Mucosal Immunity and the Gut-Microbiota-Brain-Axis in Neuroimmune Disease
Abstract
:1. Introduction
2. Architecture of the Gastrointestinal Immune System
2.1. Intestinal Epithelial Cells (IECs)
2.2. Lamina Propria (LP)
2.3. Peyer’s Patches (PPs)
2.4. Mesenteric Lymph Nodes and Lymphatics
2.5. Appendix
3. Innate Immunity at the GI Mucosal Surface
3.1. Mucus Production by the Intestinal Epithelium
3.2. Pattern Recognition Receptors and Innate Lymphoid GI Cells
3.3. Antimicrobial Peptides of the GI Tract
3.4. Populations of Innate Lymphoid Cells in the GI Tract
4. GI Adaptive Immunity
4.1. T Cell Subsets
4.2. B Cells and GI Antibody Production
5. Bridges between Innate and Adaptive GI Immune Responses
5.1. Conventional Dendritic Cells
5.2. Plasmacytoid Dendritic Cells
5.3. Macrophages
5.4. Natural Killer Cells
6. Human Neuroimmune Diseases with Altered Microbiomes
6.1. Alzheimer’s Disease (AD)
6.2. Autism Spectrum Disorders
6.3. Parkinson’s Disease
6.4. Myalgic Encephalomyelitis
6.5. Multiple Sclerosis
6.6. Hunting’s Disease
7. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
References
- Servin, A.L.; Coconnier, M.H. Adhesion of probiotic strains to the intestinal mucosa and interaction with pathogens. Best Pract. Res. Clin. Gastroenterol. 2003, 17, 741–754. [Google Scholar] [CrossRef]
- Desai, M.S.; Seekatz, A.M.; Koropatkin, N.M.; Kamada, N.; Hickey, C.A.; Wolter, M.; Pudlo, N.A.; Kitamoto, S.; Terrapon, N.; Muller, A.; et al. A Dietary Fiber-Deprived Gut Microbiota Degrades the Colonic Mucus Barrier and Enhances Pathogen Susceptibility. Cell 2016, 167, 1339–1353.e1321. [Google Scholar] [CrossRef] [Green Version]
- Oliva, A.; Aversano, L.; de Angelis, M.; Mascellino, M.T.; Miele, M.C.; Morelli, S.; Battaglia, R.; Iera, J.; Bruno, G.; Corazziari, E.S.; et al. Persistent Systemic Microbial Translocation, Inflammation, and Intestinal Damage During Clostridioides difficile Infection. Open Forum Infect. Dis. 2020, 7, ofz507. [Google Scholar] [CrossRef]
- Zhou, X.; Li, J.; Guo, J.; Geng, B.; Ji, W.; Zhao, Q.; Li, J.; Liu, X.; Liu, J.; Guo, Z.; et al. Gut-dependent microbial translocation induces inflammation and cardiovascular events after ST-elevation myocardial infarction. Microbiome 2018, 6, 66. [Google Scholar] [CrossRef] [Green Version]
- Vrakas, S.; Mountzouris, K.C.; Michalopoulos, G.; Karamanolis, G.; Papatheodoridis, G.; Tzathas, C.; Gazouli, M. Intestinal Bacteria Composition and Translocation of Bacteria in Inflammatory Bowel Disease. PLoS ONE 2017, 12, e0170034. [Google Scholar] [CrossRef] [Green Version]
- Kasubuchi, M.; Hasegawa, S.; Hiramatsu, T.; Ichimura, A.; Kimura, I. Dietary gut microbial metabolites, short-chain fatty acids, and host metabolic regulation. Nutrients 2015, 7, 2839–2849. [Google Scholar] [CrossRef] [Green Version]
- Candido, E.P.; Reeves, R.; Davie, J.R. Sodium butyrate inhibits histone deacetylation in cultured cells. Cell 1978, 14, 105–113. [Google Scholar] [CrossRef]
- Louis, P.; Flint, H.J. Diversity, metabolism and microbial ecology of butyrate-producing bacteria from the human large intestine. FEMS Microbiol. Lett. 2009, 294, 1–8. [Google Scholar] [CrossRef] [Green Version]
- Fung, K.Y.; Cosgrove, L.; Lockett, T.; Head, R.; Topping, D.L. A review of the potential mechanisms for the lowering of colorectal oncogenesis by butyrate. Br. J. Nutr. 2012, 108, 820–831. [Google Scholar] [CrossRef] [Green Version]
- Barter, R.; Pearse, A.G. Detection of 5-hydroxytryptamine in mammalian enterochromaffin cells. Nature 1953, 172, 810. [Google Scholar] [CrossRef]
- Yano, J.M.; Yu, K.; Donaldson, G.P.; Shastri, G.G.; Ann, P.; Ma, L.; Nagler, C.R.; Ismagilov, R.F.; Mazmanian, S.K.; Hsiao, E.Y. Indigenous bacteria from the gut microbiota regulate host serotonin biosynthesis. Cell 2015, 161, 264–276. [Google Scholar] [CrossRef] [Green Version]
- Fung, T.C.; Vuong, H.E.; Luna, C.D.G.; Pronovost, G.N.; Aleksandrova, A.A.; Riley, N.G.; Vavilina, A.; McGinn, J.; Rendon, T.; Forrest, L.R.; et al. Intestinal serotonin and fluoxetine exposure modulate bacterial colonization in the gut. Nat. Microbiol. 2019, 4, 2064–2073. [Google Scholar] [CrossRef]
- Barton, S.M.; Janve, V.A.; McClure, R.; Anderson, A.; Matsubara, J.A.; Gore, J.C.; Pham, W. Lipopolysaccharide Induced Opening of the Blood Brain Barrier on Aging 5XFAD Mouse Model. J. Alzheimer’s Dis. JAD 2019, 67, 503–513. [Google Scholar] [CrossRef]
- Chen, Y.; Xu, J.; Chen, Y. Regulation of Neurotransmitters by the Gut Microbiota and Effects on Cognition in Neurological Disorders. Nutrients 2021, 13, 2099. [Google Scholar] [CrossRef]
- Parker, A.; Fonseca, S.; Carding, S.R. Gut microbes and metabolites as modulators of blood-brain barrier integrity and brain health. Gut Microbes 2020, 11, 135–157. [Google Scholar] [CrossRef] [Green Version]
- Gwak, M.G.; Chang, S.Y. Gut-Brain Connection: Microbiome, Gut Barrier, and Environmental Sensors. Immune Netw. 2021, 21, e20. [Google Scholar] [CrossRef]
- Guy-Grand, D.; Vassalli, P. Gut intraepithelial T lymphocytes. Curr. Opin. Immunol. 1993, 5, 247–252. [Google Scholar] [CrossRef]
- Brandtzaeg, P. Food allergy: Separating the science from the mythology. Nat. Rev. Gastroenterol. Hepatol. 2010, 7, 380–400. [Google Scholar] [CrossRef]
- Boudry, G.; Yang, P.-C.; Perdue, M.H. Small Intestine, Anatomy. In Encyclopedia of Gastroenterology; Johnson, L.R., Ed.; Elsevier: New York, NY, USA, 2004; pp. 404–409. [Google Scholar]
- Adapted from Intestinal Immune System (Small Intestine) by BioRender.com. Available online: https://app.biorender.com/biorender-templates (accessed on 23 October 2022).
- Guy-Grand, D.; Cerf-Bensussan, N.; Malissen, B.; Malassis-Seris, M.; Briottet, C.; Vassalli, P. Two gut intraepithelial CD8+ lymphocyte populations with different T cell receptors: A role for the gut epithelium in T cell differentiation. J. Exp. Med. 1991, 173, 471–481. [Google Scholar] [CrossRef] [Green Version]
- Bonneville, M.; Janeway, C.A., Jr.; Ito, K.; Haser, W.; Ishida, I.; Nakanishi, N.; Tonegawa, S. Intestinal intraepithelial lymphocytes are a distinct set of gamma delta T cells. Nature 1988, 336, 479–481. [Google Scholar] [CrossRef]
- Feldman, M.; Friedman, L.S.; Brandt, L.J. Sleisenger and Fordtran’s Gastrointestinal and Liver Disease: Pathophysiology, Diagnosis, Management, 11th ed.; Elsevier: Philadelphia, PA, USA, 2020. [Google Scholar]
- Fenton, T.M.; Jorgensen, P.B.; Niss, K.; Rubin, S.J.S.; Morbe, U.M.; Riis, L.B.; da Silva, C.; Plumb, A.; Vandamme, J.; Jakobsen, H.L.; et al. Immune Profiling of Human Gut-Associated Lymphoid Tissue Identifies a Role for Isolated Lymphoid Follicles in Priming of Region-Specific Immunity. Immunity 2020, 52, 557–570.e556. [Google Scholar] [CrossRef] [PubMed]
- van Kruiningen, H.J.; West, A.B.; Freda, B.J.; Holmes, K.A. Distribution of Peyer’s patches in the distal ileum. Inflamm. Bowel Dis. 2002, 8, 180–185. [Google Scholar] [CrossRef] [PubMed]
- Cornes, J.S. Number, size, and distribution of Peyer’s patches in the human small intestine: Part I The development of Peyer’s patches. Gut 1965, 6, 225–229. [Google Scholar] [CrossRef] [Green Version]
- Neutra, M.R.; Mantis, N.J.; Kraehenbuhl, J.P. Collaboration of epithelial cells with organized mucosal lymphoid tissues. Nat. Immunol. 2001, 2, 1004–1009. [Google Scholar] [CrossRef]
- Rios, D.; Wood, M.B.; Li, J.; Chassaing, B.; Gewirtz, A.T.; Williams, I.R. Antigen sampling by intestinal M cells is the principal pathway initiating mucosal IgA production to commensal enteric bacteria. Mucosal Immunol. 2016, 9, 907–916. [Google Scholar] [CrossRef] [Green Version]
- Kobayashi, D.; Endo, M.; Ochi, H.; Hojo, H.; Miyasaka, M.; Hayasaka, H. Regulation of CCR7-dependent cell migration through CCR7 homodimer formation. Sci. Rep. 2017, 7, 8536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kobayashi, N.; Takahashi, D.; Takano, S.; Kimura, S.; Hase, K. The Roles of Peyer’s Patches and Microfold Cells in the Gut Immune System: Relevance to Autoimmune Diseases. Front. Immunol. 2019, 10, 2345. [Google Scholar] [CrossRef] [Green Version]
- Hase, K.; Kawano, K.; Nochi, T.; Pontes, G.S.; Fukuda, S.; Ebisawa, M.; Kadokura, K.; Tobe, T.; Fujimura, Y.; Kawano, S.; et al. Uptake through glycoprotein 2 of FimH(+) bacteria by M cells initiates mucosal immune response. Nature 2009, 462, 226–230. [Google Scholar] [CrossRef]
- Yanagihara, S.; Kanaya, T.; Fukuda, S.; Nakato, G.; Hanazato, M.; Wu, X.R.; Yamamoto, N.; Ohno, H. Uromodulin-SlpA binding dictates Lactobacillus acidophilus uptake by intestinal epithelial M cells. Int. Immunol. 2017, 29, 357–363. [Google Scholar] [CrossRef] [Green Version]
- Matsumura, T.; Sugawara, Y.; Yutani, M.; Amatsu, S.; Yagita, H.; Kohda, T.; Fukuoka, S.; Nakamura, Y.; Fukuda, S.; Hase, K.; et al. Botulinum toxin A complex exploits intestinal M cells to enter the host and exert neurotoxicity. Nat. Commun. 2015, 6, 6255. [Google Scholar] [CrossRef]
- Hamzaoui, N.; Kerneis, S.; Caliot, E.; Pringault, E. Expression and distribution of beta1 integrins in in vitro-induced M cells: Implications for Yersinia adhesion to Peyer’s patch epithelium. Cell. Microbiol. 2004, 6, 817–828. [Google Scholar] [CrossRef] [PubMed]
- Jensen, V.B.; Harty, J.T.; Jones, B.D. Interactions of the invasive pathogens Salmonella typhimurium, Listeria monocytogenes, and Shigella flexneri with M cells and murine Peyer’s patches. Infect. Immun. 1998, 66, 3758–3766. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jones, B.D.; Ghori, N.; Falkow, S. Salmonella typhimurium initiates murine infection by penetrating and destroying the specialized epithelial M cells of the Peyer’s patches. J. Exp. Med. 1994, 180, 15–23. [Google Scholar] [CrossRef] [Green Version]
- Kerneis, S.; Bogdanova, A.; Kraehenbuhl, J.P.; Pringault, E. Conversion by Peyer’s patch lymphocytes of human enterocytes into M cells that transport bacteria. Science 1997, 277, 949–952. [Google Scholar] [CrossRef] [PubMed]
- Ouzilou, L.; Caliot, E.; Pelletier, I.; Prevost, M.C.; Pringault, E.; Colbere-Garapin, F. Poliovirus transcytosis through M-like cells. J. Gen. Virol. 2002, 83, 2177–2182. [Google Scholar] [CrossRef] [Green Version]
