Next Article in Journal
Ruthenium Complexes with Protic Ligands: Influence of the Position of OH Groups and π Expansion on Luminescence and Photocytotoxicity
Next Article in Special Issue
The Role of Next-Generation Probiotics in Obesity and Obesity-Associated Disorders: Current Knowledge and Future Perspectives
Previous Article in Journal
Cadmium Disrupted ER Ca2+ Homeostasis by Inhibiting SERCA2 Expression and Activity to Induce Apoptosis in Renal Proximal Tubular Cells
Previous Article in Special Issue
Irradiation-Induced Dysbiosis: The Compounding Effect of High-Fat Diet on Metabolic and Immune Functions in Mice
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Gut Microbiome Composition and Its Metabolites Are a Key Regulating Factor for Malignant Transformation, Metastasis and Antitumor Immunity

by
Stefan Lozenov
1,
Boris Krastev
2,
Georgi Nikolaev
3,*,
Monika Peshevska-Sekulovska
4,
Milena Peruhova
5 and
Tsvetelina Velikova
6
1
Laboratory for Control and Monitoring of the Antibiotic Resistance, National Centre for Infectious and Parasitic Diseases, 26 Yanko Sakazov Blvd, 1504 Sofia, Bulgaria
2
Nadezhda Paradise Medical Center, 1330 Sofia, Bulgaria
3
Department of Cell and Developmental Biology, Faculty of Biology, Sofia University “St. Kliment Ohridski”, 1504 Sofia, Bulgaria
4
Department of Gastroenterology, University Hospital Lozenetz, Sofia, Medical Faculty, Sofia University “St. Kliment Ohridski”, 1407 Sofia, Bulgaria
5
Department of Gastroenterology, University Hospital Heart and Brain, 5804 Pleven, Bulgaria
6
Medical Faculty, Sofia University St. Kliment Ohridski, Kozyak 1 str., 1407 Sofia, Bulgaria
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(6), 5978; https://doi.org/10.3390/ijms24065978
Submission received: 1 February 2023 / Revised: 14 March 2023 / Accepted: 20 March 2023 / Published: 22 March 2023

Abstract

:
The genetic and metabolomic abundance of the microbiome exemplifies that the microbiome comprises a more extensive set of genes than the entire human genome, which justifies the numerous metabolic and immunological interactions between the gut microbiota, macroorganisms and immune processes. These interactions have local and systemic impacts that can influence the pathological process of carcinogenesis. The latter can be promoted, enhanced or inhibited by the interactions between the microbiota and the host. This review aimed to present evidence that interactions between the host and the gut microbiota might be a significant exogenic factor for cancer predisposition. It is beyond doubt that the cross-talk between microbiota and the host cells in terms of epigenetic modifications can regulate gene expression patterns and influence cell fate in both beneficial and adverse directions for the host’s health. Furthermore, bacterial metabolites could shift pro- and anti-tumor processes in one direction or another. However, the exact mechanisms behind these interactions are elusive and require large-scale omics studies to better understand and possibly discover new therapeutic approaches for cancer.

1. Introduction

The human gastrointestinal microbiota comprises the bacteria, archaea and eukarya species colonizing the human gut. Moreover, the total amount of genetic material exceeds that of the genetic material of the human host [1]. There are more than 1000 presently known species, distributed mainly among the phyla Bacteroidetes and Firmicutes (around 90%) and the rest among Proteobacteria, Verrucomicrobia and Actinobacteria [2]. However, the human gut microbiota demonstrates high geographical, interpersonal and age variability [1,2]. Moreover, its genetic and metabolomic abundance comprises a more extensive set of genes than the entire human genome [3]. The latter justifies the numerous metabolic and immunological interactions between the gut microbiota and the host, with variations in plasma metabolites and the host immune system activity correlating to the gut microbiota signature [4,5,6]. These host–microbiota interactions have local and systemic impacts, including on the pathological processes of carcinogenesis.
It has been demonstrated that carcinogenesis could be promoted or suppressed by the interaction between the microbiome and the host. For example, a considerable amount of evidence for the role of the gut microbiota and intestinal dysbiosis in the development of colorectal cancer exists [7,8]. There is also mounting evidence for the significance of gut microbiota for other non-intestinal locations of carcinogenesis, such as breast and lung cancer [9,10].
While the potential gut microbiota signatures are endless and numerous correlations can be made, in our opinion, it is convenient to approach the complex interactions between gut microbiota and cancer from the viewpoint of the major oncogenic and tumor-suppressive pathways involved. Therefore, this paper aims to review the most up-to-date information on the complex tumor-promoting and tumor-suppressing interplay between gut microbiota and cancer. Furthermore, we aimed to provide evidence that interactions between the host and the gut microbiota might be a significant exogenic factor for cancer predisposition.
Our search strategy was as follows. We conducted a modified form of a biomedical narrative review according to recent recommendations [11]. We performed a search through scientific databases Medline (PubMed) and Scopus. Both MeSH and relevant free-text terms were used: (“microbiome” OR “gut microbiota”) AND (“oncogenesis” OR “carcinogenesis”). Additionally, we searched for (“microbiome” OR “gut microbiota”) AND “colorectal cancer”, “interleukin pathways”, “NF-kB”, “Th17 cells”, “tumor suppression” and “HDACs”. Our search was confined to articles published up to January 2023. Secondly, relevant data were also derived from sources using the search engine Google Scholar. Finally, the references of retrieved publications were further hand-searched for supplements.

2. Local Oncogenic Effects of Gut Microbiota

The gut microbiota can promote local colonic oncogenesis through the production of cancerogenic metabolites, oncogenic exotoxins and the axis of chronic inflammation, including biofilm production, pathogenic adhesins stimulation and local immune system mediation [12].
For example, the enterotoxigenic Bacteroides fragilis strains (EBFT) produce one of the three isotypes of a 20 kDa zinc metalloprotease (BFT-1, BFT-2 and BFT-3). BFT modify the permeability of the colonic epithelium, inducing colitis, and promote cell proliferation through the NF-kB and the MAPK pathways [13]. In addition, it was revealed in a mouse model that BFT enhance the oncogenesis of colorectal cancer (CRC) through the Th17 pathway, and EBFT has been linked to an increased risk of CRC in human patients [12,13]. Recently, our team demonstrated that upregulated IL-6 is crucial for both inflammatory bowel diseases and CRC development, whereas Th17/T regulatory (Treg) cells and related genes are activated primarily in CRC [14]. It has been suggested that chronic inflammation mediated by Th17 cells and related cytokines is associated with an increased risk of malignant transformation.
Another prominent group of microbiota toxins interferes with the DNA and cell cycle of the intestinal epithelium. Colibactin is a byproduct of the pks genetic island in some Enterobacteriaceae, particularly Escherichia coli. It is an unstable compound, which, when introduced directly on the mucosal surfaces or intracellularly, causes DNA alkylation, interstrand crosslinking and double-strand breaks [15,16]. Then, DNA damage translates into mutagenesis and carcinogenesis potential, which has been demonstrated on epithelial cell lines and linked to colon cancer risk [17,18].
A Swedish study discovered colibactin-producing bacteria in 56% of the CRC samples versus 19% of control samples [19]. The cytolethal distending toxin (cdt) is another potent bacterial toxin isolated initially from E. coli and consequently described in Shigella, Campylobacter and other Gram-negative bacteria. It can be coded both on the bacterial chromosome, but also on plasmid vectors. Cdt is a virulence factor that enhances the invasive properties of its carriers through damage to the epithelial layer of the intestinal mucosa. Cdt also extensively suppresses lymphocytes and macrophages [20,21,22]. The mechanism of action of cdt relies on its structural similarity to the human DNAse I protein family that leads to cell cycle damage through the infliction of double-strand breaks. The described processes are often observed in mutagenesis and carcinogenesis [23]. It has been demonstrated in animal models that mice bearing cdt-positive Campylobacter jejuni are more likely to develop CRC and larger tumors [24].
The typhoid toxin (TT) found in Salmonella also induces double strand breaks in a similar manner, as the active part of TT actually comprises the cdt B subunit. However, TT is unique in being directly linked to an increased risk of developing CRC in humans. A recent Dutch study demonstrated a statistically significant increase in standardized incidence risk for CRC development after salmonellosis infection. The overall risk increased by 1.54 times for the age group above 20 and below 60 years, while for the age groups 20–39 and 40–49, the increases were 2.55 and 1.62 times, respectively [25,26].
Gut microbiota can also promote oncogenesis in a non-DNA-related way; intestinal commensals can promote inflammation-related epithelial cell proliferation in an IL-6 STAT3-dependent way, in IL-17C dependent way or through the PI3K-Akt-axis [27]. These proinflammatory pathways relate to specific virulence factors enhancing biofilm production, specific adhesins and immune cell recruitment in the tumor stroma. For example, Fusobacterium nucleatum was related to IL-17A-related inflammation in human CRC, and Peptostreptococcus anaerobius was described to induce a proinflammatory response in the tumor stroma, contributing to tumor progression via the recruitment of tumor-infiltrating immune cells [27,28].
The above mechanisms are further implied by novel sequencing techniques suggesting there are specific gut microbiota signatures in CRC patients. A few studies have demonstrated the loss of alpha diversity of the gut microbiota of CRC patients [29,30,31]. A large meta-analysis of 386 samples from CRC patients and 392 tumor-free controls revealed heterogenic data on the alpha diversity but demonstrated the prevalence of 29 bacterial species in the CRC samples, among which were the already discussed Fusobacterium and Peptostreptococcus [32]. The same study also demonstrated significant enrichment in the pks gene already discussed above and also of the fadA gene (and adhesin and virulence factor of F. nucleatum). Another large study demonstrated a correlation between a western style diet and high levels of pks positive E. coli and CRC [33]. Additionally, while some of the largest of these studies demonstrate significant correlations in samples from cancer patients versus healthy controls, further prospective long-term studies will be needed to further elaborate on the role of metagenomic profiles and their contribution to the development of CRC and to exclude the potential reciprocal relationship where CRC impacts the local flora.

