Open access peer-reviewed chapter

Perspective Chapter: Epigenetic Therapy – The Future Treatment for Cancer

Written By

Surya Chandra Rao Thumu, Shobha Rani Papanna and Sundru Manjulata Devi

Submitted: 03 February 2023 Reviewed: 21 February 2023 Published: 16 March 2023

DOI: 10.5772/intechopen.110641

From the Edited Volume

DNA Replication - Epigenetic Mechanisms and Gene Therapy Applications

Edited by Ziyad S. Haidar

Chapter metrics overview

213 Chapter Downloads

View Full Metrics

Abstract

Scientists have made a remarkable breakthrough by uncovering DNA and its role in living organisms. Epigenetics examines the phenotypic divergences due to DNA methylation and its effects at certain genetic spots. Epigenetic and genetic problems combine to cause cancer and its growth, as seen by frequent mutations in genes that manage the epigenome. Recently, new therapies targeting epigenetic alterations have been proposed. Drugs with longer shelf life and better absorption are also being manufactured and tested. On this aspect, CRISPR technology has been used to create various strategies for epigenetic engineering and is a practical approach to understanding and manipulating biological processes. Furthermore, studies on the advantages of probiotics have advanced previous interventional studies to recognize the molecular mechanisms involved. Numerous probiotic genomes include epigenetic components that influence gene expression for fundamental functions. Consequently, we suggest investigations incorporating genomic and meta-epigenomic information to better understand the mode of action of probiotics and their related microbiomes in epigenetic therapy. Here, we review established epigenetic discoveries, combined with the rapid advancement of immunotherapies, to create new possibilities for cancer treatment.

Keywords

  • epigenetics
  • epigenomes
  • CRISPR-cas
  • probiotics
  • microbiome
  • DNA methylation

1. Introduction

One of the greatest discoveries made in the twentieth century is recognizing that phenotypic changes are not merely based on genetic alterations but are implications due to external stimuli/epigenetic factors [1]. Epigenetic trait is a phenotype whose inheritance is stably heritable without altering the genetic code. Thus, for the normal development of an organism, regulations in the epigenome or epigenetic state are equally important as changes in gene expression. Today, epigenetics is a dynamic field aimed at studying individual and fundamental processes in mitosis and meiosis [2]. Several endogenous factors are found to be involved in modulating epigenetic states and their activity can be affected by each other. Cellular functions can be preserved for an extended period by generating and transmitting epigenetic states [2]. Genomic characteristics such as chromatin reconstitution and epigenetic factors’ alterations could play a role in epigenetic states. However, epigenetic changes must be precisely defined to obtain a complete and detailed picture [3, 4, 5]. Understanding the role of epigenetic alterations and their effect on gene expression and the awareness that epigenetic traits are reversible has opened the door to developing diagnostic tools to treat several diseases, including cancers [6].

Epigenetic alterations are essential for the regulatory processes associated with several biological phenomena, including genomic imprinting, X-chromosome inactivation, control of tissue-specific gene expression, genomic stability, repression of transposable elements, aging, and some diseases, including cancer [7]. The most common epigenetic modifications that modify DNA accessibility to transcriptional machinery and impact gene expression are acetylation, methylation, phosphorylation, biotinylation, and RNA interference [8]. Roberti et al. [9] have identified three main components of the epigenetic system: DNA methylation, post-translational histone modifications, and noncoding RNAs (ncRNAs). The clustered regularly interspaced short palindromic repeats (CRISPR)-associated protein 9 (Cas9)system (CRISPR-cas9) is a naturally occurring defense system in many bacteria and archaea to protect from invading viruses or plasmids [10]. CRISPR system with the capacity to program Cas9 has revolutionized medical research, biotechnology and agriculture. It is a precise genome editing tool that can modify specific genome regions in eukaryotic cells, particularly mammalian cells [11]. Cas proteins are endonucleases that sever the target nucleic acid sequence. Mutations in genes that encode Cas proteins can generate deactivated cas (dCas) proteins that maintain their ability to bind DNA in a manner targeted by gRNA and serve as programmable DNA binding domains [12]. CRISPRi/a domain proteins can be used in epigenetic studies to alter histone acetylation and methylation [13]. The dCas-based demethylation approach was discovered to improve cell disease conditions. Scientists have found that countering epigenetic changes can be an effective cancer therapy [7, 14].

The trillions of intestinal microflora have an intimate relationship with the host and impact the host’s physiology and pathology. They play a role in food fermentation, vitamin synthesis, and maintaining intestinal epithelial function. The microbiome affects the host’s epigenetics, immune system, and metabolism. Certain epigenetic changes after exposure to bacteria suggest a connection between the microbiome and the epigenome [15]. Investigating the epigenetics and para-epigenetics of a probiotic organism and its microbiome is a difficult but critical step in understanding the probiotic mechanism [1]. Researchers have used CRISPR-Cas to precisely alter probiotic organisms, explore epigenetics, and improve the effects of probiotics [16, 17]. Certain epigenetic changes during childhood are essential for adult health, and short-chain fatty acids (SCFAs), such as butyrate, can influence human genes by being epigenetic regulators [18, 19]. Researching the various probiotic molecules and their interactions with human epigenetic systems could reveal new health benefits and assess the safety of consuming these organisms. Current and future technologies can help researchers understand microbial meta-epigenomics and how it can improve human health. In the current study, a search strategy was assigned, and the search words with “EPIGENETICS”, CRISPR-cas9 EDITING IN EPIGENETIS” and PROBIOTICS THERAPY IN EPIGENETICS” has been used and the related articles were retrieved from PubMed, Web of Science, and Scopus databases from 2010 to Jan 2023. This article discusses epigenetic therapy and its potential new methodologies using the CRIPSR-cas system, epigenetic drugs, and epigenetic regulation by probiotic bacteria and the gut microbiota.

Advertisement

2. The emergence of the CRISPR Cas9 system in epigenome editing

Epigenetic changes at the target site can be achieved by securing the epigenetic effectors to sequence-specific DNA-binding domains (DBDs). Although the discovery of zinc fingers (ZFNs) and TALENs succeeded in DNA binding and genome editing efficacy, targeting different regions required redesigning and reengineering of these proteins. This has prevented the scientific community from using ZFN and TALEN effectively [20]. The rise of the CRISPR Cas system has trounced this major hurdle. Although the Cas9 protein of the type II Streptococcus pyogenes system is the most widely used in the CRISPR Cas system in gene knockout, the CRISPR-Cas system in its basic form has become a powerful technology to flexibly target any specific site in the genome simply by employing small guide RNAs (sgRNA). Because most Cas proteins are nucleases, mutations in Cas-encoding genes convert them to deactivated Cas (dCas) proteins, inhibiting their nuclease activity while retaining DNA binding capability. Thus, dCas proteins have become programmable DBP, and the ability to bind dCas9 to specific DNA loci can be altered using sgRNA (Figure 1) [12].

Figure 1.

Strategies for gene activation and repression using the CRISPR Cas9 system. (A) Targeting specific DNA sequence for activation or repression through (a) direct binding of effector domain (ED) to dCas9; (b) effector domains bound to the sgRNA scaffold named srRNA 2.0; and (c) recruitment of the effector domain to the target site by the Sun tag system. (B) The concept of gene inactivation through CRISPR interference (CRISPRi) using KRAB bound to dCas9.

2.1 CRISPR Cas9 in epigenome activation and repression

Soon after discovering that wild-type Cas9 could be used as a programmable endonuclease, researchers began to exploit dCas9 for gene regulation studies. For epigenome editing through CRISPR Cas9, the setup consists of a toolbox for expressing sgRNA. In these dCas9 and effector domains, the activation domains retain their function when fused to other proteins with different DNA binding moieties [21]. Several activation domains have met this criterion which includes the 16-virion protein (VP16) of the herpes simplex virus type I, the VP64 domain is a tetramer of the minimal activator VP16, p65, the largest subunit of the NF-kappa B transcription factor, the Rta domain encoded by Kaposi’s sarcoma-associated herpesvirus/human herpesvirus 8, human heat shock factor 1 (HSF1) etc. [21]. The CRISPR activation cabinet (CRISPRa) consisting of dCas9 fused to the activation domain in various combinations has been used in the transcriptional activation of genes. In vitro studies showed that the dCas9-VP64 and dCas9-p65 fusion proteins have activated the target reporter gene in the HEK293 reporter cell line [22]. However, the VPR tripartite activation domain designed and characterized by George et al. reported a robust multi-locus activation, outpacing VP64 alone. Furthermore, for nuclease and activation pursuit using the dCas9-VPR system, maximum efficiency was achieved by changing the length of the gRNAs [23].

