Next Article in Journal
Bioprospecting the Skin Microbiome: Advances in Therapeutics and Personal Care Products
Next Article in Special Issue
Gastric Microbiota Gender Differences in Subjects with Healthy Stomachs and Autoimmune Atrophic Gastritis
Previous Article in Journal
Biologically Active Compounds from Probiotic Microorganisms and Plant Extracts Used as Biopreservatives
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Probiotic-Derived Bioactive Compounds in Colorectal Cancer Treatment

Laboratory of General Biology, Department of Genetics, Development and Molecular Biology, School of Biology, Faculty of Sciences, Aristotle University of Thessaloniki, 54 124 Thessaloniki, Greece
*
Author to whom correspondence should be addressed.
Microorganisms 2023, 11(8), 1898; https://doi.org/10.3390/microorganisms11081898
Submission received: 30 June 2023 / Revised: 14 July 2023 / Accepted: 24 July 2023 / Published: 27 July 2023
(This article belongs to the Special Issue Gut Microbiota in Disease, Second Edition)

Abstract

:
Colorectal cancer (CRC) is a multifactorial disease with increased morbidity and mortality rates globally. Despite advanced chemotherapeutic approaches for the treatment of CRC, low survival rates due to the regular occurrence of drug resistance and deleterious side effects render the need for alternative anticancer agents imperative. Accumulating evidence supports that gut microbiota imbalance precedes the establishment of carcinogenesis, subsequently contributing to cancer progression and response to anticancer therapy. Manipulation of the gut microbiota composition via the administration of probiotic-derived bioactive compounds has gradually attained the interest of scientific communities as a novel therapeutic strategy for CRC. These compounds encompass miscellaneous metabolic secreted products of probiotics, including bacteriocins, short-chain fatty acids (SCFAs), lactate, exopolysaccharides (EPSs), biosurfactants, and bacterial peptides, with profound anti-inflammatory and antiproliferative properties. This review provides a classification of postbiotic types and a comprehensive summary of the current state of research on their biological role against CRC. It also describes how their intricate interaction with the gut microbiota regulates the proper function of the intestinal barrier, thus eliminating gut dysbiosis and CRC development. Finally, it discusses the future perspectives in precision-medicine approaches as well as the challenges of their synthesis and optimization of administration in clinical studies.

1. Introduction

Colorectal cancer (CRC) represents the third most prevalent form of cancer worldwide, accounting for more than 1.9 million new incidents and 900,000 deaths in 2020 [1,2]. CRC is a multifactorial and heterogeneous non-communicable disease. Approximately 65% of CRC cases develop sporadically through the accumulation of acquired somatic mutations and epigenetic modifications [3], while other cases are associated with CRC predisposition genes (25%) and hereditary syndromes (5%) [4,5]. The development of sporadic CRCs is usually a multistep and long-lasting process that involves progressive transformation of normal intestinal epithelial cells (IECs) into malignant cells [2]. Based on the epithelial lesion type and the specific molecular profile, two distinct carcinogenesis pathways have been recognized [1]: the traditional adenoma–carcinoma pathway [6] and the serrated neoplasia pathway [7].
Expedient options of CRC treatment, including surgery, radiotherapy, targeted therapy, immunotherapy, and chemotherapy [1], are based on tumor-related features [8]. Chemotherapeutic intervention with palliative purposes is often selected to enhance the survival rate of CRC patients [3]. However, the efficacy of current strategies is usually imperiled, due to drugs’ inadequate capacity to discriminate between healthy and cancer cells, thus causing intensified toxicity and undesirable effects to the human body [2]. Additionally, chemotherapy rarely accomplishes the complete eradication of malignant cells, while it can promote drug-resistance development [9]. Cancer cells display a variety of remarkable changes in their physiology [10]. Recently, this list has been expanded to incorporate the effects of gut microbiota composition on the initiation and/or progression of tumorigenesis [11].
Gut microbiota, the complex microbial population inhabiting the gastrointestinal tract (GIT), can shape oncologic outcomes in several ways [12]. Global epidemiological studies suggest that variations in gut microbiota composition and diversity, known as gut dysbiosis, can influence CRC initiation and progression [13]. The involvement of gut microbes in modulating physiological processes could be reversed in case of dysbiosis towards chronic inflammation and CRC induction [14]. Depending on the predominant gut microbiota-induced fermentation pathway, dietary patterns reflect on the enrichment of oncometabolites or tumor-suppressing metabolites [15]. Gut microbiota metabolism is closely associated with the efficacy and toxicity of traditional chemotherapeutic treatments [16]. In some cases, gut metabolites induce an immunostimulatory tumor microenvironment (TME) that advocates drugs toxicity on tumor cells [17]. However, a substantial number of patients experience treatment-associated adverse effects or even mortality due to these medications, a fact attributed to their intestinal microbial diversity [16]. Deciphering the synergistic or contradictory action of gut microbiota with anticancer agents remains a conundrum.
In recent years, probiotics have been utilized to attenuate postoperative gastrointestinal complications in eligible CRC patients undergoing chemotherapy [18]. Lactic acid bacteria (LAB), a ubiquitous group of Gram-positive microorganisms, comprise the most common type of probiotics due to their beneficial health effects on the host and their “generally recognized as safe” (GRAS) status [19]. The anticancer activity of probiotics is predicated on their ability to suppress tumor growth as well as induce cell cycle arrest and apoptosis [20,21]. Nevertheless, probiotics have been reported to biotransform chemical compounds [22] and anticancer agents [23], thus affecting drugs’ bioavailability and therapeutic outcomes, or even leading to disease exacerbation [24]. In this regard, administration of probiotic-derived bioactive compounds with profound anti-inflammatory and antiproliferative properties is now being leveraged as a novel personalized therapeutic approach in CRC treatment [25]. These secreted metabolic products purified from the probiotic cell-free supernatant (CFS) comprehend short-chain fatty acids (SCFAs), bacteriocins, exopolysaccharides (EPSs), nonribosomal lipopeptides, and other bacterial peptides [26] (Figure 1).
This review provides a concise summary of the most prominent probiotic-derived compounds and emphasizes the current knowledge regarding their biological roles in CRC treatment. Additionally, it focuses on the underlying mechanisms that configure their antiproliferative effects on malignant cells, as well as how their intricate interaction with the gut microbiota can lead to the elimination of dysbiosis. Finally, it discusses the future perspectives and challenges of their administration to individuals.

2. The Role of Gut Microbiota in CRC Development and Treatment

The human gut microbiota encompasses about a hundred trillion microorganisms, a number three times greater than the total number of human cells [5,27], while it encodes more than three million genes [28]. This diverse microbial community harbors between 500 to 1000 bacterial species, mostly belonging to the Bacteroidetes and Firmicutes phyla [29], and is strictly compartmentalized to the intestinal lumen of the GIT. The establishment of the gut microbiota occurs during infancy [30], while it stabilizes and reaches its peak complexity in adulthood [31]. Its composition varies among individuals and is predominantly shaped by various factors including nutrition [32], antibiotic treatment [33], age, and ethnicity [34]. A healthy gut microbiota plays a fundamental role in host nutrient metabolism, [35], immunomodulation [36,37], maintenance of the mucosal barrier integrity [38], production of antimicrobial and anti-inflammatory compounds [39,40], and protection against intruding pathogens [5].
The qualitative and quantitative alterations in the gut microbiota composition, namely dysbiosis, are often associated with increased susceptibility to gastrointestinal malignancies [41]. Gut dysbiosis is characterized by the overgrowth of proinflammatory bacterial species at the expense of beneficial microbes [42], thus leading to disturbance of epithelial barrier function, chronic inflammation, oxidative stress [43], and colorectal tumorigenesis [44]. In fact, approximately 20% of cancers, including CRC, are hypothesized to be attributed to microbes [45]. Several studies have indicated the association between gut microbiota dysbiosis and cancer development [46,47,48,49]. CRC occurrence is generally associated with the prevalence of specific bacterial species [50,51], such as Fusobacterium nucleatum [52], Bacteroides fragilis [53,54], Escherichia coli [55], Streptococcus bovis and Streptococcus gallolyticus [56], Helicobacter pylori [57], Salmonella typhimurium [58], Clostridium sp. [59], and Citrobacter rodentium [60]. These microorganisms induce carcinogenesis via activation of signaling pathways, toxic metabolites production, and extensive DNA damage [41].
The role of the gut microbiota in CRC development is supported by multiple studies [61,62,63,64,65,66] and explained by the “Driver-Passenger” model [67]. According to this model, “driver” pathogenic bacteria can induce colorectal tumorigenesis via the production of toxins and digestion of the protective mucus layer shielding the intestinal epithelium. The CRC microenvironment stimulates the overgrowth of specific “passenger” opportunistic bacteria that further promote dysbiosis, epithelial cell hyperproliferation, and chronic inflammation, thus leading to CRC progression [68]. In vivo studies confirmed that fecal microbiota transplantation (FMT) from CRC patients into germ-free [69] and Adenomatous polyposis coli (APC) gene knockout mice [70] facilitated intestinal tumorigenesis. Furthermore, significant differences regarding gut microbiota composition were detected between healthy individuals and CRC patients, which were correlated with the expression of genes known to promote inflammatory responses [71,72]. The gut microbiota may be harnessed through establishing microbial therapeutics as chemopreventive agents [73,74,75,76,77,78,79,80], as adjuvants to augment drug efficacy [81,82], or as diagnostic biomarkers for CRC screening [83,84,85,86,87].
Several studies have indicated that medications can significantly affect the gut microbiota, thus playing a pivotal role in disease development and therapy [85,88,89]. At the same time, gut microbes are implicated in drug pharmacokinetics [90], pinpointing this as one of the most challenging aspects of developing individual-specific anticancer agents to improve therapeutic outcomes. Surprisingly, the role of the gut microbiota in CRC therapy is rather supported by conflicting results. The contribution of the gut microbiota in enhanced therapeutic efficacy has been reported, while, concomitantly, the metabolic activity of gut bacteria is a critical trait in side effect exacerbation. For instance, irinotecan is mostly used intravenously to treat CRC. However, it commonly causes severe diarrhea upon its conversion into the active metabolic form by microbial β-glucuronidase enzymes. Those side effects are mitigated via the utilization of β-glucuronidase inhibitors [90]. Additionally, 5-Fluorouracil (5-FU) was found to inhibit the growth of CRC-related F. nucleatum, while specific intratumoral microbiota members can covert 5-FU into a nontoxic form, thus resulting in increased cancer epithelial cell growth [91].

