Microenvironmental Metabolites in the Intestine: Messengers between Health and Disease
Abstract
:1. Introduction
1.1. The Intestinal Epithelium Functions as a Barrier and Nutrient-Absorbing Organ
1.2. Principal Concepts of Intestinal Homeostasis and Metabolism
2. Metabolism and Intestinal Diseases
2.1. Metabolites Affecting Intestinal Barrier Functions
2.2. Metabolism and Colorectal Cancer Tumorigenesis
2.3. Oncogenic Signaling and Metabolic Pathways Involved in CRC Tumorigenesis
2.4. Oncometabolites: Metabolic Determinants of CRC Initiation and Progression
2.5. 2-Hydroxyglutarate (2-HG) and Kynurenine (Kyn): Oncometabolites in CRC
2.6. Microbial Metabolites, Immune System, and Host Metabolism: A Tripartite Interplay That Shapes CRC
2.7. Short-Chain Fatty Acids (SCFAs) in CRC: Friends or Foes?
2.8. Polyamines and Bile Acids: Oncometabolites Involved in CRC
3. Microbial Metabolites Modulate the Immune Microenvironment and the Efficacy of Anti-Tumor Therapies
4. Conclusions
Author Contributions
Funding
Conflicts of Interest
References
- 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]
- Bonis, V.; Rossell, C.; Gehart, H. The Intestinal Epithelium-Fluid Fate and Rigid Structure from Crypt Bottom to Villus Tip. Front. Cell Dev. Biol. 2021, 9, 661931. [Google Scholar] [CrossRef]
- Spence, J.R.; Lauf, R.; Shroyer, N.F. Vertebrate intestinal endoderm development. Dev. Dyn. 2011, 240, 501–520. [Google Scholar] [CrossRef] [Green Version]
- Barker, N. Adult intestinal stem cells: Critical drivers of epithelial homeostasis and regeneration. Nat. Rev. Mol. Cell Biol. 2014, 15, 19–33. [Google Scholar] [CrossRef] [Green Version]
- Barker, N.; van Es, J.H.; Kuipers, J.; Kujala, P.; van den Born, M.; Cozijnsen, M.; Haegebarth, A.; Korving, J.; Begthel, H.; Peters, P.J.; et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 2007, 449, 1003–1007. [Google Scholar] [CrossRef]
- Heppert, J.K.; Davison, J.M.; Kelly, C.; Mercado, G.P.; Lickwar, C.R.; Rawls, J.F. Transcriptional programmes underlying cellular identity and microbial responsiveness in the intestinal epithelium. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 7–23. [Google Scholar] [CrossRef]
- Noah, T.K.; Donahue, B.; Shroyer, N.F. Intestinal development and differentiation. Exp. Cell Res. 2011, 317, 2702–2710. [Google Scholar] [CrossRef] [Green Version]
- van der Schoor, S.R.D.; Reeds, P.J.; Stoll, B.; Henry, J.F.; Rosenberger, J.R.; Burrin, D.G.; van Goudoever, J.B. The high metabolic cost of a functional gut. Gastroenterology 2002, 123, 1931–1940. [Google Scholar] [CrossRef]
- Liu, Q.; Yu, Z.; Tian, F.; Zhao, J.; Zhang, H.; Zhai, Q.; Chen, W. Surface components and metabolites of probiotics for regulation of intestinal epithelial barrier. Microb. Cell Fact. 2020, 19, 23. [Google Scholar] [CrossRef]
- van Nuenen, M.H.M.C.; de Ligt, R.A.F.; Doornbos, R.P.; van der Woude, J.C.J.; Kuipers, E.J.; Venema, K. The influence of microbial metabolites on human intestinal epithelial cells and macrophages in vitro. FEMS Immunol. Med. Microbiol. 2005, 45, 183–189. [Google Scholar] [CrossRef]
- Vanuytsel, T.; Senger, S.; Fasano, A.; Shea-Donohue, T. Major signaling pathways in intestinal stem cells. Biochim. Biophys. Acta 2013, 1830, 2410–2426. [Google Scholar] [CrossRef] [Green Version]
- Yousefi, M.; Nakauka-Ddamba, A.; Berry, C.T.; Li, N.; Schoenberger, J.; Simeonov, K.P.; Cedeno, R.J.; Yu, Z.; Lengner, C.J. Calorie Restriction Governs Intestinal Epithelial Regeneration through Cell-Autonomous Regulation of mTORC1 in Reserve Stem Cells. Stem Cell Rep. 2018, 10, 703–711. [Google Scholar] [CrossRef] [Green Version]
- Mihaylova, M.M.; Cheng, C.W.; Cao, A.Q.; Tripathi, S.; Mana, M.D.; Bauer-Rowe, K.E.; Abu-Remaileh, M.; Clavain, L.; Erdemir, A.; Lewis, C.A.; et al. Fasting Activates Fatty Acid Oxidation to Enhance Intestinal Stem Cell Function during Homeostasis and Aging. Cell Stem Cell 2018, 22, 769–778.e764. [Google Scholar] [CrossRef] [Green Version]
- Beyaz, S.; Mana, M.D.; Roper, J.; Kedrin, D.; Saadatpour, A.; Hong, S.J.; Bauer-Rowe, K.E.; Xifaras, M.E.; Akkad, A.; Arias, E.; et al. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature 2016, 531, 53–58. [Google Scholar] [CrossRef]
