Nutrition and Cancer Risk from the Viewpoint of the Intestinal Microbiome
Abstract
:1. Introduction
2. Mechanism of Onset and Progression of Cancer
2.1. Deterioration of the Cellular Environment
2.2. Endoplasmic Reticulum STRESS
2.3. Hypofunction of Mitochondria
3. The Role of the Intestinal Microbiome
3.1. Control of the Metabolism
3.2. Production of Valuable Substances
3.3. Control of Immunity
3.4. Communication with Other Organs
4. Enterotype
- (1)
- Bifidobacterium: This genus has catalytic activity of cyclic lactic acid and the effect of the prevention of cancer development. It neutralizes waste products, such as bile acid, and is immunity-strengthening, aids mental stability, and complements the activity of other microbiota in carbohydrate and lipid metabolism [61].
- (2)
- Lactobacillus: This genus has catalytic activity of cyclic lactic acid and the effect of prevention of cancer development. It controls gene restoration and cell regeneration, is immunity-strengthening, aids mental stability, and complements the activity of other microbiota in carbohydrate and lipid metabolism [62].
- (3)
- Clostridium: This genus promotes the secretion of regulatory T cells and has useful effects on allergic diseases, autoimmune diseases, and chronic inflammatory diseases via the secretion of SCFAs [63].
- Cluster XVIII: Involved in suppression of carcinogenesis and overreaction of the immune system.
- Cluster XV: Involved in activation of macrophages and induction of apoptosis.
- Cluster IX Involved in gene restoration and induction of apoptosis.
- Cluster IV: Involved in induction of regulatory T cells.
- (4)
- (5)
- Clostridium cluster Blautia: Involved in the restoration of inflammatory tissues and mutated cells, and control of immunity [66].
- (6)
- Faecalibacterium prausnitzii: Involved in suppression of obesity and the onset of diabetes mellitus, and in induction of regulatory T cells via the production of butyric acid [67].
- (7)
- Clostridium butyricum: Involved in prevention of diseases via abnormal proliferation of resistant microbes, such as Clostridium difficile. It is a butyric acid-producing bacterium [68].
5. Improvement of Dysbiosis
5.1. Prebiotics
5.2. Probiotics
5.2.1. Background
5.2.2. Materials and Methods
5.2.3. Results
5.3. Fecal Microbiota Transplantation (FMT)
5.3.1. Background
5.3.2. Materials and Methods
5.3.3. Result
6. Discussion
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- Lozupone, C.A.; Stombaugh, J.I.; Gordon, J.I.; Jansson, J.K.; Knight, R. Diversity, stability and resilience of the human gut microbiota. Nature 2012, 489, 220–230. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yang, J.; Tan, Q.; Fu, Q.; Zhou, Y.; Hu, Y.; Tang, S.; Zhou, Y.; Zhang, J.; Qiu, J.; Lv, Q. Gastrointestinal microbiome and breast cancer: Correlations, mechanisms and potential clinical implications. Breast Cancer 2017, 24, 220–228. [Google Scholar] [CrossRef]
- Hosgood, H.D.; Cai, Q.; Hua, X.; Long, J.; Shi, J.; Wan, Y.; Yang, Y.; Abnet, C.; Bassig, B.A.; Hu, W.; et al. Variation in oral microbiome is associated with future risk of lung cancer among never-smokers. Thorax 2021, 76, 256–263. [Google Scholar] [CrossRef] [PubMed]
- Song, M.; Chan, A.T.; Sun, J. Influence of the gut microbiome, diet, and environment on risk of colorectal cancer. Gastroenterology 2020, 158, 322–340. [Google Scholar] [CrossRef] [PubMed]