- Cornes, J.S. Peyer’s patches in the human gut. Proc. R. Soc. Med. 1965, 58, 716. [Google Scholar] [CrossRef] [Green Version]
- Heel, K.A.; McCauley, R.D.; Papadimitriou, J.M.; Hall, J.C. Review: Peyer’s patches. J. Gastroenterol. Hepatol. 1997, 12, 122–136. [Google Scholar] [CrossRef]
- Chehade, M.; Mayer, L. Oral tolerance and its relation to food hypersensitivities. J. Allergy Clin. Immunol. 2005, 115, 3–12. [Google Scholar] [CrossRef]
- Chen, Y.; Kuchroo, V.K.; Inobe, J.; Hafler, D.A.; Weiner, H.L. Regulatory T cell clones induced by oral tolerance: Suppression of autoimmune encephalomyelitis. Science 1994, 265, 1237–1240. [Google Scholar] [CrossRef]
- Whitacre, C.C.; Gienapp, I.E.; Orosz, C.G.; Bitar, D.M. Oral tolerance in experimental autoimmune encephalomyelitis. III. Evidence for clonal anergy. J. Immunol. 1991, 147, 2155–2163. [Google Scholar]
- Bogunovic, M.; Ginhoux, F.; Helft, J.; Shang, L.; Hashimoto, D.; Greter, M.; Liu, K.; Jakubzick, C.; Ingersoll, M.A.; Leboeuf, M.; et al. Origin of the lamina propria dendritic cell network. Immunity 2009, 31, 513–525. [Google Scholar] [CrossRef] [PubMed]
- Matteoli, G.; Mazzini, E.; Iliev, I.D.; Mileti, E.; Fallarino, F.; Puccetti, P.; Chieppa, M.; Rescigno, M. Gut CD103+ dendritic cells express indoleamine 2,3-dioxygenase which influences T regulatory/T effector cell balance and oral tolerance induction. Gut 2010, 59, 595–604. [Google Scholar] [CrossRef] [PubMed]
- Denning, T.L.; Wang, Y.C.; Patel, S.R.; Williams, I.R.; Pulendran, B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat. Immunol. 2007, 8, 1086–1094. [Google Scholar] [CrossRef] [PubMed]
- du Pre, M.F.; Samsom, J.N. Adaptive T-cell responses regulating oral tolerance to protein antigen. Allergy 2011, 66, 478–490. [Google Scholar] [CrossRef]
- Maloy, K.J.; Powrie, F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature 2011, 474, 298–306. [Google Scholar] [CrossRef]
- Bollinger, R.R.; Barbas, A.S.; Bush, E.L.; Lin, S.S.; Parker, W. Biofilms in the large bowel suggest an apparent function of the human vermiform appendix. J. Theor. Biol. 2007, 249, 826–831. [Google Scholar] [CrossRef]
- Smith, H.F.; Fisher, R.E.; Everett, M.L.; Thomas, A.D.; Bollinger, R.R.; Parker, W. Comparative anatomy and phylogenetic distribution of the mammalian cecal appendix. J. Evol. Biol. 2009, 22, 1984–1999. [Google Scholar] [CrossRef]
- Im, G.Y.; Modayil, R.J.; Lin, C.T.; Geier, S.J.; Katz, D.S.; Feuerman, M.; Grendell, J.H. The appendix may protect against Clostridium difficile recurrence. Clin. Gastroenterol. Hepatol. 2011, 9, 1072–1077. [Google Scholar] [CrossRef]
- Merchant, R.; Mower, W.R.; Ourian, A.; Abrahamian, F.M.; Moran, G.J.; Krishnadasan, A.; Talan, D.A. Association Between Appendectomy and Clostridium difficile Infection. J. Clin. Med. Res. 2012, 4, 17–19. [Google Scholar] [CrossRef] [Green Version]
- Heindl, S.E.; Tsouklidis, N. Appendectomy as a Potential Predisposing Factor for the Development of Recurrent and Fulminant Clostridium Difficile. Cureus 2020, 12, e10091. [Google Scholar] [CrossRef]
- Shi, Y.B.; Shibata, Y.; Tanizaki, Y.; Fu, L. The development of adult intestinal stem cells: Insights from studies on thyroid hormone-dependent anuran metamorphosis. Vitam. Horm. 2021, 116, 269–293. [Google Scholar] [CrossRef] [PubMed]
- Gordon, J.I.; Schmidt, G.H.; Roth, K.A. Studies of intestinal stem cells using normal, chimeric, and transgenic mice. FASEB J. 1992, 6, 3039–3050. [Google Scholar] [CrossRef] [PubMed]
- Lueschow, S.R.; McElroy, S.J. The Paneth Cell: The Curator and Defender of the Immature Small Intestine. Front. Immunol. 2020, 11, 587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Phillips, A.D.; Brown, A.; Hicks, S.; Schuller, S.; Murch, S.H.; Walker-Smith, J.A.; Swallow, D.M. Acetylated sialic acid residues and blood group antigens localise within the epithelium in microvillous atrophy indicating internal accumulation of the glycocalyx. Gut 2004, 53, 1764–1771. [Google Scholar] [CrossRef] [Green Version]
- Pelaseyed, T.; Bergstrom, J.H.; Gustafsson, J.K.; Ermund, A.; Birchenough, G.M.; Schutte, A.; van der Post, S.; Svensson, F.; Rodriguez-Pineiro, A.M.; Nystrom, E.E.; et al. The mucus and mucins of the goblet cells and enterocytes provide the first defense line of the gastrointestinal tract and interact with the immune system. Immunol. Rev. 2014, 260, 8–20. [Google Scholar] [CrossRef] [Green Version]
- Martens, E.C.; Chiang, H.C.; Gordon, J.I. Mucosal glycan foraging enhances fitness and transmission of a saccharolytic human gut bacterial symbiont. Cell Host Microbe 2008, 4, 447–457. [Google Scholar] [CrossRef] [Green Version]
- Martens, E.C.; Neumann, M.; Desai, M.S. Interactions of commensal and pathogenic microorganisms with the intestinal mucosal barrier. Nat. Rev. Microbiol. 2018, 16, 457–470. [Google Scholar] [CrossRef]
- Corthesy, B. Multi-faceted functions of secretory IgA at mucosal surfaces. Front. Immunol. 2013, 4, 185. [Google Scholar] [CrossRef] [Green Version]
- Gibbins, H.L.; Proctor, G.B.; Yakubov, G.E.; Wilson, S.; Carpenter, G.H. SIgA binding to mucosal surfaces is mediated by mucin-mucin interactions. PLoS ONE 2015, 10, e0119677. [Google Scholar] [CrossRef] [Green Version]
- Herath, M.; Hosie, S.; Bornstein, J.C.; Franks, A.E.; Hill-Yardin, E.L. The Role of the Gastrointestinal Mucus System in Intestinal Homeostasis: Implications for Neurological Disorders. Front. Cell. Infect. Microbiol. 2020, 10, 248. [Google Scholar] [CrossRef]
- Eden, A.; Marcotte, T.D.; Heaton, R.K.; Nilsson, S.; Zetterberg, H.; Fuchs, D.; Franklin, D.; Price, R.W.; Grant, I.; Letendre, S.L.; et al. Increased Intrathecal Immune Activation in Virally Suppressed HIV-1 Infected Patients with Neurocognitive Impairment. PLoS ONE 2016, 11, e0157160. [Google Scholar] [CrossRef] [PubMed]
- Lee, B.; Moon, K.M.; Kim, C.Y. Tight Junction in the Intestinal Epithelium: Its Association with Diseases and Regulation by Phytochemicals. J. Immunol. Res. 2018, 2018, 2645465. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Minagar, A.; Ostanin, D.; Long, A.C.; Jennings, M.; Kelley, R.E.; Sasaki, M.; Alexander, J.S. Serum from patients with multiple sclerosis downregulates occludin and VE-cadherin expression in cultured endothelial cells. Mult. Scler. 2003, 9, 235–238. [Google Scholar] [CrossRef] [PubMed]
- Fiorentino, M.; Sapone, A.; Senger, S.; Camhi, S.S.; Kadzielski, S.M.; Buie, T.M.; Kelly, D.L.; Cascella, N.; Fasano, A. Blood-brain barrier and intestinal epithelial barrier alterations in autism spectrum disorders. Mol. Autism 2016, 7, 49. [Google Scholar] [CrossRef] [Green Version]
- Li, C.; Cui, L.; Yang, Y.; Miao, J.; Zhao, X.; Zhang, J.; Cui, G.; Zhang, Y. Gut Microbiota Differs Between Parkinson’s Disease Patients and Healthy Controls in Northeast China. Front. Mol. Neurosci. 2019, 12, 171. [Google Scholar] [CrossRef] [Green Version]
- Kawasaki, T.; Kawai, T. Toll-like receptor signaling pathways. Front. Immunol. 2014, 5, 461. [Google Scholar] [CrossRef] [Green Version]
- Tang, J.; Lin, G.; Langdon, W.Y.; Tao, L.; Zhang, J. Regulation of C-Type Lectin Receptor-Mediated Antifungal Immunity. Front. Immunol. 2018, 9, 123. [Google Scholar] [CrossRef] [Green Version]
- Brown, G.D.; Willment, J.A.; Whitehead, L. C-type lectins in immunity and homeostasis. Nat. Rev. Immunol. 2018, 18, 374–389. [Google Scholar] [CrossRef]
- Amarante-Mendes, G.P.; Adjemian, S.; Branco, L.M.; Zanetti, L.C.; Weinlich, R.; Bortoluci, K.R. Pattern Recognition Receptors and the Host Cell Death Molecular Machinery. Front. Immunol. 2018, 9, 2379. [Google Scholar] [CrossRef] [Green Version]
- Fukata, M.; Arditi, M. The role of pattern recognition receptors in intestinal inflammation. Mucosal Immunol. 2013, 6, 451–463. [Google Scholar] [CrossRef]
- Lundberg, K.; Rydnert, F.; Greiff, L.; Lindstedt, M. Human blood dendritic cell subsets exhibit discriminative pattern recognition receptor profiles. Immunology 2014, 142, 279–288. [Google Scholar] [CrossRef] [PubMed]
- Tatematsu, M.; Funami, K.; Seya, T.; Matsumoto, M. Extracellular RNA Sensing by Pattern Recognition Receptors. J. Innate Immun. 2018, 10, 398–406. [Google Scholar] [CrossRef] [PubMed]
- Brun, P.; Giron, M.C.; Qesari, M.; Porzionato, A.; Caputi, V.; Zoppellaro, C.; Banzato, S.; Grillo, A.R.; Spagnol, L.; de Caro, R.; et al. Toll-like receptor 2 regulates intestinal inflammation by controlling integrity of the enteric nervous system. Gastroenterology 2013, 145, 1323–1333. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dubovy, P.; Hradilova-Svizenska, I.; Brazda, V.; Joukal, M. Toll-Like Receptor 9-Mediated Neuronal Innate Immune Reaction Is Associated with Initiating a Pro-Regenerative State in Neurons of the Dorsal Root Ganglia Non-Associated with Sciatic Nerve Lesion. Int. J. Mol. Sci. 2021, 22, 7446. [Google Scholar] [CrossRef]
- Lee, J.W.; Nam, H.; Kim, L.E.; Jeon, Y.; Min, H.; Ha, S.; Lee, Y.; Kim, S.Y.; Lee, S.J.; Kim, E.K.; et al. TLR4 (toll-like receptor 4) activation suppresses autophagy through inhibition of FOXO3 and impairs phagocytic capacity of microglia. Autophagy 2019, 15, 753–770. [Google Scholar] [CrossRef]
- Barajon, I.; Serrao, G.; Arnaboldi, F.; Opizzi, E.; Ripamonti, G.; Balsari, A.; Rumio, C. Toll-like receptors 3, 4, and 7 are expressed in the enteric nervous system and dorsal root ganglia. J. Histochem. Cytochem. 2009, 57, 1013–1023. [Google Scholar] [CrossRef] [Green Version]
- Wu, J.; Fernandes-Alnemri, T.; Alnemri, E.S. Involvement of the AIM2, NLRC4, and NLRP3 inflammasomes in caspase-1 activation by Listeria monocytogenes. J. Clin. Immunol. 2010, 30, 693–702. [Google Scholar] [CrossRef] [Green Version]
- Mathur, A.; Hayward, J.A.; Man, S.M. Molecular mechanisms of inflammasome signaling. J. Leukoc. Biol. 2018, 103, 233–257. [Google Scholar] [CrossRef] [Green Version]
- Man, S.M. Inflammasomes in the gastrointestinal tract: Infection, cancer and gut microbiota homeostasis. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 721–737. [Google Scholar] [CrossRef]
- Adapted from Innate Immune System (Cellular Locations of Pattern Recognition Receptors) by BioRender.com. Available online: https://app.biorender.com/biorender-templates (accessed on 23 October 2022).