3. Probiotic Derived Bioactive Compounds and CRC

3.1. Cell-Free Supernatant

The cell-free supernatant (CFS) encompasses diverse bioactive metabolites secreted by probiotics during microbial growth [25,92] as well as the remains of the culture medium [93]. The typical procedure for CFS preparation involves two main steps: the removal of bacterial cells via centrifugation and the filtration of the emerged mixture to obtain a sterile, transparent liquid medium [94,95] (Figure 1). Optionally, the CFS can be subjected to lyophilization prior to use [96]. The compositional profile of CFS-derived compounds, ranging from proteinaceous molecules to organic acids, is generally altered by individual nutrients in the growth medium [96], thus endowing the CFS with variegated health-promoting effects [92,93].
The antiproliferative properties of CFSs against CRC cells originate from probiotics, mainly of the genus Lactobacillus [97,98,99,100,101,102,103,104,105,106,107], Bacillus [108,109], Enterococcus [110], Bifidobacterium [111], Leuconostoc [112], or commensal bacteria [113,114] and have been accredited in vitro (Table 1). The CFS can promote the activation of the intrinsic apoptotic pathway as indicated by increased caspase expression [115,116,117,118,119,120] and other major biochemical changes, including the loss of mitochondrial membrane potential and cytochrome c release, down-regulation of the anti-apoptotic BCL2 gene, and the up-regulation of the pro-apoptotic BAK, BAD, and BAX genes [121,122]. Moreover, apoptosis-associated morphological alterations such as the formation of cytoplasmic blebs, chromatin condensation, and DNA fragmentation have been observed [123,124,125,126]. In addition, the potent anti-inflammatory properties of Pediococcus acidilactici supernatant in LPS-pretreated CRC cells [127], as well as the suppression of pro-inflammatory cytokine production in pathogen-stimulated Caco-2 cells by the CFS from Lacticaseibacillus sp. [128] have been reported. Interestingly, Clostridium butyricum TO-A supernatant significantly down-regulates Toll-like receptor-4 (TLR4) expression and this effect is attributed to the high content of butyrate [129].
The anti-metastatic effects of various CFS have been previously observed [130,131,132,133,134,135]. For instance, CFS derived from Lactobacillus rhamnosus GG was found to prevent CRC cell invasion via reduction of matrix metalloproteinase-9 (MMP-9) expression and increased tight junction protein zona occludens-1 (ZO-1) and tissue inhibitor of metalloproteinase (TIMP) levels [130,131], whereas it exhibits synergistic action with 5-FU [136]. Lactiplantibacillus plantarum and Lactobacillus fermentum CFS induced a dramatic increase in apoptosis marker levels in three-dimensional (3D) spheroids of CRC cells in vitro [137,138]. In addition, various CFSs have been reported to suppress the expression of cyclin genes, thus affecting cell cycle progression [139,140]. A CFS derived from Lactobacillus plantarum CCARM 0067 resulted in Wnt/β-catenin suppression when combined with 5-FU [141], while it contributed to the restoration of sodium-coupled monocarboxylate transporter 1 (SMCT1) expression leading to butyrate-induced antiproliferative effects in 5-FU resistant CRC cells [142]. The inhibition of autophagy-related proteins and synergistic effects with chloroquine were observed in the case of Lactobacillus plantarum CFS-treated CRC cells [143]. Lastly, there is also evidence of the beneficial effects of CFSs derived from yeast cultures [144,145].
Table 1. In vitro effects of cell-free supernatants on CRC cells.
Table 1. In vitro effects of cell-free supernatants on CRC cells.
Probiotic StrainCRC Cell LineEffect/Mechanism of ActionReference
Bacillus coagulans Unique IS2COLO 205cytotoxic effect, apoptosis induction (↑ Bax/Bcl-2 ratio/
MtMP loss/cyt c release/↑ caspase-3/PARP cleavage)
[116]
Bacillus polyfermenticusHT-29, DLD-1,
Caco-2
antiproliferative activity, ErbB-2 and ErbB-3 inhibition[108]
Bacillus polyfermenticus KU3LoVo, HT-29anti-inflammatory and cytotoxic activity[109]
Bifidobacterium adolescentis SPM0212HT-29, SW-480, Caco-2dose-dependent anticancer activity, changes in cellular
morphology, ↓ TNF-α, inhibition of harmful fecal enzymes
[124]
Bifidobacterium bifidumSW742cytotoxic effect[111]
Clostridium butyricum TO-AHT-29TLR4 down-regulation[129]
Enterococcus faecium 12a
E. faecium L12b
E. hirae 20c
HCT-15dose-dependent cytotoxic effect,
apoptosis-related morphological changes
[125]
E. lactis IW5HT-29,
Caco-2
time- and dose-dependent cytotoxic activity,
extrinsic apoptotic pathway
[110]
Faecalibacterium prausnitziiHCT 116time- and dose-dependent cytotoxic activity[113]
Lacticaseibacillus paracasei SD1, Lacticaseibacillus rhamnosus SD4, SD11 and GGCaco-2dose-dependent cytotoxic effect, pro-inflammatory cytokine suppression after stimulation with pathogens[128]
Lactiplantibacillus plantarum 0991Caco-2dose-dependent antiproliferative activity,
↑ oxidative stress, intrinsic apoptotic pathway
[120]
Lactiplantibacillus plantarum L125HT-29antiproliferative, anti-clonogenic and anti-migration activity[135]
Lactiplantibacillus plantarum OC01HCT 116,
HT-29
dose-dependent cell toxicity (2D/3D-spheroid cultures),
mTOR and ERK pathways suppression,
E- to N-Cadherin switch inhibition
[138]
Levilactobacillus brevis 0983Caco-2dose-dependent antiproliferative activity,
↑ oxidative stress, intrinsic apoptotic pathway
[120]
Lactobacillus spp.
L. acidophilus ATCC 43121HT-29antiproliferative and antioxidant properties,
apoptosis induction (↑ caspase-3,-9/↑ Bax/Bcl-2 ratio)
[121]
L. acidophilus CICC 6074HT-29time- and dose-dependent cytotoxic activity,
cell cycle arrest (G0/G1), intrinsic apoptotic pathway
(MtMP loss/cyt c release/↑ BAX, CASP3, CASP9/↓ BCL2)
[117]
L. acidophilus IIA-2B4WiDrdose-dependent anticancer activity[106]
L. brevis PM177HT-29dose-dependent cytotoxic effect[101]
L. casei ATCC 334HCT 116anti-metastatic effects (↓ MMP-9/↑ ZO-1)[130]
