Next Article in Journal
Fluorescent Light Incites a Conserved Immune and Inflammatory Genetic Response within Vertebrate Organs (Danio rerio, Oryzias latipes and Mus musculus)
Next Article in Special Issue
Special Issue: MicroRNA Regulation in Health and Disease
Previous Article in Journal
Next-Generation Sequencing Enables Spatiotemporal Resolution of Human Centromere Replication Timing
Previous Article in Special Issue
Single Nucleotide Polymorphisms in MIR143 Contribute to Protection against Non-Hodgkin Lymphoma (NHL) in Caucasian Populations
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Host–MicroRNA–Microbiota Interactions in Colorectal Cancer

1
Bioinformatics and Computational Biology Program, University of Minnesota, Minneapolis, MN 55455, USA
2
Department of Surgery, University of Minnesota Medical School, Minneapolis, MN 55455, USA
3
Department of Medicine, University of Minnesota Medical School, Minneapolis, MN 55455, USA
4
Department of Genetics, Cell Biology, and Development, University of Minnesota Medical School, Minneapolis, MN 55455, USA
*
Author to whom correspondence should be addressed.
Genes 2019, 10(4), 270; https://doi.org/10.3390/genes10040270
Submission received: 29 January 2019 / Revised: 21 March 2019 / Accepted: 27 March 2019 / Published: 2 April 2019
(This article belongs to the Special Issue microRNA Regulation in Health and Disease)

Abstract

:
Changes in gut microbiota composition have consistently been observed in patients with colorectal cancer (CRC). Yet, it is not entirely clear how the gut microbiota interacts with tumor cells. We know that tumor cells undergo a drastic change in energy metabolism, mediated by microRNAs (miRNAs), and that tumor-derived miRNAs affect the stromal and immune cell fractions of the tumor microenvironment. Recent studies suggest that host intestinal miRNAs can also affect the growth and composition of the gut microbiota. Our previous CRC studies showed a high-level of interconnectedness between host miRNAs and their microbiota. Considering all the evidence to date, we postulate that the altered nutrient composition and miRNA expression in the CRC microenvironment selectively exerts pressure on the surrounding microbiota, leading to alterations in its composition. In this review article, we present our current understanding of the role of miRNAs in mediating host–microbiota interactions in CRC.

1. Introduction

An average human intestine contains more than 100 trillion bacteria (collectively known as the gut microbiota) [1]. In recent decades, a number of studies have suggested that the gut microbiota is crucial to human health and to the development of diseases, including colorectal cancer (CRC) [2,3,4,5,6,7]. Those studies determined that altered microbiota composition and function (dysbiosis) is a common signature of CRC. Bacterial candidates such as Fusobacterium nucleatum and Bacteroides fragilis are consistently enriched in tumor tissues, and included in that signature. Specific factors in those bacteria, including FadA and Fap2 protein from F. nucleatum and B. fragilis toxins, that play a role in CRC pathobiology have been identified [8,9,10,11,12,13,14,15,16]. However, our knowledge of the vast majority of other bacteria associated with the CRC microenvironment is limited. Moreover, we are just beginning to understand the complex interactions between host and microbiota in CRC, as well as other clinical disorders including neurodegenerative diseases [17].
In healthy humans, a key factor associated with microbiota variations is host genetics [18,19,20,21]. In a study of healthy twins, Goodrich et al. found that host genetics drive microbiota composition and can also affect the host metabolic phenotype [19]. Several other studies have found an association between the abundance of Bifidobacterium species and the presence of single-nucleotide polymorphisms (SNPs) in close proximity to the host lactase gene locus [18,22]. This association suggests that the Bifidobacterium species conceivably assists the host in metabolizing lactose.
A recent CRC study found that loss-of-function mutations in the mitogen-activated protein kinase (MAPK) and Wnt signaling pathways are associated with specific sets of microbiota profiles [2]. Furthermore, mutations in the tumor-suppressor adenomatous polyposis coli (APC) gene are also associated with a distinct inter-microbiota association network [2]. These findings suggest that a common genetic factor might orchestrate the dynamic host–microbiota interaction(s) and functional relationship(s). Indeed, other recent studies have provided experimental evidence that microRNAs (miRNAs) can influence the survival and composition of gut bacteria [3,23,24]. Moreover, miRNAs have important intermediate roles in regulating CRC transformation and progression via the action of signaling pathways, including MAPK, Wnt, and APC [25,26]. MicroRNAs are small noncoding RNAs (about 22 nt) that play an important role in regulating and fine-tuning gene expression [27]. In mammalian cells, miRNAs regulate gene expression through posttranscriptional modifications in two distinct, albeit paired, mechanisms. First, if the miRNA has an extensive complementary binding site in the messenger RNA (mRNA) target, then it will guide the RNA-induced silencing complex (RISC) to cleave the mRNA, thus inhibiting translation. Second, if the miRNA only partially binds to the 3’ untranslated region (3’UTR) of the mRNA, then the miRNA-RISC will act to repress mRNA translation [28]. Both mechanisms lead to the decreased translation of mRNAs, which alters their respective downstream functions. Because miRNAs can act upon mRNA targets with limited complementarity, each miRNA can target a wide range of mRNAs in mammalian cells and each mRNA can be targeted by numerous miRNAs. More than 30% of human genes are estimated to have conserved binding sites in the 3’UTR [29]. Clearly, given this vast and enormously complex regulatory network, miRNAs are immensely important in regulating critical cellular processes. We are only now beginning to understand the sophisticated cross-talk of miRNAs, not only with each other, but with the myriad of target mRNAs.
Based on mounting evidence, we postulate that the altered nutrient composition and miRNA expression in the CRC microenvironment selectively influences the surrounding microbiota, leading to alterations in its composition. In this review, we present our current understanding of the role of miRNAs in mediating host–microbiota interactions in CRC (Figure 1). After highlighting the evidence pointing to their central role, we reflect on the future direction of this rapidly evolving field.

2. Microbiota and Colorectal Cancer

In the healthy intestine, the microbiota maintains a stable structure and actively participates in energy harvesting and nutrient production from undigested food [30,31]. However, this balance is disrupted in patients with CRC. Current evidence suggests that the microbiota regulates host functions via both metabolites and secreted factors.

2.1. Microbial Metabolites

In a normal colon, the microbiota produces a vast number of metabolites. Some of them, including vitamin K, biotin, and short-chain fatty acids (SCFAs), are essential for maintaining homeostasis in the colon microenvironment [31]. In fact, the major energy source (∼70%) required by colon epithelium is butyrate, which is produced by the microbiota through fermentation of complex carbohydrates. Without the microbiota, the colon epithelium undergoes autophagy and fails to maintain its normal structure and function [32]. Similarly, mice lacking a microbiota (i.e., germ-free mice or those treated by broad-spectrum antibiotics) develop significantly fewer tumors in the colon [33,34,35]. However, in humans, using broad-spectrum antibiotics to treat CRC is not feasible, because of the risk of introducing harmful and highly resistant secondary infections such as Clostridium difficile.
In our current understanding, a few main classes of bacterial metabolites play a key role in the pathogenesis of CRC and the immune microenvironment. These metabolites include SCFAs, polyamines, secondary bile acids, and phytochemicals. Their role in CRC has been extensively reviewed and documented [31,36,37,38]. We have recently also explored the role of miRNAs in mediating the effect of microbial metabolites on CRC and its microenvironment [39].

