GLP-1 and GLP-2 Orchestrate Intestine Integrity, Gut Microbiota, and Immune System Crosstalk
Abstract
:1. Introduction
2. Receptors and Functions of GLP-1 and -2
3. GLP-1 and GLP-2 as Regulators of Intestine Integrity and Inflammation
3.1. Intestinal Barrier Integrity
3.2. Inflammation and Intestinal Failure
4. GLP-1 and GLP-2 Mediate Gut Microbiota–Immune System Crosstalk
4.1. Gut Microbiota and Immune System Interaction
4.2. GLP-1/GLP-2 and Microbiota Crosstalk: Interdependent Relationship
5. Conclusions
Author Contributions
Funding
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Baggio, L.L.; Drucker, D.J. Biology of incretins: GLP-1 and GIP. Gastroenterology 2007, 132, 2131–2157. [Google Scholar] [CrossRef] [PubMed]
- Drucker, D.J.; Habener, J.F.; Holst, J.J. Discovery, characterization, and clinical development of the glucagon-like peptides. J. Clin. Investig. 2017, 127, 4217–4227. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yusta, B.; Baggio, L.L.; Koehler, J.; Holland, D.; Cao, X.; Pinnell, L.J.; Johnson-Henry, K.C.; Yeung, W.; Surette, M.G.; Bang, K.W.A. GLP-1R agonists modulate enteric immune responses through the intestinal intraepithelial lymphocyte GLP-1R. Diabetes 2015, 64, 2537–2549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pyke, C.; Heller, R.S.; Kirk, R.K.; Ørskov, C.; Reedtz-Runge, S.; Kaastrup, P.; Hvelplund, A.; Bardram, L.; Calatayud, D.; Knudsen, L.B. GLP-1 receptor localization in monkey and human tissue: Novel distribution revealed with extensively validated monoclonal antibody. Endocrinology 2014, 155, 1280–1290. [Google Scholar] [CrossRef]
- Kedees, M.H.; Guz, Y.; Grigoryan, M.; Teitelman, G. Functional activity of murine intestinal mucosal cells is regulated by the glucagon-like peptide-1 receptor. Peptides 2013, 48, 36–44. [Google Scholar] [CrossRef] [PubMed]
- Grunddal, K.V.; Jensen, E.P.; Ørskov, C.; Andersen, D.B.; Windeløv, J.A.; Poulsen, S.S.; Rosenkilde, M.M.; Knudsen, L.B.; Pyke, C.; Holst, J.J. Expression Profile of the GLP-1 Receptor in the Gastrointestinal Tract and Pancreas in Adult Female Mice. Endocrinology 2022, 163, bqab216. [Google Scholar] [CrossRef]
- Egan, J.M.; Bulotta, A.; Hui, H.; Perfetti, R. GLP-1 receptor agonists are growth and differentiation factors for pancreatic islet beta cells. Diabetes/Metab. Res. Rev. 2003, 19, 115–123. [Google Scholar] [CrossRef]
- Talsania, T.; Anini, Y.; Siu, S.; Drucker, D.J.; Brubaker, P.L. Peripheral exendin-4 and peptide YY3–36 synergistically reduce food intake through different mechanisms in mice. Endocrinology 2005, 146, 3748–3756. [Google Scholar] [CrossRef] [Green Version]
- Kahles, F.; Meyer, C.; Möllmann, J.; Diebold, S.; Findeisen, H.M.; Lebherz, C.; Trautwein, C.; Koch, A.; Tacke, F.; Marx, N. GLP-1 secretion is increased by inflammatory stimuli in an IL-6–dependent manner, leading to hyperinsulinemia and blood glucose lowering. Diabetes 2014, 63, 3221–3229. [Google Scholar] [CrossRef] [Green Version]
- Ørgaard, A.; Holst, J.J. The role of somatostatin in GLP-1-induced inhibition of glucagon secretion in mice. Diabetologia 2017, 60, 1731–1739. [Google Scholar] [CrossRef]
- Horne, R.G.; Yu, Y.; Zhang, R.; Abdalqadir, N.; Rossi, L.; Surette, M.; Sherman, P.M.; Adeli, K. High fat-high fructose diet-induced changes in the gut microbiota associated with dyslipidemia in Syrian hamsters. Nutrients 2020, 12, 3557. [Google Scholar] [CrossRef]
- Tsai, C.H.; Hill, M.; Asa, S.L.; Brubaker, P.L.; Drucker, D.J. Intestinal growth-promoting properties of glucagon-like peptide-2 in mice. Am. J. Physiol. Endocrinol. Metab. 1997, 273, E77–E84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Thulesen, J.; Hartmann, B.; Hare, K.J.; Kissow, H.; Ørskov, C.; Holst, J.J.; Poulsen, S.S. Glucagon-like peptide 2 (GLP-2) accelerates the growth of colonic neoplasms in mice. Gut 2004, 53, 1145–1150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Estall, J.L.; Drucker, D.J. Glucagon-like peptide-2. Annu. Rev. Nutr. 2006, 26, 391–411. [Google Scholar] [CrossRef] [Green Version]
- Brubaker, P.L. Glucagon-like peptide-2 and the regulation of intestinal growth and function. Compr. Physiol. 2011, 8, 1185–1210. [Google Scholar]
- Chen, M.E.; Naeini, S.M.; Srikrishnaraj, A.; Drucker, D.J.; Fesler, Z.; Brubaker, P.L. Glucagon-like peptide-2 stimulates S-phase entry of intestinal Lgr5 + stem cells. Cell. Mol. Gastroenterol. Hepatol. 2022, 13, 1829–1842. [Google Scholar] [CrossRef]
- Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef]
- Durack, J.; Lynch, S.V. The gut microbiome: Relationships with disease and opportunities for therapy. J. Exp. Med. 2019, 216, 20–40. [Google Scholar] [CrossRef] [Green Version]
- Cani, P.D.; Moens de Hase, E.; Van Hul, M. Gut microbiota and host metabolism: From proof of concept to therapeutic intervention. Microorganisms 2021, 9, 1302. [Google Scholar] [CrossRef]
- 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]
- Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
- Wang, B.; Yao, M.; Lv, L.; Ling, Z.; Li, L. The human microbiota in health and disease. Engineering 2017, 3, 71–82. [Google Scholar] [CrossRef]
- Kaijian, H.; Zhuo-Xun, W.; Xuan-Yu, C.; Jing-Quan, W.; Dongya, Z.; Chuanxing, X.; Zhu, D.; Koya, J.B.; Liuya, W.; Li, J. Microbiota in health and diseases. Signal Transduct. Target. Ther. 2022, 7, 135. [Google Scholar]
- Gasaly, N.; De Vos, P.; Hermoso, M.A. Impact of bacterial metabolites on gut barrier function and host immunity: A focus on bacterial metabolism and its relevance for intestinal inflammation. Front. Immunol. 2021, 1807, 658354. [Google Scholar] [CrossRef] [PubMed]
- Asarat, M.; Vasiljevic, T.; Apostolopoulos, V.; Donkor, O. Short-chain fatty acids regulate secretion of IL-8 from human intestinal epithelial cell lines in vitro. Immunol. Investig. 2015, 44, 678–693. [Google Scholar] [CrossRef] [PubMed]
- Morgan, X.C.; Tickle, T.L.; Sokol, H.; Gevers, D.; Devaney, K.L.; Ward, D.V.; Reyes, J.A.; Shah, S.A.; LeLeiko, N.; Snapper, S.B. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012, 13, R79. [Google Scholar] [CrossRef]
- Krautkramer, K.A.; Fan, J.; Bäckhed, F. Gut microbial metabolites as multi-kingdom intermediates. Nat. Rev. Microbiol. 2021, 19, 77–94. [Google Scholar] [CrossRef]
- Pascale, A.; Marchesi, N.; Marelli, C.; Coppola, A.; Luzi, L.; Govoni, S.; Giustina, A.; Gazzaruso, C. Microbiota and metabolic diseases. Endocrine 2018, 61, 357–371. [Google Scholar] [CrossRef]
- Usami, M.; Kishimoto, K.; Ohata, A.; Miyoshi, M.; Aoyama, M.; Fueda, Y.; Kotani, J. Butyrate and trichostatin A attenuate nuclear factor κB activation and tumor necrosis factor α secretion and increase prostaglandin E2 secretion in human peripheral blood mononuclear cells. Nutr. Res. 2008, 28, 321–328. [Google Scholar] [CrossRef]