L. casei ATCC 393HT-29antiproliferative effect[100]
L. casei M3HT-29,
Caco-2
antiproliferative and anti-migration activity,
VEGF/MMPs signaling pathway down-regulation
[134]
L. casei strainsCaco-2dose-dependent cytotoxic effects, apoptosis induction[107]
L. crispatus SJ-3C-USHT-29anti-metastatic effects (↓ MMP2 and MMP9/
TIMP1 and TIMP2)
[131]
L. delbrueckiiSW-620dose-dependent anticancer activity, anti-metastatic effects,
cell cycle arrest (G1), intrinsic apoptotic pathway
[115]
L. delbrueckii ATCC 11842HT-29antiproliferative and antioxidant properties,
apoptosis induction (↑ caspase-3,-9/↑ Bax/Bcl-2 ratio)
[121]
L. fermentumDLD-1, HT-29,
WiDr
dose-dependent cytotoxic activity (2D/3D-spheroid
cultures), apoptosis markers, NF-κB pathway inhibition
[137]
L. fermentum NCIMB 5221SW-480, Caco-2time-dependent antiproliferative effect, apoptosis induction[98]
L. johnsonii LC1HT-29,
HT29-dx
↓ cell viability, ↑ mitochondrial ROS production[103]
L. pentosus S3
L. pentosus B281Caco-2, HT-29↓ cell proliferation, cell cycle arrest (G1), ↓ cyclin genes[139]
L. plantarum A7Caco-2, HT-29antiproliferative effect[97]
L. plantarum ATCC 14,917Caco-2time- and dose-dependent cytotoxic activity,
intrinsic apoptotic pathway (↓ BCL2/
↑ caspase-3, -9, BAK, BAD, and BAX)
[122]
L. plantarum B282Caco-2, HT-29↓ cell proliferation, cell cycle arrest (G1), ↓ cyclin genes[139]
L. plantarum CCARM 0067HT-29/5-FUR,
HCT 116/5-FUR
↓ CSCs markers, caspase-3 dependent apoptosis and
Wnt/β-catenin suppression in combination with 5-FU
[141]
HCT 116/5-FURanti-metastatic effects, ↓ CLDN-1[132]
HCT 116,
HCT 116/5-FUR
restoration of SMCT1 expression leading to butyrate-induced antiproliferative effect and apoptosis[142]
L. plantarum IIA-1A5WiDrdose-dependent anticancer activity[106]
L. plantarum KCTC 3108Caco-2↓ cell viability, ↓ autophagy-related proteins,
induction of mitochondrial dysfunction,
synergistic effect with chloroquine
[143]
L. plantarum S2 and O2HT-29,
HT29-dx
↓ cell viability, ↑ mitochondrial ROS production[103]
L. plantarum strainsHT-29antiproliferative effect, induction of apoptosis[102]
L. plantarum YYC-3HT-29,
Caco-2
antiproliferative and anti-migration activity,
VEGF/MMPs signaling pathway down-regulation
[134]
L. reuteri BCRC14625HT-29cell membrane damage, LDH release,
Bcl-2 inhibition via ↑ NO production
[101]
L. reuterii DSM 17938HT-29,
HT29-dx
↓ cell viability, ↑ mitochondrial ROS production[103]
L. reuteri NCIMB 701359SW-480, Caco-2apoptotic and antiproliferative activity[99]
L. reuteri PTCC 1655HT29-ShEanti-metastatic properties, apoptosis induction,
↓ MMP-9 and COX-2, ↑ TIMP-1
[133]
L. rhamnosus ATCC 7469Caco-2time- and dose-dependent cytotoxic activity,
intrinsic apoptotic pathway (↓ BCL2/
↑ caspase-3, -9, BAK, BAD, and BAX)
[132]
L. rhamnosus GGHCT 116anti-metastatic effects (↓ MMP-9/↑ ZO-1)[130]
HT-29anti-metastatic effects (↓ MMP2 and MMP9/
TIMP1 and TIMP2)
[131]
HT-29,
Caco-2
antiproliferative and anti-migration activity,
VEGF/MMPs signaling pathway down-regulation
[134]
HT-29,
HT29-dx
↓ cell viability, ↑ mitochondrial ROS production[103]
HCT 116,
Caco-2, HT-29
dose-dependent antiproliferative activity, mitotic arrest,
synergistic action with 5-FU
[136]
L. rhamnosus MD 14Caco-2,
HT-29
antigenotoxic and cytotoxic activity,
cell cycle arrest (G0/G1)
[105]
L. rhamnosus Y5HT-29time- and dose-dependent cytotoxic effect,
cell cycle arrest (G0/G1), ↓ CCND1, CCNE1 and ERBB2,
apoptosis induction (↑ CASP3, CASP9 and BAX/↓ BCL2)
[118]
L. salivarius RenHT-29antiproliferative activity, apoptosis induction,
AKT pathway inhibition, cyclin D1 and COX-2 suppression
[140]
Lactobacillus spp.HT-29, Caco-2cytotoxic activity, ↓ ERBB2 and ERBB3[104]
Lactobacillus spp.HT-29dose-dependent antiproliferative activity,
irregular morphology and cell condensation,
↑ caspase-3,-8 and Bax
[119]
Leuconostoc pseudomesenteroides strainsCaco-2, HT-29antioxidant and anticancer properties[112]
Pediococcus acidilactici TMAB26HT-29,
Caco-2
cytotoxic effects, anti-inflammatory properties in
LPS-pretreated cells (↓ TNF-α, IL-6/↑ IL-10)
[127]
Propionibacterium acidipropionici
Propionibacterium freudenreichii
HT-29,
Caco-2
cytotoxic activity, induction of apoptosis (MtMP loss/↑ ROS/↑ caspase-3/chromatin condensation)[123]
Propionibacterium freudenreichii
DSM 2027
HCT 116dose-dependent cytotoxic activity at 72 h[114]
Steptococcus salivarius CP163
Streptococcus salivarius CP208
HT-29antiproliferative activity, apoptosis induction
(↑ caspase-2, DNA fragmentation)
[126]
Yeasts
Kluyveromyces marxianus PCH397SW-480cytotoxic and antioxidant properties, cell cycle arrest[145]
Pichia kudriavzevii AS-12HT-29,
Caco-2
antiproliferative effect, apoptosis-related morphological
changes, apoptotis induction
(↑ BAD, CASP3, CASP8, CASP9 and Fas/↓ BCL2)
[144]
↑: increase or up-regulation, ↓: decrease or down-regulation. Colon cancer cell lines: COLO 205, HT-29, DLD-1, Caco-2, LoVo, SW-480, SW742, HCT-15, HCT 116, WiDr, SW-620, HT29-dx (doxorubicin-resistant HT-29 cells), HT-29/5-FUR (5-Fluorouracil-resistant HT-29 cells), HCT 116/5-FUR (5-Fluorouracil-resistant HCT 116 cells), HT29-ShE: E-cadherin shRNA engineered HT-29. MtMP: Mitochondrial membrane potential, cyt c: cytochrome c, PARP: Poly (ADP-ribose) polymerase, TNF-α: Tumor necrosis factor-α, TLR4: Toll-like receptor 4, mTOR: mammalian target of rapamycin, CASP: caspase gene, MMP-9: Matrix metalloproteinase-9, ZO-1: Zonula occludens-1 protein, VEGF: Vascular endothelial growth factor, MMPs: Matrix metalloproteinases, MMP: Matrix metalloproteinase gene, TIMP: Tissue inhibitor of metalloproteinase gene, NF-κB: Nuclear factor-κB, ROS: Reactive oxygen species, CSCs: Cancer stem cells, 5-FU: 5-Fluorouracil, CLDN-1: Claudin-1, SMCT1: Sodium-coupled monocarboxylate transporter 1, LDH: Lactate dehydrogenase, NO: Nitric oxide, COX-2: Cyclooxygenase-2, CCND1: cyclin D1 gene, CCNE1: cyclin E1 gene, ERBB2: ErbB-2 receptor tyrosine kinase 2 gene, LPS: Lipopolysaccharide, IL-6: Interleukin-6, IL-10: Interleukin-10, Fas: Fas cell surface death receptor gene.