2.2. Microbial Factors

Early studies consistently found a greater population of F. nucleatum in microbiota samples in patients with CRC than in healthy controls [40,41]. This bacterium is commonly found in the human oral microbiota and is frequently associated with gum diseases; it is, however, not commonly present in the gut microbiota. Through the Fap2 virulence factor, it uniquely binds with the D-galactose-β(1-3)-N-acetyl-D-galactosamine (Gal-GalNAc) carbohydrate moiety expressed on the tumor surface of CRCs [14]. Once it localizes to the CRC microenvironment, it targets the Wnt/β-catenin signaling pathway by binding, via association with the FadA virulence factor, to the E-cadherin protein on the cell surface [16]. The Wnt/β-catenin signaling pathway is critical during tumor initiation, tumor migration, and metabolic reprogramming [42,43,44,45]. The role of the Wnt/β-catenin signaling pathway in CRC has been previously reviewed [45].
Another bacterial protein targeting the same Wnt/β-catenin signaling pathway is the Bacteroides fragilis toxin (bft) produced by B. fragilis [46]. The bft virulence factor is able to bind to the E-cadherin protein, similar to that of FadA, but additionally cleaves the protein, which can alter the intestinal tight-junction function [47]. The Wnt/β-catenin pathway is a major signaling pathway that controls the expression of many important tumor-related genes, including MYC. The transcription factor MYC, transactivates miRNAs, such as the miR-17-92 cluster, that are highly expressed in CRC [48,49,50,51].
Additionally, F. nucleatum can also induce CRC cell proliferation by upregulating miR-21, via activation of the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) pathway via toll-like receptor 4 (TLR4) signaling [52]. The Escherichia coli bacterium harboring the pks genomic island also plays an important role in CRC. When CRC cells come in contact with the colibactin genotoxin produced by E. coli, the cells undergo cellular senescence [53,54]. This process is mediated by the cellular upregulation of miR-20a-5p, which results in the downregulation of sentrin-specific protease 1 (SENP1). This process then alters p53 small ubiquitin-like modifier (SUMO)ylation, which has been shown to affect the growth and metastasis of tumor cells [55].
In addition to factors that are virulent, many bacteria also produce beneficial factors that can reduce inflammation and modulate the immune system. In germ-free mice, early studies found impaired intestinal immune systems, which were amenable to treatment [56]. Specifically, the B. fragilis polysaccharide A (PSA) is one such immunomodulatory factor that maintains the proper function of CD4+ T cells [57]. Several other polysaccharides produced by B. fragilis are also beneficial in maintaining proper immune function. Immunization with B. fragilis polysaccharides, or the adoptive transfer of T cells specific to B. fragilis, can even boost the treatment effect of anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4) immunotherapy [58]. The seemingly conflicting role of B. fragilis within gut bacteria is only the tip of the iceberg in current microbiota research and the fine and highly complex balance between functions.
In light of all this evidence, we created the first system-level map of interactions between host miRNAs and the microbiota [3]. Our comprehensive map helped us analyze correlations between host miRNA expression levels and mucosa-associated microbiota profiles, specifically in patients with CRC.

3. MicroRNAs and Colorectal Cancer

Previous studies have identified numerous aberrant miRNA expression patterns in CRC [25,59,60,61,62]. Specifically, the miR-17-92 cluster, miR-21, miR-182, and miR-503 are consistently overexpressed in tumor (vs. normal) tissues [3,26,48,49,59,63,64,65,66,67,68,69,70,71]. Any alteration(s) in expression levels of these miRNAs could, in turn, affect a wide array of downstream gene targets. Together, these miRNAs regulate all aspects of tumor pathobiology, including (i) altering tumor metabolism; (ii) promoting cell proliferation; (iii) stimulating angiogenesis; (iv) down-regulating tumor-suppressor genes; (v) promoting evasion of immune surveillance; and (vi) creating a favorable tumor microenvironment that promotes invasion and metastasis.
Our laboratory previously reported that, during the adenoma to adenocarcinoma transition, miR-182 and miR-503 were sequentially overexpressed and targeted the tumor-suppressor FBXW7 gene [69]. Other researchers have observed, during CRC transformation, an increased expression of the miR-17-92 cluster and miR-21 [48,72]. In CRC adenocarcinoma, members of the miR-17-92 cluster target transforming growth factor-beta (TGF-β), which in turn stimulates angiogenesis in the tumor microenvironment, thus promoting tumor growth [70]. Additionally, miR-19, a member of the miR-17-92 cluster, downregulates expression of the tumor-suppressor phosphatase and tensin homolog (PTEN), thereby activating the protein kinase B (AKT)/mammalian target of rapamycin (mTOR) pathway in tumor cells [73]. The AKT/mTOR pathway is the main metabolic sensing pathway, responsible for regulating glucose transport into cells [74]. Since glucose is the main fuel source of CRC cells, an activated AKT/mTOR pathway promotes tumor cell proliferation [75].
The tumor-suppressor PDCD4 gene, which is commonly downregulated in CRC, is a target of miR-21 [67]. Inhibiting the PDCD4 gene can lead to an increase in the metastasis potential of tumor cells. Another important pathway commonly altered in CRC tumors is the Wnt/β-catenin pathway [25,26]. Dozens of miRNAs have been shown to extensively regulate the genes involved in the Wnt/β-catenin pathway [25].
The complex microenvironment of the CRC tumor also involves stromal cell and immune cell fractions, which can be regulated by cancer-derived miRNAs [76,77,78]. Studies have found that the miR-17-92 cluster, commonly overexpressed in CRC cells, is also upregulated in CRC stromal cells [68,72,79]. Strikingly, these miRNAs are not only endogenously produced by stromal cells, but also packaged in the microvesicles of tumor cells, and then delivered to stromal cells [80,81]. Similar intracellular regulation mediated by miRNAs is also found in immune cell fractions [82]. Additionally, endogenous miRNA dysregulation is prevalent in CRC immune cell fractions, usually as a downstream effect of tumor-secreted factors such as cytokines and chemokines [83,84,85]. Collectively, this evidence suggests that miRNAs are important in regulating tumor cells, in addition to maintaining the tumor microenvironment. It is clear that the relationship between miRNAs and CRC is multifaceted, interrelated, and highly complex.