- Azmy Nabeh, O.; Ishak Attallah, M.; El-Sayed El-Gawhary, N. The pivotal relation between glucagon-like peptides, NFκB and inflammatory bowel disease. Clin. Exp. Pharmacol. Physiol. 2020, 47, 1641–1648. [Google Scholar] [CrossRef]
- Marathe, C.S.; Rayner, C.K.; Jones, K.L.; Horowitz, M. Glucagon-like peptides 1 and 2 in health and disease: A review. Peptides 2013, 44, 75–86. [Google Scholar] [CrossRef] [PubMed]
- Rouillé, Y.; Martin, S.; Steiner, D.F. Differential processing of proglucagon by the subtilisin-like prohormone convertases PC2 and PC3 to generate either glucagon or glucagon-like peptide. J. Biol. Chem. 1995, 270, 26488–26496. [Google Scholar] [CrossRef] [PubMed]
- Furuta, M.; Yano, H.; Zhou, A.; Rouillé, Y.; Holst, J.J.; Carroll, R.; Ravazzola, M.; Orci, L.; Furuta, H.; Steiner, D.F. Defective prohormone processing and altered pancreatic islet morphology in mice lacking active SPC2. Proc. Natl. Acad. Sci. USA 1997, 94, 6646–6651. [Google Scholar] [CrossRef] [Green Version]
- Damholt, A.B.; Buchan, A.M.; Holst, J.J.; Kofod, H. Proglucagon processing profile in canine L cells expressing endogenous prohormone convertase 1/3 and prohormone convertase 2. Endocrinology 1999, 140, 4800–4808. [Google Scholar] [CrossRef]
- Lafferty, R.A.; O’Harte, F.P.M.; Irwin, N.; Gault, V.A.; Flatt, P.R. Proglucagon-Derived Peptides as Therapeutics. Front. Endocrinol. 2021, 12, 689678. [Google Scholar] [CrossRef]
- Lindquist, P.; Madsen, J.S.; Bräuner-Osborne, H.; Rosenkilde, M.M.; Hauser, A.S. Mutational Landscape of the Proglucagon-Derived Peptides. Front. Endocrinol. 2021, 12, 698511. [Google Scholar] [CrossRef] [PubMed]
- Holst, J.J. Glucagon and other proglucagon-derived peptides in the pathogenesis of obesity. Front. Nutr. 2022, 9, 964406. [Google Scholar] [CrossRef] [PubMed]
- Jacobson, A.; Yang, D.; Vella, M.; Chiu, I.M. The intestinal neuro-immune axis: Crosstalk between neurons, immune cells, and microbes. Mucosal. Immunol. 2021, 14, 555–565. [Google Scholar] [CrossRef]
- Cho, Y.M.; Merchant, C.E.; Kieffer, T.J. Targeting the glucagon receptor family for diabetes and obesity therapy. Pharmacol. Ther. 2012, 135, 247–278. [Google Scholar] [CrossRef]
- Drucker, D.J.; Nauck, M.A. The incretin system: Glucagon-like peptide-1 receptor agonists and dipeptidyl peptidase-4 inhibitors in type 2 diabetes. Lancet 2006, 368, 1696–1705. [Google Scholar] [CrossRef]
- Meier, J.J. GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus. Nat. Rev. Endocrinol. 2012, 8, 728–742. [Google Scholar] [CrossRef] [PubMed]
- Xu, G.; Stoffers, D.A.; Habener, J.F.; Bonner-Weir, S. Exendin-4 stimulates both beta-cell replication and neogenesis, resulting in increased beta-cell mass and improved glucose tolerance in diabetic rats. Diabetes 1999, 48, 2270–2276. [Google Scholar] [CrossRef]
- Hellström, P.M. GLP-1: Broadening the incretin concept to involve gut motility. Regul. Pept. 2009, 156, 9–12. [Google Scholar] [CrossRef] [PubMed]
- Abbott, C.R.; Monteiro, M.; Small, C.J.; Sajedi, A.; Smith, K.L.; Parkinson, J.R.C.; Ghatei, M.A.; Bloom, S.R. The inhibitory effects of peripheral administration of peptide YY3-36 and glucagon-like peptide-1 on food intake are attenuated by ablation of the vagal-brainstem-hypothalamic pathway. Brain Res. 2005, 1044, 127–131. [Google Scholar] [CrossRef] [PubMed]
- Pi-Sunyer, X.; Astrup, A.; Fujioka, K.; Greenway, F.; Halpern, A.; Krempf, M.; Lau, D.C.W.; Le Roux, C.W.; Violante Ortiz, R.; Jensen, C.B. A randomized, controlled trial of 3.0 mg of liraglutide in weight management. N. Engl. J. Med. 2015, 373, 11–22. [Google Scholar] [CrossRef] [PubMed]
- Kelly, A.S.; Auerbach, P.; Barrientos-Perez, M.; Gies, I.; Hale, P.M.; Marcus, C.; Mastrandrea, L.D.; Prabhu, N.; Arslanian, S. A randomized, controlled trial of liraglutide for adolescents with obesity. N. Engl. J. Med. 2020, 382, 2117–2128. [Google Scholar] [CrossRef] [PubMed]
- Hsieh, J.; Longuet, C.; Baker, C.L.; Qin, B.; Federico, L.M.; Drucker, D.J.; Adeli, K. The glucagon-like peptide 1 receptor is essential for postprandial lipoprotein synthesis and secretion in hamsters and mice. Diabetologia 2010, 53, 552–561. [Google Scholar] [CrossRef]
- Xiao, C.; Dash, S.; Morgantini, C.; Patterson, B.W.; Lewis, G.F. Sitagliptin, a DPP-4 inhibitor, acutely inhibits intestinal lipoprotein particle secretion in healthy humans. Diabetes 2014, 63, 2394–2401. [Google Scholar] [CrossRef] [Green Version]
- Tremblay, A.J.; Lamarche, B.; Deacon, C.F.; Weisnagel, S.J.; Couture, P. Effect of sitagliptin therapy on postprandial lipoprotein levels in patients with type 2 diabetes. Diabetes Obes. Metab. 2011, 13, 366–373. [Google Scholar] [CrossRef]
- Xiao, C.; Bandsma, R.H.J.; Dash, S.; Szeto, L.; Lewis, G.F. Exenatide, a glucagon-like peptide-1 receptor agonist, acutely inhibits intestinal lipoprotein production in healthy humans. Arterioscler. Thromb. Vasc. Biol. 2012, 32, 1513–1519. [Google Scholar] [CrossRef] [Green Version]
- Matikainen, N.; Mänttäri, S.; Schweizer, A.; Ulvestad, A.; Mills, D.; Dunning, B.E.; Foley, J.E.; Taskinen, M.R. Vildagliptin therapy reduces postprandial intestinal triglyceride-rich lipoprotein particles in patients with type 2 diabetes. Diabetologia 2006, 49, 2049–2057. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Farr, S.; Baker, C.; Naples, M.; Taher, J.; Iqbal, J.; Hussain, M.; Adeli, K. Central nervous system regulation of intestinal lipoprotein metabolism by glucagon-like peptide-1 via a brain-gut axis. Arterioscler. Thromb. Vasc. Biol. 2015, 35, 1092–1100. [Google Scholar] [CrossRef] [PubMed]
- Varin, E.M.; Hanson, A.A.; Beaudry, J.L.; Nguyen, M.-A.; Cao, X.; Baggio, L.L.; Mulvihill, E.E.; Drucker, D.J. Hematopoietic cell–versus enterocyte-derived dipeptidyl peptidase—4 differentially regulates triglyceride excursion in mice. JCI Insight 2020, 5, e140418. [Google Scholar] [CrossRef] [PubMed]
- Richards, P.; Parker, H.E.; Adriaenssens, A.E.; Hodgson, J.M.; Cork, S.C.; Trapp, S.; Gribble, F.M.; Reimann, F. Identification and characterization of GLP-1 receptor–expressing cells using a new transgenic mouse model. Diabetes 2014, 63, 1224–1233. [Google Scholar] [CrossRef] [Green Version]
- Nahmias, A.; Stahel, P.; Tian, L.; Xiao, C.; Lewis, G.F. GLP-1 (glucagon-like peptide-1) is physiologically relevant for chylomicron secretion beyond its known pharmacological role. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 1893–1900. [Google Scholar] [CrossRef] [PubMed]