3.2. Exopolysaccharides

Exopolysaccharides (EPSs) have gained scientific interest in recent years due to their diverse health-promoting properties [146], including the inhibition of pathogens’ adhesion to the intestinal epithelium, the enhancement of gut barrier integrity, and the regulation of mucosal immune responses [147]. Bacterial EPSs are extracellular, long-chain, high-molecular-weight polysaccharides, distinguished by their complex structures, which are strain-dependent and attributed to their distinct functions [92,96]. EPSs could be structurally divided into homopolysaccharides (HoPSs) containing a single type of monosaccharide and heteropolysaccharides (HePSs) composed of repeating units of numerous monosaccharides [25,148].
Their anticancer activity has been extensively studied [149,150,151], especially in the case of Lactobacillus-retrieved EPSs [152]. The majority of studies have designated the dose- or/and time-dependent cytotoxic effect of EPSs on CRC cell lines in vitro (Table 2) [153,154,155,156,157,158,159,160]. In several cases, EPSs trigger the intrinsic apoptotic pathway activation, indicated by the increased expression of Bax, caspase-3, and caspase-9 [161,162] and decreased levels of Bcl-2 [163,164,165]. EPSs from L. plantarum NCU116 activate the c-Jun dependent Fas/FasL-mediated apoptotic pathway through TLR2 in mouse intestinal epithelial cancer cells [166]. Additionally, apoptosis induction in EPS-treated CRC cells was confirmed by distinct apoptosis-related morphological features, such as cell shrinkage, nuclear fragmentation, and chromatin condensation [167,168,169,170]. EPSs from Lactobacillus acidophilus 10307 can inhibit the expression of genes involved in tumor angiogenesis and survival, including vascular endothelial growth factor (VEGF) and hypoxia-inducible factor-1α (HIF-1α), while they up-regulate antiangiogenic gene expression, such as tissue inhibitor of metalloproteinase-3 (TIMP-3) [171]. They can also enhance peroxisome proliferator-activated receptor-γ (PPAR-γ) expression, thus contributing to the suppression of CRC cellular growth [172]. Interestingly, cell-bound EPSs (cb-EPSs) isolated from L. acidophilus 606 were found to promote cell death via autophagy in HT-29 cells [173], while EPSs from Lactobacillus casei 01 can repair 4-nitroquinoline 1-oxide (4-NQO)-damaged IECs [174]. Probiotic yeast-derived EPSs can hinder the AKT-1, mammalian target of rapamycin (mTOR), and JAK-1 pathways to induce apoptosis in several CRC cell lines [175].

3.3. Bacteriocins

Bacteriocins encompass a heterogeneous group of extracellular, ribosomally synthesized antimicrobial peptides (AMPs) [176]. Harnessing their multifaceted functions, including the elimination of CRC-associated bacterial pathogens while avoiding disruption of the commensal microbiota [25,177], the regulation of the host’s immune responses contributing to gut homeostasis [37], and cancer-cell-specific targeting ability [178], bacteriocins possess unique features as potential anticancer agents [179,180]. Salivaricin was found to display potent antimicrobial activity against F. nucleatum in an ex vivo model of the human colon, thus reducing CRC development risk [181]. Interestingly, a recent in vitro study divulged that bacteriocins can migrate across epithelial monolayers [182], supporting their ability to disseminate across the GIT to exert their beneficial effects [183].
The selective cytotoxicity of bacteriocins against cancer cells is rather attributed to three dominant dissimilarities between cancer and normal cells. Firstly, the negatively charged plasma membrane of cancer cells, due to anionic compound overexpression, facilitates the electrostatic interactions of cationic bacteriocins with higher affinity to cancer than normal cells [178]. With regard to the fact that premalignant cells undergoing transformation into metastatic CRC forms are characterized by changes in phospholipid content [184], bacteriocins could be utilized as selective cytotoxic agents without affecting healthy cells. For instance, duramycin decreases CRC cells proliferation through binding to phosphatidylethanolamine (PE) [185]. Additionally, high membrane fluidity, a feature known to confer metastatic capability to malignant cells, enables bacteriocins to debilitate cancer cells’ membrane stability [186]. Lastly, the existence of abundant microvilli on the cancer cell surface allows a greater amount of bacteriocins to penetrate tumor cells [187].
Nisin is the prominent lantibiotic produced by Lactococcus lactis subsp. lactis, existing in four natural variants (A, Z, Q, and F), which differ in one or two amino acids [188]. Nisin variants A and Z have been extensively examined for their potential anticancer properties against CRC cells in vitro. Nisin A induces pore formation on the target cell membrane with subsequent loss of plasma membrane integrity [189] and calcium influx, thus causing cell death [190]. Nisin Z presents selective toxicity against colon cancer HT-29 cells [191]. However, nisin Z failed to affect Caco-2 cells’ membrane integrity, and this discrepancy has been attributed to the purity of the nisin samples as well as to the nisin variant employed in each study, pointing out the need to take both factors under consideration in future studies [192]. Regarding nisin’s mode of action, it induces the intrinsic apoptotic pathway in CRC cells as indicated by CASP3 and CASP9 gene up-regulation [193] as well as the increased apoptotic index (Bax/Bcl-2 ratio) in two different studies [194,195]. Additionally, nisin down-regulates the expression of metastasis-related genes such as MMPs, carcinoembryonic antigen (CEA), and carcinoembryonic cell adhesion molecule 6 (CEAM6) [196]. Following nisin treatment, decreased expression of the cyclin D1 gene in CRC cells was observed, thus unveiling its crucial role in CRC progression [195]. Information pertaining to nisin’s in vivo antitumor effects is only limited to xenograft mouse models with head and neck squamous cell carcinoma (HNSCC) [190,197].
A plethora of other bacteriocins exert antiproliferative activity against CRC cells, with a negligible effect against non-cancerous cells [198,199,200]. The in vitro cytotoxic effects of bacteriocins on CRC cells are shown in Figure 2 and summarized in Table 3. A recent meta-analysis study provided insight into the intervention of bacteriocins in various signaling cascades. For instance, they activate apoptosis via the regulation of the PI3K/AKT pathway, while they directly inhibit cyclooxygenase-2 (COX-2) expression and down-regulate the inflammatory NOD-like receptor family pyrin domain containing 3 (NLRP3) and nuclear factor-κB (NF-κB) pathways to diminish CRC-related inflammation [201]. Pediocin PA-1 induces cytotoxicity in HT-29 cells [202], possibly via interaction with TLRs based on 3D modeling approaches [203]. Plantaricin BM-1 triggers the caspase-dependent apoptotic pathway [204], while plantaricin P1053 increases the viability of normal CCD 841 cells via the activation of the epidermal growth factor receptor (EGFR) pathway [205]. Enterocin-treated cancer cells display apoptosis-like morphological changes [200,206].

3.4. Nonribosomal Lipopeptides

Nonribosomal lipopeptides derived from Bacillus subtilis are secondary bioactive molecules, synthesized by enzyme complexes, namely nonribosomal peptide synthetases (NRPS) [207]. These lipopeptides, mainly surfactin, iturin, and fengycin, exhibit significant cytotoxic activity against various CRC cell lines, thus contributing to elimination of cancer progression and metastasis [208] (Figure 2, Table 4). The underlying mechanisms involved in surfactin’s anticancer properties have recently been reviewed [209]. Surfactin can forcibly suppress CRC cell proliferation [210] via the induction of the caspase-dependent apoptotic pathway and cell cycle arrest at a certain concentration [211]. However, the major impediment to surfactin utilization as an anticancer agent is its hemolytic activity, leading to red blood cell (RBC) rupture and hemoglobin dissemination into the blood [211]. Iturin A can efficiently induce cytotoxic effects against CRC cells via multiple pathways, including initiation of paraptosis, apoptosis induction through the mitochondrial-mediated pathway, or activation of autophagy process [212]. Upon fengycin treatment in HT-29 cells, the expression of BAX, CASP3, and CASP6 genes increases, while decreased levels of Bcl-2 protein are observed, indicating that the mitochondrial pathway of apoptosis is triggered [213].

