bader, O.; de Boer, A.D.; Weig, M.; Chauhan, N. Adhesins in human fungal pathogens: Glue with plenty of stick. Eukaryot. Cell 2013, 12, 470–481. [Google Scholar] [CrossRef]
- Liu, Y.; Filler, S.G. Candida albicans Als3, a multifunctional adhesin and invasin. Eukaryot. Cell 2011, 10, 168–173. [Google Scholar] [CrossRef] [PubMed]
- Martin, H.; Kavanagh, K.; Velasco-Torrijos, T. Targeting adhesion in fungal pathogen Candida albicans. Future Med. Chem. 2021, 13, 313–334. [Google Scholar] [CrossRef] [PubMed]
- Villa, S.; Hamideh, M.; Weinstock, A.; Qasim, M.N.; Hazbun, T.R.; Sellam, A.; Hernday, A.D.; Thangamani, S. Transcriptional control of hyphal morphogenesis in Candida albicans. FEMS Yeast Res. 2020, 20, foaa005. [Google Scholar] [CrossRef] [PubMed]
- Witchley, J.N.; Penumetcha, P.; Abon, N.V.; Woolford, C.A.; Mitchell, A.P.; Noble, S.M. Candida albicans Morphogenesis Programs Control the Balance between Gut Commensalism and Invasive Infection. Cell Host Microbe 2019, 25, 432–443.e436. [Google Scholar] [CrossRef]
- Schonherr, F.A.; Sparber, F.; Kirchner, F.R.; Guiducci, E.; Trautwein-Weidner, K.; Gladiator, A.; Sertour, N.; Hetzel, U.; Le, G.T.T.; Pavelka, N.; et al. The intraspecies diversity of C. albicans triggers qualitatively and temporally distinct host responses that determine the balance between commensalism and pathogenicity. Mucosal Immunol. 2017, 10, 1335–1350. [Google Scholar] [CrossRef]
- Ost, K.S.; O’Meara, T.R.; Stephens, W.Z.; Chiaro, T.; Zhou, H.; Penman, J.; Bell, R.; Catanzaro, J.R.; Song, D.; Singh, S.; et al. Adaptive immunity induces mutualism between commensal eukaryotes. Nature 2021, 596, 114–118. [Google Scholar] [CrossRef]
- Richardson, J.P.; Mogavero, S.; Moyes, D.L.; Blagojevic, M.; Kruger, T.; Verma, A.H.; Coleman, B.M.; De La Cruz Diaz, J.; Schulz, D.; Ponde, N.O.; et al. Processing of Candida albicans Ece1p Is Critical for Candidalysin Maturation and Fungal Virulence. mBio 2018, 9, e02178-17. [Google Scholar] [CrossRef]
- Ho, J.; Wickramasinghe, D.N.; Nikou, S.A.; Hube, B.; Richardson, J.P.; Naglik, J.R. Candidalysin Is a Potent Trigger of Alarmin and Antimicrobial Peptide Release in Epithelial Cells. Cells 2020, 9, 699. [Google Scholar] [CrossRef] [PubMed]
- Ho, J.; Yang, X.; Nikou, S.A.; Kichik, N.; Donkin, A.; Ponde, N.O.; Richardson, J.P.; Gratacap, R.L.; Archambault, L.S.; Zwirner, C.P.; et al. Candidalysin activates innate epithelial immune responses via epidermal growth factor receptor. Nat Commun 2019, 10, 2297. [Google Scholar] [CrossRef] [PubMed]
- Verma, A.H.; Richardson, J.P.; Zhou, C.; Coleman, B.M.; Moyes, D.L.; Ho, J.; Huppler, A.R.; Ramani, K.; McGeachy, M.J.; Mufazalov, I.A.; et al. Oral epithelial cells orchestrate innate type 17 responses to Candida albicans through the virulence factor candidalysin. Sci. Immunol. 2017, 2, eaam8834. [Google Scholar] [CrossRef] [PubMed]
- Verma, A.H.; Zafar, H.; Ponde, N.O.; Hepworth, O.W.; Sihra, D.; Aggor, F.E.Y.; Ainscough, J.S.; Ho, J.; Richardson, J.P.; Coleman, B.M.; et al. IL-36 and IL-1/IL-17 Drive Immunity to Oral Candidiasis via Parallel Mechanisms. J. Immunol. 2018, 201, 627–634. [Google Scholar] [CrossRef]
- Mogavero, S.; Sauer, F.M.; Brunke, S.; Allert, S.; Schulz, D.; Wisgott, S.; Jablonowski, N.; Elshafee, O.; Kruger, T.; Kniemeyer, O.; et al. Candidalysin delivery to the invasion pocket is critical for host epithelial damage induced by Candida albicans. Cell Microbiol. 2021, 23, e13378. [Google Scholar] [CrossRef]
- Blagojevic, M.; Camilli, G.; Maxson, M.; Hube, B.; Moyes, D.L.; Richardson, J.P.; Naglik, J.R. Candidalysin triggers epithelial cellular stresses that induce necrotic death. Cell Microbiol. 2021, 23, e13371. [Google Scholar] [CrossRef]
- Kasper, L.; Konig, A.; Koenig, P.A.; Gresnigt, M.S.; Westman, J.; Drummond, R.A.; Lionakis, M.S.; Gross, O.; Ruland, J.; Naglik, J.R.; et al. The fungal peptide toxin Candidalysin activates the NLRP3 inflammasome and causes cytolysis in mononuclear phagocytes. Nat. Commun. 2018, 9, 4260. [Google Scholar] [CrossRef]
- Liu, J.; Willems, H.M.E.; Sansevere, E.A.; Allert, S.; Barker, K.S.; Lowes, D.J.; Dixson, A.C.; Xu, Z.; Miao, J.; DeJarnette, C.; et al. A variant ECE1 allele contributes to reduced pathogenicity of Candida albicans during vulvovaginal candidiasis. PLoS Pathog. 2021, 17, e1009884. [Google Scholar] [CrossRef]