- Gritsenko, A.; Yu, S.; Martin-Sanchez, F.; Diaz-Del-Olmo, I.; Nichols, E.M.; Davis, D.M.; Brough, D.; Lopez-Castejon, G. Priming Is Dispensable for NLRP3 Inflammasome Activation in Human Monocytes In Vitro. Front. Immunol. 2020, 11, 565924. [Google Scholar] [CrossRef]
- Hampe, J.; Grebe, J.; Nikolaus, S.; Solberg, C.; Croucher, P.J.; Mascheretti, S.; Jahnsen, J.; Moum, B.; Klump, B.; Krawczak, M.; et al. Association of NOD2 (CARD 15) genotype with clinical course of Crohn’s disease: A cohort study. Lancet 2002, 359, 1661–1665. [Google Scholar] [CrossRef]
- Nabhani, Z.A.; Dietrich, G.; Hugot, J.P.; Barreau, F. Nod2: The intestinal gate keeper. PLoS Pathog. 2017, 13, e1006177. [Google Scholar] [CrossRef] [Green Version]
- Feerick, C.L.; McKernan, D.P. Understanding the regulation of pattern recognition receptors in inflammatory diseases—A ‘Nod’ in the right direction. Immunology 2017, 150, 237–247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cleynen, I.; Boucher, G.; Jostins, L.; Schumm, L.P.; Zeissig, S.; Ahmad, T.; Andersen, V.; Andrews, J.M.; Annese, V.; Brand, S.; et al. Inherited determinants of Crohn’s disease and ulcerative colitis phenotypes: A genetic association study. Lancet 2016, 387, 156–167. [Google Scholar] [CrossRef] [Green Version]
- Patterson-Delafield, J.; Szklarek, D.; Martinez, R.J.; Lehrer, R.I. Microbicidal cationic proteins of rabbit alveolar macrophages: Amino acid composition and functional attributes. Infect. Immun. 1981, 31, 723–731. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dodd, D.; Spitzer, M.H.; van Treuren, W.; Merrill, B.D.; Hryckowian, A.J.; Higginbottom, S.K.; Le, A.; Cowan, T.M.; Nolan, G.P.; Fischbach, M.A.; et al. A gut bacterial pathway metabolizes aromatic amino acids into nine circulating metabolites. Nature 2017, 551, 648–652. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dupont, A.; Kaconis, Y.; Yang, I.; Albers, T.; Woltemate, S.; Heinbockel, L.; Andersson, M.; Suerbaum, S.; Brandenburg, K.; Hornef, M.W. Intestinal mucus affinity and biological activity of an orally administered antibacterial and anti-inflammatory peptide. Gut 2015, 64, 222–232. [Google Scholar] [CrossRef]
- Ganguly, D.; Chamilos, G.; Lande, R.; Gregorio, J.; Meller, S.; Facchinetti, V.; Homey, B.; Barrat, F.J.; Zal, T.; Gilliet, M. Self-RNA-antimicrobial peptide complexes activate human dendritic cells through TLR7 and TLR8. J. Exp. Med. 2009, 206, 1983–1994. [Google Scholar] [CrossRef]
- Masera, R.G.; Bateman, A.; Muscettola, M.; Solomon, S.; Angeli, A. Corticostatins/defensins inhibit in vitro NK activity and cytokine production by human peripheral blood mononuclear cells. Regul. Pept. 1996, 62, 13–21. [Google Scholar] [CrossRef]
- Zanetti, M.; Litteri, L.; Griffiths, G.; Gennaro, R.; Romeo, D. Stimulus-induced maturation of probactenecins, precursors of neutrophil antimicrobial polypeptides. J. Immunol. 1991, 146, 4295–4300. [Google Scholar]
- Oppenheim, F.G.; Xu, T.; McMillian, F.M.; Levitz, S.M.; Diamond, R.D.; Offner, G.D.; Troxler, R.F. Histatins, a novel family of histidine-rich proteins in human parotid secretion. Isolation, characterization, primary structure, and fungistatic effects on Candida albicans. J. Biol. Chem. 1988, 263, 7472–7477. [Google Scholar] [CrossRef]
- Yadav, J.K. Structural and functional swapping of amyloidogenic and antimicrobial peptides: Redefining the role of amyloidogenic propensity in disease and host defense. J. Pept. Sci. 2022, 28, e3378. [Google Scholar] [CrossRef] [PubMed]
- Panda, S.K.; Colonna, M. Innate Lymphoid Cells in Mucosal Immunity. Front. immunol. 2019, 10, 861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Constantinides, M.G.; McDonald, B.D.; Verhoef, P.A.; Bendelac, A. A committed precursor to innate lymphoid cells. Nature 2014, 508, 397–401. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Weizman, O.E.; Adams, N.M.; Schuster, I.S.; Krishna, C.; Pritykin, Y.; Lau, C.; Degli-Esposti, M.A.; Leslie, C.S.; Sun, J.C.; O’Sullivan, T.E. ILC1 Confer Early Host Protection at Initial Sites of Viral Infection. Cell 2017, 171, 795–808.e712. [Google Scholar] [CrossRef]
- Toki, S.; Goleniewska, K.; Zhang, J.; Zhou, W.; Newcomb, D.C.; Zhou, B.; Kita, H.; Boyd, K.L.; Peebles, R.S., Jr. TSLP and IL-33 reciprocally promote each other’s lung protein expression and ILC2 receptor expression to enhance innate type-2 airway inflammation. Allergy 2020, 75, 1606–1617. [Google Scholar] [CrossRef]
- Fallon, P.G.; Ballantyne, S.J.; Mangan, N.E.; Barlow, J.L.; Dasvarma, A.; Hewett, D.R.; McIlgorm, A.; Jolin, H.E.; McKenzie, A.N. Identification of an interleukin (IL)-25-dependent cell population that provides IL-4, IL-5, and IL-13 at the onset of helminth expulsion. J. Exp. Med. 2006, 203, 1105–1116. [Google Scholar] [CrossRef]
- Pickard, J.M.; Maurice, C.F.; Kinnebrew, M.A.; Abt, M.C.; Schenten, D.; Golovkina, T.V.; Bogatyrev, S.R.; Ismagilov, R.F.; Pamer, E.G.; Turnbaugh, P.J.; et al. Rapid fucosylation of intestinal epithelium sustains host-commensal symbiosis in sickness. Nature 2014, 514, 638–641. [Google Scholar] [CrossRef] [Green Version]
- Wolk, K.; Kunz, S.; Witte, E.; Friedrich, M.; Asadullah, K.; Sabat, R. IL-22 increases the innate immunity of tissues. Immunity 2004, 21, 241–254. [Google Scholar] [CrossRef] [Green Version]
- Eberl, G.; Marmon, S.; Sunshine, M.J.; Rennert, P.D.; Choi, Y.; Littman, D.R. An essential function for the nuclear receptor RORgamma(t) in the generation of fetal lymphoid tissue inducer cells. Nat. Immunol. 2004, 5, 64–73. [Google Scholar] [CrossRef]
- Created with BioRender.com. Available online: www.biorender.com (accessed on 23 October 2022).
- Han, L.; Wang, X.M.; Di, S.; Gao, Z.Z.; Li, Q.W.; Wu, H.R.; Wang, Q.; Zhao, L.H.; Tong, X.L. Innate Lymphoid Cells: A Link between the Nervous System and Microbiota in Intestinal Networks. Mediat. Inflamm. 2019, 2019, 1978094. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gangadharan, D.; Lambolez, F.; Attinger, A.; Wang-Zhu, Y.; Sullivan, B.A.; Cheroutre, H. Identification of pre- and postselection TCRalphabeta+ intraepithelial lymphocyte precursors in the thymus. Immunity 2006, 25, 631–641. [Google Scholar] [CrossRef] [PubMed]
- Guy-Grand, D.; Vassalli, P.; Eberl, G.; Pereira, P.; Burlen-Defranoux, O.; Lemaitre, F.; di Santo, J.P.; Freitas, A.A.; Cumano, A.; Bandeira, A. Origin, trafficking, and intraepithelial fate of gut-tropic T cells. J. Exp. Med. 2013, 210, 1839–1854. [Google Scholar] [CrossRef] [Green Version]
- Cheroutre, H.; Lambolez, F. Doubting the TCR coreceptor function of CD8alphaalpha. Immunity 2008, 28, 149–159. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sheridan, B.S.; Lefrancois, L. Intraepithelial lymphocytes: To serve and protect. Curr. Gastroenterol. Rep. 2010, 12, 513–521. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Deusch, K.; Luling, F.; Reich, K.; Classen, M.; Wagner, H.; Pfeffer, K. A major fraction of human intraepithelial lymphocytes simultaneously expresses the gamma/delta T cell receptor, the CD8 accessory molecule and preferentially uses the V delta 1 gene segment. Eur. J. Immunol. 1991, 21, 1053–1059. [Google Scholar] [CrossRef]
- Johansson-Lindbom, B.; Svensson, M.; Pabst, O.; Palmqvist, C.; Marquez, G.; Forster, R.; Agace, W.W. Functional specialization of gut CD103+ dendritic cells in the regulation of tissue-selective T cell homing. J. Exp. Med. 2005, 202, 1063–1073. [Google Scholar] [CrossRef] [Green Version]
- Johansson-Lindbom, B.; Svensson, M.; Wurbel, M.A.; Malissen, B.; Marquez, G.; Agace, W. Selective generation of gut tropic T cells in gut-associated lymphoid tissue (GALT): Requirement for GALT dendritic cells and adjuvant. J. Exp. Med. 2003, 198, 963–969. [Google Scholar] [CrossRef]
- Svensson, M.; Johansson-Lindbom, B.; Zapata, F.; Jaensson, E.; Austenaa, L.M.; Blomhoff, R.; Agace, W.W. Retinoic acid receptor signaling levels and antigen dose regulate gut homing receptor expression on CD8+ T cells. Mucosal Immunol. 2008, 1, 38–48. [Google Scholar] [CrossRef] [Green Version]
- Lefrancois, L.; Parker, C.M.; Olson, S.; Muller, W.; Wagner, N.; Schon, M.P.; Puddington, L. The role of beta7 integrins in CD8 T cell trafficking during an antiviral immune response. J. Exp. Med. 1999, 189, 1631–1638. [Google Scholar] [CrossRef] [Green Version]
- Kunkel, E.J.; Kim, C.H.; Lazarus, N.H.; Vierra, M.A.; Soler, D.; Bowman, E.P.; Butcher, E.C. CCR10 expression is a common feature of circulating and mucosal epithelial tissue IgA Ab-secreting cells. J. Clin. Investig. 2003, 111, 1001–1010. [Google Scholar] [CrossRef] [PubMed]
- Cerf-Bensussan, N.; Guy-Grand, D.; Griscelli, C. Intraepithelial lymphocytes of human gut: Isolation, characterisation and study of natural killer activity. Gut 1985, 26, 81–88. [Google Scholar] [CrossRef] [PubMed]
- Ebert, E.C. Proliferative responses of human intraepithelial lymphocytes to various T-cell stimuli. Gastroenterology 1989, 97, 1372–1381. [Google Scholar] [CrossRef]
- Viney, J.L.; Kilshaw, P.J.; MacDonald, T.T. Cytotoxic alpha/beta+ and gamma/delta+ T cells in murine intestinal epithelium. Eur. J. Immunol. 1990, 20, 1623–1626. [Google Scholar] [CrossRef] [PubMed]
- Bene, K.; Varga, Z.; Petrov, V.O.; Boyko, N.; Rajnavolgyi, E. Gut Microbiota Species Can Provoke both Inflammatory and Tolerogenic Immune Responses in Human Dendritic Cells Mediated by Retinoic Acid Receptor Alpha Ligation. Front. Immunol. 2017, 8, 427. [Google Scholar] [CrossRef] [Green Version]
- Bauche, D.; Marie, J.C. Transforming growth factor beta: A master regulator of the gut microbiota and immune cell interactions. Clin. Transl. Immunol. 2017, 6, e136. [Google Scholar] [CrossRef]
- Ivanov, I.I.; Rde, L.F.; Manel, N.; Yoshinaga, K.; Rifkin, D.B.; Sartor, R.B.; Finlay, B.B.; Littman, D.R. Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell Host Microbe 2008, 4, 337–349. [Google Scholar] [CrossRef] [Green Version]
- Brandtzaeg, P.; Farstad, I.N.; Johansen, F.E.; Morton, H.C.; Norderhaug, I.N.; Yamanaka, T. The B-cell system of human mucosae and exocrine glands. Immunol. Rev. 1999, 171, 45–87. [Google Scholar] [CrossRef]
- Brandtzaeg, P.; Halstensen, T.S.; Kett, K.; Krajci, P.; Kvale, D.; Rognum, T.O.; Scott, H.; Sollid, L.M. Immunobiology and immunopathology of human gut mucosa: Humoral immunity and intraepithelial lymphocytes. Gastroenterology 1989, 97, 1562–1584. [Google Scholar] [CrossRef]
- Brandtzaeg, P. Mucosal immunity: Induction, dissemination, and effector functions. Scand. J. Immunol. 2009, 70, 505–515. [Google Scholar] [CrossRef]
- Zikan, J.; Novotny, J.; Trapane, T.L.; Koshland, M.E.; Urry, D.W.; Bennett, J.C.; Mestecky, J. Secondary structure of the immunoglobulin J chain. Proc. Natl. Acad. Sci. USA 1985, 82, 5905–5909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brandtzaeg, P. Two types of IgA immunocytes in man. Nat. New Biol. 1973, 243, 142–143. [Google Scholar] [CrossRef] [PubMed]
- Matsumoto, M.L. Molecular Mechanisms of Multimeric Assembly of IgM and IgA. Annu. Rev. Immunol. 2022, 40, 221–247. [Google Scholar] [CrossRef]
- Brandtzaeg, P. Mucosal and glandular distribution of immunoglobulin components: Differential localization of free and bound SC in secretory epithelial cells. J. Immunol. 1974, 112, 1553–1559. [Google Scholar] [PubMed]