3.5. Other Bacterial Peptides

Miscellaneous bacterial peptides of probiotic origin have emerged as novel promising treatment strategies for CRC (Table 4) [214,215]. Enterococcal antiproliferative peptide (Entap) demonstrates cytotoxic activity against HT-29 cells via the induction of apoptosis and cell cycle arrest in the G1 phase [216]. Mixirins, cyclic acyl-peptides derived from the marine bacterium Bacillus sp., can inhibit the proliferation of human HCT 116 cells [217]. AMPs and bacterial-derived protein-based therapeutics for tackling increasing CRC morbidity rates have been well documented [218,219]. For instance, LHH1, a novel AMP produced by Lactobacillus casei HZ1, increases CRC cell membrane susceptibility, causing irreversible damages [220]. Two other peptides, namely m2163 and m2386, can penetrate the cell cytoplasm to induce apoptosis in SW-480 cancer cells [221]. Additionally, KL15, the conjugated form of m2163 and m2386 peptides resulting from in silico modifications in their sequences, not only possesses potent antimicrobial activity against pathogens but also induces necrotic cell death [222]. Probiotic-derived ferrichrome acts as a tumor-suppressive molecule via the c-Jun N-terminal kinase (JNK) signaling pathway against cancerous IECs to a greater extent than conventional chemotherapeutic drugs, including cisplatin and 5-FU [223]. The probiotic-derived P8 protein was found to eliminate metastasis [224] and suppress CRC growth via the inhibition of the Wnt signaling pathway [225], whereas mucin binding protein (MucBP) exhibits dose-dependent antiproliferative effects against HT-29 cells [226].
Table 4. In vitro effects of nonribosomal and other bacterial peptides on CRC cells.
Table 4. In vitro effects of nonribosomal and other bacterial peptides on CRC cells.
ClassBioactive CompoundCRC Cell LineEffect/Mode of ActionReference
Nonribosomal
peptides
Surfactin
(Bacillus subtilis)
LoVodose- and time-dependent cytotoxic activity,
caspase-dependent apoptosis induction,
ERK and PI3K/AKT pathways suppression,
cell cycle arrest (G0/G1)
[211]
HCT-15,
HT-29
dose-dependent cytotoxic activity[210]
Iturin A
(Bacillus subtilis)
Caco-2antitumor activity via multiple pathways:
1. intrinsic apoptotic pathway (↑ Bax, Bad/↓ Bcl-2),
2. paraptosis induction (ER dilatation, ↑ ROS
production, ↑ Ca2+ levels, mitochondrial dysfunction),
3. autophagy (↑ LC3-II/↓ LC3-I)
[212]
Fengycin
(Bacillus subtilis)
HCT-15,
HT-29
dose-dependent cytotoxic activity[210]
HT-29↓ cell proliferation, cell cycle arrest (G1),
apoptosis induction, ↑ ROS production,
↑ Bax and caspase-3, -6/↓ Bcl-2 and CDK4/cyclin D1
[213]
Other bacterial
peptides
Entap (Enterococcus sp.)HT-29apoptosis induction, cell cycle arrest (G1)[216]
Ferrichrome
(Lactobacillus casei ATCC 334)
Caco-2, SW-620, SK-CO-1tumor-suppressive effect, apoptosis induction via inhibition of JNK pathway[223]
KL15 peptide
(Lactobacillus casei ATCC 334)
SW-480,
Caco-2
antiproliferative effect, increased membrane
permeability, necrotic cell death
[222]
LHH1 peptide
(Lactobacillus casei HZ1)
HCT 116dose-dependent cytotoxic effect, apoptosis induction,
membrane damage
[220]
m2163 and m2386 peptides
(Lactobacillus casei ATCC 334)
SW-480↓ cell proliferation, extrinsic and intrinsic apoptosis induction, ↑ FasR and TRAILR1 expression (m2163)/
↑ FasR, TNFR1, and TRAILR1 (m2386)
[221]
Mixirins (Bacillus sp.)HCT 116↓ cell proliferation[217]
MucBP
(Lactobacillus casei)
HT-29dose-dependent antiproliferative effect[226]
Probiotic-derived P8 protein
(Lactobacillus rhamnosus KCTC 12202BP)
DLD-1antiproliferative and anti-migration activity,
cell cycle arrest (G2),
p53-p21-Cyclin B1/CDK1 pathway inhibition
[224]
Wnt pathway suppression (dysregulation of GSK3β transcription), cell cycle arrest[225]
↑: increase or up-regulation, ↓: decrease or down-regulation. Colon cancer cell lines: LoVo, HCT-15, HT-29, Caco-2, SW-620, SK-CO-1, SW-480, HCT 116, DLD-1. ER: Endoplasmic reticulum, ROS: Reactive oxygen species, LC3-II: Microtubule-associated protein 1A/1B-light chain 3-II, LC3-I: Microtubule-associated protein 1A/1B-light chain 3-I, CDK4: Cyclin-dependent kinase 4, JNK: c-jun N-terminal kinase, FasR: Fas receptor, TRAILR1: TRAIL receptor 1 gene, TNFR1: Tumor necrosis factor receptor-1, MucBP: Mucin binding protein, CDK1: Cyclin-dependent kinase 1, GSK3β: Glycogen synthase kinase β.

3.6. Short-Chain Fatty Acids

Short-chain fatty acids (SCFAs) constitute a group of metabolic products originating from the microbial fermentation of non-digestible carbohydrates [227]. The intestinal epithelium absorbs almost 95% of SCFAs synthesized by the gut microbiota [228]. SCFAs contribute to the maintenance of homeostasis, enhance gut barrier integrity, and participate in the energetic metabolism [229]. Upon their production, SCFAs are transported into the IECs via the SMCT1 [230]. Apart from the intestinal environment, a small amount of SCFAs that are not metabolized by colonocytes can reach systemic circulation and disseminate to distant tissues and organs [229], acting as signaling molecules with profound health benefits to the host [231].
SCFAs effects are mediated by two main pathways: the inhibition of histone deacetylases (HDACs) and the activation of cell surface G-protein-coupled receptors (GPRs), namely GPR41, GPR43, and GPR109A [232] (Figure 3). The administration of a mix of SCFAs (acetate, butyrate, and propionate) in a mouse model of colitis-associated CRC significantly reduced tumor incidence and attenuated colonic inflammation [233]. GPR43 deficiency was found to promote the progression of adenoma to adenocarcinoma in vivo [234]. SCFA administration suppressed intestinal inflammation and carcinogenesis in GPR43-deficient mice [235].
The pivotal role of SCFAs in the elimination of colorectal malignancy has already been reviewed [236,237,238]. A recent meta-analysis study demonstrated that lower fecal concentrations of the major SCFAs are correlated with increased CRC incidence [239]. The prominent mechanisms of SCFA action involve the down-regulation of genes related to DNA replication [240], the promotion of cell-cycle arrest and apoptosis [241], and the regulation of complex immune responses [242,243]. SCFAs provide resistance toward enteric bacterial pathogens associated with CRC development and progression [244].
Among the three aforementioned SCFAs, the anticancer effects of butyrate on CRC cells are most well-documented [245,246,247]. Butyrate can inhibit CRC proliferation via multiple mechanisms, such as the induction of the autophagy-mediated degradation of β-catenin [248], epigenetic reprogramming [249], the up-regulation of TLR4 expression, and the activation of the mitogen-activated protein kinase (MAPK) and NF-κΒ pathways [250]. Furthermore, it induces CRC cell ferroptosis via the CD44/Solute Carrier Family 7 Member 11 (SLC7A11) pathway and exhibits a synergistic therapeutic effect when combined with erastin, a ferroptosis-positive drug [251]. Interestingly, butyrate restores cytokine-induced barrier disruption, contributing to the maintenance of intestinal homeostasis [252].
Acetate was shown to reduce CRC proliferation and induce apoptosis, as indicated by several features such as loss of mitochondrial membrane potential, nuclear chromatin condensation, and ROS generation [253]. Upon acetate treatment, CRC cells exhibit apoptosis-related morphological features, while lysosomal membrane permeabilization with subsequent cathepsin D release in the cytosol takes place [254]. Another study suggests that acetate’s antiproliferative effect is a consequence of its impact on mitochondrial metabolism [255]. Propionate was found to down-regulate the protein arginine methyltransferase 1 (PRMT1) and regulate the mTOR pathway in HCT 116 cells [256]. Furthermore, it can suppress CRC tumorigenesis through promoting the proteasomal degradation of euchromatic histone-lysine N-methyltransferase 2 (EHMT2) through HECT domain E3 ubiquitin protein ligase 2 (HECTD2) up-regulation [257].