3. Systemic Oncogenic Effects Related to Gut Microbiota

While the local effects of gut microbiota on the intestinal epithelium, causing local inflammation and induction of IL-17 mediated stimulation of the NF-kB pathway towards enhanced proliferation, are well established, it is interesting to know if these pathways can have a more systemic impact other than on intestinal epithelium tissues [34]. For example, increased IL-17 in the bone marrow microenvironment can promote the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-kB), which in turn enhances disease activity in particular bone marrow cancers, such as multiple myeloma (MM) [34].
Short-chain fatty acids (SCFAs) are gut microbiota products that may have anti-inflammatory properties, locally and systemically. Dysbiosis shifting the intestinal microbiota balance towards bacterial species producing less SCFA, such as Klebsiella spp., could change the balance towards more prominent NF-kB activation. These observations were described in MM patients [34].
Prevotella heparinolytica, as a distinct member of the gut microbiota, is also related to the stimulation of the Th17 response, promoting NF-kB activation in the bone marrow and enhancing progression from monoclonal gammopathy of undetermined significance (MGUS) to MM [35,36].
In line with this, the NF-kB pathway is involved in many systemic hematological malignancies such as chronic lymphocytic leukemia (CLL), mucous membrane-associated lymphoma (MALT), diffuse large B-cell lymphoma (DLBCL) and acute lymphoblastic leukemia (ALL) [37]. Additionally, while the direct prognostic link between human hematological malignancies and specific gut microbiota signatures has yet to be demonstrated, a few studies have shown decreased gut microbiome diversity in patients with hematological malignancies [35].
A small American study of 51 pediatric and adolescent subjects (ALL patients and their healthy siblings) revealed a reduced microbial diversity of the gut flora in ALL patients compared to the healthy controls [38]. In addition, a Chinese group compared 25 DLBCL patients at the time of diagnosis to 26 healthy controls, demonstrating an increased abundance of Escherichia-Shigella, Enterococcus, Veillonella and Prevotella at the genus level in the lymphoma patients and a different beta-diversity pattern between DLBCL patients and healthy controls [39].
The spreading of tumors has also been related to the gut microbiota. Several mechanisms and strategies are employed by microbiota to regulate cancer progression and metastasizing.
Chronic exposure to bacterial metabolites causes chronic inflammation if the epithelium’s barrier function is compromised or if the tumor or metastases are persistently colonized with bacteria. Inflammation can then promote tumor initiation, development, progression and metastasis via pathways linked to cancer stem cell compartment control, immunosuppression, mutation induction, tumor microenvironment modulation and stromal cell modulation [40]. One of the most prominent mechanisms is bacteria’s ability to activate innate and adaptive immune cells within the tumor environment via Toll-like receptors, NOD-like receptors and other pattern recognition receptors to stimulate the production of pro-inflammatory cytokines, which not only regulate immunity and the microenvironment, but also serve as tissue-protective and repair responses in epithelial and cancer cells [41].
Additionally, in response to microbe-driven inflammation, cell plasticity causes cells to move from one stage of differentiation to another, promoting epithelial-mesenchymal transition and metastasis. Microbiota and microbial products can govern metastatic survival locally (e.g., in the liver, which is often rich in microbe-derived signals) or systemically, as in the case of metastasis in the lungs. By activating inflammation-driven NF-kB signaling, lipopolysaccharide (LPS) was demonstrated to enhance the survival and expansion of cancer cells metastasizing to the lung. LPS also increased metastatic dissemination by enhancing cancer cell integrin-mediated adherence to endothelial cells in vessels [42].
The interaction between the host`s immune system, microbiome and cancer cells is presented in Figure 1.

4. Tumor Suppressor Effects Related to Gut Microbiota

Significant amount of research has recently been directed toward elucidating how commensal bacteria impact antitumor immunity. Several hypotheses speculate that the gut microbiome has the potential to enhance the host’s immune responses against tumor cells, which are covered in detail in the following paragraphs.
The immune system evolved as an evolutionary protective mechanism to recognize and remove neoplastic cells, preventing cancer from spreading to other organs. Tumor immune surveillance involves a variety of immunological effector pathways, including innate and adaptive mechanisms [43]. Long-term chronic activation of immune cells by tumor antigens and uncontrolled inflammation associated with carcinogenesis, on the other hand, might eventually weaken antitumor immunity and accelerate tumor growth. As a result, tumor cells’ capacity to avoid and inhibit antitumor immunity is a characteristic of malignancy.
Furthermore, tumor cells avoid detection by suppressing antigen-presenting machinery and interferon (IFN) signaling pathways. Tumors also create an immunosuppressive microenvironment by recruiting myeloid-derived suppressor cells (MDSCs) and Tregs and producing pro-tumor and anti-inflammatory chemicals such as TGF-β, IL-10 and IDO [43].
However, the processes by which the gut microbiota impacts the efficacy of immunotherapy are currently being studied. Preclinical and preliminary clinical evidence from various gut microbiota modulation trials, including fecal transplantation, probiotics, consortia and nutrition, show that favorable microbiota modification is linked to enhanced intratumoral infiltration of CD8+ effector T cells. Furthermore, this infiltration of CD8+ T cells is frequently accompanied by the increased intratumoral activity of Th1 cells and dendritic cells and a decreased density of immunosuppressive cells [44].
One such proposed mechanism relies on the argument that microbiota-related antigens stimulate T cell reactivity against structurally similar tumor antigens based on the principle of cross-reactivity. Some evidence corroborating this hypothesis came from a study on long-term pancreatic cancer survivors [45].
By methods of whole-exome sequencing and in silico neoantigen prediction, the authors identified neoantigen-specific T cells in the peripheral blood and tumor-infiltrating lymphocytes which harbored specific reactivity to both tumor and bacterial epitopes [46]. This finding is part of a broader discussion of whether antigen mimicry between certain bacterial strains and tumor cells could lead to better anticancer immune surveillance.
CD4+ and CD8+ T cells are primarily responsible for tumor growth and cancer progression. As a result, the effectiveness of some immunotherapy, such as immune checkpoint inhibitors (ICIs), primarily depends on T cells [43]. Notably, the gut microbiota is a tumor-extrinsic component that can influence the effectiveness of cancer immunotherapy using ICIs by modulating antitumor defensive systems, as we also demonstrated recently [47].
As discussed above, SCFAs, aryl hydrocarbon receptors (AhR) and the Toll-like and nod-like receptor (TLR and NLR) ligands are all microbiome-derived compounds that can regulate the immune system locally and systemically. Through dietary fiber fermentation, anerobic commensal bacteria of the Bacteroidetes and Firmicutes phyla create SCFAs such as acetate, propionate and butyrate. Butyrate and propionate, generated by fiber-fermenting commensal microorganisms, are connected to the formation of Foxp3+ Tregs by blocking histone deacetylases (HDAC) within the cell [48]. Although the in vivo instability of induced Tregs (iTregs) has been demonstrated previously, in the study by Souza Vieira et al., T cells may have lost FOXP3 expression throughout the sensitization stage and therefore did not exert a significant effect in their research.
Furthermore, the gut microbiome may stimulate adaptive immune responses by targeting tumor cells. For example, Bifidobacterium spp. stimulates tumor-specific T cells, boosts CD8+ T cell accumulation in melanoma and bladder cancers and increases IFN production. This may limit cancer cell proliferation by downregulating the NF-kB signaling pathway [49,50]. Furthermore, the Bifidobacterium strain improves antitumor immune responses in mice with colon cancer by raising CD4+ and CD8+ T cells and NK cells and the CD4+/Treg, CD8+/Treg and effector CD8+/Treg ratios [51]. Furthermore, the gut microbiome influences colitis-associated carcinogenesis by modulating the number of IFN-producing CD8+ T cells [52].
The Prevotellaceae and Anaeroplasmataceae families are predictive of high and low tumor burdens of colon cancer, respectively [52]. According to Li Y et al., bacterial strains, particularly Bacteroides and Lactobacillus spp., are related to better antitumor immunity, increased tumor infiltration by tumor-specific CD45+, CD4+, and CD8+ T cells and increased IFNg, TNFa and IL-2 production. This, in turn, may limit melanoma progression in Rnf5/ mice [53]. In general, the gut microbiota and its metabolites regulate antitumor immunity by many pathways, including Th1 and Th17 cell growth, production of pro-inflammatory cytokines and activation of MDSCs and NK cells [53].
Microbiota-derived epitopes may stimulate antigen-presenting cells for more T cell growth and cytokine production, thus boosting the systemic response to cancer immunotherapy [43]. Given the importance of gut microbiota in balancing antitumor vs. pro-tumor immune responses, developing microbiome screening and treatment techniques to help tilt the balance in favor of antitumor immunity is critical. Given the harmful effects of antibiotics on several essential components of innate and adaptive immunity, it is crucial to avoid broad-spectrum antibiotics as much as possible in cancer patients undergoing ICI treatment. More clinical research is needed to establish the potential efficacy of microbiome-supportive treatments such as fecal microbiota transplantation (FMT) from healthy donors in cancer patients who require antibiotic therapy [43].
However, while the cross-reactivity hypothesis sounds plausible, there are still some controversies surrounding identifying the exact epitope structure that drives T cell cross-reactions. Additionally, microbiome could probably do much more than stimulate T cell cross-reactivity against structurally similar epitopes. For example, recent research demonstrated that among the T cell repertoire, there are subpopulations of memory cells that possess specificity to neoantigens with entirely ‘unknown’ structures. The latter are usually antigens to which the immune system has not been previously exposed, and there is no antigen mimicry [46].
Memory T cells with specific reactivity to human immunodeficiency virus (HIV) were identified in blood samples from HIV-seronegative adults, but such cellular clones could not be detected at birth. Moreover, these memory immune cells are activated when treated with specific microbiome-produced peptides [46].
All this suggests that commensal gut bacteria might play a role in generating, sustaining and stimulating memory T cells against unencountered yet viral epitopes. In addition, one could argue that a similar response could be induced against neoepitopes emerging from tumor initiation and subsequent cancer progression.
Apart from the direct effects on the adaptive immune system, the microbiome can stimulate an antitumor immune response by rendering pattern recognition receptors on cells belonging to the innate immunity, e.g., dendritic cells (DC). In vitro experiments demonstrated that under the influence of bacterial strains such as Lactobacillus, DCs become activated, undergo maturation and begin producing cytokines (IL-12, IL18 and IFN-γ) which trigger cellular cytotoxicity [54].
Lactobacillus species differentially activate Toll-like receptors and downstream signals in dendritic cells. Some probiotic strains such as Lactobacillus casei BL23 could exert antitumor properties via the stimulation of the IL-2 signaling pathway. This was well presented in a study on HPV-induced cancers in mice [55]. It was first demonstrated in vitro how this bacterial species could stimulate bone marrow DC to produce IL-2.
A recent study demonstrated the tumor suppressing properties of Lactobacillus gallinarum that were realized through the secretion of protective metabolites enhancing CRC cells apoptosis in mice models. Metabolomic studies and mass spectrometry revealed an increase in indole-3-lactic acid [56].
Thereafter, an in vivo analysis revealed an inverse correlation between IL-2 levels and tumor size, i.e., mice with higher levels of IL-2 had a much slower tumor growth. In addition to promoting IL-2-mediated antitumor activity, Lactobacillus casei BL23 was able to recruit natural killer (NK) cells with high cytotoxic potential against cancer. Other commensal species, such as Bacillus mesentericus, Clostridium butyricum and Enterococcus faecalis, can also lead to DC activation and enhanced antigen presentation of tumor antigens [57].
Furthermore, it was reported that when applied as a probiotic combination, these bacteria could skew CD4 differentiation towards the Th1 phenotype and stimulate peripheral blood mononuclear cells (PBMC) and DC. Another interesting fact is that the combination of microorganisms could induce immune changes on a grander scale than each species alone. This raises a question of potential synergism between different bacteria constituting normal gut flora.
An overview of the anticancer and tumor-promoting mechanisms of gut microbiota is presented in Figure 2.