2.1.1 Increment in the transactivation potential using dCas9

Several interesting CRISPRa studies have described successful gene activation using dCas9-based tools and increased transcriptional activation with multiple targeting of sgRNA. However, Konermann et al. [24] have introduced the concept of increased activation using single sgRNA and modification of the gRNA scaffold and have named it sgRNA2.0. The main objective was to create a flexible sgRNA capable of grafting additional activation domains to the target site dCas9. Enhanced stimulation can be achieved with a single sgRNA through multiple activation domains. A similar concept of activation domain recruitment to the target site was developed by Tanenbaum et al. [25] and named the Sun tag system. This approach uses a recombinant peptide array (SunTag) that recruits multiple antibody fusion proteins (activator domains). This system achieved higher levels of transcription activation with recruitment of VP64 to dCas9 through Suntag to activate the CXCR4 gene in K562 cell lines using a single sgRNA. It achieved 25 times higher transcription levels than conventional dCas9-VP64-mediated activation [25]. In addition to the techniques manipulated by dCas9 to increase gene expression activation, light-controlled manipulation of epigenetic and endogenous genes was introduced [24, 26, 27]. The principle involved in this mode of gene manipulation is that two interaction partners dimerize upon blue-light activation, where one partner is fused to a DNA-binding protein and the other to the transcriptional activation domain. Recently, optogenetic systems have used dCas9 and photoinducible dimerization for precise modulation of gene expression of mammalian cells.

2.1.2 dCas9-mediated transcriptional repression

Qi et al. initially developed the concept of CRISPR interference (CRISPRi) [12], where dCas9 and sg RNAs target a specific transcriptional start site and interfere with the activity of DNA binding proteins such as Polymerase II. With this development of CRISPRi, modifications have been made in which a stronger repressor complex, such as Kruppel’s associated Box (KRAB), was fused to dCas9, resulting in a stronger and more specific gene repressor than dCas9 alone [22]. KRAB has a highly conserved amino-terminal region of many Krüppel-class Cys2His2 zinc finger proteins with repressive function [28]. The KRAB domain functions with the KAP1 corepressor that recruits heterochromatin protein 1 (HP1), which recognizes and binds methylated H3K9, obtained by methyltransferase activity and recruits more HMTs, resulting in chromatin condensation of neighboring nucleosomes [29]. Several success stories reported the use of dCas9-KRAB fusion in gene repression, and this fusion system can successfully recruit chromatin-modifying complexes for amplified CRISPRi effects [21].

2.2 Epigenome editing through chromatin modulation using CRISPR Cas9

Genomic DNA is organized on a nucleosomal scale by wrapping around various histones that can be modified post-translationally using chemical moieties and/or by chemical modification, such as methylation at 5-carbon in cysteine residues (5-mC), which are collectively termed chromatin markers [2]. Gene editing through chromatin edits has become an important aspect in the epigenetic field in altering gene expression. dCas9 has been deployed to modulate various histone proteins within a specific DNA region that affects gene expression [2]. Although gene expression levels through chromatin modulation are modest compared to CRISPRi/a technologies, the highest levels of expression are achieved by H3K27ac deposition or DNA demethylation [30, 31].

2.2.1 dCas9 in methylation and demethylation of chromatin marks

Since aberrant methylation has several pathological implications, especially in several cancers, there is an urgent need to manipulate these epigenetic characteristics. Although small molecules such as 5-azacytidine that target DNA methylation as epigenetic inhibitors are in clinical use, these inhibitors target the entire genome where normal methylation occurs [20]. This limitation has been overcome by targeting specific gene loci mediated by the dCas9 system, which has been used to deposit and remove methylation from target genome sites. The deposition of DNA methylation has been achieved by fusion of dCas9 with the catalytic domain of eukaryotic DNA methyl transferase (DNMT3A) [32, 33] or prokaryotic DNA methyltransferase (MQ3) [34]. In addition to targeted DNA methylation, the active removal of methylation marks from the endogenous DNA sequence is another mode of gene expression manipulation. Ten–eleven translocation (TET) proteins (TET1, TET2 and TET3) are involved in endogenous DNA demethylation that regulates cell type-specific gene expression. Therefore, in epigenetic DNA demethylation using the CRISPR system, guidable dCas9 fused to TET-1 has been used to achieve locus-specific DNA demethylation [31, 35, 36, 37].

2.2.2 dCas9 mediated modification of histone proteins

In addition to the role of methylation and demethylation in gene regulation, epigenetic information is stored in histone proteins that wrap DNA around them to form chromatin fiber. These epigenetic features in a cell are constituted by post-translational modification of the histone tails that reveal key insights into regulatory activity. For example, mono and dimethylation at the four positions of lysine of Histone H3 (H3K4me1/2) and acetylation at the 27 positions of acetylation of Lysine (H3K27ac) are elements of active distal regulation. At the same time, trimethylation (H3K4me3) is a marker of active or poised bivalent promoters [11]. Since specific writers, correctors, and erasers regulate the modification of chromatin, researchers have used the dCas9 system to recruit histone modifiers for the methylation and demethylation of histone proteins [20]. Examples of histone-targeted epigenome editing include LSD1, a histone demethylase that removes the H3K4me2 mark, which was fused to dCas9, resulting in a substantial local decrease in the active enhancer markers H3K4me2 and H3K27ac and altered expression of target genes [38]. Unlike these findings, a fusion of histone acetyltransferase P300 with dCas9 resulted in a significant increase in local induction of H3K27ac expression in both the promoter and enhancer regions [39]. Histone modifications through the CRISPR system were used to suppress gene activity where local induction of H3K4me3, a marker of the active promoter for the re-expression of silenced targets, was used. Histone deacetylation by fusion of full-length histone deacetylases (HDAC) with dCas9 was shown to reduce H3k27ac and ultimately down-regulate target gene expression [40].

Advertisement

3. Gut microbiota and probiotics: the mysterious players in epigenetic changes

The gut microbiota/microbiome is the collective term used for the genes contributed collectively by viruses, bacteria, and fungi that are present in different regions of the human body and play a dynamic role in the health of the individual human [41]. Human gut co-habitants with various symbiotic microorganisms, including Bacteroides, Firmicutes, Actinobacteria, Proteobacteria, Fusobacteria, and Verrucomicrobia. The number and composition of these microbial species can vary according to age, diet, and lifestyle [42, 43]. The gut microbiota colonizes from the time of delivery and during breast feeding that gets continuously modulated during development due to dietary and environmental factors [44].

Food and Drug Administration (FDA) and World Health Organization (WHO) define probiotics as “live microorganisms that can provide health benefits to the host when administered in adequate amounts” [45]. Well-established probiotic bacteria includeLactobacillus rhamnosus, Lactobacillus reuteri, Bifidobacteria, Lactobacillus casei, Lactobacillus acidophilus, Bacillus coagulans, Escherichia coli strain Nissle 1917, and Enterococcus faecium SF68 [46]. In addition to protecting the intestinal tract, probiotics help improve various diseases, including cardiovascular disease, diabetes, and obesity [10, 47]. The well-known fact is that probiotics that restore intestinal microbiota balance support their efficiency in preventing the development of chronic immune-mediated diseases (Figure 2).

Figure 2.

A schematic representation of the external factors responsible for inflamed gut and the altered phenotype due to epigenetic modifications. The figure also depicts epigenetic therapy and the restoration of a healthy gut by epigenetic drugs, probiotics, and their metabolites.

As there was substantial evidence that the intestinal microbiota and probiotics impact several metabolic functions within the human body, such as digestion, absorption of nutrition, regulation of hormonal secretion, modulation of inflammation and immunity processes, synthesis of vitamins and various metabolites, dysbiosis of this microbiota negatively influences the onset of diseases such as obesity, diabetes, inflammatory bowel disease, cancer, etc. [44, 48, 49]. In recent years, the commensal microbiota has also been appreciated to play a prominent role in host epigenetics by altering gene expression, affecting homeostasis and health status [44, 50, 51]. Although the mechanism through which the gut microbiota regulates host epigenetic changes remains a mystery, this has, however, been revealed through studies with advanced computational tools for metagenomics that have focused on understanding the crosstalk between the microbiota and epigenetics [52].