- Richardson, J.P.; Brown, R.; Kichik, N.; Lee, S.; Priest, E.; Mogavero, S.; Maufrais, C.; Wickramasinghe, D.N.; Tsavou, A.; Kotowicz, N.K.; et al. Candidalysins Are a New Family of Cytolytic Fungal Peptide Toxins. mBio 2022, 13, e03510-21. [Google Scholar] [CrossRef]
- Hirakawa, M.P.; Martinez, D.A.; Sakthikumar, S.; Anderson, M.Z.; Berlin, A.; Gujja, S.; Zeng, Q.; Zisson, E.; Wang, J.M.; Greenberg, J.M.; et al. Genetic and phenotypic intra-species variation in Candida albicans. Genome Res. 2015, 25, 413–425. [Google Scholar] [CrossRef]
- MacCallum, D.M.; Castillo, L.; Nather, K.; Munro, C.A.; Brown, A.J.; Gow, N.A.; Odds, F.C. Property differences among the four major Candida albicans strain clades. Eukaryot. Cell 2009, 8, 373–387. [Google Scholar] [CrossRef]
- van Thiel, I.A.M.; Stavrou, A.A.; de Jong, A.; Theelen, B.; Davids, M.; Hakvoort, T.B.M.; Admiraal-van den Berg, I.; Weert, I.C.M.; de Kruijs, M.; Vu, D.; et al. Genetic and phenotypic diversity of fecal Candida albicans strains in irritable bowel syndrome. Sci. Rep. 2022, 12, 5391. [Google Scholar] [CrossRef]
- Pabst, O.; Slack, E. IgA and the intestinal microbiota: The importance of being specific. Mucosal. Immunol. 2020, 13, 12–21. [Google Scholar] [CrossRef] [PubMed]
- Doron, I.; Mesko, M.; Li, X.V.; Kusakabe, T.; Leonardi, I.; Shaw, D.G.; Fiers, W.D.; Lin, W.Y.; Bialt-DeCelie, M.; Roman, E.; et al. Mycobiota-induced IgA antibodies regulate fungal commensalism in the gut and are dysregulated in Crohn’s disease. Nat. Microbiol. 2021, 6, 1493–1504. [Google Scholar] [CrossRef] [PubMed]
- Jouault, T.; Sarazin, A.; Martinez-Esparza, M.; Fradin, C.; Sendid, B.; Poulain, D. Host responses to a versatile commensal: PAMPs and PRRs interplay leading to tolerance or infection by Candida albicans. Cell Microbiol. 2009, 11, 1007–1015. [Google Scholar] [CrossRef]
- Rivas, M.A.; Beaudoin, M.; Gardet, A.; Stevens, C.; Sharma, Y.; Zhang, C.K.; Boucher, G.; Ripke, S.; Ellinghaus, D.; Burtt, N.; et al. Deep resequencing of GWAS loci identifies independent rare variants associated with inflammatory bowel disease. Nat. Genet 2011, 43, 1066–1073. [Google Scholar] [CrossRef] [PubMed]
- Iliev, I.D.; Funari, V.A.; Taylor, K.D.; Nguyen, Q.; Reyes, C.N.; Strom, S.P.; Brown, J.; Becker, C.A.; Fleshner, P.R.; Dubinsky, M.; et al. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science 2012, 336, 1314–1317. [Google Scholar] [CrossRef] [PubMed]
- Mata-Martinez, P.; Bergon-Gutierrez, M.; Del Fresno, C. Dectin-1 Signaling Update: New Perspectives for Trained Immunity. Front. Immunol. 2022, 13, 812148. [Google Scholar] [CrossRef]
- Gantner, B.N.; Simmons, R.M.; Underhill, D.M. Dectin-1 mediates macrophage recognition of Candida albicans yeast but not filaments. EMBO J. 2005, 24, 1277–1286. [Google Scholar] [CrossRef]
- Cheng, S.C.; van de Veerdonk, F.L.; Lenardon, M.; Stoffels, M.; Plantinga, T.; Smeekens, S.; Rizzetto, L.; Mukaremera, L.; Preechasuth, K.; Cavalieri, D.; et al. The dectin-1/inflammasome pathway is responsible for the induction of protective T-helper 17 responses that discriminate between yeasts and hyphae of Candida albicans. J. Leukoc. Biol. 2011, 90, 357–366. [Google Scholar] [CrossRef]
- LeibundGut-Landmann, S.; Gross, O.; Robinson, M.J.; Osorio, F.; Slack, E.C.; Tsoni, S.V.; Schweighoffer, E.; Tybulewicz, V.; Brown, G.D.; Ruland, J.; et al. Syk- and CARD9-dependent coupling of innate immunity to the induction of T helper cells that produce interleukin 17. Nat. Immunol. 2007, 8, 630–638. [Google Scholar] [CrossRef] [PubMed]
- Gringhuis, S.I.; Kaptein, T.M.; Wevers, B.A.; Theelen, B.; van der Vlist, M.; Boekhout, T.; Geijtenbeek, T.B. Dectin-1 is an extracellular pathogen sensor for the induction and processing of IL-1beta via a noncanonical caspase-8 inflammasome. Nat. Immunol. 2012, 13, 246–254. [Google Scholar] [CrossRef] [PubMed]
- Luo, P.; Yang, Z.; Chen, B.; Zhong, X. The multifaceted role of CARD9 in inflammatory bowel disease. J. Cell Mol. Med. 2020, 24, 34–39. [Google Scholar] [CrossRef] [PubMed]
- Drummond, R.A.; Franco, L.M.; Lionakis, M.S. Human CARD9: A Critical Molecule of Fungal Immune Surveillance. Front. Immunol. 2018, 9, 1836. [Google Scholar] [CrossRef]
- Gavino, C.; Hamel, N.; Zeng, J.B.; Legault, C.; Guiot, M.C.; Chankowsky, J.; Lejtenyi, D.; Lemire, M.; Alarie, I.; Dufresne, S.; et al. Impaired RASGRF1/ERK-mediated GM-CSF response characterizes CARD9 deficiency in French-Canadians. J. Allergy Clin. Immunol. 2016, 137, 1178–1188.e1177. [Google Scholar] [CrossRef]
