Relationships Between Vitamin D, Gut Microbiome, and Systemic Autoimmunity

Erin Yamamoto1, Trine N. Jørgensen2
1Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, United States
2Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States

Tóm tắt

Từ khóa


Tài liệu tham khảo

Hayter, 2012, Updated assessment of the prevalence, spectrum and case definition of autoimmune disease, Autoimmun Rev., 11, 754, 10.1016/j.autrev.2012.02.001

Hart, 2011, Modulation of the immune system by UV radiation: more than just the effects of vitamin D?, Nat Rev Immunol., 11, 584, 10.1038/nri3045

Yamamoto, 2019, Immunological effects of vitamin D and their relations to autoimmunity, J Autoimmun., 100, 7, 10.1016/j.jaut.2019.03.002

Rogers, 2016, Chapter 2: genomic technologies in medicine and health: past, present, and future, Medical and Health Genomics, 15, 10.1016/B978-0-12-420196-5.00002-2

Hugon, 2015, A comprehensive repertoire of prokaryotic species identified in human beings, Lancet Infect Dis., 15, 1211, 10.1016/S1473-3099(15)00293-5

Li, 2014, An integrated catalog of reference genes in the human gut microbiome, Nat. Biotechnol., 32, 834, 10.1038/nbt.2942

Org, 2015, Unraveling the environmental and genetic interactions in atherosclerosis: central role of the gut microbiota, Atherosclerosis., 241, 387, 10.1016/j.atherosclerosis.2015.05.035

Wood, 2015, Gut reactions—can changes in the intestinal microbiome provide new insights into Parkinson disease?, Nat Rev Neurol., 11, 66, 10.1038/nrneurol.2014.256

Gomez, 2015, The gut microbiome in autoimmunity: sex matters, Clin Immunol., 159, 154, 10.1016/j.clim.2015.04.016

Chelakkot, 2018, Mechanisms regulating intestinal barrier integrity and its pathological implications, Exp Mol Med., 50, 103, 10.1038/s12276-018-0126-x

Pabst, 2012, New concepts in the generation and functions of IgA, Nat Rev Immunol., 12, 821, 10.1038/nri3322

Iwasaki, 2007, Mucosal dendritic cells, Annu Rev Immunol., 25, 381, 10.1146/annurev.immunol.25.022106.141634

Gommerman, 2014, Re-thinking the functions of IgA(+) plasma cells, Gut Microbes., 5, 652, 10.4161/19490976.2014.969977

Abt, 2012, Commensal bacteria calibrate the activation threshold of innate antiviral immunity, Immunity., 37, 158, 10.1016/j.immuni.2012.04.011

Shaw, 2012, Microbiota-induced IL-1β, but not IL-6, is critical for the development of steady-state T H 17 cells in the intestine, J Exp Med., 209, 251, 10.1084/jem.20111703

Ivanov, 2011, Modulation of immune homeostasis by commensal bacteria, Curr Opin Microbiol., 14, 106, 10.1016/j.mib.2010.12.003

Ivanov, 2009, Induction of intestinal Th17 cells by segmented filamentous bacteria, Cell., 139, 485, 10.1016/j.cell.2009.09.033

Wu, 2010, Gut-residing segmented filamentous bacteria drive autoimmune arthritis via T helper 17 cells, Immunity., 32, 815, 10.1016/j.immuni.2010.06.001

Gaboriau-Routhiau, 2009, The key role of segmented filamentous bacteria in the coordinated maturation of gut helper T cell responses, Immunity., 31, 677, 10.1016/j.immuni.2009.08.020

Atarashi, 2015, Th17 cell induction by adhesion of microbes to intestinal epithelial cells, Cell., 163, 367, 10.1016/j.cell.2015.08.058

Schade, 2016, Cell wall glycopolymers of Firmicutes and their role as nonprotein adhesins, FEBS Lett., 590, 3758, 10.1002/1873-3468.12288

Longman, 2015, The functional impact of the intestinal microbiome on mucosal immunity and systemic autoimmunity, Curr Opin Rheumatol., 27, 381, 10.1097/BOR.0000000000000190

Atarashi, 2013, Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota, Nature., 500, 232, 10.1038/nature12331