4. Host Regulation of Microbiota Mediated by MicroRNAs

In reestablishing germ-free mice with a normal microbiota, studies have found altered intestinal miRNA profiles, suggesting that the microbiota regulates host miRNA expression [86,87]. Moreover, the responses of intestinal cells to facilitating the microbiota process depends on the cell type, and intestinal epithelial stem cells are especially sensitive to microbiota reestablishment [87].
Because miRNAs are highly stable, several studies in the clinical arena were able to detect higher levels of miR-21 and miR-92a, among other miRNAs, in the fecal samples of patients with CRC [65,88,89]. This finding facilitated in developing a noninvasive CRC screening method and delineating the potential role of miRNAs in interacting with the trillions of microbes in the human gut.
Intestinal miRNAs develop from two main sources, including the host and the food [23,24]. The intestinal epithelial cells are the main contributors of host-derived miRNAs, either via shedding of cells or excretion of exosomes. Evidence has shown that miRNAs from food can be absorbed by the host and can affect host gene expression [90,91,92]. But certain food-sourced miRNAs remain stable in the digestive tract and reach the intestines [93,94]. This evidence suggests that miRNAs can mediate cross-species regulation. The idea remains nascent, so insight into how miRNAs mediate host–microbiota interactions is still limited. Liu et al. first demonstrated such regulation, showing that miRNAs present in the feces can regulate gene expression and growth of bacteria [23]. Specifically, they found that mice lacking the Dicer gene, which enables mature miRNA processing, had different microbiota profiles than wild-type mice. More importantly, the study reported that hsa-miR-515-5p promoted the growth of F. nucleatum in vitro by targeting the 16S ribosomal RNA (rRNA) gene.
Notably, however, hsa-miR-515-5p shows very low expression levels in CRC tumors, so they are not significantly different from normal tissue. Thus, interactions between hsa-miR-515-5p and F. nucleatum might not be significant in CRC pathogenesis. However, more importantly, this study found that fecal miRNA transplantation restores fecal microbiota composition in mice with Dicer gene knockout. Several recent studies found that fecal microbiota transplantation (FMT) offers a potential therapeutic benefit that enables an immunotherapeutic response [35,95,96,97,98]. Based on growing evidence, it is plausible that fecal miRNAs play an important role in modulating the CRC microbiota as well as immunotherapy responses.
Recently, Teng et al. demonstrated that miRNAs encapsulated in plant-derived exosome-like nanoparticles (ELNs) can enter bacteria and alter bacterial genes [24]. The process for bacterial uptake of ELNs is determined primarily by the lipid composition of the outer membrane. They found that ELNs enriched with phosphatidylcholine were preferentially taken up by the Ruminococcus species, whereas ELNs enriched with phosphatidic acid (PA) were primarily taken up by Lactobacillus rhamnosus. After the ELNs are taken up by specific bacteria, the miRNA contents are released into bacterial cells. Teng et al. also found that mdo-miR7267-3p encapsulated in the PA-enriched ELNs targets the Lactobacillus monooxygenase ycnE, which then increases its production of indole-3-carboxaldehyde (I3A). The I3A metabolite then promotes interleukin-22 (IL-22) production and helps repair damaged colon mucosa [99].
There is developing evidence to support the notion that host or exogenous miRNAs might be biologically active in bacteria, thereby affecting bacterial gene expression. Although small RNAs similar to miRNAs exist in bacteria and function similarly to miRNAs, it remains unknown as to how miRNAs function in bacteria [100]. Several studies have reported that exogenous miRNAs from plant or animal sources can be taken up by human cells and exert biological functions [90,91,92,93,94,101,102,103]. Additional studies are required to ascertain whether miRNAs can indeed affect bacteria and to delineate the precise mechanism(s).

5. Metabolic Changes in Colorectal Cancer and Microbiota Mediated by MicroRNAs

The prevailing “driver-passenger” model suggests that dysbiosis in the CRC microbiota is initially caused by colonization of driver bacteria. This is followed by a gradual change in the tumor microenvironment, an increase in the number of driver bacteria, and secondary colonization of passenger bacteria that benefit from the changed environment [104]. That model, together with other studies, suggest that a gradual metabolic change in the tumor microenvironment during cancer progression could be the cause of dysbiosis [31]. Again, we explored that issue in our recent review of the role of miRNAs in mediating the effect of microbial metabolites on CRC and its microenvironment [39].
One of the hallmarks of tumor growth is their increased use of glycolysis as a main energy source, known universally as the Warburg effect [105]. Because the normal colon uses butyrate as its major energy source, any change in that source preferred by proliferating tumor cells will undoubtedly profoundly alter the nutrient composition of the tumor microenvironment [106,107]. Indeed, several studies have found altered metabolite levels in CRC tissues and stools [107,108,109,110]. A significantly lower glucose level and higher levels of lactate and fatty acids have been found in CRC tumor tissues, as compared with adjacent normal tissues. In stool samples from patients with CRC, a higher level of amino acids and a lower level of fatty acids have also been observed [108]. Interestingly, the CRC microbiota has shown reduced carbohydrate metabolism and an increase in the biosynthesis of amino acids and fatty acids [41]. In CRC, the switch in the nutrient source preferred by proliferating tumor cells appears to alter the nutrient composition in the tumor microenvironment. At the same time, the nutrient metabolism of the tumor microbiota seems to complement the nutrient needs of the tumor. This could be due to factors associated with the tumor nutrient microenvironment, and by the miRNAs excreted by tumor cells, on the surrounding microbiota [3]. Given the role of miRNAs in mediating such metabolic changes, we believe that miRNAs play a central, if not critical role, in mediating host–microbiota metabolic interactions in CRC.

6. Conclusions and Perspectives

With thousands of bacterial species living in the human digestive tract, it is becoming quite evident that they profoundly affect human health. Our review of the recent literature regarding CRC underscores a complex metabolic interplay between the host and its microbiota, mediated in part by miRNAs. Based on the current literature, we offer five major points in host–microbiota interactions mediated by miRNAs (Figure 1):
  • The CRC microbiota has reduced representation of beneficial bacteria. These bacteria produce metabolites and other factors that can potentially slow CRC progression, in part via the modulation of miRNAs that regulate tumor cells.
  • Dysregulation of miRNAs in tumor cells can affect the survival, or the gene expression, of certain bacteria in the microbiota.
  • Dysregulated miRNAs in tumor cells can be packaged and delivered to both stromal and immune cell fractions, creating a more favorable microenvironment for tumor cells.
  • Overrepresentation of oncogenic bacteria in the CRC microbiota can modulate tumor cells, as well as the tumor microenvironment, through miRNA modulation, thereby resulting in a more favorable condition for tumor growth.
  • This negative feedback loop perpetuates CRC progression.
Potential methods to break such a negative feedback loop include:
  • Interfering with host-mediated microbiota modulation by designing strategies to deliver anti-miRNAs to block the effect of host-miRNAs on the microbiota.
  • Modulating the microbiota through miRNAs that promote the growth of beneficial bacteria while suppressing the growth of oncogenic bacteria, in conjunction with chemotherapy or immunotherapy.
Based on both experimental and computational data, we conclude that miRNAs mediate and critically influence host–microbiota interactions. Clearly, miRNAs are a major part of a complex web of highly dynamic interactions. Other factors, such as nutrient availability in the CRC microenvironment, could also play an important role. In the future, it will be imperative to use a combination of approaches to comprehensively survey the CRC microenvironment, in order to discover all potential players in mediating such interactions.

