The Influence of Gut Microbiota on Oxidative Stress and the Immune System
Abstract
:1. Introduction
2. The Cellular REDOX Equilibrium
2.1. Reactive Oxygen (ROS)
2.2. Sources of ROS Generation
Cellular Compartment | ROS Source | ROS | Mechanism of Action | ROS-Related Disease | Role of Gut Microbiota |
---|---|---|---|---|---|
Mitochondria | Complex I respiratory chain | O2·− | Cellular signaling, immune cell activation, energy metabolism | Inflammatory bowel disease (IBD) and colorectal cancer (CRC) | Induced by bacterial antigens, regulated by bacterial metabolites [7,33] |
Mitochondria | Complex III respiratory chain | O2·− | Cellular signaling, immune cell activation, energy metabolism | IBD, CRC | Induced by bacterial antigens, regulated by bacterial metabolites [7,33] |
Plasma membrane, vesicular membranes (endoplasmatic reticulum, endosome and lysosome) | NADPHoxidases | O2·− | Cellular signaling, host defense, immune cell activation, inflammation, oxidative burst | Inflammation, CRC, ileitis, and IBD | Induced by bacterial antigens [34,35,36] |
Lysosome | Myeloperoxidase | H2O2 | Neutrophile activation pathogen defense | IBD | Induced by bacterial antigens [37,38,39,40] |
Peroxisomes | Flavoproteins include acyl-CoA oxidases, urate oxidase, D-amino acid oxidase, D-aspartate oxidase, L-pipecolic acid oxidase, L-α-hydroxyacid oxidase, and polyamine oxidase | H2O2 | Combat/oxidize bacterial pathogens | IBD and CRC | Induced by bacterial antigens and metabolites [37,41,42,43] |
Peroxisomes | Xanthine oxidase | O2·−/H2O2 | Combat/oxidize bacterial pathogens, inflammation | Inflammation, type II diabetes | Induced by bacterial antigens and metabolites, purine degradation [37,41,42,43,44,45] |
Cytosol | Xanthine oxidase | O2·−/H2O2 | Inflammation | Inflammation, type II diabetes, gout | Purine degradation [37,44,45] |
2.3. Oxidative Stress
3. REDOX Regulation of the Immune System
3.1. Activation-Induced Oxidative Signaling in Immune Cells
3.1.1. T-Cell Activation-Induced Oxidative Signaling
3.1.2. B-Cell Activation-Induced Oxidative Signaling
3.1.3. Macrophages and Oxidative Signaling
3.1.4. Dendritic Cells and Oxidative Signaling
3.2. ROS, the Oxidative Burst, and the Inflammasome
4. Gut Microbiome and the REDOX Status
4.1. Gut Bacteria and Chronic Inflammation
4.2. Direct Effects of the Microbiome on the REDOX Status in the Gut
4.3. Indirect Effects of the Microbiome on the Redox Status in the Gut
4.4. Therapeutic Interventions Reconstituting a Physiologic REDOX State in the Gut
5. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Reth, M. Hydrogen peroxide as second messenger in lymphocyte activation. Nat. Immunol. 2002, 3, 1129–1134. [Google Scholar] [CrossRef] [PubMed]
- Schroeder, A.; Warnken, U.; Röth, D.; Klika, K.D.; Vobis, D.; Barnert, A.; Bujupi, F.; Oberacker, T.; Schnölzer, M.; Nicolay, J.P.; et al. Targeting Thioredoxin-1 by dimethyl fumarate induces ripoptosome-mediated cell death. Sci. Rep. 2017, 7, 43168. [Google Scholar] [CrossRef]
- Dröge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002, 82, 47–95. [Google Scholar] [CrossRef] [PubMed]
- Flohé, L. Looking Back at the Early Stages of Redox Biology. Antioxidants 2020, 9, 1254. [Google Scholar] [CrossRef] [PubMed]
- Zuo, J.; Zhang, Z.; Luo, M.; Zhou, L.; Nice, E.C.; Zhang, W.; Wang, C.; Huang, C. Redox signaling at the crossroads of human health and disease. MedComm 2022, 3, e127. [Google Scholar] [CrossRef]
- Andrés, C.M.C.; La Pérez de Lastra, J.M.; Andrés Juan, C.; Plou, F.J.; Pérez-Lebeña, E. Superoxide Anion Chemistry-Its Role at the Core of the Innate Immunity. Int. J. Mol. Sci. 2023, 24, 1841. [Google Scholar] [CrossRef] [PubMed]
- Kamiński, M.M.; Röth, D.; Krammer, P.H.; Gülow, K. Mitochondria as oxidative signaling organelles in T-cell activa-tion: Physiological role and pathological implications. Arch. Immunol. Ther. Exp. 2013, 61, 367–384. [Google Scholar] [CrossRef] [PubMed]
- De Vos, W.M.; Tilg, H.; van Hul, M.; Cani, P.D. Gut microbiome and health: Mechanistic insights. Gut 2022, 71, 1020–1032. [Google Scholar] [CrossRef]
- McCord, J.M.; Fridovich, I. Superoxide dismutase. An enzymic function for erythrocuprein (hemocuprein). J. Biol. Chem. 1969, 244, 6049–6055. [Google Scholar] [CrossRef]
- Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S.; Kalayci, O. Oxidative stress and antioxidant defense. World Allergy Organ. J. 2012, 5, 9–19. [Google Scholar] [CrossRef]
- Brand, M.D. Riding the tiger—Physiological and pathological effects of superoxide and hydrogen peroxide generated in the mitochondrial matrix. Crit. Rev. Biochem. Mol. Biol. 2020, 55, 592–661. [Google Scholar] [CrossRef] [PubMed]
- Sies, H.; Jones, D.P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 2020, 21, 363–383. [Google Scholar] [CrossRef] [PubMed]
- Vona, R.; Pallotta, L.; Cappelletti, M.; Severi, C.; Matarrese, P. The Impact of Oxidative Stress in Human Pathology: Focus on Gastrointestinal Disorders. Antioxidants 2021, 10, 201. [Google Scholar] [CrossRef] [PubMed]
- Jomova, K.; Makova, M.; Alomar, S.Y.; Alwasel, S.H.; Nepovimova, E.; Kuca, K.; Rhodes, C.J.; Valko, M. Essential metals in health and disease. Chem. Biol. Interact. 2022, 367, 110173. [Google Scholar] [CrossRef] [PubMed]
- Torti, F.M.; Torti, S.V. Regulation of ferritin genes and protein. Blood 2002, 99, 3505–3516. [Google Scholar] [CrossRef]