- Mathias, A.; Corthesy, B. N-Glycans on secretory component: Mediators of the interaction between secretory IgA and gram-positive commensals sustaining intestinal homeostasis. Gut Microbes 2011, 2, 287–293. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kafkova, L.R.; Brokesova, D.; Krupka, M.; Stehlikova, Z.; Dvorak, J.; Coufal, S.; Fajstova, A.; Srutkova, D.; Stepanova, K.; Hermanova, P.; et al. Secretory IgA N-glycans contribute to the protection against E. coli O55 infection of germ-free piglets. Mucosal Immunol. 2021, 14, 511–522. [Google Scholar] [CrossRef]
- Brandtzaeg, P. Human secretory immunoglobulin M. An immunochemical and immunohistochemical study. Immunology 1975, 29, 559–570. [Google Scholar]
- Mantis, N.J.; Rol, N.; Corthesy, B. Secretory IgA’s complex roles in immunity and mucosal homeostasis in the gut. Mucosal Immunol. 2011, 4, 603–611. [Google Scholar] [CrossRef] [Green Version]
- Brandtzaeg, P.; Pabst, R. Let’s go mucosal: Communication on slippery ground. Trends Immunol. 2004, 25, 570–577. [Google Scholar] [CrossRef]
- de Kivit, S.; van Hoffen, E.; Korthagen, N.; Garssen, J.; Willemsen, L.E. Apical TLR ligation of intestinal epithelial cells drives a Th1-polarized regulatory or inflammatory type effector response in vitro. Immunobiology 2011, 216, 518–527. [Google Scholar] [CrossRef]
- Neish, A.S.; Gewirtz, A.T.; Zeng, H.; Young, A.N.; Hobert, M.E.; Karmali, V.; Rao, A.S.; Madara, J.L. Prokaryotic regulation of epithelial responses by inhibition of IkappaB-alpha ubiquitination. Science 2000, 289, 1560–1563. [Google Scholar] [CrossRef] [PubMed]
- Artis, D. Epithelial-cell recognition of commensal bacteria and maintenance of immune homeostasis in the gut. Nat. Rev. Immunol. 2008, 8, 411–420. [Google Scholar] [CrossRef]
- Mora, J.R.; Iwata, M.; Eksteen, B.; Song, S.Y.; Junt, T.; Senman, B.; Otipoby, K.L.; Yokota, A.; Takeuchi, H.; Ricciardi-Castagnoli, P.; et al. Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science 2006, 314, 1157–1160. [Google Scholar] [CrossRef] [PubMed]
- Brandtzaeg, P. Function of mucosa-associated lymphoid tissue in antibody formation. Immunol. Investig. 2010, 39, 303–355. [Google Scholar] [CrossRef] [PubMed]
- Barone, F.; Patel, P.; Sanderson, J.D.; Spencer, J. Gut-associated lymphoid tissue contains the molecular machinery to support T-cell-dependent and T-cell-independent class switch recombination. Mucosal Immunol. 2009, 2, 495–503. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Seo, G.Y.; Jang, Y.S.; Kim, J.; Choe, J.; Han, H.J.; Lee, J.M.; Kang, S.H.; Rhee, K.J.; Park, S.R.; Kim, W.S.; et al. Retinoic acid acts as a selective human IgA switch factor. Hum. Immunol. 2014, 75, 923–929. [Google Scholar] [CrossRef]
- Guo, C.J.; Allen, B.M.; Hiam, K.J.; Dodd, D.; van Treuren, W.; Higginbottom, S.; Nagashima, K.; Fischer, C.R.; Sonnenburg, J.L.; Spitzer, M.H.; et al. Depletion of microbiome-derived molecules in the host using Clostridium genetics. Science 2019, 366, eaav1282. [Google Scholar] [CrossRef]
- Dillon, S.M.; Lee, E.J.; Kotter, C.V.; Austin, G.L.; Dong, Z.; Hecht, D.K.; Gianella, S.; Siewe, B.; Smith, D.M.; Landay, A.L.; et al. An altered intestinal mucosal microbiome in HIV-1 infection is associated with mucosal and systemic immune activation and endotoxemia. Mucosal Immunol. 2014, 7, 983–994. [Google Scholar] [CrossRef] [Green Version]
- Morgan, X.C.; Tickle, T.L.; Sokol, H.; Gevers, D.; Devaney, K.L.; Ward, D.V.; Reyes, J.A.; Shah, S.A.; LeLeiko, N.; Snapper, S.B.; et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012, 13, R79. [Google Scholar] [CrossRef]
- Saulnier, D.M.; Riehle, K.; Mistretta, T.A.; Diaz, M.A.; Mandal, D.; Raza, S.; Weidler, E.M.; Qin, X.; Coarfa, C.; Milosavljevic, A.; et al. Gastrointestinal microbiome signatures of pediatric patients with irritable bowel syndrome. Gastroenterology 2011, 141, 1782–1791. [Google Scholar] [CrossRef] [Green Version]
- Pietrzak, B.; Tomela, K.; Olejnik-Schmidt, A.; Mackiewicz, A.; Schmidt, M. Secretory IgA in Intestinal Mucosal Secretions as an Adaptive Barrier against Microbial Cells. Int. J. Mol. Sci. 2020, 21, 9254. [Google Scholar] [CrossRef] [PubMed]
- Erkocoglu, M.; Metin, A.; Kaya, A.; Ozcan, C.; Akan, A.; Civelek, E.; Capanoglu, M.; Ginis, T.; Kocabas, C.N. Allergic and autoimmune disorders in families with selective IgA deficiency. Turk. J. Med. Sci. 2017, 47, 592–598. [Google Scholar] [CrossRef] [PubMed]
- Yel, L. Selective IgA deficiency. J. Clin. Immunol. 2010, 30, 10–16. [Google Scholar] [CrossRef] [PubMed]
- Brandtzaeg, P.; Karlsson, G.; Hansson, G.; Petruson, B.; Bjorkander, J.; Hanson, L.A. The clinical condition of IgA-deficient patients is related to the proportion of IgD- and IgM-producing cells in their nasal mucosa. Clin. Exp. Immunol. 1987, 67, 626–636. [Google Scholar]
- Steinman, R.M.; Cohn, Z.A. Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, quantitation, tissue distribution. J. Exp. Med. 1973, 137, 1142–1162. [Google Scholar] [CrossRef] [Green Version]
- Romani, N.; Gruner, S.; Brang, D.; Kampgen, E.; Lenz, A.; Trockenbacher, B.; Konwalinka, G.; Fritsch, P.O.; Steinman, R.M.; Schuler, G. Proliferating dendritic cell progenitors in human blood. J. Exp. Med. 1994, 180, 83–93. [Google Scholar] [CrossRef] [Green Version]
- Asselin-Paturel, C.; Boonstra, A.; Dalod, M.; Durand, I.; Yessaad, N.; Dezutter-Dambuyant, C.; Vicari, A.; O’Garra, A.; Biron, C.; Briere, F.; et al. Mouse type I IFN-producing cells are immature APCs with plasmacytoid morphology. Nat. Immunol. 2001, 2, 1144–1150. [Google Scholar] [CrossRef]
- Jung, S.; Unutmaz, D.; Wong, P.; Sano, G.; de los Santos, K.; Sparwasser, T.; Wu, S.; Vuthoori, S.; Ko, K.; Zavala, F.; et al. In Vivo depletion of CD11c+ dendritic cells abrogates priming of CD8+ T cells by exogenous cell-associated antigens. Immunity 2002, 17, 211–220. [Google Scholar] [CrossRef] [Green Version]
- Allenspach, E.J.; Lemos, M.P.; Porrett, P.M.; Turka, L.A.; Laufer, T.M. Migratory and lymphoid-resident dendritic cells cooperate to efficiently prime naive CD4 T cells. Immunity 2008, 29, 795–806. [Google Scholar] [CrossRef] [Green Version]
- Piccioli, D.; Tavarini, S.; Borgogni, E.; Steri, V.; Nuti, S.; Sammicheli, C.; Bardelli, M.; Montagna, D.; Locatelli, F.; Wack, A. Functional specialization of human circulating CD16 and CD1c myeloid dendritic-cell subsets. Blood 2007, 109, 5371–5379. [Google Scholar] [CrossRef] [Green Version]
- Inaba, K.; Inaba, M.; Naito, M.; Steinman, R.M. Dendritic cell progenitors phagocytose particulates, including bacillus Calmette-Guerin organisms, and sensitize mice to mycobacterial antigens in vivo. J. Exp. Med. 1993, 178, 479–488. [Google Scholar] [CrossRef] [PubMed]
- Munawara, U.; Perveen, K.; Small, A.G.; Putty, T.; Quach, A.; Gorgani, N.N.; Hii, C.S.; Abbott, C.A.; Ferrante, A. Human Dendritic Cells Express the Complement Receptor Immunoglobulin Which Regulates T Cell Responses. Front. Immunol. 2019, 10, 2892. [Google Scholar] [CrossRef] [PubMed]
- Sallusto, F.; Cella, M.; Danieli, C.; Lanzavecchia, A. Dendritic cells use macropinocytosis and the mannose receptor to concentrate macromolecules in the major histocompatibility complex class II compartment: Downregulation by cytokines and bacterial products. J. Exp. Med. 1995, 182, 389–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Colledge, L.; Bennett, C.L.; Reay, P.A.; Blackburn, C.C. Rapid constitutive generation of a specific peptide-MHC class II complex from intact exogenous protein in immature murine dendritic cells. Eur. J. Immunol. 2002, 32, 3246–3255. [Google Scholar] [CrossRef]
- Caparros, E.; Munoz, P.; Sierra-Filardi, E.; Serrano-Gomez, D.; Puig-Kroger, A.; Rodriguez-Fernandez, J.L.; Mellado, M.; Sancho, J.; Zubiaur, M.; Corbi, A.L. DC-SIGN ligation on dendritic cells results in ERK and PI3K activation and modulates cytokine production. Blood 2006, 107, 3950–3958. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ouaaz, F.; Arron, J.; Zheng, Y.; Choi, Y.; Beg, A.A. Dendritic cell development and survival require distinct NF-kappaB subunits. Immunity 2002, 16, 257–270. [Google Scholar] [CrossRef] [Green Version]
- Tseng, S.Y.; Otsuji, M.; Gorski, K.; Huang, X.; Slansky, J.E.; Pai, S.I.; Shalabi, A.; Shin, T.; Pardoll, D.M.; Tsuchiya, H. B7-DC, a new dendritic cell molecule with potent costimulatory properties for T cells. J. Exp. Med. 2001, 193, 839–846. [Google Scholar] [CrossRef]
- Sallusto, F.; Schaerli, P.; Loetscher, P.; Schaniel, C.; Lenig, D.; Mackay, C.R.; Qin, S.; Lanzavecchia, A. Rapid and coordinated switch in chemokine receptor expression during dendritic cell maturation. Eur. J. Immunol. 1998, 28, 2760–2769. [Google Scholar] [CrossRef]
- Proietto, A.I.; O’Keeffe, M.; Gartlan, K.; Wright, M.D.; Shortman, K.; Wu, L.; Lahoud, M.H. Differential production of inflammatory chemokines by murine dendritic cell subsets. Immunobiology 2004, 209, 163–172. [Google Scholar] [CrossRef]
- Oppmann, B.; Lesley, R.; Blom, B.; Timans, J.C.; Xu, Y.; Hunte, B.; Vega, F.; Yu, N.; Wang, J.; Singh, K.; et al. Novel p19 protein engages IL-12p40 to form a cytokine, IL-23, with biological activities similar as well as distinct from IL-12. Immunity 2000, 13, 715–725. [Google Scholar] [CrossRef] [Green Version]
- Hsieh, C.S.; Macatonia, S.E.; Tripp, C.S.; Wolf, S.F.; O’Garra, A.; Murphy, K.M. Development of TH1 CD4+ T cells through IL-12 produced by Listeria-induced macrophages. Science 1993, 260, 547–549. [Google Scholar] [CrossRef] [PubMed]
- Jang, M.H.; Sougawa, N.; Tanaka, T.; Hirata, T.; Hiroi, T.; Tohya, K.; Guo, Z.; Umemoto, E.; Ebisuno, Y.; Yang, B.G.; et al. CCR7 is critically important for migration of dendritic cells in intestinal lamina propria to mesenteric lymph nodes. J. Immunol. 2006, 176, 803–810. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chirdo, F.G.; Millington, O.R.; Beacock-Sharp, H.; Mowat, A.M. Immunomodulatory dendritic cells in intestinal lamina propria. Eur. J. Immunol. 2005, 35, 1831–1840. [Google Scholar] [CrossRef] [PubMed]
- Kelsall, B.L.; Strober, W. Distinct populations of dendritic cells are present in the subepithelial dome and T cell regions of the murine Peyer’s patch. J. Exp. Med. 1996, 183, 237–247. [Google Scholar] [CrossRef]
- Coombes, J.L.; Siddiqui, K.R.; Arancibia-Carcamo, C.V.; Hall, J.; Sun, C.M.; Belkaid, Y.; Powrie, F. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J. Exp. Med. 2007, 204, 1757–1764. [Google Scholar] [CrossRef]
- Mora, J.R.; Bono, M.R.; Manjunath, N.; Weninger, W.; Cavanagh, L.L.; Rosemblatt, M.; von Andrian, U.H. Selective imprinting of gut-homing T cells by Peyer’s patch dendritic cells. Nature 2003, 424, 88–93. [Google Scholar] [CrossRef]
- Meredith, M.M.; Liu, K.; Darrasse-Jeze, G.; Kamphorst, A.O.; Schreiber, H.A.; Guermonprez, P.; Idoyaga, J.; Cheong, C.; Yao, K.H.; Niec, R.E.; et al. Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage. J. Exp. Med. 2012, 209, 1153–1165. [Google Scholar] [CrossRef] [Green Version]
- Guilliams, M.; Ginhoux, F.; Jakubzick, C.; Naik, S.H.; Onai, N.; Schraml, B.U.; Segura, E.; Tussiwand, R.; Yona, S. Dendritic cells, monocytes and macrophages: A unified nomenclature based on ontogeny. Nat. Rev. Immunol. 2014, 14, 571–578. [Google Scholar] [CrossRef] [Green Version]
- Shortman, K.; Liu, Y.J. Mouse and human dendritic cell subtypes. Nat. Rev. Immunol. 2002, 2, 151–161. [Google Scholar] [CrossRef]
- Zeng, R.; Oderup, C.; Yuan, R.; Lee, M.; Habtezion, A.; Hadeiba, H.; Butcher, E.C. Retinoic acid regulates the development of a gut-homing precursor for intestinal dendritic cells. Mucosal Immunol. 2013, 6, 847–856. [Google Scholar] [CrossRef] [Green Version]