Acknowledgments

S.S. is supported by research grants funded by the NIH R03CA219129 and Chainbreaker Grant; C.Y., by the MnDrive—University of Minnesota Informatics Institute graduate fellowship. Due to space restrictions, we cannot cite many other significant contributions made by numerous researchers and laboratories in this potentially important and rapidly progressing field. We thank Mary Knatterud for assisting in manuscript editing.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Turnbaugh, P.J.; Ley, R.E.; Hamady, M.; Fraser-Liggett, C.M.; Knight, R.; Gordon, J.I. The human microbiome project. Nature 2007, 449, 804–810. [Google Scholar] [CrossRef]
  2. Burns, M.B.; Montassier, E.; Abrahante, J.; Priya, S.; Niccum, D.E.; Khoruts, A.; Starr, T.K.; Knights, D.; Blekhman, R. Colorectal cancer mutational profiles correlate with defined microbial communities in the tumor microenvironment. PLoS Genet. 2018, 14, e1007376. [Google Scholar] [CrossRef]
  3. Yuan, C.; Burns, M.B.; Subramanian, S.; Blekhman, R. Interaction between host microRNAs and the gut microbiota in colorectal cancer. mSystems 2018, 3, e00205-17. [Google Scholar] [CrossRef] [PubMed]
  4. Dejea, C.M.; Wick, E.C.; Hechenbleikner, E.M.; White, J.R.; Mark Welch, J.L.; Rossetti, B.J.; Peterson, S.N.; Snesrud, E.C.; Borisy, G.G.; Lazarev, M.; et al. Microbiota organization is a distinct feature of proximal colorectal cancers. Proc. Natl. Acad. Sci. USA 2014, 111, 18321–18326. [Google Scholar] [CrossRef]
  5. Kostic, A.D.; Gevers, D.; Pedamallu, C.S.; Michaud, M.; Duke, F.; Earl, A.M.; Ojesina, A.I.; Jung, J.; Bass, A.J.; Tabernero, J.; et al. Genomic analysis identifies association of Fusobacterium with colorectal carcinoma. Genome Res. 2012, 22, 292–298. [Google Scholar] [CrossRef]
  6. Shah, M.S.; DeSantis, T.; Yamal, J.-M.; Weir, T.; Ryan, E.P.; Cope, J.L.; Hollister, E.B. Re-purposing 16S rRNA gene sequence data from within case paired tumor biopsy and tumor-adjacent biopsy or fecal samples to identify microbial markers for colorectal cancer. PLoS ONE 2018, 13, e0207002. [Google Scholar] [CrossRef]
  7. García-Castillo, V.; Sanhueza, E.; McNerney, E.; Onate, S.A.; García, A. Microbiota dysbiosis: A new piece in the understanding of the carcinogenesis puzzle. J. Med. Microbiol. 2016, 65, 1347–1362. [Google Scholar] [CrossRef] [PubMed]
  8. Boleij, A.; Hechenbleikner, E.M.; Goodwin, A.C.; Badani, R.; Stein, E.M.; Lazarev, M.G.; Ellis, B.; Carroll, K.C.; Albesiano, E.; Wick, E.C.; et al. The Bacteroides fragilis toxin gene is prevalent in the colon mucosa of colorectal cancer patients. Clin. Infect. Dis. 2015, 60, 208–215. [Google Scholar] [CrossRef] [PubMed]
  9. Toprak, N.U.; Yagci, A.; Gulluoglu, B.M.; Akin, M.L.; Demirkalem, P.; Celenk, T.; Soyletir, G. A possible role of Bacteroides fragilis enterotoxin in the aetiology of colorectal cancer. Clin. Microbiol. Infect. 2006, 12, 782–786. [Google Scholar] [CrossRef]
  10. Wu, S.; Shin, J.; Zhang, G.; Cohen, M.; Franco, A.; Sears, C.L. The Bacteroides fragilis toxin binds to a specific intestinal epithelial cell receptor. Infect. Immun. 2006, 74, 5382–5390. [Google Scholar] [CrossRef]
  11. Kostic, A.D.; Chun, E.; Robertson, L.; Glickman, J.N.; Gallini, C.A.; Michaud, M.; Clancy, T.E.; Chung, D.C.; Lochhead, P.; Hold, G.L.; et al. Fusobacterium nucleatum potentiates intestinal tumorigenesis and modulates the tumor-immune microenvironment. Cell Host Microbe 2013, 14, 207–215. [Google Scholar] [CrossRef]
  12. Mima, K.; Cao, Y.; Chan, A.T.; Qian, Z.R.; Nowak, J.A.; Masugi, Y.; Shi, Y.; Song, M.; da Silva, A.; Gu, M.; et al. Fusobacterium nucleatum in colorectal carcinoma tissue according to tumor location. Clin. Transl. Gastroenterol. 2016, 7, e200. [Google Scholar] [CrossRef]
  13. Mima, K.; Nishihara, R.; Qian, Z.R.; Cao, Y.; Sukawa, Y.; Nowak, J.A.; Yang, J.; Dou, R.; Masugi, Y.; Song, M.; et al. Fusobacterium nucleatum in colorectal carcinoma tissue and patient prognosis. Gut 2016, 65, 1973–1980. [Google Scholar] [CrossRef]
  14. Abed, J.; Emgård, J.E.M.; Zamir, G.; Faroja, M.; Almogy, G.; Grenov, A.; Sol, A.; Naor, R.; Pikarsky, E.; Atlan, K.A.; et al. Fap2 mediates Fusobacterium nucleatum colorectal adenocarcinoma enrichment by binding to tumor-expressed Gal-GalNAc. Cell Host Microbe 2016, 20, 215–225. [Google Scholar] [CrossRef]
  15. Rubinstein, M.R.; Baik, J.E.; Lagana, S.M.; Han, R.P.; Raab, W.J.; Sahoo, D.; Dalerba, P.; Wang, T.C.; Han, Y.W. Fusobacterium nucleatum promotes colorectal cancer by inducing Wnt/β-catenin modulator Annexin A1. EMBO Rep. 2019. [Google Scholar] [CrossRef]
  16. Rubinstein, M.R.; Wang, X.; Liu, W.; Hao, Y.; Cai, G.; Han, Y.W. Fusobacterium nucleatum promotes colorectal carcinogenesis by modulating E-cadherin/β-catenin signaling via its FadA adhesin. Cell Host Microbe 2013, 14, 195–206. [Google Scholar] [CrossRef]
  17. Schroeder, B.O.; Bäckhed, F. Signals from the gut microbiota to distant organs in physiology and disease. Nat. Med. 2016, 22, 1079–1089. [Google Scholar] [CrossRef]
  18. Blekhman, R.; Goodrich, J.K.; Huang, K.; Sun, Q.; Bukowski, R.; Bell, J.T.; Spector, T.D.; Keinan, A.; Ley, R.E.; Gevers, D.; et al. Host genetic variation impacts microbiome composition across human body sites. Genome Biol. 2015, 16, 191. [Google Scholar] [CrossRef] [PubMed]
  19. Goodrich, J.K.; Waters, J.L.; Poole, A.C.; Sutter, J.L.; Koren, O.; Blekhman, R.; Beaumont, M.; Van Treuren, W.; Knight, R.; Bell, J.T.; et al. Human genetics shape the gut microbiome. Cell 2014, 159, 789–799. [Google Scholar] [CrossRef]
  20. Bongers, G.; Pacer, M.E.; Geraldino, T.H.; Chen, L.; He, Z.; Hashimoto, D.; Furtado, G.C.; Ochando, J.; Kelley, K.A.; Clemente, J.C.; et al. Interplay of host microbiota, genetic perturbations, and inflammation promotes local development of intestinal neoplasms in mice. J. Exp. Med. 2014, 211, 457–472. [Google Scholar] [CrossRef]
  21. Davenport, E.R.; Cusanovich, D.A.; Michelini, K.; Barreiro, L.B.; Ober, C.; Gilad, Y. Genome-wide association studies of the human gut microbiota. PLoS ONE 2015, 10, e0140301. [Google Scholar] [CrossRef]