5. Role of the Microbiota-Produced Metabolites

Commensal bacteria provide neoantigens to the host immune system and produce a wide range of metabolites, some of which have notable tumor-suppressing features. SCFAs are among the best-studied byproducts of the gut microbiome, with butyrate and propionate most famous for their antitumor properties. Butyrate is produced during the fermentation of fiber-rich food in the colon [58]. It plays an essential role in mitochondrial beta-oxidation. As tumor cells rely on alternative pathways for energy metabolism (aerobic glycolysis), they cannot utilize butyrate efficiently, and it accumulates in their nuclei. This leads to histone deacetylase (HDAC) inhibition and, ultimately, to tumor cell apoptosis [58].
In vivo experiments have demonstrated that when tumor-bearing mice are colonized with butyrate-producing bacteria, such as Ruminococcus, Clostridium, Eubacterium and Coprococcus, their tumors exhibit diminished tumor growth compared to their counterparts whose microbiome is incapable of producing butyrate [58]. Butyrate and propionate can also exert a local antitumor effect on intestinal mucosa by suppressing inflammation predisposed to carcinogenesis. In addition, both metabolites are known to interact with G-protein coupled receptors (GPCR) such as GPR109A and GRP43 [59,60].
After binding GPR109A, propionate induces inflammatory cytokine secretion from colonic macrophages and dendritic cells. This leads to T cell differentiation towards the immunosuppressive T regulatory phenotype. Butyrate also has an affinity to GPR109A, which is how this metabolite stimulates the production of IL-18, a cytokine with a defined role in preventing the development of colon tumors [61]. On the other hand, butyrate suppresses IL-18 production by suppressing caspase 1 or NLRP3 inflammasome [62].
Moreover, butyrate has remarkable anticancer properties due to its potential to skew a negative impact on the cell development of malignant colonocytes while being the primary energy source for normal colonocytes. It was shown that butyrate suppresses glucose metabolism of colorectal cancer cells via the GPR109a-AKT signaling pathway, thus enhancing chemotherapy [63].
Data from in vivo models of GPR109A-deficient mice support the antitumor properties of SCFAs. These animals showed an increased propensity to form colon tumors, suggesting that interactions between SCFA and GPCR might be responsible for the tumor-protective effect of a healthy diet and normal gut flora [64]. There is also speculation that the balance between pro- and anti-inflammatory regulation depends on the concentration of SCFAs, where lower levels of butyrate could induce T reg differentiation and higher concentrations could skew T cells towards the inflammatory Th1 phenotype [64].
Besides the production of metabolites, the gut microbiome is also involved in metabolizing endogenous substances such as bile acids. Under the influence of intestinal flora, primary bile acids are converted to secondary bile acids, and both (primary and secondary bile acids) can exert immunoregulatory effects, mostly in the liver, where they enter via the enterohepatic circulation [65].
Recent data suggest that by the controlling bile acid metabolism, the gut microbiome could either augment or impair local hepatic antitumor immunity [66]. Authors have reported that manipulating the gut bacterial composition in mouse models could increase the accumulation of CXCR6+ NK cells in the liver and enhance interferon-y production. Moreover, these NK cells showed selective antitumor activity against liver malignancies [66]. A proposed mechanism behind CXCR6+ NK accumulation in the liver is based on the microbiome-controlled conversion of primary bile acids into secondary ones. It was demonstrated that when mice were treated with antibiotics against Gram-positive bacteria, this impaired bile acid conversion and increased primary bile acids. The latter led to increased CXCL16 expression from sinusoidal endothelial cells, which bonded to CXCR6 on CXCR6 + NK cells and enhanced their accumulation in the liver [66].
Other metabolites, such as trimethylamine-N-oxide (TMAO), hydrogen sulfide (H2S), N-nitroso compounds (NOCs), DCA, deoxycholic acids, HCAs, heterocyclic amines, etc., are critical signaling molecules that mediate the cross-talk between the host and the microbiota and play a significant role in colorectal carcinogenesis [67].
Furthermore, these metabolites are related to the etiology and severity of CRC. It was shown that some of these metabolites could act directly on the integrity of the mucosal barrier and immune responses in the gut, triggering the release of proinflammatory cytokines (i.e., TNFa and IL-17) [67]. This usually correlates with immune escape and the immunosuppression state that promote tumorigenesis. Additionally, the mentioned metabolites may damage DNA via several pathways. For example, DCAs promote tumor formation through the RAS-ERK1/2 signaling pathway, the Wnt/b-catenin signaling pathway and the PKC/p38 MAPK signaling pathway. Trimethylamine (TMA), produced from choline, carnitine and phosphatidylcholine from food processing by microbiota and then transported and oxidized to TMAO in the liver, increases the risk of developing CRC. Experimental models showed that hydrogen sulfide damages DNA through free radical oxygen and genotoxicity. NOCs also lead to DNA damage and NOC-induced DNA adducts by different mechanisms: inducing mutations in the K-ras gene (G→A) and oxidative stress. Experimental studies showed that HCA is also associated with DNA damage and the formation of DNA adducts. Interestingly, lactate metabolites also promote CRC cell proliferation, invasion and migration by providing environmental conditions and stimulating the glycolytic metabolism and angiogenesis [67].
The microbiota-derived metabolites are presented in Table 1.
It is essential to mention that these metabolites usually act together. For example, TMAO participates in NOC formation, leading to DNA damage and epigenetic changes [67].
In numerous studies, however, alteration of the gut microbiota has been linked to gastrointestinal disorders such as CRC, although cancer is considered a multifactorial and multistage disease. However, the function of microorganisms in the initiation and progression of colorectal cancer has become apparent. Convincing models have been presented to depict the complex and dynamic mechanisms and shifts in carcinogenesis [12,100].
Gut microbiota metabolites have also been assessed in human serum through untargeted and targeted metabolic approaches. A recent comprehensive metabolomic study among 44 healthy adenoma and CRC subjects demonstrated the distinct metabolic signature of patients with adenoma and CRC, correlated to the distinct increased bacterial species in adenoma and CRC subjects [101].

6. Histone Deacetylases Pathways and Tumor Cell Apoptosis

Microbiota-related epigenetic changes include DNA and histone modifications (i.e., acetylation and methylation) and non-coding RNAs (i.e., microRNAs and miR). Epigenetic regulation by the gut microbiota has been intensively studied in recent years, with many mechanisms and clinical implications demonstrated [102,103,104].
It was demonstrated that the gut epithelium secretes a range of miRNAs which can penetrate bacteria and impact their transcription and change the microbial community’s structure and diversity. Similarly, the gut microbiota produces several metabolites (i.e., butyrate and bile acids) which can modulate the human metabolism, including BMI, insulin secretion and fat synthesis. Dietary nutrient intake (e.g., folic acid, methionine and vitamin B12) supplies methyl donors that change host DNA methylation, which may modulate the inflammatory state of the intestine. Specific microbial metabolites can alter DNA methylation, histone acetylation and miRNAs, impairing intestinal homeostasis and suppressing beneficial microbiota while boosting the richness of harmful bacteria and promoting colorectal cancer [104,105].
We summarize the cross-talk between microbiota and epigenetic modifications in Figure 3.
The role of epigenetic modifications by histone-modifying enzymes in terms of cellular transformations is also an area of intensive research. Histone acetylation is a chromatin modification generally associated with opening the chromatin and allowing access to transcription factors. Acetylation happens on lysine residues by histone acetylases (HATs) and is reversed by histone deacetylases (HDACs) which have been found to play a particular role in aging and carcinogenesis. HATs and HDACs themselves are subject to regulation by post-translational modifications, protein–protein interactions, availability of cofactors and various signaling pathways [106].
The interaction between histone acetylation status and the gut microbiome is now recognized, although this was observed decades ago [102,107]. In the growing amount of possible molecular biomarkers relating gut microbes to health and disease, HDACs are attracting more attention due to the development of HDAC inhibitors as potential antitumor therapeutic agents [107].
HDACs may be involved in CRC onset and development in at least three directions.
Firstly, SCFAs, the primary metabolites produced by the gut microbiota, may act alone as HDAC inhibitors and thus promote a more transcriptionally active state of chromatin [108]. In addition, butyrate, propionate and acetate produced by the microbiota have been shown to act as such inhibitors and also increase the cytokine production of Th1 and CD4+ T cells, which could influence apoptosis and cell cycle arrest of cancer cells [109].
Another significant histone modification in the intestinal epithelium, crotonylation, is also stabilized by butyrate by inhibiting class I HDACs, which are related to cancer pathways [110]. Furthermore, the activity of HDAC3 that is highly expressed in the intestinal epithelium is also affected by SCFAs and seems to integrate multiple microbiota-derived signals [111,112]. Similar data are available for HDAC1 and HDAC2 [113,114,115]. Furthermore, HDACs may also control cell proliferation and apoptosis via modulation of the acetylation status of non-histone proteins such as p53 and tubulin [116].