- Yamamura, K.; Baba, Y.; Nakagawa, S.; Mima, K.; Miyake, K.; Nakamura, K.; Sawayama, H.; Kinoshita, K.; Ishimoto, T.; Iwatsuki, M.; et al. Human microbiome fusobacterium nucleatum in esophageal cancer tissue is associated with prognosis. Clin. Cancer Res. 2016, 22, 5574–5581. [Google Scholar] [CrossRef] [Green Version]
- Riquelme, E.; Zhang, Y.; Zhang, L.; Montiel, M.; Zoltan, M.; Dong, W.; Quesada, P.; Sahin, I.; Chandra, V.; San Lucas, A.; et al. Tumor microbiome diversity and composition influence pancreatic cancer outcomes. Cell 2019, 178, 795–806. [Google Scholar] [CrossRef]
- Zhang, X.; Coker, O.O.; Chu, E.S.; Fu, K.; Lau, H.C.H.; Wang, Y.X.; Chan, A.W.H.; Wei, H.; Yang, X.; Sung, J.J.Y.; et al. Dietary cholesterol drives fatty liver-associated liver cancer by modulating gut microbiota and metabolites. Gut 2021, 70, 761–774. [Google Scholar] [CrossRef]
- Jia, W.; Xie, G.; Jia, W. Bile acid-microbiota crosstalk in gastrointestinal inflammation and carcinogenesis. Nat. Rev. Gastroenterol. Hepatol. 2018, 15, 111–128. [Google Scholar] [CrossRef] [Green Version]
- Ma, C.; Han, M.; Heinrich, B.; Fu, Q.; Zhang, Q.; Sandhu, M.; Agdashian, D.; Terabe, M.; Berzofsky, J.A.; Fako, V.; et al. Gut microbiome-mediated bile acid metabolism regulates liver cancer via NKT cells. Science 2018, 360, eaan5931. [Google Scholar] [CrossRef] [Green Version]
- Huybrechts, I.; Zouiouich, S.; Loobuyck, A.; Vandenbulcke, Z.; Vogtmann, E.; Pisanu, S.; Iguacel, I.; Scalbert, A.; Indave, I.; Smelov, V.; et al. The human microbiome in relation to cancer risk: A systematic review of epidemiologic studies. Cancer Epidemiol. Biomark. Prev. 2020, 29, 1856–1868. [Google Scholar] [CrossRef]
- Goodman, B.; Gardner, H. The microbiome and cancer. J. Pathol. 2018, 244, 667–676. [Google Scholar] [CrossRef] [Green Version]
- Elinav, E.; Garrett, W.S.; Trinchieri, G.; Wargo, J. The cancer microbiome. Nat. Rev. Cancer 2019, 19, 371–376. [Google Scholar] [CrossRef]
- Schwabe, R.F.; Jobin, C. The microbiome and cancer. Nat. Rev. Cancer 2013, 13, 800–812. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wilson, A.S.; Koller, K.R.; Ramaboli, M.C.; Nesengani, L.T.; Ocvirk, S.; Chen, C.; Flanagan, C.A.; Sapp, F.R.; Merritt, Z.T.; Bhatti, F.; et al. Diet and the human gut microbiome: An international review. Dig. Dis. Sci. 2020, 65, 723–740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Yadav, M.; Verma, M.K.; Chauhan, N.S. A review of metabolic potential of human gut microbiome in human nutrition. Arch. Microbiol. 2018, 200, 203–217. [Google Scholar] [CrossRef]
- Frame, L.A.; Costa, E.; Jackson, S.A. Current explorations of nutrition and the gut microbiome: A comprehensive evaluation of the review literature. Nutr. Rev. 2020, 78, 798–812. [Google Scholar] [CrossRef] [PubMed]
- Kolodziejczyk, A.A.; Zheng, D.; Elinav, E. Diet-microbiota interactions and personalized nutrition. Nat. Rev. Microbiol. 2019, 17, 742–753. [Google Scholar] [CrossRef]
- David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563. [Google Scholar] [CrossRef] [Green Version]
- Marchesi, J.R.; Adams, D.H.; Fava, F.; Hermes, G.D.; Hirschfield, G.M.; Hold, G.; Quraishi, M.N.; Kinross, J.; Smidt, H.; Tuohy, K.M.; et al. The gut microbiota and host health: A new clinical frontier. Gut 2016, 65, 330–339. [Google Scholar] [CrossRef] [Green Version]
- Skrypnik, K.; Suliburska, J. Association between the gut microbiota and mineral metabolism. J. Sci. Food Agric. 2018, 98, 2449–2460. [Google Scholar] [CrossRef]