  22. Goodrich, J.K.; Davenport, E.R.; Beaumont, M.; Jackson, M.A.; Knight, R.; Ober, C.; Spector, T.D.; Bell, J.T.; Clark, A.G.; Ley, R.E. Genetic determinants of the gut microbiome in UK twins. Cell Host Microbe 2016, 19, 731–743. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, S.; da Cunha, A.P.; Rezende, R.M.; Cialic, R.; Wei, Z.; Bry, L.; Comstock, L.E.; Gandhi, R.; Weiner, H.L. The host shapes the gut microbiota via fecal microrna. Cell Host Microbe 2016, 19, 32–43. [Google Scholar] [CrossRef] [PubMed]
  24. Teng, Y.; Ren, Y.; Sayed, M.; Hu, X.; Lei, C.; Kumar, A.; Hutchins, E.; Mu, J.; Deng, Z.; Luo, C.; et al. Plant-derived exosomal microRNAs shape the gut microbiota. Cell Host Microbe 2018, 24, 637–652.e8. [Google Scholar] [CrossRef] [PubMed]
  25. Slattery, M.L.; Mullany, L.E.; Sakoda, L.C.; Samowitz, W.S.; Wolff, R.K.; Stevens, J.R.; Herrick, J.S. Expression of Wnt-signaling pathway genes and their associations with miRNAs in colorectal cancer. Oncotarget 2018, 9, 6075–6085. [Google Scholar] [CrossRef] [PubMed]
  26. Li, Y.; Lauriola, M.; Kim, D.; Francesconi, M.; D’Uva, G.; Shibata, D.; Malafa, M.P.; Yeatman, T.J.; Coppola, D.; Solmi, R.; et al. Adenomatous polyposis coli (APC) regulates miR17-92 cluster through β-catenin pathway in colorectal cancer. Oncogene 2016, 35, 4558–4568. [Google Scholar] [CrossRef] [PubMed]
  27. Bartel, D.P. MicroRNAs: Genomics, biogenesis, mechanism, and function. Cell 2004, 116, 281–297. [Google Scholar] [CrossRef]
  28. Fabian, M.R.; Sonenberg, N. The mechanics of miRNA-mediated gene silencing: A look under the hood of miRISC. Nat. Struct. Mol. Biol. 2012, 19, 586–593. [Google Scholar] [CrossRef]
  29. Lewis, B.P.; Burge, C.B.; Bartel, D.P. Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005, 120, 15–20. [Google Scholar] [CrossRef]
  30. The Human Microbiome Project Consortium. Structure, function and diversity of the healthy human microbiome. Nature 2012, 486, 207–214. [Google Scholar] [CrossRef] [PubMed]
  31. Louis, P.; Hold, G.L.; Flint, H.J. The gut microbiota, bacterial metabolites and colorectal cancer. Nat. Rev. Microbiol. 2014, 12, 661–672. [Google Scholar] [CrossRef]
  32. Donohoe, D.R.; Garge, N.; Zhang, X.; Sun, W.; O’Connell, T.M.; Bunger, M.K.; Bultman, S.J. The microbiome and butyrate regulate energy metabolism and autophagy in the mammalian colon. Cell Metab. 2011, 13, 517–526. [Google Scholar] [CrossRef]
  33. Dove, W.F.; Clipson, L.; Gould, K.A.; Luongo, C.; Marshall, D.J.; Moser, A.R.; Newton, M.A.; Jacoby, R.F. Intestinal neoplasia in the ApcMin mouse: Independence from the microbial and natural killer (beige locus) status. Cancer Res. 1997, 57, 812–814. [Google Scholar] [PubMed]
  34. Zackular, J.P.; Baxter, N.T.; Chen, G.Y.; Schloss, P.D. Manipulation of the gut microbiota reveals role in colon tumorigenesis. mSphere 2016, 1. [Google Scholar] [CrossRef] [PubMed]
  35. Vivarelli, S.; Salemi, R.; Candido, S.; Falzone, L.; Santagati, M.; Stefani, S.; Torino, F.; Banna, G.L.; Tonini, G.; Libra, M. Gut microbiota and cancer: From pathogenesis to therapy. Cancers 2019, 11, 38. [Google Scholar] [CrossRef]
  36. Blacher, E.; Levy, M.; Tatirovsky, E.; Elinav, E. Microbiome-modulated metabolites at the interface of host immunity. J. Immunol. 2017, 198, 572–580. [Google Scholar] [CrossRef]
  37. Johnson, C.H.; Spilker, M.E.; Goetz, L.; Peterson, S.N.; Siuzdak, G. Metabolite and microbiome interplay in cancer immunotherapy. Cancer Res. 2016, 76, 6146–6152. [Google Scholar] [CrossRef]
  38. O’Keefe, S.J.D. Diet, microorganisms and their metabolites, and colon cancer. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 691–706. [Google Scholar] [CrossRef]
  39. Yuan, C.; Subramanian, S. MicroRNA mediated tumor-microbiota metabolic interactions in colorectal cancer. DNA Cell Biol. 2019. [Google Scholar] [CrossRef] [PubMed]
  40. Castellarin, M.; Warren, R.L.; Freeman, J.D.; Dreolini, L.; Krzywinski, M.; Strauss, J.; Barnes, R.; Watson, P.; Allen-Vercoe, E.; Moore, R.A.; et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012, 22, 299–306. [Google Scholar] [CrossRef]
  41. Burns, M.B.; Lynch, J.; Starr, T.K.; Knights, D.; Blekhman, R. Virulence genes are a signature of the microbiome in the colorectal tumor microenvironment. Genome Med. 2015, 7, 55. [Google Scholar] [CrossRef] [PubMed]
  42. 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]
  43. Konsavage, W.M.; Kyler, S.L.; Rennoll, S.A.; Jin, G.; Yochum, G.S. Wnt/β-catenin signaling regulates Yes-associated protein (YAP) gene expression in colorectal carcinoma cells. J. Biol. Chem. 2012, 287, 11730–11739. [Google Scholar] [CrossRef]
  44. Strillacci, A.; Valerii, M.C.; Sansone, P.; Caggiano, C.; Sgromo, A.; Vittori, L.; Fiorentino, M.; Poggioli, G.; Rizzello, F.; Campieri, M.; et al. Loss of miR-101 expression promotes Wnt/β-catenin signalling pathway activation and malignancy in colon cancer cells. J. Pathol. 2013, 229, 379–389. [Google Scholar] [CrossRef]
  45. Bienz, M.; Clevers, H. Linking colorectal cancer to Wnt signaling. Cell 2000, 103, 311–320. [Google Scholar] [CrossRef]
  46. Wu, S.; Morin, P.J.; Maouyo, D.; Sears, C.L. Bacteroides fragilis enterotoxin induces c-Myc expression and cellular proliferation. Gastroenterology 2003, 124, 392–400. [Google Scholar] [CrossRef] [PubMed]
  47. Wu, S.; Lim, K.C.; Huang, J.; Saidi, R.F.; Sears, C.L. Bacteroides fragilis enterotoxin cleaves the zonula adherens protein, E-cadherin. Proc. Natl. Acad. Sci. USA 1998, 95, 14979–14984. [Google Scholar] [CrossRef]
  48. Diosdado, B.; van de Wiel, M.A.; Terhaar Sive Droste, J.S.; Mongera, S.; Postma, C.; Meijerink, W.J.H.J.; Carvalho, B.; Meijer, G.A. MiR-17-92 cluster is associated with 13q gain and c-Myc expression during colorectal adenoma to adenocarcinoma progression. Br. J. Cancer 2009, 101, 707–714. [Google Scholar] [CrossRef]
  49. Mogilyansky, E.; Rigoutsos, I. The miR-17/92 cluster: A comprehensive update on its genomics, genetics, functions and increasingly important and numerous roles in health and disease. Cell Death Differ. 2013, 20, 1603–1614. [Google Scholar] [CrossRef]
