Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 -- 2186 2023-04-13 19:01:52 |
2 format correct + 4 word(s) 2190 2023-04-14 03:39:04 | |
3 format correct -4 word(s) 2186 2023-04-14 03:40:51 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Pellegrino, A.; Coppola, G.; Santopaolo, F.; Gasbarrini, A.; Ponziani, F.R. Role of Akkermansia in IBD and Cancer. Encyclopedia. Available online: https://encyclopedia.pub/entry/43044 (accessed on 28 March 2024).
Pellegrino A, Coppola G, Santopaolo F, Gasbarrini A, Ponziani FR. Role of Akkermansia in IBD and Cancer. Encyclopedia. Available at: https://encyclopedia.pub/entry/43044. Accessed March 28, 2024.
Pellegrino, Antonio, Gaetano Coppola, Francesco Santopaolo, Antonio Gasbarrini, Francesca Romana Ponziani. "Role of Akkermansia in IBD and Cancer" Encyclopedia, https://encyclopedia.pub/entry/43044 (accessed March 28, 2024).
Pellegrino, A., Coppola, G., Santopaolo, F., Gasbarrini, A., & Ponziani, F.R. (2023, April 13). Role of Akkermansia in IBD and Cancer. In Encyclopedia. https://encyclopedia.pub/entry/43044
Pellegrino, Antonio, et al. "Role of Akkermansia in IBD and Cancer." Encyclopedia. Web. 13 April, 2023.
Role of Akkermansia in IBD and Cancer
Edit

Akkermansia muciniphila (A. muciniphila) represents approximately 1–3% of the total gut microbiota in healthy people; it is a non-motile, Gram-negative, non-spore-forming, oval-shaped bacterium belonging to the Verrucomicrobia phylum, and it is the first and only member of the phylum Verrucomicrobia found in the human gut. Its key features are the ability to produce short-chain fatty acids (SCFAs, energy source for colonocytes and anti-inflammatory molecules), to promote mucin turnover and thickening thereby reinforcing the intestinal barrier and to interact with host receptors with its exposed active molecules thus influencing inflammation and metabolism. A. muciniphila can be used as a biomarker of a healthy host metabolic profile and that its depletion represents a signature of intestinal dysbiosis across different gastrointestinal and extraintestinal diseases as inflammatory bowel disease and some cancer types. The molecular mechanism beneath the action of this bacteria in the abovementioned diseases and how A. Muciniphila can modulate the response to both conventional and targeted cancer therapy are explored. 

gut IBD Akkermansia muciniphila cancer cancer therapy immunotherapy

1. Introduction

Akkermansia muciniphila (A. muciniphila) represents approximately 1–3% of the total gut microbiota in healthy people; it is a non-motile, Gram-negative, non-spore-forming, oval-shaped bacterium belonging to the Verrucomicrobia phylum, and it is the first and only member of the phylum Verrucomicrobia found in the human gut [1][2][3]. A. muciniphila was originally considered a strict anaerobe, but it was recently proved to tolerate small amounts of oxygen and was therefore reclassified as an aerotolerant anaerobe [4].One of the distinguishing features of A. muciniphila is its ability to degrade intestinal mucin glycoproteins via various enzymes and to use them as a sole source of carbon and nitrogen; this process leads to the production of short-chain fatty acids (SCFAs). Due to this process of degradation, A. muciniphila promotes mucin turnover and thickening, thereby reinforcing the intestinal barrier and reducing gut permeability to microbial products. A further barrier-reinforcing mechanism is the A. muciniphila-induced production of antimicrobial peptides from Paneth cells. SCFAs derived from gut mucin glycoproteins are absorbed in the colon and serve as an energy source for colonocytes, inducing regulatory T cells and exerting anti-inflammatory effects [4][5][6][7][8].
SCFAs are subsequently used by other bacteria in the gut microbial community, such as Anaerostipes caccae, Anaerobutyricum hallii, and Faecalibacterium prausnitzii, to further produce butyrate and propionate [1][6][9][10][11].
In addition to metabolites, the effects of A. muciniphila are mediated by exposed active molecules; among these, Amuc_1100 (an outer membrane protein involved in pili formation) can replicate almost all of the effects of live A. muciniphila through Toll-like receptor 2 (TLR2) sensing [12][13][14]. TLRs are expressed by a wide range of immune, epithelial, and endothelial cells whose main role is the recognition of microbial structures, capable of stimulating pro- and anti-inflammatory responses with further implications in the regulation of host metabolism [14][15]. The heat stability of these proteins explains why A. muciniphila retains most of its effects even after pasteurization. In 2021, the safety of pasteurized A. muciniphila was positively assessed by the European Food Safety Authority (EFSA) and the Panel on Nutrition, Novel Foods and Food Allergens (NDA) [16], and its production represents the beginning of the new generation of probiotics [17].