- Zhang, Z.; Li, J.; Zheng, W.; Zhao, G.; Zhang, H.; Wang, X.; Guo, Y.; Qin, C.; Shi, Y. Peripheral Lymphoid Volume Expansion and Maintenance Are Controlled by Gut Microbiota via RALDH+ Dendritic Cells. Immunity 2016, 44, 330–342. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zeng, R.; Bscheider, M.; Lahl, K.; Lee, M.; Butcher, E.C. Generation and transcriptional programming of intestinal dendritic cells: Essential role of retinoic acid. Mucosal Immunol. 2016, 9, 183–193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goverse, G.; Molenaar, R.; Macia, L.; Tan, J.; Erkelens, M.N.; Konijn, T.; Knippenberg, M.; Cook, E.C.; Hanekamp, D.; Veldhoen, M.; et al. Diet-Derived Short Chain Fatty Acids Stimulate Intestinal Epithelial Cells To Induce Mucosal Tolerogenic Dendritic Cells. J. Immunol. 2017, 198, 2172–2181. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Penna, G.; Vulcano, M.; Roncari, A.; Facchetti, F.; Sozzani, S.; Adorini, L. Cutting edge: Differential chemokine production by myeloid and plasmacytoid dendritic cells. J. Immunol. 2002, 169, 6673–6676. [Google Scholar] [CrossRef]
- Cella, M.; Jarrossay, D.; Facchetti, F.; Alebardi, O.; Nakajima, H.; Lanzavecchia, A.; Colonna, M. Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I interferon. Nat. Med. 1999, 5, 919–923. [Google Scholar] [CrossRef]
- Young, L.J.; Wilson, N.S.; Schnorrer, P.; Proietto, A.; Broeke, T.T.; Matsuki, Y.; Mount, A.M.; Belz, G.T.; O’Keeffe, M.; Ohmura-Hoshino, M.; et al. Differential MHC class II synthesis and ubiquitination confers distinct antigen-presenting properties on conventional and plasmacytoid dendritic cells. Nat. Immunol. 2008, 9, 1244–1252. [Google Scholar] [CrossRef]
- GeurtsvanKessel, C.H.; Willart, M.A.; van Rijt, L.S.; Muskens, F.; Kool, M.; Baas, C.; Thielemans, K.; Bennett, C.; Clausen, B.E.; Hoogsteden, H.C.; et al. Clearance of influenza virus from the lung depends on migratory langerin+CD11b- but not plasmacytoid dendritic cells. J. Exp. Med. 2008, 205, 1621–1634. [Google Scholar] [CrossRef] [Green Version]
- Tel, J.; Schreibelt, G.; Sittig, S.P.; Mathan, T.S.; Buschow, S.I.; Cruz, L.J.; Lambeck, A.J.; Figdor, C.G.; de Vries, I.J. Human plasmacytoid dendritic cells efficiently cross-present exogenous Ags to CD8+ T cells despite lower Ag uptake than myeloid dendritic cell subsets. Blood 2013, 121, 459–467. [Google Scholar] [CrossRef] [Green Version]
- Dzionek, A.; Sohma, Y.; Nagafune, J.; Cella, M.; Colonna, M.; Facchetti, F.; Gunther, G.; Johnston, I.; Lanzavecchia, A.; Nagasaka, T.; et al. BDCA-2, a novel plasmacytoid dendritic cell-specific type II C-type lectin, mediates antigen capture and is a potent inhibitor of interferon alpha/beta induction. J. Exp. Med. 2001, 194, 1823–1834. [Google Scholar] [CrossRef]
- Meyer-Wentrup, F.; Benitez-Ribas, D.; Tacken, P.J.; Punt, C.J.; Figdor, C.G.; de Vries, I.J.; Adema, G.J. Targeting DCIR on human plasmacytoid dendritic cells results in antigen presentation and inhibits IFN-alpha production. Blood 2008, 111, 4245–4253. [Google Scholar] [CrossRef] [Green Version]
- Tel, J.; Smits, E.L.; Anguille, S.; Joshi, R.N.; Figdor, C.G.; de Vries, I.J. Human plasmacytoid dendritic cells are equipped with antigen-presenting and tumoricidal capacities. Blood 2012, 120, 3936–3944. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Honda, K.; Yanai, H.; Negishi, H.; Asagiri, M.; Sato, M.; Mizutani, T.; Shimada, N.; Ohba, Y.; Takaoka, A.; Yoshida, N.; et al. IRF-7 is the master regulator of type-I interferon-dependent immune responses. Nature 2005, 434, 772–777. [Google Scholar] [CrossRef] [PubMed]
- Hjorton, K.; Hagberg, N.; Israelsson, E.; Jinton, L.; Berggren, O.; Sandling, J.K.; Thorn, K.; Mo, J.; Consortium, D.; Eloranta, M.L.; et al. Cytokine production by activated plasmacytoid dendritic cells and natural killer cells is suppressed by an IRAK4 inhibitor. Arthritis Res. Ther. 2018, 20, 238. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- O’Keeffe, M.; Hochrein, H.; Vremec, D.; Caminschi, I.; Miller, J.L.; Anders, E.M.; Wu, L.; Lahoud, M.H.; Henri, S.; Scott, B.; et al. Mouse plasmacytoid cells: Long-lived cells, heterogeneous in surface phenotype and function, that differentiate into CD8(+) dendritic cells only after microbial stimulus. J. Exp. Med. 2002, 196, 1307–1319. [Google Scholar] [CrossRef] [PubMed]
- Grouard, G.; Rissoan, M.C.; Filgueira, L.; Durand, I.; Banchereau, J.; Liu, Y.J. The enigmatic plasmacytoid T cells develop into dendritic cells with interleukin (IL)-3 and CD40-ligand. J. Exp. Med. 1997, 185, 1101–1111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Contractor, N.; Louten, J.; Kim, L.; Biron, C.A.; Kelsall, B.L. Cutting edge: Peyer’s patch plasmacytoid dendritic cells (pDCs) produce low levels of type I interferons: Possible role for IL-10, TGFbeta, and prostaglandin E2 in conditioning a unique mucosal pDC phenotype. J. Immunol. 2007, 179, 2690–2694. [Google Scholar] [CrossRef] [Green Version]
- Wendland, M.; Czeloth, N.; Mach, N.; Malissen, B.; Kremmer, E.; Pabst, O.; Forster, R. CCR9 is a homing receptor for plasmacytoid dendritic cells to the small intestine. Proc. Natl. Acad. Sci. USA 2007, 104, 6347–6352. [Google Scholar] [CrossRef] [Green Version]
- Tezuka, H.; Abe, Y.; Asano, J.; Sato, T.; Liu, J.; Iwata, M.; Ohteki, T. Prominent role for plasmacytoid dendritic cells in mucosal T cell-independent IgA induction. Immunity 2011, 34, 247–257. [Google Scholar] [CrossRef] [Green Version]
- Kang, H.K.; Liu, M.; Datta, S.K. Low-dose peptide tolerance therapy of lupus generates plasmacytoid dendritic cells that cause expansion of autoantigen-specific regulatory T cells and contraction of inflammatory Th17 cells. J. Immunol. 2007, 178, 7849–7858. [Google Scholar] [CrossRef] [Green Version]
- Kuwana, M.; Kaburaki, J.; Wright, T.M.; Kawakami, Y.; Ikeda, Y. Induction of antigen-specific human CD4(+) T cell anergy by peripheral blood DC2 precursors. Eur. J. Immunol. 2001, 31, 2547–2557. [Google Scholar] [CrossRef]
- Baumgart, D.C.; Metzke, D.; Guckelberger, O.; Pascher, A.; Grotzinger, C.; Przesdzing, I.; Dorffel, Y.; Schmitz, J.; Thomas, S. Aberrant plasmacytoid dendritic cell distribution and function in patients with Crohn’s disease and ulcerative colitis. Clin. Exp. Immunol. 2011, 166, 46–54. [Google Scholar] [CrossRef] [PubMed]
- Patterson, S.; Rae, A.; Hockey, N.; Gilmour, J.; Gotch, F. Plasmacytoid dendritic cells are highly susceptible to human immunodeficiency virus type 1 infection and release infectious virus. J. Virol. 2001, 75, 6710–6713. [Google Scholar] [CrossRef] [Green Version]
- Boichuk, S.V.; Khaiboullina, S.F.; Ramazanov, B.R.; Khasanova, G.R.; Ivanovskaya, K.A.; Nizamutdinov, E.Z.; Sharafutdinov, M.R.; Martynova, E.V.; DeMeirleir, K.L.; Hulstaert, J.; et al. Gut-Associated Plasmacytoid Dendritic Cells Display an Immature Phenotype and Upregulated Granzyme B in Subjects with HIV/AIDS. Front. immunol. 2015, 6, 485. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dzionek, A.; Fuchs, A.; Schmidt, P.; Cremer, S.; Zysk, M.; Miltenyi, S.; Buck, D.W.; Schmitz, J. BDCA-2, BDCA-3, and BDCA-4: Three markers for distinct subsets of dendritic cells in human peripheral blood. J. Immunol. 2000, 165, 6037–6046. [Google Scholar] [CrossRef] [PubMed]
- de Heer, H.J.; Hammad, H.; Soullie, T.; Hijdra, D.; Vos, N.; Willart, M.A.; Hoogsteden, H.C.; Lambrecht, B.N. Essential role of lung plasmacytoid dendritic cells in preventing asthmatic reactions to harmless inhaled antigen. J. Exp. Med. 2004, 200, 89–98. [Google Scholar] [CrossRef] [PubMed]
- Wollenberg, A.; Wagner, M.; Gunther, S.; Towarowski, A.; Tuma, E.; Moderer, M.; Rothenfusser, S.; Wetzel, S.; Endres, S.; Hartmann, G. Plasmacytoid dendritic cells: A new cutaneous dendritic cell subset with distinct role in inflammatory skin diseases. J. Investig. Dermatol. 2002, 119, 1096–1102. [Google Scholar] [CrossRef] [Green Version]
- Zhang, C.; Yang, M.; Ericsson, A.C. Function of Macrophages in Disease: Current Understanding on Molecular Mechanisms. Front. Immunol. 2021, 12, 620510. [Google Scholar] [CrossRef]
- Yona, S.; Kim, K.W.; Wolf, Y.; Mildner, A.; Varol, D.; Breker, M.; Strauss-Ayali, D.; Viukov, S.; Guilliams, M.; Misharin, A.; et al. Fate mapping reveals origins and dynamics of monocytes and tissue macrophages under homeostasis. Immunity 2013, 38, 79–91. [Google Scholar] [CrossRef] [Green Version]
- Mantovani, A.; Sica, A.; Sozzani, S.; Allavena, P.; Vecchi, A.; Locati, M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004, 25, 677–686. [Google Scholar] [CrossRef]
- Gordon, S.; Martinez, F.O. Alternative activation of macrophages: Mechanism and functions. Immunity 2010, 32, 593–604. [Google Scholar] [CrossRef] [Green Version]
- Xue, J.; Schmidt, S.V.; Sander, J.; Draffehn, A.; Krebs, W.; Quester, I.; De Nardo, D.; Gohel, T.D.; Emde, M.; Schmidleithner, L.; et al. Transcriptome-based network analysis reveals a spectrum model of human macrophage activation. Immunity 2014, 40, 274–288. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, S.H.; Starkey, P.M.; Gordon, S. Quantitative analysis of total macrophage content in adult mouse tissues. Immunochemical studies with monoclonal antibody F4/80. J. Exp. Med. 1985, 161, 475–489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Viola, M.F.; Boeckxstaens, G. Niche-specific functional heterogeneity of intestinal resident macrophages. Gut 2021, 70, 1383–1395. [Google Scholar] [CrossRef] [PubMed]
- Bain, C.C.; Bravo-Blas, A.; Scott, C.L.; Perdiguero, E.G.; Geissmann, F.; Henri, S.; Malissen, B.; Osborne, L.C.; Artis, D.; Mowat, A.M. Constant replenishment from circulating monocytes maintains the macrophage pool in the intestine of adult mice. Nat. Immunol. 2014, 15, 929–937. [Google Scholar] [CrossRef]
- de Schepper, S.; Verheijden, S.; Aguilera-Lizarraga, J.; Viola, M.F.; Boesmans, W.; Stakenborg, N.; Voytyuk, I.; Schmidt, I.; Boeckx, B.; de Casterle, I.D.; et al. Self-Maintaining Gut Macrophages Are Essential for Intestinal Homeostasis. Cell 2019, 176, 676. [Google Scholar] [CrossRef] [Green Version]
- Bujko, A.; Atlasy, N.; Landsverk, O.J.B.; Richter, L.; Yaqub, S.; Horneland, R.; Oyen, O.; Aandahl, E.M.; Aabakken, L.; Stunnenberg, H.G.; et al. Transcriptional and functional profiling defines human small intestinal macrophage subsets. J. Exp. Med. 2018, 215, 441–458. [Google Scholar] [CrossRef] [Green Version]
- Bharat, A.; Bhorade, S.M.; Morales-Nebreda, L.; McQuattie-Pimentel, A.C.; Soberanes, S.; Ridge, K.; DeCamp, M.M.; Mestan, K.K.; Perlman, H.; Budinger, G.R.; et al. Flow Cytometry Reveals Similarities Between Lung Macrophages in Humans and Mice. Am. J. Respir. Cell. Mol. Biol. 2016, 54, 147–149. [Google Scholar] [CrossRef] [Green Version]
- Schridde, A.; Bain, C.C.; Mayer, J.U.; Montgomery, J.; Pollet, E.; Denecke, B.; Milling, S.W.F.; Jenkins, S.J.; Dalod, M.; Henri, S.; et al. Tissue-specific differentiation of colonic macrophages requires TGFbeta receptor-mediated signaling. Mucosal Immunol. 2017, 10, 1387–1399. [Google Scholar] [CrossRef] [Green Version]
- Shaw, T.N.; Houston, S.A.; Wemyss, K.; Bridgeman, H.M.; Barbera, T.A.; Zangerle-Murray, T.; Strangward, P.; Ridley, A.J.L.; Wang, P.; Tamoutounour, S.; et al. Tissue-resident macrophages in the intestine are long lived and defined by Tim-4 and CD4 expression. J. Exp. Med. 2018, 215, 1507–1518. [Google Scholar] [CrossRef]
- Hume, D.A.; Allan, W.; Hogan, P.G.; Doe, W.F. Immunohistochemical characterisation of macrophages in human liver and gastrointestinal tract: Expression of CD4, HLA-DR, OKM1, and the mature macrophage marker 25F9 in normal and diseased tissue. J. Leukoc. Biol. 1987, 42, 474–484. [Google Scholar] [CrossRef]