7. Cancer Immunotherapy and Microbiome

Recent preclinical studies using cell culture and animal models, human clinical studies, and meta-analyses of clinical studies have revealed that the gut microbiota alters the host’s response to various anticancer drugs, with immunomodulation emerging as one of the central mechanisms facilitating these differential responses. Dysbiosis is not only a result, but also a cause, of varied reactions to treatment. Increased intestinal diversity, for example, was predictive of a lower mortality in patients receiving allogeneic hematopoietic stem cell transplantation (allo-HSCT) for treating hematological malignancies [117].
The fact that immunological modulation caused by increased microbial diversity determines the severity of graft vs. host illness is an essential factor for patients starting allo-HSCT. Furthermore, compositional changes caused by therapy might also be involved [118,119]. For example, checkpoint inhibitors, such as other cancer medicines, have significant inter-individual heterogeneity in patient responses [47].
A rising amount of research has already offered considerable insight into the effect of the gut microbiota on the xenobiotic metabolism, which might significantly impact future disease treatment. For example, gut microbial xenobiotic metabolites have altered bioavailability, bioactivity and toxicity [120,121]. In addition, they can interfere with the functions of human xenobiotic-metabolizing enzymes to influence the fate of other exogenic molecules.
However, bacteria may become pharmacological targets in the not-too-distant future. Microbial drug targets may also reduce the negative effects of many chemotherapeutics on the GI tract (i.e., adverse effects, such as those caused by irinotecan (camptothecin)). For example, the microbiota can reactivate the active form of the chemotherapeutic agent (SN38) in the GI lumen, which is then glucuronidated in the liver to generate the inactive SN38-G, which is eliminated via the GI system. Then, increased SN38 levels in the colon produce severe and possibly fatal diarrhea, necessitating dosage reduction and frequent dose adjustments [122]. In addition, incorporating fiber-rich, prebiotic foods, restricting red meat intake and lowering obesity rates could assist in reducing the global tumor burden in the long run by the beneficial effects caused by these actions, such as the production of SCFAs during the bacterial fermentation of plant-based fibers, which provide broad protection against the development of cancer [122].
Since dysbiosis appears to be a carcinogenesis precursor, it not only predates illness onset but also spreads during tumor growth. Therefore, maintaining eubiotics, or an optimum microbiota composition, is critical for avoiding disease-causing events. As a result, new, specialized, narrow-range antibiotics that preferentially target infections or pathobionts while maintaining eubiosis are needed [118,123,124,125,126].
Speaking of microbiota, precision medicine that offers medical therapies tailored to each patient’s genetic makeup and variances in lifestyle and environment also pays attention to microbiome interventions [127,128]. Given the wide variety of impacts that the microbiota has on human health, changes in patient composition should be considered when determining who will benefit from a given treatment technique. For example, as previously stated, the presence or absence of particular bacterial community members, or even their metabolites, can affect the occurrence, severity and therapy of cancer and may serve as prognostic biomarkers. Furthermore, greater access to centralized, cloud-based repositories for whole-genome and transcriptome sequencing information would aid computer scientists’ data mining methodologies. Combining pharmacogenomics data with unique microbial organisms or their particular metabolites will most likely allow for accurate dosage, symptom control and enhanced treatment responses in the future [129].
Furthermore, because antibiotics might alter the makeup of the gut microbiota, interfering with the impact of immunotherapy, the link between antibiotic-associated dysbiosis and immunotherapy is a hot issue. For example, Elkrief et al. discovered that using antibiotics before immunotherapy, such as anti-PD-1/PD-L1 and anti-CTLA-4, was an independent risk factor for lower progression-free survival (hazard ratio = 0.32, 95% confidence range = 0.13–0.83, p = 0.02) [128]. Furthermore, patients who received antibiotics before immunotherapy had a lower chance of responding effectively and a better prognosis.
Aside from using the gut microbiota to predict immunotherapy success, several clinical trials have focused on adjusting the gut microbiota composition to overcome anti-PD-1/PD-L1 resistance, including using FMT [129]. Even though numerous preclinical studies have shown that the gut microbiota regulates the host systemic immune response, modulates immunotherapy efficacy and influences treatment-induced adverse effects, the regulatory function of certain commensal bacteria requires further investigation, particularly for extrapolation from the mouse model to humans. The findings of these ongoing investigations may give more consistent data to demonstrate the viability of improving immunotherapy efficacy by modifying the gut microbiota makeup. It is worth noting, however, that the original gut mucosa commensals interfere with the colonization of supplementary probiotics. In addition, resistance to probiotic colonization varies amongst populations and may be impacted by baseline commensal status. As a result, the patient’s commensal background should be considered when modifying gut microbiota through therapies such as fecal transplantation [129].
In the case of CRC, several approaches have been investigated to target and modulate the gut microbiota composition, including both microbial physiology and/or their metabolites that cause or contribute to CRC directly or indirectly, for example, dietary interventions, antibiotic treatments, probiotics, prebiotics and postbiotics, as well as FMT. In addition, several experimental investigations have advanced our understanding of the function of gut biomodulators and microbe-based therapy as anticancer agents. Still, a practical clinical application in CRC prevention and management remains mainly missing. Finally, studies are being conducted to determine the effectiveness of tailored diets and biomodulators in restoring a eubiotic state to prevent and treat CRC [130,131], since the microbiota could facilitate cancer progression [132,133].
Our review has some limitations associated with the nature of the narrative review, and the data were not further analyzed statistically. However, we did our best to present the data comprehensively and discuss the studies critically. Furthermore, among the strengths of our review is that we gave the information logically by showing the data from animal studies and then described clinical trials and investigations that translated big data into practice. In addition, our review agglomerates gut microbial metabolites and signaling pathways of the related immune process, which has not been covered elsewhere.

8. Conclusions

In this review, we presented the available data and evidence supporting the idea that interaction between the host and the gut microbiota might be a significant exogenic factor for cancer predisposition. Various mechanisms by which bacteria could induce or prevent carcinogenesis are attaining more and more attention these days. The more we investigate, the more we learn about how diverse these mechanisms and pathways could be. We already know that the balance between commensal and pathogenic gut bacteria could affect intestinal mucosa permeability, local and systemic immunity and inflammation.
In addition, bacterial metabolites could stimulate either pro- or anti-tumor processes. Finally, it is beyond doubt that the cross-talk between microbiota and the host cells in terms of epigenetic modifications can regulate gene expression patterns, which may further influence cell fate in beneficial or adverse directions. However, the exact mechanisms behind these interactions are elusive. Therefore, to better understand them and possibly discover new therapeutic approaches, there is a need for large-scale omics studies.

Author Contributions

Conceptualization, B.K. and S.L.; methodology, G.N.; validation, B.K., G.N. and T.V.; formal analysis, B.K.; investigation, M.P.-S.; resources, M.P.; data curation, B.K.; writing—original draft preparation, B.K., S.L., G.N., M.P.-S. and M.P.; writing—review and editing, T.V.; supervision, T.V.; project administration, B.K.; funding acquisition, G.N. All authors have read and agreed to the published version of the manuscript.

Funding

The research is supported by the Ministry of Education and Science of Bulgaria, Grant DO1-308/20.12.2021 “INFRAAC” of NRRI 2021–2027. The APC was funded by the Ministry of Education and Science, Grant DO1-308/20.12.2021 “INFRAAC” of NRRI 2021–2027.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

T.V. is Grantee of the European Union-Next Generation EU, through the National Recovery and Resilience Plan of the Republic of Bulgaria, project № BG-RRP-2.004-0008-C01.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study, in the collection, analyses, or interpretation of data, in the writing of the manuscript, or in the decision to publish the results.