Due to its many beneficial effects, it is not surprising that A. muciniphila can be used as a biomarker of a healthy host metabolic profile, and that its depletion represents a signature of intestinal dysbiosis across different gastrointestinal and extraintestinal diseases. A reduced abundance of A. muciniphila in the gut microbial community has been related in fact to several diseases, such as obesity, type 2 diabetes, inflammatory bowel disease and some cancer types; conversely, the administration of live A. muciniphila has shown a protective role even in the pathogenesis of cardiovascular disease in mice [18][19][20][21].

2. Akkermansia muciniphila and Inflammatory Bowel Diseases

The gut microbiota plays an undeniable role in the pathogenesis of inflammatory bowel diseases (IBD), and the modulation of the gut microbiota represents one of the most promising challenges in IBD therapy [22][23][24][25][26].
Many case–control studies have documented a significant decrease in the relative abundance of A. muciniphila both in ulcerative colitis (UC) and Crohn’s disease (CD) compared to healthy controls [27][28][29][30], with only one study showing an opposite trend in a group of patients affected by CD [31].
As previously mentioned, A. muciniphila exerts an anti-inflammatory effect within the intestinal microecology. Among the underlying mechanisms proposed, the production of SCFAs is the most deeply investigated; the production of SCFAs has been demonstrated to protect against colitis by increasing the number of forkhead box P3 (Foxp3+) regulatory T cells in the colon and through the activation of the G-protein coupled receptor 43 (GPR43) expressed by immune cells and colonic epithelium [32][33][34]. Wang et al. observed that the administration of A. muciniphila could improve dextran sulfate sodium (DSS)-induced colitis in mice by reducing macrophage and CD8+ cytotoxic T lymphocyte levels in the colon [35], while Bian et al. reported a downregulation of pro-inflammatory cytokines and chemokines [36]. Additionally, the administration of A. muciniphila enhances intestinal stem cell proliferation and Paneth and goblet cell differentiation in the small intestine and colon of both healthy mice and mice with gut damage [37].
A. muciniphila also restored the mRNA expression of tight junction proteins such as zonulin-1, occludin, and claudin-1 in mouse models of DSS-induced colitis, thereby reducing gut permeability and reshaping the intestinal microbiota, leading it toward eubiosis; these effects are related to the administration of Amuc:2109, a β-acetylaminohexosidase secreted by this microorganism [38].
However, an increased abundance of A. muciniphila was reported in some preclinical models of gut inflammation [39][40][41]. Interestingly, when administered to mice with non-DSS-induced colitis, A. muciniphila was associated with symptoms worsening; A. muciniphila administration also exacerbated the symptoms of Salmonella-typhimurium-induced gut inflammation in a mouse model with a background microbiota of eight bacterial species [42], and it was possibly implicated in the worsening of colitis in IL10 −/− mice.
The discrepancy in the effects of this bacterial species could allow for several interpretations, being possibly biased by the different mouse models used; moreover, it can be speculated that the increased abundance of A. muciniphila in colitis models could represent a causative factor or rather, a reactive response. When A. muciniphila was administered in the IL-10 −/− mice colonized with a simplified human gut microbiota, it did not promote inflammation, suggesting that other environmental conditions could be involved [43][44].
Finally, there are few studies on the predictive effect of Akkermansia after FMT in patients with IBD. Zhang et al. demonstrated that washed microbiota transplantation (WMT) significantly increased the colonization rate of Akkermansia and that there was a positive correlation between the abundance of patient’s and donor’s Akkermansia abundance after WMT, speculating its possible role as a predictive factor of WMT efficacy [27]. Similar results were obtained by Kump et al. in treatment-refractory patients with UC; indeed, the stool of donors with a higher bacterial richness and a higher relative abundance of A. muciniphila, Ruminococcaceae, and Ruminococcus spp. were more likely to induce remission in these patients. In particular, A. muciniphila was nearly absent in baseline samples but was significantly increased the day after FMT in patients achieving remission [45].
In conclusion, current evidence, although conflicting to some degree, paves the way for a potential role of A. muciniphila in IBD treatment [46].

3. Akkermansia muciniphila and Cancer

Colorectal cancer (CRC) is one of the most common and lethal cancers in the world. Although obesity, Western dietary habits, smoking, and heavy alcohol consumption are the better-known risk factors for CRC, the intestinal environment has also received widespread attention in this field. It has been demonstrated in humans and in animal models that gut dysbiosis may promote colon carcinogenesis via multiple mechanisms, including the development of chronic inflammation and the production of genotoxins and other microbial products [47][48][49][50].