- Glocker, E.O.; Hennigs, A.; Nabavi, M.; Schaffer, A.A.; Woellner, C.; Salzer, U.; Pfeifer, D.; Veelken, H.; Warnatz, K.; Tahami, F.; et al. A homozygous CARD9 mutation in a family with susceptibility to fungal infections. N. Engl. J. Med. 2009, 361, 1727–1735. [Google Scholar] [CrossRef]
- McGovern, D.P.; Gardet, A.; Torkvist, L.; Goyette, P.; Essers, J.; Taylor, K.D.; Neale, B.M.; Ong, R.T.; Lagace, C.; Li, C.; et al. Genome-wide association identifies multiple ulcerative colitis susceptibility loci. Nat. Genet 2010, 42, 332–337. [Google Scholar] [CrossRef]
- Zhernakova, A.; Festen, E.M.; Franke, L.; Trynka, G.; van Diemen, C.C.; Monsuur, A.J.; Bevova, M.; Nijmeijer, R.M.; van ‘t Slot, R.; Heijmans, R.; et al. Genetic analysis of innate immunity in Crohn’s disease and ulcerative colitis identifies two susceptibility loci harboring CARD9 and IL18RAP. Am. J. Hum. Genet 2008, 82, 1202–1210. [Google Scholar] [CrossRef]
- Sokol, H.; Conway, K.L.; Zhang, M.; Choi, M.; Morin, B.; Cao, Z.; Villablanca, E.J.; Li, C.; Wijmenga, C.; Yun, S.H.; et al. Card9 mediates intestinal epithelial cell restitution, T-helper 17 responses, and control of bacterial infection in mice. Gastroenterology 2013, 145, 591–601.e593. [Google Scholar] [CrossRef]
- Lamas, B.; Richard, M.L.; Leducq, V.; Pham, H.P.; Michel, M.L.; Da Costa, G.; Bridonneau, C.; Jegou, S.; Hoffmann, T.W.; Natividad, J.M.; et al. CARD9 impacts colitis by altering gut microbiota metabolism of tryptophan into aryl hydrocarbon receptor ligands. Nat. Med. 2016, 22, 598–605. [Google Scholar] [CrossRef]
- Danne, C.; Michaudel, C.; Skerniskyte, J.; Planchais, J.; Magniez, A.; Agus, A.; Michel, M.L.; Lamas, B.; Da Costa, G.; Spatz, M.; et al. CARD9 in neutrophils protects from colitis and controls mitochondrial metabolism and cell survival. Gut 2023, 72, 1081–1092. [Google Scholar] [CrossRef] [PubMed]
- Fournier, B.M.; Parkos, C.A. The role of neutrophils during intestinal inflammation. Mucosal. Immunol. 2012, 5, 354–366. [Google Scholar] [CrossRef] [PubMed]
- Chen, Y.H.; Yeung, F.; Lacey, K.A.; Zaldana, K.; Lin, J.D.; Bee, G.C.W.; McCauley, C.; Barre, R.S.; Liang, S.H.; Hansen, C.B.; et al. Rewilding of laboratory mice enhances granulopoiesis and immunity through intestinal fungal colonization. Sci. Immunol. 2023, 8, eadd6910. [Google Scholar] [CrossRef] [PubMed]
- Pelletier, M.; Maggi, L.; Micheletti, A.; Lazzeri, E.; Tamassia, N.; Costantini, C.; Cosmi, L.; Lunardi, C.; Annunziato, F.; Romagnani, S.; et al. Evidence for a cross-talk between human neutrophils and Th17 cells. Blood 2010, 115, 335–343. [Google Scholar] [CrossRef] [PubMed]
- Kvedaraite, E. Neutrophil-T cell crosstalk in inflammatory bowel disease. Immunology 2021, 164, 657–664. [Google Scholar] [CrossRef]
- Swidergall, M.; Khalaji, M.; Solis, N.V.; Moyes, D.L.; Drummond, R.A.; Hube, B.; Lionakis, M.S.; Murdoch, C.; Filler, S.G.; Naglik, J.R. Candidalysin Is Required for Neutrophil Recruitment and Virulence During Systemic Candida albicans Infection. J. Infect Dis. 2019, 220, 1477–1488. [Google Scholar] [CrossRef]
- Gander-Bui, H.T.T.; Schlafli, J.; Baumgartner, J.; Walthert, S.; Genitsch, V.; van Geest, G.; Galvan, J.A.; Cardozo, C.; Graham Martinez, C.; Grans, M.; et al. Targeted removal of macrophage-secreted interleukin-1 receptor antagonist protects against lethal Candida albicans sepsis. Immunity 2023, 56, 1743–1760.e9. [Google Scholar] [CrossRef]
- Kusaka, S.; Nishida, A.; Takahashi, K.; Bamba, S.; Yasui, H.; Kawahara, M.; Inatomi, O.; Sugimoto, M.; Andoh, A. Expression of human cathelicidin peptide LL-37 in inflammatory bowel disease. Clin. Exp. Immunol. 2018, 191, 96–106. [Google Scholar] [CrossRef]
- Urban, C.F.; Ermert, D.; Schmid, M.; Abu-Abed, U.; Goosmann, C.; Nacken, W.; Brinkmann, V.; Jungblut, P.R.; Zychlinsky, A. Neutrophil extracellular traps contain calprotectin, a cytosolic protein complex involved in host defense against Candida albicans. PLoS Pathog. 2009, 5, e1000639. [Google Scholar] [CrossRef]
- Branzk, N.; Lubojemska, A.; Hardison, S.E.; Wang, Q.; Gutierrez, M.G.; Brown, G.D.; Papayannopoulos, V. Neutrophils sense microbe size and selectively release neutrophil extracellular traps in response to large pathogens. Nat. Immunol. 2014, 15, 1017–1025. [Google Scholar] [CrossRef]
- Steffen, U.; Koeleman, C.A.; Sokolova, M.V.; Bang, H.; Kleyer, A.; Rech, J.; Unterweger, H.; Schicht, M.; Garreis, F.; Hahn, J.; et al. IgA subclasses have different effector functions associated with distinct glycosylation profiles. Nat. Commun. 2020, 11, 120. [Google Scholar] [CrossRef]