- Kiessling, M.K.; Klemke, C.D.; Kaminski, M.M.; Galani, I.E.; Krammer, P.H.; Gülow, K. Inhibition of constitutively activated nuclear factor-kappaB induces reactive oxygen species- and iron-dependent cell death in cutaneous T-cell lymphoma. Cancer Res. 2009, 69, 2365–2374. [Google Scholar] [CrossRef]
- Davies, M.J. The oxidative environment and protein damage. Biochim. Biophys. Acta 2005, 1703, 93–109. [Google Scholar] [CrossRef]
- Cinelli, M.A.; Do, H.T.; Miley, G.P.; Silverman, R.B. Inducible nitric oxide synthase: Regulation, structure, and inhibition. Med. Res. Rev. 2020, 40, 158–189. [Google Scholar] [CrossRef]
- Palmer, R.M.; Rees, D.D.; Ashton, D.S.; Moncada, S. L-arginine is the physiological precursor for the formation of nitric oxide in endothelium-dependent relaxation. Biochem. Biophys. Res. Commun. 1988, 153, 1251–1256. [Google Scholar] [CrossRef]
- Radi, R. Oxygen radicals, nitric oxide, and peroxynitrite: Redox pathways in molecular medicine. Proc. Natl. Acad. Sci. USA 2018, 115, 5839–5848. [Google Scholar] [CrossRef]
- Boveris, A.; Cadenas, E.; Stoppani, A.O. Role of ubiquinone in the mitochondrial generation of hydrogen peroxide. Biochem. J. 1976, 156, 435–444. [Google Scholar] [CrossRef] [PubMed]
- Boveris, A.; Chance, B. The mitochondrial generation of hydrogen peroxide. General properties and effect of hyperbaric oxygen. Biochem. J. 1973, 134, 707–716. [Google Scholar] [CrossRef] [PubMed]
- Loschen, G.; Azzi, A.; Richter, C.; Flohé, L. Superoxide radicals as precursors of mitochondrial hydrogen peroxide. FEBS Lett. 1974, 42, 68–72. [Google Scholar] [CrossRef] [PubMed]
- Kamiński, M.M.; Sauer, S.W.; Kamiński, M.; Opp, S.; Ruppert, T.; Grigaravičius, P.; Grudnik, P.; Gröne, H.-J.; Krammer, P.H.; Gülow, K. T cell activation is driven by an ADP-dependent glucokinase linking enhanced glycolysis with mitochondrial reactive oxygen species generation. Cell Rep. 2012, 2, 1300–1315. [Google Scholar] [CrossRef] [PubMed]
- Kaminski, M.M.; Sauer, S.W.; Klemke, C.-D.; Süss, D.; Okun, J.G.; Krammer, P.H.; Gülow, K. Mitochondrial reactive oxygen species control T cell activation by regulating IL-2 and IL-4 expression: Mechanism of ciprofloxa-cin-mediated immunosuppression. J. Immunol. 2010, 184, 4827–4841. [Google Scholar] [CrossRef]
- Kaminski, M.; Kiessling, M.; Süss, D.; Krammer, P.H.; Gülow, K. Novel role for mitochondria: Protein kinase Cθ-dependent oxidative signaling organelles in activation-induced T-cell death. Mol. Cell. Biol. 2007, 27, 3625–3639. [Google Scholar] [CrossRef]
- Weinberg, S.E.; Singer, B.D.; Steinert, E.M.; Martinez, C.A.; Mehta, M.M.; Martínez-Reyes, I.; Gao, P.; Helmin, K.A.; Abdala-Valencia, H.; Sena, L.A.; et al. Mitochondrial complex III is essential for suppressive function of regulatory T cells. Nature 2019, 565, 495–499. [Google Scholar] [CrossRef]
- Sena, L.A.; Li, S.; Jairaman, A.; Prakriya, M.; Ezponda, T.; Hildeman, D.A.; Wang, C.-R.; Schumacker, P.T.; Licht, J.D.; Perlman, H.; et al. Mitochondria are required for antigen-specific T cell activation through reactive oxygen species signaling. Immunity 2013, 38, 225–236. [Google Scholar] [CrossRef]
- Pecchillo Cimmino, T.; Ammendola, R.; Cattaneo, F.; Esposito, G. NOX Dependent ROS Generation and Cell Metabolism. Int. J. Mol. Sci. 2023, 24, 2086. [Google Scholar] [CrossRef]
- Di Marzo, N.; Chisci, E.; Giovannoni, R. The Role of Hydrogen Peroxide in Redox-Dependent Signaling: Homeo-static and Pathological Responses in Mammalian Cells. Cells 2018, 7, 156. [Google Scholar] [CrossRef]
- Neish, A.S. Redox signaling mediated by the gut microbiota. Free Radic. Res. 2013, 47, 950–957. [Google Scholar] [CrossRef] [PubMed]
- Bedard, K.; Krause, K.-H. The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiol. Rev. 2007, 87, 245–313. [Google Scholar] [CrossRef] [PubMed]
- Jackson, D.N.; Theiss, A.L. Gut bacteria signaling to mitochondria in intestinal inflammation and cancer. Gut Microbes 2020, 11, 285–304. [Google Scholar] [CrossRef]
- Aviello, G.; Knaus, U.G. NADPH oxidases and ROS signaling in the gastrointestinal tract. Mucosal Immunol. 2018, 11, 1011–1023. [Google Scholar] [CrossRef]
- Vermot, A.; Petit-Härtlein, I.; Smith, S.M.E.; Fieschi, F. NADPH Oxidases (NOX): An Overview from Discovery, Molecular Mechanisms to Physiology and Pathology. Antioxidants 2021, 10, 890. [Google Scholar] [CrossRef] [PubMed]
- Jackson, S.H.; Devadas, S.; Kwon, J.; Pinto, L.A.; Williams, M.S. T cells express a phagocyte-type NADPH oxidase that is activated after T cell receptor stimulation. Nat. Immunol. 2004, 5, 818–827. [Google Scholar] [CrossRef] [PubMed]
- Singh, V.; Ahlawat, S.; Mohan, H.; Gill, S.S.; Sharma, K.K. Balancing reactive oxygen species generation by rebooting gut microbiota. J. Appl. Microbiol. 2022, 132, 4112–4129. [Google Scholar] [CrossRef]
- Aratani, Y. Myeloperoxidase: Its role for host defense, inflammation, and neutrophil function. Arch. Biochem. Biophys. 2018, 640, 47–52. [Google Scholar] [CrossRef]
- Chami, B.; Martin, N.J.J.; Dennis, J.M.; Witting, P.K. Myeloperoxidase in the inflamed colon: A novel target for treating inflammatory bowel disease. Arch. Biochem. Biophys. 2018, 645, 61–71. [Google Scholar] [CrossRef]
- Winterbourn, C.C.; Kettle, A.J.; Hampton, M.B. Reactive Oxygen Species and Neutrophil Function. Annu. Rev. Biochem. 2016, 85, 765–792. [Google Scholar] [CrossRef]
- Sandalio, L.M.; Rodríguez-Serrano, M.; Romero-Puertas, M.C.; Del Río, L.A. Role of peroxisomes as a source of reactive oxygen species (ROS) signaling molecules. Subcell. Biochem. 2013, 69, 231–255. [Google Scholar] [CrossRef] [PubMed]
- Fransen, M.; Nordgren, M.; Wang, B.; Apanasets, O. Role of peroxisomes in ROS/RNS-metabolism: Implications for human disease. Biochim. Biophys. Acta 2012, 1822, 1363–1373. [Google Scholar] [CrossRef] [PubMed]
- Di Cara, F.; Bülow, M.H.; Simmonds, A.J.; Rachubinski, R.A. Dysfunctional peroxisomes compromise gut structure and host defense by increased cell death and Tor-dependent autophagy. Mol. Biol. Cell 2018, 29, 2766–2783. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Li, Y.; Liao, W.; Huang, J.; Liu, Y.; Li, Z.; Tang, J. Gut microbiota remodeling: A promising therapeutic strategy to confront hyperuricemia and gout. Front. Cell. Infect. Microbiol. 2022, 12, 935723. [Google Scholar] [CrossRef]
- Yu, Y.; Liu, Q.; Li, H.; Wen, C.; He, Z. Alterations of the Gut Microbiome Associated with the Treatment of Hyperuricaemia in Male Rats. Front. Microbiol. 2018, 9, 2233. [Google Scholar] [CrossRef] [PubMed]
- Sies, H. (Ed.) Oxidative Stress: Introductory Remarks. In Oxidative Stress; Academic Press: London, UK, 1985; pp. 1–8. ISBN 9780126427608. [Google Scholar]
- Oberacker, T.; Bajorat, J.; Ziola, S.; Schroeder, A.; Röth, D.; Kastl, L.; Edgar, B.A.; Wagner, W.; Gülow, K.; Krammer, P.H. Enhanced expression of thioredoxin-interacting-protein regulates oxidative DNA damage and aging. FEBS Lett. 2018, 592, 2297–2307. [Google Scholar] [CrossRef]
- Rahman, K. Studies on free radicals, antioxidants, and co-factors. Clin. Interv. Aging. 2007, 2, 219–236. [Google Scholar]
- Schrader, M.; Fahimi, H.D. Peroxisomes and oxidative stress. Biochim. Biophys. Acta 2006, 1763, 1755–1766. [Google Scholar] [CrossRef]
- Gergely, P.; Niland, B.; Gonchoroff, N.; Pullmann, R.; Phillips, P.E.; Perl, A. Persistent mitochondrial hyperpolarization, increased reactive oxygen intermediate production, and cytoplasmic alkalinization characterize altered IL-10 signaling in patients with systemic lupus erythematosus. J. Immunol. 2002, 169, 1092–1101. [Google Scholar] [CrossRef]
- Nagy, G.; Koncz, A.; Perl, A. T cell activation-induced mitochondrial hyperpolarization is mediated by Ca2+- and redox-dependent production of nitric oxide. J. Immunol. 2003, 171, 5188–5197. [Google Scholar] [CrossRef]
- Williams, M.S.; Noguchi, S.; Henkart, P.A.; Osawa, Y. Nitric oxide synthase plays a signaling role in TCR-triggered apoptotic death. J. Immunol. 1998, 161, 6526–6531. [Google Scholar] [CrossRef] [PubMed]
- Sena, L.A.; Chandel, N.S. Physiological roles of mitochondrial reactive oxygen species. Mol. Cell 2012, 48, 158–167. [Google Scholar] [CrossRef] [PubMed]
- Röth, D.; Krammer, P.H.; Gülow, K. Dynamin related protein 1-dependent mitochondrial fission regulates oxidative signalling in T cells. FEBS Lett. 2014, 588, 1749–1754. [Google Scholar] [CrossRef]
- Kamiński, M.M.; Röth, D.; Sass, S.; Sauer, S.W.; Krammer, P.H.; Gülow, K. Manganese superoxide dismutase: A regulator of T cell activation-induced oxidative signaling and cell death. Biochim. Biophys. Acta 2012, 1823, 1041–1052. [Google Scholar] [CrossRef] [PubMed]
- Devadas, S.; Zaritskaya, L.; Rhee, S.G.; Oberley, L.; Williams, M.S. Discrete generation of superoxide and hydrogen peroxide by T cell receptor stimulation: Selective regulation of mitogen-activated protein kinase activation and fas ligand expression. J. Exp. Med. 2002, 195, 59–70. [Google Scholar] [CrossRef] [PubMed]
- Ozay, E.I.; Sherman, H.L.; Mello, V.; Trombley, G.; Lerman, A.; Tew, G.N.; Yadava, N.; Minter, L.M. Rotenone Treatment Reveals a Role for Electron Transport Complex I in the Subcellular Localization of Key Transcriptional Regulators During T Helper Cell Differentiation. Front. Immunol. 2018, 9, 1284. [Google Scholar] [CrossRef]
- Yin, M.; O’Neill, L.A.J. The role of the electron transport chain in immunity. FASEB J. 2021, 35, e21974. [Google Scholar] [CrossRef]
- Wheeler, M.L.; DeFranco, A.L. Prolonged production of reactive oxygen species in response to B cell receptor stimulation promotes B cell activation and proliferation. J. Immunol. 2012, 189, 4405–4416. [Google Scholar] [CrossRef]
- Waters, L.R.; Ahsan, F.M.; Wolf, D.M.; Shirihai, O.; Teitell, M.A. Initial B Cell Activation Induces Metabolic Reprogramming and Mitochondrial Remodeling. iScience 2018, 5, 99–109. [Google Scholar] [CrossRef]
- Bassoy, E.Y.; Walch, M.; Martinvalet, D. Reactive Oxygen Species: Do They Play a Role in Adaptive Immunity? Front. Immunol. 2021, 12, 755856. [Google Scholar] [CrossRef]
- Jha, A.K.; Huang, S.C.-C.; Sergushichev, A.; Lampropoulou, V.; Ivanova, Y.; Loginicheva, E.; Chmielewski, K.; Stew-art, K.M.; Ashall, J.; Everts, B.; et al. Network integration of parallel metabolic and transcriptional data reveals metabolic modules that regulate macrophage polarization. Immunity 2015, 42, 419–430. [Google Scholar] [CrossRef]
- Feingold, K.R.; Shigenaga, J.K.; Kazemi, M.R.; McDonald, C.M.; Patzek, S.M.; Cross, A.S.; Moser, A.; Grunfeld, C. Mechanisms of triglyceride accumulation in activated macrophages. J. Leukoc. Biol. 2012, 92, 829–839. [Google Scholar] [CrossRef]
- Van Uden, P.; Kenneth, N.S.; Rocha, S. Regulation of hypoxia-inducible factor-1alpha by NF-kappaB. Biochem. J. 2008, 412, 477–484. [Google Scholar] [CrossRef]
- Freemerman, A.J.; Johnson, A.R.; Sacks, G.N.; Milner, J.J.; Kirk, E.L.; Troester, M.A.; Macintyre, A.N.; Go-raksha-Hicks, P.; Rathmell, J.C.; Makowski, L. Metabolic reprogramming of macrophages: Glucose transporter 1 (GLUT1)-mediated glucose metabolism drives a proinflammatory phenotype. J. Biol. Chem. 2014, 289, 7884–7896. [Google Scholar] [CrossRef]
- Wang, T.; Liu, H.; Lian, G.; Zhang, S.-Y.; Wang, X.; Jiang, C. HIF1α-Induced Glycolysis Metabolism Is Essential to the Activation of Inflammatory Macrophages. Mediat. Inflamm. 2017, 2017, 9029327. [Google Scholar] [CrossRef]
- Pavlou, S.; Wang, L.; Xu, H.; Chen, M. Higher phagocytic activity of thioglycollate-elicited peritoneal macrophages is related to metabolic status of the cells. J. Inflamm. 2017, 14, 4. [Google Scholar] [CrossRef]
- Van den Bossche, J.; O’Neill, L.A.; Menon, D. Macrophage Immunometabolism: Where Are We (Going)? Trends Immunol. 2017, 38, 395–406. [Google Scholar] [CrossRef]
- Boscá, L.; González-Ramos, S.; Prieto, P.; Fernández-Velasco, M.; Mojena, M.; Martín-Sanz, P.; Alemany, S. Metabolic signatures linked to macrophage polarization: From glucose metabolism to oxidative phosphorylation. Biochem. Soc. Trans. 2015, 43, 740–744. [Google Scholar] [CrossRef]
- Burova, E.; Borodkina, A.; Shatrova, A.; Nikolsky, N. Sublethal oxidative stress induces the premature senescence of human mesenchymal stem cells derived from endometrium. Oxid. Med. Cell. Longev. 2013, 2013, 474931. [Google Scholar] [CrossRef]
- Li, H.; Luo, Y.-F.; Wang, Y.-S.; Yang, Q.; Xiao, Y.-L.; Cai, H.-R.; Xie, C.-M. Using ROS as a Second Messenger, NADPH Oxidase 2 Mediates Macrophage Senescence via Interaction with NF-κB during Pseudomonas aeruginosa Infection. Oxid. Med. Cell. Longev. 2018, 2018, 9741838. [Google Scholar] [CrossRef]
- Elder, S.S.; Emmerson, E. Senescent cells and macrophages: Key players for regeneration? Open Biol. 2020, 10, 200309. [Google Scholar] [CrossRef]
- Brown, L.A.S.; Ping, X.-D.; Harris, F.L.; Gauthier, T.W. Glutathione availability modulates alveolar macrophage function in the chronic ethanol-fed rat. Am. J. Physiol. Lung Cell. Mol. Physiol. 2007, 292, L824–L832. [Google Scholar] [CrossRef]
- Fitzpatrick, A.M.; Teague, W.G.; Burwell, L.; Brown, M.S.; Brown, L.A.S. Glutathione oxidation is associated with airway macrophage functional impairment in children with severe asthma. Pediatr. Res. 2011, 69, 154–159. [Google Scholar] [CrossRef] [PubMed]
- Gallo, P.M.; Gallucci, S. The dendritic cell response to classic, emerging, and homeostatic danger signals. Implications for autoimmunity. Front. Immunol. 2013, 4, 138. [Google Scholar] [CrossRef]
- Thwe, P.M.; Pelgrom, L.R.; Cooper, R.; Beauchamp, S.; Reisz, J.A.; D’Alessandro, A.; Everts, B.; Amiel, E. Cell-Intrinsic Glycogen Metabolism Supports Early Glycolytic Reprogramming Required for Dendritic Cell Immune Responses. Cell Metab. 2017, 26, 558–567.e5. [Google Scholar] [CrossRef]
- Krawczyk, C.M.; Holowka, T.; Sun, J.; Blagih, J.; Amiel, E.; DeBerardinis, R.J.; Cross, J.R.; Jung, E.; Thompson, C.B.; Jones, R.G.; et al. Toll-like receptor-induced changes in glycolytic metabolism regulate dendritic cell activation. Blood 2010, 115, 4742–4749. [Google Scholar] [CrossRef]
- Sukhbaatar, N.; Hengstschläger, M.; Weichhart, T. mTOR-Mediated Regulation of Dendritic Cell Differentiation and Function. Trends Immunol. 2016, 37, 778–789. [Google Scholar] [CrossRef]
- Everts, B.; Amiel, E.; van der Windt, G.J.W.; Freitas, T.C.; Chott, R.; Yarasheski, K.E.; Pearce, E.L.; Pearce, E.J. Commitment to glycolysis sustains survival of NO-producing inflammatory dendritic cells. Blood 2012, 120, 1422–1431. [Google Scholar] [CrossRef]
- Amiel, E.; Everts, B.; Fritz, D.; Beauchamp, S.; Ge, B.; Pearce, E.L.; Pearce, E.J. Mechanistic target of rapamycin inhibition extends cellular lifespan in dendritic cells by preserving mitochondrial function. J. Immunol. 2014, 193, 2821–2830. [Google Scholar] [CrossRef]
- Morris, G.; Gevezova, M.; Sarafian, V.; Maes, M. Redox regulation of the immune response. Cell. Mol. Immunol. 2022, 19, 1079–1101. [Google Scholar] [CrossRef]
- Mantegazza, A.R.; Savina, A.; Vermeulen, M.; Pérez, L.; Geffner, J.; Hermine, O.; Rosenzweig, S.D.; Faure, F.; Amigorena, S. NADPH oxidase controls phagosomal pH and antigen cross-presentation in human dendritic cells. Blood 2008, 112, 4712–4722. [Google Scholar] [CrossRef]
- Paardekooper, L.M.; Dingjan, I.; Linders, P.T.A.; Staal, A.H.J.; Cristescu, S.M.; Verberk, W.C.E.P.; van den Bogaart, G. Human Monocyte-Derived Dendritic Cells Produce Millimolar Concentrations of ROS in Phagosomes Per Second. Front. Immunol. 2019, 10, 1216. [Google Scholar] [CrossRef]
- Oberkampf, M.; Guillerey, C.; Mouriès, J.; Rosenbaum, P.; Fayolle, C.; Bobard, A.; Savina, A.; Ogier-Denis, E.; Enninga, J.; Amigorena, S.; et al. Mitochondrial reactive oxygen species regulate the induction of CD8+ T cells by plasmacytoid dendritic cells. Nat. Commun. 2018, 9, 2241. [Google Scholar] [CrossRef]
- Mittal, M.; Siddiqui, M.R.; Tran, K.; Reddy, S.P.; Malik, A.B. Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 2014, 20, 1126–1167. [Google Scholar] [CrossRef]
- Babior, B.M. NADPH oxidase: An update. Blood 1999, 93, 1464–1476. [Google Scholar] [CrossRef]
- Nunoi, H.; Nakamura, H.; Nishimura, T.; Matsukura, M. Recent topics and advanced therapies in chronic granulomatous disease. Hum. Cell 2023, 36, 515–527. [Google Scholar] [CrossRef]
- Rokutan, K.; Kawahara, T.; Kuwano, Y.; Tominaga, K.; Sekiyama, A.; Teshima-Kondo, S. NADPH oxidases in the gastrointestinal tract: A potential role of Nox1 in innate immune response and carcinogenesis. Antioxid. Redox Signal. 2006, 8, 1573–1582. [Google Scholar] [CrossRef]
- van der Post, S.; Birchenough, G.M.H.; Held, J.M. NOX1-dependent redox signaling potentiates colonic stem cell proliferation to adapt to the intestinal microbiota by linking EGFR and TLR activation. Cell Rep. 2021, 35, 108949. [Google Scholar] [CrossRef]
- Bulua, A.C.; Simon, A.; Maddipati, R.; Pelletier, M.; Park, H.; Kim, K.-Y.; Sack, M.N.; Kastner, D.L.; Siegel, R.M. Mitochondrial reactive oxygen species promote production of proinflammatory cytokines and are elevated in TNFR1-associated periodic syndrome (TRAPS). J. Exp. Med. 2011, 208, 519–533. [Google Scholar] [CrossRef]
- Zhou, R.; Yazdi, A.S.; Menu, P.; Tschopp, J. A role for mitochondria in NLRP3 inflammasome activation. Nature 2011, 469, 221–225. [Google Scholar] [CrossRef]
- Dostert, C.; Pétrilli, V.; van Bruggen, R.; Steele, C.; Mossman, B.T.; Tschopp, J. Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science 2008, 320, 674–677. [Google Scholar] [CrossRef]
- Schroder, K.; Tschopp, J. The inflammasomes. Cell 2010, 140, 821–832. [Google Scholar] [CrossRef]
- Zhou, R.; Tardivel, A.; Thorens, B.; Choi, I.; Tschopp, J. Thioredoxin-interacting protein links oxidative stress to inflammasome activation. Nat. Immunol. 2010, 11, 136–140. [Google Scholar] [CrossRef]
- Berg, G.; Rybakova, D.; Fischer, D.; Cernava, T.; Vergès, M.-C.C.; Charles, T.; Chen, X.; Cocolin, L.; Eversole, K.; Corral, G.H.; et al. Microbiome definition re-visited: Old concepts and new challenges. Microbiome 2020, 8, 103. [Google Scholar] [CrossRef]
- Bermon, S.; Petriz, B.; Kajėnienė, A.; Prestes, J.; Castell, L.; Franco, O.L. The microbiota: An exercise immunology perspective. Exerc. Immunol. Rev. 2015, 21, 70–79. [Google Scholar] [PubMed]
- Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef]
- Cho, I.; Blaser, M.J. The human microbiome: At the interface of health and disease. Nature reviews. Genetics 2012, 13, 260–270. [Google Scholar] [CrossRef]
- Yilmaz, B.; Juillerat, P.; Øyås, O.; Ramon, C.; Bravo, F.D.; Franc, Y.; Fournier, N.; Michetti, P.; Mueller, C.; Geuking, M.; et al. Microbial network disturbances in relapsing refractory Crohn’s disease. Nat. Med. 2019, 25, 323–336. [Google Scholar] [CrossRef]
- 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]
- Cohen, L.J.; Cho, J.H.; Gevers, D.; Chu, H. Genetic Factors and the Intestinal Microbiome Guide Development of Microbe-Based Therapies for Inflammatory Bowel Diseases. Gastroenterology 2019, 156, 2174–2189. [Google Scholar] [CrossRef] [PubMed]
- Caparrós, E.; Wiest, R.; Scharl, M.; Rogler, G.; Gutiérrez Casbas, A.; Yilmaz, B.; Wawrzyniak, M.; Francés, R. Dysbi-otic microbiota interactions in Crohn’s disease. Gut Microbes 2021, 13, 1949096. [Google Scholar] [CrossRef]
- Morais, L.H.; Schreiber, H.L.; Mazmanian, S.K. The gut microbiota-brain axis in behaviour and brain disorders. Nature reviews. Microbiology 2021, 19, 241–255. [Google Scholar] [CrossRef]
- Wang, Y.; Zhang, Z.; Li, B.; He, B.; Li, L.; Nice, E.C.; Zhang, W.; Xu, J. New Insights into the Gut Microbiota in Neurodegenerative Diseases from the Perspective of Redox Homeostasis. Antioxidants 2022, 11, 2287. [Google Scholar] [CrossRef]
- Shandilya, S.; Kumar, S.; Kumar Jha, N.; Kumar Kesari, K.; Ruokolainen, J. Interplay of gut microbiota and oxidative stress: Perspective on neurodegeneration and neuroprotection. J. Adv. Res. 2022, 38, 223–244. [Google Scholar] [CrossRef]
- Tranah, T.H.; Edwards, L.A.; Schnabl, B.; Shawcross, D.L. Targeting the gut-liver-immune axis to treat cirrhosis. Gut 2021, 70, 982–994. [Google Scholar] [CrossRef]
- Ni, Q.; Ye, Z.; Wang, Y.; Chen, J.; Zhang, W.; Ma, C.; Li, K.; Liu, Y.; Liu, L.; Han, Z.; et al. Gut Microbial Dysbiosis and Plasma Metabolic Profile in Individuals with Vitiligo. Front. Microbiol. 2020, 11, 592248. [Google Scholar] [CrossRef]
- Ni, Q.; Zhang, P.; Li, Q.; Han, Z. Oxidative Stress and Gut Microbiome in Inflammatory Skin Diseases. Front. Cell Dev. Biol. 2022, 10, 849985. [Google Scholar] [CrossRef]
- Haderer, M.; Neubert, P.; Rinner, E.; Scholtis, A.; Broncy, L.; Gschwendtner, H.; Kandulski, A.; Pavel, V.; Mehrl, A.; Brochhausen, C.; et al. Novel pathomechanism for spontaneous bacterial peritonitis: Disruption of cell junctions by cellular and bacterial proteases. Gut 2022, 71, 580–592. [Google Scholar] [CrossRef] [PubMed]
- Albillos, A.; de Gottardi, A.; Rescigno, M. The gut-liver axis in liver disease: Pathophysiological basis for therapy. J. Hepatol. 2020, 72, 558–577. [Google Scholar] [CrossRef]
- Bao, M.; Wang, K.; Li, J.; Li, Y.; Zhu, H.; Lu, M.; Zhang, Y.; Fan, Q.; Han, L.; Wang, K.; et al. ROS Scavenging and inflammation-directed polydopamine nanoparticles regulate gut immunity and flora therapy in inflammatory bowel disease. Acta Biomater. 2023, 161, 250–264. [Google Scholar] [CrossRef]
- O’Neill, S.; Brault, J.; Stasia, M.-J.; Knaus, U.G. Genetic disorders coupled to ROS deficiency. Redox Biol. 2015, 6, 135–156. [Google Scholar] [CrossRef]
- Thiagarajah, J.R.; Chang, J.; Goettel, J.A.; Verkman, A.S.; Lencer, W.I. Aquaporin-3 mediates hydrogen peroxide-dependent responses to environmental stress in colonic epithelia. Proc. Natl. Acad. Sci. USA 2017, 114, 568–573. [Google Scholar] [CrossRef]
- Rogler, G. Resolution of inflammation in inflammatory bowel disease. Lancet Gastroenterol. Hepatol. 2017, 2, 521–530. [Google Scholar] [CrossRef]
- Banan, A.; Choudhary, S.; Zhang, Y.; Fields, J.Z.; Keshavarzian, A. Ethanol-induced barrier dysfunction and its pre-vention by growth factors in human intestinal monolayers: Evidence for oxidative and cytoskeletal mechanisms. J. Pharmacol. Exp. Ther. 1999, 291, 1075–1085. [Google Scholar]
- Rao, R.; Baker, R.D.; Baker, S.S. Inhibition of oxidant-induced barrier disruption and protein tyrosine phosphoryla-tion in Caco-2 cell monolayers by epidermal growth factor. Biochem. Pharmacol. 1999, 57, 685–695. [Google Scholar] [CrossRef]
- Pacher, P.; Beckman, J.S.; Liaudet, L. Nitric oxide and peroxynitrite in health and disease. Physiol. Rev. 2007, 87, 315–424. [Google Scholar] [CrossRef]
- Babbs, C.F. Oxygen radicals in ulcerative colitis. Free. Radic. Biol. Med. 1992, 13, 169–181. [Google Scholar] [CrossRef]
- McBee, M.E.; Chionh, Y.H.; Sharaf, M.L.; Ho, P.; Cai, M.W.L.; Dedon, P.C. Production of Superoxide in Bacteria Is Stress- and Cell State-Dependent: A Gating-Optimized Flow Cytometry Method that Minimizes ROS Measurement Artifacts with Fluorescent Dyes. Front. Microbiol. 2017, 8, 459. [Google Scholar] [CrossRef]
- Imlay, J.A. Cellular defenses against superoxide and hydrogen peroxide. Annu. Rev. Biochem. 2008, 77, 755–776. [Google Scholar] [CrossRef]
- Voltan, S.; Martines, D.; Elli, M.; Brun, P.; Longo, S.; Porzionato, A.; Macchi, V.; D’Incà, R.; Scarpa, M.; Palù, G.; et al. Lactobacillus crispatus M247-derived H2O2 acts as a signal transducing molecule activating peroxisome prolif-erator activated receptor-gamma in the intestinal mucosa. Gastroenterology 2008, 135, 1216–1227. [Google Scholar] [CrossRef]
- Ito, A.; Sato, Y.; Kudo, S.; Sato, S.; Nakajima, H.; Toba, T. The screening of hydrogen peroxide-producing lactic acid bacteria and their application to inactivating psychrotrophic food-borne pathogens. Curr. Microbiol. 2003, 47, 231–236. [Google Scholar] [CrossRef]
- Vetrano, S.; Rescigno, M.; Cera, M.R.; Correale, C.; Rumio, C.; Doni, A.; Fantini, M.; Sturm, A.; Borroni, E.; Repici, A.; et al. Unique role of junctional adhesion molecule-a in maintaining mucosal homeostasis in inflammatory bowel disease. Gastroenterology 2008, 135, 173–184. [Google Scholar] [CrossRef] [PubMed]
- Zeissig, S.; Bürgel, N.; Günzel, D.; Richter, J.; Mankertz, J.; Wahnschaffe, U.; Kroesen, A.J.; Zeitz, M.; Fromm, M.; Schulzke, J.-D. Changes in expression and distribution of claudin 2, 5 and 8 lead to discontinuous tight junctions and barrier dysfunction in active Crohn’s disease. Gut 2007, 56, 61–72. [Google Scholar] [CrossRef] [PubMed]
- Fantini, M.C.; Guadagni, I. From inflammation to colitis-associated colorectal cancer in inflammatory bowel disease: Pathogenesis and impact of current therapies. Dig. Liver Dis. 2021, 53, 558–565. [Google Scholar] [CrossRef]
- Rogler, G. Chronic ulcerative colitis and colorectal cancer. Cancer Lett. 2014, 345, 235–241. [Google Scholar] [CrossRef] [PubMed]
- Bardelčíková, A.; Šoltys, J.; Mojžiš, J. Oxidative Stress, Inflammation and Colorectal Cancer: An Overview. Antioxidants 2023, 12, 901. [Google Scholar] [CrossRef] [PubMed]
- Kelder, T.; Stroeve, J.H.M.; Bijlsma, S.; Radonjic, M.; Roeselers, G. Correlation network analysis reveals relationships between diet-induced changes in human gut microbiota and metabolic health. Nutr. Diabetes 2014, 4, e122. [Google Scholar] [CrossRef]
- Rajilić-Stojanović, M.; Jonkers, D.M.; Salonen, A.; Hanevik, K.; Raes, J.; Jalanka, J.; de Vos, W.M.; Manichanh, C.; Golic, N.; Enck, P.; et al. Intestinal microbiota and diet in IBS: Causes, consequences, or epiphenomena? Am. J. Gastroenterol. 2015, 110, 278–287. [Google Scholar] [CrossRef]
- Chooruk, A.; Piwat, S.; Teanpaisan, R. Antioxidant activity of various oral Lactobacillus strains. J. Appl. Microbiol. 2017, 123, 271–279. [Google Scholar] [CrossRef]
- Zorov, D.B.; Plotnikov, E.Y.; Silachev, D.N.; Zorova, L.D.; Pevzner, I.B.; Zorov, S.D.; Babenko, V.A.; Jankauskas, S.S.; Popkov, V.A.; Savina, P.S. Microbiota and mitobiota. Putting an equal sign between mitochondria and bacteria. Biochemistry 2014, 79, 1017–1031. [Google Scholar] [CrossRef]
- Wang, Y.; Wu, Y.; Wang, Y.; Xu, H.; Mei, X.; Yu, D.; Wang, Y.; Li, W. Antioxidant Properties of Probiotic Bacteria. Nutrients 2017, 9, 521. [Google Scholar] [CrossRef] [PubMed]
- Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [PubMed]
- Guarner, F.; Khan, A.G.; Garisch, J.; Eliakim, R.; Gangl, A.; Thomson, A.; Krabshuis, J.; Lemair, T.; Kaufmann, P.; de Paula, J.A.; et al. World Gastroenterology Organisation Global Guidelines: Probiotics and prebiotics October 2011. J. Clin. Gastroenterol. 2012, 46, 468–481. [Google Scholar] [CrossRef] [PubMed]
- Wieërs, G.; Belkhir, L.; Enaud, R.; Leclercq, S.; Philippart de Foy, J.-M.; Dequenne, I.; de Timary, P.; Cani, P.D. How Probiotics Affect the Microbiota. Front. Cell. Infect. Microbiol. 2019, 9, 454. [Google Scholar] [CrossRef]
- Kucharzik, T.; Dignass, A.; Areya, R.; Bokemeyer, B.; Esters, P.; Herrlinger, K.; Kannengiesser, K.; Kienle, P.; Langhorst, J.; Lügering, A.; et al. Aktualisierte S3-Leitlinie Colitis ulcerosa (Version 6.1). Available online: https://www.dgvs.de/wp-content/uploads/2023/03/LL-CU_Leitlinie_final_20.03.223.pdf (accessed on 20 March 2023).
- Su, G.L.; Ko, C.W.; Bercik, P.; Falck-Ytter, Y.; Sultan, S.; Weizman, A.V.; Morgan, R.L. AGA Clinical Practice Guidelines on the Role of Probiotics in the Management of Gastrointestinal Disorders. Gastroenterology 2020, 159, 697–705. [Google Scholar] [CrossRef]
- Ragan, M.V.; Wala, S.J.; Goodman, S.D.; Bailey, M.T.; Besner, G.E. Next-Generation Probiotic Therapy to Protect the Intestines from Injury. Front. Cell. Infect. Microbiol. 2022, 12, 863949. [Google Scholar] [CrossRef]
- Clostridioides Difficile. Available online: https://www.rki.de/DE/Content/Infekt/EpidBull/Merkblaetter/Ratgeber_Clostridium.html (accessed on 1 May 2023).
- Layer, P.; Andresen, V.; Allescher, H.; Bischoff, S.C.; Claßen, M.; Elsenbruch, S.; Freitag, M.; Frieling, T.; Gebhard, M.; Goebel-Stengel, M.; et al. Update S3-Leitlinie Reizdarmsyndrom: Definition, Pathophysiologie, Diagnostik und Therapie. Gemeinsame Leitlinie der Deutschen Gesellschaft für Gastroenterologie, Verdauungs- und Stoffwechselkrankheiten (DGVS) und der Deutschen Gesellschaft für Neurogastroenterologie und Motilität (DGNM)—Juni 2021—AWMF-Registriernummer: 021/016. Z. Gastroenterol. 2021, 59, 1323–1415. [Google Scholar] [CrossRef]
- Feng, T.; Wang, J. Oxidative stress tolerance and antioxidant capacity of lactic acid bacteria as probiotic: A systematic review. Gut Microbes 2020, 12, 1801944. [Google Scholar] [CrossRef]
- Chiarugi, P.; Pani, G.; Giannoni, E.; Taddei, L.; Colavitti, R.; Raugei, G.; Symons, M.; Borrello, S.; Galeotti, T.; Ramponi, G. Reactive oxygen species as essential mediators of cell adhesion: The oxidative inhibition of a FAK tyrosine phosphatase is required for cell adhesion. J. Cell Biol. 2003, 161, 933–944. [Google Scholar] [CrossRef]
- Diaz, B.; Shani, G.; Pass, I.; Anderson, D.; Quintavalle, M.; Courtneidge, S.A. Tks5-dependent, nox-mediated generation of reactive oxygen species is necessary for invadopodia formation. Sci. Signal. 2009, 2, ra53. [Google Scholar] [CrossRef]
- Wentworth, C.C.; Jones, R.M.; Kwon, Y.M.; Nusrat, A.; Neish, A.S. Commensal-epithelial signaling mediated via formyl peptide receptors. Am. J. Pathol. 2010, 177, 2782–2790. [Google Scholar] [CrossRef] [PubMed]
- Swanson, P.A.; Kumar, A.; Samarin, S.; Vijay-Kumar, M.; Kundu, K.; Murthy, N.; Hansen, J.; Nusrat, A.; Neish, A.S. Enteric commensal bacteria potentiate epithelial restitution via reactive oxygen species-mediated inactivation of focal adhesion kinase phosphatases. Proc. Natl. Acad. Sci. USA 2011, 108, 8803–8808. [Google Scholar] [CrossRef] [PubMed]
- Migeotte, I.; Communi, D.; Parmentier, M. Formyl peptide receptors: A promiscuous subfamily of G pro-tein-coupled receptors controlling immune responses. Cytokine Growth Factor Rev. 2006, 17, 501–519. [Google Scholar] [CrossRef]
- Tiso, M.; Schechter, A.N. Nitrate reduction to nitrite, nitric oxide and ammonia by gut bacteria under physiological conditions. PLoS ONE 2015, 10, e0119712. [Google Scholar] [CrossRef]
- Silva, Y.P.; Bernardi, A.; Frozza, R.L. The Role of Short-Chain Fatty Acids from Gut Microbiota in Gut-Brain Communication. Front. Endocrinol. 2020, 11, 25. [Google Scholar] [CrossRef]
- Hoyles, L.; Snelling, T.; Umlai, U.-K.; Nicholson, J.K.; Carding, S.R.; Glen, R.C.; McArthur, S. Microbiome-host sys-tems interactions: Protective effects of propionate upon the blood-brain barrier. Microbiome 2018, 6, 55. [Google Scholar] [CrossRef]
- Peng, L.; Li, Z.-R.; Green, R.S.; Holzman, I.R.; Lin, J. Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers. J. Nutr. 2009, 139, 1619–1625. [Google Scholar] [CrossRef]
- Saleri, R.; Borghetti, P.; Ravanetti, F.; Cavalli, V.; Ferrari, L.; de Angelis, E.; Andrani, M.; Martelli, P. Effects of dif-ferent short-chain fatty acids (SCFA) on gene expression of proteins involved in barrier function in IPEC-J2. Porc. Health Manag. 2022, 8, 21. [Google Scholar] [CrossRef]
- Pérez-Reytor, D.; Puebla, C.; Karahanian, E.; García, K. Use of Short-Chain Fatty Acids for the Recovery of the In-testinal Epithelial Barrier Affected by Bacterial Toxins. Front. Physiol. 2021, 12, 650313. [Google Scholar] [CrossRef]
- González-Bosch, C.; Boorman, E.; Zunszain, P.A.; Mann, G.E. Short-chain fatty acids as modulators of redox signaling in health and disease. Redox Biol. 2021, 47, 102165. [Google Scholar] [CrossRef]
- Guo, W.; Liu, J.; Sun, J.; Gong, Q.; Ma, H.; Kan, X.; Cao, Y.; Wang, J.; Fu, S. Butyrate alleviates oxidative stress by regulating NRF2 nuclear accumulation and H3K9/14 acetylation via GPR109A in bovine mammary epithelial cells and mammary glands. Free Radic. Biol. Med. 2020, 152, 728–742. [Google Scholar] [CrossRef] [PubMed]
- Yamamoto, M.; Kensler, T.W.; Motohashi, H. The KEAP1-NRF2 System: A Thiol-Based Sensor-Effector Apparatus for Maintaining Redox Homeostasis. Physiol. Rev. 2018, 98, 1169–1203. [Google Scholar] [CrossRef]
- Azad, M.A.K.; Sarker, M.; Li, T.; Yin, J. Probiotic Species in the Modulation of Gut Microbiota: An Overview. Biomed Res. Int. 2018, 2018, 9478630. [Google Scholar] [CrossRef] [PubMed]
- Park, J.-S.; Choi, J.W.; Jhun, J.; Kwon, J.Y.; Lee, B.-I.; Yang, C.W.; Park, S.-H.; Cho, M.-L. Lactobacillus acidophilus Improves Intestinal Inflammation in an Acute Colitis Mouse Model by Regulation of Th17 and Treg Cell Balance and Fibrosis Development. J. Med. Food 2018, 21, 215–224. [Google Scholar] [CrossRef]