A. muciniphila depletion is a feature of CRC-associated dysbiosis. In models of colitis-associated CRC (CAC), the administration of pasteurized A. muciniphila or Amuc_1100 alone improved symptoms, delayed tumor development, and decreased the number and area of tumor lesions by attenuating DNA damage, cell apoptosis, and abnormal proliferation; the beneficial effects of A. muciniphila were associated with the expansion of cytotoxic T-lymphocytes in the colon and mesenteric lymph nodes and with the modulation of macrophages subpopulations, thus explaining how A. muciniphila influences inflammation-associated tumorigenesis [35].
Another study further confirmed that the abundance of A. muciniphila is significantly reduced in humans with CRC and that its supplementation can inhibit colonic tumorigenesis in ApcMin/+ mice via the expansion of M1-like macrophages in colonic tissue. Tumor-associated macrophages (TAMs) can assume a pro-inflammatory polarization (M1) or an anti-inflammatory polarization (M2), with only the former helping to suppress cancer cells. Even this effect is mediated by the interaction between A. muciniphila and the TLR2 expressed by macrophages [51][52][53].
Apart from its immunomodulatory effects, A. muciniphila can also directly interfere with colon carcinogenesis through the production of Amuc_1434, an enzyme that can degrade Mucin2, the main component of the intestinal mucus layer, which is highly expressed in mucinous CRC. Amuc_1434 showed a protective effect on tumor protein 53 (p53) expression in vitro, resulting in the blockade of the G0/G1 cell cycle phase and the promotion of CRC cells apoptosis [54][55].
Conversely, it was observed that A. muciniphila abundance was increased in two different cohorts of patients affected by CRC, as well as in a cohort of patients with esophageal and gastric cancers compared with healthy controls [56][57]. However, according to Weil et al., this observation can be related to an increased substrate availability rather than to a detrimental role of this bacterium, considering the overexpression of MUC1 and MUC5AC in CRCs [57].
Besides CRC, A. muciniphila was found to be more abundant in patients with non-small-cell lung cancer (NSCLC) and to gradually decrease during the progression from cirrhosis to hepatocellular carcinoma [58][59][60].
In recent years, gut microbiota modulation applied to cancer therapy is certainly a topic of growing interest in either treatment efficacy or tolerability [61][62][63].
There are some data regarding the possible role of A. muciniphila in both conventional and targeted anticancer therapy. For instance, A. muciniphila could improve the antitumor effect of cisplatin; in mouse models of lung cancer, the administration of A. muciniphila in combination with cis-diamminedichloroplatinum (CDDP) was associated with reduced tumor growth, the downregulation of ki-67, p53, factor-associated suicide (Fas) ligand proteins, and the upregulation of Fas proteins [61]. Moreover, the administration of A. muciniphila in this setting increased the production of pro-inflammatory cytokines and suppressed the development of T-reg lymphocytes, thus suggesting that it could modulate the immune microenvironment toward an inflammatory response, counteracting tumor immune escape. A. muciniphila has been proven to enhance the antitumor efficacy of interleukin (IL)-2; in murine models of melanoma and CRC, the combined administration of IL-2 and A. muciniphila reduced the tumor burden and improved survival compared with IL-2 treatment alone, primarily by stimulating the response of CD4+ and CD8+ T cells against cancer cells and by decreasing the number and the activity of T-regs. These beneficial effects were at least partially mediated by the interaction between a specific membrane protein and TLR2 [64]. The current literature also shows a peculiar interplay between A. muciniphila and abiraterone acetate (AA), an inhibitor of androgen biosynthesis for the treatment of prostate cancer (PCa) refractory to androgen deprivation therapy (ADT). In a cohort of PCa patients, AA administration increased the abundance of A. muciniphila, with this effect probably resulting from the interaction between the conjugated acetate portion of AA and A. muciniphila [65][66][67]. Unfortunately, the authors did not explore the contribution of A. muciniphila to the efficacy of AA. In a later study, the intravenous administration of A. muciniphila-derived extracellular vesicles in PCa-bearing, immune-competent mice operated as an immune modulator; it was associated with the increased activation of CD8+ T cells and tumor-killing M1 macrophages, resulting in a reduced tumor mass [68].
To further clarify the role of Akkermansia in cancer immunotherapy, Xu et al. evaluated the effects of the modulation of the gut microbiome on the response to immune checkpoint inhibitors (ICIs). In CRC mouse models, the exposure to several broad-spectrum antibiotics interfered with the efficacy of programmed cell death protein 1 (PD-1) antibodies, depending on the type of antibiotic and the resulting changes in the gut microbiota composition. A. muciniphila was found to be enriched in the vancomycin-treated group and associated with a better outcome; according to the authors, Akkermansia could preserve the efficacy of anti-PD-1 therapy by modulating the metabolism of glycerophospholipids, which influence the expression of immune-related cytokines IFN-γ and IL-2 in the tumor microenvironment [69]. Other published studies in patients with hepatocellular carcinoma and melanoma highlighted the contribution of the gut microbiome to the response to immunotherapy, and A. muciniphila emerged as a key element associated with treatment efficacy [70][71][72][73][74].