- Liew, W.P.; Mohd-Redzwan, S. Mycotoxin: Its impact on gut health and microbiota. Front. Cell. Infect. Microbiol. 2018, 8, 60. [Google Scholar] [CrossRef] [Green Version]
- Avgerinos, K.I.; Spyrou, N.; Mantzoros, C.S.; Dalamaga, M. Obesity and cancer risk: Emerging biological mechanisms and perspectives. Metabolism 2019, 92, 121–135. [Google Scholar] [CrossRef] [PubMed]
- Donati Zeppa, S.; Agostini, D.; Gervasi, M.; Annibalini, G.; Amatori, S.; Ferrini, F.; Sisti, D.; Piccoli, G.; Barbieri, E.; Sestili, P.; et al. Mutual interactions among exercise, sport supplements and microbiota. Nutrients 2019, 12, 17. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cubillos-Ruiz, J.R.; Bettigole, S.E.; Glimcher, L.H. Tumorigenic and immunosuppressive effects of endoplasmic reticulum stress in cancer. Cell 2017, 168, 692–706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Urra, H.; Dufey, E.; Avril, T.; Chevet, E.; Hetz, C. Endoplasmic reticulum stress and the hallmarks of cancer. Trends Cancer 2016, 2, 252–262. [Google Scholar] [CrossRef]
- Oakes, S.A. Endoplasmic reticulum stress signaling in cancer cells. Am. J. Pathol. 2020, 190, 934–946. [Google Scholar] [CrossRef] [Green Version]
- Qi, Z.; Chen, L. Endoplasmic reticulum stress and autophagy. Adv. Exp. Med. Biol. 2019, 1206, 167–177. [Google Scholar]
- Jeong, S.Y.; Seol, D.W. The role of mitochondria in apoptosis. BMB Rep. 2008, 41, 11–22. [Google Scholar] [CrossRef] [Green Version]
- Sinha, K.; Das, J.; Pal, P.B.; Sil, P.C. Oxidative stress: The mitochondria-dependent and mitochondria-independent pathway of apoptosis. Arch. Toxicol. 2013, 87, 1157–1180. [Google Scholar] [CrossRef] [PubMed]
- Abate, M.; Festa, A.; Falco, M.; Lombardi, A.; Luce, A.; Grimaldi, A.; Zappavigna, S.; Sperlongano, P.; Irace, C.; Caraglia, M.; et al. Mitochondria as playmarkers of apoptosis, autophagy and senescence. Semin. Cell. Dev. Biol. 2020, 98, 139–153. [Google Scholar] [CrossRef]
- Benschop, R.J.; Rodriguez-Feuerhahn, M.; Schedlowski, M. Catecholamine-induced leukocytosis: Early observations, current research, and future directions. Brain Behav. Immun. 1996, 10, 77–91. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nakai, A.; Hayano, Y.; Furuta, F.; Noda, M.; Suzuki, K. Control of lymphocyte egress from lymph nodes through β2-adrenergic receptors. J. Exp. Med. 2014, 211, 2583–2598. [Google Scholar] [CrossRef]
- Grebe, K.M.; Hickman, H.D.; Irvine, K.R.; Takeda, K.; Bennink, J.R.; Yewdell, J.W. Sympathetic nervous system control of anti-influenza CD8+ T cell responses. Proc. Natl. Acad. Sci. USA 2009, 106, 5300–5305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Herve, J.; Dubreil, L.; Tardif, V.; Terme, M.; Pogu, S.; Anegon, I.; Rozec, B.; Gauthier, C.; Bach, J.M.; Blancou, P. β2-Adrenoreceptor agonist inhibits antigen cross-presentation by dendritic cells. J. Immunol. 2013, 190, 3163–3171. [Google Scholar] [CrossRef] [Green Version]
- Hu, Z.; Chen, R.; Cai, Z.; Yu, L.; Fei, Y.; Weng, L.; Wang, J.; Ge, X.; Zhu, T.; Wang, J.; et al. Salmeterol attenuates the inflammatory response in asthma and decreases the pro-inflammatory cytokine secretion of dendritic cells. Cell. Mol. Immunol. 2012, 9, 267–275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Nijhuis, L.E.; Olivier, B.J.; Dhawan, S.; Hibers, F.W.; Boon, L.; Wolkers, M.C.; Samsom, J.N.; de Jonge, W.J. Adrenergic β2 receptor activation stimulates anti-inflammatory properties of dendritic cells in vitro. PLoS ONE 2014, 9, e85086. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Reynolds, A.; Mann, J.; Cummings, J.; Winter, N.; Mete, E.; Te Morenga, L. Carbohydrate quality and human health: A series of systematic reviews and meta-analyses. Lancet 2019, 393, 434–445. [Google Scholar] [CrossRef] [Green Version]