Atarashi, 2011, Induction of colonic regulatory T cells by indigenous clostridium species, Science., 331, 337, 10.1126/science.1198469

Wesemann, 2013, Microbial colonization influences early B-lineage development in the gut lamina propria, Nature., 501, 112, 10.1038/nature12496

Crabbé, 1970, Immunohistochemical observations on lymphoid tissues from conventional and germ-free mice, Lab Invest., 22, 448

Moreau, 1978, Increase in the population of duodenal immunoglobulin A plasmocytes in axenic mice associated with different living or dead bacterial strains of intestinal origin, Infect Immun., 21, 532, 10.1128/IAI.21.2.532-539.1978

Rosser, 2014, Regulatory B cells are induced by gut microbiota-driven interleukin-1β and interleukin-6 production, Nat Med., 20, 1334, 10.1038/nm.3680

Ramakrishna, 2019, Bacteroides fragilis polysaccharide A induces IL-10 secreting B and T cells that prevent viral encephalitis, Nat Commun., 10, 2153, 10.1038/s41467-019-09884-6

Mishima, 2019, Microbiota maintain colonic homeostasis by activating TLR2/MyD88/PI3K signaling in IL-10–producing regulatory B cells, J Clin Invest., 129, 3702, 10.1172/JCI93820

Mishima, 2015, Resident bacteria-stimulated IL-10-secreting B cells ameliorate T cell-mediated colitis by inducing Tr-1 cells that require IL-27-signaling, Cell Mol Gastroenterol Hepatol., 1, 295, 10.1016/j.jcmgh.2015.01.002

Wexler, 2017, An insider's perspective: bacteroides as a window into the microbiome, Nat Microbiol., 2, 17026, 10.1038/nmicrobiol.2017.26

Dasgupta, 2014, Plasmacytoid dendritic cells mediate anti-inflammatory responses to a gut commensal molecule via both innate and adaptive mechanisms, Cell Host Microbe, 15, 413, 10.1016/j.chom.2014.03.006

Round, 2009, The gut microbiota shapes intestinal immune responses during health and disease, Nat Rev Immunol., 9, 313, 10.1038/nri2515

Ochoa-Reparaz, 2010, Central nervous system demyelinating disease protection by the human commensal bacteroides fragilis depends on polysaccharide a expression, J Immunol., 185, 4101, 10.4049/jimmunol.1001443

Baker, 2017, Memory B cells are major targets for effective immunotherapy in relapsing multiple sclerosis, EBioMedicine., 16, 41, 10.1016/j.ebiom.2017.01.042

Vinolo, 2011, Regulation of inflammation by short chain fatty acids, Nutrients., 3, 858, 10.3390/nu3100858

Koh, 2016, From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites, Cell., 165, 1332, 10.1016/j.cell.2016.05.041

den Besten, 2013, The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism, J Lipid Res., 54, 2325, 10.1194/jlr.R036012

Arpaia, 2013, Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation, Nature., 504, 451, 10.1038/nature12726

Smith, 2013, The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis, Science., 341, 569, 10.1126/science.1241165

Chang, 2014, The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition, Proc Natl Acad Sci USA., 111, 2247, 10.1073/pnas.1322269111

Braniste, 2014, The gut microbiota influences blood-brain barrier permeability in mice, Sci Transl Med., 6, 263ra158, 10.1126/scitranslmed.3009759

Wang, 2011, The G-protein-coupled bile acid receptor, Gpbar1 (TGR5), negatively regulates hepatic inflammatory response through antagonizing nuclear factor κ light-chain enhancer of activated B cells (NF-κB) in mice, Hepatology., 54, 1421, 10.1002/hep.24525

Biagioli, 2017, The bile acid receptor GPBAR1 regulates the M1/M2 phenotype of intestinal macrophages and activation of GPBAR1 rescues mice from murine colitis, J Immunol., 199, 718, 10.4049/jimmunol.1700183

Melhem, 2019, Metabolite-sensing G protein-coupled receptors connect the diet-microbiota-metabolites axis to inflammatory bowel disease, Cells., 8, 450, 10.3390/cells8050450

Mu, 2017, Antibiotics ameliorate lupus-like symptoms in mice, Sci Rep., 7, 1, 10.1038/s41598-017-14223-0