- Drucker, D.J.; Erlich, P.; Asa, S.L.; Brubaker, P.L. Induction of intestinal epithelial proliferation by glucagon-like peptide 2. Proc. Natl. Acad. Sci. USA 1996, 93, 7911–7916. [Google Scholar] [CrossRef] [Green Version]
- Buchman, A.L.; Katz, S.; Fang, J.C.; Bernstein, C.N.; Abou-Assi, S.G.; Teduglutide Study, G. Teduglutide, a novel mucosally active analog of glucagon-like peptide-2 (GLP-2) for the treatment of moderate to severe Crohn’s disease. Inflamm. Bowel Dis. 2010, 16, 962–973. [Google Scholar] [CrossRef]
- Reiner, J.; Thiery, J.; Held, J.; Berlin, P.; Skarbaliene, J.; Vollmar, B.; Jaster, R.; Eriksson, P.O.; Lamprecht, G.; Witte, M. The dual GLP-1 and GLP-2 receptor agonist dapiglutide promotes barrier function in murine short bowel. Ann. N.Y. Acad. Sci. 2022, 1514, 132–141. [Google Scholar] [CrossRef]
- Eliasson, J.; Hvistendahl, M.K.; Freund, N.; Bolognani, F.; Meyer, C.; Jeppesen, P.B. Apraglutide, a novel glucagon-like peptide-2 analog, improves fluid absorption in patients with short bowel syndrome intestinal failure: Findings from a placebo-controlled, randomized phase 2 trial. J. Parenter. Enter. Nutr. 2022, 46, 896–904. [Google Scholar] [CrossRef]
- Dash, S.; Xiao, C.; Morgantini, C.; Connelly, P.W.; Patterson, B.W.; Lewis, G.F. Glucagon-like peptide-2 regulates release of chylomicrons from the intestine. Gastroenterology 2014, 147, 1275–1284.e1274. [Google Scholar] [CrossRef] [Green Version]
- Hein, G.J.; Baker, C.; Hsieh, J.; Farr, S.; Adeli, K. GLP-1 and GLP-2 as yin and yang of intestinal lipoprotein production: Evidence for predominance of GLP-2-stimulated postprandial lipemia in normal and insulin-resistant states. Diabetes 2013, 62, 373–381. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hsieh, J.; Trajcevski, K.E.; Farr, S.L.; Baker, C.L.; Lake, E.J.; Taher, J.; Iqbal, J.; Hussain, M.M.; Adeli, K. Glucagon-Like Peptide 2 (GLP-2) Stimulates Postprandial Chylomicron Production and Postabsorptive Release of Intestinal Triglyceride Storage Pools via Induction of Nitric Oxide Signaling in Male Hamsters and Mice. Endocrinology 2015, 156, 3538–3547. [Google Scholar] [CrossRef] [PubMed]
- Hartmann, B.; Harr, M.B.; Jeppesen, P.B.; Wojdemann, M.; Deacon, C.F.; Mortensen, P.B.; Holst, J.J. In vivo and in vitro degradation of glucagon-like peptide-2 in humans. J. Clin. Endocrinol. Metab. 2000, 85, 2884–2888. [Google Scholar] [CrossRef] [PubMed]
- Drucker, D.J. Glucagon-like peptide 2. J. Clin. Endocrinol. Metab. 2001, 86, 1759–1764. [Google Scholar] [CrossRef] [PubMed]
- Yusta, B.; Matthews, D.; Koehler, J.A.; Pujadas, G.; Kaur, K.D.; Drucker, D.J. Localization of glucagon-like peptide-2 receptor expression in the mouse. Endocrinology 2019, 160, 1950–1963. [Google Scholar] [CrossRef]
- Velázquez, E.; Blázquez, E.; Ruiz-Albusac, J.M. Glucagon-like peptide-2 (GLP-2) modulates the cGMP Signalling pathway by regulating the expression of the soluble guanylyl cyclase receptor subunits in cultured rat astrocytes. Mol. Neurobiol. 2012, 46, 242–250. [Google Scholar] [CrossRef]
- Munroe, D.G.; Gupta, A.K.; Kooshesh, F.; Vyas, T.B.; Rizkalla, G.; Wang, H.; Demchyshyn, L.; Yang, Z.-J.; Kamboj, R.K.; Chen, H. Prototypic G protein-coupled receptor for the intestinotrophic factor glucagon-like peptide 2. Proc. Natl. Acad. Sci. USA 1999, 96, 1569–1573. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, W.; Chen, L.-N.; Zhou, Q.; Zhao, L.-H.; Yang, D.; Zhang, H.; Cong, Z.; Shen, D.-D.; Zhao, F.; Zhou, F. A unique hormonal recognition feature of the human glucagon-like peptide-2 receptor. Cell Res. 2019, 30, 1098–1108. [Google Scholar] [CrossRef]
- Sun, L.; Pang, Y.; Wang, X.; Wu, Q.; Liu, H.; Liu, B.; Liu, G.; Ye, M.; Kong, W.; Jiang, C. Ablation of gut microbiota alleviates obesity-induced hepatic steatosis and glucose intolerance by modulating bile acid metabolism in hamsters. Acta Pharm. Sin. B 2019, 9, 702–710. [Google Scholar] [CrossRef]
- Sun, H.; Meng, K.; Hou, L.; Shang, L.; Yan, J. GLP-2 decreases food intake in the dorsomedial hypothalamic nucleus (DMH) through Exendin (9–39) in male Sprague-Dawley (SD) rats. Physiol. Behav. 2021, 229, 113253. [Google Scholar] [CrossRef]
- Hansen, N.L.; Brønden, A.; Nexøe-Larsen, C.C.; Christensen, A.S.; Sonne, D.P.; Rehfeld, J.F.; Wever Albretchsen, N.J.; Hartmann, B.; Vilsbøll, T.; Holst, J.J.; et al. Glucagon-like peptide 2 inhibits postprandial gallbladder emptying in man: A randomized, double-blinded, crossover study. Clin. Transl. Gastroenterol. 2020, 11, e00257. [Google Scholar] [CrossRef] [PubMed]
- Wang, Y.; Guan, X. GLP-2 potentiates L-type Ca2+ channel activity associated with stimulated glucose uptake in hippocampal neurons. Am. J. Physiol. Endocrinol. Metab. 2010, 298, E156–E166. [Google Scholar] [CrossRef] [PubMed]
- Sasaki-Hamada, S.; Fujiwara, A.; Iwai, T.; Oka, J.-I. GLP-2 restores impairments in spatial working memory and hippocampal LTD via the MEK/ERK pathway in juvenile-onset diabetes rats. Behav. Brain Res. 2021, 406, 113235. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Z.; Hao, L.; Shi, M.; Yu, Z.; Shao, S.; Yuan, Y.; Zhang, Z.; Hölscher, C. Neuroprotective Effects of a GLP-2 Analogue in the MPTP Parkinson’s Disease Mouse Model. J. Park. Dis. 2021, 11, 529–543. [Google Scholar] [CrossRef] [PubMed]
- Hsieh, J.; Longuet, C.; Maida, A.; Bahrami, J.; Xu, E.; Baker, C.L.; Brubaker, P.L.; Drucker, D.J.; Adeli, K. Glucagon-like peptide-2 increases intestinal lipid absorption and chylomicron production via CD36. Gastroenterology 2009, 137, 997–1005. [Google Scholar] [CrossRef] [PubMed]
- Grande, E.M.; Raka, F.; Hoffman, S.; Adeli, K. GLP-2 Regulation of Dietary Fat Absorption and Intestinal Chylomicron Production via Neuronal Nitric Oxide Synthase (nNOS) Signaling. Diabetes 2022, 71, 1388–1399. [Google Scholar] [CrossRef]
- Stahel, P.; Xiao, C.; Davis, X.; Tso, P.; Lewis, G.F. Glucose and GLP-2 (Glucagon-Like Peptide-2) mobilize intestinal triglyceride by distinct mechanisms. Arterioscler. Thromb. Vasc. Biol. 2019, 39, 1565–1573. [Google Scholar] [CrossRef] [Green Version]
- Greenwood-Van Meerveld, B.; Johnson, A.C.; Grundy, D. Gastrointestinal Physiology and Function. Handb Exp Pharm. 2017, 239, 1–16. [Google Scholar]
- Wang, S.Z.; Yu, Y.J.; Adeli, K. Role of Gut Microbiota in Neuroendocrine Regulation of Carbohydrate and Lipid Metabolism via the Microbiota-Gut-Brain-Liver Axis. Microorganisms 2020, 8, 527. [Google Scholar] [CrossRef] [Green Version]
- Ringseis, R.; Gessner, D.K.; Eder, K. The Gut-Liver Axis in the Control of Energy Metabolism and Food Intake in Animals. Annu Rev. Anim. Biosci. 2020, 8, 295–319. [Google Scholar] [CrossRef] [Green Version]
- 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] [PubMed] [Green Version]