Finally, in patients with NSCLC and renal cell carcinoma (RCC) undergoing immunotherapy with ICIs, FMT from treatment responders to germ-free mice resulted in increased efficacy of immunotherapy. A. muciniphila was found to be more abundant and associated with treatment response, and the oral administration of A. muciniphila improved PD-1 blockade effectiveness, once again through the modulation of the immune response, specifically by promoting the recruitment of CD4+ T cells [70].
In conclusion, even in the setting of the modulation of the response to cancer therapy, current evidence, although limited, shows a promising role for A. muciniphila. Further studies are needed to clarify its potential in this field. 

References

  1. Derrien, M.; Vaughan, E.E.; Plugge, C.M.; de Vos, W.M. Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int. J. Syst. Evol. Microbiol. 2004, 54, 1469–1476.
  2. Ropot, A.V.; Karamzin, A.M.; Sergeyev, O.V. Cultivation of the Next-Generation Probiotic Akkermansia muciniphila, Methods of Its Safe Delivery to the Intestine, and Factors Contributing to Its Growth In Vivo. Curr. Microbiol. 2020, 77, 1363–1372.
  3. Zhang, T.; Li, Q.; Cheng, L.; Buch, H.; Zhang, F. Akkermansia muciniphila is a promising probiotic. Microb. Biotechnol. 2019, 12, 1109–1125.
  4. Reunanen, J.; Kainulainen, V.; Huuskonen, L.; Ottman, N.; Belzer, C.; Huhtinen, H.; de Vos, W.M.; Satokari, R. Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity of the Epithelial Cell Layer. Appl. Environ. Microbiol. 2015, 81, 3655–3662.
  5. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071.
  6. Belzer, C.; de Vos, W.M. Microbes inside—From diversity to function: The case of Akkermansia. ISME J. 2012, 6, 1449–1458.
  7. Ottman, N.; Geerlings, S.Y.; Aalvink, S.; de Vos, W.M.; Belzer, C. Action and function of Akkermansia muciniphila in microbiome ecology, health and disease. Best Pract. Res. Clin. Gastroenterol. 2017, 31, 637–642.
  8. Brodmann, T.; Endo, A.; Gueimonde, M.; Vinderola, G.; Kneifel, W.; de Vos, W.M.; Salminen, S.; Gómez-Gallego, C. Safety of novel microbes for human consumption: Practical examples of assessment in the European Union. Front. Microbiol. 2017, 8, 1725.
  9. Derrien, M.; Belzer, C.; de Vos, W.M. Akkermansia muciniphila and its role in regulating host functions. Microb. Pathog. 2017, 106, 171–181.
  10. van Passel, M.W.J.; Kant, R.; Zoetendal, E.G.; Plugge, C.M.; Derrien, M.; Malfatti, S.A.; Chain, P.S.G.; Woyke, T.; Palva, A.; de Vos, W.M.; et al. The genome of Akkermansia muciniphila, a dedicated intestinal mucin degrader, and its use in exploring intestinal metagenomes. PLoS ONE 2011, 6, e16876.
  11. van der Hee, B.; Wells, J.M. Microbial Regulation of Host Physiology by Short-chain Fatty Acids. Trends Microbiol. 2021, 29, 700–712.
  12. Kang, C.-S.; Ban, M.; Choi, E.-J.; Moon, H.-G.; Jeon, J.-S.; Kim, D.-K.; Park, S.-K.; Jeon, S.G.; Roh, T.-Y.; Myung, S.-J.; et al. Extracellular vesicles derived from gut microbiota, especially Akkermansia muciniphila, protect the progression of dextran sulfate sodium-induced colitis. PLoS ONE 2013, 8, e76520.
  13. Chelakkot, C.; Choi, Y.; Kim, D.-K.; Park, H.T.; Ghim, J.; Kwon, Y.; Jeon, J.; Kim, M.-S.; Jee, Y.-K.; Gho, Y.S.; et al. Akkermansia muciniphila-derived extracellular vesicles influence gut permeability through the regulation of tight junctions. Exp. Mol. Med. 2018, 50, e450.
  14. Plovier, H.; Everard, A.; Druart, C.; Depommier, C.; Van Hul, M.; Geurts, L.; Chilloux, J.; Ottman, N.; Duparc, T.; Lichtenstein, L.; et al. A purified membrane protein from Akkermansia muciniphila or the pasteurized bacterium improves metabolism in obese and diabetic mice. Nat. Med. 2017, 23, 107–113.
  15. Di Lorenzo, A.; Bolli, E.; Tarone, L.; Cavallo, F.; Conti, L. Toll-Like Receptor 2 at the Crossroad between Cancer Cells, the Immune System, and the Microbiota. Int. J. Mol. Sci. 2020, 21, 9418.
  16. EFSA Panel on Nutrition; Novel Foods and Food Allergens (NDA); Turck, D.; Bohn, T.; Castenmiller, J.; De Henauw, S.; Hirsch-Ernst, K.I.; Maciuk, A.; Mangelsdorf, I.; McArdle, H.J.; et al. Safety of pasteurised Akkermansia muciniphila as a novel food pursuant to Regulation (EU) 2015/2283. EFSA J. 2021, 19, e06780.
  17. Iwaza, R.; Wasfy, R.M.; Dubourg, G.; Raoult, D.; Lagier, J.-C. Akkermansia muciniphila: The state of the art, 18 years after its first discovery. Front. Gastroenterol. 2022, 1.
  18. Saarela, M.H. Safety aspects of next generation probiotics. Current Opinion in Food Science 2019, 30, 8–13.
  19. Lopetuso, L.R.; Quagliariello, A.; Schiavoni, M.; Petito, V.; Russo, A.; Reddel, S.; Del Chierico, F.; Ianiro, G.; Scaldaferri, F.; Neri, M.; et al. Towards a disease-associated common trait of gut microbiota dysbiosis: The pivotal role of Akkermansia muciniphila. Dig. Liver Dis. 2020, 52, 1002–1010.
  20. Almeida, D.; Machado, D.; Andrade, J.C.; Mendo, S.; Gomes, A.M.; Freitas, A.C. Evolving trends in next-generation probiotics: A 5W1H perspective. Crit. Rev. Food Sci. Nutr. 2020, 60, 1783–1796.
  21. Li, J.; Lin, S.; Vanhoutte, P.M.; Woo, C.W.; Xu, A. Akkermansia Muciniphila Protects Against Atherosclerosis by Preventing Metabolic Endotoxemia-Induced Inflammation in Apoe-/- Mice. Circulation 2016, 133, 2434–2446.
  22. Lee, M.; Chang, E.B. Inflammatory Bowel Diseases (IBD) and the Microbiome-Searching the Crime Scene for Clues. Gastroenterology 2021, 160, 524–537.
  23. Liu, S.; Zhao, W.; Lan, P.; Mou, X. The microbiome in inflammatory bowel diseases: From pathogenesis to therapy. Protein Cell 2021, 12, 331–345.
  24. Nishida, A.; Nishino, K.; Sakai, K.; Owaki, Y.; Noda, Y.; Imaeda, H. Can control of gut microbiota be a future therapeutic option for inflammatory bowel disease? World J. Gastroenterol. 2021, 27, 3317–3326.
  25. Zheng, L.; Wen, X.-L. Gut microbiota and inflammatory bowel disease: The current status and perspectives. World J. Clin. Cases 2021, 9, 321–333.
  26. Quaglio, A.E.V.; Grillo, T.G.; De Oliveira, E.C.S.; Di Stasi, L.C.; Sassaki, L.Y. Gut microbiota, inflammatory bowel disease and colorectal cancer. World J. Gastroenterol. 2022, 28, 4053–4060.
  27. Zhang, T.; Li, P.; Wu, X.; Lu, G.; Marcella, C.; Ji, X.; Ji, G.; Zhang, F. Alterations of Akkermansia muciniphila in the inflammatory bowel disease patients with washed microbiota transplantation. Appl. Microbiol. Biotechnol. 2020, 104, 10203–10215.
  28. Lo Presti, A.; Del Chierico, F.; Altomare, A.; Zorzi, F.; Cella, E.; Putignani, L.; Guarino, M.P.L.; Monteleone, G.; Cicala, M.; Angeletti, S.; et al. Exploring the genetic diversity of the 16S rRNA gene of Akkermansia muciniphila in IBD and IBS. Future Microbiol. 2019, 14, 1497–1509.
  29. Lo Sasso, G.; Khachatryan, L.; Kondylis, A.; Battey, J.N.D.; Sierro, N.; Danilova, N.A.; Grigoryeva, T.V.; Markelova, M.I.; Khusnutdinova, D.R.; Laikov, A.V.; et al. Inflammatory Bowel Disease-Associated Changes in the Gut: Focus on Kazan Patients. Inflamm. Bowel Dis. 2021, 27, 418–433.
  30. Png, C.W.; Lindén, S.K.; Gilshenan, K.S.; Zoetendal, E.G.; McSweeney, C.S.; Sly, L.I.; McGuckin, M.A.; Florin, T.H.J. Mucolytic bacteria with increased prevalence in IBD mucosa augment in vitro utilization of mucin by other bacteria. Am. J. Gastroenterol. 2010, 105, 2420–2428.
  31. Danilova, N.A.; Abdulkhakov, S.R.; Grigoryeva, T.V.; Markelova, M.I.; Vasilyev, I.Y.; Boulygina, E.A.; Ardatskaya, M.D.; Pavlenko, A.V.; Tyakht, A.V.; Odintsova, A.K.; et al. Markers of dysbiosis in patients with ulcerative colitis and Crohn’s disease. Ter. Arkh. 2019, 91, 17–24.
  32. Zhai, R.; Xue, X.; Zhang, L.; Yang, X.; Zhao, L.; Zhang, C. Strain-Specific Anti-inflammatory Properties of Two Akkermansia muciniphila Strains on Chronic Colitis in Mice. Front. Cell. Infect. Microbiol. 2019, 9, 239.
  33. Maslowski, K.M.; Vieira, A.T.; Ng, A.; Kranich, J.; Sierro, F.; Yu, D.; Schilter, H.C.; Rolph, M.S.; Mackay, F.; Artis, D.; et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature 2009, 461, 1282–1286.
  34. Smith, P.M.; Howitt, M.R.; Panikov, N.; Michaud, M.; Gallini, C.A.; Bohlooly, -Y.M.; Glickman, J.N.; Garrett, W.S. The microbial metabolites, short-chain fatty acids, regulate colonic Treg cell homeostasis. Science 2013, 341, 569–573.
  35. Wang, L.; Tang, L.; Feng, Y.; Zhao, S.; Han, M.; Zhang, C.; Yuan, G.; Zhu, J.; Cao, S.; Wu, Q.; et al. A purified membrane protein from Akkermansia muciniphila or the pasteurised bacterium blunts colitis associated tumourigenesis by modulation of CD8+ T cells in mice. Gut 2020, 69, 1988–1997.
  36. Bian, X.; Wu, W.; Yang, L.; Lv, L.; Wang, Q.; Li, Y.; Ye, J.; Fang, D.; Wu, J.; Jiang, X.; et al. Administration of Akkermansia muciniphila Ameliorates Dextran Sulfate Sodium-Induced Ulcerative Colitis in Mice. Front. Microbiol. 2019, 10, 2259.
  37. Kim, S.; Shin, Y.-C.; Kim, T.-Y.; Kim, Y.; Lee, Y.-S.; Lee, S.-H.; Kim, M.-N.; Eunjo, O.; Kim, K.S.; Kweon, M.-N. Mucin degrader Akkermansia muciniphila accelerates intestinal stem cell-mediated epithelial development. Gut Microbes 2021, 13, 1–20.
  38. Qian, K.; Chen, S.; Wang, J.; Sheng, K.; Wang, Y.; Zhang, M. A β-N-acetylhexosaminidase Amuc_2109 from Akkermansia muciniphila protects against dextran sulfate sodium-induced colitis in mice by enhancing intestinal barrier and modulating gut microbiota. Food Funct. 2022, 13, 2216–2227.
  39. Håkansson, Å.; Tormo-Badia, N.; Baridi, A.; Xu, J.; Molin, G.; Hagslätt, M.L.; Karlsson, C.; Jeppsson, B.; Cilio, C.M.; Ahrné, S. Immunological alteration and changes of gut microbiota after dextran sulfate sodium (DSS) administration in mice. Clin. Exp. Med. 2015, 15, 107–120.
  40. Chen, H.; Xia, Y.; Zhu, S.; Yang, J.; Yao, J.; Di, J.; Liang, Y.; Gao, R.; Wu, W.; Yang, Y.; et al. Lactobacillus plantarum LP-Onlly alters the gut flora and attenuates colitis by inducing microbiome alteration in interleukin-10 knockout mice. Mol. Med. Report. 2017, 16, 5979–5985.
  41. Zhu, W.; Yan, J.; Zhi, C.; Zhou, Q.; Yuan, X. 1,25(OH)2D3 deficiency-induced gut microbial dysbiosis degrades the colonic mucus barrier in Cyp27b1 knockout mouse model. Gut Pathog. 2019, 11, 8.
  42. Ganesh, B.P.; Klopfleisch, R.; Loh, G.; Blaut, M. Commensal Akkermansia muciniphila exacerbates gut inflammation in Salmonella Typhimurium-infected gnotobiotic mice. PLoS ONE 2013, 8, e74963.
  43. Seregin, S.S.; Golovchenko, N.; Schaf, B.; Chen, J.; Pudlo, N.A.; Mitchell, J.; Baxter, N.T.; Zhao, L.; Schloss, P.D.; Martens, E.C.; et al. NLRP6 Protects Il10-/- Mice from Colitis by Limiting Colonization of Akkermansia muciniphila. Cell Rep. 2017, 19, 733–745.
  44. Ring, C.; Klopfleisch, R.; Dahlke, K.; Basic, M.; Bleich, A.; Blaut, M. Akkermansia muciniphila strain ATCC BAA-835 does not promote short-term intestinal inflammation in gnotobiotic interleukin-10-deficient mice. Gut Microbes 2019, 10, 188–203.
  45. Kump, P.; Wurm, P.; Gröchenig, H.P.; Wenzl, H.; Petritsch, W.; Halwachs, B.; Wagner, M.; Stadlbauer, V.; Eherer, A.; Hoffmann, K.M.; et al. The taxonomic composition of the donor intestinal microbiota is a major factor influencing the efficacy of faecal microbiota transplantation in therapy refractory ulcerative colitis. Aliment. Pharmacol. Ther. 2018, 47, 67–77.
  46. Zhang, T.; Ji, X.; Lu, G.; Zhang, F. The potential of Akkermansia muciniphila in inflammatory bowel disease. Appl. Microbiol. Biotechnol. 2021, 105, 5785–5794.
  47. Siegel, R.L.; Miller, K.D.; Goding Sauer, A.; Fedewa, S.A.; Butterly, L.F.; Anderson, J.C.; Cercek, A.; Smith, R.A.; Jemal, A. Colorectal cancer statistics, 2020. CA Cancer J. Clin. 2020, 70, 145–164.
  48. 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.
  49. Sobhani, I.; Bergsten, E.; Couffin, S.; Amiot, A.; Nebbad, B.; Barau, C.; de’Angelis, N.; Rabot, S.; Canoui-Poitrine, F.; Mestivier, D.; et al. Colorectal cancer-associated microbiota contributes to oncogenic epigenetic signatures. Proc. Natl. Acad. Sci. USA 2019, 116, 24285–24295.
  50. Gao, Z.-Y.; Cui, Z.; Yan, Y.-Q.; Ning, L.-J.; Wang, Z.-H.; Hong, J. Microbe-based management for colorectal cancer. Chin. Med. J. 2021, 134, 2922–2930.
  51. Fan, L.; Xu, C.; Ge, Q.; Lin, Y.; Wong, C.C.; Qi, Y.; Ye, B.; Lian, Q.; Zhuo, W.; Si, J.; et al. Muciniphila Suppresses Colorectal Tumorigenesis by Inducing TLR2/NLRP3-Mediated M1-Like TAMs. Cancer Immunol. Res. 2021, 9, 1111–1124.
  52. Ren, J.; Sui, H.; Fang, F.; Li, Q.; Li, B. The application of ApcMin/+ mouse model in colorectal tumor researches. J. Cancer Res. Clin. Oncol. 2019, 145, 1111–1122.
  53. Yunna, C.; Mengru, H.; Lei, W.; Weidong, C. Macrophage M1/M2 polarization. Eur. J. Pharmacol. 2020, 877, 173090.
  54. Meng, X.; Zhang, J.; Wu, H.; Yu, D.; Fang, X. Akkermansia muciniphila Aspartic Protease Amuc_1434* Inhibits Human Colorectal Cancer LS174T Cell Viability via TRAIL-Mediated Apoptosis Pathway. Int. J. Mol. Sci. 2020, 21, 3385.
  55. Meng, X.; Wang, W.; Lan, T.; Yang, W.; Yu, D.; Fang, X.; Wu, H. A Purified Aspartic Protease from Akkermansia Muciniphila Plays an Important Role in Degrading Muc2. Int. J. Mol. Sci. 2019, 21, 72.
  56. Li, N.; Bai, C.; Zhao, L.; Ge, Y.; Li, X. Characterization of the fecal microbiota in gastrointestinal cancer patients and healthy people. Clin. Transl. Oncol. 2022, 24, 1134–1147.
  57. Weir, T.L.; Manter, D.K.; Sheflin, A.M.; Barnett, B.A.; Heuberger, A.L.; Ryan, E.P. Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults. PLoS ONE 2013, 8, e70803.
  58. Vernocchi, P.; Gili, T.; Conte, F.; Del Chierico, F.; Conta, G.; Miccheli, A.; Botticelli, A.; Paci, P.; Caldarelli, G.; Nuti, M.; et al. Network Analysis of Gut Microbiome and Metabolome to Discover Microbiota-Linked Biomarkers in Patients Affected by Non-Small Cell Lung Cancer. Int. J. Mol. Sci. 2020, 21, 8730.
  59. Lapidot, Y.; Amir, A.; Nosenko, R.; Uzan-Yulzari, A.; Veitsman, E.; Cohen-Ezra, O.; Davidov, Y.; Weiss, P.; Bradichevski, T.; Segev, S.; et al. Alterations in the gut microbiome in the progression of cirrhosis to hepatocellular carcinoma. mSystems 2020, 5, e00153-20.
  60. Ponziani, F.R.; Nicoletti, A.; Gasbarrini, A.; Pompili, M. Diagnostic and therapeutic potential of the gut microbiota in patients with early hepatocellular carcinoma. Ther. Adv. Med. Oncol. 2019, 11, 1758835919848184.
  61. Chen, Z.; Qian, X.; Chen, S.; Fu, X.; Ma, G.; Zhang, A. Akkermansia muciniphila Enhances the Antitumor Effect of Cisplatin in Lewis Lung Cancer Mice. J. Immunol. Res. 2020, 2020, 2969287.
  62. Viaud, S.; Saccheri, F.; Mignot, G.; Yamazaki, T.; Daillère, R.; Hannani, D.; Enot, D.P.; Pfirschke, C.; Engblom, C.; Pittet, M.J.; et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013, 342, 971–976.
  63. Iida, N.; Dzutsev, A.; Stewart, C.A.; Smith, L.; Bouladoux, N.; Weingarten, R.A.; Molina, D.A.; Salcedo, R.; Back, T.; Cramer, S.; et al. Commensal bacteria control cancer response to therapy by modulating the tumor microenvironment. Science 2013, 342, 967–970.
  64. Shi, L.; Sheng, J.; Chen, G.; Zhu, P.; Shi, C.; Li, B.; Park, C.; Wang, J.; Zhang, B.; Liu, Z.; et al. Combining IL-2-based immunotherapy with commensal probiotics produces enhanced antitumor immune response and tumor clearance. J. Immunother. Cancer 2020, 8.
  65. Daisley, B.A.; Chanyi, R.M.; Abdur-Rashid, K.; Al, K.F.; Gibbons, S.; Chmiel, J.A.; Wilcox, H.; Reid, G.; Anderson, A.; Dewar, M.; et al. Abiraterone acetate preferentially enriches for the gut commensal Akkermansia muciniphila in castrate-resistant prostate cancer patients. Nat. Commun. 2020, 11, 4822.
  66. Dao, M.C.; Everard, A.; Aron-Wisnewsky, J.; Sokolovska, N.; Prifti, E.; Verger, E.O.; Kayser, B.D.; Levenez, F.; Chilloux, J.; Hoyles, L.; et al. Akkermansia muciniphila and improved metabolic health during a dietary intervention in obesity: Relationship with gut microbiome richness and ecology. Gut 2016, 65, 426–436.
  67. Alard, J.; Lehrter, V.; Rhimi, M.; Mangin, I.; Peucelle, V.; Abraham, A.-L.; Mariadassou, M.; Maguin, E.; Waligora-Dupriet, A.-J.; Pot, B.; et al. Beneficial metabolic effects of selected probiotics on diet-induced obesity and insulin resistance in mice are associated with improvement of dysbiotic gut microbiota. Environ. Microbiol. 2016, 18, 1484–1497.
  68. Luo, Z.-W.; Xia, K.; Liu, Y.-W.; Liu, J.-H.; Rao, S.-S.; Hu, X.-K.; Chen, C.-Y.; Xu, R.; Wang, Z.-X.; Xie, H. Extracellular Vesicles from Akkermansia muciniphila Elicit Antitumor Immunity Against Prostate Cancer via Modulation of CD8+ T Cells and Macrophages. Int. J. Nanomedicine 2021, 16, 2949–2963.
  69. 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.
  70. 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.
  71. Zheng, Y.; Wang, T.; Tu, X.; Huang, Y.; Zhang, H.; Tan, D.; Jiang, W.; Cai, S.; Zhao, P.; Song, R.; et al. Gut microbiome affects the response to anti-PD-1 immunotherapy in patients with hepatocellular carcinoma. J. Immunother. Cancer 2019, 7, 193.
  72. Matson, V.; Fessler, J.; Bao, R.; Chongsuwat, T.; Zha, Y.; Alegre, M.-L.; Luke, J.J.; Gajewski, T.F. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science 2018, 359, 104–108.
  73. Ponziani, F.R.; De Luca, A.; Picca, A.; Marzetti, E.; Petito, V.; Del Chierico, F.; Reddel, S.; Paroni Sterbini, F.; Sanguinetti, M.; Putignani, L.; et al. Gut dysbiosis and fecal calprotectin predict response to immune checkpoint inhibitors in patients with hepatocellular carcinoma. Hepatol. Commun. 2022, 6, 1492–1501.
  74. Gopalakrishnan, V.; Spencer, C.N.; Nezi, L.; Reuben, A.; Andrews, M.C.; Karpinets, T.V.; Prieto, P.A.; Vicente, D.; Hoffman, K.; Wei, S.C.; et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science 2018, 359, 97–103.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : , , , ,
View Times: 433
Revisions: 3 times (View History)
Update Date: 14 Apr 2023
1000/1000