- Mana, M.D.; Hussey, A.M.; Tzouanas, C.N.; Imada, S.; Barrera Millan, Y.; Bahceci, D.; Saiz, D.R.; Webb, A.T.; Lewis, C.A.; Carmeliet, P.; et al. High-fat diet-activated fatty acid oxidation mediates intestinal stemness and tumorigenicity. Cell Rep. 2021, 35, 109212. [Google Scholar] [CrossRef]
- Xie, Y.; Ding, F.; Di, W.; Lv, Y.; Xia, F.; Sheng, Y.; Yu, J.; Ding, G. Impact of a high-fat diet on intestinal stem cells and epithelial barrier function in middle-aged female mice. Mol. Med. Rep. 2020, 21, 1133–1144. [Google Scholar] [CrossRef] [Green Version]
- Xie, R.; Sun, Y.; Wu, J.; Huang, S.; Jin, G.; Guo, Z.; Zhang, Y.; Liu, T.; Liu, X.; Cao, X.; et al. Maternal High Fat Diet Alters Gut Microbiota of Offspring and Exacerbates DSS-Induced Colitis in Adulthood. Front. Immunol. 2018, 9, 2608. [Google Scholar] [CrossRef]
- Aliluev, A.; Tritschler, S.; Sterr, M.; Oppenlander, L.; Hinterdobler, J.; Greisle, T.; Irmler, M.; Beckers, J.; Sun, N.; Walch, A.; et al. Diet-induced alteration of intestinal stem cell function underlies obesity and prediabetes in mice. Nat. Metab. 2021, 3, 1202–1216. [Google Scholar] [CrossRef]
- Rodríguez-Colman, M.J.; Schewe, M.; Meerlo, M.; Stigter, E.; Gerrits, J.; Pras-Raves, M.; Sacchetti, A.; Hornsveld, M.; Oost, K.C.; Snippert, H.J.; et al. Interplay between metabolic identities in the intestinal crypt supports stem cell function. Nature 2017, 543, 424–427. [Google Scholar] [CrossRef]
- Rath, E.; Berger, E.; Messlik, A.; Nunes, T.; Liu, B.; Kim, S.C.; Hoogenraad, N.; Sans, M.; Sartor, R.B.; Haller, D. Induction of dsRNA-activated protein kinase links mitochondrial unfolded protein response to the pathogenesis of intestinal inflammation. Gut 2012, 61, 1269–1278. [Google Scholar] [CrossRef]
- Khaloian, S.; Rath, E.; Hammoudi, N.; Gleisinger, E.; Blutke, A.; Giesbertz, P.; Berger, E.; Metwaly, A.; Waldschmitt, N.; Allez, M.; et al. Mitochondrial impairment drives intestinal stem cell transition into dysfunctional Paneth cells predicting Crohn’s disease recurrence. Gut 2020, 69, 1939–1951. [Google Scholar] [CrossRef]
- 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] [Green Version]
- Ghosh, S.; Whitley, C.S.; Haribabu, B.; Jala, V.R. Regulation of Intestinal Barrier Function by Microbial Metabolites. Cell. Mol. Gastroenterol. Hepatol. 2021, 11, 1463–1482. [Google Scholar] [CrossRef]
- Konjar, Š.; Pavšič, M.; Veldhoen, M. Regulation of Oxygen Homeostasis at the Intestinal Epithelial Barrier Site. Int. J. Mol. Sci. 2021, 22, 9170. [Google Scholar] [CrossRef]
- Manresa, M.C.; Taylor, C.T. Hypoxia Inducible Factor (HIF) Hydroxylases as Regulators of Intestinal Epithelial Barrier Function. Cell. Mol. Gastroenterol. Hepatol. 2017, 3, 303–315. [Google Scholar] [CrossRef] [Green Version]
- Glover, L.E.; Colgan, S.P. Epithelial Barrier Regulation by Hypoxia-Inducible Factor. Ann. Am. Thorac. Soc. 2017, 14, S233–S236. [Google Scholar] [CrossRef]
- Louis, N.A.; Hamilton, K.E.; Canny, G.; Shekels, L.L.; Ho, S.B.; Colgan, S.P. Selective induction of mucin-3 by hypoxia in intestinal epithelia. J. Cell. Biochem. 2006, 99, 1616–1627. [Google Scholar] [CrossRef] [PubMed]
- Sonnenburg, J.L.; Xu, J.; Leip, D.D.; Chen, C.H.; Westover, B.P.; Weatherford, J.; Buhler, J.D.; Gordon, J.I. Glycan foraging in vivo by an intestine-adapted bacterial symbiont. Science 2005, 307, 1955–1959. [Google Scholar] [CrossRef] [Green Version]
- Tailford, L.E.; Crost, E.H.; Kavanaugh, D.; Juge, N. Mucin glycan foraging in the human gut microbiome. Front. Genet. 2015, 6, 81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Camilleri, M.; Lyle, B.J.; Madsen, K.L.; Sonnenburg, J.; Verbeke, K.; Wu, G.D. Role for diet in normal gut barrier function: Developing guidance within the framework of food-labeling regulations. Am. J. Physiol. Gastrointest. Liver Physiol. 2019, 317, G17–G39. [Google Scholar] [CrossRef]
- Gálvez, J. Role of Th17 Cells in the Pathogenesis of Human IBD. ISRN Inflamm. 2014, 2014, 928461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hinrichsen, F.; Hamm, J.; Westermann, M.; Schröder, L.; Shima, K.; Mishra, N.; Walker, A.; Sommer, N.; Klischies, K.; Prasse, D.; et al. Microbial regulation of hexokinase 2 links mitochondrial metabolism and cell death in colitis. Cell Metab. 2021, 33, 2355–2366.e2358. [Google Scholar] [CrossRef] [PubMed]
