Potential roles of enteric glial cells in Crohn's disease: A critical review
Received date: 31 Jan 2023
Revised date: 23 Jul 2023
Accepted date: 26 Jul 2023
Published date: 20 Jan 2024
Copyright
Enteric glial cells in the enteric nervous system are critical for the regulation of gastrointestinal homeostasis. Increasing evidence suggests two-way communication between enteric glial cells and both enteric neurons and immune cells. These interactions may be important in the pathogenesis of Crohn's disease (CD), a chronic relapsing disease characterized by a dysregulated immune response. Structural abnormalities in glial cells have been identified in CD. Furthermore, classical inflammatory pathways associated with CD (e.g., the nuclear factor kappa-B pathway) function in enteric glial cells. However, the specific mechanisms by which enteric glial cells contribute to CD have not been summarized in detail. In this review, we describe the possible roles of enteric glial cells in the pathogenesis of CD, including the roles of glia–immune interactions, neuronal modulation, neural plasticity, and barrier integrity. Additionally, the implications for the development of therapeutic strategies for CD based on enteric glial cell-mediated pathogenic processes are discussed.
Xinyi Mao , Jun Shen . Potential roles of enteric glial cells in Crohn's disease: A critical review[J]. Cell Proliferation, 2024 , 57(1) : e13536 . DOI: 10.1111/cpr.13536
1 |
Torres J, Mehandru S, Colombel JF, Peyrin-Biroulet L. Crohn's disease. Lancet. 2017;389(10080):1741-1755.
|
2 |
Kikut J, Drozd A, Mokrzycka M, Grzybowska-Chlebowczyk U, Ziętek M, Szczuko M. Are EPA and DHA derivatives involved in IBD remission? J Clin Med. 2022;11(9):2388.
|
3 |
Schwanke RC, Marcon R, Bento AF, Calixto JB. EPA- and DHA-derived resolvins' actions in inflammatory bowel disease. Eur J Pharmacol. 2016;785:156-164.
|
4 |
Serhan CN. Pro-resolving lipid mediators are leads for resolution physiology. Nature. 2014;510(7503):92-101.
|
5 |
Jessen KR, Mirsky R. Astrocyte-like glia in the peripheral nervous system: an immunohistochemical study of enteric glia. J Neurosci. 1983;3(11):2206-2218.
|
6 |
Rühl A. Glial cells in the gut. Neurogastroenterol Motil. 2005;17(6):777-790.
|
7 |
Bush TG, Savidge TC, Freeman TC, et al. Fulminant jejuno-ileitis following ablation of enteric glia in adult transgenic mice. Cell. 1998;93(2):189-201.
|
8 |
Cabarrocas J, Savidge TC, Liblau RS. Role of enteric glial cells in inflammatory bowel disease. Glia. 2003;41(1):81-93.
|
9 |
Verkhratsky A, Nedergaard M. Physiology of astroglia. Physiol Rev. 2018;98(1):239-389.
|
10 |
Seguella L, Gulbransen BD. Enteric glial biology, intercellular signalling and roles in gastrointestinal disease. Nat Rev Gastroenterol Hepatol. 2021;18(8):571-587.
|
11 |
Abdo H, Derkinderen P, Gomes P, et al. Enteric glial cells protect neurons from oxidative stress in part via reduced glutathione. FASEB J. 2010;24(4):1082-1094.
|
12 |
Gabella G. Fine structure of the myenteric plexus in the Guinea-pig ileum. J Anat. 1972;111(Pt 1):69-97.
|
13 |
Vergnolle N, Cirillo C. Neurons and glia in the enteric nervous system and epithelial barrier function. Physiology (Bethesda). 2018;33(4):269-280.
|
14 |
McClain JL, Fried DE, Gulbransen BD. Agonist-evoked Ca(2+) signaling in enteric glia drives neural programs that regulate intestinal motility in mice. Cell Mol Gastroenterol Hepatol. 2015;1(6):631-645.
|
15 |
Fung C, Boesmans W, Cirillo C, Foong JPP, Bornstein JC, vanden Berghe P. VPAC receptor subtypes tune purinergic neuron-to-glia communication in the murine submucosal plexus. Front Cell Neurosci. 2017;11:118.
|
16 |
Gulbransen BD, Bashashati M, Hirota SA, et al. Activation of neuronal P2X7 receptor-pannexin-1 mediates death of enteric neurons during colitis. Nat Med. 2012;18(4):600-604.