Ochoa-Reparaz, 2009, Role of gut commensal microflora in the development of experimental autoimmune encephalomyelitis, J Immunol., 183, 6041, 10.4049/jimmunol.0900747

Lee, 2011, Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis, Proc Natl Acad Sci USA., 108, 4615, 10.1073/pnas.1000082107

East, 1969, Autoimmune reactions and malignant changes in germ-free New Zealand Black mice, Clin Exp Immunol., 4, 621

Hernández-Chirlaque, 2016, Germ-free and antibiotic-treated mice are highly susceptible to epithelial injury in DSS colitis, J Crohn's Colitis., 10, 1324, 10.1093/ecco-jcc/jjw096

Liu, 2016, Role of the gut microbiome in modulating arthritis progression in mice, Sci Rep., 6, 30594, 10.1038/srep30594

Pearson, 1963, Adjuvant arthritis induced in germ-free rats, Exp Biol Med., 112, 91, 10.3181/00379727-112-27959

Kohashi, 1979, Susceptibility to adjuvant-induced arthritis among germfree, specific-pathogen-free, and conventional rats, Infect Immun., 26, 791, 10.1128/IAI.26.3.791-794.1979

Miyake, 2015, Dysbiosis in the gut microbiota of patients with multiple sclerosis, with a striking depletion of species belonging to clostridia XIVa and IV clusters B, PLoS ONE., 10, e0137429, 10.1371/journal.pone.0137429

Chen, 2016, Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls, Sci Rep., 6, 1, 10.1038/srep28484

Jangi, 2016, Alterations of the human gut microbiome in multiple sclerosis, Nat Commun., 7, 12015, 10.1038/ncomms12015

Cekanaviciute, 2017, Gut bacteria from multiple sclerosis patients modulate human T cells and exacerbate symptoms in mouse models, Proc Natl Acad Sci USA., 114, 10713, 10.1073/pnas.1711235114

Berer, 2017, Gut microbiota from multiple sclerosis patients enables spontaneous autoimmune encephalomyelitis in mice, Proc Natl Acad Sci USA., 114, 10719, 10.1073/pnas.1711233114

Gevers, 2014, The treatment-naive microbiome in new-onset Crohn's disease, Cell Host Microbe., 15, 382, 10.1016/j.chom.2014.02.005

Ohkusa, 2003, Induction of experimental ulcerative colitis by Fusobacterium varium isolated from colonic mucosa of patients with ulcerative colitis, Gut., 52, 79, 10.1136/gut.52.1.79

Ohkusa, 2009, Commensal bacteria can enter colonic epithelial cells and induce proinflammatory cytokine secretion: a possible pathogenic mechanism of ulcerative colitis, J Med Microbiol., 58, 535, 10.1099/jmm.0.005801-0

Frank, 2011, Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases, Inflamm Bowel Dis., 17, 179, 10.1002/ibd.21339

Martinez-Medina, 2006, Abnormal microbiota composition in the ileocolonic mucosa of Crohn's disease patients as revealed by polymerase chain reaction-denaturing gradient gel electrophoresis, Inflamm Bowel Dis., 12, 1136, 10.1097/01.mib.0000235828.09305.0c

Pascal, 2017, A microbial signature for Crohn's disease, Gut., 66, 813, 10.1136/gutjnl-2016-313235

Chen, 2016, An expansion of rare lineage intestinal microbes characterizes rheumatoid arthritis, Genome Med., 8, 43, 10.1186/s13073-016-0299-7

Zhang, 2015, The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment, Nat Med., 21, 895, 10.1038/nm.3914

Scher, 2013, Expansion of intestinal Prevotella copri correlates with enhanced susceptibility to arthritis, eLife., 2, e01202, 10.7554/eLife.01202.028

Maeda, 2016, Dysbiosis contributes to arthritis development via activation of autoreactive T cells in the intestine, Arthritis Rheumatol., 68, 2646, 10.1002/art.39783

Alpizar-Rodriguez, 2019, Prevotella copri in individuals at risk for rheumatoid arthritis, Ann Rheum Dis., 78, 590, 10.1136/annrheumdis-2018-214514

Pianta, 2017, Evidence of the immune relevance of Prevotella copri, a gut microbe, in patients with rheumatoid arthritis, Arthritis Rheumatol., 69, 964, 10.1002/art.40003