  50. O’Donnell, K.A.; Wentzel, E.A.; Zeller, K.I.; Dang, C.V.; Mendell, J.T. c-Myc-regulated microRNAs modulate E2F1 expression. Nature 2005, 435, 839–843. [Google Scholar] [CrossRef]
  51. Dang, C.V. c-Myc target genes involved in cell growth, apoptosis, and metabolism. Mol. Cell Biol. 1999, 19, 1–11. [Google Scholar] [CrossRef] [PubMed]
  52. Yang, Y.; Weng, W.; Peng, J.; Hong, L.; Yang, L.; Toiyama, Y.; Gao, R.; Liu, M.; Yin, M.; Pan, C.; et al. Fusobacterium nucleatum increases proliferation of colorectal cancer cells and tumor development in mice by activating Toll-Like receptor 4 signaling to nuclear factor-κB, and up-regulating expression of microRNA-21. Gastroenterology 2017, 152, 851–866.e24. [Google Scholar] [CrossRef]
  53. Dalmasso, G.; Cougnoux, A.; Delmas, J.; Darfeuille-Michaud, A.; Bonnet, R. The bacterial genotoxin colibactin promotes colon tumor growth by modifying the tumor microenvironment. Gut Microbes 2014, 5, 675–680. [Google Scholar] [CrossRef] [PubMed]
  54. Cougnoux, A.; Dalmasso, G.; Martinez, R.; Buc, E.; Delmas, J.; Gibold, L.; Sauvanet, P.; Darcha, C.; Déchelotte, P.; Bonnet, M.; et al. Bacterial genotoxin colibactin promotes colon tumour growth by inducing a senescence-associated secretory phenotype. Gut 2014, 63, 1932–1942. [Google Scholar] [CrossRef] [PubMed]
  55. Baek, S.H. A novel link between SUMO modification and cancer metastasis. Cell Cycle 2006, 5, 1492–1495. [Google Scholar] [CrossRef]
  56. Gordon, H.A. Morphological and physiological characterization of germfree life. Ann. N.Y. Acad. Sci. 1959, 78, 208–220. [Google Scholar] [CrossRef] [PubMed]
  57. Mazmanian, S.K.; Liu, C.H.; Tzianabos, A.O.; Kasper, D.L. An immunomodulatory molecule of symbiotic bacteria directs maturation of the host immune system. Cell 2005, 122, 107–118. [Google Scholar] [CrossRef]
  58. Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Waldschmitt, N.; Flament, C.; Rusakiewicz, S.; Routy, B.; Roberti, M.P.; Duong, C.P.M.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2015, 350, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
  59. Sarver, A.L.; French, A.J.; Borralho, P.M.; Thayanithy, V.; Oberg, A.L.; Silverstein, K.A.T.; Morlan, B.W.; Riska, S.M.; Boardman, L.A.; Cunningham, J.M.; et al. Human colon cancer profiles show differential microRNA expression depending on mismatch repair status and are characteristic of undifferentiated proliferative states. BMC Cancer 2009, 9, 401. [Google Scholar] [CrossRef]
  60. Sarver, A.L.; Sarver, A.E.; Yuan, C.; Subramanian, S. OMCD: Oncomir cancer database. BMC Cancer 2018, 18, 1223. [Google Scholar] [CrossRef] [PubMed]
  61. Wong, N.W.; Chen, Y.; Chen, S.; Wang, X. OncomiR: An online resource for exploring pan-cancer microRNA dysregulation. Bioinformatics 2018, 34, 713–715. [Google Scholar] [CrossRef]
  62. Falzone, L.; Scola, L.; Zanghì, A.; Biondi, A.; Di Cataldo, A.; Libra, M.; Candido, S. Integrated analysis of colorectal cancer microRNA datasets: Identification of microRNAs associated with tumor development. Aging 2018, 10, 1000–1014. [Google Scholar] [CrossRef] [PubMed]
  63. Koga, Y.; Yasunaga, M.; Takahashi, A.; Kuroda, J.; Moriya, Y.; Akasu, T.; Fujita, S.; Yamamoto, S.; Baba, H.; Matsumura, Y. MicroRNA expression profiling of exfoliated colonocytes isolated from feces for colorectal cancer screening. Cancer Prev. Res. (Phila Pa) 2010, 3, 1435–1442. [Google Scholar] [CrossRef] [PubMed]
  64. Fang, L.; Li, H.; Wang, L.; Hu, J.; Jin, T.; Wang, J.; Yang, B.B. MicroRNA-17-5p promotes chemotherapeutic drug resistance and tumour metastasis of colorectal cancer by repressing PTEN expression. Oncotarget 2014, 5, 2974–2987. [Google Scholar] [CrossRef] [PubMed]
  65. Wu, C.W.; Ng, S.S.M.; Dong, Y.J.; Ng, S.C.; Leung, W.W.; Lee, C.W.; Wong, Y.N.; Chan, F.K.L.; Yu, J.; Sung, J.J.Y. Detection of miR-92a and miR-21 in stool samples as potential screening biomarkers for colorectal cancer and polyps. Gut 2012, 61, 739–745. [Google Scholar] [CrossRef] [PubMed]
  66. Kulda, V.; Pesta, M.; Topolcan, O.; Liska, V.; Treska, V.; Sutnar, A.; Rupert, K.; Ludvikova, M.; Babuska, V.; Holubec, L.; et al. Relevance of miR-21 and miR-143 expression in tissue samples of colorectal carcinoma and its liver metastases. Cancer Genet. Cytogenet. 2010, 200, 154–160. [Google Scholar] [CrossRef]
  67. Asangani, I.A.; Rasheed, S.A.K.; Nikolova, D.A.; Leupold, J.H.; Colburn, N.H.; Post, S.; Allgayer, H. MicroRNA-21 (miR-21) post-transcriptionally downregulates tumor suppressor Pdcd4 and stimulates invasion, intravasation and metastasis in colorectal cancer. Oncogene 2008, 27, 2128–2136. [Google Scholar] [CrossRef] [PubMed]
  68. Bullock, M.D.; Pickard, K.M.; Nielsen, B.S.; Sayan, A.E.; Jenei, V.; Mellone, M.; Mitter, R.; Primrose, J.N.; Thomas, G.J.; Packham, G.K.; et al. Pleiotropic actions of miR-21 highlight the critical role of deregulated stromal microRNAs during colorectal cancer progression. Cell Death Dis. 2013, 4, e684. [Google Scholar] [CrossRef]
  69. Li, L.; Sarver, A.L.; Khatri, R.; Hajeri, P.B.; Kamenev, I.; French, A.J.; Thibodeau, S.N.; Steer, C.J.; Subramanian, S. Sequential expression of miR-182 and miR-503 cooperatively targets FBXW7, contributing to the malignant transformation of colon adenoma to adenocarcinoma. J. Pathol. 2014, 234, 488–501. [Google Scholar] [CrossRef]
  70. Dews, M.; Fox, J.L.; Hultine, S.; Sundaram, P.; Wang, W.; Liu, Y.Y.; Furth, E.; Enders, G.H.; El-Deiry, W.; Schelter, J.M.; et al. The myc-miR-17~92 axis blunts TGFβ signaling and production of multiple TGF β-dependent antiangiogenic factors. Cancer Res. 2010, 70, 8233–8246. [Google Scholar] [CrossRef]
  71. Hu, S.; Liu, L.; Chang, E.B.; Wang, J.-Y.; Raufman, J.-P. Butyrate inhibits pro-proliferative miR-92a by diminishing c-Myc-induced miR-17-92a cluster transcription in human colon cancer cells. Mol. Cancer 2015, 14, 180. [Google Scholar] [CrossRef] [PubMed]
  72. Yamamichi, N.; Shimomura, R.; Inada, K.; Sakurai, K.; Haraguchi, T.; Ozaki, Y.; Fujita, S.; Mizutani, T.; Furukawa, C.; Fujishiro, M.; et al. Locked nucleic acid in situ hybridization analysis of miR-21 expression during colorectal cancer development. Clin. Cancer Res. 2009, 15, 4009–4016. [Google Scholar] [CrossRef] [PubMed]