|
17 |
Geboes K, Collins S. Structural abnormalities of the nervous system in Crohn's disease and ulcerative colitis. Neurogastroenterol Motil. 1998;10(3):189-202.
|
18 |
Cornet A, Savidge TC, Cabarrocas J, et al. Enterocolitis induced by autoimmune targeting of enteric glial cells: a possible mechanism in Crohn's disease? Proc Natl Acad Sci U S A. 2001;98(23):13306-13311.
|
19 |
Atreya I, Atreya R, Neurath MF. NF-kappaB in inflammatory bowel disease. J Intern Med. 2008;263(6):591-596.
|
20 |
O'Connor PM, Lapointe TK, Beck PL, et al. Mechanisms by which inflammation may increase intestinal cancer risk in inflammatory bowel disease. Inflamm Bowel Dis. 2010;16(8):1411-1420.
|
21 |
Han YM, Koh J, Kim JW, et al. NF-kappa B activation correlates with disease phenotype in Crohn's disease. PloS One. 2017;12(7):e0182071.
|
22 |
Ibiza S, García-Cassani B, Ribeiro H, et al. Glial-cell-derived neuroregulators control type 3 innate lymphoid cells and gut defence. Nature. 2016;535(7612):440-443.
|
23 |
Grubišić V, McClain JL, Fried DE, et al. Enteric glia modulate macrophage phenotype and visceral sensitivity following inflammation. Cell Rep. 2020;32(10):108100.
|
24 |
Progatzky F, Shapiro M, Chng SH, et al. Regulation of intestinal immunity and tissue repair by enteric glia. Nature. 2021;599(7883):125-130.
|
25 |
Palmela C, Chevarin C, Xu Z, et al. Adherent-invasive Escherichia coli in inflammatory bowel disease. Gut. 2018;67(3):574-587.
|
26 |
Liu Y, van Kruiningen HJ, West AB, Cartun RW, Cortot A, Colombel JF. Immunocytochemical evidence of Listeria, Escherichia coli, and Streptococcus antigens in Crohn's disease. Gastroenterology. 1995;108(5):1396-1404.
|
27 |
Elliott TR, Hudspith BN, Wu G, et al. Quantification and characterization of mucosa-associated and intracellular Escherichia coli in inflammatory bowel disease. Inflamm Bowel Dis. 2013;19(11):2326-2338.
|
28 |
Turco F, Sarnelli G, Cirillo C, et al. Enteroglial-derived S100B protein integrates bacteria-induced toll-like receptor signalling in human enteric glial cells. Gut. 2014;63(1):105-115.
|
29 |
Esposito G, Capoccia E, Turco F, et al. Palmitoylethanolamide improves colon inflammation through an enteric glia/toll like receptor 4-dependent PPAR-α activation. Gut. 2014;63(8):1300-1312.
|
30 |
Schmidt AM, Yan SD, Yan SF, Stern DM. The multiligand receptor RAGE as a progression factor amplifying immune and inflammatory responses. J Clin Invest. 2001;108(7):949-955.
|
31 |
MacEachern SJ, Patel BA, Keenan CM, et al. Inhibiting inducible nitric oxide synthase in enteric glia restores electrogenic ion transport in mice with colitis. Gastroenterology. 2015;149(2):445-55.e3.
|
32 |
Hibbs JB, Taintor RR, Vavrin Z, Rachlin EM. Nitric oxide: a cytotoxic activated macrophage effector molecule. Biochem Biophys Res Commun. 1988;157(1):87-94.
|
33 |
Jones RJ, Jourd'heuil D, Salerno JC, Smith SME, Singer HA. iNOS regulation by calcium/calmodulin-dependent protein kinase II in vascular smooth muscle. Am J Physiol Heart Circ Physiol. 2007;292(6):H2634-H2642.
|
34 |
Brown IA, McClain JL, Watson RE, et al. Enteric glia mediate neuron death in colitis through purinergic pathways that require connexin-43 and nitric oxide. Cell Mol Gastroenterol Hepatol. 2016;2(1):77-91.
|
35 |
Grubišić V, Gulbransen BD. Enteric glia: the most alimentary of all glia. J Physiol. 2017;595(2):557-570.