4. Challenges and Future Perspectives

Accumulating evidence has elucidated that gut microbiota dysbiosis contributes perilously to CRC occurrence and progression. Detrimental opportunistic pathogens can reconstruct the composition of colonic commensal bacteria, thus favoring the creation of a microenvironment susceptible to carcinogenesis. Conventional CRC treatments are characterized by the insufficient ability to specifically target cancer cells, while they are accompanied by chemoresistance development and numerous side effects in the host. Probiotic supplementation has been recommended as an effective complementary therapy for the elimination of gastrointestinal discomfort and the attenuation of gut dysbiosis in CRC patients undergoing chemotherapy [18]. However, despite probiotics’ accredited beneficial health effects, including epithelial colonization, the restoration of microbial diversity, and the detoxification of carcinogens, a few studies have highlighted impediments to their utilization [20]. In that context, miscellaneous bioactive compounds of probiotic origin with antiproliferative properties, such as bacteriocins, SCFAs, and EPSs could be exploited as conceivable anticancer agents [258]. Bacteriocins have been predominantly used as natural biopreservatives in the food industry, with nisin being the only bacteriocin licensed by the Food and Drug Administration (FDA) as a “GRAS” additive [259]. Apart from food applications, bacteriocins are now receiving increased attention as promising anticancer agents due to their specificity against cancer cells with limited or no effect on healthy cells [178]. Nevertheless, despite their advantageous properties of relevance to medical use, including biocompatibility, biodegradability, and lack of immunogenicity [180], they display some fundamental shortcomings, such as decreased bioavailability and susceptibility to proteolytic enzymes during exposure in the GIT, when orally administrated [259]. To overcome such limitations, multiple strategies varying from encapsulation technologies to bioengineering and semi-synthetic techniques could be recruited to improve their physicochemical characteristics and biological activity, while high-throughput sequencing may enhance the discovery of new bacteriocins [260]. Additionally, the costly production pertaining to bacteriocins’ efficient purification [261] as well as their high complexity due to extended posttranslational modifications [260] remain important hindrances towards their large-scale manufacturing. Interestingly, as progressive increases in multidrug-resistant infections have been declared a global health emergency [262], bacteriocins are considered as next-generation antibiotics [259], as well as feasible microbiome- editing tools [263] due to their potent antimicrobial activity. In fact, bacteriocins’ antimicrobial and toxic effects, as well as their biosafety in in vivo systems have been recently addressed [262,264]. They were found to act as immunostimulatory molecules contributing to the reduction of infection-associated parameters, including biochemical and histopathological biomarkers [262]. Furthermore, the anticancer effects of microcin E492 were tested in zebrafish xenografts, showing significant reduction in tumor growth [265], while nisin was found to act synergistically with the chemotherapeutic 5-FU in murine skin cancer models [266]. However, there is still inadequate research regarding their toxicity and therapeutic efficacy in vivo, a crucial prerequisite towards clinical trials. Indeed, in vitro experiments may not necessarily align with in vivo studies. Hence, further investigations should be conducted to decipher the delivery strategies, route of administration, and pharmacokinetic parameters of bacteriocins.
EPSs are complex, multifunctional carbohydrates excreted from probiotic bacteria, which have recently gained research attention with regard to pharmaceutical and therapeutic applications [150] due to their favorable health-promoting properties, such as immune system modulation, free radicals scavenging, and inhibition of cancer cell growth [267]. They are considered promising substitutes for synthetic anticancer drugs owing to their unique features, including biocompatibility, thermal stability, biodegradability, and nontoxic nature. The diverse physicochemical characteristics of EPSs (e.g., monosaccharide composition, branching degree, electric charge, molecular weight) are directly associated with their functionality [268]. Therefore, specific methodologies should be implemented to elucidate their chemical structure and provide an insight on their structure-dependent functional benefits [267,268,269,270]. Additionally, the high production costs and low polysaccharide yields as well as the time-consuming processes required for EPS purification currently restrict their commercialization [269]. Regarding this aspect, genetic and metabolic engineering could facilitate EPS production yield [268]. Detailed studies should also be performed to evaluate their safety via the application of targeted high-throughput screening strategies [271]. Only limited research has been performed in vivo, supporting probiotic EPSs’ ability to alleviate intestinal inflammation via gut microbiota modulation [272,273,274], contribute to the maintenance of the epithelial barrier [275], exert anticancer effects [276,277], and attenuate 5-FU-induced toxicity in animal models [278]. Nevertheless, there is still a lack of clinical evidence to proceed into human administration.
SCFAs represent the largest group of bioactive compounds residing in the intestinal lumen with a profound contribution to gut immunity stimulation. SCFAs also act as signaling molecules upon their dissemination into the bloodstream, thus promoting health benefits for the host [37]. Given their protective anti-inflammatory and antioxidant features [279], SCFAs have been found to eliminate the severity of conventional anticancer drugs’ GIT toxicities [280]. They can also exert anticancer properties via the regulation of immune response, a reduction in HDAC activity [281], and the repairment of intestinal microecology [282], thus promoting chemosensitivity and cell growth inhibition, or they can act as antitumor adjuvant drugs [283]. Patients responding to chemotherapy, immunotherapy, and radiotherapy treatments were found to exhibit a higher abundance of SCFA-producing microbes and higher levels of fecal and plasma SCFAs. Furthermore, recent studies suggest that SCFA-based interventional strategies could be implemented to promote cancer treatment efficacy and decrease the adverse side effects commonly caused by chemotherapeutics [284,285]. However, the majority of them focus on defining the correlation between SCFAs levels and therapeutic outcomes rather than assessing SCFAs’ utilization for medical purposes. Inconsistent results derived from in vitro and in vivo studies may be attributed to different methodological approaches as well as interindividual variations in SCFA production [284]. These limitations should be taken under consideration prior to experimental designs. Hence, the lack of pre-clinical and clinical evidence still impedes the evaluation of SCFAs’ local and systemic effects and the determination of the favorable route of administration to the host. In this context, more elaborative investigations are required to extrapolate conclusions regarding the multifaceted interactions occurring in the gut and configure the criteria of SCFA utilization based on the distinctiveness of each individual.

Author Contributions

Conceptualization, M.T.; writing—original draft preparation, C.T.; writing—review and editing, M.T. and C.T.; figure editing, C.T.; supervision, M.T. All authors have read and agreed to the published version of the manuscript.

Funding

This study was partially financially supported by the Graduate Program “Applications of Biology” of the School of Biology, Aristotle University of Thessaloniki.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Dekker, E.; Tanis, P.J.; Vleugels, J.L.A.; Kasi, P.M.; Wallace, M.B. Colorectal cancer. Lancet 2019, 394, 1467–1480. [Google Scholar] [CrossRef]
  2. Hossain, M.S.; Karuniawati, H.; Jairoun, A.A.; Urbi, Z.; Ooi, D.J.; John, A.; Lim, Y.C.; Kibria, K.M.K.; Mohiuddin, A.M.; Ming, L.C.; et al. Colorectal Cancer: A Review of Carcinogenesis, Global Epidemiology, Current Challenges, Risk Factors, Preventive and Treatment Strategies. Cancers 2022, 14, 1732. [Google Scholar] [CrossRef]
  3. Huang, Z.; Yang, M. Molecular Network of Colorectal Cancer and Current Therapeutic Options. Front. Oncol. 2022, 12, 852927. [Google Scholar] [CrossRef]
  4. Katsaounou, K.; Nicolaou, E.; Vogazianos, P.; Brown, C.; Stavrou, M.; Teloni, S.; Hatzis, P.; Agapiou, A.; Fragkou, E.; Tsiaoussis, G.; et al. Colon Cancer: From Epidemiology to Prevention. Metabolites 2022, 12, 499. [Google Scholar] [CrossRef]
  5. Pandey, H.; Tang, D.W.T.; Wong, S.H.; Lal, D. Gut Microbiota in Colorectal Cancer: Biological Role and Therapeutic Opportunities. Cancers 2023, 15, 866. [Google Scholar] [CrossRef]
  6. Fearon, E.R.; Vogelstein, B. A genetic model for colorectal tumorigenesis. Cell 1990, 61, 759–767. [Google Scholar] [CrossRef]
  7. Schmitt, M.; Greten, F.R. The inflammatory pathogenesis of colorectal cancer. Nat. Rev. Immunol. 2021, 21, 653–667. [Google Scholar] [CrossRef]
  8. Mármol, I.; Sánchez-de-Diego, C.; Pradilla Dieste, A.; Cerrada, E.; Rodriguez Yoldi, M.J. Colorectal Carcinoma: A General Overview and Future Perspectives in Colorectal Cancer. Int. J. Mol. Sci. 2017, 18, 197. [Google Scholar] [CrossRef] [Green Version]
  9. Vasan, N.; Baselga, J.; Hyman, D.M. A view on drug resistance in cancer. Nature 2019, 575, 299–309. [Google Scholar] [CrossRef] [Green Version]
  10. Hanahan, D.; Weinberg, R.A. Hallmarks of cancer: The next generation. Cell 2011, 144, 646–674. [Google Scholar] [CrossRef] [Green Version]
  11. Hanahan, D. Hallmarks of Cancer: New Dimensions. Cancer Discov. 2022, 12, 31–46. [Google Scholar] [CrossRef]
  12. Chrysostomou, D.; Roberts, L.A.; Marchesi, J.R.; Kinross, J.M. Gut Microbiota Modulation of Efficacy and Toxicity of Cancer Chemotherapy and Immunotherapy. Gastroenterology 2023, 164, 198–213. [Google Scholar] [CrossRef]
  13. Vitali, F.; Tortora, K.; Di Paola, M.; Bartolucci, G.; Menicatti, M.; De Filippo, C.; Caderni, G. Intestinal microbiota profiles in a genetic model of colon tumorigenesis correlates with colon cancer biomarkers. Sci. Rep. 2022, 12, 1432. [Google Scholar] [CrossRef]
  14. Sadrekarimi, H.; Gardanova, Z.R.; Bakhshesh, M.; Ebrahimzadeh, F.; Yaseri, A.F.; Thangavelu, L.; Hasanpoor, Z.; Zadeh, F.A.; Kahrizi, M.S. Emerging role of human microbiome in cancer development and response to therapy: Special focus on intestinal microflora. J. Transl. Med. 2022, 20, 301. [Google Scholar] [CrossRef]
  15. O’Keefe, S.J. Diet, microorganisms and their metabolites, and colon cancer. Nat. Rev. Gastroenterol. Hepatol. 2016, 13, 691–706. [Google Scholar] [CrossRef]
  16. Mueller, A.-L.; Brockmueller, A.; Fahimi, N.; Ghotbi, T.; Hashemi, S.; Sadri, S.; Khorshidi, N.; Kunnumakkara, A.B.; Shakibaei, M. Bacteria-Mediated Modulatory Strategies for Colorectal Cancer Treatment. Biomedicines 2022, 10, 832. [Google Scholar] [CrossRef]
  17. Liu, Y.; Lau, H.C.; Yu, J. Microbial metabolites in colorectal tumorigenesis and cancer therapy. Gut Microbes 2023, 15, 2203968. [Google Scholar] [CrossRef]
  18. Huang, F.; Li, S.; Chen, W.; Han, Y.; Yao, Y.; Yang, L.; Li, Q.; Xiao, Q.; Wei, J.; Liu, Z.; et al. Postoperative Probiotics Administration Attenuates Gastrointestinal Complications and Gut Microbiota Dysbiosis Caused by Chemotherapy in Colorectal Cancer Patients. Nutrients 2023, 15, 356. [Google Scholar] [CrossRef]
  19. Garbacz, K. Anticancer activity of lactic acid bacteria. Semin. Cancer Biol. 2022, 86, 356–366. [Google Scholar] [CrossRef]
  20. Lu, K.; Dong, S.; Wu, X.; Jin, R.; Chen, H. Probiotics in Cancer. Front. Oncol. 2021, 11, 638148. [Google Scholar] [CrossRef]
  21. Dicks, L.M.T.; Vermeulen, W. Do Bacteria Provide an Alternative to Cancer Treatment and What Role Does Lactic Acid Bacteria Play? Microorganisms 2022, 10, 1733. [Google Scholar] [CrossRef]
  22. Kyrila, G.; Katsoulas, A.; Schoretsaniti, V.; Rigopoulos, A.; Rizou, E.; Doulgeridou, S.; Sarli, V.; Samanidou, V.; Touraki, M. Bisphenol A removal and degradation pathways in microorganisms with probiotic properties. J. Hazard. Mater. 2021, 413, 125363. [Google Scholar] [CrossRef]
  23. Mavromatis, P.; Stampouli, K.; Vliora, A.; Mayilyan, A.; Samanidou, V.; Touraki, M. Development of an HPLC-DAD Method for the Extraction and Quantification of 5-Fluorouracil, Uracil, and 5-Fluorodeoxyuridin Monophosphate in Cells and Culture Media of Lactococcus lactis. Separations 2022, 9, 376. [Google Scholar] [CrossRef]
  24. Purdel, C.; Ungurianu, A.; Adam-Dima, I.; Margină, D. Exploring the potential impact of probiotic use on drug metabolism and efficacy. Biomed. Pharmacother. 2023, 161, 114468. [Google Scholar] [CrossRef]
  25. Song, D.; Wang, X.; Ma, Y.; Liu, N.N.; Wang, H. Beneficial insights into postbiotics against colorectal cancer. Front. Nutr. 2023, 10, 1111872. [Google Scholar] [CrossRef]
  26. Szydłowska, A.; Sionek, B. Probiotics and Postbiotics as the Functional Food Components Affecting the Immune Response. Microorganisms 2023, 11, 104. [Google Scholar] [CrossRef]
  27. Cheng, W.Y.; Wu, C.-Y.; Yu, J. The role of gut microbiota in cancer treatment: Friend or foe? Gut 2020, 69, 1867–1876. [Google Scholar] [CrossRef]
  28. Rinninella, E.; Raoul, P.; Cintoni, M.; Franceschi, F.; Miggiano, G.A.D.; Gasbarrini, A.; Mele, M.C. What is the Healthy Gut Microbiota Composition? A Changing Ecosystem across Age, Environment, Diet, and Diseases. Microorganisms 2019, 7, 14. [Google Scholar] [CrossRef] [Green Version]
  29. Karpiński, T.M.; Ożarowski, M.; Stasiewicz, M. Carcinogenic microbiota and its role in colorectal cancer development. Semin. Cancer Biol. 2022, 86, 420–430. [Google Scholar] [CrossRef]
  30. Yao, Y.; Cai, X.; Ye, Y.; Wang, F.; Chen, F.; Zheng, C. The Role of Microbiota in Infant Health: From Early Life to Adulthood. Front. Immunol. 2021, 12, 708472. [Google Scholar] [CrossRef]
  31. Thriene, K.; Michels, K.B. Human Gut Microbiota Plasticity throughout the Life Course. Int. J. Environ. Res. Public Health 2023, 20, 1463. [Google Scholar] [CrossRef]
  32. Fan, Y.; Pedersen, O. Gut microbiota in human metabolic health and disease. Nat. Rev. Microbiol. 2021, 19, 55–71. [Google Scholar] [CrossRef]
  33. Patangia, D.V.; Anthony Ryan, C.; Dempsey, E.; Paul Ross, R.; Stanton, C. Impact of antibiotics on the human microbiome and consequences for host health. Microbiologyopen 2022, 11, e1260. [Google Scholar] [CrossRef]
  34. Brooks, A.W.; Priya, S.; Blekhman, R.; Bordenstein, S.R. Gut microbiota diversity across ethnicities in the United States. PLoS Biol. 2018, 16, e2006842. [Google Scholar] [CrossRef] [Green Version]
  35. Jandhyala, S.M.; Talukdar, R.; Subramanyam, C.; Vuyyuru, H.; Sasikala, M.; Nageshwar Reddy, D. Role of the normal gut microbiota. World J. Gastroenterol. 2015, 21, 8787–8803. [Google Scholar] [CrossRef]
  36. Mukherji, R.; Weinberg, B.A. The gut microbiome and potential implications for early-onset colorectal cancer. Future Med. 2020, 9. [Google Scholar] [CrossRef]
  37. Thoda, C.; Touraki, M. Immunomodulatory Properties of Probiotics and Their Derived Bioactive Compounds. Appl. Sci. 2023, 13, 4726. [Google Scholar] [CrossRef]
  38. Ghosh, S.; Whitley, C.S.; Haribabu, B.; Jala, V.R. Regulation of Intestinal Barrier Function by Microbial Metabolites. Cell Mol. Gastroenterol. Hepatol. 2021, 11, 1463–1482. [Google Scholar] [CrossRef]
  39. Valdes, A.M.; Walter, J.; Segal, E.; Spector, T.D. Role of the gut microbiota in nutrition and health. BMJ 2018, 361, k2179. [Google Scholar] [CrossRef] [Green Version]