- Canales-Herrerias, P.; Garcia-Carmona, Y.; Shang, J.; Meringer, H.; Yee, D.S.; Radigan, L.; Buta, S.; Martinez-Delgado, G.; Tankelevich, M.; Helmus, D.; et al. Selective IgA2 deficiency in a patient with small intestinal Crohn’s disease. J. Clin. Investig. 2023, 133, e167742. [Google Scholar] [CrossRef] [PubMed]
- Urban, C.F.; Reichard, U.; Brinkmann, V.; Zychlinsky, A. Neutrophil extracellular traps capture and kill Candida albicans yeast and hyphal forms. Cell Microbiol. 2006, 8, 668–676. [Google Scholar] [CrossRef] [PubMed]
- Li, T.; Wang, C.; Liu, Y.; Li, B.; Zhang, W.; Wang, L.; Yu, M.; Zhao, X.; Du, J.; Zhang, J.; et al. Neutrophil Extracellular Traps Induce Intestinal Damage and Thrombotic Tendency in Inflammatory Bowel Disease. J. Crohns Colitis 2020, 14, 240–253. [Google Scholar] [CrossRef] [PubMed]
- Dinallo, V.; Marafini, I.; Di Fusco, D.; Laudisi, F.; Franze, E.; Di Grazia, A.; Figliuzzi, M.M.; Caprioli, F.; Stolfi, C.; Monteleone, I.; et al. Neutrophil Extracellular Traps Sustain Inflammatory Signals in Ulcerative Colitis. J. Crohns Colitis 2019, 13, 772–784. [Google Scholar] [CrossRef]
- Neri, B.; Mossa, M.; Scucchi, L.; Sena, G.; Palmieri, G.; Biancone, L. Histological scores in inflammatory bowel disease. J. Dig. Dis. 2021, 22, 9–22. [Google Scholar] [CrossRef]
- Konikoff, M.R.; Denson, L.A. Role of fecal calprotectin as a biomarker of intestinal inflammation in inflammatory bowel disease. Inflamm. Bowel Dis. 2006, 12, 524–534. [Google Scholar] [CrossRef]
- Caer, C.; Wick, M.J. Human Intestinal Mononuclear Phagocytes in Health and Inflammatory Bowel Disease. Front. Immunol. 2020, 11, 410. [Google Scholar] [CrossRef]
- Muehlhoefer, A.; Saubermann, L.J.; Gu, X.; Luedtke-Heckenkamp, K.; Xavier, R.; Blumberg, R.S.; Podolsky, D.K.; MacDermott, R.P.; Reinecker, H.C. Fractalkine is an epithelial and endothelial cell-derived chemoattractant for intraepithelial lymphocytes in the small intestinal mucosa. J. Immunol. 2000, 164, 3368–3376. [Google Scholar] [CrossRef]
- Sans, M.; Danese, S.; de la Motte, C.; de Souza, H.S.; Rivera-Reyes, B.M.; West, G.A.; Phillips, M.; Katz, J.A.; Fiocchi, C. Enhanced recruitment of CX3CR1+ T cells by mucosal endothelial cell-derived fractalkine in inflammatory bowel disease. Gastroenterology 2007, 132, 139–153. [Google Scholar] [CrossRef]
- Bernardo, D.; Marin, A.C.; Fernandez-Tome, S.; Montalban-Arques, A.; Carrasco, A.; Tristan, E.; Ortega-Moreno, L.; Mora-Gutierrez, I.; Diaz-Guerra, A.; Caminero-Fernandez, R.; et al. Human intestinal pro-inflammatory CD11c(high)CCR2(+)CX3CR1(+) macrophages, but not their tolerogenic CD11c(-)CCR2(-)CX3CR1(-) counterparts, are expanded in inflammatory bowel disease. Mucosal Immunol. 2018, 11, 1114–1126. [Google Scholar] [CrossRef] [PubMed]
- Kuboi, Y.; Nishimura, M.; Ikeda, W.; Nakatani, T.; Seki, Y.; Yamaura, Y.; Ogawa, K.; Hamaguchi, A.; Muramoto, K.; Mizuno, K.; et al. Blockade of the fractalkine-CX3CR1 axis ameliorates experimental colitis by dislodging venous crawling monocytes. Int. Immunol. 2019, 31, 287–302. [Google Scholar] [CrossRef] [PubMed]
- Leonardi, I.; Li, X.; Semon, A.; Li, D.; Doron, I.; Putzel, G.; Bar, A.; Prieto, D.; Rescigno, M.; McGovern, D.P.B.; et al. CX3CR1(+) mononuclear phagocytes control immunity to intestinal fungi. Science 2018, 359, 232–236. [Google Scholar] [CrossRef]
- Malik, A.; Sharma, D.; Malireddi, R.K.S.; Guy, C.S.; Chang, T.C.; Olsen, S.R.; Neale, G.; Vogel, P.; Kanneganti, T.D. SYK-CARD9 Signaling Axis Promotes Gut Fungi-Mediated Inflammasome Activation to Restrict Colitis and Colon Cancer. Immunity 2018, 49, 515–530.e515. [Google Scholar] [CrossRef] [PubMed]
- Doron, I.; Leonardi, I.; Li, X.V.; Fiers, W.D.; Semon, A.; Bialt-DeCelie, M.; Migaud, M.; Gao, I.H.; Lin, W.Y.; Kusakabe, T.; et al. Human gut mycobiota tune immunity via CARD9-dependent induction of anti-fungal IgG antibodies. Cell 2021, 184, 1017–1031.e1014. [Google Scholar] [CrossRef]
- Brand, S.; Hofbauer, K.; Dambacher, J.; Schnitzler, F.; Staudinger, T.; Pfennig, S.; Seiderer, J.; Tillack, C.; Konrad, A.; Goke, B.; et al. Increased expression of the chemokine fractalkine in Crohn’s disease and association of the fractalkine receptor T280M polymorphism with a fibrostenosing disease Phenotype. Am. J. Gastroenterol. 2006, 101, 99–106. [Google Scholar] [CrossRef]
- Galvez, J. Role of Th17 Cells in the Pathogenesis of Human IBD. ISRN Inflamm. 2014, 2014, 928461. [Google Scholar] [CrossRef]
- Fuss, I.J. Is the Th1/Th2 paradigm of immune regulation applicable to IBD? Inflamm. Bowel Dis. 2008, 14 (Suppl. 2), S110–S112. [Google Scholar] [CrossRef]
- Puel, A.; Cypowyj, S.; Marodi, L.; Abel, L.; Picard, C.; Casanova, J.L. Inborn errors of human IL-17 immunity underlie chronic mucocutaneous candidiasis. Curr. Opin. Allergy Clin. Immunol. 2012, 12, 616–622. [Google Scholar] [CrossRef]
- Levy, R.; Okada, S.; Beziat, V.; Moriya, K.; Liu, C.; Chai, L.Y.; Migaud, M.; Hauck, F.; Al Ali, A.; Cyrus, C.; et al. Genetic, immunological, and clinical features of patients with bacterial and fungal infections due to inherited IL-17RA deficiency. Proc. Natl. Acad. Sci. USA 2016, 113, E8277–E8285. [Google Scholar] [CrossRef]
- Fujino, S.; Andoh, A.; Bamba, S.; Ogawa, A.; Hata, K.; Araki, Y.; Bamba, T.; Fujiyama, Y. Increased expression of interleukin 17 in inflammatory bowel disease. Gut 2003, 52, 65–70. [Google Scholar] [CrossRef]
- Bacher, P.; Hohnstein, T.; Beerbaum, E.; Rocker, M.; Blango, M.G.; Kaufmann, S.; Rohmel, J.; Eschenhagen, P.; Grehn, C.; Seidel, K.; et al. Human Anti-fungal Th17 Immunity and Pathology Rely on Cross-Reactivity against Candida albicans. Cell 2019, 176, 1340–1355.e1315. [Google Scholar] [CrossRef]
- Caprioli, F.; Pallone, F.; Monteleone, G. Th17 immune response in IBD: A new pathogenic mechanism. J. Crohns Colitis 2008, 2, 291–295. [Google Scholar] [CrossRef] [PubMed]
- Atarashi, K.; Tanoue, T.; Ando, M.; Kamada, N.; Nagano, Y.; Narushima, S.; Suda, W.; Imaoka, A.; Setoyama, H.; Nagamori, T.; et al. Th17 Cell Induction by Adhesion of Microbes to Intestinal Epithelial Cells. Cell 2015, 163, 367–380. [Google Scholar] [CrossRef] [PubMed]
- Jiang, W.; Su, J.; Zhang, X.; Cheng, X.; Zhou, J.; Shi, R.; Zhang, H. Elevated levels of Th17 cells and Th17-related cytokines are associated with disease activity in patients with inflammatory bowel disease. Inflamm. Res. 2014, 63, 943–950. [Google Scholar] [CrossRef]
- Chassaing, B.; Aitken, J.D.; Malleshappa, M.; Vijay-Kumar, M. Dextran sulfate sodium (DSS)-induced colitis in mice. Curr. Protoc. Immunol. 2014, 104, 15–25. [Google Scholar] [CrossRef] [PubMed]
- Prieto, D.; Correia, I.; Pla, J.; Roman, E. Adaptation of Candida albicans to commensalism in the gut. Future Microbiol. 2016, 11, 567–583. [Google Scholar] [CrossRef]
- Machata, S.; Sreekantapuram, S.; Hunniger, K.; Kurzai, O.; Dunker, C.; Schubert, K.; Kruger, W.; Schulze-Richter, B.; Speth, C.; Rambach, G.; et al. Significant Differences in Host-Pathogen Interactions Between Murine and Human Whole Blood. Front. Immunol. 2020, 11, 565869. [Google Scholar] [CrossRef]
- Yang, X.O.; Chang, S.H.; Park, H.; Nurieva, R.; Shah, B.; Acero, L.; Wang, Y.H.; Schluns, K.S.; Broaddus, R.R.; Zhu, Z.; et al. Regulation of inflammatory responses by IL-17F. J. Exp. Med. 2008, 205, 1063–1075. [Google Scholar] [CrossRef]
- Jawhara, S.; Thuru, X.; Standaert-Vitse, A.; Jouault, T.; Mordon, S.; Sendid, B.; Desreumaux, P.; Poulain, D. Colonization of mice by Candida albicans is promoted by chemically induced colitis and augments inflammatory responses through galectin-3. J. Infect. Dis. 2008, 197, 972–980. [Google Scholar] [CrossRef]
- Gehlhaar, A.; Inala, A.; Llivichuzhca-Loja, D.; Silva, T.N.; Adegboye, C.Y.; O’Connell, A.E.; Konnikova, L. Insights into the Role of Commensal-Specific T Cells in Intestinal Inflammation. J. Inflamm. Res. 2022, 15, 1873–1887. [Google Scholar] [CrossRef]
- Hegazy, A.N.; West, N.R.; Stubbington, M.J.T.; Wendt, E.; Suijker, K.I.M.; Datsi, A.; This, S.; Danne, C.; Campion, S.; Duncan, S.H.; et al. Circulating and Tissue-Resident CD4(+) T Cells With Reactivity to Intestinal Microbiota Are Abundant in Healthy Individuals and Function Is Altered During Inflammation. Gastroenterology 2017, 153, 1320–1337.e1316. [Google Scholar] [CrossRef] [PubMed]
- Martini, G.R.; Tikhonova, E.; Rosati, E.; DeCelie, M.B.; Sievers, L.K.; Tran, F.; Lessing, M.; Bergfeld, A.; Hinz, S.; Nikolaus, S.; et al. Selection of cross-reactive T cells by commensal and food-derived yeasts drives cytotoxic T(H)1 cell responses in Crohn’s disease. Nat. Med. 2023, 29, 2602–2614. [Google Scholar] [CrossRef] [PubMed]
- Hackstein, C.P.; Costigan, D.; Drexhage, L.; Pearson, C.; Bullers, S.; Ilott, N.; Akther, H.D.; Gu, Y.; FitzPatrick, M.E.B.; Harrison, O.J.; et al. A conserved population of MHC II-restricted, innate-like, commensal-reactive T cells in the gut of humans and mice. Nat. Commun. 2022, 13, 7472. [Google Scholar] [CrossRef] [PubMed]