|
36 |
Elia PP, Tolentino YF, Bernardazzi C, de Souza HS. The role of innate immunity receptors in the pathogenesis of inflammatory bowel disease. Mediators Inflamm. 2015;2015:936193.
|
37 |
Walev I, Reske K, Palmer M, Valeva A, Bhakdi S. Potassium-inhibited processing of IL-1 beta in human monocytes. EMBO J. 1995;14(8):1607-1614.
|
38 |
Ferrari D, Pizzirani C, Adinolfi E, et al. The P2X7 receptor: a key player in IL-1 processing and release. J Immunol. 2006;176(7):3877-3883.
|
39 |
Adinolfi E, Giuliani AL, de Marchi E, et al. The P2X7 receptor: a main player in inflammation. Biochem Pharmacol. 2018;151:234-244.
|
40 |
Pelegrin P, Surprenant A. Pannexin-1 mediates large pore formation and interleukin-1beta release by the ATP-gated P2X7 receptor. EMBO J. 2006;25(21):5071-5082.
|
41 |
Martinon F, Burns K, Tschopp J. The inflammasome: a molecular platform triggering activation of inflammatory caspases and processing of proIL-beta. Mol Cell. 2002;10(2):417-426.
|
42 |
Flamant M, Aubert P, Rolli-Derkinderen M, et al. Enteric glia protect against Shigella flexneri invasion in intestinal epithelial cells: a role for S-nitrosoglutathione. Gut. 2011;60(4):473-484.
|
43 |
Chow AK, Gulbransen BD. Potential roles of enteric glia in bridging neuroimmune communication in the gut. Am J Physiol Gastrointest Liver Physiol. 2017;312(2):G145-g52.
|
44 |
Boschetti G, Nancey S, Sardi F, Roblin X, Flourié B, Kaiserlian D. Therapy with anti-TNFα antibody enhances number and function of Foxp3(+) regulatory T cells in inflammatory bowel diseases. Inflamm Bowel Dis. 2011;17(1):160-170.
|
45 |
Bernink JH, Peters CP, Munneke M, et al. Human type 1 innate lymphoid cells accumulate in inflamed mucosal tissues. Nat Immunol. 2013;14(3):221-229.
|
46 |
Zhou L, Chu C, Teng F, et al. Innate lymphoid cells support regulatory T cells in the intestine through interleukin-2. Nature. 2019;568(7752):405-409.
|
47 |
Artis D, Spits H. The biology of innate lymphoid cells. Nature. 2015;517(7534):293-301.
|
48 |
Tang Y, Tan SA, Iqbal A, Li J, Glover SC. STAT3 genotypic variant rs744166 and increased tyrosine phosphorylation of STAT3 in IL-23 responsive innate lymphoid cells during pathogenesis of Crohn's disease. J Immunol Res. 2019;2019:9406146.
|
49 |
Monteleone G, Trapasso F, Parrello T, et al. Bioactive IL-18 expression is up-regulated in Crohn's disease. J Immunol. 1999;163(1):143-147.
|
50 |
Muñoz M, Eidenschenk C, Ota N, et al. Interleukin-22 induces interleukin-18 expression from epithelial cells during intestinal infection. Immunity. 2015;42(2):321-331.
|
51 |
Lucarini E, Seguella L, Vincenzi M, et al. Role of enteric glia as bridging element between gut inflammation and visceral pain consolidation during acute colitis in rats. Biomedicine. 2021;9(11):1671.
|
52 |
Baillie JK, Arner E, Daub C, et al. Analysis of the human monocyte-derived macrophage transcriptome and response to lipopolysaccharide provides new insights into genetic aetiology of inflammatory bowel disease. PLoS Genet. 2017;13(3):e1006641.
|
53 |
Mortha A, Chudnovskiy A, Hashimoto D, et al. Microbiota-dependent crosstalk between macrophages and ILC3 promotes intestinal homeostasis. Science. 2014;343(6178):1249288.
|
54 |
Britton GJ, Contijoch EJ, Mogno I, et al. Microbiotas from humans with inflammatory bowel disease Alter the balance of gut Th17 and RORγt(+) regulatory T cells and exacerbate colitis in mice. Immunity. 2019;50(1):212-24.e4.
|
55 |
Geboes K, Rutgeerts P, Ectors N, et al. Major histocompatibility class II expression on the small intestinal nervous system in Crohn's disease. Gastroenterology. 1992;103(2):439-447.
|
56 |
Fujino S, Andoh A, Bamba S, et al. Increased expression of interleukin 17 in inflammatory bowel disease. Gut. 2003;52(1):65-70.
|
57 |
Niessner M, Volk BA. Altered Th1/Th2 cytokine profiles in the intestinal mucosa of patients with inflammatory bowel disease as assessed by quantitative reversed transcribed polymerase chain reaction (RT-PCR). Clin Exp Immunol. 1995;101(3):428-435.
|
58 |
Chang JT. Pathophysiology of inflammatory bowel diseases. N Engl J Med. 2020;383(27):2652-2664.
|
59 |
Chow AK, Grubišić V, Gulbransen BD. Enteric glia regulate lymphocyte activation via autophagy-mediated MHC-II expression. Cell Mol Gastroenterol Hepatol. 2021;12(4):1215-1237.
|
60 |
Coccia EM, Remoli ME, Di Giacinto C, et al. Cholera toxin subunit B inhibits IL-12 and IFN-{gamma} production and signaling in experimental colitis and Crohn's disease. Gut. 2005;54(11):1558-1564.
|
61 |
Maloy KJ, Powrie F. Intestinal homeostasis and its breakdown in inflammatory bowel disease. Nature. 2011;474(7351):298-306.
|
62 |
Ochoa-Cortes F, Turco F, Linan-Rico A, et al. Enteric glial cells: a new frontier in neurogastroenterology and clinical target for inflammatory bowel diseases. Inflamm Bowel Dis. 2016;22(2):433-449.
|
63 |
Bassotti G, Villanacci V, Antonelli E, Morelli A, Salerni B. Enteric glial cells: new players in gastrointestinal motility? Lab Invest. 2007;87(7):628-632.
|
64 |
Di Virgilio F, Dal Ben D, Sarti AC, et al. The P2X7 receptor in infection and inflammation. Immunity. 2017;47(1):15-31.
|
65 |
de Gassart A, Martinon F. Pyroptosis: Caspase-11 unlocks the gates of death. Immunity. 2015;43(5):835-837.
|
66 |
McClain J, Grubišić V, Fried D, et al. Ca2+ responses in enteric glia are mediated by connexin-43 hemichannels and modulate colonic transit in mice. Gastroenterology. 2014;146(2):497-507.e1.
|
67 |
Delvalle NM, Fried DE, Rivera-Lopez G, Gaudette L, Gulbransen BD. Cholinergic activation of enteric glia is a physiological mechanism that contributes to the regulation of gastrointestinal motility. Am J Physiol Gastrointest Liver Physiol. 2018;315(4):G473-g83.
|
68 |
Green CL, Ho W, Sharkey KA, McKay DM. Dextran sodium sulfate-induced colitis reveals nicotinic modulation of ion transport via iNOS-derived NO. Am J Physiol Gastrointest Liver Physiol. 2004;287(3):G706-G714.
|
69 |
Zhang N, Weber A, Li B, et al. An inducible nitric oxide synthase-luciferase reporter system for in vivo testing of anti-inflammatory compounds in transgenic mice. J Immunol. 2003;170(12):6307-6319.
|
70 |
Auteri M, Zizzo MG, Serio R. GABA and GABA receptors in the gastrointestinal tract: from motility to inflammation. Pharmacol Res. 2015;93:11-21.
|
71 |
Fletcher EL, Clark MJ, Furness JB. Neuronal and glial localization of GABA transporter immunoreactivity in the myenteric plexus. Cell Tissue Res. 2002;308(3):339-346.
|
72 |
Fried DE, Watson RE, Robson SC, Gulbransen BD. Ammonia modifies enteric neuromuscular transmission through glial γ-aminobutyric acid signaling. Am J Physiol Gastrointest Liver Physiol. 2017;313(6):G570-G580.
|
73 |
von Boyen GB, Schulte N, Pflüger C, et al. Distribution of enteric glia and GDNF during gut inflammation. BMC Gastroenterol. 2011;11:3.
|
74 |
Sokol H, Polin V, Lavergne-Slove A, et al. Plexitis as a predictive factor of early postoperative clinical recurrence in Crohn's disease. Gut. 2009;58(9):1218-1225.
|
75 |
Delvalle NM, Dharshika C, Morales-Soto W, Fried DE, Gaudette L, Gulbransen BD. Communication between enteric neurons, glia, and nociceptors underlies the effects of tachykinins on neuroinflammation. Cell Mol Gastroenterol Hepatol. 2018;6(3):321-344.
|
76 |
Hirano T, Hirayama D, Wagatsuma K, Yamakawa T, Yokoyama Y, Nakase H. Immunological mechanisms in inflammation-associated colon carcinogenesis. Int J Mol Sci. 2020;21(9):3062.
|
77 |
Prado GN, Taylor L, Zhou X, Ricupero D, Mierke DF, Polgar P. Mechanisms regulating the expression, self-maintenance, and signaling-function of the bradykinin B2 and B1 receptors. J Cell Physiol. 2002;193(3):275-286.
|
78 |
Vezzani A, Viviani B. Neuromodulatory properties of inflammatory cytokines and their impact on neuronal excitability. Neuropharmacology. 2015;96:70-82.
|
79 |
Murakami M, Ohta T, Ito S. Interleukin-1beta enhances the action of bradykinin in rat myenteric neurons through up-regulation of glial B1 receptor expression. Neuroscience. 2008;151(1):222-231.
|
80 |
Murakami M, Ohta T, Otsuguro K, et al. Involvement of prostaglandin E(2) derived from enteric glial cells in the action of bradykinin in cultured rat myenteric neurons. Neuroscience. 2007;145(2):642-653.
|
81 |
Gilks WP, Abou-Sleiman PM, Gandhi S, et al. A common LRRK2 mutation in idiopathic Parkinson's disease. Lancet. 2005;365(9457):415-416.
|
82 |
Hui KY, Fernandez-Hernandez H, Hu J, et al. Functional variants in the LRRK2 gene confer shared effects on risk for Crohn's disease and Parkinson's disease. Sci Transl Med. 2018;10(423):eaai7795.
|
83 |
Gardet A, Benita Y, Li C, et al. LRRK2 is involved in the IFN-gamma response and host response to pathogens. J Immunol. 2010;185(9):5577-5585.
|
84 |
de Guilhem De Lataillade A, Caillaud M, Oullier T, et al. LRRK2 expression in normal and pathologic human gut and in rodent enteric neural cell lines. J Neurochem. 2022;164:193-209.
|
85 |
Munoz L, Kavanagh ME, Phoa AF, et al. Optimisation of LRRK2 inhibitors and assessment of functional efficacy in cell-based models of neuroinflammation. Eur J Med Chem. 2015;95:29-34.
|
86 |
Streubel-Gallasch L, Giusti V, Sandre M, et al. Parkinson's disease-associated LRRK2 interferes with astrocyte-mediated alpha-synuclein clearance. Mol Neurobiol. 2021;58(7):3119-3140.
|
87 |
Dugan LL, Kim JS, Zhang Y, et al. Differential effects of cAMP in neurons and astrocytes. Role of B-raf. J Biol Chem. 1999;274(36):25842-25848.
|
88 |
Savidge TC, Newman P, Pothoulakis C, et al. Enteric glia regulate intestinal barrier function and inflammation via release of S-nitrosoglutathione. Gastroenterology. 2007;132(4):1344-1358.
|
89 |
Yu YB, Li YQ. Enteric glial cells and their role in the intestinal epithelial barrier. World J Gastroenterol. 2014;20(32):11273-11280.
|
90 |
Bauman BD, Meng J, Zhang L, et al. Enteric glial-mediated enhancement of intestinal barrier integrity is compromised by morphine. J Surg Res. 2017;219:214-221.
|
91 |
Pochard C, Coquenlorge S, Jaulin J, et al. Defects in 15-HETE production and control of epithelial permeability by human enteric glial cells from patients with Crohn's disease. Gastroenterology. 2016;150(1):168-180.
|
92 |
Meir M, Flemming S, Burkard N, et al. Glial cell line-derived neurotrophic factor promotes barrier maturation and wound healing in intestinal epithelial cells in vitro. Am J Physiol Gastrointest Liver Physiol. 2015;309(8):G613-G624.
|
93 |
Xiao W, Wang W, Chen W, et al. GDNF is involved in the barrier-inducing effect of enteric glial cells on intestinal epithelial cells under acute ischemia reperfusion stimulation. Mol Neurobiol. 2014;50(2):274-289.
|
94 |
Zhang DK, He FQ, Li TK, et al. Glial-derived neurotrophic factor regulates intestinal epithelial barrier function and inflammation and is therapeutic for murine colitis. J Pathol. 2010;222(2):213-222.
|
95 |
von Boyen GB, Steinkamp M, Geerling I, et al. Proinflammatory cytokines induce neurotrophic factor expression in enteric glia: a key to the regulation of epithelial apoptosis in Crohn's disease. Inflamm Bowel Dis. 2006;12(5):346-354.
|
96 |
Steinkamp M, Geerling I, Seufferlein T, et al. Glial-derived neurotrophic factor regulates apoptosis in colonic epithelial cells. Gastroenterology. 2003;124(7):1748-1757.
|
97 |
Chen H, Han T, Gao L, Zhang DK. The involvement of glial cell-derived neurotrophic factor in inflammatory bowel disease. J Interferon Cytokine Res. 2022;42(1):1-7.
|
98 |
Meir M, Kannapin F, Diefenbacher M, et al. Intestinal epithelial barrier maturation by enteric glial cells is GDNF-dependent. Int J Mol Sci. 2021;22(4):1887.
|
99 |
Patel A, Harker N, Moreira-Santos L, et al. Differential RET signaling pathways drive development of the enteric lymphoid and nervous systems. Sci Signal. 2012;5(235):ra55.
|
100 |
Sonnenberg GF, Fouser LA, Artis D. Border patrol: regulation of immunity, inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat Immunol. 2011;12(5):383-390.
|
101 |
Meir M, Burkard N, Ungewiß H, et al. Neurotrophic factor GDNF regulates intestinal barrier function in inflammatory bowel disease. J Clin Invest. 2019;129(7):2824-2840.
|
102 |
Brun P, Gobbo S, Caputi V, et al. Toll like receptor-2 regulates production of glial-derived neurotrophic factors in murine intestinal smooth muscle cells. Mol Cell Neurosci. 2015;68:24-35.
|
103 |
Young HM, Hearn CJ, Farlie PG, Canty AJ, Thomas PQ, Newgreen DF. GDNF is a chemoattractant for enteric neural cells. Dev Biol. 2001;229(2):503-516.
|
104 |
Cintrón-Colón AF, Almeida-Alves G, Boynton AM, Spitsbergen JM. GDNF synthesis, signaling, and retrograde transport in motor neurons. Cell Tissue Res. 2020;382(1):47-56.
|
105 |
Li Z, Zhang X, Zhou H, Liu W, Li J. Exogenous S-nitrosoglutathione attenuates inflammatory response and intestinal epithelial barrier injury in endotoxemic rats. J Trauma Acute Care Surg. 2016;80(6):977-984.
|
106 |
Fanning AS, Ma TY, Anderson JM. Isolation and functional characterization of the actin binding region in the tight junction protein ZO-1. FASEB J. 2002;16(13):1835-1837.
|
107 |
Grubišić V, Gulbransen BD. Enteric glial activity regulates secretomotor function in the mouse colon but does not acutely affect gut permeability. J Physiol. 2017;595(11):3409-3424.
|
108 |
Kabouridis PS, Lasrado R, McCallum S, et al. Microbiota controls the homeostasis of glial cells in the gut lamina propria. Neuron. 2015;85(2):289-295.
|
109 |
Neves AR, Castelo-Branco MT, Figliuolo VR, et al. Overexpression of ATP-activated P2X7 receptors in the intestinal mucosa is implicated in the pathogenesis of Crohn's disease. Inflamm Bowel Dis. 2014;20(3):444-457.
|
110 |
Eser A, Colombel JF, Rutgeerts P, et al. Safety and efficacy of an oral inhibitor of the purinergic receptor P2X7 in adult patients with moderately to severely active Crohn's disease: a randomized placebo-controlled, double-blind, Phase IIa Study. Inflamm Bowel Dis. 2015;21(10):2247-2253.
|
111 |
Ali Z, Laurijssens B, Ostenfeld T, et al. Pharmacokinetic and pharmacodynamic profiling of a P2X7 receptor allosteric modulator GSK1482160 in healthy human subjects. Br J Clin Pharmacol. 2013;75(1):197-207.
|
112 |
Hofman P, Cherfils-Vicini J, Bazin M, et al. Genetic and pharmacological inactivation of the purinergic P2RX7 receptor dampens inflammation but increases tumor incidence in a mouse model of colitis-associated cancer. Cancer Res. 2015;75(5):835-845.
|
113 |
Burnstock G, Jacobson KA, Christofi FL. Purinergic drug targets for gastrointestinal disorders. Curr Opin Pharmacol. 2017;37:131-141.
|
114 |
Lara R, Adinolfi E, Harwood CA, et al. P2X7 in cancer: from molecular mechanisms to therapeutics. Front Pharmacol. 2020;11:793.
|
115 |
Diezmos EF, Markus I, Perera DS, et al. Blockade of Pannexin-1 channels and purinergic P2X7 receptors shows protective effects against cytokines-induced colitis of human colonic mucosa. Front Pharmacol. 2018;9:865.
|
116 |
Good ME, Chiu YH, Poon IKH, et al. Pannexin 1 channels as an unexpected new target of the anti-hypertensive drug spironolactone. Circ Res. 2018;122(4):606-615.
|
117 |
Zhang J, Wang XJ, Wu LJ, et al. Herb-partitioned moxibustion alleviates colonic inflammation in Crohn's disease rats by inhibiting hyperactivation of the NLRP3 inflammasome via regulation of the P2X7R-Pannexin-1 signaling pathway. PLoS One. 2021;16(5):e0252334.
|
118 |
Di Cesare ML, Marcoli M, Micheli L, et al. Oxaliplatin evokes P2X7-dependent glutamate release in the cerebral cortex: a pain mechanism mediated by pannexin 1. Neuropharmacology. 2015;97:133-141.
|
119 |
Bravo D, Maturana CJ, Pelissier T, Hernández A, Constandil L. Interactions of pannexin 1 with NMDA and P2X7 receptors in central nervous system pathologies: possible role on chronic pain. Pharmacol Res. 2015;101:86-93.
|
120 |
Watkins LR, Hutchinson MR, Ledeboer A, Wieseler-Frank J, Milligan ED, Maier SF. Norman cousins lecture. Glia as the "bad guys": implications for improving clinical pain control and the clinical utility of opioids. Brain Behav Immun. 2007;21(2):131-146.
|
121 |
Liu SL, Li YH, Shi GY, et al. A novel inhibitory effect of naloxone on macrophage activation and atherosclerosis formation in mice. J Am Coll Cardiol. 2006;48(9):1871-1879.
|
122 |
Younger J, Parkitny L, McLain D. The use of low-dose naltrexone (LDN) as a novel anti-inflammatory treatment for chronic pain. Clin Rheumatol. 2014;33(4):451-459.
|
123 |
Smith JP, Bingaman SI, Ruggiero F, et al. Therapy with the opioid antagonist naltrexone promotes mucosal healing in active Crohn's disease: a randomized placebo-controlled trial. Dig Dis Sci. 2011;56(7):2088-2097.
|
124 |
Smith JP, Stock H, Bingaman S, Mauger D, Rogosnitzky M, Zagon IS. Low-dose naltrexone therapy improves active Crohn's disease. Am J Gastroenterol. 2007;102(4):820-828.
|
125 |
Huang YF, Li QP, Dou YX, et al. Therapeutic effect of Brucea javanica oil emulsion on experimental Crohn's disease in rats: involvement of TLR4/ NF-κB signaling pathway. Biomed Pharmacother. 2019;114:108766.
|
126 |
Costa DVS, Bon-Frauches AC, Silva A, et al. 5-fluorouracil induces enteric neuron death and glial activation during intestinal mucositis via a S100B-RAGE-NFκB-dependent pathway. Sci Rep. 2019;9(1):665.
|
127 |
Morales-Soto W, Gulbransen BD. Enteric glia: a new player in abdominal pain. Cell Mol Gastroenterol Hepatol. 2019;7(2):433-445.
|
128 |
Fox AD, Kripke SA, De Paula J, et al. Effect of a glutamine-supplemented enteral diet on methotrexate-induced enterocolitis. JPEN J Parenter Enteral Nutr. 1988;12(4):325-331.
|
129 |
Coëffier M, Marion R, Ducrotté P, Déchelotte P. Modulating effect of glutamine on IL-1beta-induced cytokine production by human gut. Clin Nutr. 2003;22(4):407-413.
|
130 |
Akobeng AK, Elawad M, Gordon M. Glutamine for induction of remission in Crohn's disease. Cochrane Database Syst Rev. 2016;2:Cd007348.
|
131 |
Severo JS, da Silva Barros VJ, Alves da Silva AC, et al. Effects of glutamine supplementation on inflammatory bowel disease: a systematic review of clinical trials. Clin Nutr ESPEN. 2021;42:53-60.
|
132 |
Szatkowski P, Krzysciak W, Mach T, Owczarek D, Brzozowski B, Szczeklik K. Nuclear factor-κB: importance, induction of inflammation, and effects of pharmacological modulators in Crohn's disease. J Physiol Pharmacol. 2020;71(4).
|
133 |
Cheng WX, Ren Y, Lu MM, et al. Palmitoylation in Crohn's disease: current status and future directions. World J Gastroenterol. 2021;27(48):8201-8215.
|
134 |
Hanauer SB, Sandborn WJ, Rutgeerts P, et al. Human anti-tumor necrosis factor monoclonal antibody (adalimumab) in Crohn's disease: the CLASSIC-I trial. Gastroenterology. 2006;130(2):323-333.
|
135 |
Jilani NZ, Akobeng AK. Adalimumab for maintenance of clinical response and remission in patients with Crohn's disease: the CHARM trial. Colombel JF, Sandborn WJ, Rutgeerts P et al. Gastroenterology 2007;132:52-65. J Pediatr Gastroenterol Nutr. 2008;46(2):226-227.
|
136 |
Sandborn WJ, Feagan BG, Stoinov S, et al. Certolizumab pegol for the treatment of Crohn's disease. N Engl J Med. 2007;357(3):228-238.
|
137 |
Cushing K, Higgins PDR. Management of Crohn disease: a review. Jama. 2021;325(1):69-80.
|
138 |
Feagan BG, Sandborn WJ, Gasink C, et al. Ustekinumab as induction and maintenance therapy for Crohn's disease. N Engl J Med. 2016;375(20):1946-1960.
|
139 |
Fang L, Pang Z, Shu W, et al. Anti-TNF therapy induces CD4+ T-cell production of IL-22 and promotes epithelial repairs in patients with Crohn's disease. Inflamm Bowel Dis. 2018;24(8):1733-1744.
|
140 |
Valès S, Bacola G, Biraud M, et al. Tumor cells hijack enteric glia to activate colon cancer stem cells and stimulate tumorigenesis. EBioMedicine. 2019;49:172-188.
|
141 |
Selgrad M, De Giorgio R, Fini L, et al. JC virus infects the enteric glia of patients with chronic idiopathic intestinal pseudo-obstruction. Gut. 2009;58(1):25-32.
|
142 |
Seelig DM, Mason GL, Telling GC, Hoover EA. Chronic wasting disease prion trafficking via the autonomic nervous system. Am J Pathol. 2011;179(3):1319-1328.
|
143 |
Liu C, Yang J. Enteric glial cells in immunological disorders of the gut. Front Cell Neurosci. 2022;16:895871.
|
144 |
Zoumboulakis D, Cirella KR, Gougeon PY, Lourenssen SR, Blennerhassett MG. MMP-9 processing of intestinal smooth muscle-derived GDNF is required for neurotrophic action on enteric neurons. Neuroscience. 2020;443:8-18.
|
145 |
Brun P, Giron MC, Qesari M, et al. Toll-like receptor 2 regulates intestinal inflammation by controlling integrity of the enteric nervous system. Gastroenterology. 2013;145(6):1323-1333.
|
146 |
Matteoli G. Enteric glial cells: new players in mucosal defence against bacteria? Gut. 2011;60(4):429-430.
|
147 |
Kinchen J, Chen HH, Parikh K, et al. Structural remodeling of the human colonic mesenchyme in inflammatory bowel disease. Cell. 2018;175(2):372-86.e17.
|
148 |
Smillie CS, Biton M, Ordovas-Montanes J, et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell. 2019;178(3):714-30.e22.
|
149 |
Liñán-Rico A, Turco F, Ochoa-Cortes F, et al. Molecular signaling and dysfunction of the human reactive enteric glial cell phenotype: implications for GI infection, IBD, POI, neurological, motility, and GI disorders. Inflamm Bowel Dis. 2016;22(8):1812-1834.
|
/
〈 | 〉 |