- Geuking, M.B.; Cahenzli, J.; Lawson, M.A.E.; Ng, D.C.K.; Slack, E.; Hapfelmeier, S.; McCoy, K.D.; Macpherson, A.J. Intestinal bacterial colonization induces mutualistic regulatory T cell responses. Immunity 2011, 34, 794–806. [Google Scholar] [CrossRef]
- Mazmanian, S.K.; Liu, C.H.; Tzianabos, A.O.; Kasper, D.L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005, 122, 107–118. [Google Scholar] [CrossRef] [PubMed]
- Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K.; et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232–236. [Google Scholar] [CrossRef]
- Lin, S.; Li, Y.; Zamyatnin, A.A.; Werner, J.; Bazhin, A.V. Reactive oxygen species and colorectal cancer. J. Cell. Physiol. 2018, 233, 5119–5132. [Google Scholar] [CrossRef]
- Pircalabioru, G.; Aviello, G.; Kubica, M.; Zhdanov, A.; Paclet, M.-H.; Brennan, L.; Hertzberger, R.; Papkovsky, D.; Bourke, B.; Knaus, U.G. Defensive Mutualism Rescues NADPH Oxidase Inactivation in Gut Infection. Cell Host Microbe 2016, 19, 651–663. [Google Scholar] [CrossRef]
- Eeckhaut, V.; Machiels, K.; Perrier, C.; Romero, C.; Maes, S.; Flahou, B.; Steppe, M.; Haesebrouck, F.; Sas, B.; Ducatelle, R.; et al. Butyricicoccus pullicaecorum in inflammatory bowel disease. Gut 2013, 62, 1745–1752. [Google Scholar] [CrossRef]
- Martín, R.; Miquel, S.; Benevides, L.; Bridonneau, C.; Robert, V.; Hudault, S.; Chain, F.; Berteau, O.; Azevedo, V.; Chatel, J.M.; et al. Functional Characterization of Novel Faecalibacterium prausnitzii Strains Isolated from Healthy Volunteers: A Step Forward in the Use of F. prausnitzii as a Next-Generation Probiotic. Front. Microbiol. 2017, 8, 1226. [Google Scholar] [CrossRef] [PubMed]
- Hu, W.; Gao, W.; Liu, Z.; Fang, Z.; Wang, H.; Zhao, J.; Zhang, H.; Lu, W.; Chen, W. Specific Strains of Faecalibacterium prausnitzii Ameliorate Nonalcoholic Fatty Liver Disease in Mice in Association with Gut Microbiota Regulation. Nutrients 2022, 14, 2945. [Google Scholar] [CrossRef] [PubMed]
- Mah, C.; Jayawardana, T.; Leong, G.; Koentgen, S.; Lemberg, D.; Connor, S.J.; Rokkas, T.; Grimm, M.C.; Leach, S.T.; Hold, G.L. Assessing the Relationship between the Gut Microbiota and Inflammatory Bowel Disease Therapeutics: A Systematic Review. Pathogens 2023, 12, 262. [Google Scholar] [CrossRef] [PubMed]
- Glassner, K.L.; Abraham, B.P.; Quigley, E.M.M. The microbiome and inflammatory bowel disease. J. Allergy Clin. Immunol. 2020, 145, 16–27. [Google Scholar] [CrossRef]
- Round, J.L.; Lee, S.M.; Li, J.; Tran, G.; Jabri, B.; Chatila, T.A.; Mazmanian, S.K. The Toll-like receptor 2 pathway establishes colonization by a commensal of the human microbiota. Science 2011, 332, 974–977. [Google Scholar] [CrossRef]
- Hrncir, T.; Hrncirova, L.; Kverka, M.; Hromadka, R.; Machova, V.; Trckova, E.; Kostovcikova, K.; Kralickova, P.; Krejsek, J.; Tlaskalova-Hogenova, H. Gut Microbiota and NAFLD: Pathogenetic Mechanisms, Microbiota Signa-tures, and Therapeutic Interventions. Microorganisms 2021, 9, 957. [Google Scholar] [CrossRef]
- Widhiati, S.; Purnomosari, D.; Wibawa, T.; Soebono, H. The role of gut microbiome in inflammatory skin disorders: A systematic review. Dermatol. Rep. 2022, 14, 9188. [Google Scholar] [CrossRef]
- Kim, J.; Lee, H.K. Potential Role of the Gut Microbiome in Colorectal Cancer Progression. Front. Immunol. 2021, 12, 807648. [Google Scholar] [CrossRef]
- Obermeier, F.; Hofmann, C.; Falk, W. Inflammatory bowel diseases: When natural friends turn into enemies-the importance of CpG motifs of bacterial DNA in intestinal homeostasis and chronic intestinal inflammation. Int. J. Inflam. 2010, 2010, 641910. [Google Scholar] [CrossRef]
- Hofmann, C.; Dunger, N.; Doser, K.; Lippert, E.; Siller, S.; Edinger, M.; Falk, W.; Obermeier, F. Physiologic TLR9-CpG-DNA interaction is essential for the homeostasis of the intestinal immune system. Inflamm. Bowel Dis. 2014, 20, 136–143. [Google Scholar] [CrossRef]
- Cheung, E.C.; Vousden, K.H. The role of ROS in tumour development and progression. Nat. Rev. Cancer 2022, 22, 280–297. [Google Scholar] [CrossRef] [PubMed]
- Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef] [PubMed]
- Bell, H.N.; Rebernick, R.J.; Goyert, J.; Singhal, R.; Kuljanin, M.; Kerk, S.A.; Huang, W.; Das, N.K.; Andren, A.; Solanki, S.; et al. Reuterin in the healthy gut microbiome suppresses colorectal cancer growth through altering redox balance. Cancer Cell 2022, 40, 185–200.e6. [Google Scholar] [CrossRef] [PubMed]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kunst, C.; Schmid, S.; Michalski, M.; Tümen, D.; Buttenschön, J.; Müller, M.; Gülow, K. The Influence of Gut Microbiota on Oxidative Stress and the Immune System. Biomedicines 2023, 11, 1388. https://doi.org/10.3390/biomedicines11051388
Kunst C, Schmid S, Michalski M, Tümen D, Buttenschön J, Müller M, Gülow K. The Influence of Gut Microbiota on Oxidative Stress and the Immune System. Biomedicines. 2023; 11(5):1388. https://doi.org/10.3390/biomedicines11051388
Chicago/Turabian StyleKunst, Claudia, Stephan Schmid, Marlen Michalski, Deniz Tümen, Jonas Buttenschön, Martina Müller, and Karsten Gülow. 2023. "The Influence of Gut Microbiota on Oxidative Stress and the Immune System" Biomedicines 11, no. 5: 1388. https://doi.org/10.3390/biomedicines11051388
APA StyleKunst, C., Schmid, S., Michalski, M., Tümen, D., Buttenschön, J., Müller, M., & Gülow, K. (2023). The Influence of Gut Microbiota on Oxidative Stress and the Immune System. Biomedicines, 11(5), 1388. https://doi.org/10.3390/biomedicines11051388