- Kleinschek, M.A.; Boniface, K.; Sadekova, S.; Grein, J.; Murphy, E.E.; Turner, S.P.; Raskin, L.; Desai, B.; Faubion, W.A.; de Waal Malefyt, R.; et al. Circulating and gut-resident human Th17 cells express CD161 and promote intestinal inflammation. J. Exp. Med. 2009, 206, 525–534. [Google Scholar] [CrossRef]
- McCarthy, N.E.; Eberl, M. Human gammadelta T-Cell Control of Mucosal Immunity and Inflammation. Front. Immunol. 2018, 9, 985. [Google Scholar] [CrossRef]
- Catalan-Serra, I.; Sandvik, A.K.; Bruland, T.; Andreu-Ballester, J.C. Gammadelta T Cells in Crohn’s Disease: A New Player in the Disease Pathogenesis? J. Crohns Colitis 2017, 11, 1135–1145. [Google Scholar] [CrossRef]
- Ali, Z.; Shao, L.; Halliday, L.; Reichenberg, A.; Hintz, M.; Jomaa, H.; Chen, Z.W. Prolonged (E)-4-hydroxy-3-methyl-but-2-enyl pyrophosphate-driven antimicrobial and cytotoxic responses of pulmonary and systemic Vgamma2Vdelta2 T cells in macaques. J. Immunol. 2007, 179, 8287–8296. [Google Scholar] [CrossRef]
- Lo Presti, E.; Di Mitri, R.; Mocciaro, F.; Di Stefano, A.B.; Scibetta, N.; Unti, E.; Cicero, G.; Pecoraro, G.; Conte, E.; Dieli, F.; et al. Characterization of gammadelta T Cells in Intestinal Mucosa From Patients With Early-Onset or Long-Standing Inflammatory Bowel Disease and Their Correlation with Clinical Status. J. Crohns Colitis 2019, 13, 873–883. [Google Scholar] [CrossRef]
- McCarthy, N.E.; Bashir, Z.; Vossenkamper, A.; Hedin, C.R.; Giles, E.M.; Bhattacharjee, S.; Brown, S.G.; Sanders, T.J.; Whelan, K.; MacDonald, T.T.; et al. Proinflammatory Vdelta2+ T cells populate the human intestinal mucosa and enhance IFN-gamma production by colonic alphabeta T cells. J. Immunol. 2013, 191, 2752–2763. [Google Scholar] [CrossRef]
- McCarthy, N.E.; Hedin, C.R.; Sanders, T.J.; Amon, P.; Hoti, I.; Ayada, I.; Baji, V.; Giles, E.M.; Wildemann, M.; Bashir, Z.; et al. Azathioprine therapy selectively ablates human Vdelta2(+) T cells in Crohn’s disease. J. Clin. Investig. 2015, 125, 3215–3225. [Google Scholar] [CrossRef] [PubMed]
- Steenholt, J.V.; Nielsen, C.; Baudewijn, L.; Staal, A.; Rasmussen, K.S.; Sabir, H.J.; Barington, T.; Husby, S.; Toft-Hansen, H. The composition of T cell subtypes in duodenal biopsies are altered in coeliac disease patients. PLoS ONE 2017, 12, e0170270. [Google Scholar] [CrossRef] [PubMed]
- Maher, C.O.; Dunne, K.; Comerford, R.; O’Dea, S.; Loy, A.; Woo, J.; Rogers, T.R.; Mulcahy, F.; Dunne, P.J.; Doherty, D.G. Candida albicans stimulates IL-23 release by human dendritic cells and downstream IL-17 secretion by Vdelta1 T cells. J. Immunol. 2015, 194, 5953–5960. [Google Scholar] [CrossRef] [PubMed]
- Conti, H.R.; Peterson, A.C.; Brane, L.; Huppler, A.R.; Hernandez-Santos, N.; Whibley, N.; Garg, A.V.; Simpson-Abelson, M.R.; Gibson, G.A.; Mamo, A.J.; et al. Oral-resident natural Th17 cells and gammadelta T cells control opportunistic Candida albicans infections. J. Exp. Med. 2014, 211, 2075–2084. [Google Scholar] [CrossRef]
- Aggor, F.E.Y.; Break, T.J.; Trevejo-Nunez, G.; Whibley, N.; Coleman, B.M.; Bailey, R.D.; Kaplan, D.H.; Naglik, J.R.; Shan, W.; Shetty, A.C.; et al. Oral epithelial IL-22/STAT3 signaling licenses IL-17-mediated immunity to oral mucosal candidiasis. Sci. Immunol. 2020, 5, eaba0570. [Google Scholar] [CrossRef] [PubMed]
- Singh, S.; Murad, M.H.; Fumery, M.; Sedano, R.; Jairath, V.; Panaccione, R.; Sandborn, W.J.; Ma, C. Comparative efficacy and safety of biologic therapies for moderate-to-severe Crohn’s disease: A systematic review and network meta-analysis. Lancet Gastroenterol. Hepatol. 2021, 6, 1002–1014. [Google Scholar] [CrossRef] [PubMed]
- Jain, U.; Ver Heul, A.M.; Xiong, S.; Gregory, M.H.; Demers, E.G.; Kern, J.T.; Lai, C.W.; Muegge, B.D.; Barisas, D.A.G.; Leal-Ekman, J.S.; et al. Debaryomyces is enriched in Crohn’s disease intestinal tissue and impairs healing in mice. Science 2021, 371, 1154–1159. [Google Scholar] [CrossRef]
- Break, T.J.; Oikonomou, V.; Dutzan, N.; Desai, J.V.; Swidergall, M.; Freiwald, T.; Chauss, D.; Harrison, O.J.; Alejo, J.; Williams, D.W.; et al. Aberrant type 1 immunity drives susceptibility to mucosal fungal infections. Science 2021, 371, eaay5731. [Google Scholar] [CrossRef]
- Puel, A.; Casanova, J.L. Comment on “Aberrant type 1 immunity drives susceptibility to mucosal fungal infections”. Science 2021, 373, eabi5459. [Google Scholar] [CrossRef]