He, 2016, Alterations of the gut microbiome in Chinese patients with systemic lupus erythematosus, Gut Pathog., 8, 64, 10.1186/s13099-016-0146-9

van der Meulen, 2019, Shared gut, but distinct oral microbiota composition in primary Sjögren's syndrome and systemic lupus erythematosus, J Autoimmun., 97, 77, 10.1016/j.jaut.2018.10.009

Varela, 2013, Colonisation by Faecalibacterium prausnitzii and maintenance of clinical remission in patients with ulcerative colitis, Aliment Pharmacol Therap., 38, 151, 10.1111/apt.12365

Hickey, 2015, Colitogenic bacteroides thetaiotaomicron antigens access host immune cells in a sulfatase-dependent manner via outer membrane vesicles, Cell Host Microbe., 17, 672, 10.1016/j.chom.2015.04.002

Zhou, 2016, Lower Level of Bacteroides in the gut microbiota is associated with inflammatory bowel disease: a meta-analysis, BioMed Res Int., 2016, 1, 10.1155/2016/5828959

Hevia, 2014, Intestinal dysbiosis associated with systemic lupus erythematosus, mBio., 5, 1, 10.1128/mBio.01548-14

Rodríguez-Carrio, 2017, Intestinal dysbiosis is associated with altered short-chain fatty acids and serum-free fatty acids in systemic lupus erythematosus, Front Immunol., 8, 23, 10.3389/fimmu.2017.00023

Cantarel, 2015, Gut microbiota in multiple sclerosis, J Invest Med., 63, 729, 10.1097/JIM.0000000000000192

Kostic, 2014, The microbiome in inflammatory bowel disease: current status and the future ahead, Gastroenterology., 146, 1489, 10.1053/j.gastro.2014.02.009

Manichanh, 2012, The gut microbiota in IBD, Nat Rev Gastroenterol Hepatol., 9, 599, 10.1038/nrgastro.2012.152

Martinez, 2008, Unstable composition of the fecal microbiota in ulcerative colitis during clinical remission, Am J Gastroenterol., 103, 643, 10.1111/j.1572-0241.2007.01592.x

Martinez-Medina, 2009, Molecular diversity of Escherichia coli in the human gut: new ecological evidence supporting the role of adherent-invasive E. coli (AIEC) in Crohn's disease, Inflamm Bowel Dis., 15, 872, 10.1002/ibd.20860

Santoru, 2017, Cross sectional evaluation of the gut-microbiome metabolome axis in an Italian cohort of IBD patients, Sci Rep., 7, 9523, 10.1038/s41598-017-10034-5

Sokol, 2008, Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients, Proc Natl Acad Sci USA., 105, 16731, 10.1073/pnas.0804812105

Willing, 2009, Twin studies reveal specific imbalances in the mucosa-associated microbiota of patients with ileal Crohn's disease, Inflamm Bowel Dis., 15, 653, 10.1002/ibd.20783

Morgan, 2012, Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment, Genome Biol., 13, R79, 10.1186/gb-2012-13-9-r79

Lopez-Siles, 2017, Faecalibacterium prausnitzii: from microbiology to diagnostics and prognostics, ISME J., 11, 841, 10.1038/ismej.2016.176

Wrzosek, 2013, Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence the production of mucus glycans and the development of goblet cells in the colonic epithelium of a gnotobiotic model rodent, BMC Biol., 11, 61, 10.1186/1741-7007-11-61

Delday, 2019, Bacteroides thetaiotaomicron ameliorates colon inflammation in preclinical models of Crohn's disease, Inflamm Bowel Dis., 25, 85, 10.1093/ibd/izy281

Bloom, 2011, Commensal bacteroides species induce colitis in host-genotype-specific fashion in a mouse model of inflammatory bowel disease, Cell Host Microbe, 9, 390, 10.1016/j.chom.2011.04.009

Fukuda, 2011, Bifidobacteria can protect from enteropathogenic infection through production of acetate, Nature., 469, 543, 10.1038/nature09646

Burrello, 2018, Therapeutic faecal microbiota transplantation controls intestinal inflammation through IL10 secretion by immune cells, Nat Commun., 9, 5184, 10.1038/s41467-018-07359-8