- 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] [PubMed] [Green Version]
- Kissow, H.; Hartmann, B.; Holst, J.J.; Viby, N.-E.; Hansen, L.r.S.; Rosenkilde, M.M.; Hare, K.J.; Poulsen, S.S. Glucagon-like peptide-1 (GLP-1) receptor agonism or DPP-4 inhibition does not accelerate neoplasia in carcinogen treated mice. Regul. Pept. 2012, 179, 91–100. [Google Scholar] [CrossRef] [PubMed]
- Kissow, H.; Hartmann, B.; Holst, J.J.; Poulsen, S.S. Glucagon-like peptide-1 as a treatment for chemotherapy-induced mucositis. Gut 2013, 62, 1724–1733. [Google Scholar] [CrossRef]
- Koehler, J.A.; Baggio, L.L.; Yusta, B.; Longuet, C.; Rowland, K.J.; Cao, X.; Holland, D.; Brubaker, P.L.; Drucker, D.J. GLP-1R agonists promote normal and neoplastic intestinal growth through mechanisms requiring Fgf7. Cell Metab. 2015, 21, 379–391. [Google Scholar] [CrossRef] [Green Version]
- Bang-Berthelsen, C.H.; Holm, T.L.; Pyke, C.; Simonsen, L.; Søkilde, R.; Pociot, F.; Heller, R.S.; Folkersen, L.; Kvist, P.H.; Jackerott, M. GLP-1 induces barrier protective expression in Brunner’s glands and regulates colonic inflammation. Inflamm. Bow. Dis. 2016, 22, 2078–2097. [Google Scholar] [CrossRef] [Green Version]
- Nozu, T.; Miyagishi, S.; Kumei, S.; Nozu, R.; Takakusaki, K.; Okumura, T. Glucagone-like peptide-1 analog, liraglutide, improves visceral sensation and gut permeability in rats. J. Gastroenterol. Hepatol. 2018, 33, 232–239. [Google Scholar] [CrossRef] [Green Version]
- Hunt, J.E.; Holst, J.J.; Jeppesen, P.B.; Kissow, H. GLP-1 and intestinal diseases. Biomedicines 2021, 9, 383. [Google Scholar] [CrossRef]
- Panaro, B.L.; Yusta, B.; Matthews, D.; Koehler, J.A.; Song, Y.; Sandoval, D.A.; Drucker, D.J. Intestine-selective reduction of Gcg expression reveals the importance of the distal gut for GLP-1 secretion. Mol. Metab. 2020, 37, 100990. [Google Scholar] [CrossRef]
- Lu, V.B.; Rievaj, J.; O’Flaherty, E.A.; Smith, C.A.; Pais, R.; Pattison, L.A.; Tolhurst, G.; Leiter, A.B.; Bulmer, D.C.; Gribble, F.M. Adenosine triphosphate is co-secreted with glucagon-like peptide-1 to modulate intestinal enterocytes and afferent neurons. Nat. Commun. 2019, 10, 1029. [Google Scholar] [CrossRef] [Green Version]
- Osinski, C.; Le Gléau, L.; Poitou, C.; de Toro-Martin, J.; Genser, L.; Fradet, M.; Soula, H.A.; Leturque, A.; Blugeon, C.; Jourdren, L. Type 2 diabetes is associated with impaired jejunal enteroendocrine GLP-1 cell lineage in human obesity. Int. J. Obes. 2021, 45, 170–183. [Google Scholar] [CrossRef] [PubMed]
- Harpain, F.; Schlager, L.; Hütterer, E.; Dawoud, C.; Kirchnawy, S.; Stift, J.; Krotka, P.; Stift, A. Teduglutide in short bowel syndrome patients: A way back to normal life? J. Parenter. Enter. Nutr. 2022, 46, 300–309. [Google Scholar] [CrossRef] [PubMed]
- Bremholm, L.; Hornum, M.; Henriksen, B.M.; Larsen, S.; Holst, J.J. Glucagon-like peptide-2 increases mesenteric blood flow in humans. Scand. J. Gastroenterol. 2009, 44, 314–319. [Google Scholar] [CrossRef] [PubMed]
- Glerup, P.; Sonne, K.; Berner-Hansen, M.; Skarbaliene, J. Short-versus long-term, gender and species differences in the intestinotrophic effects of long-acting glucagon-like peptide 2 analog. Physiol. Res. 2022, 71, 323. [Google Scholar] [CrossRef] [PubMed]
- Zhang, Q.; Liu, M.; Li, S.; Xu, Z.; Wang, J.; Wang, Y.; Fei, Z.; Huang, W.; Sun, H. Oral Bifidobacterium longum expressing GLP-2 improves nutrient assimilation and nutritional homeostasis in mice. J. Microbiol. Methods 2018, 145, 87–92. [Google Scholar] [CrossRef]
- Feng, Y.; Demehri, F.R.; Xiao, W.; Tsai, Y.-H.; Jones, J.C.; Brindley, C.D.; Threadgill, D.W.; Holst, J.J.; Hartmann, B.; Barrett, T.A. Interdependency of EGF and GLP-2 signaling in attenuating mucosal atrophy in a mouse model of parenteral nutrition. Cell. Mol. Gastroenterol. Hepatol. 2017, 3, 447–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, E.S.; Keam, S.J. Teduglutide: A review in short bowel syndrome. Drugs 2017, 77, 345–352. [Google Scholar] [CrossRef]
- Kounatidis, D.; Vallianou, N.G.; Tsilingiris, D.; Christodoulatos, G.S.; Geladari, E.; Stratigou, T.; Karampela, I.; Dalamaga, M. Therapeutic Potential of GLP-2 Analogs in Gastrointestinal Disorders: Current Knowledge, Nutritional Aspects, and Future Perspectives. Curr. Nutr. Rep. 2022. [Google Scholar] [CrossRef]
- Brandtzaeg, P.; Kiyono, H.; Pabst, R.; Russell, M.W. Terminology: Nomenclature of mucosa-associated lymphoid tissue. Mucosal Immunol. 2008, 1, 31–37. [Google Scholar] [CrossRef] [Green Version]
- Moens, E.; Veldhoen, M. Epithelial barrier biology: Good fences make good neighbours. Immunology 2011, 135, 1–8. [Google Scholar] [CrossRef]
- McGhee, J.R.; Fujihashi, K. Inside the mucosal immune system. PLoS Biol. 2012, 10, e1001397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zietek, T.; Rath, E. Inflammation meets metabolic disease: Gut feeling mediated by GLP-1. Front. Immunol. 2016, 7, 154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Arakawa, M.; Mita, T.; Azuma, K.; Ebato, C.; Goto, H.; Nomiyama, T.; Fujitani, Y.; Hirose, T.; Kawamori, R.; Watada, H. Inhibition of monocyte adhesion to endothelial cells and attenuation of atherosclerotic lesion by a glucagon-like peptide-1 receptor agonist, exendin-4. Diabetes 2010, 59, 1030–1037. [Google Scholar] [CrossRef] [PubMed]
- Al-Dwairi, A.; Alqudah, T.E.; Al-Shboul, O.; Alqudah, M.; Mustafa, A.G.; Alfaqih, M.A. Glucagon-like peptide-1 exerts anti-inflammatory effects on mouse colon smooth muscle cells through the cyclic adenosine monophosphate/nuclear factor-κB pathway in vitro. J. Inflamm. Res. 2018, 11, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, S.; Kahles, F.; Rattik, S.; Nairz, M.; McAlpine, C.S.; Anzai, A.; Selgrade, D.; Fenn, A.M.; Chan, C.T.; Mindur, J.E. Gut intraepithelial T cells calibrate metabolism and accelerate cardiovascular disease. Nature 2019, 566, 115–119. [Google Scholar] [CrossRef] [PubMed]
- Tsalamandris, S.; Antonopoulos, A.S.; Oikonomou, E.; Papamikroulis, G.-A.; Vogiatzi, G.; Papaioannou, S.; Deftereos, S.; Tousoulis, D. The role of inflammation in diabetes: Current concepts and future perspectives. Eur. Cardiol. Rev. 2018, 14, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ramesh, P.; Yeo, J.L.; Brady, E.M.; McCann, G.P. Role of inflammation in diabetic cardiomyopathy. Ther. Adv. Endocrinol. Metab. 2022, 13, 20420188221083530. [Google Scholar] [CrossRef]
- Lopez-Candales, A.; Burgos, P.M.H.n.; Hernandez-Suarez, D.F.; Harris, D. Linking chronic inflammation with cardiovascular disease: From normal aging to the metabolic syndrome. J. Nat. Sci. 2017, 3, e341. [Google Scholar]