- Bain, C.C.; Scott, C.L.; Uronen-Hansson, H.; Gudjonsson, S.; Jansson, O.; Grip, O.; Guilliams, M.; Malissen, B.; Agace, W.W.; Mowat, A.M. Resident and pro-inflammatory macrophages in the colon represent alternative context-dependent fates of the same Ly6Chi monocyte precursors. Mucosal Immunol. 2013, 6, 498–510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hume, D.A.; Perry, V.H.; Gordon, S. The mononuclear phagocyte system of the mouse defined by immunohistochemical localisation of antigen F4/80: Macrophages associated with epithelia. Anat. Rec. 1984, 210, 503–512. [Google Scholar] [CrossRef] [PubMed]
- Ueda, Y.; Kayama, H.; Jeon, S.G.; Kusu, T.; Isaka, Y.; Rakugi, H.; Yamamoto, M.; Takeda, K. Commensal microbiota induce LPS hyporesponsiveness in colonic macrophages via the production of IL-10. Int. Immunol. 2010, 22, 953–962. [Google Scholar] [CrossRef] [Green Version]
- Chieppa, M.; Rescigno, M.; Huang, A.Y.; Germain, R.N. Dynamic imaging of dendritic cell extension into the small bowel lumen in response to epithelial cell TLR engagement. J. Exp. Med. 2006, 203, 2841–2852. [Google Scholar] [CrossRef]
- Schulz, O.; Jaensson, E.; Persson, E.K.; Liu, X.; Worbs, T.; Agace, W.W.; Pabst, O. Intestinal CD103+, but not CX3CR1+, antigen sampling cells migrate in lymph and serve classical dendritic cell functions. J. Exp. Med. 2009, 206, 3101–3114. [Google Scholar] [CrossRef] [PubMed]
- Cummings, R.J.; Barbet, G.; Bongers, G.; Hartmann, B.M.; Gettler, K.; Muniz, L.; Furtado, G.C.; Cho, J.; Lira, S.A.; Blander, J.M. Different tissue phagocytes sample apoptotic cells to direct distinct homeostasis programs. Nature 2016, 539, 565–569. [Google Scholar] [CrossRef]
- Cerovic, V.; Houston, S.A.; Scott, C.L.; Aumeunier, A.; Yrlid, U.; Mowat, A.M.; Milling, S.W. Intestinal CD103(-) dendritic cells migrate in lymph and prime effector T cells. Mucosal Immunol. 2013, 6, 104–113. [Google Scholar] [CrossRef] [Green Version]
- Muntjewerff, E.M.; Meesters, L.D.; van den Bogaart, G. Antigen Cross-Presentation by Macrophages. Front. immunol. 2020, 11, 1276. [Google Scholar] [CrossRef]
- Mazzini, E.; Massimiliano, L.; Penna, G.; Rescigno, M. Oral tolerance can be established via gap junction transfer of fed antigens from CX3CR1(+) macrophages to CD103(+) dendritic cells. Immunity 2014, 40, 248–261. [Google Scholar] [CrossRef] [Green Version]
- Muller, P.A.; Koscso, B.; Rajani, G.M.; Stevanovic, K.; Berres, M.L.; Hashimoto, D.; Mortha, A.; Leboeuf, M.; Li, X.M.; Mucida, D.; et al. Crosstalk between Muscularis Macrophages and Enteric Neurons Regulates Gastrointestinal Motility. Cell 2014, 158, 1210. [Google Scholar] [CrossRef] [Green Version]
- van der Zanden, E.P.; Snoek, S.A.; Heinsbroek, S.E.; Stanisor, O.I.; Verseijden, C.; Boeckxstaens, G.E.; Peppelenbosch, M.P.; Greaves, D.R.; Gordon, S.; de Jonge, W.J. Vagus nerve activity augments intestinal macrophage phagocytosis via nicotinic acetylcholine receptor alpha4beta2. Gastroenterology 2009, 137, 1029–1039.e4. [Google Scholar] [CrossRef] [PubMed]
- Paul, S.; Lal, G. The Molecular Mechanism of Natural Killer Cells Function and Its Importance in Cancer Immunotherapy. Front. Immunol. 2017, 8, 1124. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lanier, L.L. Up on the tightrope: Natural killer cell activation and inhibition. Nat. Immunol. 2008, 9, 495–502. [Google Scholar] [CrossRef] [PubMed]
- Koutsakos, M.; McWilliam, H.E.G.; Aktepe, T.E.; Fritzlar, S.; Illing, P.T.; Mifsud, N.A.; Purcell, A.W.; Rockman, S.; Reading, P.C.; Vivian, J.P.; et al. Downregulation of MHC Class I Expression by Influenza A and B Viruses. Front. Immunol. 2019, 10, 1158. [Google Scholar] [CrossRef]
- Holmes, T.D.; El-Sherbiny, Y.M.; Davison, A.; Clough, S.L.; Blair, G.E.; Cook, G.P. A human NK cell activation/inhibition threshold allows small changes in the target cell surface phenotype to dramatically alter susceptibility to NK cells. J. Immunol. 2011, 186, 1538–1545. [Google Scholar] [CrossRef] [Green Version]
- Sordo-Bahamonde, C.; Lorenzo-Herrero, S.; Payer, A.R.; Gonzalez, S.; Lopez-Soto, A. Mechanisms of Apoptosis Resistance to NK Cell-Mediated Cytotoxicity in Cancer. Int. J. Mol. Sci. 2020, 21, 3726. [Google Scholar] [CrossRef]
- Metkar, S.S.; Wang, B.; Aguilar-Santelises, M.; Raja, S.M.; Uhlin-Hansen, L.; Podack, E.; Trapani, J.A.; Froelich, C.J. Cytotoxic cell granule-mediated apoptosis: Perforin delivers granzyme B-serglycin complexes into target cells without plasma membrane pore formation. Immunity 2002, 16, 417–428. [Google Scholar] [CrossRef] [Green Version]
- de la Roche, M.; Asano, Y.; Griffiths, G.M. Origins of the cytolytic synapse. Nat. Rev. Immunol. 2016, 16, 421–432. [Google Scholar] [CrossRef]
- Sonar, S.; Lal, G. Role of Tumor Necrosis Factor Superfamily in Neuroinflammation and Autoimmunity. Front. immunol. 2015, 6, 364. [Google Scholar] [CrossRef] [Green Version]
- MacFarlane, M.; Ahmad, M.; Srinivasula, S.M.; Fernandes-Alnemri, T.; Cohen, G.M.; Alnemri, E.S. Identification and molecular cloning of two novel receptors for the cytotoxic ligand TRAIL. J. Biol. Chem. 1997, 272, 25417–25420. [Google Scholar] [CrossRef] [Green Version]
- Vincenz, C.; Dixit, V.M. Fas-associated death domain protein interleukin-1beta-converting enzyme 2 (FLICE2), an ICE/Ced-3 homologue, is proximally involved in CD95- and p55-mediated death signaling. J. Biol. Chem. 1997, 272, 6578–6583. [Google Scholar] [CrossRef] [Green Version]
- Krug, A.; French, A.R.; Barchet, W.; Fischer, J.A.; Dzionek, A.; Pingel, J.T.; Orihuela, M.M.; Akira, S.; Yokoyama, W.M.; Colonna, M. TLR9-dependent recognition of MCMV by IPC and DC generates coordinated cytokine responses that activate antiviral NK cell function. Immunity 2004, 21, 107–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Takai, T.; Li, M.; Sylvestre, D.; Clynes, R.; Ravetch, J.V. FcR gamma chain deletion results in pleiotrophic effector cell defects. Cell 1994, 76, 519–529. [Google Scholar] [CrossRef]
- Reefman, E.; Kay, J.G.; Wood, S.M.; Offenhauser, C.; Brown, D.L.; Roy, S.; Stanley, A.C.; Low, P.C.; Manderson, A.P.; Stow, J.L. Cytokine secretion is distinct from secretion of cytotoxic granules in NK cells. J. Immunol. 2010, 184, 4852–4862. [Google Scholar] [CrossRef] [PubMed]
- Wang, R.; Jaw, J.J.; Stutzman, N.C.; Zou, Z.; Sun, P.D. Natural killer cell-produced IFN-gamma and TNF-alpha induce target cell cytolysis through up-regulation of ICAM-1. J. Leukoc. Biol. 2012, 91, 299–309. [Google Scholar] [CrossRef] [Green Version]
- Quatrini, L.; Chiesa, M.D.; Sivori, S.; Mingari, M.C.; Pende, D.; Moretta, L. Human NK cells, their receptors and function. Eur. J. Immunol. 2021, 51, 1566–1579. [Google Scholar] [CrossRef]
- Cooper, M.A.; Fehniger, T.A.; Caligiuri, M.A. The biology of human natural killer-cell subsets. Trends Immunol. 2001, 22, 633–640. [Google Scholar] [CrossRef]
- Carson, W.E.; Fehniger, T.A.; Caligiuri, M.A. CD56bright natural killer cell subsets: Characterization of distinct functional responses to interleukin-2 and the c-kit ligand. Eur. J. Immunol. 1997, 27, 354–360. [Google Scholar] [CrossRef]
- Trinchieri, G. Biology of natural killer cells. Adv. Immunol. 1989, 47, 187–376. [Google Scholar] [CrossRef]
- Satoh-Takayama, N.; Vosshenrich, C.A.; Lesjean-Pottier, S.; Sawa, S.; Lochner, M.; Rattis, F.; Mention, J.J.; Thiam, K.; Cerf-Bensussan, N.; Mandelboim, O.; et al. Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity 2008, 29, 958–970. [Google Scholar] [CrossRef]
- Dowling, L.R.; Strazzari, M.R.; Keely, S.; Kaiko, G.E. Enteric nervous system and intestinal epithelial regulation of the gut-brain axis. J. Allergy Clin. Immunol. 2022, 150, 513–522. [Google Scholar] [CrossRef] [PubMed]
- Gubert, C.; Gasparotto, J.; Morais, L.H. Convergent pathways of the gut microbiota-brain axis and neurodegenerative disorders. Gastroenterol. Rep. 2022, 10, goac017. [Google Scholar] [CrossRef] [PubMed]
- Serrano-Pozo, A.; Frosch, M.P.; Masliah, E.; Hyman, B.T. Neuropathological alterations in Alzheimer disease. Cold Spring Harb. Perspect. Med. 2011, 1, a006189. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kohler, C.A.; Maes, M.; Slyepchenko, A.; Berk, M.; Solmi, M.; Lanctot, K.L.; Carvalho, A.F. The Gut-Brain Axis, Including the Microbiome, Leaky Gut and Bacterial Translocation: Mechanisms and Pathophysiological Role in Alzheimer’s Disease. Curr. Pharm. Des. 2016, 22, 6152–6166. [Google Scholar] [CrossRef] [PubMed]
- Minter, M.R.; Hinterleitner, R.; Meisel, M.; Zhang, C.; Leone, V.; Zhang, X.; Oyler-Castrillo, P.; Zhang, X.; Musch, M.W.; Shen, X.; et al. Antibiotic-induced perturbations in microbial diversity during post-natal development alters amyloid pathology in an aged APPSWE/PS1DeltaE9 murine model of Alzheimer’s disease. Sci. Rep. 2017, 7, 10411. [Google Scholar] [CrossRef] [Green Version]
- Kobayashi, Y.; Sugahara, H.; Shimada, K.; Mitsuyama, E.; Kuhara, T.; Yasuoka, A.; Kondo, T.; Abe, K.; Xiao, J.Z. Therapeutic potential of Bifidobacterium breve strain A1 for preventing cognitive impairment in Alzheimer’s disease. Sci. Rep. 2017, 7, 13510. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, X.; Sun, G.; Feng, T.; Zhang, J.; Huang, X.; Wang, T.; Xie, Z.; Chu, X.; Yang, J.; Wang, H.; et al. Sodium oligomannate therapeutically remodels gut microbiota and suppresses gut bacterial amino acids-shaped neuroinflammation to inhibit Alzheimer’s disease progression. Cell Res. 2019, 29, 787–803. [Google Scholar] [CrossRef] [Green Version]
- Vogt, N.M.; Kerby, R.L.; Dill-McFarland, K.A.; Harding, S.J.; Merluzzi, A.P.; Johnson, S.C.; Carlsson, C.M.; Asthana, S.; Zetterberg, H.; Blennow, K.; et al. Gut microbiome alterations in Alzheimer’s disease. Sci. Rep. 2017, 7, 13537. [Google Scholar] [CrossRef] [Green Version]
- Maqsood, R.; Stone, T.W. The Gut-Brain Axis, BDNF, NMDA and CNS Disorders. Neurochem. Res. 2016, 41, 2819–2835. [Google Scholar] [CrossRef]
- Dagda, R.K.; Banerjee, T.D. Role of protein kinase A in regulating mitochondrial function and neuronal development: Implications to neurodegenerative diseases. Rev. Neurosci. 2015, 26, 359–370. [Google Scholar] [CrossRef] [Green Version]
- Horvath, K.; Papadimitriou, J.C.; Rabsztyn, A.; Drachenberg, C.; Tildon, J.T. Gastrointestinal abnormalities in children with autistic disorder. J. Pediatr. 1999, 135, 559–563. [Google Scholar] [CrossRef]
- McElhanon, B.O.; McCracken, C.; Karpen, S.; Sharp, W.G. Gastrointestinal symptoms in autism spectrum disorder: A meta-analysis. Pediatrics 2014, 133, 872–883. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- D’Eufemia, P.; Celli, M.; Finocchiaro, R.; Pacifico, L.; Viozzi, L.; Zaccagnini, M.; Cardi, E.; Giardini, O. Abnormal intestinal permeability in children with autism. Acta Paediatr. 1996, 85, 1076–1079. [Google Scholar] [CrossRef]
- de Magistris, L.; Familiari, V.; Pascotto, A.; Sapone, A.; Frolli, A.; Iardino, P.; Carteni, M.; de Rosa, M.; Francavilla, R.; Riegler, G.; et al. Alterations of the intestinal barrier in patients with autism spectrum disorders and in their first-degree relatives. J. Pediatr. Gastroenterol. Nutr. 2010, 51, 418–424. [Google Scholar] [CrossRef] [PubMed]
- de Angelis, M.; Piccolo, M.; Vannini, L.; Siragusa, S.; de Giacomo, A.; Serrazzanetti, D.I.; Cristofori, F.; Guerzoni, M.E.; Gobbetti, M.; Francavilla, R. Fecal microbiota and metabolome of children with autism and pervasive developmental disorder not otherwise specified. PLoS ONE 2013, 8, e76993. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Finegold, S.M.; Dowd, S.E.; Gontcharova, V.; Liu, C.; Henley, K.E.; Wolcott, R.D.; Youn, E.; Summanen, P.H.; Granpeesheh, D.; Dixon, D.; et al. Pyrosequencing study of fecal microflora of autistic and control children. Anaerobe 2010, 16, 444–453. [Google Scholar] [CrossRef]