- He, C.; Deng, J.; Hu, X.; Zhou, S.; Wu, J.; Xiao, D.; Darko, K.O.; Huang, Y.; Tao, T.; Peng, M.; et al. Vitamin A inhibits the action of LPS on the intestinal epithelial barrier function and tight junction proteins. Food Funct. 2019, 10, 1235–1242. [Google Scholar] [CrossRef] [PubMed]
- Stio, M.; Retico, L.; Annese, V.; Bonanomi, A.G. Vitamin D regulates the tight-junction protein expression in active ulcerative colitis. Scand. J. Gastroenterol. 2016, 51, 1193–1199. [Google Scholar] [CrossRef] [PubMed]
- Ooi, J.H.; Li, Y.; Rogers, C.J.; Cantorna, M.T. Vitamin D regulates the gut microbiome and protects mice from dextran sodium sulfate-induced colitis. J. Nutr. 2013, 143, 1679–1686. [Google Scholar] [CrossRef]
- de La Serre, C.B.; Ellis, C.L.; Lee, J.; Hartman, A.L.; Rutledge, J.C.; Raybould, H.E. Propensity to high-fat diet-induced obesity in rats is associated with changes in the gut microbiota and gut inflammation. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 299, G440–G448. [Google Scholar] [CrossRef]
- Keum, N.; Giovannucci, E. Global burden of colorectal cancer: Emerging trends, risk factors and prevention strategies. Nat. Rev. Gastroenterol. Hepatol. 2019, 16, 713–732. [Google Scholar] [CrossRef]
- Burnett-Hartman, A.N.; Lee, J.K.; Demb, J.; Gupta, S. An Update on the Epidemiology, Molecular Characterization, Diagnosis, and Screening Strategies for Early-Onset Colorectal Cancer. Gastroenterology 2021, 160, 1041–1049. [Google Scholar] [CrossRef] [PubMed]
- Fearon, E.R. Molecular genetics of colorectal cancer. Annu. Rev. Pathol. 2011, 6, 479–507. [Google Scholar] [CrossRef]
- Kinzler, K.W.; Vogelstein, B. Lessons from Hereditary Colorectal Cancer. Cell 1996, 87, 159–170. [Google Scholar] [CrossRef] [Green Version]
- Guinney, J.; Dienstmann, R.; Wang, X.; de Reyniès, A.; Schlicker, A.; Soneson, C.; Marisa, L.; Roepman, P.; Nyamundanda, G.; Angelino, P.; et al. The consensus molecular subtypes of colorectal cancer. Nat. Med. 2015, 21, 1350–1356. [Google Scholar] [CrossRef]
- La Vecchia, S.; Sebastian, C. Metabolic pathways regulating colorectal cancer initiation and progression. Semin. Cell Dev. Biol. 2020, 98, 63–70. [Google Scholar] [CrossRef]
- Shim, H.; Dolde, C.; Lewis, B.C.; Wu, C.S.; Dang, G.; Jungmann, R.A.; Dalla-Favera, R.; Dang, C.V. c-Myc transactivation of LDH-A: Implications for tumor metabolism and growth. Proc. Natl. Acad. Sci. USA 1997, 94, 6658–6663. [Google Scholar] [CrossRef] [Green Version]
- Baysal, B.E.; Ferrell, R.E.; Willett-Brozick, J.E.; Lawrence, E.C.; Myssiorek, D.; Bosch, A.; van der Mey, A.; Taschner, P.E.; Rubinstein, W.S.; Myers, E.N.; et al. Mutations in SDHD, a mitochondrial complex II gene, in hereditary paraganglioma. Science 2000, 287, 848–851. [Google Scholar] [CrossRef] [PubMed]
- Green, A.; Beer, P. Somatic mutations of IDH1 and IDH2 in the leukemic transformation of myeloproliferative neoplasms. N. Engl. J. Med. 2010, 362, 369–370. [Google Scholar] [CrossRef]
- Hao, H.X.; Khalimonchuk, O.; Schraders, M.; Dephoure, N.; Bayley, J.P.; Kunst, H.; Devilee, P.; Cremers, C.W.; Schiffman, J.D.; Bentz, B.G.; et al. SDH5, a gene required for flavination of succinate dehydrogenase, is mutated in paraganglioma. Science 2009, 325, 1139–1142. [Google Scholar] [CrossRef] [Green Version]
- Schiliro, C.; Firestein, B.L. Mechanisms of Metabolic Reprogramming in Cancer Cells Supporting Enhanced Growth and Proliferation. Cells 2021, 10, 1056. [Google Scholar] [CrossRef]
- Ying, H.; Kimmelman, A.C.; Lyssiotis, C.A.; Hua, S.; Chu, G.C.; Fletcher-Sananikone, E.; Locasale, J.W.; Son, J.; Zhang, H.; Coloff, J.L.; et al. Oncogenic Kras Maintains Pancreatic Tumors through Regulation of Anabolic Glucose Metabolism. Cell 2012, 149, 656–670. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Toda, K.; Kawada, K.; Iwamoto, M.; Inamoto, S.; Sasazuki, T.; Shirasawa, S.; Hasegawa, S.; Sakai, Y. Metabolic Alterations Caused by KRAS Mutations in Colorectal Cancer Contribute to Cell Adaptation to Glutamine Depletion by Upregulation of Asparagine Synthetase. Neoplasia 2016, 18, 654–665. [Google Scholar] [CrossRef] [Green Version]