- Lebrun, L.J.; Lenaerts, K.; Kiers, D.; de Barros, J.-P.P.; Le Guern, N.; Plesnik, J.; Thomas, C.; Bourgeois, T.; Dejong, C.H.C.; Kox, M. Enteroendocrine L cells sense LPS after gut barrier injury to enhance GLP-1 secretion. Cell Rep. 2017, 21, 1160–1168. [Google Scholar] [CrossRef] [Green Version]
- Ellingsgaard, H.; Hauselmann, I.; Schuler, B.; Habib, A.M.; Baggio, L.L.; Meier, D.T.; Eppler, E.; Bouzakri, K.; Wueest, S.; Muller, Y.D. Interleukin-6 enhances insulin secretion by increasing glucagon-like peptide-1 secretion from L cells and alpha cells. Nat. Med. 2011, 17, 1481–1489. [Google Scholar] [CrossRef] [Green Version]
- Nguyen, A.T.; Mandard, S.; Dray, C.; Deckert, V.; Valet, P.; Besnard, P.; Drucker, D.J.; Lagrost, L.; Grober, J. Lipopolysaccharides-mediated increase in glucose-stimulated insulin secretion: Involvement of the GLP-1 pathway. Diabetes 2014, 63, 471–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, Y.S.; Park, M.S.; Choung, J.S.; Kim, S.S.; Oh, H.H.; Choi, C.S.; Ha, S.Y.; Kang, Y.; Kim, Y.; Jun, H.S. Glucagon-like peptide-1 inhibits adipose tissue macrophage infiltration and inflammation in an obese mouse model of diabetes. Diabetologia 2012, 55, 2456–2468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bartz, S.; Mody, A.; Hornik, C.; Bain, J.; Muehlbauer, M.; Kiyimba, T.; Kiboneka, E.; Stevens, R.; Bartlett, J.; St Peter, J.V. Severe acute malnutrition in childhood: Hormonal and metabolic status at presentation, response to treatment, and predictors of mortality. J. Clin. Endocrinol. Metab. 2014, 99, 2128–2137. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Robinson, E.; Cassidy, R.S.; Tate, M.; Zhao, Y.; Lockhart, S.; Calderwood, D.; Church, R.; McGahon, M.K.; Brazil, D.P.; McDermott, B.J. Exendin-4 protects against post-myocardial infarction remodelling via specific actions on inflammation and the extracellular matrix. Basic Res. Cardiol. 2015, 110, 20. [Google Scholar] [CrossRef] [PubMed]
- Buldak, L.; Machnik, G.; Buldak, R.J.; Labuzek, K.; Boldys, A.; Belowski, D.; Basiak, M.; Okopień, B. Exenatide (a GLP-1 agonist) expresses anti-inflammatory properties in cultured human monocytes/macrophages in a protein kinase A and B/Akt manner. Pharmacol. Rep. 2016, 68, 329–337. [Google Scholar]
- Guo, C.; Huang, T.; Chen, A.; Chen, X.; Wang, L.; Shen, F.; Gu, X. Glucagon-like peptide 1 improves insulin resistance in vitro through anti-inflammation of macrophages. Braz. J. Med. Biol. Res. 2016, 49, e5826. [Google Scholar] [CrossRef]
- Mitchell, P.D.; Salter, B.M.; Oliveria, J.-P.; El-Gammal, A.; Tworek, D.; Smith, S.G.; Sehmi, R.; Gauvreau, G.M.; Butler, M.; O’Byrne, P.M. Glucagon-like peptide-1 receptor expression on human eosinophils and its regulation of eosinophil activation. Clin. Exp. Allergy 2017, 47, 331–338. [Google Scholar] [CrossRef]
- Steven, S.; Jurk, K.; Kopp, M.; Kröller-Schön, S.; Mikhed, Y.; Schwierczek, K.; Roohani, S.; Kashani, F.; Oelze, M.; Klein, T. Glucagon-like peptide-1 receptor signalling reduces microvascular thrombosis, nitro-oxidative stress and platelet activation in endotoxaemic mice. Br. J. Pharmacol. 2016, 174, 1620–1632. [Google Scholar] [CrossRef] [Green Version]
- Furukawa, S.; Fujita, T.; Shimabukuro, M.; Iwaki, M.; Yamada, Y.; Nakajima, Y.; Nakayama, O.; Makishima, M.; Matsuda, M.; Shimomura, I. Increased oxidative stress in obesity and its impact on metabolic syndrome. J. Clin. Investig. 2004, 114, 1752–1761. [Google Scholar] [CrossRef]
- Dandona, P.; Aljada, A.; Bandyopadhyay, A. Inflammation: The link between insulin resistance, obesity and diabetes. Trends Immunol. 2004, 25, 4–7. [Google Scholar] [CrossRef]
- Pang, J.; Feng, J.N.; Ling, W.; Jin, T. The anti-inflammatory feature of glucagon-like peptide-1 and its based diabetes drugs-Therapeutic potential exploration in lung injury. Acta Pharm. Sin. B 2022. [Google Scholar] [CrossRef]
- Viswanathan, P.; Chaudhuri, A.; Bhatia, R.; Al-Atrash, F.; Mohanty, P.; Dandona, P. Exenatide therapy in obese patients with type 2 diabetes mellitus treated with insulin. Endocr. Pract. 2007, 13, 444–450. [Google Scholar] [CrossRef] [PubMed]
- Wu, J.-D.; Xu, X.-H.; Zhu, J.; Ding, B.; Du, T.-X.; Gao, G.; Mao, X.-M.; Ye, L.; Lee, K.-O.; Ma, J.-H. Effect of exenatide on inflammatory and oxidative stress markers in patients with type 2 diabetes mellitus. Diabetes Technol. Ther. 2011, 13, 143–148. [Google Scholar] [CrossRef] [PubMed]
- Chaudhuri, A.; Ghanim, H.; Vora, M.; Sia, C.L.; Korzeniewski, K.; Dhindsa, S.; Makdissi, A.; Dandona, P. Exenatide exerts a potent antiinflammatory effect. J. Clin. Endocrinol. Metab. 2012, 97, 198–207. [Google Scholar] [CrossRef] [Green Version]
- Hogan, A.E.; Gaoatswe, G.; Lynch, L.; Corrigan, M.A.; Woods, C.; O’Connell, J.; O’Shea, D. Glucagon-like peptide 1 analogue therapy directly modulates innate immune-mediated inflammation in individuals with type 2 diabetes mellitus. Diabetologia 2012, 57, 781–784. [Google Scholar] [CrossRef]
- Zatorski, H.; Salaga, M.; Fichna, J. Role of glucagon-like peptides in inflammatory bowel diseases—current knowledge and future perspectives. Naunyn-Schmiedeberg’s Arch. Pharmacol. 2019, 392, 1321–1330. [Google Scholar] [CrossRef] [Green Version]
- Anbazhagan, A.N.; Thaqi, M.; Priyamvada, S.; Jayawardena, D.; Kumar, A.; Gujral, T.; Chatterjee, I.; Mugarza, E.; Saksena, S.; Onyuksel, H. GLP-1 nanomedicine alleviates gut inflammation. Nanomed. Nanotechnol. Biol. Med. 2017, 13, 659–665. [Google Scholar] [CrossRef] [Green Version]
- Lourie, J. A Novel Use of Liraglutide: Induction of Partial Remission in Ulcerative Colitis and Ankylosing Spondylitis. Clin. Med. Rev. Case Rep 2019, 6, 6–8. [Google Scholar]
- Robertson, M.D.; Livesey, G.; Morgan, L.M.; Hampton, S.M.; Mathers, J.C. The influence of the colon on postprandial glucagon-like peptide 1 (7-36) amide concentration in man. J. Endocrinol. 1999, 161, 25–32. [Google Scholar] [CrossRef] [Green Version]
- Keller, J.; Binnewies, U.; Rösch, M.; Juul Holst, J.; Beglinger, C.; Andresen, V.; Layer, P. Gastric emptying and disease activity in inflammatory bowel disease. Eur. J. Clin. Investig. 2015, 45, 1234–1242. [Google Scholar] [CrossRef]
- Xie, S.; Liu, B.; Fu, S.; Wang, W.; Yin, Y.; Li, N.; Chen, W.; Liu, J.; Liu, D. GLP-2 suppresses LPS-induced inflammation in macrophages by inhibiting ERK phosphorylation and NF-κB activation. Cell. Physiol. Biochem. 2014, 34, 590–602. [Google Scholar] [CrossRef] [PubMed]
- Deng, G.; Lei, Q.; Gao, X.; Zhang, Y.; Zheng, H.; Bi, J.; Wang, X. Glucagon-Like Peptide-2 Modulates Enteric Paneth Cells Immune Response and Alleviates Gut Inflammation During Intravenous Fluid Infusion in Mice With a Central Catheter. Front. Nutr. 2021, 8, 688715. [Google Scholar] [CrossRef] [PubMed]
- Wang, C.; Fan, W.; Feng, X.; Zhang, Y.; Liu, C.; Liu, Z. The roles of the glucagon-like peptide-2 and the serum TGF-β1 levels in the intestinal barrier and immune functions in rats with obstructive jaundice. Am. J. Transl. Res. 2021, 13, 10449. [Google Scholar] [PubMed]
- Martin, G.R.; Wallace, L.E.; Sigalet, D.L. Glucagon-like peptide-2 induces intestinal adaptation in parenterally fed rats with short bowel syndrome. Am. J. Physiol. Gastrointest. Liver Physiol. 2004, 286, 964–972. [Google Scholar] [CrossRef]
- Chang, Y.; Deng, Q.; Zhang, Z.; Zhao, H.; Tang, J.; Chen, X.; Liu, G.; Tian, G.; Cai, J.; Jia, G. Glucagon-like peptide 2 attenuates intestinal mucosal barrier injury through the MLCK/pMLC signaling pathway in a piglet model. J. Cell. Physiol. 2021, 236, 3015–3032. [Google Scholar] [CrossRef] [PubMed]
- Norona, J.; Apostolova, P.; Schmidt, D.; Ihlemann, R.; Reischmann, N.; Taylor, G.; Köhler, N.; de Heer, J.; Heeg, S.; Andrieux, G. Glucagon-like peptide 2 for intestinal stem cell and Paneth cell repair during graft-versus-host disease in mice and humans. Blood 2020, 136, 1442–1455. [Google Scholar] [CrossRef]
- Madnawat, H.; Welu, A.L.; Gilbert, E.J.; Taylor, D.B.; Jain, S.; Manithody, C.; Blomenkamp, K.; Jain, A.K. Mechanisms of parenteral nutrition–associated liver and gut injury. Nutr. Clin. Pract. 2020, 35, 63–71. [Google Scholar] [CrossRef] [Green Version]
- Hellerman Itzhaki, M.; Singer, P. Advances in medical nutrition therapy: Parenteral nutrition. Nutrients 2020, 12, 717. [Google Scholar] [CrossRef] [Green Version]
- Belkaid, Y.; Hand, T.W. Role of the microbiota in immunity and inflammation. Cell 2014, 157, 121–141. [Google Scholar] [CrossRef] [Green Version]
- Belkaid, Y.; Harrison, O.J. Homeostatic immunity and the microbiota. Immunity 2017, 46, 562–576. [Google Scholar] [CrossRef] [Green Version]
- Hooper, L.V.; Littman, D.R.; Macpherson, A.J. Interactions between the microbiota and the immune system. Science 2012, 336, 1268–1273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Klein, J.R. T-cell activation in the curious world of the intestinal intraepithelial lymphocyte. Immunol. Res. 2004, 30, 327–337. [Google Scholar] [CrossRef]
- Kamada, N.; Seo, S.-U.; Chen, G.Y.; Núñez, G. Role of the gut microbiota in immunity and inflammatory disease. Nat. Rev. Immunol. 2015, 13, 321–335. [Google Scholar] [CrossRef] [PubMed]
- Francino, M.P. Early development of the gut microbiota and immune health. Pathogens 2014, 3, 769–790. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Worthington, J.J. The intestinal immunoendocrine axis: Novel cross-talk between enteroendocrine cells and the immune system during infection and inflammatory disease. Biochem. Soc. Trans. 2015, 43, 727–733. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Watnick, P.I.; Jugder, B.-E. Microbial control of intestinal homeostasis via enteroendocrine cell innate immune signaling. Trends Microbiol. 2020, 28, 141–149. [Google Scholar] [CrossRef] [PubMed]
- Roed, S.N.; Wismann, P.; Underwood, C.R.; Kulahin, N.; Iversen, H.; Cappelen, K.A.; Schäffer, L.; Lehtonen, J.; Hecksher-Soerensen, J.; Secher, A. Real-time trafficking and signaling of the glucagon-like peptide-1 receptor. Mol. Cell. Endocrinol. 2014, 382, 938–949. [Google Scholar] [CrossRef]
- Clarke, G.; Stilling, R.M.; Kennedy, P.J.; Stanton, C.; Cryan, J.F.; Dinan, T.G. Minireview: Gut microbiota: The neglected endocrine organ. Mol. Endocrinol. 2014, 28, 1221–1238. [Google Scholar] [CrossRef] [Green Version]
- Hacquard, S.; Garrido-Oter, R.; González, A.; Spaepen, S.; Ackermann, G.; Lebeis, S.; McHardy, A.C.; Dangl, J.L.; Knight, R.; Ley, R. Microbiota and host nutrition across plant and animal kingdoms. Cell Host Microbe 2015, 17, 603–616. [Google Scholar] [CrossRef] [Green Version]
- Lynch, J.B.; Hsiao, E.Y. Microbiomes as sources of emergent host phenotypes. Science 2019, 365, 1405–1409. [Google Scholar] [CrossRef]
- Cani, P.D.; Van Hul, M. Mediterranean diet, gut microbiota and health: When age and calories do not add up! Gut 2020, 69, 1167–1168. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xiao, H.; Kang, S. The Role of the Gut Microbiome in Energy Balance With a Focus on the Gut-Adipose Tissue Axis. Front Genet 2020, 11, 297. [Google Scholar] [CrossRef] [PubMed]
- Turnbaugh, P.J.; Bäckhed, F.; Fulton, L.; Gordon, J.I. Diet-induced obesity is linked to marked but reversible alterations in the mouse distal gut microbiome. Cell Host Microbe 2008, 3, 213–223. [Google Scholar] [CrossRef] [Green Version]
- Cahenzli, J.; Köller, Y.; Wyss, M.; Geuking, M.B.; McCoy, K.D. Intestinal microbial diversity during early-life colonization shapes long-term IgE levels. Cell Host Microbe 2013, 14, 559–570. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bauer, H.; Horowitz, R.E.; Levenson, S.M.; Popper, H. The response of the lymphatic tissue to the microbial flora. Studies on germfree mice. Am. J. Pathol. 1963, 42, 471. [Google Scholar] [PubMed]
- Herbst, T.; Sichelstiel, A.; Schär, C.; Yadava, K.; Bürki, K.; Cahenzli, J.; McCoy, K.; Marsland, B.J.; Harris, N.L. Dysregulation of allergic airway inflammation in the absence of microbial colonization. Am. J. Respir. Crit. Care Med. 2013, 184, 198–205. [Google Scholar] [CrossRef]
- Hapfelmeier, S.; Lawson, M.A.E.; Slack, E.; Kirundi, J.K.; Stoel, M.; Heikenwalder, M.; Cahenzli, J.; Velykoredko, Y.; Balmer, M.L.; Endt, K. Reversible microbial colonization of germ-free mice reveals the dynamics of IgA immune responses. Science 2010, 328, 1705–1709. [Google Scholar] [CrossRef] [Green Version]
- Umesaki, Y.; Setoyama, H.; Matsumoto, S.; Okada, Y. Expansion of alpha beta T-cell receptor-bearing intestinal intraepithelial lymphocytes after microbial colonization in germ-free mice and its independence from thymus. Immunology 1993, 79, 32. [Google Scholar]
- Wells, J.M.; Rossi, O.; Meijerink, M.; van Baarlen, P. Epithelial crosstalk at the microbiota–mucosal interface. Proc. Natl. Acad. Sci. USA 2011, 108, 4607–4614. [Google Scholar] [CrossRef] [Green Version]
- Tan, T.G.; Sefik, E.; Geva-Zatorsky, N.; Kua, L.; Naskar, D.; Teng, F.; Pasman, L.; Ortiz-Lopez, A.; Jupp, R.; Wu, H.-J.J. Identifying species of symbiont bacteria from the human gut that, alone, can induce intestinal Th17 cells in mice. Proc. Natl. Acad. Sci. USA 2016, 113, E8141–E8150. [Google Scholar] [CrossRef] [Green Version]