- Parracho, H.M.; Bingham, M.O.; Gibson, G.R.; McCartney, A.L. Differences between the gut microflora of children with autistic spectrum disorders and that of healthy children. J. Med. Microbiol. 2005, 54, 987–991. [Google Scholar] [CrossRef]
- Finegold, S.M.; Molitoris, D.; Song, Y.; Liu, C.; Vaisanen, M.L.; Bolte, E.; McTeague, M.; Sandler, R.; Wexler, H.; Marlowe, E.M.; et al. Gastrointestinal microflora studies in late-onset autism. Clin. Infect. Dis. 2002, 35, S6–S16. [Google Scholar] [CrossRef]
- Ristori, M.V.; Quagliariello, A.; Reddel, S.; Ianiro, G.; Vicari, S.; Gasbarrini, A.; Putignani, L. Autism, Gastrointestinal Symptoms and Modulation of Gut Microbiota by Nutritional Interventions. Nutrients 2019, 11, 2812. [Google Scholar] [CrossRef] [Green Version]
- Garcia-Gutierrez, E.; Narbad, A.; Rodriguez, J.M. Autism Spectrum Disorder Associated With Gut Microbiota at Immune, Metabolomic, and Neuroactive Level. Front. Neurosci. 2020, 14, 578666. [Google Scholar] [CrossRef]
- Braniste, V.; Al-Asmakh, M.; Kowal, C.; Anuar, F.; Abbaspour, A.; Toth, M.; Korecka, A.; Bakocevic, N.; Ng, L.G.; Kundu, P.; et al. The gut microbiota influences blood-brain barrier permeability in mice. Sci. Transl. Med. 2014, 6, 263ra158. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Adams, J.B.; Johansen, L.J.; Powell, L.D.; Quig, D.; Rubin, R.A. Gastrointestinal flora and gastrointestinal status in children with autism--comparisons to typical children and correlation with autism severity. BMC Gastroenterol. 2011, 11, 22. [Google Scholar] [CrossRef] [Green Version]
- Grimaldi, R.; Gibson, G.R.; Vulevic, J.; Giallourou, N.; Castro-Mejia, J.L.; Hansen, L.H.; Leigh Gibson, E.; Nielsen, D.S.; Costabile, A. A prebiotic intervention study in children with autism spectrum disorders (ASDs). Microbiome 2018, 6, 133. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sandler, R.H.; Finegold, S.M.; Bolte, E.R.; Buchanan, C.P.; Maxwell, A.P.; Vaisanen, M.L.; Nelson, M.N.; Wexler, H.M. Short-term benefit from oral vancomycin treatment of regressive-onset autism. J. Child Neurol. 2000, 15, 429–435. [Google Scholar] [CrossRef] [PubMed]
- Vila, M.; Przedborski, S. Genetic clues to the pathogenesis of Parkinson’s disease. Nat. Med. 2004, 10, S58–S62. [Google Scholar] [CrossRef] [PubMed]
- Driver, J.A.; Logroscino, G.; Gaziano, J.M.; Kurth, T. Incidence and remaining lifetime risk of Parkinson disease in advanced age. Neurology 2009, 72, 432–438. [Google Scholar] [CrossRef]
- Maggi, G.; Trojano, L.; Barone, P.; Santangelo, G. Sleep Disorders and Cognitive Dysfunctions in Parkinson’s Disease: A Meta-Analytic Study. Neuropsychol. Rev. 2021, 31, 643–682. [Google Scholar] [CrossRef]
- Pardo-Rodriguez, M.; Bojorges-Valdez, E.; Yanez-Suarez, O. Disruption of the Cortical-Vagal Communication Network in Parkinson’s Disease. Annu. Int. Conf. IEEE Eng. Med. Biol. Soc. 2021, 2021, 5842–5845. [Google Scholar] [CrossRef]
- Pierzchlinska, A.; Kwasniak-Butowska, M.; Slawek, J.; Drozdzik, M.; Bialecka, M. Arterial Blood Pressure Variability and Other Vascular Factors Contribution to the Cognitive Decline in Parkinson’s Disease. Molecules 2021, 26, 1523. [Google Scholar] [CrossRef]
- Xu, Z.; Anderson, K.N.; Saffari, S.E.; Lawson, R.A.; Chaudhuri, K.R.; Brooks, D.; Pavese, N. Progression of sleep disturbances in Parkinson’s disease: A 5-year longitudinal study. J. Neurol. 2021, 268, 312–320. [Google Scholar] [CrossRef]
- Fasano, A.; Visanji, N.P.; Liu, L.W.; Lang, A.E.; Pfeiffer, R.F. Gastrointestinal dysfunction in Parkinson’s disease. Lancet Neurol. 2015, 14, 625–639. [Google Scholar] [CrossRef]
- Zhu, Y.; Yuan, M.; Liu, Y.; Yang, F.; Chen, W.Z.; Xu, Z.Z.; Xiang, Z.B.; Xu, R.S. Association between inflammatory bowel diseases and Parkinson’s disease: Systematic review and meta-analysis. Neural Regen. Res. 2022, 17, 344–353. [Google Scholar] [CrossRef] [PubMed]
- Dumitrescu, L.; Marta, D.; Danau, A.; Lefter, A.; Tulba, D.; Cozma, L.; Manole, E.; Gherghiceanu, M.; Ceafalan, L.C.; Popescu, B.O. Serum and Fecal Markers of Intestinal Inflammation and Intestinal Barrier Permeability Are Elevated in Parkinson’s Disease. Front. Neurosci. 2021, 15, 689723. [Google Scholar] [CrossRef]
- Su, Y.; Liu, N.; Zhang, Z.; Li, H.; Ma, J.; Yuan, Y.; Shi, M.; Liu, J.; Zhao, Z.; Zhang, Z.; et al. Cholecystokinin and glucagon-like peptide-1 analogues regulate intestinal tight junction, inflammation, dopaminergic neurons and alpha-synuclein accumulation in the colon of two Parkinson’s disease mouse models. Eur. J. Pharmacol. 2022, 926, 175029. [Google Scholar] [CrossRef] [PubMed]
- Clairembault, T.; Leclair-Visonneau, L.; Coron, E.; Bourreille, A.; le Dily, S.; Vavasseur, F.; Heymann, M.F.; Neunlist, M.; Derkinderen, P. Structural alterations of the intestinal epithelial barrier in Parkinson’s disease. Acta Neuropathol. Commun. 2015, 3, 12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Halliday, G.M.; del Tredici, K.; Braak, H. Critical appraisal of brain pathology staging related to presymptomatic and symptomatic cases of sporadic Parkinson’s disease. J. Neural Transm. 2006, 70, 99–103. [Google Scholar] [CrossRef]
- Forsyth, C.B.; Shannon, K.M.; Kordower, J.H.; Voigt, R.M.; Shaikh, M.; Jaglin, J.A.; Estes, J.D.; Dodiya, H.B.; Keshavarzian, A. Increased intestinal permeability correlates with sigmoid mucosa alpha-synuclein staining and endotoxin exposure markers in early Parkinson’s disease. PLoS ONE 2011, 6, e28032. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Unger, M.M.; Spiegel, J.; Dillmann, K.U.; Grundmann, D.; Philippeit, H.; Burmann, J.; Fassbender, K.; Schwiertz, A.; Schafer, K.H. Short chain fatty acids and gut microbiota differ between patients with Parkinson’s disease and age-matched controls. Parkinsonism Relat. Disord. 2016, 32, 66–72. [Google Scholar] [CrossRef]
- Scheperjans, F.; Aho, V.; Pereira, P.A.; Koskinen, K.; Paulin, L.; Pekkonen, E.; Haapaniemi, E.; Kaakkola, S.; Eerola-Rautio, J.; Pohja, M.; et al. Gut microbiota are related to Parkinson’s disease and clinical phenotype. Mov. Disord. Off. J. Mov. Disord. Soc. 2015, 30, 350–358. [Google Scholar] [CrossRef]
- Keshavarzian, A.; Green, S.J.; Engen, P.A.; Voigt, R.M.; Naqib, A.; Forsyth, C.B.; Mutlu, E.; Shannon, K.M. Colonic bacterial composition in Parkinson’s disease. Mov. Disord. Off. J. Mov. Disord. Soc. 2015, 30, 1351–1360. [Google Scholar] [CrossRef]
- Sampson, T.R.; Debelius, J.W.; Thron, T.; Janssen, S.; Shastri, G.G.; Ilhan, Z.E.; Challis, C.; Schretter, C.E.; Rocha, S.; Gradinaru, V.; et al. Gut Microbiota Regulate Motor Deficits and Neuroinflammation in a Model of Parkinson’s Disease. Cell 2016, 167, 1469–1480.e1412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ciaramella, A.; Salani, F.; Bizzoni, F.; Pontieri, F.E.; Stefani, A.; Pierantozzi, M.; Assogna, F.; Caltagirone, C.; Spalletta, G.; Bossu, P. Blood dendritic cell frequency declines in idiopathic Parkinson’s disease and is associated with motor symptom severity. PLoS ONE 2013, 8, e65352. [Google Scholar] [CrossRef] [PubMed]
- Liu, W.; Liu, X.; Li, Y.; Zhao, J.; Liu, Z.; Hu, Z.; Wang, Y.; Yao, Y.; Miller, A.W.; Su, B.; et al. LRRK2 promotes the activation of NLRC4 inflammasome during Salmonella Typhimurium infection. J. Exp. Med. 2017, 214, 3051–3066. [Google Scholar] [CrossRef] [Green Version]
- Gardet, A.; Benita, Y.; Li, C.; Sands, B.E.; Ballester, I.; Stevens, C.; Korzenik, J.R.; Rioux, J.D.; Daly, M.J.; Xavier, R.J.; et al. LRRK2 is involved in the IFN-gamma response and host response to pathogens. J. Immunol. 2010, 185, 5577–5585. [Google Scholar] [CrossRef] [PubMed]
- Daher, J.P.L. Interaction of LRRK2 and alpha-Synuclein in Parkinson’s Disease. Adv. Neurobiol. 2017, 14, 209–226. [Google Scholar] [CrossRef]
- Stolzenberg, E.; Berry, D.; Yang, D.; Lee, E.Y.; Kroemer, A.; Kaufman, S.; Wong, G.C.L.; Oppenheim, J.J.; Sen, S.; Fishbein, T.; et al. A Role for Neuronal Alpha-Synuclein in Gastrointestinal Immunity. J. Innate Immun. 2017, 9, 456–463. [Google Scholar] [CrossRef]
- Piancone, F.; Saresella, M.; la Rosa, F.; Marventano, I.; Meloni, M.; Navarro, J.; Clerici, M. Inflammatory Responses to Monomeric and Aggregated alpha-Synuclein in Peripheral Blood of Parkinson Disease Patients. Front. Neurosci. 2021, 15, 639646. [Google Scholar] [CrossRef]
- Barbut, D.; Stolzenberg, E.; Zasloff, M. Gastrointestinal Immunity and Alpha-Synuclein. J. Parkinsons Dis. 2019, 9, S313–S322. [Google Scholar] [CrossRef] [Green Version]
- Schmidt, S.H.; Knape, M.J.; Boassa, D.; Mumdey, N.; Kornev, A.P.; Ellisman, M.H.; Taylor, S.S.; Herberg, F.W. The dynamic switch mechanism that leads to activation of LRRK2 is embedded in the DFGpsi motif in the kinase domain. Proc. Natl. Acad. Sci. USA 2019, 116, 14979–14988. [Google Scholar] [CrossRef] [Green Version]
- Kasen, A.; Houck, C.; Burmeister, A.R.; Sha, Q.; Brundin, L.; Brundin, P. Upregulation of alpha-synuclein following immune activation: Possible trigger of Parkinson’s disease. Neurobiol. Dis. 2022, 166, 105654. [Google Scholar] [CrossRef]
- Carruthers, B.M.; van de Sande, M.I.; De Meirleir, K.L.; Klimas, N.G.; Broderick, G.; Mitchell, T.; Staines, D.; Powles, A.C.; Speight, N.; Vallings, R.; et al. Myalgic encephalomyelitis: International Consensus Criteria. J. Intern. Med. 2011, 270, 327–338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fukuda, K.; Straus, S.E.; Hickie, I.; Sharpe, M.C.; Dobbins, J.G.; Komaroff, A. The chronic fatigue syndrome: A comprehensive approach to its definition and study. International Chronic Fatigue Syndrome Study Group. Ann. Intern. Med. 1994, 121, 953–959. [Google Scholar] [CrossRef] [PubMed]
- Tomas, C.; Brown, A.; Strassheim, V.; Elson, J.L.; Newton, J.; Manning, P. Cellular bioenergetics is impaired in patients with chronic fatigue syndrome. PLoS ONE 2017, 12, e0186802. [Google Scholar] [CrossRef] [Green Version]
- Morris, G.; Maes, M. Mitochondrial dysfunctions in myalgic encephalomyelitis/chronic fatigue syndrome explained by activated immuno-inflammatory, oxidative and nitrosative stress pathways. Metab. Brain Dis. 2014, 29, 19–36. [Google Scholar] [CrossRef] [PubMed]
- Myhill, S.; Booth, N.E.; McLaren-Howard, J. Chronic fatigue syndrome and mitochondrial dysfunction. Int. J. Clin. Exp. Med. 2009, 2, 1–16. [Google Scholar]
- Klimas, N.G.; Koneru, A.O. Chronic fatigue syndrome: Inflammation, immune function, and neuroendocrine interactions. Curr. Rheumatol. Rep. 2007, 9, 482–487. [Google Scholar] [CrossRef]
- Konstantinov, K.; von Mikecz, A.; Buchwald, D.; Jones, J.; Gerace, L.; Tan, E.M. Autoantibodies to nuclear envelope antigens in chronic fatigue syndrome. J. Clin. Investig. 1996, 98, 1888–1896. [Google Scholar] [CrossRef] [Green Version]
- Loebel, M.; Grabowski, P.; Heidecke, H.; Bauer, S.; Hanitsch, L.G.; Wittke, K.; Meisel, C.; Reinke, P.; Volk, H.D.; Fluge, O.; et al. Antibodies to beta adrenergic and muscarinic cholinergic receptors in patients with Chronic Fatigue Syndrome. Brain Behav. Immun. 2016, 52, 32–39. [Google Scholar] [CrossRef] [Green Version]
- de Beeck, K.O.; Vermeersch, P.; Verschueren, P.; Westhovens, R.; Marien, G.; Blockmans, D.; Bossuyt, X. Antinuclear antibody detection by automated multiplex immunoassay in untreated patients at the time of diagnosis. Autoimmun. Rev. 2012, 12, 137–143. [Google Scholar] [CrossRef]