  73. Grillari, J.; Hackl, M.; Grillari-Voglauer, R. miR-17-92 cluster: Ups and downs in cancer and aging. Biogerontology 2010, 11, 501–506. [Google Scholar] [CrossRef] [PubMed]
  74. Makinoshima, H.; Takita, M.; Saruwatari, K.; Umemura, S.; Obata, Y.; Ishii, G.; Matsumoto, S.; Sugiyama, E.; Ochiai, A.; Abe, R.; et al. Signaling through the phosphatidylinositol 3-kinase (PI3K)/mammalian target of rapamycin (mTOR) axis is responsible for aerobic glycolysis mediated by glucose transporter in epidermal growth factor receptor (EGFR)-mutated lung adenocarcinoma. J. Biol. Chem. 2015, 290, 17495–17504. [Google Scholar] [CrossRef] [PubMed]
  75. Weijenberg, M.P.; Hughes, L.A.E.; Bours, M.J.L.; Simons, C.C.J.M.; van Engeland, M.; van den Brandt, P.A. The mTOR pathway and the role of energy balance throughout life in colorectal cancer etiology and prognosis: Unravelling mechanisms through a multidimensional molecular epidemiologic approach. Curr. Nutr. Rep. 2013, 2, 19–26. [Google Scholar] [CrossRef] [PubMed]
  76. Kohlhapp, F.J.; Mitra, A.K.; Lengyel, E.; Peter, M.E. MicroRNAs as mediators and communicators between cancer cells and the tumor microenvironment. Oncogene 2015, 34, 5857–5868. [Google Scholar] [CrossRef]
  77. Kosaka, N.; Yoshioka, Y.; Hagiwara, K.; Tominaga, N.; Katsuda, T.; Ochiya, T. Trash or Treasure: Extracellular microRNAs and cell-to-cell communication. Front. Genet. 2013, 4, 173. [Google Scholar] [CrossRef]
  78. Runtsch, M.C.; Round, J.L.; O’Connell, R.M. MicroRNAs and the regulation of intestinal homeostasis. Front. Genet. 2014, 5, 347. [Google Scholar] [CrossRef]
  79. Nielsen, B.S.; Jørgensen, S.; Fog, J.U.; Søkilde, R.; Christensen, I.J.; Hansen, U.; Brünner, N.; Baker, A.; Møller, S.; Nielsen, H.J. High levels of microRNA-21 in the stroma of colorectal cancers predict short disease-free survival in stage II colon cancer patients. Clin. Exp. Metastasis 2011, 28, 27–38. [Google Scholar] [CrossRef]
  80. Dews, M.; Homayouni, A.; Yu, D.; Murphy, D.; Sevignani, C.; Wentzel, E.; Furth, E.E.; Lee, W.M.; Enders, G.H.; Mendell, J.T.; et al. Augmentation of tumor angiogenesis by a Myc-activated microRNA cluster. Nat. Genet. 2006, 38, 1060–1065. [Google Scholar] [CrossRef]
  81. Zhuang, G.; Wu, X.; Jiang, Z.; Kasman, I.; Yao, J.; Guan, Y.; Oeh, J.; Modrusan, Z.; Bais, C.; Sampath, D.; et al. Tumour-secreted miR-9 promotes endothelial cell migration and angiogenesis by activating the JAK-STAT pathway. EMBO J. 2012, 31, 3513–3523. [Google Scholar] [CrossRef]
  82. Fanini, F.; Fabbri, M. Cancer-derived exosomic microRNAs shape the immune system within the tumor microenvironment: State of the art. Semin. Cell Dev. Biol. 2017, 67, 23–28. [Google Scholar] [CrossRef] [PubMed]
  83. Sonda, N.; Simonato, F.; Peranzoni, E.; Calì, B.; Bortoluzzi, S.; Bisognin, A.; Wang, E.; Marincola, F.M.; Naldini, L.; Gentner, B.; et al. miR-142-3p prevents macrophage differentiation during cancer-induced myelopoiesis. Immunity 2013, 38, 1236–1249. [Google Scholar] [CrossRef] [PubMed]
  84. Zhang, M.; Liu, Q.; Mi, S.; Liang, X.; Zhang, Z.; Su, X.; Liu, J.; Chen, Y.; Wang, M.; Zhang, Y.; et al. Both miR-17-5p and miR-20a alleviate suppressive potential of myeloid-derived suppressor cells by modulating STAT3 expression. J. Immunol. 2011, 186, 4716–4724. [Google Scholar] [CrossRef]
  85. Mei, S.; Xin, J.; Liu, Y.; Zhang, Y.; Liang, X.; Su, X.; Yan, H.; Huang, Y.; Yang, R. MicroRNA-200c promotes suppressive potential of myeloid-derived suppressor cells by modulating PTEN and FOG2 expression. PLoS ONE 2015, 10, e0135867. [Google Scholar] [CrossRef]
  86. Dalmasso, G.; Nguyen, H.T.T.; Yan, Y.; Laroui, H.; Charania, M.A.; Ayyadurai, S.; Sitaraman, S.V.; Merlin, D. Microbiota modulate host gene expression via microRNAs. PLoS ONE 2011, 6, e19293. [Google Scholar] [CrossRef] [PubMed]
  87. Peck, B.C.E.; Mah, A.T.; Pitman, W.A.; Ding, S.; Lund, P.K.; Sethupathy, P. Functional transcriptomics in diverse intestinal epithelial cell types reveals robust microrna sensitivity in intestinal stem cells to microbial status. J. Biol. Chem. 2017, 292, 2586–2600. [Google Scholar] [CrossRef]
  88. Phua, L.C.; Chue, X.P.; Koh, P.K.; Cheah, P.Y.; Chan, E.C.Y.; Ho, H.K. Global fecal microRNA profiling in the identification of biomarkers for colorectal cancer screening among Asians. Oncol. Rep. 2014, 32, 97–104. [Google Scholar] [CrossRef]
  89. Rotelli, M.T.; Di Lena, M.; Cavallini, A.; Lippolis, C.; Bonfrate, L.; Chetta, N.; Portincasa, P.; Altomare, D.F. Fecal microRNA profile in patients with colorectal carcinoma before and after curative surgery. Int. J. Colorectal Dis. 2015, 30, 891–898. [Google Scholar] [CrossRef]
  90. Baier, S.R.; Nguyen, C.; Xie, F.; Wood, J.R.; Zempleni, J. MicroRNAs are absorbed in biologically meaningful amounts from nutritionally relevant doses of cow milk and affect gene expression in peripheral blood mononuclear cells, HEK-293 kidney cell cultures, and mouse livers. J. Nutr. 2014, 144, 1495–1500. [Google Scholar] [CrossRef] [PubMed]
  91. Izumi, H.; Tsuda, M.; Sato, Y.; Kosaka, N.; Ochiya, T.; Iwamoto, H.; Namba, K.; Takeda, Y. Bovine milk exosomes contain microRNA and mRNA and are taken up by human macrophages. J. Dairy Sci. 2015, 98, 2920–2933. [Google Scholar] [CrossRef]
  92. Zhang, L.; Hou, D.; Chen, X.; Li, D.; Zhu, L.; Zhang, Y.; Li, J.; Bian, Z.; Liang, X.; Cai, X.; et al. Exogenous plant MIR168a specifically targets mammalian LDLRAP1: Evidence of cross-kingdom regulation by microRNA. Cell Res. 2012, 22, 107–126. [Google Scholar] [CrossRef]
  93. Mu, J.; Zhuang, X.; Wang, Q.; Jiang, H.; Deng, Z.-B.; Wang, B.; Zhang, L.; Kakar, S.; Jun, Y.; Miller, D.; et al. Interspecies communication between plant and mouse gut host cells through edible plant derived exosome-like nanoparticles. Mol. Nutr. Food Res. 2014, 58, 1561–1573. [Google Scholar] [CrossRef]
  94. Wolf, T.; Baier, S.R.; Zempleni, J. The intestinal transport of bovine milk exosomes is mediated by endocytosis in human colon carcinoma Caco-2 cells and rat small intestinal IEC-6 cells. J. Nutr. 2015, 145, 2201–2206. [Google Scholar] [CrossRef]