- Schoeler, M.; Caesar, R. Dietary lipids, gut microbiota and lipid metabolism. Rev. Endocr. Metab. Disord. 2019, 20, 461–472. [Google Scholar] [CrossRef] [Green Version]
- Kumar, J.; Rani, K.; Datt, C. Molecular link between dietary fibre, gut microbiota and health. Mol. Biol. Rep. 2020, 47, 6229–6237. [Google Scholar] [CrossRef]
- Tan, J.; McKenzie, C.; Potamitis, M.; Thorburn, A.N.; Mackay, C.R.; Macia, L. The role of short-chain fatty acids in health and disease. Adv. Immunol. 2014, 121, 91–119. [Google Scholar]
- Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [Green Version]
- Sivaprakasam, S.; Prasad, P.D.; Singh, N. Benefits of short-chain fatty acids and their receptors in inflammation and carcinogenesis. Pharmacol. Ther. 2016, 164, 144–151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lee, L.-H.; Letchumanan, V.; Tan, L.T.-H.; Ser, H.-L.; Law, J.W.-F. Gut-skin axis: Decoding the link between the gut microbiome and hives. Gut 2020, 9, A17–A18. [Google Scholar]
- Johnson, D.; Letchumanan, V.; Thurairajasingam, S.; Lee, L.-H. A revolutionizing approach to autism spectrum disorder using the microbiome. Nutrients 2020, 12, 1983. [Google Scholar] [CrossRef]
- Nguyen, T.T.; Kosciolek, T.; Eyler, L.T.; Knight, R.; Jeste, D.V. Overview and systematic review of studies of microbiome in schizophrenia and bipolar disorder. J. Psychiatr. Res. 2018, 99, 50–61. [Google Scholar] [CrossRef] [PubMed]
- Nikolova, V.L.; Cleare, A.J.; Young, A.H.; Stone, J.M. Updated review and meta-analysis of probiotics for the treatment of clinical depression: Adjunctive vs. stand-alone treatment. J. Clin. Med. 2021, 10, 647. [Google Scholar] [CrossRef]
- Battistini, C.; Ballan, R.; Herkenhoff, M.E.; Saad, S.M.I.; Sun, J. Vitamin D modulates intestinal microbiota in inflammatory bowel diseases. Int. J. Mol. Sci. 2020, 22, 362. [Google Scholar] [CrossRef] [PubMed]
- Blachier, F.; Andriamihaja, M.; Larraufie, P.; Ahn, E.; Lan, A.; Kim, E. Production of hydrogen sulfide by the intestinal microbiota and epithelial cells and consequences for the colonic and rectal mucosa. Am. J. Physiol. Gastrointest. Liver Physiol. 2021, 320, G125–G135. [Google Scholar] [CrossRef] [PubMed]
- Couto, M.R.; Gonçalves, P.; Magro, F.; Martel, F. Microbiota-derived butyrate regulates intestinal inflammation: Focus on inflammatory bowel disease. Pharmacol. Res. 2020, 159, 104947. [Google Scholar] [CrossRef] [PubMed]
- Louis, P.; Flint, H.J. Formation of propionate and butyrate by the human colonic microbiota. Environ. Microbiol. 2017, 19, 29–41. [Google Scholar] [CrossRef] [Green Version]
- Cheng, H.; Guan, X.; Chen, D.; Ma, W. The Th17/Treg cell balance: A gut microbiota-modulated story. Microorganisms 2019, 7, 583. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mayer, E.A.; Tillisch, K.; Gupta, A. Gut/brain axis and the microbiota. J. Clin. Investig. 2015, 125, 926–938. [Google Scholar] [CrossRef]