- Tanaka, S.; Kuratsune, H.; Hidaka, Y.; Hakariya, Y.; Tatsumi, K.I.; Takano, T.; Kanakura, Y.; Amino, N. Autoantibodies against muscarinic cholinergic receptor in chronic fatigue syndrome. Int. J. Mol. Med. 2003, 12, 225–230. [Google Scholar] [CrossRef]
- Singh, S.; Stafford, P.; Schlauch, K.A.; Tillett, R.R.; Gollery, M.; Johnston, S.A.; Khaiboullina, S.F.; de Meirleir, K.L.; Rawat, S.; Mijatovic, T.; et al. Humoral Immunity Profiling of Subjects with Myalgic Encephalomyelitis Using a Random Peptide Microarray Differentiates Cases from Controls with High Specificity and Sensitivity. Mol. Neurobiol. 2018, 55, 633–641. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fujii, H.; Sato, W.; Kimura, Y.; Matsuda, H.; Ota, M.; Maikusa, N.; Suzuki, F.; Amano, K.; Shin, I.; Yamamura, T.; et al. Altered Structural Brain Networks Related to Adrenergic/Muscarinic Receptor Autoantibodies in Chronic Fatigue Syndrome. J. Neuroimaging 2020, 30, 822–827. [Google Scholar] [CrossRef] [PubMed]
- Wirth, K.; Scheibenbogen, C. A Unifying Hypothesis of the Pathophysiology of Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS): Recognitions from the finding of autoantibodies against ss2-adrenergic receptors. Autoimmun. Rev. 2020, 19, 102527. [Google Scholar] [CrossRef] [PubMed]
- Salim, S.Y.; Soderholm, J.D. Importance of disrupted intestinal barrier in inflammatory bowel diseases. Inflamm. Bowel Dis. 2011, 17, 362–381. [Google Scholar] [CrossRef]
- Lupo, G.F.D.; Rocchetti, G.; Lucini, L.; Lorusso, L.; Manara, E.; Bertelli, M.; Puglisi, E.; Capelli, E. Potential role of microbiome in Chronic Fatigue Syndrome/Myalgic Encephalomyelits (CFS/ME). Sci. Rep. 2021, 11, 7043. [Google Scholar] [CrossRef]
- Maes, M.; Mihaylova, I.; Leunis, J.C. Increased serum IgA and IgM against LPS of enterobacteria in chronic fatigue syndrome (CFS): Indication for the involvement of gram-negative enterobacteria in the etiology of CFS and for the presence of an increased gut-intestinal permeability. J. Affect. Disord. 2007, 99, 237–240. [Google Scholar] [CrossRef]
- Giloteaux, L.; Goodrich, J.K.; Walters, W.A.; Levine, S.M.; Ley, R.E.; Hanson, M.R. Reduced diversity and altered composition of the gut microbiome in individuals with myalgic encephalomyelitis/chronic fatigue syndrome. Microbiome 2016, 4, 30. [Google Scholar] [CrossRef] [Green Version]
- Younas, M.; Psomas, C.; Reynes, C.; Cezar, R.; Kundura, L.; Portales, P.; Merle, C.; Atoui, N.; Fernandez, C.; le Moing, V.; et al. Microbial Translocation Is Linked to a Specific Immune Activation Profile in HIV-1-Infected Adults With Suppressed Viremia. Front. Immunol. 2019, 10, 2185. [Google Scholar] [CrossRef]
- Fremont, M.; Coomans, D.; Massart, S.; de Meirleir, K. High-throughput 16S rRNA gene sequencing reveals alterations of intestinal microbiota in myalgic encephalomyelitis/chronic fatigue syndrome patients. Anaerobe 2013, 22, 50–56. [Google Scholar] [CrossRef] [Green Version]
- Toubal, A.; Nel, I.; Lotersztajn, S.; Lehuen, A. Mucosal-associated invariant T cells and disease. Nat. Rev. Immunol. 2019, 19, 643–657. [Google Scholar] [CrossRef]
- Eckle, S.B.; Corbett, A.J.; Keller, A.N.; Chen, Z.; Godfrey, D.I.; Liu, L.; Mak, J.Y.; Fairlie, D.P.; Rossjohn, J.; McCluskey, J. Recognition of Vitamin B Precursors and Byproducts by Mucosal Associated Invariant T Cells. J. Biol. Chem. 2015, 290, 30204–30211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Gold, M.C.; Cerri, S.; Smyk-Pearson, S.; Cansler, M.E.; Vogt, T.M.; Delepine, J.; Winata, E.; Swarbrick, G.M.; Chua, W.J.; Yu, Y.Y.; et al. Human mucosal associated invariant T cells detect bacterially infected cells. PLoS Biol. 2010, 8, e1000407. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ussher, J.E.; Bilton, M.; Attwod, E.; Shadwell, J.; Richardson, R.; de Lara, C.; Mettke, E.; Kurioka, A.; Hansen, T.H.; Klenerman, P.; et al. CD161++ CD8+ T cells, including the MAIT cell subset, are specifically activated by IL-12+IL-18 in a TCR-independent manner. Eur. J. Immunol. 2014, 44, 195–203. [Google Scholar] [CrossRef] [PubMed]
- Serriari, N.E.; Eoche, M.; Lamotte, L.; Lion, J.; Fumery, M.; Marcelo, P.; Chatelain, D.; Barre, A.; Nguyen-Khac, E.; Lantz, O.; et al. Innate mucosal-associated invariant T (MAIT) cells are activated in inflammatory bowel diseases. Clin. Exp. Immunol. 2014, 176, 266–274. [Google Scholar] [CrossRef]
- Cliff, J.M.; King, E.C.; Lee, J.S.; Sepulveda, N.; Wolf, A.S.; Kingdon, C.; Bowman, E.; Dockrell, H.M.; Nacul, L.; Lacerda, E.; et al. Cellular Immune Function in Myalgic Encephalomyelitis/Chronic Fatigue Syndrome (ME/CFS). Front. Immunol. 2019, 10, 796. [Google Scholar] [CrossRef]
- Laufer, G.; Mayer, J.; Mueller, B.F.; Mueller-Lantzsch, N.; Ruprecht, K. Analysis of transcribed human endogenous retrovirus W env loci clarifies the origin of multiple sclerosis-associated retrovirus env sequences. Retrovirology 2009, 6, 37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- de Meirleir, K.L.; Khaiboullina, S.F.; Fremont, M.; Hulstaert, J.; Rizvanov, A.A.; Palotas, A.; Lombardi, V.C. Plasmacytoid dendritic cells in the duodenum of individuals diagnosed with myalgic encephalomyelitis are uniquely immunoreactive to antibodies to human endogenous retroviral proteins. In Vivo 2013, 27, 177–187. [Google Scholar]
- Bergallo, M.; Galliano, I.; Montanari, P.; Zaniol, E.; Graziano, E.; Calvi, C.; Alliaudi, C.; Dapra, V.; Savino, F. Modulation of human endogenous retroviruses -H, -W and -K transcription by microbes. Microbes Infect. 2020, 22, 366–370. [Google Scholar] [CrossRef]
- Yu, P.; Lubben, W.; Slomka, H.; Gebler, J.; Konert, M.; Cai, C.; Neubrandt, L.; da Costa, O.P.; Paul, S.; Dehnert, S.; et al. Nucleic acid-sensing Toll-like receptors are essential for the control of endogenous retrovirus viremia and ERV-induced tumors. Immunity 2012, 37, 867–879. [Google Scholar] [CrossRef] [Green Version]
- Ascherio, A. Environmental factors in multiple sclerosis. Expert Rev. Neurother. 2013, 13, 3–9. [Google Scholar] [CrossRef]
- Forte, G.I.; Ragonese, P.; Salemi, G.; Scola, L.; Candore, G.; D’Amelio, M.; Crivello, A.; Di Benedetto, N.; Nuzzo, D.; Giacalone, A.; et al. Search for genetic factors associated with susceptibility to multiple sclerosis. Ann. N. Y. Acad. Sci. 2006, 1067, 264–269. [Google Scholar] [CrossRef] [PubMed]
- Ghezzi, A.; Zaffaroni, M.; Caputo, D.; Marforio, S.; Montanini, R. Cerebral evoked potentials and cerebrospinal fluid examination in progressive spastic paraparesis (possible multiple sclerosis). Riv. Neurobiol. 1984, 30, 544–550. [Google Scholar] [PubMed]
- Levinthal, D.J.; Rahman, A.; Nusrat, S.; O’Leary, M.; Heyman, R.; Bielefeldt, K. Adding to the burden: Gastrointestinal symptoms and syndromes in multiple sclerosis. Mult. Scler. Int. 2013, 2013, 319201. [Google Scholar] [CrossRef] [Green Version]
- Chen, J.; Chia, N.; Kalari, K.R.; Yao, J.Z.; Novotna, M.; Soldan, M.M.; Luckey, D.H.; Marietta, E.V.; Jeraldo, P.R.; Chen, X.; et al. Multiple sclerosis patients have a distinct gut microbiota compared to healthy controls. Sci. Rep. 2016, 6, 28484. [Google Scholar] [CrossRef] [Green Version]
- Miyake, S.; Kim, S.; Suda, W.; Oshima, K.; Nakamura, M.; Matsuoka, T.; Chihara, N.; Tomita, A.; Sato, W.; Kim, S.W.; et al. Dysbiosis in the Gut Microbiota of Patients with Multiple Sclerosis, with a Striking Depletion of Species Belonging to Clostridia XIVa and IV Clusters. PLoS ONE 2015, 10, e0137429. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cantarel, B.L.; Waubant, E.; Chehoud, C.; Kuczynski, J.; DeSantis, T.Z.; Warrington, J.; Venkatesan, A.; Fraser, C.M.; Mowry, E.M. Gut microbiota in multiple sclerosis: Possible influence of immunomodulators. J. Investig. Med. 2015, 63, 729–734. [Google Scholar] [CrossRef] [Green Version]
- Ott, S.J.; Musfeldt, M.; Wenderoth, D.F.; Hampe, J.; Brant, O.; Folsch, U.R.; Timmis, K.N.; Schreiber, S. Reduction in diversity of the colonic mucosa associated bacterial microflora in patients with active inflammatory bowel disease. Gut 2004, 53, 685–693. [Google Scholar] [CrossRef] [Green Version]
- Manichanh, C.; Rigottier-Gois, L.; Bonnaud, E.; Gloux, K.; Pelletier, E.; Frangeul, L.; Nalin, R.; Jarrin, C.; Chardon, P.; Marteau, P.; et al. Reduced diversity of faecal microbiota in Crohn’s disease revealed by a metagenomic approach. Gut 2006, 55, 205–211. [Google Scholar] [CrossRef] [Green Version]
- Lee, Y.K.; Menezes, J.S.; Umesaki, Y.; Mazmanian, S.K. Proinflammatory T-cell responses to gut microbiota promote experimental autoimmune encephalomyelitis. Proc. Natl. Acad. Sci. USA 2010, 108, 4615–4622. [Google Scholar] [CrossRef] [Green Version]
- Joossens, M.; Huys, G.; Cnockaert, M.; de Preter, V.; Verbeke, K.; Rutgeerts, P.; Vandamme, P.; Vermeire, S. Dysbiosis of the faecal microbiota in patients with Crohn’s disease and their unaffected relatives. Gut 2011, 60, 631–637. [Google Scholar] [CrossRef] [Green Version]
- Walker, F.O. Huntington’s Disease. Semin. Neurol. 2007, 27, 143–150. [Google Scholar] [CrossRef] [PubMed]
- Ross, C.A.; Tabrizi, S.J. Huntington’s disease: From molecular pathogenesis to clinical treatment. Lancet Neurol. 2011, 10, 83–98. [Google Scholar] [CrossRef]
- Kong, G.; Cao, K.L.; Judd, L.M.; Li, S.; Renoir, T.; Hannan, A.J. Microbiome profiling reveals gut dysbiosis in a transgenic mouse model of Huntington’s disease. Neurobiol. Dis. 2020, 135, 104268. [Google Scholar] [CrossRef] [PubMed]
- Gubert, C.; Choo, J.M.; Love, C.J.; Kodikara, S.; Masson, B.A.; Liew, J.J.M.; Wang, Y.; Kong, G.; Narayana, V.K.; Renoir, T.; et al. Faecal microbiota transplant ameliorates gut dysbiosis and cognitive deficits in Huntington’s disease mice. Brain Commun. 2022, 4, fcac205. [Google Scholar] [CrossRef]
- Du, G.; Dong, W.; Yang, Q.; Yu, X.; Ma, J.; Gu, W.; Huang, Y. Altered Gut Microbiota Related to Inflammatory Responses in Patients With Huntington’s Disease. Front. Immunol. 2020, 11, 603594. [Google Scholar] [CrossRef] [PubMed]
- Stan, T.L.; Soylu-Kucharz, R.; Burleigh, S.; Prykhodko, O.; Cao, L.; Franke, N.; Sjogren, M.; Haikal, C.; Hallenius, F.; Bjorkqvist, M. Increased intestinal permeability and gut dysbiosis in the R6/2 mouse model of Huntington’s disease. Sci. Rep. 2020, 10, 18270. [Google Scholar] [CrossRef] [PubMed]
- Wasser, C.I.; Mercieca, E.C.; Kong, G.; Hannan, A.J.; McKeown, S.J.; Glikmann-Johnston, Y.; Stout, J.C. Gut dysbiosis in Huntington’s disease: Associations among gut microbiota, cognitive performance and clinical outcomes. Brain Commun. 2020, 2, fcaa110. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
Sterling, K.G.; Dodd, G.K.; Alhamdi, S.; Asimenios, P.G.; Dagda, R.K.; De Meirleir, K.L.; Hudig, D.; Lombardi, V.C. Mucosal Immunity and the Gut-Microbiota-Brain-Axis in Neuroimmune Disease. Int. J. Mol. Sci. 2022, 23, 13328. https://doi.org/10.3390/ijms232113328
Sterling KG, Dodd GK, Alhamdi S, Asimenios PG, Dagda RK, De Meirleir KL, Hudig D, Lombardi VC. Mucosal Immunity and the Gut-Microbiota-Brain-Axis in Neuroimmune Disease. International Journal of Molecular Sciences. 2022; 23(21):13328. https://doi.org/10.3390/ijms232113328
Chicago/Turabian StyleSterling, Kathryn G., Griffin Kutler Dodd, Shatha Alhamdi, Peter G. Asimenios, Ruben K. Dagda, Kenny L. De Meirleir, Dorothy Hudig, and Vincent C. Lombardi. 2022. "Mucosal Immunity and the Gut-Microbiota-Brain-Axis in Neuroimmune Disease" International Journal of Molecular Sciences 23, no. 21: 13328. https://doi.org/10.3390/ijms232113328