References

  1. Thursby, E.; Juge, N. Introduction to the human gut microbiota. Biochem. J. 2017, 474, 1823–1836. [Google Scholar] [CrossRef] [PubMed]
  2. Klement, R.J.; Pazienza, V. Impact of Different Types of Diet on Gut Microbiota Profiles and Cancer Prevention and Treatment. Medicina 2019, 55, 84. [Google Scholar] [CrossRef] [Green Version]
  3. Grice, E.A.; Segre, J.A. The human microbiome: Our second genome. Annu. Rev. Genomics Hum. Genet. 2012, 13, 151–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Van Treuren, W.; Dodd, D. Microbial Contribution to the Human Metabolome: Implications for Health and Disease. Annu. Rev. Pathol. 2020, 15, 345–369. [Google Scholar] [CrossRef] [Green Version]
  5. Mair, R.D.; Sirich, T.L.; Plummer, N.S.; Meyer, T.W. Characteristics of Colon-Derived Uremic Solutes. Clin. J. Am. Soc. Nephrol. 2018, 13, 1398–1404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Riazi-Rad, F.; Behrouzi, A.; Mazaheri, H.; Katebi, A.; Ajdary, S. Impact of gut microbiota on immune system. Acta Microbiol. Immunol. Hung. 2021, 68, 135–144. [Google Scholar] [CrossRef]
  7. Chen, W.; Liu, F.; Ling, Z.; Tong, X.; Xiang, C. Human intestinal lumen and mucosa-associated microbiota in patients with colorectal cancer. PLoS ONE 2012, 7, e39743. [Google Scholar] [CrossRef]
  8. Gao, Z.; Guo, B.; Gao, R.; Zhu, Q.; Qin, H. Microbiotammuneiss is associated with colorectal cancer. Front. Microbiol. 2015, 6, 20. [Google Scholar] [CrossRef]
  9. Xuan, C.; Shamonki, J.M.; Chung, A.; Dinome, M.L.; Chung, M.; Sieling, P.A.; Lee, D.J. Microbial dysbiosis is associated with human breast cancer. PLoS ONE 2014, 9, e83744. [Google Scholar] [CrossRef] [Green Version]
  10. Zhao, Y.; Liu, Y.; Li, S.; Peng, Z.; Liu, X.; Chen, J.; Zheng, X. Role of lung and gut microbiota on lung cancer pathogenesis. J. Cancer Res. Clin. Oncol. 2021, 147, 2177–2186. [Google Scholar] [CrossRef]
  11. Gasparyan, A.Y.; Ayvazyan, L.; Blackmore, H.; Kitas, G.D. Writing a narrative biomedical review: Considerations for authors, peer reviewers, and editors. Rheumatol. Int. 2011, 31, 1409–1417. [Google Scholar] [CrossRef] [PubMed]
  12. Cheng, Y.; Ling, Z.; Li, L. The Intestinal Microbiota and Colorectal Cancer. Front. Immunol. 2020, 11, 615056. [Google Scholar] [CrossRef] [PubMed]
  13. Hwang, S.; Lee, C.G.; Jo, M.; Park, C.O.; Gwon, S.Y.; Hwang, S.; Yi, H.C.; Lee, S.Y.; Eom, Y.B.; Karim, B.; et al. Enterotoxigenic Bacteroides fragilis infection exacerbates tumorigenesis in AOM/DSS mouse model. Int. J. Med. Sci. 2020, 17, 145–152. [Google Scholar] [CrossRef] [Green Version]
  14. Velikova, T.V.; Miteva, L.; Stanilov, N.; Spassova, Z.; Stanilova, S.A. Interleukin-6 compared to the other Th17/Treg related cytokines in inflammatory bowel disease and colorectal cancer. World J. Gastroenterol. 2020, 26, 1912–1925. [Google Scholar] [CrossRef] [PubMed]
  15. Wernke, K.M.; Xue, M.; Tirla, A.; Kim, C.S.; Crawford, J.M.; Herzon, S.B. Structure and bioactivity of colibactin. Bioorg. Med. Chem. Lett. 2020, 30, 127280. [Google Scholar] [CrossRef]
  16. Dougherty, M.W.; Jobin, C. Shining a Light on Colibactin Biology. Toxins 2021, 13, 346. [Google Scholar] [CrossRef]
  17. Iftekhar, A.; Berger, H.; Bouznad, N.; Heuberger, J.; Boccellato, F.; Dobrindt, U.; Hermeking, H.; Sigal, M.; Meyer, T.F. Genomic aberrations after short-term exposure to colibactin-producing E. coli transform primary colon epithelial cells. Nat. Commun. 2021, 12, 1003. [Google Scholar] [CrossRef]
  18. Strakova, N.; Korena, K.; Karpiskova, R. Klebsiella pneumoniae producing bacterial toxin colibactin as a risk of colorectal cancer development—A systematic review. Toxicon 2021, 197, 126–135. [Google Scholar] [CrossRef]
  19. Eklöf, V.; Löfgren-Burström, A.; Zingmark, C.; Edin, S.; Larsson, P.; Karling, P.; Alexeyev, O.; Rutegård, J.; Wikberg, M.L.; Palmqvist, R. Cancer-associated fecal microbial markers in colorectal cancer detection. Int. J. Cancer 2017, 141, 2528–2536. [Google Scholar] [CrossRef] [Green Version]
  20. Jinadasa, R.N.; Bloom, S.E.; Weiss, R.S.; Duhamel, G.E. Cytolethal distending toxin: A conserved bacterial genotoxin that blocks cell cycle progression, leading to apoptosis of a broad range of mammalian cell lineages. Microbiology 2011, 157 Pt 7, 1851–1875. [Google Scholar] [CrossRef] [Green Version]
  21. Shenker, B.J.; Walker, L.M.; Zekavat, Z.; Ojcius, D.M.; Huang, P.R.; Boesze-Battaglia, K. Cytolethal distending toxin-induced release of interleukin-1β by human macrophages is dependent upon activation of glycogen synthase kinase 3β, spleen tyrosine kinase (Syk) and the noncanonical inflammasome. Cell. Microbiol. 2020, 22, e13194. [Google Scholar] [CrossRef] [PubMed]
  22. Boesze-Battaglia, K.; Dhingra, A.; Walker, L.M.; Zekavat, A.; Shenker, B.J. Internalization and Intoxication of Human Macrophages by the Active Subunit of the Aggregatibacter actinomycetemcomitans Cytolethal Distending Toxin Is Dependent Upon Cellugyrin (Synaptogyrin-2). Front. Immunol. 2020, 11, 1262. [Google Scholar] [CrossRef] [PubMed]
  23. Frisan, T. Bacterial genotoxins: The long journey to the nucleus of mammalian cells. Biochim. Biophys. Acta 2016, 1858, 567–575. [Google Scholar] [CrossRef] [PubMed]
  24. He, Z.; Gharaibeh, R.Z.; Newsome, R.C.; Pope, J.L.; Dougherty, M.W.; Tomkovich, S.; Pons, B.; Mirey, G.; Vignard, J.; Hendrixson, D.R.; et al. Campylobacter jejuni promotes colorectal tumorigenesis through the action of cytolethal distending toxin. Gut 2019, 68, 289–300. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Martin, O.C.B.; Bergonzini, A.; Amico, F.; Chen, P.; Shay, J.W.; Dupuy, J.; Svensson, M.; Masucci, M.G.; Frisan, T. Infection with genotoxin-producing Salmonella enterica synergises with loss of the tumour suppressor APC in promoting genomic instability via the PI3K pathway in colonic epithelial cells. Cell. Microbiol. 2019, 21, e13099. [Google Scholar] [CrossRef] [Green Version]
  26. Mughini-Gras, L.; Schaapveld, M.; Kramers, J.; Mooij, S.; Neefjes-Borst, E.A.; Pelt, W.V.; Neefjes, J. Increased colon cancer risk after severe Salmonella infection. PLoS ONE 2018, 13, e0189721. [Google Scholar] [CrossRef] [Green Version]
  27. Ye, X.; Wang, R.; Bhattacharya, R.; Boulbes, D.R.; Fan, F.; Xia, L.; Adoni, H.; Ajami, N.J.; Wong, M.C.; Smith, D.P.; et al. Fusobacterium Nucleatum Subspecies Animalis Influences Proinflammatory Cytokine Expression and Monocyte Activation in Human Colorectal Tumors. Cancer Prev. Res. 2017, 10, 398–409. [Google Scholar] [CrossRef] [Green Version]
  28. Sears, C.L.; Pardoll, D.M. Perspective: Alpha-bugs, their microbial partners, and the link to colon cancer. J. Infect. Dis. 2011, 203, 306–311. [Google Scholar] [CrossRef] [Green Version]
  29. Chen, L.; Wang, W.; Zhou, R.; Ng, S.C.; Li, J.; Huang, M.; Zhou, F.; Wang, X.; Shen, B.; A Kamm, M.; et al. Characteristics of fecal and mucosa-associated microbiota in Chinese patients with inflammatory bowel disease. Medicine 2014, 93, e51. [Google Scholar] [CrossRef]
  30. Saffarian, A.; Mulet, C.; Regnault, B.; Amiot, A.; Tran-Van-Nhieu, J.; Ravel, J.; Sobhani, I.; Sansonetti, P.J.; Pédron, T.; Barnich, N.; et al. Crypt- and Mucosa-Associated Core Microbiotas in Humans and Their Alteration in Colon Cancer Patients. MBio 2019, 10, 1315–1319. [Google Scholar] [CrossRef] [Green Version]
  31. Ahn, J.; Sinha, R.; Pei, Z.; Dominianni, C.; Wu, J.; Shi, J.; Goedert, J.J.; Hayes, R.B.; Yang, L. Human gut microbiome and risk for colorectal cancer. J. Natl. Cancer Inst. 2013, 105, 1907–1911. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Wirbel, J.; Pyl, P.T.; Kartal, E.; Zych, K.; Kashani, A.; Milanese, A.; Fleck, J.S.; Voigt, A.Y.; Palleja, A.; Ponnudurai, R.; et al. Meta-analysis of fecal metagenomes reveals global microbial signatures that are specific for colorectal cancer. Nat. Med. 2019, 25, 679–689. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Arima, K.; Zhong, R.; Ugai, T.; Zhao, M.; Haruki, K.; Akimoto, N.; Lau, M.C.; Okadome, K.; Mehta, R.S.; Väyrynen, J.P.; et al. Western-Style Diet, pks Island-Carrying Escherichia coli, and Colorectal Cancer: Analyses from Two Large Prospective Cohort Studies. Gastroenterology 2022, 163, 862–874. [Google Scholar] [CrossRef] [PubMed]
  34. Jian, X.; Zhu, Y.; Ouyang, J.; Wang, Y.; Lei, Q.; Xia, J.; Guan, Y.; Zhang, J.; Guo, J.; He, Y.; et al. Alterations of gut microbiome accelerate multiple myeloma progression by increasing the relative abundances of nitrogen-recycling bacteria. Microbiome 2020, 8, 74. [Google Scholar] [CrossRef]
  35. Ahmed, N.; Ghannoum, M.; Gallogly, M.; de Lima, M.; Malek, E. Influence of gut microbiome on multiple myeloma: Friend or foe? J. Immunother. Cancer 2020, 8, e000576. [Google Scholar] [CrossRef]
  36. Calcinotto, A.; Brevi, A.; Chesi, M.; Ferrarese, R.; Garcia Perez, L.; Grioni, M.; Kumar, S.; Garbitt, V.M.; Sharik, M.E.; Henderson, K.J.; et al. Microbiota-driven interleukin-17-producing cells and eosinophils synergize to accelerate multiple myeloma progression. Nat. Commun. 2018, 9, 4832. [Google Scholar] [CrossRef] [Green Version]
  37. Imbert, V.; Peyron, J.F. NF-κB in Hematological Malignancies. Biomedicines 2017, 5, 27. [Google Scholar] [CrossRef] [Green Version]
  38. Rajagopala, S.V.; Yooseph, S.; Harkins, D.M.; Moncera, K.J.; Zabokrtsky, K.B.; Torralba, M.G.; Tovchigrechko, A.; Highlander, S.K.; Pieper, R.; Sender, L.; et al. Gastrointestinal microbial populations can distinguish pediatric and adolescent Acute Lymphoblastic Leukemia (ALL) at the time of disease diagnosis. BMC Genom. 2016, 17, 635. [Google Scholar] [CrossRef] [Green Version]
  39. Yuan, L.; Wang, W.; Zhang, W.; Zhang, Y.; Wei, C.; Li, J.; Zhou, D. Gut Microbiota in Untreated Diffuse Large B Cell Lymphoma Patients. Front. Microbiol. 2021, 12, 646361. [Google Scholar] [CrossRef]
  40. Robles, A.I.; Traverso, G.; Zhang, M.; Roberts, N.J.; Khan, M.A.; Joseph, C.; Lauwers, G.Y.; Selaru, F.M.; Popoli, M.; Pittman, M.E.; et al. Whole-Exome Sequencing Analyses of Inflammatory Bowel Disease-Associated Colorectal Cancers. Gastroenterology 2016, 150, 931–943. [Google Scholar] [CrossRef] [Green Version]
  41. Greten, F.R.; Grivennikov, S.I. Inflammation and Cancer: Triggers, Mechanisms, and Consequences. Immunity 2019, 51, 27–41. [Google Scholar] [CrossRef] [PubMed]
  42. Andreeva, N.V.; Gabbasova, R.R.; Grivennikov, S.I. Microbiome in cancer progression and therapy. Curr. Opin. Microbiol. 2020, 56, 118–126. [Google Scholar] [CrossRef] [PubMed]
  43. Aghamajidi, A.; Maleki Vareki, S. The Effect of the Gut Microbiota on Systemic and Antitumor Immunity and Response to Systemic Therapy against Cancer. Cancers 2022, 14, 3563. [Google Scholar] [CrossRef]
  44. Baruch, E.N.; Wang, J.; Wargo, J.A. Gut Microbiota and Antitumor Immunity: Potential Mechanisms for Clinical Effect. Cancer Immunol. Res. 2021, 9, 365–370. [Google Scholar] [CrossRef] [PubMed]
  45. Balachandran, V.P.; Łuksza, M.; Zhao, J.N.; Makarov, V.; Moral, J.A.; Remark, R.; Herbst, B.; Askan, G.; Bhanot, U.; Senbabaoglu, Y.; et al. Identification of unique neoantigen qualities in long-term survivors of pancreatic cancer. Nature 2017, 551, 512–516. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Su, L.F.; Kidd, B.A.; Han, A.; Kotzin, J.J.; Davis, M.M. Virus-specific CD4(+) memory-phenotype T cells are abundant in unexposed adults. Immunity 2013, 38, 373–383. [Google Scholar] [CrossRef] [Green Version]
  47. Velikova, T.; Krastev, B.; Lozenov, S.; Gencheva, R.; Peshevska-Sekulovska, M.; Nikolaev, G.; Peruhova, M. Antibiotic-Related Changes in Microbiome: The Hidden Villain behind Colorectal Carcinoma Immunotherapy Failure. Int. J. Mol. Sci. 2021, 22, 1754. [Google Scholar] [CrossRef]
  48. Vieira, R.S.; Castoldi, A.; Basso, P.J.; Hiyane, M.I.; Câmara, N.O.S.; Almeida, R.R. Butyrate Attenuates Lung Inflammation by Negatively Modulating Th9 Cells. Front. Immunol. 2019, 10, 67. [Google Scholar] [CrossRef]
  49. 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] [Green Version]
  50. Jiang, L.; Xiao, X.; Ren, J.; Tang, Y.; Weng, H.; Yang, Q.; Wu, M.; Tang, W. Proteomic analysis of bladder cancer indicates Prx-I as a key molecule in BI-TK/GCV treatment system. PLoS ONE 2014, 9, e98764. [Google Scholar] [CrossRef] [Green Version]
  51. Yoon, Y.; Kim, G.; Jeon, B.N.; Fang, S.; Park, H. Bifidobacterium Strain-Specific Enhances the Efficacy of Cancer Therapeutics in Tumor-Bearing Mice. Cancers 2021, 13, 957. [Google Scholar] [CrossRef]
  52. Demin, N.N.; Karmanova, I.G.; Rubinskaya, N.L.; Khomutetskaya, O.E. Comparative neurochemical and physiological characteristics of catalepsy-type rest and sleep. Neurosci. Behav. Physiol. 1978, 9, 98–102. [Google Scholar] [CrossRef] [PubMed]
  53. Li, Y.; Tinoco, R.; Elmen, L.; Segota, I.; Xian, Y.; Fujita, Y.; Sahu, A.; Zarecki, R.; Marie, K.; Feng, Y.; et al. Gut microbiota dependent antitumor immunity restricts melanoma growth in Rnf5(-/-) mice. Nat. Commun. 2019, 10, 1492. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Ghoneum, M.; Felo, N.; Agrawal, S.; Agrawal, A. A novel kefir product (PFT) activates dendritic cells to induce CD4+T and CD8+T cell responses in vitro. Int. J. Immunopathol. Pharmacol. 2015, 28, 488–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Jacouton, E.; Michel, M.L.; Torres-Maravilla, E.; Chain, F.; Langella, P.; Bermúdez-Humarán, L.G. Elucidating the Immune-Related Mechanisms by Which Probiotic Strain Lactobacillus casei BL23 Displays Anti-tumoral Properties. Front. Microbiol. 2019, 9, 3281. [Google Scholar] [CrossRef] [PubMed]
  56. Sugimura, N.; Li, Q.; Chu, E.S.H.; Lau, H.C.H.; Fong, W.; Liu, W.; Liang, C.; Nakatsu, G.; Su, A.C.Y.; Coker, O.O.; et al. Lactobacillus gallinarum modulates the gut microbiota and produces anti-cancer metabolites to protect against colorectal tumourigenesis. Gut 2022, 71, 2011–2021. [Google Scholar] [CrossRef] [PubMed]
  57. Hua, M.C.; Lin, T.Y.; Lai, M.W.; Kong, M.S.; Chang, H.J.; Chen, C.C. Probiotic Bio-Three induces Th1 and mmuneflammatory effects in PBMC and dendritic cells. World J. Gastroenterol. 2010, 16, 3529–3540. [Google Scholar] [CrossRef] [PubMed]
  58. Donohoe, D.R.; Holley, D.; Collins, L.B.; Montgomery, S.A.; Whitmore, A.C.; Hillhouse, A.; Curry, K.P.; Renner, S.W.; Greenwalt, A.; Ryan, E.P.; et al. A gnotobiotic mouse model demonstrates that dietary fiber protects against colorectal tumorigenesis in a microbiota- and butyrate-dependent manner. Cancer Discov. 2014, 4, 1387–1397. [Google Scholar] [CrossRef] [Green Version]
  59. Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.D.; Manicassamy, S.; Munn, D.H.; et al. Activation of Gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef] [Green Version]
  60. 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. [Google Scholar] [CrossRef] [Green Version]
  61. Salcedo, R.; Worschech, A.; Cardone, M.; Jones, Y.; Gyulai, Z.; Dai, R.M.; Wang, E.; Ma, W.; Haines, D.; O’hUigin, C.; et al. MyD88-mediated signaling prevents development of adenocarcinomas of the colon: Role of interleukin 18. J. Exp. Med. 2010, 207, 1625–1636. [Google Scholar] [CrossRef] [PubMed]
  62. Swanson, K.V.; Deng, M.; Ting, J.P. The NLRP3 inflammasome: Molecular activation and regulation to therapeutics. Nat. Rev. Immunol. 2019, 19, 477–489. [Google Scholar] [CrossRef] [PubMed]
  63. Geng, H.W.; Yin, F.Y.; Zhang, Z.F.; Gong, X.; Yang, Y. Butyrate Suppresses Glucose Metabolism of Colorectal Cancer Cells via GPR109a-AKT Signaling Pathway and Enhances Chemotherapy. Front. Mol. Biosci. 2021, 8, 634874. [Google Scholar] [CrossRef] [PubMed]
  64. Kespohl, M.; Vachharajani, N.; Luu, M.; Harb, H.; Pautz, S.; Wolff, S.; Sillner, N.; Walker, A.; Schmitt-Kopplin, P.; Boettger, T.; et al. The Microbial Metabolite Butyrate Induces Expression of Th1-Associated Factors in CD4+ T Cells. Front. Immunol. 2017, 8, 1036. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Liu, X.; Chen, Y.; Zhang, S.; Dong, L. Gut microbiota-mediated immunomodulation in tumor. J. Exp. Clin. Cancer Res. 2021, 40, 221. [Google Scholar] [CrossRef]
  66. 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]
  67. Zhang, W.; An, Y.; Qin, X.; Wu, X.; Wang, X.; Hou, H.; Song, X.; Liu, T.; Wang, B.; Huang, X.; et al. Gut Microbiota-Derived Metabolites in Colorectal Cancer: The Bad and the Challenges. Front. Oncol. 2021, 11, 739648. [Google Scholar] [CrossRef]
  68. Liu, T.; Song, X.; Khan, S.; Li, Y.; Guo, Z.; Li, C.; Wang, S.; Dong, W.; Liu, W.; Wang, B.; et al. The Gut Microbiota at the Intersection of Bile Acids and Intestinal Carcinogenesis: An Old Story, Yet Mesmerizing. Int. J. Cancer 2020, 146, 1780–1790. [Google Scholar] [CrossRef]
  69. Megna, B.W.; Carney, P.R.; Nukaya, M.; Geiger, P.; Kennedy, G.D. Indole-3-carbinol induces tumor cell death: Function follows form. J. Surg. Res. 2016, 204, 47–54. [Google Scholar] [CrossRef] [Green Version]
  70. Bae, S.; Ulrich, C.M.; Neuhouser, M.L.; Malysheva, O.; Bailey, L.B.; Xiao, L.; Brown, E.C.; Cushing-Haugen, K.L.; Zheng, Y.; Cheng, T.-Y.D.; et al. Plasma Choline Metabolites and Colorectal Cancer Risk in the Women’s Health Initiative Observational Study. Cancer Res. 2014, 74, 7442–7452. [Google Scholar] [CrossRef] [Green Version]
  71. Liu, X.; Liu, H.; Yuan, C.; Zhang, Y.; Wang, W.; Hu, S.; Liu, L.; Wang, Y. Preoperative Serum TMAO Level is a New Prognostic Marker for Colorectal Cancer. Biomark. Med. 2017, 11, 443–447. [Google Scholar] [CrossRef] [PubMed]
  72. Xu, R.; Wang, Q.; Li, L. A Genome-Wide Systems Analysis Reveals Strong Link Between Colorectal Cancer and Trimethylamine N-Oxide (TMAO), a Gut Microbial Metabolite of Dietary Meat and Fat. BMC Genom. 2015, 16 (Suppl. S7), S4. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Yazici, C.; Wolf, P.G.; Kim, H.; Cross, T.L.; Vermillion, K.; Carroll, T.; Augustus, G.J.; Mutlu, E.; Tussing-Humphreys, L.; Braunschweig, C.; et al. Race-Dependent Association of Sulfidogenic Bacteria with Colorectal Cancer. Gut 2017, 66, 1983–1994. [Google Scholar] [CrossRef]
  74. Nguyen, L.H.; Ma, W.; Wang, D.D.; Cao, Y.; Mallick, H.; Gerbaba, T.K.; Lloyd-Price, J.; Abu-Ali, G.; Hall, A.B.; Sikavi, D.; et al. Association Between Sulfur-Metabolizing Bacterial Communities in Stool and Risk of Distal Colorectal Cancer in Men. Gastroenterology 2020, 158, 1313–1325. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Attene-Ramos, M.S.; Wagner, E.D.; Gaskins, H.R.; Plewa, M.J. Hydrogen Sulfide Induces Direct Radical-Associated DNA Damage. Mol. Cancer Res. 2007, 5, 455–459. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Bayerdörffer, E.; Mannes, G.A.; Richter, W.O.; Ochsenkühn, T.; Wiebecke, B.; Köpcke, W.; Paumgartner, G. Increased Serum Deoxycholic Acid Levels in Men with Colorectal Adenomas. Gastroenterology 1993, 104, 145–151. [Google Scholar] [CrossRef]