- Hutton, J.E.; Wang, X.; Zimmerman, L.J.; Slebos, R.J.C.; Trenary, I.A.; Young, J.D.; Li, M.; Liebler, D.C. Oncogenic KRAS and BRAF Drive Metabolic Reprogramming in Colorectal Cancer. Mol. Cell. Proteom. 2016, 15, 2924–2938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pate, K.T.; Stringari, C.; Sprowl-Tanio, S.; Wang, K.; TeSlaa, T.; Hoverter, N.P.; McQuade, M.M.; Garner, C.; Digman, M.A.; Teitell, M.A.; et al. Wnt signaling directs a metabolic program of glycolysis and angiogenesis in colon cancer. EMBO J. 2014, 33, 1454–1473. [Google Scholar] [CrossRef]
- Sandoval, I.T.; Delacruz, R.G.; Miller, B.N.; Hill, S.; Olson, K.A.; Gabriel, A.E.; Boyd, K.; Satterfield, C.; Van Remmen, H.; Rutter, J.; et al. A metabolic switch controls intestinal differentiation downstream of Adenomatous polyposis coli (APC). Elife 2017, 6, 248. [Google Scholar] [CrossRef] [Green Version]
- Olivares, O.; Däbritz, J.H.M.; King, A.; Gottlieb, E.; Halsey, C. Research into cancer metabolomics: Towards a clinical metamorphosis. Semin. Cell Dev. Biol. 2015, 43, 52–64. [Google Scholar] [CrossRef] [Green Version]
- Thompson, C.B. Metabolic enzymes as oncogenes or tumor suppressors. N. Engl. J. Med. 2009, 360, 813–815. [Google Scholar] [CrossRef] [Green Version]
- Ward, P.S.; Thompson, C.B. Metabolic reprogramming: A cancer hallmark even warburg did not anticipate. Cancer Cell 2012, 21, 297–308. [Google Scholar] [CrossRef] [Green Version]
- Yang, M.; Soga, T.; Pollard, P.J.; Adam, J. The emerging role of fumarate as an oncometabolite. Front. Oncol. 2012, 2, 85. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Hirschey, M.D.; DeBerardinis, R.J.; Diehl, A.M.E.; Drew, J.E.; Frezza, C.; Green, M.F.; Jones, L.W.; Ko, Y.H.; Le, A.; Lea, M.A.; et al. Dysregulated metabolism contributes to oncogenesis. Semin. Cancer Biol. 2015, 35, S129–S150. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Pollard, P.J.; Brière, J.J.; Alam, N.A.; Barwell, J.; Barclay, E.; Wortham, N.C.; Hunt, T.; Mitchell, M.; Olpin, S.; Moat, S.J.; et al. Accumulation of Krebs cycle intermediates and over-expression of HIF1alpha in tumours which result from germline FH and SDH mutations. Hum. Mol. Genet. 2005, 14, 2231–2239. [Google Scholar] [CrossRef] [PubMed]
- Ward, P.S.; Patel, J.; Wise, D.R.; Abdel-Wahab, O.; Bennett, B.D.; Coller, H.A.; Cross, J.R.; Fantin, V.R.; Hedvat, C.V.; Perl, A.E.; et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 2010, 17, 225–234. [Google Scholar] [CrossRef] [Green Version]
- Cheng, Q.Y.; Xiong, J.; Huang, W.; Ma, Q.; Ci, W.; Feng, Y.Q.; Yuan, B.F. Sensitive Determination of Onco-metabolites of D- and L-2-hydroxyglutarate Enantiomers by Chiral Derivatization Combined with Liquid Chromatography/Mass Spectrometry Analysis. Sci. Rep. 2015, 5, 15217. [Google Scholar] [CrossRef] [Green Version]
- Du, X.; Hu, H. The Roles of 2-Hydroxyglutarate. Front. Cell Dev. Biol. 2021, 9, 486. [Google Scholar] [CrossRef]
- Lokody, I. Metabolism: IDH2 drives cancer in vivo. Nat. Rev. Cancer 2013, 13, 756–757. [Google Scholar] [CrossRef]
- Shelar, S.; Shim, E.H.; Brinkley, G.J.; Kundu, A.; Carobbio, F.; Poston, T.; Tan, J.; Parekh, V.; Benson, D.; Crossman, D.K.; et al. Biochemical and Epigenetic Insights into L-2-Hydroxyglutarate, a Potential Therapeutic Target in Renal Cancer. Clin. Cancer Res. 2018, 24, 6433–6446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, F.; Travins, J.; DeLaBarre, B.; Penard-Lacronique, V.; Schalm, S.; Hansen, E.; Straley, K.; Kernytsky, A.; Liu, W.; Gliser, C.; et al. Targeted inhibition of mutant IDH2 in leukemia cells induces cellular differentiation. Science 2013, 340, 622–626. [Google Scholar] [CrossRef]
- Bunse, L.; Pusch, S.; Bunse, T.; Sahm, F.; Sanghvi, K.; Friedrich, M.; Alansary, D.; Sonner, J.K.; Green, E.; Deumelandt, K.; et al. Suppression of antitumor T cell immunity by the oncometabolite (R)-2-hydroxyglutarate. Nat. Med. 2018, 24, 1192–1203. [Google Scholar] [CrossRef]
- Richardson, L.G.; Choi, B.D.; Curry, W.T. (R)-2-hydroxyglutarate drives immune quiescence in the tumor microenvironment of IDH-mutant gliomas. Transl. Cancer Res. 2019, 8, S167–S170. [Google Scholar] [CrossRef]
- Kohanbash, G.; Carrera, D.A.; Shrivastav, S.; Ahn, B.J.; Jahan, N.; Mazor, T.; Chheda, Z.S.; Downey, K.M.; Watchmaker, P.B.; Beppler, C.; et al. Isocitrate dehydrogenase mutations suppress STAT1 and CD8+ T cell accumulation in gliomas. J. Clin. Investig. 2017, 127, 1425–1437. [Google Scholar] [CrossRef] [PubMed]
- Böttcher, M.; Renner, K.; Berger, R.; Mentz, K.; Thomas, S.; Cardenas-Conejo, Z.E.; Dettmer, K.; Oefner, P.J.; Mackensen, A.; Kreutz, M.; et al. D-2-hydroxyglutarate interferes with HIF-1α stability skewing T-cell metabolism towards oxidative phosphorylation and impairing Th17 polarization. Oncoimmunology 2018, 7, e1445454. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, L.; Srensen, M.D.; Kristensen, B.W.; Reifenberger, G.; McIntyre, T.M.; Lin, F. D-2-Hydroxyglutarate Is an Intercellular Mediator in IDH-Mutant Gliomas Inhibiting Complement and T Cells. Clin. Cancer Res. 2018, 24, 5381–5391. [Google Scholar] [CrossRef] [Green Version]
- Colvin, H.; Nishida, N.; Konno, M.; Haraguchi, N.; Takahashi, H.; Nishimura, J.; Hata, T.; Kawamoto, K.; Asai, A.; Tsunekuni, K.; et al. Oncometabolite D-2-Hydroxyglurate Directly Induces Epithelial-Mesenchymal Transition and is Associated with Distant Metastasis in Colorectal Cancer. Sci. Rep. 2016, 6, 36289. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kim, E.R.; Chang, D.K. Colorectal cancer in inflammatory bowel disease: The risk, pathogenesis, prevention and diagnosis. World J. Gastroenterol. 2014, 20, 9872–9881. [Google Scholar] [CrossRef]
- Han, J.; Jackson, D.; Holm, J.; Turner, K.; Ashcraft, P.; Wang, X.; Cook, B.; Arning, E.; Genta, R.M.; Venuprasad, K.; et al. Elevated d-2-hydroxyglutarate during colitis drives progression to colorectal cancer. Proc. Natl. Acad. Sci. USA 2018, 115, 1057–1062. [Google Scholar] [CrossRef] [Green Version]
- Venkateswaran, N.; Conacci-Sorrell, M. Kynurenine: An oncometabolite in colon cancer. Cell Stress 2020, 4, 24–26. [Google Scholar] [CrossRef] [Green Version]
- Heng, B.; Lim, C.K.; Lovejoy, D.B.; Bessede, A.; Gluch, L.; Guillemin, G.J. Understanding the role of the kynurenine pathway in human breast cancer immunobiology. Oncotarget 2016, 7, 6506–6520. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Opitz, C.A.; Litzenburger, U.M.; Sahm, F.; Ott, M.; Tritschler, I.; Trump, S.; Schumacher, T.; Jestaedt, L.; Schrenk, D.; Weller, M.; et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 2011, 478, 197–203. [Google Scholar] [CrossRef] [PubMed]
- Taguchi, A.; Niwa, M.; Hoshi, M.; Saito, K.; Masutani, T.; Hisamatsu, K.; Kobayashi, K.; Hatano, Y.; Tomita, H.; Hara, A. Indoleamine 2,3-dioxygenase 1 is upregulated in activated microglia in mice cerebellum during acute viral encephalitis. Neurosci. Lett. 2014, 564, 120–125. [Google Scholar] [CrossRef] [PubMed]
- Brandacher, G.; Perathoner, A.; Ladurner, R.; Schneeberger, S.; Obrist, P.; Winkler, C.; Werner, E.R.; Werner-Felmayer, G.; Weiss, H.G.; Göbel, G.; et al. Prognostic value of indoleamine 2,3-dioxygenase expression in colorectal cancer: Effect on tumor-infiltrating T cells. Clin. Cancer Res. 2006, 12, 1144–1151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bishnupuri, K.S.; Alvarado, D.M.; Khouri, A.N.; Shabsovich, M.; Chen, B.; Dieckgraefe, B.K.; Ciorba, M.A. IDO1 and Kynurenine Pathway Metabolites Activate PI3K-Akt Signaling in the Neoplastic Colon Epithelium to Promote Cancer Cell Proliferation and Inhibit Apoptosis. Cancer Res. 2019, 79, 1138–1150. [Google Scholar] [CrossRef] [Green Version]
- Venkateswaran, N.; Lafita-Navarro, M.C.; Hao, Y.H.; Kilgore, J.A.; Perez-Castro, L.; Braverman, J.; Borenstein-Auerbach, N.; Kim, M.; Lesner, N.P.; Mishra, P.; et al. MYC promotes tryptophan uptake and metabolism by the kynurenine pathway in colon cancer. Genes Dev. 2019, 33, 1236–1251. [Google Scholar] [CrossRef] [Green Version]
- Hanahan, D.; Weinberg, R.A. The hallmarks of cancer. Cell 2000, 100, 57–70. [Google Scholar] [CrossRef] [Green Version]
- Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
- Rossi, T.; Vergara, D.; Fanini, F.; Maffia, M.; Bravaccini, S.; Pirini, F. Microbiota-Derived Metabolites in Tumor Progression and Metastasis. Int. J. Mol. Sci. 2020, 21, 5786. [Google Scholar] [CrossRef]
- Wu, Y.; Jiao, N.; Zhu, R.; Zhang, Y.; Wu, D.; Wang, A.-J.; Fang, S.; Tao, L.; Li, Y.; Cheng, S.; et al. Identification of microbial markers across populations in early detection of colorectal cancer. Nat. Commun. 2021, 12, 3063. [Google Scholar] [CrossRef]
- Zou, S.; Fang, L.; Lee, M.-H. Dysbiosis of gut microbiota in promoting the development of colorectal cancer. Gastroenterol. Rep. 2018, 6, 1–12. [Google Scholar] [CrossRef] [Green Version]
- Pickard, J.M.; Zeng, M.Y.; Caruso, R.; Núñez, G. Gut microbiota: Role in pathogen colonization, immune responses, and inflammatory disease. Immunol. Rev. 2017, 279, 70–89. [Google Scholar] [CrossRef] [PubMed]
- Bultman, S.J. Emerging roles of the microbiome in cancer. Carcinogenesis 2014, 35, 249–255. [Google Scholar] [CrossRef] [Green Version]
- Kosumi, K.; Mima, K.; Baba, H.; Ogino, S. Dysbiosis of the gut microbiota and colorectal cancer: The key target of molecular pathological epidemiology. J. Lab. Precis. Med. 2018, 3, 76. [Google Scholar] [CrossRef] [PubMed]
- 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]
- Pan, P.; Oshima, K.; Huang, Y.W.; Agle, K.A.; Drobyski, W.R.; Chen, X.; Zhang, J.; Yearsley, M.M.; Yu, J.; Wang, L.S. Loss of FFAR2 promotes colon cancer by epigenetic dysregulation of inflammation suppressors. Int. J. Cancer 2018, 143, 886–896. [Google Scholar] [CrossRef] [PubMed]
- Tang, Y.; Chen, Y.; Jiang, H.; Robbins, G.T.; Nie, D. G-protein-coupled receptor for short-chain fatty acids suppresses colon cancer. Int. J. Cancer 2011, 128, 847–856. [Google Scholar] [CrossRef]
- Dengler, F.; Kraetzig, A.; Gäbel, G. Butyrate Protects Porcine Colon Epithelium from Hypoxia-Induced Damage on a Functional Level. Nutrients 2021, 13, 305. [Google Scholar] [CrossRef]
- Hamer, H.M.; Jonkers, D.; Venema, K.; Vanhoutvin, S.; Troost, F.J.; Brummer, R.J. Review article: The role of butyrate on colonic function. Aliment. Pharmacol. Ther. 2008, 27, 104–119. [Google Scholar] [CrossRef] [PubMed]
- Peng, Y.; Nie, Y.; Yu, J.; Wong, C.C. Microbial Metabolites in Colorectal Cancer: Basic and Clinical Implications. Metabolites 2021, 11, 159. [Google Scholar] [CrossRef]
- Belcheva, A.; Irrazabal, T.; Robertson, S.J.; Streutker, C.; Maughan, H.; Rubino, S.; Moriyama, E.H.; Copeland, J.K.; Surendra, A.; Kumar, S.; et al. Gut microbial metabolism drives transformation of MSH2-deficient colon epithelial cells. Cell 2014, 158, 288–299. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, H.M.; Yu, Y.N.; Wang, J.L.; Lin, Y.W.; Kong, X.; Yang, C.Q.; Yang, L.; Liu, Z.J.; Yuan, Y.Z.; Liu, F.; et al. Decreased dietary fiber intake and structural alteration of gut microbiota in patients with advanced colorectal adenoma. Am. J. Clin. Nutr. 2013, 97, 1044–1052. [Google Scholar] [CrossRef] [Green Version]
- Bultman, S.J. Molecular pathways: Gene-environment interactions regulating dietary fiber induction of proliferation and apoptosis via butyrate for cancer prevention. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2014, 20, 799–803. [Google Scholar] [CrossRef] [Green Version]
- Soda, K. The mechanisms by which polyamines accelerate tumor spread. J. Exp. Clin. Cancer Res. 2011, 30, 95. [Google Scholar] [CrossRef] [Green Version]
- Thomas, T.; Thomas, T.J. Polyamine metabolism and cancer. J. Cell Mol. Med. 2003, 7, 113–126. [Google Scholar] [CrossRef] [PubMed]
- Shah, P.; Swiatlo, E. A multifaceted role for polyamines in bacterial pathogens. Mol. Microbiol. 2008, 68, 4–16. [Google Scholar] [CrossRef]
- Johnson, C.H.; Dejea, C.M.; Edler, D.; Hoang, L.T.; Santidrian, A.F.; Felding, B.H.; Ivanisevic, J.; Cho, K.; Wick, E.C.; Hechenbleikner, E.M.; et al. Metabolism links bacterial biofilms and colon carcinogenesis. Cell Metab. 2015, 21, 891–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Goodwin, A.C.; Destefano Shields, C.E.; Wu, S.; Huso, D.L.; Wu, X.; Murray-Stewart, T.R.; Hacker-Prietz, A.; Rabizadeh, S.; Woster, P.M.; Sears, C.L.; et al. Polyamine catabolism contributes to enterotoxigenic Bacteroides fragilis-induced colon tumorigenesis. Proc. Natl. Acad. Sci. USA 2011, 108, 15354–15359. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, C.; Ruan, P.; Zhao, Y.; Li, X.; Wang, J.; Wu, X.; Liu, T.; Wang, S.; Hou, J.; Li, W.; et al. Spermidine/spermine N1-acetyltransferase regulates cell growth and metastasis via AKT/β-catenin signaling pathways in hepatocellular and colorectal carcinoma cells. Oncotarget 2017, 8, 1092–1109. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cook, J.W.; Kennaway, E.L.; Kennaway, N.M. Production of Tumours in Mice by Deoxycholic Acid. Nature 1940, 145, 627. [Google Scholar] [CrossRef]
- Bernstein, C.; Holubec, H.; Bhattacharyya, A.K.; Nguyen, H.; Payne, C.M.; Zaitlin, B.; Bernstein, H. Carcinogenicity of deoxycholate, a secondary bile acid. Arch. Toxicol. 2011, 85, 863–871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Ocvirk, S.; Wilson, A.S.; Posma, J.M.; Li, J.V.; Koller, K.R.; Day, G.M.; Flanagan, C.A.; Otto, J.E.; Sacco, P.E.; Sacco, F.D.; et al. A prospective cohort analysis of gut microbial co-metabolism in Alaska Native and rural African people at high and low risk of colorectal cancer. Am. J. Clin. Nutr. 2020, 111, 406–419. [Google Scholar] [CrossRef]
- Yachida, S.; Mizutani, S.; Shiroma, H.; Shiba, S.; Nakajima, T.; Sakamoto, T.; Watanabe, H.; Masuda, K.; Nishimoto, Y.; Kubo, M.; et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat. Med. 2019, 25, 968–976. [Google Scholar] [CrossRef]
- Liu, L.; Dong, W.; Wang, S.; Zhang, Y.; Liu, T.; Xie, R.; Wang, B.; Cao, H. Deoxycholic acid disrupts the intestinal mucosal barrier and promotes intestinal tumorigenesis. Food Funct. 2018, 9, 5588–5597. [Google Scholar] [CrossRef] [Green Version]
- Payne, C.M.; Weber, C.; Crowley-Skillicorn, C.; Dvorak, K.; Bernstein, H.; Bernstein, C.; Holubec, H.; Dvorakova, B.; Garewal, H. Deoxycholate induces mitochondrial oxidative stress and activates NF-κB through multiple mechanisms in HCT-116 colon epithelial cells. Carcinogenesis 2007, 28, 215–222. [Google Scholar] [CrossRef]
- Soderholm, A.T.; Pedicord, V.A. Intestinal epithelial cells: At the interface of the microbiota and mucosal immunity. Immunology 2019, 158, 267–280. [Google Scholar] [CrossRef] [Green Version]
- Zheng, D.; Liwinski, T.; Elinav, E. Interaction between microbiota and immunity in health and disease. Cell Res. 2020, 30, 492–506. [Google Scholar] [CrossRef]
- Haghikia, A.; Jörg, S.; Duscha, A.; Berg, J.; Manzel, A.; Waschbisch, A.; Hammer, A.; Lee, D.H.; May, C.; Wilck, N.; et al. Dietary Fatty Acids Directly Impact Central Nervous System Autoimmunity via the Small Intestine. Immunity 2015, 43, 817–829. [Google Scholar] [CrossRef] [Green Version]
- Mariño, E.; Richards, J.L.; McLeod, K.H.; Stanley, D.; Yap, Y.A.; Knight, J.; McKenzie, C.; Kranich, J.; Oliveira, A.C.; Rossello, F.J.; et al. Gut microbial metabolites limit the frequency of autoimmune T cells and protect against type 1 diabetes. Nat. Immunol. 2017, 18, 552–562. [Google Scholar] [CrossRef]
- Tan, J.; McKenzie, C.; Vuillermin, P.J.; Goverse, G.; Vinuesa, C.G.; Mebius, R.E.; Macia, L.; Mackay, C.R. Dietary Fiber and Bacterial SCFA Enhance Oral Tolerance and Protect against Food Allergy through Diverse Cellular Pathways. Cell Rep. 2016, 15, 2809–2824. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.D.; Manicassamy, S.; Munn, D.H.; et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Luu, M.; Weigand, K.; Wedi, F.; Breidenbend, C.; Leister, H.; Pautz, S.; Adhikary, T.; Visekruna, A. Regulation of the effector function of CD8+ T cells by gut microbiota-derived metabolite butyrate. Sci. Rep. 2018, 8, 14430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abshire, D.; Lang, M.K. The Evolution of Radiation Therapy in Treating Cancer. Semin. Oncol. Nurs. 2018, 34, 151–157. [Google Scholar] [CrossRef]
- Liu, J.; Liu, C.; Yue, J. Radiotherapy and the gut microbiome: Facts and fiction. Radiat. Oncol. 2021, 16, 9. [Google Scholar] [CrossRef]
- Jang, B.S.; Chang, J.H.; Chie, E.K.; Kim, K.; Park, J.W.; Kim, M.J.; Song, E.J.; Nam, Y.D.; Kang, S.W.; Jeong, S.Y.; et al. Gut Microbiome Composition Is Associated with a Pathologic Response After Preoperative Chemoradiation in Patients with Rectal Cancer. Int. J. Radiat. Oncol. Biol. Phys. 2020, 107, 736–746. [Google Scholar] [CrossRef]
- Kim, Y.S.; Kim, J.; Park, S.-J. High-throughput 16S rRNA gene sequencing reveals alterations of mouse intestinal microbiota after radiotherapy. Anaerobe 2015, 33, 1–7. [Google Scholar] [CrossRef]
- Yang, K.; Hou, Y.; Zhang, Y.; Liang, H.; Sharma, A.; Zheng, W.; Wang, L.; Torres, R.; Tatebe, K.; Chmura, S.J.; et al. Suppression of local type I interferon by gut microbiota–derived butyrate impairs antitumor effects of ionizing radiation. J. Exp. Med. 2021, 218, e20201915. [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, 1–27. [Google Scholar] [CrossRef]
- Li, Y.; Dong, J.; Xiao, H.; Zhang, S.; Wang, B.; Cui, M.; Fan, S. Gut commensal derived-valeric acid protects against radiation injuries. Gut Microbes 2020, 11, 789–806. [Google Scholar] [CrossRef] [PubMed]
- Jiang, T.; Zhou, C. The past, present and future of immunotherapy against tumor. Transl. Lung Cancer Res. 2015, 4, 253–264. [Google Scholar] [CrossRef]
- Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef]
- Sears, C.L.; Pardoll, D.M. The intestinal microbiome influences checkpoint blockade. Nat. Med. 2018, 24, 254–255. [Google Scholar] [CrossRef]
- Xu, X.; Lv, J.; Guo, F.; Li, J.; Jia, Y.; Jiang, D.; Wang, N.; Zhang, C.; Kong, L.; Liu, Y.; et al. Gut Microbiome Influences the Efficacy of PD-1 Antibody Immunotherapy on MSS-Type Colorectal Cancer via Metabolic Pathway. Front. Microbiol. 2020, 11, 814. [Google Scholar] [CrossRef]
- Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Botticelli, A.; Vernocchi, P.; Marini, F.; Quagliariello, A.; Cerbelli, B.; Reddel, S.; Del Chierico, F.; Di Pietro, F.; Giusti, R.; Tomassini, A.; et al. Gut metabolomics profiling of non-small cell lung cancer (NSCLC) patients under immunotherapy treatment. J. Transl. Med. 2020, 18, 49. [Google Scholar] [CrossRef]
- Andriamihaja, M.; Lan, A.; Beaumont, M.; Audebert, M.; Wong, X.; Yamada, K.; Yin, Y.; Tomé, D.; Carrasco-Pozo, C.; Gotteland, M.; et al. The deleterious metabolic and genotoxic effects of the bacterial metabolite p-cresol on colonic epithelial cells. Free Radic. Biol. Med. 2015, 85, 219–227. [Google Scholar] [CrossRef]
- Verbeke, K.A.; Boobis, A.R.; Chiodini, A.; Edwards, C.A.; Franck, A.; Kleerebezem, M.; Nauta, A.; Raes, J.; van Tol, E.A.F.; Tuohy, K.M. Towards microbial fermentation metabolites as markers for health benefits of prebiotics. Nutr. Res. Rev. 2015, 28, 42–66. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Metabolite | Origin | Mechanism | Effect | Reference |
---|---|---|---|---|
SCFAs (butyrate, propionate, acetate) | Food | Treg-mediated immunity | Gut-immune regulation | |
Microbial fermentation products | Inhibition of HDACs | Increased intestinal barrier integrity | [29,30,31] | |
Tumor cells apoptosis | Tumor protective | |||
Butyrate | Food | Proliferation | Proliferation in Apcmin/+, Msh2−/− mice | [94,95] |
Microbial fermentation products | Enhanced β-catenin activity | Tumor promoting | ||
Vitamin A/D | Food | Reduction in cytokines levels | Increased intestinal barrier integrity | [33,34,35] |
Upregulation of Tight junctions | Tumor protective | |||
Increased intestinal barrier integrity | ||||
Polyamines | Food, microbial contents | Upregulation of spermine oxidase | Inflammation | |
Upregulation of EMT genes | Proliferation | [99,101,102] | ||
Tumor promoting | ||||
Deoxycholic acid (DCA) | Cholesterol/secondary bile acids | Pro-inflammatory genes/anti-apoptotic genes | Intestinal barrier dysfunction | |
NLRP3 activation | Cytokine inflammation | [107,108] | ||
Intestinal barrier permeability | CSC proliferation | |||
Tumor promoting | ||||
2-hydroxyglutarate (2-HG) | Reduced form of α-KG | Promotion of EMT genes | Invasiveness and metastasis | |
Downregulation of D2HGDH | EMT phenotype | [69,71,72] | ||
Apoptosis resistance | ||||
Tumor promoting | ||||
Kynurenine | Diet-derived tryptophan | PI3K-AKT-induced proliferation | Proliferation | |
Upregulation of membrane carrier LAT1 | Biomass, protein synthesis | [78,79] | ||
Tumor promoting |
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
Zaurito, A.E.; Tschurtschenthaler, M. Microenvironmental Metabolites in the Intestine: Messengers between Health and Disease. Metabolites 2022, 12, 46. https://doi.org/10.3390/metabo12010046
Zaurito AE, Tschurtschenthaler M. Microenvironmental Metabolites in the Intestine: Messengers between Health and Disease. Metabolites. 2022; 12(1):46. https://doi.org/10.3390/metabo12010046
Chicago/Turabian StyleZaurito, Antonio Enrico, and Markus Tschurtschenthaler. 2022. "Microenvironmental Metabolites in the Intestine: Messengers between Health and Disease" Metabolites 12, no. 1: 46. https://doi.org/10.3390/metabo12010046
APA StyleZaurito, A. E., & Tschurtschenthaler, M. (2022). Microenvironmental Metabolites in the Intestine: Messengers between Health and Disease. Metabolites, 12(1), 46. https://doi.org/10.3390/metabo12010046