- Kisand, K.; Meager, A.; Hayday, A.; Willcox, N. Comment on “Aberrant type 1 immunity drives susceptibility to mucosal fungal infections”. Science 2021, 373, eabi6235. [Google Scholar] [CrossRef]
- Break, T.J.; Oikonomou, V.; Dutzan, N.; Desai, J.V.; Swidergall, M.; Freiwald, T.; Chauss, D.; Harrison, O.J.; Alejo, J.; Williams, D.W.; et al. Response to Comments on “Aberrant type 1 immunity drives susceptibility to mucosal fungal infections”. Science 2021, 373, eabi8835. [Google Scholar] [CrossRef]
- Philippot, Q.; Casanova, J.L.; Puel, A. Candidiasis in patients with APS-1: Low IL-17, high IFN-gamma, or both? Curr. Opin. Immunol. 2021, 72, 318–323. [Google Scholar] [CrossRef] [PubMed]
- El Hage Chehade, N.; Ghoneim, S.; Shah, S.; Chahine, A.; Mourad, F.H.; Francis, F.F.; Binion, D.G.; Farraye, F.A.; Hashash, J.G. Efficacy of Fecal Microbiota Transplantation in the Treatment of Active Ulcerative Colitis: A Systematic Review and Meta-Analysis of Double-Blind Randomized Controlled Trials. Inflamm. Bowel Dis. 2022, 29, 808–817. [Google Scholar] [CrossRef] [PubMed]
- Fehily, S.R.; Basnayake, C.; Wright, E.K.; Kamm, M.A. Fecal microbiota transplantation therapy in Crohn’s disease: Systematic review. J. Gastroenterol. Hepatol. 2021, 36, 2672–2686. [Google Scholar] [CrossRef]
- Kedia, S.; Virmani, S.; Vuyyuru, S.K.; Kumar, P.; Kante, B.; Sahu, P.; Kaushal, K.; Farooqui, M.; Singh, M.; Verma, M.; et al. Faecal microbiota transplantation with anti-inflammatory diet (FMT-AID) followed by anti-inflammatory diet alone is effective in inducing and maintaining remission over 1 year in mild to moderate ulcerative colitis: A randomised controlled trial. Gut 2022, 71, 2401–2413. [Google Scholar] [CrossRef] [PubMed]
- Quraishi, M.N.; Widlak, M.; Bhala, N.; Moore, D.; Price, M.; Sharma, N.; Iqbal, T.H. Systematic review with meta-analysis: The efficacy of faecal microbiota transplantation for the treatment of recurrent and refractory Clostridium difficile infection. Aliment Pharmacol. Ther. 2017, 46, 479–493. [Google Scholar] [CrossRef]
- Imdad, A.; Nicholson, M.R.; Tanner-Smith, E.E.; Zackular, J.P.; Gomez-Duarte, O.G.; Beaulieu, D.B.; Acra, S. Fecal transplantation for treatment of inflammatory bowel disease. Cochrane Database Syst. Rev. 2018, 11, CD012774. [Google Scholar] [CrossRef]
- Moayyedi, P.; Surette, M.G.; Kim, P.T.; Libertucci, J.; Wolfe, M.; Onischi, C.; Armstrong, D.; Marshall, J.K.; Kassam, Z.; Reinisch, W.; et al. Fecal Microbiota Transplantation Induces Remission in Patients With Active Ulcerative Colitis in a Randomized Controlled Trial. Gastroenterology 2015, 149, 102–109.e106. [Google Scholar] [CrossRef]
- Paramsothy, S.; Kamm, M.A.; Kaakoush, N.O.; Walsh, A.J.; van den Bogaerde, J.; Samuel, D.; Leong, R.W.L.; Connor, S.; Ng, W.; Paramsothy, R.; et al. Multidonor intensive faecal microbiota transplantation for active ulcerative colitis: A randomised placebo-controlled trial. Lancet 2017, 389, 1218–1228. [Google Scholar] [CrossRef]
- Fuentes, S.; Rossen, N.G.; van der Spek, M.J.; Hartman, J.H.; Huuskonen, L.; Korpela, K.; Salojarvi, J.; Aalvink, S.; de Vos, W.M.; D’Haens, G.R.; et al. Microbial shifts and signatures of long-term remission in ulcerative colitis after faecal microbiota transplantation. ISME J. 2017, 11, 1877–1889. [Google Scholar] [CrossRef]
- Paramsothy, S.; Nielsen, S.; Kamm, M.A.; Deshpande, N.P.; Faith, J.J.; Clemente, J.C.; Paramsothy, R.; Walsh, A.J.; van den Bogaerde, J.; Samuel, D.; et al. Specific Bacteria and Metabolites Associated With Response to Fecal Microbiota Transplantation in Patients With Ulcerative Colitis. Gastroenterology 2019, 156, 1440–1454.e1442. [Google Scholar] [CrossRef] [PubMed]
- Leonardi, I.; Paramsothy, S.; Doron, I.; Semon, A.; Kaakoush, N.O.; Clemente, J.C.; Faith, J.J.; Borody, T.J.; Mitchell, H.M.; Colombel, J.F.; et al. Fungal Trans-kingdom Dynamics Linked to Responsiveness to Fecal Microbiota Transplantation (FMT) Therapy in Ulcerative Colitis. Cell Host Microbe 2020, 27, 823–829.e823. [Google Scholar] [CrossRef] [PubMed]
- Sendid, B.; Salvetat, N.; Sarter, H.; Loridant, S.; Cunisse, C.; Francois, N.; Aijjou, R.; Gele, P.; Leroy, J.; Deplanque, D.; et al. A Pilot Clinical Study on Post-Operative Recurrence Provides Biological Clues for a Role of Candida Yeasts and Fluconazole in Crohn’s Disease. J. Fungi 2021, 7, 324. [Google Scholar] [CrossRef] [PubMed]
- Zwolinska-Wcislo, M.; Brzozowski, T.; Budak, A.; Kwiecien, S.; Sliwowski, Z.; Drozdowicz, D.; Trojanowska, D.; Rudnicka-Sosin, L.; Mach, T.; Konturek, S.J.; et al. Effect of Candida colonization on human ulcerative colitis and the healing of inflammatory changes of the colon in the experimental model of colitis ulcerosa. J. Physiol. Pharmacol. 2009, 60, 107–118. [Google Scholar]
- Jena, A.; Dutta, U.; Shah, J.; Sharma, V.; Prasad, K.K.; Shivaprakash, R.M.; Mandavdhare, H.S.; Samanta, J.; Sharma, P.; Popli, P.; et al. Oral Fluconazole Therapy in Patients With Active Ulcerative Colitis Who Have Detectable Candida in the Stool: A Double-Blind Randomized Placebo-controlled Trial. J. Clin. Gastroenterol. 2022, 56, 705–711. [Google Scholar] [CrossRef]
- Edwards, J.E., Jr.; Schwartz, M.M.; Schmidt, C.S.; Sobel, J.D.; Nyirjesy, P.; Schodel, F.; Marchus, E.; Lizakowski, M.; DeMontigny, E.A.; Hoeg, J.; et al. A Fungal Immunotherapeutic Vaccine (NDV-3A) for Treatment of Recurrent Vulvovaginal Candidiasis-A Phase 2 Randomized, Double-Blind, Placebo-Controlled Trial. Clin. Infect. Dis. 2018, 66, 1928–1936. [Google Scholar] [CrossRef]
- Cao, Z.; Conway, K.L.; Heath, R.J.; Rush, J.S.; Leshchiner, E.S.; Ramirez-Ortiz, Z.G.; Nedelsky, N.B.; Huang, H.; Ng, A.; Gardet, A.; et al. Ubiquitin Ligase TRIM62 Regulates CARD9-Mediated Anti-fungal Immunity and Intestinal Inflammation. Immunity 2015, 43, 715–726. [Google Scholar] [CrossRef]
- Leshchiner, E.S.; Rush, J.S.; Durney, M.A.; Cao, Z.; Dancik, V.; Chittick, B.; Wu, H.; Petrone, A.; Bittker, J.A.; Phillips, A.; et al. Small-molecule inhibitors directly target CARD9 and mimic its protective variant in inflammatory bowel disease. Proc. Natl. Acad. Sci. USA 2017, 114, 11392–11397. [Google Scholar] [CrossRef]
- Hueber, W.; Sands, B.E.; Lewitzky, S.; Vandemeulebroecke, M.; Reinisch, W.; Higgins, P.D.; Wehkamp, J.; Feagan, B.G.; Yao, M.D.; Karczewski, M.; et al. Secukinumab, a human anti-IL-17A monoclonal antibody, for moderate to severe Crohn’s disease: Unexpected results of a randomised, double-blind placebo-controlled trial. Gut 2012, 61, 1693–1700. [Google Scholar] [CrossRef]
- Targan, S.R.; Feagan, B.; Vermeire, S.; Panaccione, R.; Melmed, G.Y.; Landers, C.; Li, D.; Russell, C.; Newmark, R.; Zhang, N.; et al. A Randomized, Double-Blind, Placebo-Controlled Phase 2 Study of Brodalumab in Patients With Moderate-to-Severe Crohn’s Disease. Am. J. Gastroenterol. 2016, 111, 1599–1607. [Google Scholar] [CrossRef]
- Colombel, J.F.; Sendid, B.; Jouault, T.; Poulain, D. Secukinumab failure in Crohn’s disease: The yeast connection? Gut 2013, 62, 800–801. [Google Scholar] [CrossRef] [PubMed]
- Langley, R.G.; Elewski, B.E.; Lebwohl, M.; Reich, K.; Griffiths, C.E.; Papp, K.; Puig, L.; Nakagawa, H.; Spelman, L.; Sigurgeirsson, B.; et al. Secukinumab in plaque psoriasis—Results of two phase 3 trials. N. Engl. J. Med. 2014, 371, 326–338. [Google Scholar] [CrossRef]
- Deng, Z.; Wang, S.; Wu, C.; Wang, C. IL-17 inhibitor-associated inflammatory bowel disease: A study based on literature and database analysis. Front. Pharmacol. 2023, 14, 1124628. [Google Scholar] [CrossRef] [PubMed]
- Davidson, L.; van den Reek, J.; Bruno, M.; van Hunsel, F.; Herings, R.M.C.; Matzaraki, V.; Boahen, C.K.; Kumar, V.; Groenewoud, H.M.M.; van de Veerdonk, F.L.; et al. Risk of candidiasis associated with interleukin-17 inhibitors: A real-world observational study of multiple independent sources. Lancet Reg. Health Eur. 2022, 13, 100266. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Carlson, S.L.; Mathew, L.; Savage, M.; Kok, K.; Lindsay, J.O.; Munro, C.A.; McCarthy, N.E. Mucosal Immunity to Gut Fungi in Health and Inflammatory Bowel Disease. J. Fungi 2023, 9, 1105. https://doi.org/10.3390/jof9111105
Carlson SL, Mathew L, Savage M, Kok K, Lindsay JO, Munro CA, McCarthy NE. Mucosal Immunity to Gut Fungi in Health and Inflammatory Bowel Disease. Journal of Fungi. 2023; 9(11):1105. https://doi.org/10.3390/jof9111105
Chicago/Turabian StyleCarlson, Sean L., Liya Mathew, Michael Savage, Klaartje Kok, James O. Lindsay, Carol A. Munro, and Neil E. McCarthy. 2023. "Mucosal Immunity to Gut Fungi in Health and Inflammatory Bowel Disease" Journal of Fungi 9, no. 11: 1105. https://doi.org/10.3390/jof9111105
APA StyleCarlson, S. L., Mathew, L., Savage, M., Kok, K., Lindsay, J. O., Munro, C. A., & McCarthy, N. E. (2023). Mucosal Immunity to Gut Fungi in Health and Inflammatory Bowel Disease. Journal of Fungi, 9(11), 1105. https://doi.org/10.3390/jof9111105