  77. Bayerdörffer, E.; Mannes, G.A.; Ochsenkühn, T.; Dirschedl, P.; Wiebecke, B.; Paumgartner, G. Unconjugated Secondary Bile Acids in the Serum of Patients with Colorectal Adenomas. Gut 1995, 36, 268–273. [Google Scholar] [CrossRef] [Green Version]
  78. Ocvirk, S.; Wilson, A.S.; Posma, J.M.; Li, J.V.; Koller, K.R.; Day, G.M.; Flanagan, C.A.; Otto, J.E.; Sacco, P.E.; Sacco, F.D.; et al. A Prospective Cohort Analysis of Gut Microbial Co-Metabolism in Alaska Native and Rural African People at High and Low Risk of Colorectal Cancer. Am. J. Clin. Nutr. 2020, 111, 406–419. [Google Scholar] [CrossRef]
  79. Liu, L.; Dong, W.; Wang, S.; Zhang, Y.; Liu, T.; Xie, R.; Wang, B.; Cao, H. Deoxycholic Acid Disrupts the Intestinal Mucosal Barrier and Promotes Intestinal Tumorigenesis. Food Funct. 2018, 9, 5588–5597. [Google Scholar] [CrossRef] [Green Version]
  80. Dong, W.; Liu, L.; Dou, Y.; Xu, M.; Liu, T.; Wang, S.; Zhang, Y.; Deng, B.; Wang, B.; Cao, H. Deoxycholic Acid Activates Epidermal Growth Factor Receptor and Promotes Intestinal Carcinogenesis by ADAM17-Dependent Ligand Release. J. Cell. Mol. Med. 2018, 22, 4263–4273. [Google Scholar] [CrossRef]
  81. Cao, H.; Xu, M.; Dong, W.; Deng, B.; Wang, S.; Zhang, Y.; Wang, S.; Luo, S.; Wang, W.; Qi, Y.; et al. Secondary Bile Acid-Induced Dysbiosis Promotes Intestinal Carcinogenesis. Int. J. Cancer 2017, 140, 2545–2556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Lee, H.Y.; Crawley, S.; Hokari, R.; Kwon, S.; Kim, Y.S. Bile Acid Regulates MUC2 Transcription in Colon Cancer Cells via Positive EGFR/PKC/Ras/ERK/CREB, PI3K/Akt/IkappaB/NF-kappaB and P38/MSK1/CREB Pathways and Negative JNK/c-Jun/AP-1 Pathway. Int. J. Oncol. 2010, 36, 941–953. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Fu, T.; Coulter, S.; Yoshihara, E.; Oh, T.G.; Fang, S.; Cayabyab, F.; Zhu, Q.; Zhang, T.; Leblanc, M.; Liu, S.; et al. FXR Regulates Intestinal Cancer Stem Cell Proliferation. Cell 2019, 176, 1098–1112.e18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Jean-Louis, S.; Akare, S.; Ali, M.A.; Mash, E.A., Jr.; Meuillet, E.; Martinez, J.D. Deoxycholic Acid Induces Intracellular Signaling Through Membrane Perturbations. J. Biol. Chem. 2006, 281, 14948–14960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Zhu, Y.; Wang, P.P.; Zhao, J.; Green, R.; Sun, Z.; Roebothan, B.; Squires, J.; Buehler, S.; Dicks, E.; Zhao, J.; et al. Dietary N-Nitroso Compounds and Risk of Colorectal Cancer: A Case-Control Study in Newfoundland and Labrador and Ontario, Canada. Br. J. Nutr. 2014, 111, 1109–1117. [Google Scholar] [CrossRef] [Green Version]
  86. Hebels, D.G.; Briedé, J.J.; Khampang, R.; Kleinjans, J.C.; de Kok, T.M. Radical Mechanisms in Nitrosamine- and Nitrosamide-Induced Whole-Genome Gene Expression Modulations in Caco-2 Cells. Toxicol. Sci. 2010, 116, 194–205. [Google Scholar] [CrossRef] [Green Version]
  87. Hebels, D.G.; Jennen, D.G.; Kleinjans, J.C.; de Kok, T.M. Molecular Signatures of N-Nitroso Compounds in Caco-2 Cells: Implications for Colon Carcinogenesis. Toxicol. Sci. 2009, 108, 290–300. [Google Scholar] [CrossRef] [Green Version]
  88. Gottschalg, E.; Scott, G.B.; Burns, P.A.; Shuker, D.E. Potassium Diazoacetate-Induced P53 Mutations In Vitro in Relation to Formation of O6-Carboxymethyl- and O6-Methyl-2′-Deoxyguanosine DNA Adducts: Relevance for Gastrointestinal Cancer. Carcinogenesis 2007, 28, 356–362. [Google Scholar] [CrossRef] [Green Version]
  89. Lewin, M.H.; Bailey, N.; Bandaletova, T.; Bowman, R.; Cross, A.J.; Pollock, J.; Shuker, D.E.G.; Bingham, S.A. Red Meat Enhances the Colonic Formation of the DNA Adduct O6-Carboxymethyl Guanine: Implications for Colorectal Cancer Risk. Cancer Res. 2006, 66, 1859–1865. [Google Scholar] [CrossRef] [Green Version]
  90. Helmus, D.S.; Thompson, C.L.; Zelenskiy, S.; Tucker, T.C.; Li, L. Red Meat-Derived Heterocyclic Amines Increase Risk of Colon Cancer: A Population-Based Case-Control Study. Nutr. Cancer 2013, 65, 1141–1150. [Google Scholar] [CrossRef] [Green Version]
  91. Cross, A.J.; Ferrucci, L.M.; Risch, A.; Graubard, B.I.; Ward, M.H.; Park, Y.; Hollenbeck, A.R.; Schatzkin, A.; Sinha, R. A Large Prospective Study of Meat Consumption and Colorectal Cancer Risk: An Investigation of Potential Mechanisms Underlying This Association. Cancer Res. 2010, 70, 2406–2414. [Google Scholar] [CrossRef] [Green Version]
  92. Hasegawa, R.; Sano, M.; Tamano, S.; Imaida, K.; Shirai, T.; Nagao, M.; Sugimura, T.; Ito, N. Dose-Dependence of 2-Amino-1-Methyl-6-Phenylimidazo [4,5-B]-Pyridine (PhIP) Carcinogenicity in Rats. Carcinogenesis 1993, 14, 2553–2557. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, R.; Lin, X.; Li, Z.; Li, Q.; Bi, K. Quantitative Metabolomics for Investigating the Value of Polyamines in the Early Diagnosis and Therapy of Colorectal Cancer. Oncotarget 2017, 9, 4583–4592. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Manna, S.K.; Tanaka, N.; Krausz, K.W.; Haznadar, M.; Xue, X.; Matsubara, T.; Bowman, E.D.; Fearon, E.R.; Harris, C.C.; Shah, Y.M.; et al. Biomarkers of Coordinate Metabolic Reprogramming in Colorectal Tumors in Mice and Humans. Gastroenterology 2014, 146, 1313–1324. [Google Scholar] [CrossRef] [Green Version]
  95. Guo, Y.; Ye, Q.; Deng, P.; Cao, Y.; He, D.; Zhou, Z.; Wang, C.; Zaytseva, Y.Y.; Schwartz, C.E.; Lee, E.Y.; et al. Spermine Synthase and MYC Cooperate to Maintain Colorectal Cancer Cell Survival by Repressing Bim Expression. Nat. Commun. 2020, 11, 3243. [Google Scholar] [CrossRef] [PubMed]
  96. Clausen, M.R.; Mortensen, P.B. Fecal Ammonia in Patients with Adenomatous Polyps and Cancer of the Colon. Nutr. Cancer 1992, 18, 175–180. [Google Scholar] [CrossRef] [PubMed]
  97. Visek, W.J. Diet and Cell Growth Modulation by Ammonia. Am. J. Clin. Nutr. 1978, 31, S216–S220. [Google Scholar] [CrossRef]
  98. Cheng, C.; Xie, Z.; Li, Y.; Wang, J.; Qin, C.; Zhang, Y. PTBP1 Knockdown Overcomes the Resistance to Vincristine and Oxaliplatin in Drug-Resistant Colon Cancer Cells Through Regulation of Glycolysis. BioMed Pharm. 2018, 108, 194–200. [Google Scholar] [CrossRef]
  99. Haas, R.; Smith, J.; Rocher-Ros, V.; Nadkarni, S.; Montero-Melendez, T.; D’Acquisto, F.; Bland, E.J.; Bombardieri, M.; Pitzalis, C.; Perretti, M.; et al. Lactate Regulates Metabolic and Pro-Inflammatory Circuits in Control of T Cell Migration and Effector Functions. PloS Biol. 2015, 13, e1002202. [Google Scholar] [CrossRef]
  100. Gao, R.; Gao, Z.; Huang, L.; Qin, H. Gut microbiota and colorectal cancer. Eur. J. Clin. Microbiol. Infect. Dis. 2017, 36, 757–769. [Google Scholar] [CrossRef] [Green Version]
  101. Chen, F.; Dai, X.; Zhou, C.; Li, K.-X.; Zhang, Y.-J.; Lou, X.-Y.; Zhu, Y.-M.; Sun, Y.-L.; Peng, B.-X.; Cui, W. Integrated analysis of the faecal metagenome and serum metabolome reveals the role of gut microbiome-associated metabolites in the detection of colorectal cancer and adenoma. Gut 2022, 71, 1315–1325. [Google Scholar] [CrossRef] [PubMed]
  102. Woo, V.; Alenghat, T. Epigenetic regulation by gut microbiota. Gut Microbes. 2022, 14, 2022407. [Google Scholar] [CrossRef] [PubMed]
  103. Sharma, M.; Li, Y.; Stoll, M.L.; Tollefsbol, T.O. The Epigenetic Connection Between the Gut Microbiome in Obesity and Diabetes. Front. Genet. 2020, 10, 1329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Wu, Y.; Wang, C.Z.; Wan, J.Y.; Yao, H.; Yuan, C.S. Dissecting the Interplay Mechanism between Epigenetics and Gut Microbiota: Health Maintenance and Disease Prevention. Int. J. Mol. Sci. 2021, 22, 6933. [Google Scholar] [CrossRef] [PubMed]
  105. Peruhova, M.; Peshevska-Sekulovska, M.; Krastev, B.; Panayotova, G.; Georgieva, V.; Konakchieva, R.; Nikolaev, G.; Velikova, T.V. What could microRNA expression tell us more about colorectal serrated pathway carcinogenesis? World J. Gastroenterol. 2020, 26, 6556–6571. [Google Scholar] [CrossRef] [PubMed]
  106. Ropero, S.; Esteller, M. The role of histone deacetylases (HDACs) in human cancer. Mol. Oncol. 2007, 1, 19–25. [Google Scholar] [CrossRef] [Green Version]
  107. Boffa, L.C.; Lupton, J.R.; Mariani, M.R.; Ceppi, M.; Newmark, H.L.; Scalmati, A.; Lipkin, M. Modulation of colonic epithelial cell proliferation, histone acetylation, and luminal short chain fatty acids by variation of dietary fiber (wheat bran) in rats. Cancer Res. 1992, 52, 5906–5912. [Google Scholar]
  108. Miro-Blanch, J.; Yanes, O. Epigenetic Regulation at the Interplay Between Gut Microbiota and Host Metabolism. Front. Genet. 2019, 10, 638. [Google Scholar] [CrossRef]
  109. Yang, W.; Yu, T.; Huang, X.; Bilotta, A.J.; Xu, L.; Lu, Y.; Sun, J.; Pan, F.; Zhou, J.; Zhang, W.; et al. Intestinal microbiota-derived short-chain fatty acids regulation of immune cell IL-22 production and gut immunity. Nat. Commun. 2020, 11, 4457. [Google Scholar] [CrossRef]
  110. Fellows, R.; Denizot, J.; Stellato, C.; Cuomo, A.; Jain, P.; Stoyanova, E.; Balázsi, S.; Hajnády, Z.; Liebert, A.; Kazakevych, J.; et al. Microbiota derived short chain fatty acids promote histone crotonylation in the colon through histone deacetylases. Nat. Commun. 2018, 9, 105. [Google Scholar] [CrossRef] [Green Version]
  111. Kaiko, G.E.; Ryu, S.H.; Koues, O.I.; Collins, P.L.; Solnica-Krezel, L.; Pearce, E.J.; Pearce, E.L.; Oltz, E.M.; Stappenbeck, T.S. The Colonic Crypt Protects Stem Cells from Microbiota-Derived Metabolites. Cell 2016, 165, 1708–1720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  112. Wu, S.E.; Hashimoto-Hill, S.; Woo, V.; Eshleman, E.M.; Whitt, J.; Engleman, L.; Karns, R.; Denson, L.A.; Haslam, D.B.; Alenghat, T. Microbiota-derived metabolite promotes HDAC3 activity in the gut. Nature 2020, 586, 108–112. [Google Scholar] [CrossRef]
  113. Turgeon, N.; Gagné, J.M.; Blais, M.; Gendron, F.P.; Boudreau, F.; Asselin, C. The acetylome regulators Hdac1 and Hdac2 differently modulate intestinal epithelial cell dependent homeostatic responses in experimental colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 2014, 306, G594–G605. [Google Scholar] [CrossRef]
  114. Gonneaud, A.; Turgeon, N.; Boudreau, F.; Perreault, N.; Rivard, N.; Asselin, C. Distinct Roles for Intestinal Epithelial Cell-Specific Hdac1 and Hdac2 in the Regulation of Murine Intestinal Homeostasis. J. Cell. Physiol. 2016, 231, 436–448. [Google Scholar] [CrossRef] [PubMed]
  115. Zimberlin, C.D.; Lancini, C.; Sno, R.; Rosekrans, S.L.; McLean, C.M.; Vlaming, H.; van den Brink, G.R.; Bots, M.; Medema, J.P.; Dannenberg, J.H. HDAC1 and HDAC2 collectively regulate intestinal stem cell homeostasis. FASEB J. 2015, 29, 2070–2080. [Google Scholar] [CrossRef]
  116. Liu, S.; Chang, W.; Jin, Y.; Feng, C.; Wu, S.; He, J.; Xu, T. The function of histone acetylation in cervical cancer development. Biosci. Rep. 2019, 39, BSR20190527. [Google Scholar] [CrossRef] [PubMed]
  117. Taur, Y.; Jenq, R.R.; Perales, M.A.; Littmann, E.R.; Morjaria, S.; Ling, L.; No, D.; Gobourne, A.; Viale, A.; Dahi, P.B.; et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood 2014, 124, 1174–1182. [Google Scholar] [CrossRef]
  118. Bhatt, A.P.; Redinbo, M.R.; Bultman, S.J. The role of the microbiome in cancer development and therapy. CA Cancer J. Clin. 2017, 67, 326–344. [Google Scholar] [CrossRef] [Green Version]
  119. Peruhova, M.; Peshevska-Sekulovska, M.; Velikova, T. Interactions between human microbiome, liver diseases, and immunosuppression after liver transplant. World J. Immunol. 2021, 11, 11–16. [Google Scholar] [CrossRef]
  120. Nakov, R.; Velikova, T. Chemical Metabolism of Xenobiotics by Gut Microbiota. Curr. Drug. Metab. 2020, 21, 260–269. [Google Scholar] [CrossRef]
  121. Velikova, T.; Nakov, R.; Ianiro, G. 2.10—Medication and Health Risks Associated with Neglected Side Effects on Gut Microbiota. In Comprehensive Gut Microbiota; Elsevier: Amsterdam, The Netherlands, 2022; Volume 112, p. 124. [Google Scholar] [CrossRef]
  122. Wallace, B.D.; Wang, H.; Lane, K.T.; Scott, J.E.; Orans, J.; Koo, J.S.; Venkatesh, M.; Jobin, C.; Yeh, L.-A.; Mani, S.; et al. Alleviating cancer drug toxicity by inhibiting a bacterial enzyme. Science 2010, 330, 831–835. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Zitvogel, L.; Galluzzi, L.; Viaud, S.; Vétizou, M.; Daillère, R.; Merad, M.; Kroemer, G. Cancer and the gut microbiota: An unexpected link. Sci. Transl. Med. 2015, 7, 271ps1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Raza, M.H.; Gul, K.; Arshad, A.; Riaz, N.; Waheed, U.; Rauf, A.; Aldakheel, F.; Alduraywish, S.; Rehman, M.U.; Abdullah, M.; et al. Microbiota in cancer development and treatment. J. Cancer Res. Clin. Oncol. 2019, 145, 49–63. [Google Scholar] [CrossRef] [PubMed]
  125. Hanus, M.; Parada-Venegas, D.; Landskron, G.; Wielandt, A.M.; Hurtado, C.; Alvarez, K.; Hermoso, M.A.; López-Köstner, F.; De la Fuente, M. Immune System, Microbiota, and Microbial Metabolites: The Unresolved Triad in Colorectal Cancer Microenvironment. Front. Immunol. 2021, 12, 612826. [Google Scholar] [CrossRef] [PubMed]
  126. Alhinai, E.A.; Walton, G.E.; Commane, D.M. The Role of the Gut Microbiota in Colorectal Cancer Causation. Int. J. Mol. Sci. 2019, 20, 5295. [Google Scholar] [CrossRef] [Green Version]
  127. Liu, Y.; Baba, Y.; Ishimoto, T.; Gu, X.; Zhang, J.; Nomoto, D.; Okadome, K.; Baba, H.; Qiu, P. Gut microbiome in gastrointestinal cancer: A friend or foe? Int. J. Biol. Sci. 2022, 18, 4101–4117. [Google Scholar] [CrossRef]
  128. Elkrief, A.; Derosa, L.; Kroemer, G.; Zitvogel, L.; Routy, B. The negative impact of antibiotics on outcomes in cancer patients treated with immunotherapy: A new independent prognostic factor? Ann. Oncol. 2019, 30, 1572–1579. [Google Scholar] [CrossRef] [Green Version]
  129. Yi, M.; Jiao, D.; Qin, S.; Chu, Q.; Li, A.; Wu, K. Manipulating Gut Microbiota Composition to Enhance the Therapeutic Effect of Cancer Immunotherapy. Integr. Cancer Ther. 2019, 18, 1534735419876351. [Google Scholar] [CrossRef] [Green Version]
  130. Perillo, F.; Amoroso, C.; Strati, F.; Giuffrè, M.R.; Díaz-Basabe, A.; Lattanzi, G.; Facciotti, F. Gut Microbiota Manipulation as a Tool for Colorectal Cancer Management: Recent Advances in Its Use for Therapeutic Purposes. Int. J. Mol. Sci. 2020, 21, 5389. [Google Scholar] [CrossRef]
  131. Fong, W.; Li, Q.; Yu, J. Gut microbiota modulation: A novel strategy for prevention and treatment of colorectal cancer. Oncogene 2020, 39, 4925–4943. [Google Scholar] [CrossRef]
  132. Kim, J.; Lee, H.K. Potential Role of the Gut Microbiome in Colorectal Cancer Progression. Front. Immunol. 2022, 12, 807648. [Google Scholar] [CrossRef] [PubMed]
  133. Wen, X.; Ye, X.; Yang, X.; Jiang, R.; Qian, C.; Wang, X. The cross-talk between intestinal bacterial microbiota and immune cells in colorectal cancer progression. Clin. Transl. Oncol. 2022, 25, 620–632. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Immune surveillance and tumor development are connected with the state of health of the microbiome composition or dysbiosis.
Figure 1. Immune surveillance and tumor development are connected with the state of health of the microbiome composition or dysbiosis.
Ijms 24 05978 g001
Figure 2. Tumor promoting and anticancer mechanisms and pathways of gut microbiota and their metabolites.
Figure 2. Tumor promoting and anticancer mechanisms and pathways of gut microbiota and their metabolites.
Ijms 24 05978 g002
Figure 3. Cross-talk between microbiota and epigenetic modifications associated with gut homeostasis or disease (including cancer). Arrows represent influence or development, and an arrow followed by | represents suppression.
Figure 3. Cross-talk between microbiota and epigenetic modifications associated with gut homeostasis or disease (including cancer). Arrows represent influence or development, and an arrow followed by | represents suppression.
Ijms 24 05978 g003
Table 1. Microbiota-derived metabolites, their primary sources and their effects on carcinogenesis.
Table 1. Microbiota-derived metabolites, their primary sources and their effects on carcinogenesis.
Microbiota-Derived MetaboliteSourceEffectsReferences
SCFAAnaerobesAntitumor effect, reducing inflammation[58,59,60,61,62,63,64]
Secondary bile acidsB. fragilis, Bacteroides vulgatus, Clostridium perfringens, Eubacterium, Lactobacillus and BifidobacteriumContribute to CRC progression[65,66,68]
IndolesGram-positive anaerobe (i.e., P. anaerobius)Tumor prevention [69,70]
TMAOGram-negative, Gram-positive, anaerobe (i.e., E. coli, Clostridium and Desulfovibrios)Positive association with CRC risk, new prognostic marker[71,72]
H2SGram-negative anaerobes (i.e., F. nucleatum and Desulfovibrios)Potential environmental risk factors for CRC[73,74,75]
DCAGram-positive, Gram-negative anaerobes (i.e., Desulfovibrios and Clostridium)Positive associations with colorectal adenomas and CRC; contributes to CRC development and carcinogenesis promotion[76,77,78,79,80,81,82,83,84]
NOCsFacultative and anaerobesPositive association with CRC risk[85,86,87,88,89]
HCAsBacteroides, LactobacilliPositive association with CRC risk; Bacteroides convert HCA to carcinogens and Lactobacilli reduce their mutagenic effect[90,91,92]
PolyaminesGram-negative anaerobes (i.e., B. fragilis and F. nucleatum)Positive association with CRC [93,94,95]
AmmoniaGram-negative anaerobes, clostridia, enterobacteria and Bacillus spp. Gram-positive non-sporing anaerobes, streptococci Contribute to CRC development and promote neoplastic transformation[96,97]
LactateLactobacillus, Leuconostoc, Pediococcus, Lactococcus and StreptococcusPromote CRC[98,99]
SCFAs—short-chain fatty acids; H2S—hydrogen sulfide; TMAO—trimethylamine-N-oxide; DCA—deoxycholic acids; HCAs—heterocyclic amines; NOCs—N-nitroso compounds; TMAO—trimethylamine-N-oxide; TMA—trimethylamine.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Lozenov, S.; Krastev, B.; Nikolaev, G.; Peshevska-Sekulovska, M.; Peruhova, M.; Velikova, T. Gut Microbiome Composition and Its Metabolites Are a Key Regulating Factor for Malignant Transformation, Metastasis and Antitumor Immunity. Int. J. Mol. Sci. 2023, 24, 5978. https://doi.org/10.3390/ijms24065978

AMA Style

Lozenov S, Krastev B, Nikolaev G, Peshevska-Sekulovska M, Peruhova M, Velikova T. Gut Microbiome Composition and Its Metabolites Are a Key Regulating Factor for Malignant Transformation, Metastasis and Antitumor Immunity. International Journal of Molecular Sciences. 2023; 24(6):5978. https://doi.org/10.3390/ijms24065978

Chicago/Turabian Style

Lozenov, Stefan, Boris Krastev, Georgi Nikolaev, Monika Peshevska-Sekulovska, Milena Peruhova, and Tsvetelina Velikova. 2023. "Gut Microbiome Composition and Its Metabolites Are a Key Regulating Factor for Malignant Transformation, Metastasis and Antitumor Immunity" International Journal of Molecular Sciences 24, no. 6: 5978. https://doi.org/10.3390/ijms24065978

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