  95. 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. [Google Scholar] [CrossRef]
  96. 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. [Google Scholar] [CrossRef]
  97. 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]
  98. Gopalakrishnan, V.; Helmink, B.A.; Spencer, C.N.; Reuben, A.; Wargo, J.A. The influence of the gut microbiome on cancer, immunity, and cancer immunotherapy. Cancer Cell 2018, 33, 570–580. [Google Scholar] [CrossRef]
  99. Zelante, T.; Iannitti, R.G.; Cunha, C.; De Luca, A.; Giovannini, G.; Pieraccini, G.; Zecchi, R.; D’Angelo, C.; Massi-Benedetti, C.; Fallarino, F.; et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 2013, 39, 372–385. [Google Scholar] [CrossRef]
  100. Gottesman, S.; Storz, G. Bacterial small RNA regulators: Versatile roles and rapidly evolving variations. Cold Spring Harb. Perspect. Biol. 2011, 3. [Google Scholar] [CrossRef]
  101. Lässer, C.; Alikhani, V.S.; Ekström, K.; Eldh, M.; Paredes, P.T.; Bossios, A.; Sjöstrand, M.; Gabrielsson, S.; Lötvall, J.; Valadi, H. Human saliva, plasma and breast milk exosomes contain RNA: Uptake by macrophages. J. Transl. Med. 2011, 9, 9. [Google Scholar] [CrossRef] [PubMed]
  102. Deng, Z.; Rong, Y.; Teng, Y.; Mu, J.; Zhuang, X.; Tseng, M.; Samykutty, A.; Zhang, L.; Yan, J.; Miller, D.; et al. Broccoli-derived nanoparticle inhibits mouse colitis by activating dendritic cell AMP-activated protein kinase. Mol. Ther. 2017, 25, 1641–1654. [Google Scholar] [CrossRef]
  103. Ju, S.; Mu, J.; Dokland, T.; Zhuang, X.; Wang, Q.; Jiang, H.; Xiang, X.; Deng, Z.-B.; Wang, B.; Zhang, L.; et al. Grape exosome-like nanoparticles induce intestinal stem cells and protect mice from DSS-induced colitis. Mol. Ther. 2013, 21, 1345–1357. [Google Scholar] [CrossRef]
  104. Tjalsma, H.; Boleij, A.; Marchesi, J.R.; Dutilh, B.E. A bacterial driver-passenger model for colorectal cancer: Beyond the usual suspects. Nat. Rev. Microbiol. 2012, 10, 575–582. [Google Scholar] [CrossRef]
  105. Warburg, O. On the origin of cancer cells. Science 1956, 123, 309–314. [Google Scholar] [CrossRef]
  106. Bishop, K.S.; Xu, H.; Marlow, G. Epigenetic regulation of gene expression induced by butyrate in colorectal cancer: Involvement of microRNA. Genet. Epigenet. 2017, 9, 1179237X17729900. [Google Scholar] [CrossRef]
  107. Hu, S.; Dong, T.S.; Dalal, S.R.; Wu, F.; Bissonnette, M.; Kwon, J.H.; Chang, E.B. The microbe-derived short chain fatty acid butyrate targets miRNA-dependent p21 gene expression in human colon cancer. PLoS ONE 2011, 6, e16221. [Google Scholar] [CrossRef]
  108. 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. [Google Scholar] [CrossRef]
  109. Hirayama, A.; Kami, K.; Sugimoto, M.; Sugawara, M.; Toki, N.; Onozuka, H.; Kinoshita, T.; Saito, N.; Ochiai, A.; Tomita, M.; et al. Quantitative metabolome profiling of colon and stomach cancer microenvironment by capillary electrophoresis time-of-flight mass spectrometry. Cancer Res. 2009, 69, 4918–4925. [Google Scholar] [CrossRef]
  110. Brown, D.G.; Rao, S.; Weir, T.L.; O’Malia, J.; Bazan, M.; Brown, R.J.; Ryan, E.P. Metabolomics and metabolic pathway networks from human colorectal cancers, adjacent mucosa, and stool. Cancer Metab. 2016, 4, 11. [Google Scholar] [CrossRef]
Figure 1. Host–microRNA–microbiota interactions in colorectal cancer. Microbiota composition has a functional effect on the cancer cells, via stromal and tumor infiltrating immune cells by regulating various cellular process (1). Microbial-metabolites and other secreted factors affect miRNA/gene expression profiles in cells present in the tumor microenvironment. In turn, tumor cells affect the microbiota composition of the stromal and tumor infiltrating immune cells through shedding of epithelial cells and/or secreting extracellular vesicles (EVs) containing miRNAs (2). The tumor-miRNAs alter the microbiota composition by affecting the gene expression of the microbiota and by delivering cancer-secreted metabolites (3). The tumor-derived miRNAs also have a role in regulating stromal and tumor infiltrating immune cells by affecting gene expression through miRNAs delivered in EVs (4). Such interactions will finally create a favorable microenvironment for tumor cells that include angiogenesis, immune evasion, and microbiota composition (5).
Figure 1. Host–microRNA–microbiota interactions in colorectal cancer. Microbiota composition has a functional effect on the cancer cells, via stromal and tumor infiltrating immune cells by regulating various cellular process (1). Microbial-metabolites and other secreted factors affect miRNA/gene expression profiles in cells present in the tumor microenvironment. In turn, tumor cells affect the microbiota composition of the stromal and tumor infiltrating immune cells through shedding of epithelial cells and/or secreting extracellular vesicles (EVs) containing miRNAs (2). The tumor-miRNAs alter the microbiota composition by affecting the gene expression of the microbiota and by delivering cancer-secreted metabolites (3). The tumor-derived miRNAs also have a role in regulating stromal and tumor infiltrating immune cells by affecting gene expression through miRNAs delivered in EVs (4). Such interactions will finally create a favorable microenvironment for tumor cells that include angiogenesis, immune evasion, and microbiota composition (5).
Genes 10 00270 g001

Share and Cite

MDPI and ACS Style

Yuan, C.; Steer, C.J.; Subramanian, S. Host–MicroRNA–Microbiota Interactions in Colorectal Cancer. Genes 2019, 10, 270. https://doi.org/10.3390/genes10040270

AMA Style

Yuan C, Steer CJ, Subramanian S. Host–MicroRNA–Microbiota Interactions in Colorectal Cancer. Genes. 2019; 10(4):270. https://doi.org/10.3390/genes10040270

Chicago/Turabian Style

Yuan, Ce, Clifford J. Steer, and Subbaya Subramanian. 2019. "Host–MicroRNA–Microbiota Interactions in Colorectal Cancer" Genes 10, no. 4: 270. https://doi.org/10.3390/genes10040270

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop