Critical Signaling Transduction Pathways and Intestinal Barrier: Implications for Pathophysiology and Therapeutics
Abstract
:1. Introduction
2. Components of the Intestinal Barrier
2.1. Intestinal Epithelium
2.2. Tight Junctions
2.3. Intestinal Microbiota
3. Pathological Mechanisms of Intestinal Barrier Disruption
4. Signal Transduction Pathway
4.1. PI3K/Akt/mTOR Signaling Pathway
4.2. MAPK Signaling Pathway
4.3. AMPK Signaling Pathway
4.4. JAK-STAT Signaling Pathway
4.5. NF-κB Signaling Pathway
5. Methods of Literature Review
6. Limitations and Challenges
6.1. The Nature of Animal Model
6.2. The Physiological Differences between the Intestines
6.3. Lack of Effective Drugs
7. Future Perspectives
8. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Conflicts of Interest
Abbreviations
References
- Wang, Y.; Song, W.; Wang, J.; Wang, T.; Xiong, X.; Qi, Z.; Fu, W.; Yang, X.; Chen, Y.G. Single-cell transcriptome analysis reveals differential nutrient absorption functions in human intestine. J. Exp. Med. 2020, 217, e20191130. [Google Scholar] [CrossRef] [PubMed]
- Chelakkot, C.; Ghim, J.; Ryu, S.H. Mechanisms regulating intestinal barrier integrity and its pathological implications. Exp. Mol. Med. 2018, 50, 1–9. [Google Scholar] [CrossRef] [PubMed]
- Zhang, X.; Wu, J.; Liu, Q.; Li, X.; Li, S.; Chen, J.; Hong, Z.; Wu, X.; Zhao, Y.; Ren, J. mtDNA-STING pathway promotes necroptosis-dependent enterocyte injury in intestinal ischemia reperfusion. Cell. Death Dis. 2020, 11, 1050. [Google Scholar] [CrossRef]
- Stevens, B.R.; Goel, R.; Seungbum, K.; Richards, E.M.; Holbert, R.C.; Pepine, C.J.; Raizada, M.K. Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression. Gut 2018, 67, 1555–1557. [Google Scholar] [CrossRef] [PubMed]
- Langer, V.; Vivi, E.; Regensburger, D.; Winkler, T.H.; Waldner, M.J.; Rath, T.; Schmid, B.; Skottke, L.; Lee, S.; Jeon, N.L.; et al. IFN-gamma drives inflammatory bowel disease pathogenesis through VE-cadherin-directed vascular barrier disruption. J. Clin. Investig. 2019, 129, 4691–4707. [Google Scholar] [CrossRef] [PubMed]
- Chu, H.; Khosravi, A.; Kusumawardhani, I.P.; Kwon, A.H.; Vasconcelos, A.C.; Cunha, L.D.; Mayer, A.E.; Shen, Y.; Wu, W.L.; Kambal, A.; et al. Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science 2016, 352, 1116–1120. [Google Scholar] [CrossRef] [PubMed]
- Sikalidis, A.K.; Maykish, A. The Gut Microbiome and Type 2 Diabetes Mellitus: Discussing a Complex Relationship. Biomedicines 2020, 8, 8. [Google Scholar] [CrossRef] [PubMed]
- Stephens, M.; von der Weid, P.Y. Lipopolysaccharides modulate intestinal epithelial permeability and inflammation in a species-specific manner. Gut Microbes 2020, 11, 421–432. [Google Scholar] [CrossRef]
- Hu, Q.; Ren, H.; Li, G.; Wang, D.; Zhou, Q.; Wu, J.; Zheng, J.; Huang, J.; Slade, D.A.; Wu, X.; et al. STING-mediated intestinal barrier dysfunction contributes to lethal sepsis. EBioMedicine 2019, 41, 497–508. [Google Scholar] [CrossRef]
- Kim, A.; Seong, K.M.; Choi, Y.Y.; Shim, S.; Park, S.; Lee, S.S. Inhibition of EphA2 by Dasatinib Suppresses Radiation-Induced Intestinal Injury. Int. J. Mol. Sci. 2020, 21, 9096. [Google Scholar] [CrossRef]
- Kaminsky, L.W.; Al-Sadi, R.; Ma, T.Y. IL-1beta and the Intestinal Epithelial Tight Junction Barrier. Front. Immunol. 2021, 12, 767456. [Google Scholar] [CrossRef] [PubMed]
- Li, M.; Oshima, T.; Ito, C.; Yamada, M.; Tomita, T.; Fukui, H.; Miwa, H. Glutamine Blocks Interleukin-13-Induced Intestinal Epithelial Barrier Dysfunction. Digestion 2021, 102, 170–179. [Google Scholar] [CrossRef] [PubMed]
- Cao, S.; Wu, H.; Wang, C.; Zhang, Q.; Jiao, L.; Lin, F.; Hu, C.H. Diquat-induced oxidative stress increases intestinal permeability, impairs mitochondrial function, and triggers mitophagy in piglets. J. Anim. Sci. 2018, 96, 1795–1805. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Wang, Q.; Wang, X.; Zhu, L.; Chen, J.; Zhang, B.; Chen, Y.; Yuan, Z. Gut microbial dysbiosis is associated with development and progression of radiation enteritis during pelvic radiotherapy. J. Cell. Mol. Med. 2019, 23, 3747–3756. [Google Scholar] [CrossRef] [PubMed]
- Akbarali, H.I.; Muchhala, K.H.; Jessup, D.K.; Cheatham, S. Chemotherapy induced gastrointestinal toxicities. Adv. Cancer Res. 2022, 155, 131–166. [Google Scholar] [CrossRef] [PubMed]
- Trefts, E.; Shaw, R.J. AMPK: Restoring metabolic homeostasis over space and time. Mol. Cell. 2021, 81, 3677–3690. [Google Scholar] [CrossRef] [PubMed]
- Chou, W.C.; Rampanelli, E.; Li, X.; Ting, J.P. Impact of intracellular innate immune receptors on immunometabolism. Cell. Mol. Immunol. 2022, 19, 337–351. [Google Scholar] [CrossRef]
- He, J.; Wang, M.; Yang, L.; Xin, H.; Bian, F.; Jiang, G.; Zhang, X. Astragaloside IV Alleviates Intestinal Barrier Dysfunction via the AKT-GSK3beta-beta-Catenin Pathway in Peritoneal Dialysis. Front. Pharmacol. 2022, 13, 873150. [Google Scholar] [CrossRef]
- Xu, H.; Wang, Y.; Jurutka, P.W.; Wu, S.; Chen, Y.; Cao, C.; Chen, G.; Tian, B.; Wang, S.; Cheng, S. 16alpha-Hydroxytrametenolic Acid from Poria cocos Improves Intestinal Barrier Function Through the Glucocorticoid Receptor-Mediated PI3K/Akt/NF-kappaB Pathway. J. Agric. Food Chem. 2019, 67, 10871–10879. [Google Scholar] [CrossRef]
- Gao, H.N.; Hu, H.; Wen, P.C.; Lian, S.; Xie, X.L.; Song, H.L.; Yang, Z.N.; Ren, F.Z. Yak milk-derived exosomes alleviate lipopolysaccharide-induced intestinal inflammation by inhibiting PI3K/AKT/C3 pathway activation. J. Dairy. Sci. 2021, 104, 8411–8424. [Google Scholar] [CrossRef]
- Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-kappaB signaling in inflammation. Signal. Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef] [PubMed]
- Peterson, L.W.; Artis, D. Intestinal epithelial cells: Regulators of barrier function and immune homeostasis. Nat. Rev. Immunol. 2014, 14, 141–153. [Google Scholar] [CrossRef] [PubMed]
- Liu, W.; He, K.; Wu, D.; Zhou, L.; Li, G.; Lin, Z.; Yang, X.; Liu, J.; Pui Man Hoi, M. Natural Dietary Compound Xanthohumol Regulates the Gut Microbiota and Its Metabolic Profile in a Mouse Model of Alzheimer’s Disease. Molecules 2022, 27, 1281. [Google Scholar] [CrossRef] [PubMed]
- Clevers, H. The intestinal crypt, a prototype stem cell compartment. Cell 2013, 154, 274–284. [Google Scholar] [CrossRef] [PubMed]
- Birchenough, G.M.H.; Johansson, M.E.V. Forming a mucus barrier along the colon. Science 2020, 370, 402–403. [Google Scholar] [CrossRef]
- Lichtenstein, G.R. Goblet cells make more than just mucus. Gastroenterology 2000, 118, 1272–1274. [Google Scholar] [CrossRef] [PubMed]
- Garabedian, E.M.; Roberts, L.J.; McNevin, M.S.; Gordon, J.I. Examining the role of Paneth cells in the small intestine by lineage ablation in transgenic mice. J. Biol. Chem. 1997, 272, 23729–23740. [Google Scholar] [CrossRef]
- Ayabe, T.; Satchell, D.P.; Wilson, C.L.; Parks, W.C.; Selsted, M.E.; Ouellette, A.J. Secretion of microbicidal alpha-defensins by intestinal Paneth cells in response to bacteria. Nat. Immunol. 2000, 1, 113–118. [Google Scholar] [CrossRef]
- Moran, G.W.; Leslie, F.C.; Levison, S.E.; Worthington, J.; McLaughlin, J.T. Enteroendocrine cells: Neglected players in gastrointestinal disorders? Ther. Adv. Gastroenterol. 2008, 1, 51–60. [Google Scholar] [CrossRef]
- Muller, T.D.; Finan, B.; Clemmensen, C.; DiMarchi, R.D.; Tschop, M.H. The New Biology and Pharmacology of Glucagon. Physiol. Rev. 2017, 97, 721–766. [Google Scholar] [CrossRef]
- Gribble, F.M.; Reimann, F. Enteroendocrine Cells: Chemosensors in the Intestinal Epithelium. Annu. Rev. Physiol. 2016, 78, 277–299. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Jang, M.H.; Kweon, M.N.; Iwatani, K.; Yamamoto, M.; Terahara, K.; Sasakawa, C.; Suzuki, T.; Nochi, T.; Yokota, Y.; Rennert, P.D.; et al. Intestinal villous M cells: An antigen entry site in the mucosal epithelium. Proc. Natl. Acad. Sci. USA 2004, 101, 6110–6115. [Google Scholar] [CrossRef] [PubMed]
- Allaire, J.M.; Crowley, S.M.; Law, H.T.; Chang, S.Y.; Ko, H.J.; Vallance, B.A. The Intestinal Epithelium: Central Coordinator of Mucosal Immunity. Trends Immunol. 2018, 39, 677–696. [Google Scholar] [CrossRef] [PubMed]
- Howitt, M.R.; Lavoie, S.; Michaud, M.; Blum, A.M.; Tran, S.V.; Weinstock, J.V.; Gallini, C.A.; Redding, K.; Margolskee, R.F.; Osborne, L.C.; et al. Tuft cells, taste-chemosensory cells, orchestrate parasite type 2 immunity in the gut. Science 2016, 351, 1329–1333. [Google Scholar] [CrossRef] [PubMed]
- Gerbe, F.; Sidot, E.; Smyth, D.J.; Ohmoto, M.; Matsumoto, I.; Dardalhon, V.; Cesses, P.; Garnier, L.; Pouzolles, M.; Brulin, B.; et al. Intestinal epithelial tuft cells initiate type 2 mucosal immunity to helminth parasites. Nature 2016, 529, 226–230. [Google Scholar] [CrossRef] [PubMed]
- Schneider, C.; O’Leary, C.E.; Locksley, R.M. Regulation of immune responses by tuft cells. Nat. Rev. Immunol. 2019, 19, 584–593. [Google Scholar] [CrossRef]
- Furuse, M.; Hirase, T.; Itoh, M.; Nagafuchi, A.; Yonemura, S.; Tsukita, S.; Tsukita, S. Occludin: A novel integral membrane protein localizing at tight junctions. J. Cell. Biol. 1993, 123 Pt 2, 1777–1788. [Google Scholar] [CrossRef]
- Furuse, M.; Fujita, K.; Hiiragi, T.; Fujimoto, K.; Tsukita, S. Claudin-1 and -2: Novel integral membrane proteins localizing at tight junctions with no sequence similarity to occludin. J. Cell. Biol. 1998, 141, 1539–1550. [Google Scholar] [CrossRef]
- Kuo, W.T.; Zuo, L.; Odenwald, M.A.; Madha, S.; Singh, G.; Gurniak, C.B.; Abraham, C.; Turner, J.R. The Tight Junction Protein ZO-1 Is Dispensable for Barrier Function but Critical for Effective Mucosal Repair. Gastroenterology 2021, 161, 1924–1939. [Google Scholar] [CrossRef]
- Otani, T.; Nguyen, T.P.; Tokuda, S.; Sugihara, K.; Sugawara, T.; Furuse, K.; Miura, T.; Ebnet, K.; Furuse, M. Claudins and JAM-A coordinately regulate tight junction formation and epithelial polarity. J. Cell. Biol. 2019, 218, 3372–3396. [Google Scholar] [CrossRef] [PubMed]
- Garcia, M.A.; Nelson, W.J.; Chavez, N. Cell-Cell Junctions Organize Structural and Signaling Networks. Cold Spring Harb. Perspect. Biol. 2018, 10, a029181. [Google Scholar] [CrossRef] [PubMed]
- Rosenthal, R.; Gunzel, D.; Krug, S.M.; Schulzke, J.D.; Fromm, M.; Yu, A.S. Claudin-2-mediated cation and water transport share a common pore. Acta Physiol. 2017, 219, 521–536. [Google Scholar] [CrossRef] [PubMed]
- Qiu, S.; Li, P.; Zhao, H.; Li, X. Maresin 1 alleviates dextran sulfate sodium-induced ulcerative colitis by regulating NRF2 and TLR4/NF-kB signaling pathway. Int. Immunopharmacol. 2020, 78, 106018. [Google Scholar] [CrossRef] [PubMed]
- Li, B.L.; Zhao, D.Y.; Du, P.L.; Wang, X.T.; Yang, Q.; Cai, Y.R. Luteolin alleviates ulcerative colitis through SHP-1/STAT3 pathway. Inflamm. Res. Off. J. Eur. Histamine Res. Soc. 2021, 70, 705–717. [Google Scholar] [CrossRef] [PubMed]
- Yang, S.; Hu, T.; Liu, H.; Lv, Y.L.; Zhang, W.; Li, H.; Xuan, L.; Gong, L.L.; Liu, L.H. Akebia saponin D ameliorates metabolic syndrome (MetS) via remodeling gut microbiota and attenuating intestinal barrier injury. Biomed. Pharmacother. Biomed. Pharmacother. 2021, 138, 111441. [Google Scholar] [CrossRef] [PubMed]
- Senchukova, M.A. Microbiota of the gastrointestinal tract: Friend or foe? World J. Gastroenterol. 2023, 29, 19–42. [Google Scholar] [CrossRef]
- Sender, R.; Fuchs, S.; Milo, R. Are We Really Vastly Outnumbered? Revisiting the Ratio of Bacterial to Host Cells in Humans. Cell 2016, 164, 337–340. [Google Scholar] [CrossRef]
- Martinez-Guryn, K.; Leone, V.; Chang, E.B. Regional Diversity of the Gastrointestinal Microbiome. Cell Host Microbe 2019, 26, 314–324. [Google Scholar] [CrossRef]
- Marchesi, J.R.; Adams, D.H.; Fava, F.; Hermes, G.D.; Hirschfield, G.M.; Hold, G.; Quraishi, M.N.; Kinross, J.; Smidt, H.; Tuohy, K.M.; et al. The gut microbiota and host health: A new clinical frontier. Gut 2016, 65, 330–339. [Google Scholar] [CrossRef]
- Moore, W.E.; Holdeman, L.V. Human fecal flora: The normal flora of 20 Japanese-Hawaiians. Appl. Microbiol. 1974, 27, 961–979. [Google Scholar] [CrossRef] [PubMed]
- Kumar, H.; Schutz, F.; Bhardwaj, K.; Sharma, R.; Nepovimova, E.; Dhanjal, D.S.; Verma, R.; Kumar, D.; Kuca, K.; Cruz-Martins, N. Recent advances in the concept of paraprobiotics: Nutraceutical/functional properties for promoting children health. Crit. Rev. Food Sci. Nutr. 2021, 63, 3943–3958. [Google Scholar] [CrossRef] [PubMed]
- Duncan, S.H.; Holtrop, G.; Lobley, G.E.; Calder, A.G.; Stewart, C.S.; Flint, H.J. Contribution of acetate to butyrate formation by human faecal bacteria. Br. J. Nutr. 2004, 91, 915–923. [Google Scholar] [CrossRef] [PubMed]
- Hernandez, M.A.G.; Canfora, E.E.; Jocken, J.W.E.; Blaak, E.E. The Short-Chain Fatty Acid Acetate in Body Weight Control and Insulin Sensitivity. Nutrients 2019, 11, 1943. [Google Scholar] [CrossRef]
- Cheng, S.; Ma, X.; Geng, S.; Jiang, X.; Li, Y.; Hu, L.; Li, J.; Wang, Y.; Han, X. Fecal Microbiota Transplantation Beneficially Regulates Intestinal Mucosal Autophagy and Alleviates Gut Barrier Injury. mSystems 2018, 3, e00137-18. [Google Scholar] [CrossRef] [PubMed]
- El-Salhy, M.; Hatlebakk, J.G.; Gilja, O.H.; Brathen Kristoffersen, A.; Hausken, T. Efficacy of faecal microbiota transplantation for patients with irritable bowel syndrome in a randomised, double-blind, placebo-controlled study. Gut 2020, 69, 859–867. [Google Scholar] [CrossRef] [PubMed]
- Maynard, C.L.; Elson, C.O.; Hatton, R.D.; Weaver, C.T. Reciprocal interactions of the intestinal microbiota and immune system. Nature 2012, 489, 231–241. [Google Scholar] [CrossRef]
- Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
- Stern, A.; Mick, E.; Tirosh, I.; Sagy, O.; Sorek, R. CRISPR targeting reveals a reservoir of common phages associated with the human gut microbiome. Genome Res. 2012, 22, 1985–1994. [Google Scholar] [CrossRef]
- Hsu, B.B.; Gibson, T.E.; Yeliseyev, V.; Liu, Q.; Lyon, L.; Bry, L.; Silver, P.A.; Gerber, G.K. Dynamic Modulation of the Gut Microbiota and Metabolome by Bacteriophages in a Mouse Model. Cell. Host Microbe 2019, 25, 803–814. [Google Scholar] [CrossRef]
- Shuwen, H.; Kefeng, D. Intestinal phages interact with bacteria and are involved in human diseases. Gut Microbes 2022, 14, 2113717. [Google Scholar] [CrossRef]
- Ge, T.; Yao, X.; Zhao, H.; Yang, W.; Zou, X.; Peng, F.; Li, B.; Cui, R. Gut microbiota and neuropsychiatric disorders: Implications for neuroendocrine-immune regulation. Pharmacol. Res. 2021, 173, 105909. [Google Scholar] [CrossRef] [PubMed]
- Aron-Wisnewsky, J.; Warmbrunn, M.V.; Nieuwdorp, M.; Clement, K. Metabolism and Metabolic Disorders and the Microbiome: The Intestinal Microbiota Associated With Obesity, Lipid Metabolism, and Metabolic Health-Pathophysiology and Therapeutic Strategies. Gastroenterology 2021, 160, 573–599. [Google Scholar] [CrossRef] [PubMed]
- Tang, W.H.W.; Backhed, F.; Landmesser, U.; Hazen, S.L. Intestinal Microbiota in Cardiovascular Health and Disease: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2019, 73, 2089–2105. [Google Scholar] [CrossRef] [PubMed]