Burrello, 2019, Fecal microbiota transplantation controls murine chronic intestinal inflammation by modulating immune cell functions and gut microbiota composition, Cells., 8, 517, 10.3390/cells8060517

Moayyedi, 2015, Fecal microbiota transplantation induces remission in patients with active ulcerative colitis in a randomized controlled trial, Gastroenterology., 149, 102, 10.1053/j.gastro.2015.04.001

Rossen, 2015, Findings from a randomized controlled trial of fecal transplantation for patients with ulcerative colitis, Gastroenterology., 149, 110, 10.1053/j.gastro.2015.03.045

Costello, 2017, Op036 Short duration, low intensity pooled faecal microbiota transplantation induces remission in patients with mild-moderately active ulcerative colitis: a randomised controlled trial, J Crohn, 39, 11, 10.1093/ecco-jcc/jjx002.035

Paramsothy, 2017, Faecal microbiota transplantation for inflammatory bowel disease: a systematic review and meta-analysis, J Crohn's Colitis., 11, 1180, 10.1093/ecco-jcc/jjx063

Narula, 2017, Systematic review and meta-analysis, Inflamm Bowel Dis., 23, 1702, 10.1097/MIB.0000000000001228

Shi, 2016, Fecal microbiota transplantation for ulcerative colitis: a systematic review and meta-analysis U, PLoS ONE., 11, e0157259, 10.1371/journal.pone.0157259

Vermeire, 2016, Donor species richness determines faecal microbiota transplantation success in inflammatory bowel disease, J Crohn's Colitis., 10, 387, 10.1093/ecco-jcc/jjv203

Kohashi, 1985, Reverse effect of gram-positive bacteria vs. gram-negative bacteria on adjuvant-induced arthritis in germfree rats, Microbiol Immunol., 29, 487, 10.1111/j.1348-0421.1985.tb00851.x

Eerola, 1994, Intestinal flora in early rheumatoid arthritis, Br J Rheumatol., 33, 1030, 10.1093/rheumatology/33.11.1030

Luo, 2017, Gut microbiota in human systemic lupus erythematosus and a mouse model of lupus A, Appl Environ Microbiol., 84, e02288, 10.1128/AEM.02288-17

Johnson, 2015, Impact of dietary deviation on disease progression and gut microbiome composition in lupus-prone SNF1 mice, Clin Exp Immunol., 181, 323, 10.1111/cei.12609

Manfredo Vieira, 2018, Translocation of a gut pathobiont drives autoimmunity in mice and humans, Science., 359, 1156, 10.1126/science.aar7201

Varrin-Doyer, 2012, Aquaporin 4-specific T cells in neuromyelitis optica exhibit a Th17 bias and recognize Clostridium ABC transporter, Ann Neurol., 72, 53, 10.1002/ana.23651

Luthold, 2017, Gut microbiota interactions with the immunomodulatory role of vitamin D in normal individuals, Metabolism., 69, 76, 10.1016/j.metabol.2017.01.007

Ooi, 2013, Vitamin D regulates the gut microbiome and protects mice from dextran sodium sulfate – induced colitis 1 – 3, J Nutr., 143, 1679, 10.3945/jn.113.180794

Assa, 2014, Vitamin D deficiency promotes epithelial barrier dysfunction and intestinal inflammation, J Infect Dis., 210, 1296, 10.1093/infdis/jiu235

Jin, 2015, Lack of vitamin D receptor causes dysbiosis and changes the functions of the murine intestinal microbiome, Clin Ther., 37, 996, 10.1016/j.clinthera.2015.04.004

Wu, 2015, Intestinal epithelial vitamin D receptor deletion leads to defective autophagy in colitis, Gut., 64, 1082, 10.1136/gutjnl-2014-307436

Wang, 2016, Genome-wide association analysis identifies variation in vitamin D receptor and other host factors influencing the gut microbiota, Nat Genet., 48, 1396, 10.1038/ng.3695

Wu, 2011, Linking long-term dietary patterns with gut microbial enterotypes, Science., 334, 105, 10.1126/science.1208344

Bashir, 2016, Effects of high doses of vitamin D 3 on mucosa - associated gut microbiome vary between regions of the human gastrointestinal tract, Eur J Nutr., 55, 1479, 10.1007/s00394-015-0966-2