- Louwies, T.; Johnson, A.C.; Orock, A.; Yuan, T.; Greenwood-Van Meerveld, B. The microbiota-gut-brain axis: An emerging role for the epigenome. Exp. Biol. Med. 2020, 245, 138–145. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kikuchi, K.; Saigusa, D.; Kanemitsu, Y.; Matsumoto, Y.; Thanai, P.; Suzuki, N.; Mise, K.; Yamaguchi, H.; Nakamura, T.; Asaji, K.; et al. Gut microbiome-derived phenyl sulfate contributes to albuminuria in diabetic kidney disease. Nat. Commun. 2019, 10, 1835. [Google Scholar] [CrossRef] [PubMed]
- Lee, C.J.; Sears, C.L.; Maruthur, N. Gut microbiome and its role in obesity and insulin resistance. Ann. N. Y. Acad. Sci. 2020, 1461, 37–52. [Google Scholar] [CrossRef]
- Tang, W.H.; Wang, Z.; Levison, B.S.; Koeth, R.A.; Britt, E.B.; Fu, X.; Wu, Y.; Hazen, S.L. Intestinal microbial metabolism of phosphatidylcholine and cardiovascular risk. N. Engl. J. Med. 2013, 368, 1575–1584. [Google Scholar] [CrossRef] [Green Version]
- Sasabe, J.; Miyoshi, Y.; Rakoff-Nahoum, S.; Zhang, T.; Mita, M.; Davis, B.M.; Hamase, K.; Waldor, M.K. Interplay between microbial d-amino acids and host d-amino acid oxidase modifies murine mucosal defence and gut microbiota. Nat. Microbiol. 2016, 1, 16125. [Google Scholar] [CrossRef]
- Nakade, Y.; Iwata, Y.; Furuichi, K.; Mita, M.; Hamase, K.; Konno, R.; Miyake, T.; Sakai, N.; Kitajima, S.; Toyama, T.; et al. Gut microbiota-derived D-serine protects against acute kidney injury. JCI Insight 2018, 3, e97957. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Wang, Z.; Roberts, A.B.; Buffa, J.A.; Levison, B.S.; Zhu, W.; Org, E.; Gu, X.; Huang, Y.; Zamanian-Daryoush, M.; Culley, M.K.; et al. Non-lethal inhibition of gut microbial trimethylamine production for the treatment of atherosclerosis. Cell 2015, 163, 1585–1595. [Google Scholar] [CrossRef] [Green Version]
- Vandeputte, D.; Kathagen, G.; D’hoe, K.; Vieira-Silva, S.; Valles-Colomer, M.; Sabino, J.; Wang, J.; Tito, R.Y.; De Commer, L.; Darzi, Y.; et al. Quantitative microbiome profiling links gut community variation to microbial load. Nature 2017, 551, 507–511. [Google Scholar] [CrossRef]
- Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.L.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Abedin-Do, A.; Taherian-Esfahani, Z.; Ghafouri-Fard, S.; Ghafouri-Fard, S.; Motevaseli, E. Immunomodulatory effects of Lactobacillus strains: Emphasis on their effects on cancer cells. Immunotherapy 2015, 7, 1307–1329. [Google Scholar] [CrossRef] [PubMed]
- Atarashi, K.; Tanoue, T.; Oshima, K.; Suda, W.; Nagano, Y.; Nishikawa, H.; Fukuda, S.; Saito, T.; Narushima, S.; Hase, K.; et al. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature 2013, 500, 232–236. [Google Scholar] [CrossRef] [PubMed]
- Macchione, I.G.; Lopetuso, L.R.; Ianiro, G.; Napoli, M.; Gibiino, G.; Rizzatti, G.; Petito, V.; Gasbarrini, A.; Scaldaferri, F. Akkermansia muciniphila: Key player in metabolic and gastrointestinal disorders. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 8075–8083. [Google Scholar]
- Depommier, C.; Everard, A.; Druart, C.; Plovier, H.; Van Hul, M.; Vieira-Silva, S.; Falony, G.; Raes, J.; Maiter, D.; Delzenne, N.M.; et al. Supplementation with Akkermansia muciniphila in overweight and obese human volunteers: A proof-of-concept exploratory study. Nat. Med. 2019, 25, 1096–1103. [Google Scholar] [CrossRef] [PubMed]