3.1 Gut microbiota and host epigenetic features

Host epigenetics can influence the shaping of the microbiota in the intestine. Studies have shown that host miRNAs could control microbial gene transcription, thus modeling the structure of the microbial community, vice versa was observed where the gut microbiota and its metabolites were found to regulate epigenetic changes and influence host metabolism [41]. Short-chain fatty acids (SCFAs) are the main metabolites in host epigenome modulation that are produced by intestinal microbes and are found to affect body health and disease status [53, 54]. The gut microbiota synthesizes many bioactive compounds that serve as epigenetic substrates, cofactors, or regulators of epigenetic enzyme activity [55, 56]. For example, folate and B vitamins (B2, B12) donate methyl groups for DNA or histone methylation. SCFAs inhibit the deacetylase activity of histone deacetylases (HDACs), leading to changes in chromatin [57, 58]. Studies in rodent models have shown that the gut microbiota was found to influence host epigenetic transcriptome markers. Alteration of intestinal microflora in normal, modified, and germ-free (GF) mice was found to affect the modification of N6-methyladenosine (m6A) in the cecum and the decrease in expression in the liver, which ultimately affected host metabolism, inflammation, and antibacterial process. Akkermansia muciniphila and Lactobacillus plantarum could influence the modification of m6A in mono-associated mice [59]. This microbiota was also found to modulate host miRNA expression patterns to maintain intestinal homeostasis [60]. The intestinal microflora negatively regulates miR-107 expression in dendritic cells and macrophages, thus affecting the activity of the MyD88 and NF-B pathways, which subsequently influenced immune homeostasis and the expression of the target gene IL-23p19, which are responsible for oral cancer [61]. Probiotic supplementation could prevent and treat colon cancer by regulating miRNAs [6]. Coculture of probiotic Leuconostoc mesenteroides with HT-29 demonstrated effective down-regulation of miRNA-21 and miRNA-200b, thus promoting colon cancer cell apoptosis [62].

3.2 Probiotic and host epigenetics

Probiotics reduce the risk of certain diseases primarily through clinically validated mechanisms involving epigenetic regulation. The intestinal flora in the large intestine undergoes fermentation to transform dietary fiber into SCFAs, with butyrate being one of the major SCFAs produced [41]. SCFAs such as acetate, propionate, and butyrate are found to modulate the host’s immune system [63]. Gut bacteria and some probiotics produce enzymes such as methyltransferases, acetyltransferases, deacetylases, Bir A ligase, phosphotransferases, kinases, and synthetases. Furthermore, they produce S-adenosylmethionine (SAM), acetyl-CoA, NAD+, α-KG, and ATP, which are essential cofactors for many epigenetic processes that regulate DNA methylation, post-translational histone modifications, and nucleosome position [41, 44, 51]. Butyrate is an anti-inflammatory molecule produced by the gut microbiota and some probiotics can influence the host’s epigenome by decreasing intestinal permeability, preventing autoimmunity for type 1 diabetes (T1D), and protecting against autoimmunity of islets [64, 65].

SCFAs are the main probiotic metabolites that have been indicated to have a protective function in the intestine and exert anti-inflammatory effects in several animal and human models. SCFAs exert health-promoting actions by lowering intestinal pH, acting as energy sources for colon-inhabiting microbes, stimulating colonic blood flow, helping in the contraction of smooth muscle cells, enhancing transepithelial chloride secretion, and aiding in the proliferation of colonic epithelial cells. These SCFAs are the class of epigenetic drugs with histone deacetylase inhibitory activity (HDACi) that plays a vital role as anticancer agents with strong antiproliferative effects on tumor cells [66, 67]. The administration of SCFAs, such as butyrate and acetate, has shown promising results in ameliorating inflammatory lesions in mouse models of allergic airway disease and colitis [68]. The probiotic bacterium Propionibacterium freudenreichii produced a high level of SCFA, acetate, and propionate [69]. Butyrate, acetate, polyphenols, and vitamins are the main metabolites of intestinal microbes that participate in epigenetic processes. Table 1 lists some examples of probiotic bacteria that modulate the epigenetic makeup with their functional attributes. Studies have shown that butyrate can reduce inflammation (by increasing IL-10 expression) in inflammatory bowel disease and help protect against colitis and mortality [81]. Another LMW associated with epigenetic regulation is acetate, which is one of the immunomodulatory peptide drugs, “Glatiramer Acetate”. This was reported to increase the expression of the FOXP3 transcription factor (Forkhead box P3), which increased Treg cell proliferation and differentiation. This reduced the rate of diabetes and insulitis in NOD mice [82]. These SCFAs inhibit HDAC activity, protect against neurodegenerative diseases, and promote immunoglobulin secretion and intestinal mucosal barrier function [83]. Vitamins such as B2, B12, and B6 are essential for an enzyme that systemizes SA, the primary methyl-donating substrate for DNMT and HHMT. Probiotic species such as Lactobacillus and Bifidobacterium can confer better protective effects by biotinylating proteins in the intestines. This process is important for DNA repair and chromatin structure [84].

BacteriaEpigenetic ChangesHealth effectReference
LactobacilliDown-regulated miRNAs (miR-200b, miR-215, and miR-192)Maintain homeostasis and shape the host response to infection[70]
Leuconostoc mesenteroidesModulates miRNA-21, miRNA-200bPromote the apoptosis of colon cancer cells[62]
Fusobacterium nucleatumModulates the Toll-like receptor, miRNAs (miR-4802 and miR-18a)Improve the response to chemotherapy, reduce cancer recurrence,[71]
VSL#3 (VSL Pharmaceuticals, Ft Lauderdale, FL, USA): 3 × 1011 CFU/g of bifidobacteria (B. longum, B. infantis, and B. breve), lactobacilli (L. acidophilus, L. casei, L. delbrueckii subsp. L. bulgaricus and L. plantarum) and Streptococcus salivarius subsp. thermophilus.Increases IL-10 production from Peyer patches and the spleen, along with increased expression of IL-10 in the pancreas.Prevent the development of autoimmune diabetes in NOD mice and Induced immunomodulation by a reduction in insulitis severity[72]
Lactobacillus johnsonii N6.2-MediatedIncreases the cytokines IL-6 and IL-23 within the mesenteric lymph nodes of BBDP fed LjN6.2.Confirms the resistance of T1D[73]
Lactococcus lactisIncreases the frequencies of local Tregs accumulated in the pancreatic isletsRestorative therapy for autoimmune diabetes[74]
Bifidobacterium spp.Reduces the expression of insulin receptor substrate 1, protein kinase B (Akt / PBB), IKKa and IkBa, protein-1 (MCP-1) and interleukin-6, (IL-6)Promotes recovery of β-cells of pancreas cell-cells and increases insulin sensitivity in mice by enhancing the function of the insulin signaling pathway as a promising strategy for the treatment of diabetes.[75]
Lactobacillus reuteriinhibits osteoblast TNF-a signaling and osteoblast maturation markers.Blocks the loss of bones[76]
Lactobacillus kefiranofaciens M and Lactobacillus kefiri KStimulates GLP-1 secretion, inhibits pro-inflammatory and inflammatory cytokine production, increases IL-10 production, and modifies the intestinal microbiota cytokine TNFa and THPotentially inhibits type 1 diabetes progression in vitro and in vivo by improving GLP-1[77]
Lactobacillus brevis KLDS1.0727 and KLDS 1.0373GABA overexpressionInhibits the development of T1D in diabetics mice model[78]
Lactobacillus johnsonii N6.2decreases the K: T ratio significantly (CD45RO + CD183 + CD196–) and cytotoxic CD8+ T cells.Reduced risk of T1D[79]
Lactobacillus rhamnosus GG and Bifidobacterium lactis Bb12Prevents islet cell destructionReduce the risk of autoimmunity[80]

Table 1.

Probiotic bacteria with their epigenetic changes.

Advertisement

4. Epigenetic therapies

Epigenetic therapies are a promising new area of treatment for cancer and other diseases. They involve manipulating gene expression using drugs, probiotic bacteria, enzymes, or other molecules that interact with DNA. The gut microbiota and probiotics can affect the host epigenome by activating epigenetically silenced genes in cancer cells. Butyrate, produced by gut microbiota can prevent colon cancer and other diseases by reducing pro-angiogenic factors such as EGF and HIF 1α [85]. Sodium butyrate is an HDAC inhibitor and can increase cell death in human medulloblastoma cells [86]. Below we have discussed the epigenetic treatment for major organs that are effected by cancer, including lung, liver, gastric colon, breast, etc.

4.1 Epigenetic therapy in gastric cancer (GC)

GC is one of the most fatal forms of cancer in the world. Its grim prognosis is due to the complexity of the disease, late diagnosis, and unsatisfactory treatments. In addition to genetic changes and external influences, studies have shown that modifications in epigenetic processes are instrumental in the emergence and development of gastric tumors, which has become a distinctive feature of GC [87]. In most cases, it is caused by Helicobacter pylori infection characterized by chronic gastritis and peptic ulcers. H. pylori harboring the pathogenicity island of the cytotoxins-associated gene (CagPAI) induces the dephosphorylation of histone H3S10, H3 threonine 3, and the deacetylation of H3K23 in gastric epithelial cells [88]. DNMT inhibitors (DNMTi) and histone deacetylase inhibitors (HDACi) have been shown to be the most effective epigenetic drugs for treating GC in animal models. Nucleoside analogues of DNMTi (such as 5-azacitidine and 5-aza-dC or decitabine (DAC)) and non-nucleoside analogues of DNMTi (such as hydralazine) are separated according to their ability to integrate into freshly produced DNA [89]. A total of 485,512 methylation locations (482,421 at CpG sites and 3091 at non-CpG sites) from 55 cancer-related genes showed that epigenetic aberrations could alter various cancer-related pathways [87]. With the addition of the epigenetic drug to neoadjuvant epirubicin-oxaliplatin-capecitabine, a response rate of 67% was observed in patients with well-differentiated gastro-esophageal tumors, including 25% with a complete response. Hypomethylation of tumor-associated loci was observed at all 5-azacitidine doses, and the degrees of hypomethylation were correlated with a therapeutic response [90]. Gut bacteria can help fight cancer by producing bioactive metabolites that inhibit HDAC and HAT. These metabolites like SCFAs, propionate and butyrate can alter the epigenetic profile of cancer cells, making them more susceptible to treatment [91]. However, changing epigenetic profiles may represent new treatment approaches for GC. Cancer heterogeneity can be overcome and cancer homeostasis can be reprogrammed so that it is more likely to react to cytotoxic drugs or immune checkpoint inhibitors if epigenetic processes are targeted.

4.2 Epigenetic therapy in colon cancer

Colon cancer is one of the most fatal forms of the disease worldwide. Its genesis is linked to the collection of genetic and epigenetic modifications in the epithelial cells of the colon that cause them to morph into adenocarcinomas. In the last 10 years, there has been enormous progress in understanding cancer epigenetics, notably aberrant DNA methylation [92]. Microbial exposure was perceived to induce DNA hypomethylation, which increases the expression and activity of the DNA demethylase enzymes Tet3 and Dnmt1 [50, 93]. DNA methylation in colon biopsies is correlated with microbial composition, inflammation status, and disease classification in ulcerative colitis and Crohn’s disease patients [85]. Ulcerative colitis is associated with colorectal cancer, since Fusobacterium is associated with increased DNA methylation [94, 95, 96]. Yu et al. [71] demonstrated DNA methylation, particularly in the 3’CpG islands of glycosylation genes involved in cell maturation, which was substantially hampered in the absence of the intestinal microbiota. Changes in the fecal microbiota with decreased SCFA (acetate, propionate, and butyrate) have been attributed to the development and progression of colon cancer [97]. Studies have shown a low level of butyrate-producing bacteria in patients with colorectal cancer with a concurrent increase in mucin-degrading species such as Akkermansia muciniphila [98]. Probiotics can reduce the risk of colon cancer by improving intestinal microbiota balance. They can also reduce side effects of chemotherapy and radiation therapy by regulating neutrophil function and supporting anti-inflammatory activity by inducing TNF-α and miRNA-dependent expression of p21 gene [99, 100].

4.3 Epigenetic therapy in breast, ovarian, and endometrial cancer

Most ovarian cancers, 14–24%, are inherited diseases caused by gene mutations in BRCA1 and BRCA2 [101]. When BRCA1, BRCA2, BRIP1, RAD51C, RAD51D, and FANCM are altered, genomic instability results in ovarian cancer with an early somatic mutation in TP53 [102]. The epigenetic environment of these tumors can contribute to increased immune activity. Although durable and long-lasting responses have been shown in solid tumors such as melanoma, lung cancer, and renal cell carcinoma [103], checkpoint blockade therapies have not been successful in ovarian cancer. Six genes that help control the cell cycle—BRCA1, CDKN2A, RASSF1A, LOTI, DAPK, and ICAM-l—are suppressed when hypermethylated gene promoters are detected in cancer. Studies have shown that extensive cessation of CpG hypermethylation in ovarian cancer leads to slower growth of cancer cells [104]. The research found that ITF2357 and 5-azacytidine (AZA) inhibited the DNMT1 enzyme and stimulated T cell participation by upregulating ERV expression in a mouse model of ovarian tumor. This activated a type I interferon response by increasing the expression of endogenous retroviruses (ERV), pieces of ancient viral DNA that make up 8% of our genome [105]. Moufarrij et al. [106] demonstrated that DNMTi decitabine successfully treats ovarian cancer that does not respond to chemotherapy. Furthermore, Cicek et al. [107] determined that the epigenetic drugs can enhance expression of tumor antigen NY-ESO-1 by changing its methylation status. It is important to study the combined pharmacodynamics and multifactorial mechanisms of epigenetic drugs when combined with targeted or immune therapies.

The gut microbiome has been observed to modulate estrogen metabolism [108]. Shimizu et al. [109] showed a significant increase in the reproductive capacity of germ-free mice with the introduction of microbials. Bacterial introduction normalized the estrous cycle and increased the copulation and implantation rates. An aggregate of enteric bacterial genes in the human gut microbiome can influence the estrabolome. Bacterial β-glucuronidases and β-glucuronides enhance estrogen deconjugation and conjugation [110]. Deconjugation results in the re-absorption of free estrogens, leading to the development of estrogen-driven cancers such as breast, ovarian, and endometrial cancers. Daidzein, a class of hydroxy isoflavones, is converted to dihydrodaidzein, S-(−)equol, and O-desmethyl angiotensin by the concentration of equol in the urine of intestinal bacteria has been correlated with a reduced risk of breast cancer. Therefore, epigenetic modifications can affect cancer progression [111].

4.4 Epigenetic therapy in liver and lung cancer

Tumor forms of cancer are thought to result from oncogene activation and TSG silencing due to mutations in epigenetic regulatory pathways. These epigenetic alterations can also be related to resistance to chemotherapy [112]. Studies show that methylation of certain genes (like CDK2A, p16, CDH13, RASSF1A, and APC) correlates with the recurrence of stage I non-small cell lung cancer (NSCLC) after surgical resection and epigenetic changes that affect p16, and p16 expression are associated with reduced survival after early-stage NSCLC resection [113]. In a phase I/II trial, patients with advanced untreated NSCLC received an 8-hour continuous infusion of high-dose decitabine (200 to 660 mg/m2). Only one patient completed more than one cycle, affecting the efficacy of treatment [114]. Pharmacodynamic studies have found that one-third of patients with lung cancer have higher levels of p16, MAGE-3, and NY-ESO-1 [115]. Histone modifications, HAT, and HDAC inhibitors are currently used in lung cancer treatment, but are still a few years away from being used as lung cancer biomarkers and guide therapy [116].

Dietary regulation, balanced gut microflora, and epigenetic modification can be useful in preventing liver and lung cancer. Studies have shown that chromatin modifications and other epigenetics (especially miRNA) are critical to the development of chronic obstructive lung disease (COPD) and lung cancer [117, 118]. SCFAs produced by the gut microbiota bind to the G protein-coupled receptor 43 (GPR43), affecting inflammatory responses [30]. GPR43 is important in reducing inflammation in models of colitis, arthritis, and asthma [68].

Advertisement

5. Conclusions

As scientists continue to explore the potential applications of epigenetic reprogramming, the possibilities are endless. This research could transform the way a disease is treated and prevent it from occurring in the first place. There is much to learn about this process, but the medical and scientific communities are excited to explore the potential of these organisms. Epigenetic detection has been an important advance in cancer research. By shedding light on how cancer develops and progresses, new treatments have been developed on the basis of these findings. In spite of many challenges like aberrant promoter methylation, inactivated mutations, tumor micro-environment, and external factors, these developments could have a significant impact on the fight against cancer. Epigenetic therapies are a promising new field of medicine that has the potential to revolutionize healthcare. Epigenetic drugs and other therapies are being studied for their potential to treat a wide range of diseases, including cancer. The gut microbiome can be used to manage and treat cancer and gut-related diseases. The bioactive metabolites produced by beneficial probiotic microbes affect the epigenome and the differentiation and functioning of various immune cells, enterocytes, and pancreatic cells. Modifying the intestinal microbiome by repairing dysbiosis through diet intervention or fecal transplants would be a promising approach to treating metabolic syndromes. Additionally, several clinical trials of epigenetic therapy are currently underway and could offer insight into the potential of epigenetic therapies. A new generation of epigenetic therapies could revolutionize healthcare care and change the way we live in the near future.

Advertisement

Acknowledgments

The authors are grateful to the editors for invitation of the book chapter. The authors SCR and SPR sincerely thank the founder of SVR BioScience Research Services, Dr. Sundru Manjulata Devi for giving an opportunity to contribute to the review.

Advertisement

Conflict of interest

The authors declare no conflict of interest.

Advertisement

Authorship criteria

Concept and design of the book chapter was conceived by SMD. The epigenetic changes and therapy was contributed by SMD. SCR contributed and formulated the for role of CRISR-Cas9 and SPR promoted the role of gut microbiota and probiotics in epigenetic changes. Editing, revision and final approval was made by SMD, SCR and SPR.

References

  1. 1. Morovic W, Budinoff CR. Epigenetics: A new frontier in probiotic research. Trends in Microbiology. 2021;29:117-126. DOI: 10.1016/j.tim.2020.04.008
  2. 2. Nakamura M, Gao Y, Dominguez AA, Qi LS. CRISPR technologies for precise epigenome editing. Nature Cell Biology. 2021;23:11-22. DOI: 10.1038/s41556-020-00620-7
  3. 3. Keung AJ, Joung JK, Khalil AS, Collins JJ. Chromatin regulation at the frontier of synthetic biology. Nature Reviews. Genetics. 2015;16:159-171. DOI: 10.1038/nrg3900
  4. 4. Holtzman L, Gersbach CA. Editing the epigenome: Reshaping the genomic landscape. Annual Review of Genomics and Human Genetics. 2018;19:43-71. DOI: 10.1146/annurev-genom-083117-021632
  5. 5. Gasperini M, Tome JM, Shendure J. Towards a comprehensive catalogue of validated and target-linked human enhancers. Nature Reviews. Genetics. 2020;21:292-310. DOI: 10.1038/s41576-019-0209-0
  6. 6. Kumar M, Hemalatha R, Kumar R, Nagpal R, Devraj JP, Verma V, et al. Probiotics, prebiotics and synbiotics—A review. Cell. 2021;8:93-95. DOI: 10.4049/jimmunol.1001864
  7. 7. Jones PA, Issa J-PJ, Baylin S. Targeting the cancer epigenome for therapy. Nature Reviews. Genetics. 2016;17:630-641. DOI: 10.1038/nrg.2016.93
  8. 8. Wang Y, Yan L, Zhang Z, Prado E, Fu L, Xu X, et al. Epigenetic regulation and its therapeutic potential in pulmonary hypertension. Frontiers in Pharmacology. 2018;9:241. DOI: 10.3389/fphar.2018.00241
  9. 9. Roberti A, Valdes AF, Torrecillas R, Fraga MF, Fernandez AF. Epigenetics in cancer therapy and nanomedicine. Clinical Epigenetics. 2019;11:81. DOI: 10.1186/s13148-019-0675-4
  10. 10. Kesika P, Sivamaruthi BS, Chaiyasut C. Do probiotics improve the health status of individuals with diabetes mellitus? A review on outcomes of clinical trials. BioMed Research International. 2019;2019:1531567. DOI: 10.1155/2019/1531567
  11. 11. Xiong X, Chen M, Lim WA, Zhao D, Qi LS. CRISPR/Cas9 for human genome engineering and disease research. Annual Review of Genomics and Human Genetics. 2016;17:131-154. DOI: 10.1146/annurev-genom-083115-022258
  12. 12. Qi LS, Larson MH, Gilbert LA, Doudna JA, Weissman JS, Arkin AP, et al. Repurposing CRISPR as an RNA-guided platform for sequence-specific control of gene expression. Cell. 2013;152:1173-1183. DOI: 10.1016/j.cell.2013.02.022
  13. 13. Morton AR, Dogan-Artun N, Faber ZJ, MacLeod G, Bartels CF, Piazza MS, et al. Functional enhancers shape extrachromosomal oncogene amplifications. Cell. 2019;179:1330-1341.e13. DOI: 10.1016/j.cell.2019.10.039
  14. 14. Mirabella AC, Foster BM, Bartke T. Chromatin deregulation in disease. Chromosoma. 2016;125:75-93. DOI: 10.1007/s00412-015-0530-0
  15. 15. Franzago M, Fraticelli F, Nicolucci A, Celentano C, Liberati M, Stuppia L, et al. Molecular analysis of a genetic variants panel related to nutrients and metabolism: Association with susceptibility to gestational diabetes and Cardiometabolic risk in affected women. Journal Diabetes Research. 2017;2017:4612623. DOI: 10.1155/2017/4612623
  16. 16. Hidalgo-Cantabrana C, O’Flaherty S, Barrangou R. CRISPR-based engineering of next-generation lactic acid bacteria. Current Opinion in Microbiology. 2017;37:79-87. DOI: 10.1016/j.mib.2017.05.015
  17. 17. Pan M, Barrangou R. Combining omics technologies with CRISPR-based genome editing to study food microbes. Current Opinion in Biotechnology. 2020;61:198-208. DOI: 10.1016/j.copbio.2019.12.027
  18. 18. Canani RB, di Costanzo M, Leone L, Bedogni G, Brambilla P, Cianfarani S, et al. Epigenetic mechanisms elicited by nutrition in early life. Nutrition Research Reviews. 2011;24:198-205. DOI: 10.1017/s0954422411000102
  19. 19. Delgado S, Sánchez B, Margolles A, Ruas-Madiedo P, Ruiz L. Molecules produced by probiotics and intestinal microorganisms with immunomodulatory activity. Nutrients. 2020;12:391. DOI: 10.3390/nu12020391
  20. 20. Adli M. The CRISPR tool kit for genome editing and beyond. Nature Communications. 2018;9:1911. DOI: 10.1038/s41467-018-04252-2
  21. 21. Tadić V, Josipović G, Zoldoš V, Vojta A. CRISPR/Cas9-based epigenome editing: An overview of dCas9-based tools with special emphasis on off-target activity. Methods. 2019;164-165:109-119. DOI: 10.1016/j.ymeth.2019.05.003
  22. 22. Gilbert LA, Larson MH, Morsut L, Liu Z, Brar GA, Torres SE, et al. CRISPR-mediated modular RNA-guided regulation of transcription in eukaryotes. Cell. 2013;154:442-451. DOI: 10.1016/j.cell.2013.06.044
  23. 23. Kiani S, Chavez A, Tuttle M, Hall RN, Chari R, Ter-Ovanesyan D, et al. Cas9 gRNA engineering for genome editing, activation and repression. Nature Methods. 2015;12:1051-1054. DOI: 10.1038/nmeth.3580
  24. 24. Konermann S, Brigham MD, Trevino A, Hsu PD, Heidenreich M, Cong L, et al. Optical control of mammalian endogenous transcription and epigenetic states. Nature. 2013;500:472-476. DOI: 10.1038/nature12466
  25. 25. Tanenbaum ME, Gilbert LA, Qi LS, Weissman JS, Vale RD. A protein-tagging system for signal amplification in gene expression and fluorescence imaging. Cell. 2014;159:635-646. DOI: 10.1016/j.cell.2014.09.039
  26. 26. Konermann S, Brigham MD, Trevino AE, Joung J, Abudayyeh OO, Barcena C, et al. Genome-scale transcriptional activation by an engineered CRISPR-Cas9 complex. Nature. 2015;517:583-588. DOI: 10.1038/nature14136
  27. 27. Polstein LR, Gersbach CA. Light-inducible spatiotemporal control of gene activation by customizable zinc finger transcription factors. Journal of the American Chemical Society. 2012;134:16480-16483. DOI: 10.1021/ja3065667
  28. 28. Margolin JF, Friedman JR, Meyer WK, Vissing H, Thiesen HJ, Rauscher FJ 3rd. Krüppel-associated boxes are potent transcriptional repression domains. Proceedings of the National Academy of Sciences of the United States of America. 1994;91:4509-4513. DOI: 10.1073/pnas.91.10.4509
  29. 29. Fritsch L, Robin P, Mathieu JRR, Souidi M, Hinaux H, Rougeulle C, et al. A subset of the histone H3 lysine 9 methyltransferases Suv39h1, G9a, GLP, and SETDB1 participate in a multimeric complex. Molecular Cell. 2010;37:46-56. DOI: 10.1016/j.molcel.2009.12.017
  30. 30. Lu Y, Chan Y-T, Tan H-Y, Li S, Wang N, Feng Y. Epigenetic regulation in human cancer: The potential role of epi-drug in cancer therapy. Molecular Cancer. 2020;19:79. DOI: 10.1186/s12943-020-01197-3
  31. 31. Morita S, Noguchi H, Horii T, Nakabayashi K, Kimura M, Okamura K, et al. Targeted DNA demethylation in vivo using dCas9–peptide repeat and scFv–TET1 catalytic domain fusions. Nature Biotechnology. 2016;34:1060-1065. DOI: 10.1038/nbt.3658
  32. 32. Amabile A, Migliara A, Capasso P, Biffi M, Cittaro D, Naldini L, et al. Inheritable silencing of endogenous genes by hit-and-run targeted epigenetic editing. Cell. 2016;167:219-232.e14. DOI: 10.1016/j.cell.2016.09.006
  33. 33. Vojta A, Dobrinić P, Tadić V, Bočkor L, Korać P, Julg B, et al. Repurposing the CRISPR-Cas9 system for targeted DNA methylation. Nucleic Acids Research. 2016;44:5615-5628. DOI: 10.1093/nar/gkw159
  34. 34. Lei Y, Zhang X, Su J, Jeong M, Gundry MC, Huang Y-H, et al. Targeted DNA methylation in vivo using an engineered dCas9-MQ1 fusion protein. Nature Communications. 2017;8:16026. DOI: 10.1038/ncomms16026
  35. 35. Liu J, Yue Y, Han D, Wang X, Fu Y, Zhang L, et al. A METTL3-METTL14 complex mediates mammalian nuclear RNA N6-adenosine methylation. Nature Chemical Biology. 2014;10:93-95. DOI: 10.1038/nchembio.1432
  36. 36. Xu X, Tao Y, Gao X, Zhang L, Li X, Zou W, et al. A CRISPR-based approach for targeted DNA demethylation. Cell Discovery. 2016;2:16009. DOI: 10.1038/celldisc.2016.9
  37. 37. Choudhury SR, Cui Y, Lubecka K, Stefanska B, Irudayaraj J. CRISPR-dCas9 mediated TET1 targeting for selective DNA demethylation at BRCA1 promoter. Oncotarget. 2016;7:46545-46556. DOI: 10.18632/oncotarget.10234
  38. 38. Kearns NA, Pham H, Tabak B, Genga RM, Silverstein NJ, Garber M, et al. Functional annotation of native enhancers with a Cas9-histone demethylase fusion. Nature Methods. 2015;12:401-403. DOI: 10.1038/nmeth.3325
  39. 39. Hilton IB, D’Ippolito AM, Vockley CM, Thakore PI, Crawford GE, Reddy TE, et al. Epigenome editing by a CRISPR-Cas9-based acetyltransferase activates genes from promoters and enhancers. Nature Biotechnology. 2015;33:510-517. DOI: 10.1038/nbt.3199
  40. 40. Kwon DY, Zhao Y-T, Lamonica JM, Zhou Z. Locus-specific histone deacetylation using a synthetic CRISPR-Cas9-based HDAC. Nature Communications. 2017;8:15315. DOI: 10.1038/ncomms15315
  41. 41. Paul B, Barnes S, Demark-Wahnefried W, Morrow C, Salvador C, Skibola C, et al. Influences of diet and the gut microbiome on epigenetic modulation in cancer and other diseases. Clinical Epigenetics. 2015;7:112. DOI: 10.1186/s13148-015-0144-7
  42. 42. Hollister EB, Gao C, Versalovic J. Compositional and functional features of the gastrointestinal microbiome and their effects on human health. Gastroenterology. 2014;146:1449-1458. DOI: 10.1053/j.gastro.2014.01.052
  43. 43. Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano GAD, Gasbarrini A, et al. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms. 2019;7:14. DOI: 10.3390/microorganisms7010014
  44. 44. Sharma M, Li Y, Stoll ML, Tollefsbol TO. The epigenetic connection between the gut microbiome in obesity and diabetes. Frontiers in Genetics. 2020;10:1329. DOI: 10.3389/fgene.2019.01329
  45. 45. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, et al. The international scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nature Reviews Gastroenterology & Hepatology. 2014;11:506-514. DOI: 10.1038/nrgastro.2014.66
  46. 46. Pandey KR, Naik SR, Vakil B. Probiotics, prebiotics and synbiotics—A review. Journal of Food Science and Technology. 2015;52:7577-7587. DOI: 10.1007/s13197-015-1921-1
  47. 47. Moludi J, Maleki V, Jafari-Vayghyan H, Vaghef-Mehrabany E, Alizadeh M. Metabolic endotoxemia and cardiovascular disease: A systematic review about potential roles of prebiotics and probiotics. Clinical and Experimental Pharmacology & Physiology. 2020;47:927-939. DOI: 10.1111/1440-1681.13250
  48. 48. Cani PD, Everard A, Duparc T. Gut microbiota, enteroendocrine functions and metabolism. Current Opinion in Pharmacology. 2013;13:935-940. DOI: 10.1016/j.coph.2013.09.008
  49. 49. Louis P, Hold GL, Flint HJ. The gut microbiota, bacterial metabolites and colorectal cancer. Nature Reviews. Microbiology. 2014;12:661-672. DOI: 10.1038/nrmicro3344
  50. 50. Ansari I, Raddatz G, Gutekunst J, Ridnik M, Cohen D, Abu-Remaileh M, et al. The microbiota programs DNA methylation to control intestinal homeostasis and inflammation. Nature Microbiology. 2020;5:610-619. DOI: 10.1038/s41564-019-0659-3
  51. 51. Sharma S, Kelly TK, Jones PA. Epigenetics in cancer. Carcinogenesis. 2010;31:27-36. DOI: 10.1093/carcin/bgp220
  52. 52. Woo V, Alenghat T. Epigenetic regulation by gut microbiota. Gut Microbes. 2022;14:2022407. DOI: 10.1080/19490976.2021.2022407
  53. 53. Wang Y, Kuang Z, Yu X, Ruhn KA, Kubo M, Hooper L, et al. The intestinal microbiota regulates body composition through NFIL3 and the circadian clock. Science. 2017;357:912-916. DOI: 10.1126/science.aan0677
  54. 54. Cervantes-Barragan L, Chai JN, MaD T, di Luccia B, Ahern PP, Merriman J, et al. Lactobacillus reuteri induces gut intraepithelial CD4 + CD8αα + T cells. Science. 1979;2017(357):806-810. DOI: 10.1126/science.aah5825
  55. 55. Miro-Blanch J, Yanes O. Epigenetic regulation at the interplay between gut microbiota and host metabolism. Frontiers in Genetics. 2019;10:638. DOI: 10.3389/fgene.2019.00638
  56. 56. Shock T, Badang L, Ferguson B, Martinez-Guryn K. The interplay between diet, gut microbes, and host epigenetics in health and disease. The Journal of Nutritional Biochemistry. 2021;95:108631. DOI: 10.1016/j.jnutbio.2021.108631
  57. 57. Arpaia N, Campbell C, Fan X, Dikiy S, van der Veeken J, deRoos P, et al. Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation. Nature. 2013;504:451-455. DOI: 10.1038/nature12726
  58. 58. Chang P, v, Hao L, Offermanns S, Medzhitov R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proceedings of the National Academy of Sciences of the United States of America. 2014;111:2247-2252. DOI: 10.1073/pnas.1322269111
  59. 59. Jabs S, Biton A, Bécavin C, Nahori M-A, Ghozlane A, Pagliuso A, et al. Impact of the gut microbiota on the m(6)a epitranscriptome of mouse cecum and liver. Nature Communications. 2020;11:1344. DOI: 10.1038/s41467-020-15126-x
  60. 60. Dalmasso G, Nguyen HTT, Yan Y, Laroui H, Charania MA, Ayyadurai S, et al. Microbiota modulate host gene expression via microRNAs. PLoS One. 2011;6:e19293-e19293. DOI: 10.1371/journal.pone.0019293
  61. 61. Xue X, Cao AT, Cao X, Yao S, Carlsen ED, Soong L, et al. Downregulation of microRNA-107 in intestinal CD11c(+) myeloid cells in response to microbiota and proinflammatory cytokines increases IL-23p19 expression. European Journal of Immunology. 2014;44:673-682. DOI: 10.1002/eji.201343717
  62. 62. Zununi Vahed S, Barzegari A, Rahbar Saadat Y, Goreyshi A, Omidi Y. Leuconostoc mesenteroides-derived anticancer pharmaceuticals hinder inflammation and cell survival in colon cancer cells by modulating NF-κB/AKT/PTEN/MAPK pathways. Biomedicine & Pharmacotherapy. 2017;94:1094-1100. DOI: 10.1016/j.biopha.2017.08.033
  63. 63. Hullar MAJ, Fu BC. Diet, the gut microbiome, and epigenetics. Cancer Journal. 2014;20:170-175. DOI: 10.1097/PPO.0000000000000053
  64. 64. Endesfelder D, Zu Castell W, Ardissone A, Davis-Richardson AG, Achenbach P, Hagen M, et al. Compromised gut microbiota networks in children with anti-islet cell autoimmunity. Diabetes. 2014;63:2006-2014. DOI: 10.2337/db13-1676
  65. 65. Waldecker M, Kautenburger T, Daumann H, Busch C, Schrenk D. Inhibition of histone-deacetylase activity by short-chain fatty acids and some polyphenol metabolites formed in the colon. The Journal of Nutritional Biochemistry. 2008;19:587-593. DOI: 10.1016/j.jnutbio.2007.08.002
  66. 66. Marks PA, Richon VM, Miller T, Kelly WK. Histone deacetylase inhibitors. Adv Cancer Res. 2004;91:137-168. DOI: 10.1016/S0065-230X(04)91004-4
  67. 67. Acharya MR, Sparreboom A, Venitz J, Figg WD. Rational development of histone deacetylase inhibitors as anticancer agents: A review. Molecular Pharmacology. 2005;68:917-932. DOI: 10.1124/mol.105.014167
  68. 68. Maslowski KM, Vieira AT, Ng A, Kranich J, Sierro F, Yu D, et al. Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43. Nature. 2009;461:1282-1286. DOI: 10.1038/nature08530
  69. 69. Lan A, Lagadic-Gossmann D, Lemaire C, Brenner C, Jan G. Acidic extracellular pH shifts colorectal cancer cell death from apoptosis to necrosis upon exposure to propionate and acetate, major end-products of the human probiotic propionibacteria. Apoptosis. 2006;12:573-591. DOI: 10.1007/s10495-006-0010-3
  70. 70. Wieërs G, Belkhir L, Enaud R, Leclercq S, Philippart de Foy J-M, Dequenne I, et al. How probiotics affect the microbiota. Frontiers in Cellular and Infection Microbiology. 2020;9:454. DOI: 10.3389/fcimb.2019.00454
  71. 71. Yu T, Guo F, Yu Y, Sun T, Ma D, Han J, et al. Fusobacterium nucleatum promotes Chemoresistance to colorectal cancer by modulating autophagy. Cell. 2017;170:548-563.e16. DOI: 10.1016/j.cell.2017.07.008
  72. 72. Calcinaro F, Dionisi S, Marinaro M, Candeloro P, Bonato V, Marzotti S, et al. Oral probiotic administration induces interleukin-10 production and prevents spontaneous autoimmune diabetes in the non-obese diabetic mouse. Diabetologia. 2005;48:1565-1575. DOI: 10.1007/s00125-005-1831-2
  73. 73. Lau K, Benitez P, Ardissone A, Wilson TD, Collins EL, Lorca G, et al. Inhibition of type 1 diabetes correlated to a lactobacillus johnsonii N6.2-mediated Th17 bias. The Journal of Immunology. 2011;186:3538-3546. DOI: 10.4049/jimmunol.1001864
  74. 74. Takiishi T, Korf H, van Belle TL, Robert S, Grieco FA, Caluwaerts S, et al. Reversal of autoimmune diabetes by restoration of antigen-specific tolerance using genetically modified Lactococcus lactis in mice. The Journal of Clinical Investigation. 2012;122:1717-1725. DOI: 10.1172/JCI60530
  75. 75. Le TKC, Hosaka T, Nguyen TT, Kassu A, Dang TO, Tran HB, et al. Bifidobacterium species lower serum glucose, increase expressions of insulin signaling proteins, and improve adipokine profile in diabetic mice. Biomedical Research. 2015;36:63-70. DOI: 10.2220/biomedres.36.63
  76. 76. Zhang J, Motyl KJ, Irwin R, MacDougald OA, Britton RA, McCabe LR. Loss of bone and Wnt10b expression in male type 1 diabetic mice is blocked by the probiotic lactobacillus reuteri. Endocrinology. 2015;156:3169-3182. DOI: 10.1210/EN.2015-1308
  77. 77. Wei S-H, Chen Y-P, Chen M-J. Selecting probiotics with the abilities of enhancing GLP-1 to mitigate the progression of type 1 diabetes in vitro and in vivo. Journal of Functional Foods. 2015;18:473-486. DOI: 10.1016/j.jff.2015.08.016
  78. 78. Abdelazez A, Abdelmotaal H, Evivie SE, Melak S, Jia F-F, Khoso MH, et al. Screening potential probiotic characteristics of lactobacillus brevis strains in vitro and intervention effect on type I diabetes in vivo. BioMed Research International. 2018;2018:7356173. DOI: 10.1155/2018/7356173
  79. 79. Marcial GE, Ford AL, Haller MJ, Gezan SA, Harrison NA, Cai D, et al. Lactobacillus johnsonii N6.2 modulates the host immune responses: A double-blind, randomized trial in healthy adults. Frontiers in Immunology. 2017;8:655. DOI: 10.3389/fimmu.2017.00655
  80. 80. Groele L, Szajewska H, Szypowska A. Effects of lactobacillus rhamnosus GG and Bifidobacterium lactis Bb12 on beta-cell function in children with newly diagnosed type 1 diabetes: Protocol of a randomised controlled trial. BMJ Open. 2017;7:e017178-e017178. DOI: 10.1136/bmjopen-2017-017178
  81. 81. Sokol H, Seksik P, Furet JP, Firmesse O, Nion-Larmurier I, Beaugerie L, et al. Low counts of Faecalibacterium prausnitzii in colitis microbiota. Inflammatory Bowel Diseases. 2009;15:1183-1189. DOI: 10.1002/ibd.20903
  82. 82. Cui J, Li F, Shi Z-L. Origin and evolution of pathogenic coronaviruses. Nature Reviews. Microbiology. 2019;17:181-192. DOI: 10.1038/s41579-018-0118-9
  83. 83. Kibe R, Kurihara S, Sakai Y, Suzuki H, Ooga T, Sawaki E, et al. Upregulation of colonic luminal polyamines produced by intestinal microbiota delays senescence in mice. Scientific Reports. 2014;4:4548. DOI: 10.1038/srep04548
  84. 84. Dai Z, Zhang J, Wu Q , Chen J, Liu J, Wang L, et al. The role of microbiota in the development of colorectal cancer. International Journal of Cancer. 2019;145:2032-2041. DOI: 10.1002/ijc.32017
  85. 85. Ryan FJ, Ahern AM, Fitzgerald RS, Laserna-Mendieta EJ, Power EM, Clooney AG, et al. Colonic microbiota is associated with inflammation and host epigenomic alterations in inflammatory bowel disease. Nature Communications. 2020;11:1512. DOI: 10.1038/s41467-020-15342-5
  86. 86. Nör C, Sassi FA, de Farias CB, Schwartsmann G, Abujamra AL, Lenz G, et al. The histone deacetylase inhibitor sodium butyrate promotes cell death and differentiation and reduces Neurosphere formation in human Medulloblastoma cells. Molecular Neurobiology. 2013;48:533-543. DOI: 10.1007/s12035-013-8441-7
  87. 87. Canale M, Casadei-Gardini A, Ulivi P, Arechederra M, Berasain C, Lollini P-L, et al. Epigenetic mechanisms in gastric cancer: Potential new therapeutic opportunities. International Journal of Molecular Sciences. 2020;21:5500. DOI: 10.3390/ijms21155500
  88. 88. Ding S-Z, Goldberg JB, Hatakeyama M. Helicobacter pylori infection, oncogenic pathways and epigenetic mechanisms in gastric carcinogenesis. Future Oncology. 2010;6:851-862. DOI: 10.2217/fon.10.37
  89. 89. Zhou Z, Li H-Q , Liu F. DNA methyltransferase inhibitors and their therapeutic potential. Current Topics in Medicinal Chemistry. 2019;18:2448-2457. DOI: 10.2174/1568026619666181120150122
  90. 90. Schneider BJ, Shah MA, Klute K, Ocean A, Popa E, Altorki N, et al. Phase I study of epigenetic priming with Azacitidine prior to standard neoadjuvant chemotherapy for patients with Resectable gastric and esophageal adenocarcinoma: Evidence of tumor Hypomethylation as an indicator of major histopathologic response. Clinical Cancer Research. 2017;23:2673-2680. DOI: 10.1158/1078-0432.CCR-16-1896
  91. 91. Sun D, Chen Y, Fang J-Y. Influence of the microbiota on epigenetics in colorectal cancer. National Science Review. 2019;6:1138-1148. DOI: 10.1093/nsr/nwy160
  92. 92. Lao VV, Grady WM. Epigenetics and colorectal cancer. Nature Reviews. Gastroenterology & Hepatology. 2011;8:686-700. DOI: 10.1038/nrgastro.2011.173
  93. 93. Poupeau A, Garde C, Sulek K, Citirikkaya K, Treebak JT, Arumugam M, et al. Genes controlling the activation of natural killer lymphocytes are epigenetically remodeled in intestinal cells from germ-free mice. The FASEB Journal. 2019;33:2719-2731. DOI: 10.1096/fj.201800787R
  94. 94. Eaden JA, Abrams KR, Mayberry JF. The risk of colorectal cancer in ulcerative colitis: A meta-analysis. Gut. 2001;48:526-535. DOI: 10.1136/gut.48.4.526
  95. 95. Tahara T, Hirata I, Nakano N, Tahara S, Horiguchi N, Kawamura T, et al. Potential link between fusobacterium enrichment and DNA methylation accumulation in the inflammatory colonic mucosa in ulcerative colitis. Oncotarget. 2017;8:61917-61926. DOI: 10.18632/oncotarget.18716
  96. 96. Linson EA, Hanauer SB. Epidemiology of colorectal cancer in inflammatory bowel disease—The evolving landscape. Current Gastroenterology Reports. 2021;23(9):1-16. DOI: 10.1007/s11894-021-00816-3
  97. 97. Ohigashi S, Sudo K, Kobayashi D, Takahashi O, Takahashi T, Asahara T, et al. Changes of the intestinal microbiota, short chain fatty acids, and fecal pH in patients with colorectal cancer. Digestive Diseases and Sciences. 2013;58:1717-1726. DOI: 10.1007/s10620-012-2526-4
  98. 98. Weir TL, Manter DK, Sheflin AM, Barnett BA, Heuberger AL, Ryan EP. Stool microbiome and metabolome differences between colorectal cancer patients and healthy adults. PLoS One. 2013;8:e70803-e70803. DOI: 10.1371/journal.pone.0070803
  99. 99. Hýžd’alová M, Hofmanová J, Pacherník J, Vaculová A, Kozubík A. The interaction of butyrate with TNF-α during differentiation and apoptosis of colon epithelial cells: Role of NF-κB activation. Cytokine. 2008;44:33-43. DOI: 10.1016/j.cyto.2008.06.003
  100. 100. Hu S, Dong TS, Dalal SR, Wu F, Bissonnette M, Kwon JH, et al. The microbe-derived short chain fatty acid butyrate targets miRNA-dependent p21 gene expression in human colon cancer. PLoS One. 2011;6:e16221-e16221. DOI: 10.1371/journal.pone.0016221
  101. 101. Walsh T, Casadei S, Lee MK, Pennil CC, Nord AS, Thornton AM, et al. Mutations in 12 genes for inherited ovarian, fallopian tube, and peritoneal carcinoma identified by massively parallel sequencing. Proceedings of the National Academy of Sciences of the United States of America. 2011;108:18032-18037. DOI: 10.1073/pnas.1115052108
  102. 102. Kroeger PT Jr, Drapkin R. Pathogenesis and heterogeneity of ovarian cancer. Current Opinion in Obstetrics & Gynecology. 2017;29:26-34. DOI: 10.1097/GCO.0000000000000340
  103. 103. Jelinic P, Ricca J, van Oudenhove E, Olvera N, Merghoub T, Levine DA, et al. Immune-active microenvironment in small cell carcinoma of the ovary, hypercalcemic type: Rationale for immune checkpoint blockade. Journal of the National Cancer Institute. 2018;110:787-790. DOI: 10.1093/jnci/djx277
  104. 104. Ahluwalia A, Yan P, Hurteau JA, Bigsby RM, Jung SH, Huang TH-M, et al. DNA methylation and ovarian cancer. Gynecologic Oncology. 2001;82:261-268. DOI: 10.1006/gyno.2001.6291
  105. 105. Stone ML, Chiappinelli KB, Li H, Murphy LM, Travers ME, Topper MJ, et al. Epigenetic therapy activates type I interferon signaling in murine ovarian cancer to reduce immunosuppression and tumor burden. Proceedings of the National Academy of Sciences of the United States of America. 2017;114:E10981-E10990. DOI: 10.1073/pnas.1712514114
  106. 106. Moufarrij S, Dandapani M, Arthofer E, Gomez S, Srivastava A, Lopez-Acevedo M, et al. Epigenetic therapy for ovarian cancer: Promise and progress. Clinical Epigenetics. 2019;11:7. DOI: 10.1186/s13148-018-0602-0
  107. 107. Cicek MS, Koestler DC, Fridley BL, Kalli KR, Armasu SM, Larson MC, et al. Epigenome-wide ovarian cancer analysis identifies a methylation profile differentiating clear-cell histology with epigenetic silencing of the HERG K+ channel. Human Molecular Genetics. 2013;22:3038-3047. DOI: 10.1093/hmg/ddt160
  108. 108. Fuhrman BJ, Feigelson HS, Flores R, Gail MH, Xu X, Ravel J, et al. Associations of the fecal microbiome with urinary estrogens and estrogen metabolites in postmenopausal women. The Journal of Clinical Endocrinology and Metabolism. 2014;99:4632-4640. DOI: 10.1210/jc.2014-2222
  109. 109. Shimizu K, Muranaka Y, Fujimura R, Ishida H, Tazume S, Shimamura T. Normalization of reproductive function in germfree mice following bacterial contamination. Experimental Animals. 1998;47:151-158. DOI: 10.1538/expanim.47.151
  110. 110. Plottel CS, Blaser MJ. Microbiome and malignancy. Cell Host & Microbe. 2011;10:324-335. DOI: 10.1016/j.chom.2011.10.003
  111. 111. Rowland IR, Wiseman H, Sanders TAB, Adlercreutz H, Bowey EA. Interindividual variation in metabolism of soy Isoflavones and Lignans: Influence of habitual diet on Equol production by the gut microflora. Nutrition and Cancer. 2000;36:27-32. DOI: 10.1207/s15327914nc3601_5
  112. 112. Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nature Reviews. Genetics. 2002;3:415-428. DOI: 10.1038/nrg816
  113. 113. Brock M, Hooker CM, Ota-Machida E, Han Y, Guo M, Ames S, et al. DNA methylation markers and early recurrence in stage I lung cancer. New England Journal of Medicine. 2008;358:1118-1128. DOI: 10.1056/nejmoa0706550
  114. 114. Momparler RL, Ayoub J. Potential of 5-aza-2′-deoxycytidine (Decitabine) a potent inhibitor of DNA methylation for therapy of advanced non-small cell lung cancer. Lung Cancer. 2001;34:111-115. DOI: 10.1016/s0169-5002(01)00397-x
  115. 115. Schrump DS, Fischette MR, Nguyen DM, Zhao M, Li X, Kunst TF, et al. Phase I study of decitabine-mediated gene expression in patients with cancers involving the lungs, esophagus, or pleura. Clinical Cancer Research. 2006;12:5777-5785. DOI: 10.1158/1078-0432.ccr-06-0669
  116. 116. Ansari J, Shackelford RE, El-Osta H. Epigenetics in non-small cell lung cancer: From basics to therapeutics. Translational Lung Cancer Research. 2016;5:155-171. DOI: 10.21037/tlcr.2016.02.02
  117. 117. Yao H, Rahman I. Current concepts on the role of inflammation in COPD and lung cancer. Current Opinion in Pharmacology. 2009;9:375-383. DOI: 10.1016/j.coph.2009.06.009
  118. 118. Shin JI, Brusselle GG. Mechanistic links between COPD and lung cancer: A role of microRNA let-7? Nature Reviews. Cancer. 2013;14:70. DOI: 10.1038/nrc3477-c1

Written By

Surya Chandra Rao Thumu, Shobha Rani Papanna and Sundru Manjulata Devi

Submitted: 03 February 2023 Reviewed: 21 February 2023 Published: 16 March 2023