Seura, 2017, The relationship between habitual dietary intake and gut microbiota in young Japanese women, J Nutr Sci Vitaminol., 63, 396, 10.3177/jnsv.63.396

Greenstein, 2012, Vitamins A &D inhibit the growth of mycobacteria in radiometric culture, PLoS ONE., 7, e29631, 10.1371/journal.pone.0029631

Szaleniec, 2018, Bacterial steroid hydroxylases: enzyme classes, their functions and comparison of their catalytic mechanisms, Appl Microbiol Biotechnol., 102, 8153, 10.1007/s00253-018-9239-3

Sugimoto, 2008, Crystal structure of CYP105A1 (P450SU-1) in complex with 1α,25-dihydroxy vitamin D3†,‡, Biochemistry., 47, 4017, 10.1021/bi7023767

Geer, 2010, The NCBI BioSystems database, Nucleic Acids Res., 38, D492, 10.1093/nar/gkp858

Bora, 2018, The gut microbiota regulates endocrine vitamin D metabolism through fibroblast growth factor 23, Front Immunol., 9, 408, 10.3389/fimmu.2018.00408

Schäffler, 2018, Vitamin D administration leads to a shift of the intestinal bacterial composition in Crohn's disease patients, but not in healthy controls, J Digest Dis., 19, 225, 10.1111/1751-2980.12591

Garg, 2018, The effect of Vitamin D on intestinal inflammation and faecal microbiota in patients with ulcerative colitis, J Crohn's Colitis., 12, 963, 10.1093/ecco-jcc/jjy052

Ghaly, 2018, High dose vitamin D supplementation alters faecal microbiome and predisposes mice to more severe colitis, Sci Rep., 8, 11511, 10.1038/s41598-018-29759-y

Kang, 2013, Extracellular vesicles derived from gut microbiota, especially Akkermansia muciniphila, protect the progression of dextran sulfate sodium-induced colitis D, PLoS ONE., 8, e76520, 10.1371/journal.pone.0076520

Assa, 2015, Vitamin D deficiency predisposes to adherent-invasive Escherichia coli-induced barrier dysfunction and experimental colonic injury, Inflamm Bowel Dis., 21, 297, 10.1097/MIB.0000000000000282

Chen, 2015, Protective effect of 1,25-dihydroxyvitamin d3 on lipopolysaccharide-induced intestinal epithelial tight junction injury in caco-2 cell monolayers, Inflammation., 38, 375, 10.1007/s10753-014-0041-9

Kong, 2008, Novel role of the vitamin D receptor in maintaining the integrity of the intestinal mucosal barrier, Am J Physiol Gastrointest Liver Physiol., 294, 208, 10.1152/ajpgi.00398.2007

Zhao, 2012, Protective role of 1, 25 (OH) 2 vitamin D 3 in the mucosal injury and epithelial barrier disruption in DSS-induced acute colitis in mice, BMC Gastroenterol., 25, 1, 10.1186/1471-230X-12-1

Zhang, 2015, Tight junction CLDN2 gene is a direct target of the vitamin D receptor, Sci Rep., 5, 10642, 10.1038/srep10642

He, 2018, Gut epithelial vitamin d receptor regulates microbiota-dependent mucosal inflammation by suppressing intestinal epithelial cell apoptosis, Endocrinology., 159, 967, 10.1210/en.2017-00748

Gombart, 2005, Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1,25-dihydroxyvitamin D3, FASEB J., 19, 1067, 10.1096/fj.04-3284com

Wang, 2004, Cutting edge: 1,25-dihydroxyvitamin D 3 is a direct inducer of antimicrobial peptide gene expression, J Immunol., 173, 2909, 10.4049/jimmunol.173.5.2909

Lagishetty, 2010, Vitamin D deficiency in mice impairs colonic antibacterial activity and predisposes to colitis, Endocrinology., 151, 2423, 10.1210/en.2010-0089

Bevins, 2011, Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis, Nat Rev Microbiol., 9, 356, 10.1038/nrmicro2546

Vandamme, 2012, A comprehensive summary of LL-37, the factotum human cathelicidin peptide, Cell Immunol., 280, 22, 10.1016/j.cellimm.2012.11.009