- Benítez-Páez, A.; Gómez Del Pugar, E.M.; López-Almela, I.; Moya-Pérez, Á.; Codoñer-Franch, P.; Sanz, Y. Depletion of Blautia species in the microbiota of obese children relates to intestinal inflammation and metabolic phenotype worsening. mSystems 2020, 5, e00857-19. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sokol, H.; Pigneur, B.; Watterlot, L.; Lakhdari, O.; Bermúdez-Humarán, L.G.; Gratadoux, J.J.; Blugeon, S.; Bridonneau, C.; Furet, J.P.; Corthier, G.; et al. Faecalibacterium prausnitzii is an anti-inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc. Natl. Acad. Sci. USA 2008, 105, 16731–16736. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chen, D.; Jin, D.; Huang, S.; Wu, J.; Xu, M.; Liu, T.; Dong, W.; Liu, X.; Wang, S.; Zhong, W.; et al. Clostridium butyricum, a butyrate-producing probiotic, inhibits intestinal tumor development through modulating Wnt signaling and gut microbiota. Cancer Lett. 2020, 469, 456–467. [Google Scholar] [CrossRef]
- Gibson, G.R.; Roberfroid, M.B. Dietary modulation of the human colonic microbiota: Introducing the concept of prebiotics. J. Nutr. 1995, 125, 1401–1412. [Google Scholar] [CrossRef]
- Holscher, H.D. Dietary fiber and prebiotics and the gastrointestinal microbiota. Gut Microbes 2017, 8, 172–184. [Google Scholar] [CrossRef]
- Korczak, R.; Kamil, A.; Fleige, L.; Donovan, S.M.; Slavin, J.L. Dietary fiber and digestive health in children. Nutr. Rev. 2017, 75, 241–259. [Google Scholar] [CrossRef] [PubMed]
- Fuller, S.; Beck, E.; Salman, H.; Tapsell, L. New horizons for the study of dietary fiber and health: A review. Plant. Foods Hum. Nutr. 2016, 71, 1–12. [Google Scholar] [CrossRef] [PubMed]
- Zheng, B.; Shen, H.; Han, H.; Han, T.; Qin, Y. Dietary fiber intake and reduced risk of ovarian cancer: A meta-analysis. Nutr. J. 2018, 17, 99. [Google Scholar] [CrossRef] [PubMed]
- Gill, S.K.; Rossi, M.; Bajka, B.; Whelan, K. Dietary fibre in gastrointestinal health and disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 101–116. [Google Scholar] [CrossRef]
- Hijová, E.; Bertková, I.; Štofilová, J. Dietary fibre as prebiotics in nutrition. Cent. Eur. J. Public Health 2019, 27, 251–255. [Google Scholar] [CrossRef] [Green Version]
- Csernus, B.; Czeglédi, L. Physiological, antimicrobial, intestine morphological, and immunological effects of fructooligosaccharides in pigs. Arch. Anim. Breed. 2020, 63, 325–335. [Google Scholar] [CrossRef]
- Scalabrin, D.M.; Mitmesser, S.H.; Welling, G.W.; Harris, C.L.; Marunycz, J.D.; Walker, D.C.; Bos, N.A.; Tölkkö, S.; Salminen, S.; Vanderhoof, J.A. New prebiotic blend of polydextrose and galacto-oligosaccharides has a bifidogenic effect in young infants. J. Pediatr. Gastroenterol. Nutr. 2012, 54, 343–352. [Google Scholar] [CrossRef]
- Marizzoni, M.; Cattaneo, A.; Mirabelli, P.; Festari, C.; Lopizzo, N.; Nicolosi, V.; Mombelli, E.; Mazzelli, M.; Luongo, D.; Naviglio, D.; et al. Short-Chain fatty acids and lipopolysaccharide as mediators between gut dysbiosis and amyloid pathology in Alzheimer’s disease. J. Alzheimers Dis. 2020, 78, 683–697. [Google Scholar] [CrossRef] [PubMed]
- Capuano, E.; Dekker, M.; Verkerk, R.; Oliviero, T. Food as pharma? The case of glucosinolates. Curr. Pharm. Des. 2017, 23, 2697–2721. [Google Scholar] [CrossRef]
- Zhu, P.; Yu, H.; Zhou, K.; Bai, Y.; Qi, R.; Zhang, S. 3,3′-Diindolylmethane modulates aryl hydrocarbon receptor of esophageal squamous cell carcinoma to reverse epithelial-mesenchymal transition through repressing RhoA/ROCK1-mediated COX2/PGE(2) pathway. J. Exp. Clin. Cancer Res. 2020, 39, 113. [Google Scholar] [CrossRef]
- Zhu, P.; Zhou, K.; Lu, S.; Bai, Y.; Qi, R.; Zhang, S. Modulation of aryl hydrocarbon receptor inhibits esophageal squamous cell carcinoma progression by repressing COX2/PGE2/STAT3 axis. J. Cell. Commun. Signal. 2020, 14, 175–192. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, Y.; Imamura, R.; Koyama, Y.; Kondo, M.; Kobayashi, H.; Nonomura, N.; Shimada, S. Renoprotective and neuroprotective effects of enteric hydrogen generation from Si-based agent. Sci. Rep. 2020, 10, 5859. [Google Scholar] [CrossRef] [Green Version]
- Imamura, R.; Kawamura, M.; Taniguchi, A.; Kobayashi, Y.; Nakazawa, S.; Kato, T.; Abe, T.; Uemura, M.; Kobayashi, H.; Nonomura, N. Efficacy of a Si-based agent against developing renal failure in a rat remnant kidney model. Biochem. Biophys. Res. Commun. 2020, 533, 698–703. [Google Scholar] [CrossRef] [PubMed]
- Tohyama, K.; Kobayashi, Y.; Kan, T.; Yazawa, K.; Tereshima, T.; Mutani, M. Effect of Lactobacilli on urinary indicant excretion in gnotobiotic rats and in man. Microbiol. Immunol. 1981, 25, 101–112. [Google Scholar] [CrossRef] [PubMed]
- Nakanishi, M.; Ohki, M.; Mukai, J.; Uchida, K.; Harano, Y. Development of simple measurement method of urinary indoxyl sulfate reflecting intestinal environment. J. Jpn. Mibyou Syst. Assoc. 2017, 23, 1–5. [Google Scholar]
- Johnson, D.; Thurairajasingam, S.; Letchumanan, V.; Chan, K.-G.; Lee, L.-H. Exploring the role and potential of probiotics in the field of mental health: Major depressive disorder. Nutrients 2021, 13, 1728. [Google Scholar] [CrossRef]
- Lee, J.-Y.; Chu, S.-H.; Jeon, J.Y.; Lee, M.-K.; Park, J.-H.; Lee, D.-C.; Lee, J.-W.; Kim, N.-K. Effects of 12 weeks of probiotic supplementation on quality of life in colorectal cancer survivors: A double-blind, randomized, placebo-controlled trial. Dig. Liver Dis. 2014, 46, 1126–1132. [Google Scholar] [CrossRef]
- Szulińska, M.; Łoniewski, I.; Van Hemert, S.; Sobieska, M.; Bogdański, P. Dose-dependent effects of multispecies probiotic supplementation on the lipopolysaccharide (LPS) level and cardiometabolic profile in obese postmenopausal women: A 12-week randomized clinical trial. Nutrients 2018, 10, 773. [Google Scholar] [CrossRef] [Green Version]
- Chao, L.; Liu, C.; Sutthawongwadee, S.; Li, Y.; Lv, W.; Chen, W.; Yu, L.; Zhou, J.; Guo, A.; Li, Z. Effects of probiotics on depressive or anxiety variables in healthy participants under stress conditions or with a depressive or anxiety diagnosis: A meta-analysis of randomized controlled trials. Front. Neurol. 2020, 11, 421. [Google Scholar] [CrossRef]
- Antushevich, H. Fecal microbiota transplantation in disease therapy. Clin. Chim. Acta 2020, 503, 90–98. [Google Scholar] [CrossRef]
- Baruch, E.N.; Youngster, I.; Ben-Betzalel, G.; Ortenberg, R.; Lahat, A.; Katz, L.; Adler, K.; Dick-Necula, D.; Raskin, S.; Bloch, N.; et al. Fecal microbiota transplant promotes response in immunotherapy-refractory melanoma patients. Science 2021, 371, 602–609. [Google Scholar] [CrossRef]
- Wang, Y.; Wiesnoski, D.H.; Helmink, B.A.; Gopalakrishnan, V.; Choi, K.; DuPont, H.L.; Jiang, Z.D.; Abu-Sbeih, H.; Sanchez, C.A.; Chang, C.C.; et al. Fecal microbiota transplantation for refractory immune checkpoint inhibitor-associated colitis. Nat. Med. 2018, 24, 1804–1808. [Google Scholar] [CrossRef] [PubMed]
- Bárcena, C.; Valdés-Mas, R.; Mayoral, P.; Garabaya, C.; Durand, S.; Rodríguez, F.; Fernández-García, M.T.; Salazar, N.; Nogacka, A.M.; Garatachea, N.; et al. Healthspan and lifespan extension by fecal microbiota transplantation into progeroid mice. Nat. Med. 2019, 25, 1234–1242. [Google Scholar] [CrossRef] [PubMed]
- Van Nood, E.; Vrieze, A.; Nieudorp, M.; Fuentes, S.; Zoetendal, E.G.; de Vos, W.M.; Visser, C.E.; Kuijper, E.J.; Bartelsman, J.F.; Tijssen, J.G.; et al. Duodenal infusion of donor feces for recurrent Clostridium difficile. N. Engl. J. Med. 2013, 368, 407–415. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Xue, L.J.; Yang, X.Z.; Tong, Q.; Shen, P.; Ma, S.J.; Wu, S.N.; Zheng, J.L.; Wang, H.G. Fecal microbiota transplantation therapy for Parkinson’s disease: A preliminary study. Medicine 2020, 99, e22035. [Google Scholar] [CrossRef] [PubMed]
- Sokol, H.; Landman, C.; Seksik, P.; Berard, L.; Montil, M.; Nion-Larmurier, I.; Bourrier, A.; Le Gall, G.; Lalande, V.; De Rougemont, A.; et al. Fecal microbiota transplantation to maintain remission in Crohn’s disease: A pilot randomized controlled study. Microbiome 2020, 8, 12. [Google Scholar] [CrossRef]
- Chen, D.; Wu, J.; Jin, D.; Wang, B.; Cao, H. Fecal microbiota transplantation in cancer management: Current status and perspectives. Int. J. Cancer 2019, 145, 2021–2031. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Shimizu, S.; Dan, K.; Tanaka, C.; Tanaka, M.; Tanaka, Y.; Shirotani, M.; Kitamura, K.; Yorozu, K.; Oehorumu, M.; Tsukamoto, G. Ultrafine bubble water usefulness in fecal microbiota transplantation: Recognition of transplanted microbiota in intestinal epithelial cells. Bioact. Compd. Health Dis. 2020, 3, 141–153. [Google Scholar] [CrossRef]
- Routy, B.; Le Chatelier, E.; Derosa, L.; Duong, C.P.M.; Alou, M.T.; Daillère, R.; Fluckiger, A.; Messaoudene, M.; Rauber, C.; Roberti, M.P.; et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science 2018, 359, 91–97. [Google Scholar] [CrossRef] [Green Version]
- 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] [Green Version]
- Integrative HMP(iHMP) Research Network Consortium. The Integrative Human Microbiome Project. Nature 2019, 569, 641–648. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Rapp, K.M.; Jenkins, J.P.; Betenbaugh, M.J. Partners for life: Building microbial consortia for the future. Curr. Opin. Biotechnol. 2020, 66, 292–300. [Google Scholar] [CrossRef] [PubMed]
Profile (Age, Sex, Cancer, Stage) | Before (μM/g Creatinine) | After (μM/g Creatinine) | |
---|---|---|---|
1 | 42, F, ovarian, I | 333 | 278 |
2 | 70, F, breast, II | 622 | 489 |
3 | 73, M, oropharyngeal, III | 326 | 482 |
4 | 58, F, colorectal, III | 350 | 180 |
5 | 50, F, melanoma, III | 405 | 367 |
6 | 43, F, breast, III | 247 | 350 |
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2021 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
Tanaka, Y.; Shimizu, S.; Shirotani, M.; Yorozu, K.; Kitamura, K.; Oehorumu, M.; Kawai, Y.; Fukuzawa, Y. Nutrition and Cancer Risk from the Viewpoint of the Intestinal Microbiome. Nutrients 2021, 13, 3326. https://doi.org/10.3390/nu13103326
Tanaka Y, Shimizu S, Shirotani M, Yorozu K, Kitamura K, Oehorumu M, Kawai Y, Fukuzawa Y. Nutrition and Cancer Risk from the Viewpoint of the Intestinal Microbiome. Nutrients. 2021; 13(10):3326. https://doi.org/10.3390/nu13103326
Chicago/Turabian StyleTanaka, Yoshimu, Shin Shimizu, Masahiko Shirotani, Kensho Yorozu, Kunihiro Kitamura, Masayuki Oehorumu, Yuichi Kawai, and Yoshitaka Fukuzawa. 2021. "Nutrition and Cancer Risk from the Viewpoint of the Intestinal Microbiome" Nutrients 13, no. 10: 3326. https://doi.org/10.3390/nu13103326