- Olszak, T.; An, D.; Zeissig, S.; Vera, M.P.; Richter, J.; Franke, A.; Glickman, J.N.; Siebert, R.; Baron, R.M.; Kasper, D.L. Microbial exposure during early life has persistent effects on natural killer T cell function. Science 2012, 336, 489–493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jiao, Y.; Wu, L.; Huntington, N.D.; Zhang, X. Crosstalk between gut microbiota and innate immunity and its implication in autoimmune diseases. Front. Immunol. 2020, 11, 282. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Danne, C.; Ryzhakov, G.; Martínez-López, M.; Ilott, N.E.; Franchini, F.; Cuskin, F.; Lowe, E.C.; Bullers, S.J.; Arthur, J.S.C.; Powrie, F. A large polysaccharide produced by Helicobacter hepaticus induces an anti-inflammatory gene signature in macrophages. Cell Host Microbe 2017, 22, 733–745.e735. [Google Scholar] [CrossRef] [Green Version]
- Bansal, T.; Alaniz, R.C.; Wood, T.K.; Jayaraman, A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc. Natl. Acad. Sci. USA 2010, 107, 228–233. [Google Scholar] [CrossRef] [Green Version]
- Bevins, C.L.; Salzman, N.H. Paneth cells, antimicrobial peptides and maintenance of intestinal homeostasis. Nat. Rev. Microbiol. 2011, 9, 356–368. [Google Scholar] [CrossRef] [PubMed]
- Ehmann, D.; Wendler, J.; Koeninger, L.; Larsen, I.S.; Klag, T.; Berger, J.; Marette, A.; Schaller, M.; Stange, E.F.; Malek, N.P. Paneth cell α-defensins HD-5 and HD-6 display differential degradation into active antimicrobial fragments. Proc. Natl. Acad. Sci. USA 2019, 116, 3746–3751. [Google Scholar] [CrossRef] [Green Version]
- Zheng, Y.; Wu, Y.; Tao, L.; Chen, X.; Jones, T.J.; Wang, K.; Hu, F. Chinese Propolis Prevents Obesity and Metabolism Syndromes Induced by a High Fat Diet and Accompanied by an Altered Gut Microbiota Structure in Mice. Nutrients 2020, 12, 959. [Google Scholar] [CrossRef]
- Peterson, D.A.; McNulty, N.P.; Guruge, J.L.; Gordon, J.I. IgA response to symbiotic bacteria as a mediator of gut homeostasis. Cell Host Microbe 2007, 2, 328–339. [Google Scholar] [CrossRef] [Green Version]
- Benckert, J.; Schmolka, N.; Kreschel, C.; Zoller, M.J.; Sturm, A.; Wiedenmann, B.; Wardemann, H. The majority of intestinal IgA+ and IgG+ plasmablasts in the human gut are antigen-specific. J. Clin. Investig. 2011, 121, 1946–1955. [Google Scholar] [CrossRef] [Green Version]
- Janeway Jr, C.A.; Medzhitov, R. Innate immune recognition. Annu. Rev. Immunol. 2002, 20, 197–216. [Google Scholar] [CrossRef] [Green Version]
- Rakoff-Nahoum, S.; Paglino, J.; Eslami-Varzaneh, F.; Edberg, S.; Medzhitov, R. Recognition of commensal microflora by toll-like receptors is required for intestinal homeostasis. Cell 2004, 118, 229–241. [Google Scholar] [CrossRef] [Green Version]
- Carvalho, F.A.; Koren, O.; Goodrich, J.K.; Johansson, M.E.V.; Nalbantoglu, I.; Aitken, J.D.; Su, Y.; Chassaing, B.; Walters, W.A.; González, A. Transient inability to manage proteobacteria promotes chronic gut inflammation in TLR5-deficient mice. Cell Host Microbe 2012, 12, 139–152. [Google Scholar] [CrossRef] [Green Version]
- Vijay-Kumar, M.; Aitken, J.D.; Carvalho, F.A.; Cullender, T.C.; Mwangi, S.; Srinivasan, S.; Sitaraman, S.V.; Knight, R.; Ley, R.E.; Gewirtz, A.T. Metabolic syndrome and altered gut microbiota in mice lacking Toll-like receptor 5. Science 2010, 328, 228–231. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef] [PubMed]
- Yadav, H.; Lee, J.H.; Lloyd, J.; Walter, P.; Rane, S.G. Beneficial metabolic effects of a probiotic via butyrate-induced GLP-1 hormone secretion. J. Biol. Chem. 2013, 288, 25088–25097. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schulthess, J.; Pandey, S.; Capitani, M.; Rue-Albrecht, K.C.; Arnold, I.; Franchini, F.; Chomka, A.; Ilott, N.E.; Johnston, D.G.W.; Pires, E. The short chain fatty acid butyrate imprints an antimicrobial program in macrophages. Immunity 2019, 50, 432–445.e437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Canfora, E.E.; Jocken, J.W.; Blaak, E.E. Short-chain fatty acids in control of body weight and insulin sensitivity. Nat. Rev. Endocrinol. 2015, 11, 577–591. [Google Scholar] [CrossRef] [PubMed]
- Wu, K.; Yuan, Y.; Yu, H.; Dai, X.; Wang, S.; Sun, Z.; Wang, F.; Fei, H.; Lin, Q.; Jiang, H. The gut microbial metabolite trimethylamine N-oxide aggravates GVHD by inducing M1 macrophage polarization in mice. Blood 2020, 136, 501–515. [Google Scholar] [CrossRef]
- Walker, A.W.; Sanderson, J.D.; Churcher, C.; Parkes, G.C.; Hudspith, B.N.; Rayment, N.; Brostoff, J.; Parkhill, J.; Dougan, G.; Petrovska, L. High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease. BMC Microbiol. 2012, 11, 7. [Google Scholar] [CrossRef] [Green Version]
- Wang, Z.-K.; Yang, Y.-S.; Chen, Y.; Yuan, J.; Sun, G.; Peng, L.-H. Intestinal microbiota pathogenesis and fecal microbiota transplantation for inflammatory bowel disease. World J. Gastroenterol. WJG 2014, 20, 14805. [Google Scholar] [CrossRef]
- Vijay, A.; Valdes, A.M. Role of the gut microbiome in chronic diseases: A narrative review. Eur. J. Clin. Nutr. 2021, 76, 489–501. [Google Scholar] [CrossRef] [PubMed]
- Covasa, M.; Stephens, R.W.; Toderean, R.; Cobuz, C. Intestinal sensing by gut microbiota: Targeting gut peptides. Front. Endocrinol. 2019, 10, 82. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Psichas, A.; Sleeth, M.L.; Murphy, K.G.; Brooks, L.; Bewick, G.A.; Hanyaloglu, A.C.; Ghatei, M.A.; Bloom, S.R.; Frost, G. The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via free fatty acid receptor 2 in rodents. Int. J. Obes. (Lond.) 2015, 39, 424–429. [Google Scholar] [CrossRef] [Green Version]
- Tolhurst, G.; Heffron, H.; Lam, Y.S.; Parker, H.E.; Habib, A.M.; Diakogiannaki, E.; Cameron, J.; Grosse, J.; Reimann, F.; Gribble, F.M. Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via the G-protein-coupled receptor FFAR2. Diabetes 2012, 61, 364–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Li, Y.; Kokrashvili, Z.; Mosinger, B.; Margolskee, R.F. Gustducin couples fatty acid receptors to GLP-1 release in colon. Am. J. Physiol. Endocrinol. Metab. 2013, 304, E651–E660. [Google Scholar] [CrossRef] [Green Version]
- Christiansen, C.B.; Gabe, M.B.N.; Svendsen, B.; Dragsted, L.O.; Rosenkilde, M.M.; Holst, J.J. The impact of short-chain fatty acids on GLP-1 and PYY secretion from the isolated perfused rat colon. Am. J. Physiol. Gastrointest. Liver Physiol. 2018, 315, G53–G65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chimerel, C.; Emery, E.; Summers, D.K.; Keyser, U.; Gribble, F.M.; Reimann, F. Bacterial metabolite indole modulates incretin secretion from intestinal enteroendocrine L cells. Cell Rep. 2014, 9, 1202–1208. [Google Scholar] [CrossRef]
- Wichmann, A.; Allahyar, A.; Greiner, T.U.; Plovier, H.; Lundén, G.Ö.; Larsson, T.; Drucker, D.J.; Delzenne, N.M.; Cani, P.D.; Bäckhed, F. Microbial modulation of energy availability in the colon regulates intestinal transit. Cell Host Microbe 2013, 14, 582–590. [Google Scholar] [CrossRef] [Green Version]
- Brooks, L.; Viardot, A.; Tsakmaki, A.; Stolarczyk, E.; Howard, J.K.; Cani, P.D.; Everard, A.; Sleeth, M.L.; Psichas, A.; Anastasovskaj, J. Fermentable carbohydrate stimulates FFAR2-dependent colonic PYY cell expansion to increase satiety. Mol. Metab. 2017, 6, 48–60. [Google Scholar] [CrossRef] [Green Version]
- Zhao, L.; Chen, Y.; Xia, F.; Abudukerimu, B.; Zhang, W.; Guo, Y.; Wang, N.; Lu, Y. A glucagon-like peptide-1 receptor agonist lowers weight by modulating the structure of gut microbiota. Front. Endocrinol. 2018, 9, 233. [Google Scholar] [CrossRef] [Green Version]
- Madsen, M.S.A.; Holm, J.B.; Pallejà, A.; Wismann, P.; Fabricius, K.; Rigbolt, K.; Mikkelsen, M.; Sommer, M.; Jelsing, J.; Nielsen, H.B. Metabolic and gut microbiome changes following GLP-1 or dual GLP-1/GLP-2 receptor agonist treatment in diet-induced obese mice. Sci. Rep. 2019, 9, 15582. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Grasset, E.; Puel, A.; Charpentier, J.; Collet, X.; Christensen, J.E.; Tercé, F.o.; Burcelin, R.m. A specific gut microbiota dysbiosis of type 2 diabetic mice induces GLP-1 resistance through an enteric NO-dependent and gut-brain axis mechanism. Cell Metab. 2017, 25, 1075–1090.e1075. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- He, Y.; Wu, W.; Zheng, H.-M.; Li, P.; McDonald, D.; Sheng, H.-F.; Chen, M.-X.; Chen, Z.-H.; Ji, G.-Y.; Zheng, Z.-D.-X. Regional variation limits applications of healthy gut microbiome reference ranges and disease models. Nat. Med. 2018, 24, 1532–1535. [Google Scholar] [CrossRef]
- Wang, Z.; Saha, S.; Van Horn, S.; Thomas, E.; Traini, C.; Sathe, G.; Rajpal, D.K.; Brown, J.R. Gut microbiome differences between metformin-and liraglutide-treated T2 DM subjects. Endocrinol. Diabetes Metab. 2018, 1, e00009. [Google Scholar] [CrossRef] [Green Version]
- Shang, J.; Liu, F.; Zhang, B.; Dong, K.; Lu, M.; Jiang, R.; Xu, Y.; Diao, L.; Zhao, J.; Tang, H. Liraglutide-induced structural modulation of the gut microbiota in patients with type 2 diabetes mellitus. PeerJ 2021, 9, e11128. [Google Scholar] [CrossRef]
- Montandon, S.A.; Jornayvaz, F.o.R. Effects of antidiabetic drugs on gut microbiota composition. Genes 2017, 8, 250. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, L.; Li, P.; Tang, Z.; Yan, X.; Feng, B. Structural modulation of the gut microbiota and the relationship with body weight: Compared evaluation of liraglutide and saxagliptin treatment. Sci. Rep. 2016, 6, 33251. [Google Scholar] [CrossRef]
- Smits, M.M.; Tonneijck, L.; Muskiet, M.H.A.; Hoekstra, T.; Kramer, M.H.H.; Diamant, M.; Nieuwdorp, M.; Groen, A.K.; Cahen, D.L.; van Raalte, D.l.H. Biliary effects of liraglutide and sitagliptin, a 12-week randomized placebo-controlled trial in type 2 diabetes patients. Diabetes Obes. Metab. 2016, 18, 1217–1225. [Google Scholar] [CrossRef]
- Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.l.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2016, 110, 9066–9071. [Google Scholar] [CrossRef] [Green Version]
- Yan, M.; Song, M.-M.; Bai, R.-X.; Cheng, S.; Yan, W.-M. Effect of Roux-en-Y gastric bypass surgery on intestinal Akkermansia muciniphila. World J. Gastrointest. Surg. 2016, 8, 301. [Google Scholar] [CrossRef] [PubMed]
- Dao, M.C.; Everard, A.; Aron-Wisnewsky, J.; Sokolovska, N.; Prifti, E.; Verger, E.O.; Kayser, B.D.; Levenez, F.; Chilloux, J.; Hoyles, L. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: Relationship with gut microbiome richness and ecology. Gut 2016, 65, 426–436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tsai, C.-Y.; Lu, H.-C.; Chou, Y.-H.; Liu, P.-Y.; Chen, H.-Y.; Huang, M.-C.; Lin, C.-H.; Tsai, C.-N. Gut Microbial Signatures for Glycemic Responses of GLP-1 Receptor Agonists in Type 2 Diabetic Patients: A Pilot Study. Front. Endocrinol. 2022, 12, 814770. [Google Scholar] [CrossRef]
- De Vadder, F.; Kovatcheva-Datchary, P.; Goncalves, D.; Vinera, J.; Zitoun, C.; Duchampt, A.; Backhed, F.; Mithieux, G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell 2014, 156, 84–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wahlstrom, A.; Sayin, S.I.; Marschall, H.U.; Backhed, F. Intestinal Crosstalk between Bile Acids and Microbiota and Its Impact on Host Metabolism. Cell Metab. 2016, 24, 41–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Dalile, B.; Van Oudenhove, L.; Vervliet, B.; Verbeke, K. The role of short-chain fatty acids in microbiota-gut-brain communication. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 461–478. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed] [Green Version]
- Cani, P.D.; Possemiers, S.; Van de Wiele, T.; Guiot, Y.; Everard, A.; Rottier, O.; Geurts, L.; Naslain, D.; Neyrinck, A.; Lambert, D.M. Changes in gut microbiota control inflammation in obese mice through a mechanism involving GLP-2-driven improvement of gut permeability. Gut 2009, 58, 1091–1103. [Google Scholar] [CrossRef] [PubMed]
- Li, D.; Yang, Y.; Yin, X.; Liu, Y.; Xu, H.; Ni, Y.; Hang, P.; Niu, S.; Zhang, H.; Ding, W. Glucagon-like peptide (GLP)-2 improved colonizing bacteria and reduced severity of ulcerative colitis by enhancing the diversity and abundance of intestinal mucosa. Bioengineered 2021, 12, 5195–5209. [Google Scholar] [CrossRef]
- Wu, J.; Ren, W.; Li, L.; Luo, M.; Xu, K.; Shen, J.; Wang, J.; Chang, G.; Lu, Y.; Qi, Y. Effect of aging and glucagon-like peptide 2 on intestinal microbiota in SD rats. Aging Dis. 2018, 9, 566. [Google Scholar] [CrossRef] [Green Version]
- Simon, M.-C.; Strassburger, K.; Nowotny, B.; Kolb, H.; Nowotny, P.; Burkart, V.; Zivehe, F.; Hwang, J.-H.; Stehle, P.; Pacini, G. Intake of Lactobacillus reuteri improves incretin and insulin secretion in glucose-tolerant humans: A proof of concept. Diabetes Care 2015, 38, 1827–1834. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Abdalqadir, N.; Adeli, K. GLP-1 and GLP-2 Orchestrate Intestine Integrity, Gut Microbiota, and Immune System Crosstalk. Microorganisms 2022, 10, 2061. https://doi.org/10.3390/microorganisms10102061
Abdalqadir N, Adeli K. GLP-1 and GLP-2 Orchestrate Intestine Integrity, Gut Microbiota, and Immune System Crosstalk. Microorganisms. 2022; 10(10):2061. https://doi.org/10.3390/microorganisms10102061
Chicago/Turabian StyleAbdalqadir, Nyan, and Khosrow Adeli. 2022. "GLP-1 and GLP-2 Orchestrate Intestine Integrity, Gut Microbiota, and Immune System Crosstalk" Microorganisms 10, no. 10: 2061. https://doi.org/10.3390/microorganisms10102061
APA StyleAbdalqadir, N., & Adeli, K. (2022). GLP-1 and GLP-2 Orchestrate Intestine Integrity, Gut Microbiota, and Immune System Crosstalk. Microorganisms, 10(10), 2061. https://doi.org/10.3390/microorganisms10102061