- Medzhitov, R. Inflammation 2010: New adventures of an old flame. Cell 2010, 140, 771–776. [Google Scholar] [CrossRef] [PubMed]
- Holthaus, D.; Kraft, M.R.; Krug, S.M.; Wolf, S.; Muller, A.; Delgado Betancourt, E.; Schorr, M.; Holland, G.; Knauf, F.; Schulzke, J.D.; et al. Dissection of Barrier Dysfunction in Organoid-Derived Human Intestinal Epithelia Induced by Giardia duodenalis. Gastroenterology 2022, 162, 844–858. [Google Scholar] [CrossRef] [PubMed]
- Qu, W.; Zhang, L.; Ao, J. Radiotherapy Induces Intestinal Barrier Dysfunction by Inhibiting Autophagy. ACS Omega 2020, 5, 12955–12963. [Google Scholar] [CrossRef]
- Huang, Y.; Feng, Y.; Wang, Y.; Wang, P.; Wang, F.; Ren, H. Severe Burn-Induced Intestinal Epithelial Barrier Dysfunction Is Associated With Endoplasmic Reticulum Stress and Autophagy in Mice. Front. Physiol. 2018, 9, 441. [Google Scholar] [CrossRef]
- Huang, Y.; Zhou, J.; Wang, S.; Xiong, J.; Chen, Y.; Liu, Y.; Xiao, T.; Li, Y.; He, T.; Li, Y.; et al. Indoxyl sulfate induces intestinal barrier injury through IRF1-DRP1 axis-mediated mitophagy impairment. Theranostics 2020, 10, 7384–7400. [Google Scholar] [CrossRef]
- Bain, C.C.; Mowat, A.M. Macrophages in intestinal homeostasis and inflammation. Immunol. Rev. 2014, 260, 102–117. [Google Scholar] [CrossRef]
- Saez, A.; Herrero-Fernandez, B.; Gomez-Bris, R.; Sanchez-Martinez, H.; Gonzalez-Granado, J.M. Pathophysiology of Inflammatory Bowel Disease: Innate Immune System. Int. J. Mol. Sci. 2023, 24, 1526. [Google Scholar] [CrossRef] [PubMed]
- Dong, J.; Liang, W.; Wang, T.; Sui, J.; Wang, J.; Deng, Z.; Chen, D. Saponins regulate intestinal inflammation in colon cancer and IBD. Pharmacol. Res. 2019, 144, 66–72. [Google Scholar] [CrossRef] [PubMed]
- Haberman, Y.; Karns, R.; Dexheimer, P.J.; Schirmer, M.; Somekh, J.; Jurickova, I.; Braun, T.; Novak, E.; Bauman, L.; Collins, M.H.; et al. Ulcerative colitis mucosal transcriptomes reveal mitochondriopathy and personalized mechanisms underlying disease severity and treatment response. Nat. Commun. 2019, 10, 38. [Google Scholar] [CrossRef] [PubMed]
- Scherz-Shouval, R.; Elazar, Z. ROS, mitochondria and the regulation of autophagy. Trends Cell. Biol. 2007, 17, 422–427. [Google Scholar] [CrossRef]
- Zhou, X.; Wang, W.; Wang, C.; Zheng, C.; Xu, X.; Ni, X.; Hu, S.; Cai, B.; Sun, L.; Shi, K.; et al. DPP4 Inhibitor Attenuates Severe Acute Pancreatitis-Associated Intestinal Inflammation via Nrf2 Signaling. Oxid. Med. Cell. Longev. 2019, 2019, 6181754. [Google Scholar] [CrossRef] [PubMed]
- Van der Flier, L.G.; Clevers, H. Stem cells, self-renewal, and differentiation in the intestinal epithelium. Annu. Rev. Physiol. 2009, 71, 241–260. [Google Scholar] [CrossRef] [PubMed]
- Bhattacharyya, A.; Chattopadhyay, R.; Mitra, S.; Crowe, S.E. Oxidative stress: An essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol. Rev. 2014, 94, 329–354. [Google Scholar] [CrossRef] [PubMed]
- Hasegawa, T.; Mizugaki, A.; Inoue, Y.; Kato, H.; Murakami, H. Cystine reduces tight junction permeability and intestinal inflammation induced by oxidative stress in Caco-2 cells. Amino Acids 2021, 53, 1021–1032. [Google Scholar] [CrossRef]
- Diao, X.; Yamada, K.; Shibata, Y.; Imada, C. Metabolites Produced by a New Lactiplantibacillus plantarum Strain BF1-13 Isolated from Deep Seawater of Izu-Akazawa Protect the Intestinal Epithelial Barrier from the Dysfunction Induced by Hydrogen Peroxide. Mar. Drugs 2022, 20, 87. [Google Scholar] [CrossRef]
- Chen, Y.E.; Xu, S.J.; Lu, Y.Y.; Chen, S.X.; Du, X.H.; Hou, S.Z.; Huang, H.Y.; Liang, J. Asperuloside suppressing oxidative stress and inflammation in DSS-induced chronic colitis and RAW 264.7 macrophages via Nrf2/HO-1 and NF-kappaB pathways. Chem. Biol. Interact. 2021, 344, 109512. [Google Scholar] [CrossRef]
- Zhuang, Y.; Wu, H.; Wang, X.; He, J.; He, S.; Yin, Y. Resveratrol Attenuates Oxidative Stress-Induced Intestinal Barrier Injury through PI3K/Akt-Mediated Nrf2 Signaling Pathway. Oxid. Med. Cell. Longev. 2019, 2019, 7591840. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.; Yang, C.L.; Sha, Y.K.; Wu, Y.; Liu, Z.Y.; Yuan, Z.H.; Sun, Z.L. Koumine Alleviates Lipopolysaccharide-Induced Intestinal Barrier Dysfunction in IPEC-J2 Cells by Regulating Nrf2/NF-kappaB Pathway. Am. J. Chin. Med. 2020, 48, 127–142. [Google Scholar] [CrossRef] [PubMed]
- Lin, Y.; Zheng, X.; Chen, J.; Luo, D.; Xie, J.; Su, Z.; Huang, X.; Yi, X.; Wei, L.; Cai, J.; et al. Protective Effect of Bruguiera gymnorrhiza (L.) Lam. Fruit on Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice: Role of Keap1/Nrf2 Pathway and Gut Microbiota. Front. Pharmacol. 2019, 10, 1602. [Google Scholar] [CrossRef]
- Shi, J.; Xie, Q.; Yue, Y.; Chen, Q.; Zhao, L.; Evivie, S.E.; Li, B.; Huo, G. Gut microbiota modulation and anti-inflammatory properties of mixed lactobacilli in dextran sodium sulfate-induced colitis in mice. Food Funct. 2021, 12, 5130–5143. [Google Scholar] [CrossRef] [PubMed]
- Saeedi, B.J.; Liu, K.H.; Owens, J.A.; Hunter-Chang, S.; Camacho, M.C.; Eboka, R.U.; Chandrasekharan, B.; Baker, N.F.; Darby, T.M.; Robinson, B.S.; et al. Gut-Resident Lactobacilli Activate Hepatic Nrf2 and Protect Against Oxidative Liver Injury. Cell Metab. 2020, 31, 956–968.e5. [Google Scholar] [CrossRef]
- Hansson, G.C. Mucins and the Microbiome. Annu. Rev. Biochem. 2020, 89, 769–793. [Google Scholar] [CrossRef] [PubMed]
- Bel, S.; Pendse, M.; Wang, Y.; Li, Y.; Ruhn, K.A.; Hassell, B.; Leal, T.; Winter, S.E.; Xavier, R.J.; Hooper, L.V. Paneth cells secrete lysozyme via secretory autophagy during bacterial infection of the intestine. Science 2017, 357, 1047–1052. [Google Scholar] [CrossRef] [PubMed]
- Singh, R.; Balasubramanian, I.; Zhang, L.; Gao, N. Metaplastic Paneth Cells in Extra-Intestinal Mucosal Niche Indicate a Link to Microbiome and Inflammation. Front. Physiol. 2020, 11, 280. [Google Scholar] [CrossRef]
- Stagg, A.J. Intestinal Dendritic Cells in Health and Gut Inflammation. Front. Immunol. 2018, 9, 2883. [Google Scholar] [CrossRef]
- Kanaya, T.; Williams, I.R.; Ohno, H. Intestinal M cells: Tireless samplers of enteric microbiota. Traffic 2020, 21, 34–44. [Google Scholar] [CrossRef]
- Rooks, M.G.; Garrett, W.S. Gut microbiota, metabolites and host immunity. Nat. Rev. Immunol. 2016, 16, 341–352. [Google Scholar] [CrossRef] [PubMed]
- Kogut, M.H.; Lee, A.; Santin, E. Microbiome and pathogen interaction with the immune system. Poult. Sci. 2020, 99, 1906–1913. [Google Scholar] [CrossRef] [PubMed]
- Kelly, D.; Conway, S.; Aminov, R. Commensal gut bacteria: Mechanisms of immune modulation. Trends Immunol. 2005, 26, 326–333. [Google Scholar] [CrossRef] [PubMed]
- Mehandru, S.; Colombel, J.F. The intestinal barrier, an arbitrator turned provocateur in IBD. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 83–84. [Google Scholar] [CrossRef] [PubMed]
- Somineni, H.K.; Kugathasan, S. The Microbiome in Patients With Inflammatory Diseases. Clin. Gastroenterol. Hepatol. 2019, 17, 243–255. [Google Scholar] [CrossRef] [PubMed]
- Pimentel, M.; Lembo, A. Microbiome and Its Role in Irritable Bowel Syndrome. Dig. Dis. Sci. 2020, 65, 829–839. [Google Scholar] [CrossRef]
- Wong, S.H.; Yu, J. Gut microbiota in colorectal cancer: Mechanisms of action and clinical applications. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 690–704. [Google Scholar] [CrossRef]
- Ramirez, J.; Guarner, F.; Bustos Fernandez, L.; Maruy, A.; Sdepanian, V.L.; Cohen, H. Antibiotics as Major Disruptors of Gut Microbiota. Front. Cell. Infect. Microbiol. 2020, 10, 572912. [Google Scholar] [CrossRef]
- Weiss, G.A.; Hennet, T. Mechanisms and consequences of intestinal dysbiosis. Cell. Mol. Life Sci. 2017, 74, 2959–2977. [Google Scholar] [CrossRef]
- Wu, Z.; Huang, S.; Li, T.; Li, N.; Han, D.; Zhang, B.; Xu, Z.Z.; Zhang, S.; Pang, J.; Wang, S.; et al. Gut microbiota from green tea polyphenol-dosed mice improves intestinal epithelial homeostasis and ameliorates experimental colitis. Microbiome 2021, 9, 184. [Google Scholar] [CrossRef]
- Nakamoto, N.; Sasaki, N.; Aoki, R.; Miyamoto, K.; Suda, W.; Teratani, T.; Suzuki, T.; Koda, Y.; Chu, P.S.; Taniki, N.; et al. Gut pathobionts underlie intestinal barrier dysfunction and liver T helper 17 cell immune response in primary sclerosing cholangitis. Nat. Microbiol. 2019, 4, 492–503. [Google Scholar] [CrossRef]
- Yu, J.; Zhang, Y.; McIlroy, J.; Rordorf-Nikolic, T.; Orr, G.A.; Backer, J.M. Regulation of the p85/p110 phosphatidylinositol 3′-kinase: Stabilization and inhibition of the p110alpha catalytic subunit by the p85 regulatory subunit. Mol. Cell. Biol. 1998, 18, 1379–1387. [Google Scholar] [CrossRef]
- Burke, J.E.; Williams, R.L. Synergy in activating class I PI3Ks. Trends Biochem. Sci. 2015, 40, 88–100. [Google Scholar] [CrossRef]
- Hoxhaj, G.; Manning, B.D. The PI3K-AKT network at the interface of oncogenic signalling and cancer metabolism. Nat. Rev. Cancer 2020, 20, 74–88. [Google Scholar] [CrossRef] [PubMed]
- Mohseni, A.H.; Casolaro, V.; Bermudez-Humaran, L.G.; Keyvani, H.; Taghinezhad, S.S. Modulation of the PI3K/Akt/mTOR signaling pathway by probiotics as a fruitful target for orchestrating the immune response. Gut Microbes 2021, 13, 1–17. [Google Scholar] [CrossRef] [PubMed]
- Shao, Y.; Wolf, P.G.; Guo, S.; Guo, Y.; Gaskins, H.R.; Zhang, B. Zinc enhances intestinal epithelial barrier function through the PI3K/AKT/mTOR signaling pathway in Caco-2 cells. J. Nutr. Biochem. 2017, 43, 18–26. [Google Scholar] [CrossRef] [PubMed]
- Yanase, K.; Funaguchi, N.; Iihara, H.; Yamada, M.; Kaito, D.; Endo, J.; Ito, F.; Ohno, Y.; Tanaka, H.; Itoh, Y.; et al. Prevention of radiation esophagitis by polaprezinc (zinc L-carnosine) in patients with non-small cell lung cancer who received chemoradiotherapy. Int. J. Clin. Exp. Med. 2015, 8, 16215–16222. [Google Scholar]
- Watari, I.; Oka, S.; Tanaka, S.; Aoyama, T.; Imagawa, H.; Shishido, T.; Yoshida, S.; Chayama, K. Effectiveness of polaprezinc for low-dose aspirin-induced small-bowel mucosal injuries as evaluated by capsule endoscopy: A pilot randomized controlled study. BMC Gastroenterol. 2013, 13, 108. [Google Scholar] [CrossRef]
- Choi, H.S.; Lim, J.Y.; Chun, H.J.; Lee, M.; Kim, E.S.; Keum, B.; Seo, Y.S.; Jeen, Y.T.; Um, S.H.; Lee, H.S.; et al. The effect of polaprezinc on gastric mucosal protection in rats with ethanol-induced gastric mucosal damage: Comparison study with rebamipide. Life Sci. 2013, 93, 69–77. [Google Scholar] [CrossRef]
- Jin, H.O.; Hong, S.E.; Kim, J.Y.; Jang, S.K.; Park, I.C. Amino acid deprivation induces AKT activation by inducing GCN2/ATF4/REDD1 axis. Cell. Death Dis. 2021, 12, 1127. [Google Scholar] [CrossRef]
- Sikalidis, A.K. Amino acids and immune response: A role for cysteine, glutamine, phenylalanine, tryptophan and arginine in T-cell function and cancer? Pathol. Oncol. Res. 2015, 21, 9–17. [Google Scholar] [CrossRef] [PubMed]
- Ma, X.M.; Blenis, J. Molecular mechanisms of mTOR-mediated translational control. Nat. Rev. Mol. Cell. Biol. 2009, 10, 307–318. [Google Scholar] [CrossRef]
- Li, N.; Neu, J. Glutamine deprivation alters intestinal tight junctions via a PI3-K/Akt mediated pathway in Caco-2 cells. J. Nutr. 2009, 139, 710–714. [Google Scholar] [CrossRef]
- Chen, M.; Fu, Q.; Song, X.; Muhammad, A.; Jia, R.; Zou, Y.; Yin, L.; Li, L.; He, C.; Ye, G.; et al. Preparation of resveratrol dry suspension and its immunomodulatory and anti-inflammatory activity in mice. Pharm. Biol. 2020, 58, 8–15. [Google Scholar] [CrossRef] [PubMed]
- Storniolo, C.E.; Moreno, J.J. Resveratrol Analogs with Antioxidant Activity Inhibit Intestinal Epithelial Cancer Caco-2 Cell Growth by Modulating Arachidonic Acid Cascade. J. Agric. Food Chem. 2019, 67, 819–828. [Google Scholar] [CrossRef] [PubMed]
- Corpas, R.; Grinan-Ferre, C.; Rodriguez-Farre, E.; Pallas, M.; Sanfeliu, C. Resveratrol Induces Brain Resilience Against Alzheimer Neurodegeneration Through Proteostasis Enhancement. Mol. Neurobiol. 2019, 56, 1502–1516. [Google Scholar] [CrossRef]
- Zhang, B.; Xu, Y.; Lv, H.; Pang, W.; Wang, J.; Ma, H.; Wang, S. Intestinal pharmacokinetics of resveratrol and regulatory effects of resveratrol metabolites on gut barrier and gut microbiota. Food Chem. 2021, 357, 129532. [Google Scholar] [CrossRef]
- Mayangsari, Y.; Suzuki, T. Resveratrol Ameliorates Intestinal Barrier Defects and Inflammation in Colitic Mice and Intestinal Cells. J. Agric. Food Chem. 2018, 66, 12666–12674. [Google Scholar] [CrossRef]
- He, S.; Guo, Y.; Zhao, J.; Xu, X.; Wang, N.; Liu, Q. Ferulic Acid Ameliorates Lipopolysaccharide-Induced Barrier Dysfunction via MicroRNA-200c-3p-Mediated Activation of PI3K/AKT Pathway in Caco-2 Cells. Front. Pharmacol. 2020, 11, 376. [Google Scholar] [CrossRef]
- Cao, B.; Zhao, R.Y.; Li, H.H.; Xu, X.M.; Cui, H.; Deng, H.; Chen, L.; Wei, B. Oral administration of asparagine and 3-indolepropionic acid prolongs survival time of rats with traumatic colon injury. Mil. Med. Res. 2022, 9, 37. [Google Scholar] [CrossRef]
- Miao, Y.; Lv, Q.; Qiao, S.; Yang, L.; Tao, Y.; Yan, W.; Wang, P.; Cao, N.; Dai, Y.; Wei, Z. Alpinetin improves intestinal barrier homeostasis via regulating AhR/suv39h1/TSC2/mTORC1/autophagy pathway. Toxicol. Appl. Pharmacol. 2019, 384, 114772. [Google Scholar] [CrossRef] [PubMed]
- Li, M.Y.; Luo, H.J.; Wu, X.; Liu, Y.H.; Gan, Y.X.; Xu, N.; Zhang, Y.M.; Zhang, S.H.; Zhou, C.L.; Su, Z.R.; et al. Anti-Inflammatory Effects of Huangqin Decoction on Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice Through Regulation of the Gut Microbiota and Suppression of the Ras-PI3K-Akt-HIF-1alpha and NF-kappaB Pathways. Front. Pharmacol. 2019, 10, 1552. [Google Scholar] [CrossRef] [PubMed]
- Liu, B.; Piao, X.; Niu, W.; Zhang, Q.; Ma, C.; Wu, T.; Gu, Q.; Cui, T.; Li, S. Kuijieyuan Decoction Improved Intestinal Barrier Injury of Ulcerative Colitis by Affecting TLR4-Dependent PI3K/AKT/NF-kappaB Oxidative and Inflammatory Signaling and Gut Microbiota. Front. Pharmacol. 2020, 11, 1036. [Google Scholar] [CrossRef] [PubMed]
- Kabiri, Z.; Greicius, G.; Zaribafzadeh, H.; Hemmerich, A.; Counter, C.M.; Virshup, D.M. Wnt signaling suppresses MAPK-driven proliferation of intestinal stem cells. J. Clin. Investig. 2018, 128, 3806–3812. [Google Scholar] [CrossRef] [PubMed]
- He, D.; Wu, H.; Xiang, J.; Ruan, X.; Peng, P.; Ruan, Y.; Chen, Y.G.; Wang, Y.; Yu, Q.; Zhang, H.; et al. Gut stem cell aging is driven by mTORC1 via a p38 MAPK-p53 pathway. Nat. Commun. 2020, 11, 37. [Google Scholar] [CrossRef]
- Xiang, D.C.; Yang, J.Y.; Xu, Y.J.; Zhang, S.; Li, M.; Zhu, C.; Zhang, C.L.; Liu, D. Protective effect of Andrographolide on 5-Fu induced intestinal mucositis by regulating p38 MAPK signaling pathway. Life Sci. 2020, 252, 117612. [Google Scholar] [CrossRef] [PubMed]
- Kaur, N.; Lum, M.; Lewis, R.E.; Black, A.R.; Black, J.D. A novel anti-proliferative PKCalpha-Ras-ERK signaling axis in intestinal epithelial cells. J. Biol. Chem. 2022, 298, 102121. [Google Scholar] [CrossRef]
- Lemieux, E.; Boucher, M.J.; Mongrain, S.; Boudreau, F.; Asselin, C.; Rivard, N. Constitutive activation of the MEK/ERK pathway inhibits intestinal epithelial cell differentiation. Am. J. Physiol. Gastrointest. Liver Physiol. 2011, 301, G719–G730. [Google Scholar] [CrossRef]
- Zhang, H.; Deng, X.; Zhou, C.; Wu, W.; Zhang, H. Deoxynivalenol Induces Inflammation in IPEC-J2 Cells by Activating P38 Mapk And Erk1/2. Toxins 2020, 12, 180. [Google Scholar] [CrossRef]
- Qu, F.; Xu, W.; Deng, Z.; Xie, Y.; Tang, J.; Chen, Z.; Luo, W.; Xiong, D.; Zhao, D.; Fang, J.; et al. Fish c-Jun N-Terminal Kinase (JNK) Pathway Is Involved in Bacterial MDP-Induced Intestinal Inflammation. Front. Immunol. 2020, 11, 459. [Google Scholar] [CrossRef]
- Qi, H.; Fu, H.; Dong, X.; Feng, D.; Li, N.; Wen, C.; Nakamura, Y.; Zhu, B. Apoptosis induction is involved in UVA-induced autolysis in sea cucumber Stichopus japonicus. J. Photochem. Photobiol. B 2016, 158, 130–135. [Google Scholar] [CrossRef] [PubMed]
- Zhai, Y.; Dang, Y.; Gao, W.; Zhang, Y.; Xu, P.; Gu, J.; Ye, X. P38 and JNK signal pathways are involved in the regulation of phlorizin against UVB-induced skin damage. Exp. Dermatol. 2015, 24, 275–279. [Google Scholar] [CrossRef] [PubMed]
- Upadhyay, R.; Mohan Rao, L.J. An outlook on chlorogenic acids-occurrence, chemistry, technology, and biological activities. Crit. Rev. Food Sci. Nutr. 2013, 53, 968–984. [Google Scholar] [CrossRef] [PubMed]
- Gao, W.; Wang, C.; Yu, L.; Sheng, T.; Wu, Z.; Wang, X.; Zhang, D.; Lin, Y.; Gong, Y. Chlorogenic Acid Attenuates Dextran Sodium Sulfate-Induced Ulcerative Colitis in Mice through MAPK/ERK/JNK Pathway. Biomed. Res. Int. 2019, 2019, 6769789. [Google Scholar] [CrossRef] [PubMed]
- Ran, X.; Li, Y.; Chen, G.; Fu, S.; He, D.; Huang, B.; Wei, L.; Lin, Y.; Guo, Y.; Hu, G. Farrerol Ameliorates TNBS-Induced Colonic Inflammation by Inhibiting ERK1/2, JNK1/2, and NF-kappaB Signaling Pathway. Int. J. Mol. Sci. 2018, 19, 2037. [Google Scholar] [CrossRef] [PubMed]
- Shi, Y.; Zhou, M.; Yan, J.; Gong, Z.; Wu, J.; Chen, Y.; Chen, Y. N-Acetyl-Seryl-Aspartyl-Lysyl-Proline Mitigates Experimental Colitis Through Inhibition of Intestinal Mucosal Inflammatory Responses via MEK-ERK Signaling. Front. Pharmacol. 2020, 11, 593. [Google Scholar] [CrossRef]
- Jones, R.M.; Sloane, V.M.; Wu, H.; Luo, L.; Kumar, A.; Kumar, M.V.; Gewirtz, A.T.; Neish, A.S. Flagellin administration protects gut mucosal tissue from irradiation-induced apoptosis via MKP-7 activity. Gut 2011, 60, 648–657. [Google Scholar] [CrossRef] [PubMed]
- Li, E.; Horn, N.; Ajuwon, K.M. Mechanisms of deoxynivalenol-induced endocytosis and degradation of tight junction proteins in jejunal IPEC-J2 cells involve selective activation of the MAPK pathways. Arch. Toxicol. 2021, 95, 2065–2079. [Google Scholar] [CrossRef]
- Sadeghi, A.; Seyyed Ebrahimi, S.S.; Golestani, A.; Meshkani, R. Resveratrol Ameliorates Palmitate-Induced Inflammation in Skeletal Muscle Cells by Attenuating Oxidative Stress and JNK/NF-kappaB Pathway in a SIRT1-Independent Mechanism. J. Cell. Biochem. 2017, 118, 2654–2663. [Google Scholar] [CrossRef]
- Tang, M.; Yuan, D.; Liao, P. Berberine improves intestinal barrier function and reduces inflammation, immunosuppression, and oxidative stress by regulating the NF-kappaB/MAPK signaling pathway in deoxynivalenol-challenged piglets. Environ. Pollut. 2021, 289, 117865. [Google Scholar] [CrossRef]
- Jiang, P.; Ren, L.; Zhi, L.; Yu, Z.; Lv, F.; Xu, F.; Peng, W.; Bai, X.; Cheng, K.; Quan, L.; et al. Negative regulation of AMPK signaling by high glucose via E3 ubiquitin ligase MG53. Mol. Cell. 2021, 81, 629–637.e5. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Xia, D.; Chen, J.; Zhang, X.; Wang, H.; Huang, L.; Shen, J.; Wang, S.; Feng, Y.; He, D.; et al. Dietary fibers with different viscosity regulate lipid metabolism via ampk pathway: Roles of gut microbiota and short-chain fatty acid. Poult. Sci. 2022, 101, 101742. [Google Scholar] [CrossRef] [PubMed]
- Sanchez-Lopez, E.; Zhong, Z.; Stubelius, A.; Sweeney, S.R.; Booshehri, L.M.; Antonucci, L.; Liu-Bryan, R.; Lodi, A.; Terkeltaub, R.; Lacal, J.C.; et al. Choline Uptake and Metabolism Modulate Macrophage IL-1beta and IL-18 Production. Cell Metab. 2019, 29, 1350–1362.e7. [Google Scholar] [CrossRef] [PubMed]
- Lin, S.C.; Hardie, D.G. AMPK: Sensing Glucose as well as Cellular Energy Status. Cell Metab. 2018, 27, 299–313. [Google Scholar] [CrossRef] [PubMed]
- Furuzono, S.; Kubota, T.; Taura, J.; Konishi, M.; Naito, A.; Tsutsui, M.; Karasawa, H.; Kubota, N.; Kadowaki, T. A xanthene derivative, DS20060511, attenuates glucose intolerance by inducing skeletal muscle-specific GLUT4 translocation in mice. Commun. Biol. 2021, 4, 994. [Google Scholar] [CrossRef] [PubMed]
- Wu, W.; Wang, S.; Liu, Q.; Shan, T.; Wang, X.; Feng, J.; Wang, Y. AMPK facilitates intestinal long-chain fatty acid uptake by manipulating CD36 expression and translocation. FASEB J. 2020, 34, 4852–4869. [Google Scholar] [CrossRef] [PubMed]
- Schaffler, A.; Scholmerich, J. Innate immunity and adipose tissue biology. Trends Immunol. 2010, 31, 228–235. [Google Scholar] [CrossRef]
- Chen, L.; Vasoya, R.P.; Toke, N.H.; Parthasarathy, A.; Luo, S.; Chiles, E.; Flores, J.; Gao, N.; Bonder, E.M.; Su, X.; et al. HNF4 Regulates Fatty Acid Oxidation and Is Required for Renewal of Intestinal Stem Cells in Mice. Gastroenterology 2020, 158, 985–999.e9. [Google Scholar] [CrossRef]
- Olivier, S.; Diounou, H.; Pochard, C.; Frechin, L.; Durieu, E.; Foretz, M.; Neunlist, M.; Rolli-Derkinderen, M.; Viollet, B. Intestinal Epithelial AMPK Deficiency Causes Delayed Colonic Epithelial Repair in DSS-Induced Colitis. Cells 2022, 11, 590. [Google Scholar] [CrossRef]
- Sun, X.; Yang, Q.; Rogers, C.J.; Du, M.; Zhu, M.J. AMPK improves gut epithelial differentiation and barrier function via regulating Cdx2 expression. Cell. Death Differ. 2017, 24, 819–831. [Google Scholar] [CrossRef]
- Olivier, S.; Leclerc, J.; Grenier, A.; Foretz, M.; Tamburini, J.; Viollet, B. AMPK Activation Promotes Tight Junction Assembly in Intestinal Epithelial Caco-2 Cells. Int. J. Mol. Sci. 2019, 20, 5171. [Google Scholar] [CrossRef] [PubMed]
- Banskota, S.; Wang, H.; Kwon, Y.H.; Gautam, J.; Gurung, P.; Haq, S.; Hassan, F.M.N.; Bowdish, D.M.; Kim, J.A.; Carling, D.; et al. Salicylates Ameliorate Intestinal Inflammation by Activating Macrophage AMPK. Inflamm. Bowel Dis. 2021, 27, 914–926. [Google Scholar] [CrossRef] [PubMed]
- Li, Y.; Yan, H.; Zhang, Y.; Li, Q.; Yu, L.; Li, Q.; Liu, C.; Xie, Y.; Chen, K.; Ye, F.; et al. Alterations of the Gut Microbiome Composition and Lipid Metabolic Profile in Radiation Enteritis. Front. Cell. Infect. Microbiol. 2020, 10, 541178. [Google Scholar] [CrossRef] [PubMed]
- Beck, P.L.; Wong, J.F.; Li, Y.; Swaminathan, S.; Xavier, R.J.; Devaney, K.L.; Podolsky, D.K. Chemotherapy- and radiotherapy-induced intestinal damage is regulated by intestinal trefoil factor. Gastroenterology 2004, 126, 796–808. [Google Scholar] [CrossRef] [PubMed]
- Loge, L.; Florescu, C.; Alves, A.; Menahem, B. Radiation enteritis: Diagnostic and therapeutic issues. J. Visc. Surg. 2020, 157, 475–485. [Google Scholar] [CrossRef] [PubMed]
- Wang, Z.; Chen, Z.; Jiang, Z.; Luo, P.; Liu, L.; Huang, Y.; Wang, H.; Wang, Y.; Long, L.; Tan, X.; et al. Cordycepin prevents radiation ulcer by inhibiting cell senescence via NRF2 and AMPK in rodents. Nat. Commun. 2019, 10, 2538. [Google Scholar] [CrossRef] [PubMed]
- Wen, J.; Xu, B.; Sun, Y.; Lian, M.; Li, Y.; Lin, Y.; Chen, D.; Diao, Y.; Almoiliqy, M.; Wang, L. Paeoniflorin protects against intestinal ischemia/reperfusion by activating LKB1/AMPK and promoting autophagy. Pharmacol. Res. 2019, 146, 104308. [Google Scholar] [CrossRef] [PubMed]
- Ahmed, A.; Tait, S.W.G. Targeting immunogenic cell death in cancer. Mol. Oncol. 2020, 14, 2994–3006. [Google Scholar] [CrossRef]
- Thota, S.; Begum, R.; Kaur, G.; Bagam, P.; Dorsey, W.; Batra, S. Pentachlorophenol mediated regulation of DAMPs and inflammation: In vitro study. Toxicol. In Vitro 2022, 83, 105378. [Google Scholar] [CrossRef]
- Haddad, T.C.; Greeno, E.W. Chemotherapy-induced thrombosis. Thromb. Res. 2006, 118, 555–568. [Google Scholar] [CrossRef]
- Yu, Q.; Yang, X.; Zhang, C.; Zhang, X.; Wang, C.; Chen, L.; Liu, X.; Gu, Y.; He, X.; Hu, L.; et al. AMPK activation by ozone therapy inhibits tissue factor-triggered intestinal ischemia and ameliorates chemotherapeutic enteritis. FASEB J. 2020, 34, 13005–13021. [Google Scholar] [CrossRef] [PubMed]
- Song, M.; Zeng, Q.; Xiang, Y.; Gao, L.; Huang, J.; Huang, J.; Wu, K.; Lu, J. The antibacterial effect of topical ozone on the treatment of MRSA skin infection. Mol. Med. Rep. 2018, 17, 2449–2455. [Google Scholar] [CrossRef] [PubMed]
- Heim, M.H.; Kerr, I.M.; Stark, G.R.; Darnell, J.E., Jr. Contribution of STAT SH2 groups to specific interferon signaling by the Jak-STAT pathway. Science 1995, 267, 1347–1349. [Google Scholar] [CrossRef] [PubMed]
- Glassman, C.R.; Tsutsumi, N.; Saxton, R.A.; Lupardus, P.J.; Jude, K.M.; Garcia, K.C. Structure of a Janus kinase cytokine receptor complex reveals the basis for dimeric activation. Science 2022, 376, 163–169. [Google Scholar] [CrossRef] [PubMed]
- Dariya, B.; Muppala, S.; Srivani, G.; Momin, S.; Alam, A.; Saddala, M.S. Targeting STAT proteins via computational analysis in colorectal cancer. Mol. Cell. Biochem. 2021, 476, 165–174. [Google Scholar] [CrossRef] [PubMed]
- Richmond, C.A.; Rickner, H.; Shah, M.S.; Ediger, T.; Deary, L.; Zhou, F.; Tovaglieri, A.; Carlone, D.L.; Breault, D.T. JAK/STAT-1 Signaling Is Required for Reserve Intestinal Stem Cell Activation during Intestinal Regeneration Following Acute Inflammation. Stem Cell. Rep. 2018, 10, 17–26. [Google Scholar] [CrossRef] [PubMed]
- Srinivasan, T.; Than, E.B.; Bu, P.; Tung, K.L.; Chen, K.Y.; Augenlicht, L.; Lipkin, S.M.; Shen, X. Notch signalling regulates asymmetric division and inter-conversion between lgr5 and bmi1 expressing intestinal stem cells. Sci. Rep. 2016, 6, 26069. [Google Scholar] [CrossRef]
- Du, G.; Xiong, L.; Li, X.; Zhuo, Z.; Zhuang, X.; Yu, Z.; Wu, L.; Xiao, D.; Liu, Z.; Jie, M.; et al. Peroxisome Elevation Induces Stem Cell Differentiation and Intestinal Epithelial Repair. Dev. Cell. 2020, 53, 169–184.e11. [Google Scholar] [CrossRef]
- Yang, S.; Li, X.; Xiu, M.; Dai, Y.; Wan, S.; Shi, Y.; Liu, Y.; He, J. Flos puerariae ameliorates the intestinal inflammation of Drosophila via modulating the Nrf2/Keap1, JAK-STAT and Wnt signaling. Front. Pharmacol. 2022, 13, 893758. [Google Scholar] [CrossRef]
- Wang, L.; Hu, Y.; Song, B.; Xiong, Y.; Wang, J.; Chen, D. Targeting JAK/STAT signaling pathways in treatment of inflammatory bowel disease. Inflamm. Res. Off. J. Eur. Histamine Res. Soc. 2021, 70, 753–764. [Google Scholar] [CrossRef]
- Nunes, C.; Almeida, L.; Barbosa, R.M.; Laranjinha, J. Luteolin suppresses the JAK/STAT pathway in a cellular model of intestinal inflammation. Food Funct. 2017, 8, 387–396. [Google Scholar] [CrossRef] [PubMed]
- Hong, Z.S.; Xie, J.; Wang, X.F.; Dai, J.J.; Mao, J.Y.; Bai, Y.Y.; Sheng, J.; Tian, Y. Moringa oleifera Lam. Peptide Remodels Intestinal Mucosal Barrier by Inhibiting JAK-STAT Activation and Modulating Gut Microbiota in Colitis. Front. Immunol. 2022, 13, 924178. [Google Scholar] [CrossRef] [PubMed]
- Li, M.X.; Li, M.Y.; Lei, J.X.; Wu, Y.Z.; Li, Z.H.; Chen, L.M.; Zhou, C.L.; Su, J.Y.; Huang, G.X.; Huang, X.Q.; et al. Huangqin decoction ameliorates DSS-induced ulcerative colitis: Role of gut microbiota and amino acid metabolism, mTOR pathway and intestinal epithelial barrier. Phytomedicine 2022, 100, 154052. [Google Scholar] [CrossRef] [PubMed]
- Wen, S.H.; Li, Y.; Li, C.; Xia, Z.Q.; Liu, W.F.; Zhang, X.Y.; Lei, W.L.; Huang, W.Q.; Liu, K.X. Ischemic postconditioning during reperfusion attenuates intestinal injury and mucosal cell apoptosis by inhibiting JAK/STAT signaling activation. Shock 2012, 38, 411–419. [Google Scholar] [CrossRef] [PubMed]
- Liu, Z.; Hu, K.; Chen, Y.S.; Huang, Y.J.; Hu, Q.; Zeng, W.; Cao, Y.; Xiao, Q.; Zhang, X.K. JAK2/STAT3 inhibition attenuates intestinal ischemia-reperfusion injury via promoting autophagy: In vitro and in vivo study. Mol. Biol. Rep. 2022, 49, 2857–2867. [Google Scholar] [CrossRef]
- Jia, L.; Yu, W.; Yu, H.; Weng, Y. Electroacupuncture Pretreatment Attenuates Intestinal Injury after Autogenous Orthotopic Liver Transplantation in Rats via the JAK/STAT Pathway. Oxid. Med. Cell. Longev. 2020, 2020, 9187406. [Google Scholar] [CrossRef] [PubMed]
- Gilmore, T.D. Introduction to NF-kappaB: Players, pathways, perspectives. Oncogene 2006, 25, 6680–6684. [Google Scholar] [CrossRef] [PubMed]
- Beinke, S.; Ley, S.C. Functions of NF-kappaB1 and NF-kappaB2 in immune cell biology. Biochem. J. 2004, 382, 393–409. [Google Scholar] [CrossRef]
- Ghosh, S.; May, M.J.; Kopp, E.B. NF-kappa B and Rel proteins: Evolutionarily conserved mediators of immune responses. Annu. Rev. Immunol. 1998, 16, 225–260. [Google Scholar] [CrossRef]
- Baeuerle, P.A.; Baltimore, D. I kappa B: A specific inhibitor of the NF-kappa B transcription factor. Science 1988, 242, 540–546. [Google Scholar] [CrossRef]
- Hayden, M.S.; Ghosh, S. NF-kappaB in immunobiology. Cell Res. 2011, 21, 223–244. [Google Scholar] [CrossRef] [PubMed]
- Karin, M.; Ben-Neriah, Y. Phosphorylation meets ubiquitination: The control of NF-[kappa]B activity. Annu. Rev. Immunol. 2000, 18, 621–663. [Google Scholar] [CrossRef] [PubMed]
- Hayden, M.S.; Ghosh, S. Shared principles in NF-kappaB signaling. Cell 2008, 132, 344–362. [Google Scholar] [CrossRef] [PubMed]
- Venkatesh, M.; Mukherjee, S.; Wang, H.; Li, H.; Sun, K.; Benechet, A.P.; Qiu, Z.; Maher, L.; Redinbo, M.R.; Phillips, R.S.; et al. Symbiotic bacterial metabolites regulate gastrointestinal barrier function via the xenobiotic sensor PXR and Toll-like receptor 4. Immunity 2014, 41, 296–310. [Google Scholar] [CrossRef] [PubMed]
- Dou, W.; Zhang, J.; Sun, A.; Zhang, E.; Ding, L.; Mukherjee, S.; Wei, X.; Chou, G.; Wang, Z.T.; Mani, S. Protective effect of naringenin against experimental colitis via suppression of Toll-like receptor 4/NF-kappaB signalling. Br. J. Nutr. 2013, 110, 599–608. [Google Scholar] [CrossRef] [PubMed]
- Mahmoud, T.N.; El-Maadawy, W.H.; Kandil, Z.A.; Khalil, H.; El-Fiky, N.M.; El Alfy, T. Canna x generalis L.H. Bailey rhizome extract ameliorates dextran sulfate sodium-induced colitis via modulating intestinal mucosal dysfunction, oxidative stress, inflammation, and TLR4/NF-B and NLRP3 inflammasome pathways. J. Ethnopharmacol. 2021, 269, 113670. [Google Scholar] [CrossRef] [PubMed]
- Huang, Y.F.; Li, Q.P.; Dou, Y.X.; Wang, T.T.; Qu, C.; Liang, J.L.; Lin, Z.X.; Huang, X.Q.; Su, Z.R.; Chen, J.N.; et al. Therapeutic effect of Brucea javanica oil emulsion on experimental Crohn’s disease in rats: Involvement of TLR4/NF-kappaB signaling pathway. Biomed. Pharmacother. Biomed. Pharmacother. 2019, 114, 108766. [Google Scholar] [CrossRef]
- Huang, Y.F.; Zhou, J.T.; Qu, C.; Dou, Y.X.; Huang, Q.H.; Lin, Z.X.; Xian, Y.F.; Xie, J.H.; Xie, Y.L.; Lai, X.P.; et al. Anti-inflammatory effects of Brucea javanica oil emulsion by suppressing NF-kappaB activation on dextran sulfate sodium-induced ulcerative colitis in mice. J. Ethnopharmacol. 2017, 198, 389–398. [Google Scholar] [CrossRef]
- Guo, H.; Chou, W.C.; Lai, Y.; Liang, K.; Tam, J.W.; Brickey, W.J.; Chen, L.; Montgomery, N.D.; Li, X.; Bohannon, L.M.; et al. Multi-omics analyses of radiation survivors identify radioprotective microbes and metabolites. Science 2020, 370, eaay9097. [Google Scholar] [CrossRef]
- Shim, S.; Jang, H.S.; Myung, H.W.; Myung, J.K.; Kang, J.K.; Kim, M.J.; Lee, S.B.; Jang, W.S.; Lee, S.J.; Jin, Y.W.; et al. Rebamipide ameliorates radiation-induced intestinal injury in a mouse model. Toxicol. Appl. Pharmacol. 2017, 329, 40–47. [Google Scholar] [CrossRef]
- Zhao, Z.; Cheng, W.; Qu, W.; Shao, G.; Liu, S. Antibiotic Alleviates Radiation-Induced Intestinal Injury by Remodeling Microbiota, Reducing Inflammation, and Inhibiting Fibrosis. ACS Omega 2020, 5, 2967–2977. [Google Scholar] [CrossRef] [PubMed]
- Kojima, M.; Hosoda, H.; Date, Y.; Nakazato, M.; Matsuo, H.; Kangawa, K. Ghrelin is a growth-hormone-releasing acylated peptide from stomach. Nature 1999, 402, 656–660. [Google Scholar] [CrossRef]
- Kwak, S.Y.; Shim, S.; Park, S.; Kim, H.; Lee, S.J.; Kim, M.J.; Jang, W.S.; Kim, Y.H.; Jang, H. Ghrelin reverts intestinal stem cell loss associated with radiation-induced enteropathy by activating Notch signaling. Phytomedicine 2021, 81, 153424. [Google Scholar] [CrossRef] [PubMed]
- Wu, H.; Chen, Q.Y.; Wang, W.Z.; Chu, S.; Liu, X.X.; Liu, Y.J.; Tan, C.; Zhu, F.; Deng, S.J.; Dong, Y.L.; et al. Compound sophorae decoction enhances intestinal barrier function of dextran sodium sulfate induced colitis via regulating notch signaling pathway in mice. Biomed. Pharmacother. Biomed. Pharmacother. 2021, 133, 110937. [Google Scholar] [CrossRef] [PubMed]
- Lin, J.C.; Wu, J.Q.; Wang, F.; Tang, F.Y.; Sun, J.; Xu, B.; Jiang, M.; Chu, Y.; Chen, D.; Li, X.; et al. QingBai decoction regulates intestinal permeability of dextran sulphate sodium-induced colitis through the modulation of notch and NF-kappaB signalling. Cell Prolif. 2019, 52, e12547. [Google Scholar] [CrossRef]
- Wei, Y.Y.; Fan, Y.M.; Ga, Y.; Zhang, Y.N.; Han, J.C.; Hao, Z.H. Shaoyao decoction attenuates DSS-induced ulcerative colitis, macrophage and NLRP3 inflammasome activation through the MKP1/NF-kappaB pathway. Phytomedicine 2021, 92, 153743. [Google Scholar] [CrossRef]
- Cui, Y.; Wang, Q.; Wang, M.; Jia, J.; Wu, R. Gardenia Decoction Prevent Intestinal Mucosal Injury by Inhibiting Pro-inflammatory Cytokines and NF-kappaB Signaling. Front. Pharmacol. 2019, 10, 180. [Google Scholar] [CrossRef]
- Xuan-Qing, C.; Xiang-Yu, L.V.; Shi-Jia, L.I.U. Baitouweng decoction alleviates dextran sulfate sodium-induced ulcerative colitis by regulating intestinal microbiota and the IL-6/STAT3 signaling pathway. J. Ethnopharmacol. 2021, 265, 113357. [Google Scholar] [CrossRef]
- Zhao, Y.; Luan, H.; Jiang, H.; Xu, Y.; Wu, X.; Zhang, Y.; Li, R. Gegen Qinlian decoction relieved DSS-induced ulcerative colitis in mice by modulating Th17/Treg cell homeostasis via suppressing IL-6/JAK2/STAT3 signaling. Phytomedicine 2021, 84, 153519. [Google Scholar] [CrossRef]
- Zheng, Y.; Liang, C.; Li, Z.; Chen, J.; Chen, Z.; Jiang, Y.; Dong, Q.; Xiao, Y.; Fu, C.; Liao, W.; et al. Study on the mechanism of Huangqin Decoction on rats with ulcerative colitis of damp-heat type base on mtDNA, TLR4, p-PI3K, p-Akt protein expression and microbiota. J. Ethnopharmacol. 2022, 295, 115356. [Google Scholar] [CrossRef]
- Yan, Z.; Zhang, K.; Zhang, K.; Wang, G.; Wang, L.; Zhang, J.; Qiu, Z.; Guo, Z.; Kang, Y.; Song, X.; et al. Huang Bai Jian Pi decoction alleviates diarrhea and represses inflammatory injury via PI3K/Akt/NF-kappaB pathway: In vivo and in vitro studies. J. Ethnopharmacol. 2022, 292, 115212. [Google Scholar] [CrossRef] [PubMed]
- Lu, Z.; Xiong, W.; Xiao, S.; Lin, Y.; Yu, K.; Yue, G.; Liu, Q.; Li, F.; Liang, J. Huanglian Jiedu Decoction ameliorates DSS-induced colitis in mice via the JAK2/STAT3 signalling pathway. Chin. Med. 2020, 15, 45. [Google Scholar] [CrossRef] [PubMed]
- Yuan, Z.; Yang, L.; Zhang, X.; Ji, P.; Hua, Y.; Wei, Y. Huang-Lian-Jie-Du Decoction Ameliorates Acute Ulcerative Colitis in Mice via Regulating NF-kappaB and Nrf2 Signaling Pathways and Enhancing Intestinal Barrier Function. Front. Pharmacol. 2019, 10, 1354. [Google Scholar] [CrossRef] [PubMed]
- Zhao, Y.; Luan, H.; Gao, H.; Wu, X.; Zhang, Y.; Li, R. Gegen Qinlian decoction maintains colonic mucosal homeostasis in acute/chronic ulcerative colitis via bidirectionally modulating dysregulated Notch signaling. Phytomedicine 2020, 68, 153182. [Google Scholar] [CrossRef] [PubMed]
- Mo, X.; Tang, K.; Deng, L.; Zhou, X.; Li, X.; Zhang, Y.; Wang, J. Prevention of ulcerative colitis by Huangqin decoction: Reducing the intestinal epithelial cell apoptosis rate through the IFN-gamma/JAK/ETS signalling pathway. Pharm. Biol. 2022, 60, 1116–1125. [Google Scholar] [CrossRef]
- Wang, J.; Dempsey, E.; Corr, S.C.; Kukula-Koch, W.; Sasse, A.; Sheridan, H. The Traditional Chinese Medicine Houttuynia cordata Thunb decoction alters intestinal barrier function via an EGFR dependent MAPK (ERK1/2) signalling pathway. Phytomedicine 2022, 105, 154353. [Google Scholar] [CrossRef] [PubMed]
- Wen, Y.; Zhan, Y.; Tang, S.Y.; Liu, F.; Wang, Q.X.; Kong, P.F.; Tang, X.G. Zhizhu Decoction Alleviates Intestinal Barrier Damage via Regulating SIRT1/FoxO1 Signaling Pathway in Slow Transit Constipation Model Mice. Chin. J. Integr. Med. 2022. [Google Scholar] [CrossRef]
- Leng, J.; Huang, F.; Hai, Y.; Tian, H.; Liu, W.; Fang, Y.; Hu, Y.; Peng, J. Amelioration of non-alcoholic steatohepatitis by Qushi Huayu decoction is associated with inhibition of the intestinal mitogen-activated protein kinase pathway. Phytomedicine 2020, 66, 153135. [Google Scholar] [CrossRef]
- Wu, X.; Fu, S.; Jiang, M.; Wang, J.; Tang, H.; Fang, C.; Li, W.; Fu, C. Sanhuang Xiexin decoction ameliorates DSS-induced colitis in mice by regulating intestinal inflammation, intestinal barrier, and intestinal flora. J. Ethnopharmacol. 2022, 297, 115537. [Google Scholar] [CrossRef]
- Otterson, M.F.; Leming, S.C.; Fox, C.J.; Moulder, J.E. Propagation of giant migrating contractions between the small intestine, cecum and colon during radiation. Neurogastroenterol. Motil. Off. J. Eur. Gastrointest. Motil. Soc. 2010, 22, 919–926. [Google Scholar] [CrossRef]
- Li, N.; Zuo, B.; Huang, S.; Zeng, B.; Han, D.; Li, T.; Liu, T.; Wu, Z.; Wei, H.; Zhao, J.; et al. Spatial heterogeneity of bacterial colonization across different gut segments following inter-species microbiota transplantation. Microbiome 2020, 8, 161. [Google Scholar] [CrossRef] [PubMed]
- Heap, G.A.; So, K.; Weedon, M.; Edney, N.; Bewshea, C.; Singh, A.; Annese, V.; Beckly, J.; Buurman, D.; Chaudhary, R.; et al. Clinical Features and HLA Association of 5-Aminosalicylate (5-ASA)-induced Nephrotoxicity in Inflammatory Bowel Disease. J. Crohns Colitis 2016, 10, 149–158. [Google Scholar] [CrossRef]
- Suzuki, K.; Kakuta, Y.; Naito, T.; Takagawa, T.; Hanai, H.; Araki, H.; Sasaki, Y.; Sakuraba, H.; Sasaki, M.; Hisamatsu, T.; et al. Genetic Background of Mesalamine-induced Fever and Diarrhea in Japanese Patients with Inflammatory Bowel Disease. Inflamm. Bowel Dis. 2022, 28, 21–31. [Google Scholar] [CrossRef]
- Sandborn, W.J.; Su, C.; Sands, B.E.; D’Haens, G.R.; Vermeire, S.; Schreiber, S.; Danese, S.; Feagan, B.G.; Reinisch, W.; Niezychowski, W.; et al. Tofacitinib as Induction and Maintenance Therapy for Ulcerative Colitis. N. Engl. J. Med. 2017, 376, 1723–1736. [Google Scholar] [CrossRef] [PubMed]
- D’Haens, G.; Panes, J.; Louis, E.; Lacerda, A.; Zhou, Q.; Liu, J.; Loftus, E.V., Jr. Upadacitinib Was Efficacious and Well-tolerated Over 30 Months in Patients With Crohn’s Disease in the CELEST Extension Study. Clin. Gastroenterol. Hepatol. 2021, 20, 2337–2346. [Google Scholar] [CrossRef] [PubMed]
- Sandborn, W.J.; Ghosh, S.; Panes, J.; Schreiber, S.; D’Haens, G.; Tanida, S.; Siffledeen, J.; Enejosa, J.; Zhou, W.; Othman, A.A.; et al. Efficacy of Upadacitinib in a Randomized Trial of Patients With Active Ulcerative Colitis. Gastroenterology 2020, 158, 2139–2149.e14. [Google Scholar] [CrossRef] [PubMed]
- Chen, J.; Luo, Y.; Li, Y.; Chen, D.; Yu, B.; He, J. Chlorogenic Acid Attenuates Oxidative Stress-Induced Intestinal Epithelium Injury by Co-Regulating the PI3K/Akt and IκBα/NF-κB Signaling. Antioxidants 2021, 10, 1915. [Google Scholar] [CrossRef]
- Gao, T.; Wang, T.; Wang, Z.; Cao, J.; Dong, Y.; Chen, Y. Melatonin-mediated MT2 attenuates colitis induced by dextran sodium sulfate via PI3K/AKT/Nrf2/SIRT1/RORα/NF-κB signaling pathways. Int. Immunopharmacol. 2021, 96, 107779. [Google Scholar] [CrossRef]
- Rahmani, F.; Asgharzadeh, F.; Avan, A.; Barneh, F.; Parizadeh, M.R.; Ferns, G.A.; Ryzhikov, M.; Ahmadian, M.R.; Giovannetti, E.; Jafari, M.; et al. Rigosertib potently protects against colitis-associated intestinal fibrosis and inflammation by regulating PI3K/AKT and NF-κB signaling pathways. Life Sci. 2020, 249, 117470. [Google Scholar] [CrossRef]
- Kobayashi, T.; Motoya, S.; Nakamura, S.; Yamamoto, T.; Nagahori, M.; Tanaka, S.; Hisamatsu, T.; Hirai, F.; Nakase, H.; Watanabe, K.; et al. Discontinuation of infliximab in patients with ulcerative colitis in remission (HAYABUSA): A multicentre, open-label, randomised controlled trial. Lancet. Gastroenterol. Hepatol. 2021, 6, 429–437. [Google Scholar] [CrossRef]
- Roblin, X.; Williet, N.; Boschetti, G.; Phelip, J.M.; Del Tedesco, E.; Berger, A.E.; Vedrines, P.; Duru, G.; Peyrin-Biroulet, L.; Nancey, S.; et al. Addition of azathioprine to the switch of anti-TNF in patients with IBD in clinical relapse with undetectable anti-TNF trough levels and antidrug antibodies: A prospective randomised trial. Gut 2020, 69, 1206–1212. [Google Scholar] [CrossRef]
- Schreiber, S.; Ben-Horin, S.; Leszczyszyn, J.; Dudkowiak, R.; Lahat, A.; Gawdis-Wojnarska, B.; Pukitis, A.; Horynski, M.; Farkas, K.; Kierkus, J.; et al. Randomized Controlled Trial: Subcutaneous vs Intravenous Infliximab CT-P13 Maintenance in Inflammatory Bowel Disease. Gastroenterology 2021, 160, 2340–2353. [Google Scholar] [CrossRef]
Medicine | Signaling Pathway | Activation or Inhibition | Intestinal Pathologies | Effects | References |
---|---|---|---|---|---|
Compound sophorae decoction | Notch | Inhibition | UC | Reduced the loss of muc-2 and goblet cells. Repairs damage of the epithelial cell barrier and prevents mucosal barrier damage, thus improving permeability. | [194] |
QingBai decoction | NF-κB and Notch | Inhibition | UC, CD | Inhibits the expression of pro-inflammatory cytokines and prevents the loss of muc-2 | [195] |
Shaoyao decoction | MKP1/NF-κB/NLRP3 | Inhibition | UC | Regulates inflammatory factor homeostasis. Inhibits inflammation, necrosis, and pyroptosis. | [196] |
Gardenia decoction | NF-κB | Inhibition | Endotoxin-induced intestinal mucosal injury | Inhibits pro-inflammatory cytokines and up-regulates total antioxidant capacity. | [197] |
Baitouweng decoction | IL-6/STAT3 | Inhibition | UC | Inhibits the release of classical inflammatory factors such as interleukin 6 and STAT3 and regulates the structure of the intestinal flora. | [198] |
Gegen Qinlian decoction | IL-6/JAK2/STAT3 | Inhibition | UC | Regulates the homeostasis of Th17/Treg cells in colon tissue and inhibits the progression of the inflammatory response. | [199] |
Huangqin decoction | PI3K/AKT | Inhibition | UC | The intact mucosal layer structure of the intestinal epithelial mucosa was maintained, reducing the production of pro-inflammatory cytokines and improving the infiltration of inflammatory cells. | [200] |
Ligularia fischeri root extracts | Bcl-2/Bax | Activation | Diarrhea disease | Inhibit apoptosis of intestinal epithelial cells and release of pro-inflammatory cytokines. | [83] |
Huang Bai Jian Pi decoction | PI3K/AKT/NF-κB | Inhibition | UC | Regulates the balance of inflammatory and anti-inflammatory factors, significantly improving diarrhoeal symptoms and relieving the systemic inflammatory response. | [201] |
Huanglian Jiedu Decoction | JAK2/STAT3, NF-κB and Nrf2 | Inhibition (JAK2/STAT3, NF-κB) Activation (Nrf2) | Acute UC | Reducing the secretion of inflammatory factors and peroxidases, promoting the proliferation of intestinal epithelial cells, and reducing the risk of intestinal fibrosis. | [202,203] |
Gegen Qinlian decoction | Notch | Activation/Inhibition | Acute/chronic UC | Promotes the proliferation, differentiation, and secretion of epithelial cells to regulate the homeostasis of the colonic mucosa through bidirectional regulation. | [204] |
Huangqin decoction | IFN-γ/JAK/ETS | Inhibition | UC | Inhibits the release of inflammatory factors and apoptosis of intestinal epithelial cells. | [205] |
Houttuynia cordata Thunb decoction | MAPK (ERK1/2) | Inhibition | Colitis | Alleviation of intestinal inflammation by increasing the expression of tight junction proteins and anti-inflammatory factors. | [206] |
Zhizhu Decoction | SIRT1/FoxO1 | Activation | Slow transit constipation | Increased expression of TJ proteins and MUC2, significantly relieving intestinal inflammation and oxidative stress levels. | [207] |
Qushi Huayu decoction | MAPK | Inhibition | Intestinal leakage | Regulates the composition of the gut microbiota and promotes the expression of TJ proteins. | [208] |
Sanhuang Xiexin decoction | TLR4-MyD88-NF-κB | Inhibition | UC | Inhibit inflammatory cytokines, reduce oxidative stress levels, and restore intestinal probiotic abundance. | [209] |
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
Gao, J.; Cao, B.; Zhao, R.; Li, H.; Xu, Q.; Wei, B. Critical Signaling Transduction Pathways and Intestinal Barrier: Implications for Pathophysiology and Therapeutics. Pharmaceuticals 2023, 16, 1216. https://doi.org/10.3390/ph16091216
Gao J, Cao B, Zhao R, Li H, Xu Q, Wei B. Critical Signaling Transduction Pathways and Intestinal Barrier: Implications for Pathophysiology and Therapeutics. Pharmaceuticals. 2023; 16(9):1216. https://doi.org/10.3390/ph16091216
Chicago/Turabian StyleGao, Jingwang, Bo Cao, Ruiyang Zhao, Hanghang Li, Qixuan Xu, and Bo Wei. 2023. "Critical Signaling Transduction Pathways and Intestinal Barrier: Implications for Pathophysiology and Therapeutics" Pharmaceuticals 16, no. 9: 1216. https://doi.org/10.3390/ph16091216
APA StyleGao, J., Cao, B., Zhao, R., Li, H., Xu, Q., & Wei, B. (2023). Critical Signaling Transduction Pathways and Intestinal Barrier: Implications for Pathophysiology and Therapeutics. Pharmaceuticals, 16(9), 1216. https://doi.org/10.3390/ph16091216