  40. Peluzio, M.D.C.G.; Martinez, J.A.; Milagro, F.I. Postbiotics: Metabolites and mechanisms involved in microbiota-host interactions. Trends Food Sci. Technol. 2021, 108, 11–26. [Google Scholar] [CrossRef]
  41. Torres-Maravilla, E.; Boucard, A.-S.; Mohseni, A.H.; Taghinezhad-S, S.; Cortes-Perez, N.G.; Bermúdez-Humarán, L.G. Role of Gut Microbiota and Probiotics in Colorectal Cancer: Onset and Progression. Microorganisms 2021, 9, 1021. [Google Scholar] [CrossRef]
  42. Asseri, A.H.; Bakhsh, T.; Abuzahrah, S.S.; Ali, S.; Rather, I.A. The gut dysbiosis-cancer axis: Illuminating novel insights and implications for clinical practice. Front. Pharmacol. 2023, 14, 1208044. [Google Scholar] [CrossRef]
  43. Bardelčíková, A.; Šoltys, J.; Mojžiš, J. Oxidative Stress, Inflammation and Colorectal Cancer: An Overview. Antioxidants 2023, 12, 901. [Google Scholar] [CrossRef]
  44. Wu, W.; Ouyang, Y.; Zheng, P.; Xu, X.; He, C.; Xie, C.; Hong, J.; Lu, N.; Zhu, Y.; Li, N. Research trends on the relationship between gut microbiota and colorectal cancer: A bibliometric analysis. Front. Cell. Infect. Microbiol. 2023, 12, 1027448. [Google Scholar] [CrossRef]
  45. Schwabe, R.F.; Jobin, C. The microbiome and cancer. Nat. Rev. Cancer 2013, 13, 800–812. [Google Scholar] [CrossRef] [Green Version]
  46. Zou, S.; Fang, L.; Lee, M.H. Dysbiosis of gut microbiota in promoting the development of colorectal cancer. Gastroenterol. Rep. 2018, 6, 1–12. [Google Scholar] [CrossRef] [Green Version]
  47. Ahmad Kendong, S.M.; Raja Ali, R.A.; Nawawi, K.N.M.; Ahmad, H.F.; Mokhtar, N.M. Gut Dysbiosis and Intestinal Barrier Dysfunction: Potential Explanation for Early-Onset Colorectal Cancer. Front. Cell. Infect. Microbiol. 2021, 11, 744606. [Google Scholar] [CrossRef]
  48. Fan, X.; Jin, Y.; Chen, G.; Ma, X.; Zhang, L. Gut Microbiota Dysbiosis Drives the Development of Colorectal Cancer. Digestion 2021, 102, 508–515. [Google Scholar] [CrossRef]
  49. Yang, Y.; Du, L.; Shi, D.; Kong, C.; Liu, J.; Li, X.; Ma, Y. Dysbiosis of human gut microbiome in young-onset colorectal cancer. Nat. Commun. 2021, 12, 6757. [Google Scholar] [CrossRef]
  50. Xu, S.; Yin, W.; Zhang, Y.; Lv, Q.; Yang, Y.; He, J. Foes or Friends? Bacteria Enriched in the Tumor Microenvironment of Colorectal Cancer. Cancers 2020, 12, 372. [Google Scholar] [CrossRef] [Green Version]
  51. Parida, S.; Sharma, D. The Microbiome and Cancer: Creating Friendly Neighborhoods and Removing the Foes Within. Cancer Res. 2021, 81, 790–800. [Google Scholar] [CrossRef]
  52. Wang, N.; Fang, J.-Y. Fusobacterium nucleatum, a key pathogenic factor and microbial biomarker for colorectal cancer. Trends Microbiol. 2023, 31, 159–172. [Google Scholar] [CrossRef]
  53. Haghi, F.; Goli, E.; Mirzaei, B.; Zeighami, H. The association between fecal enterotoxigenic B. fragilis with colorectal cancer. BMC Cancer 2019, 19, 879. [Google Scholar] [CrossRef] [Green Version]
  54. Scott, N.; Whittle, E.; Jeraldo, P.; Chia, N. A systemic review of the role of enterotoxic Bacteroides fragilis in colorectal cancer. Neoplasia 2022, 29, 100797. [Google Scholar] [CrossRef]
  55. Wang, Y.; Fu, K. Genotoxins: The Mechanistic Links between Escherichia coli and Colorectal Cancer. Cancers 2023, 15, 1152. [Google Scholar] [CrossRef]
  56. Abdulamir, A.S.; Hafidh, R.R.; Abu Bakar, F. The association of Streptococcus bovis/gallolyticus with colorectal tumors: The nature and the underlying mechanisms of its etiological role. J. Exp. Clin. Cancer Res. 2011, 30, 11. [Google Scholar] [CrossRef] [Green Version]
  57. Butt, J.; Epplein, M. Helicobacter pylori and colorectal cancer-A bacterium going abroad? PLoS Pathog. 2019, 15, e1007861. [Google Scholar] [CrossRef] [Green Version]
  58. Zha, L.; Garrett, S.; Sun, J. Salmonella Infection in Chronic Inflammation and Gastrointestinal Cancer. Diseases 2019, 7, 28. [Google Scholar] [CrossRef] [Green Version]
  59. 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]
  60. Umar, S. Citrobacter Infection and Wnt signaling. Curr. Colorectal Cancer Rep. 2012, 8. [Google Scholar] [CrossRef] [Green Version]
  61. Cheng, Y.; Ling, Z.; Li, L. The Intestinal Microbiota and Colorectal Cancer. Front. Immunol. 2020, 11, 615056. [Google Scholar] [CrossRef]
  62. Rebersek, M. Gut microbiome and its role in colorectal cancer. BMC Cancer 2021, 21, 1325. [Google Scholar] [CrossRef]
  63. Ren, L.; Ye, J.; Zhao, B.; Sun, J.; Cao, P.; Yang, Y. The Role of Intestinal Microbiota in Colorectal Cancer. Front. Pharmacol. 2021, 12, 674807. [Google Scholar] [CrossRef]
  64. Khan, A.A.; Sirsat, A.T.; Singh, H.; Cash, P. Microbiota and cancer: Current understanding and mechanistic implications. Clin. Transl. Oncol. 2022, 24, 193–202. [Google Scholar] [CrossRef]
  65. Kim, J.; Lee, H.K. Potential Role of the Gut Microbiome In Colorectal Cancer Progression. Front. Immunol. 2022, 12, 807648. [Google Scholar] [CrossRef]
  66. Koyande, N.; Gangopadhyay, M.; Thatikonda, S.; Rengan, A.K. The role of gut microbiota in the development of colorectal cancer: A review. Int. J. Colorectal Dis. 2022, 37, 1509–1523. [Google Scholar] [CrossRef]
  67. Tjalsma, H.; Boleij, A.; Marchesi, J.R.; Dutilh, B.E. A bacterial driver-passenger model for colorectal cancer: Beyond the usual suspects. Nat. Rev. Microbiol. 2012, 10, 575–582. [Google Scholar] [CrossRef]
  68. Avril, M.; DePaolo, R.W. "Driver-passenger" bacteria and their metabolites in the pathogenesis of colorectal cancer. Gut Microbes 2021, 13, 1941710. [Google Scholar] [CrossRef]
  69. Wong, S.H.; Zhao, L.; Zhang, X.; Nakatsu, G.; Han, J.; Xu, W.; Xiao, X.; Kwong, T.N.Y.; Tsoi, H.; Wu, W.K.K.; et al. Gavage of fecal samples from patients with colorectal cancer promotes intestinal carcinogenesis in germ-free and conventional mice. Gastroenterology 2017, 153, 1621–1633. [Google Scholar] [CrossRef] [Green Version]
  70. Li, L.; Li, X.; Zhong, W.; Yang, M.; Xu, M.; Sun, Y.; Ma, J.; Liu, T.; Song, X.; Dong, W.; et al. Gut microbiota from colorectal cancer patients enhances the progression of intestinal adenoma in Apcmin/+ mice. EBioMedicine 2019, 48, 301–315. [Google Scholar] [CrossRef] [Green Version]
  71. Flemer, B.; Lynch, D.B.; Brown, J.M.; Jeffery, I.B.; Ryan, F.J.; Claesson, M.J.; O’Riordain, M.; Shanahan, F.; O’Toole, P.W. Tumour- associated and non-tumour-associated microbiota in colorectal cancer. Gut 2017, 66, 633–643. [Google Scholar] [CrossRef] [PubMed]
  72. Richard, M.L.; Liguori, G.; Lamas, B.; Brandi, G.; da Costa, G.; Hoffmann, T.W.; Pierluigi Di Simone, M.; Calabrese, C.; Poggioli, G.; Langella, P.; et al. Mucosa-associated microbiota dysbiosis in colitis associated cancer. Gut Microbes 2018, 9, 131–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. 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]
  74. Sepich-Poore, G.D.; Zitvogel, L.; Straussman, R.; Hasty, J.; Wargo, J.A.; Knight, R. The microbiome and human cancer. Science 2021, 371, eabc4552. [Google Scholar] [CrossRef]
  75. Li, J.; Zhu, Y.; Yang, L.; Wang, Z. Effect of gut microbiota in the colorectal cancer and potential target therapy. Discov. Oncol. 2022, 13, 51. [Google Scholar] [CrossRef]
  76. 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]
  77. Kim, S.H.; Lim, Y.J. The role of microbiome in colorectal carcinogenesis and its clinical potential as a target for cancer treatment. Intest. Res. 2022, 20, 31–42. [Google Scholar] [CrossRef]
  78. Siddiqui, R.; Boghossian, A.; Alharbi, A.M.; Alfahemi, H.; Khan, N.A. The Pivotal Role of the Gut Microbiome in Colorectal Cancer. Biology 2022, 11, 1642. [Google Scholar] [CrossRef]
  79. Wang, Y.; Li, H. Gut microbiota modulation: A tool for the management of colorectal cancer. J. Transl. Med. 2022, 20, 178. [Google Scholar] [CrossRef]
  80. Dougherty, M.W.; Jobin, C. Intestinal bacteria and colorectal cancer: Etiology and treatment. Gut Microbes. 2023, 15, 2185028. [Google Scholar] [CrossRef]
  81. 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]
  82. Sánchez-Alcoholado, L.; Ramos-Molina, B.; Otero, A.; Laborda-Illanes, A.; Ordóñez, R.; Medina, J.A.; Gómez-Millán, J.; Queipo-Ortuño, M.I. The Role of the Gut Microbiome in Colorectal Cancer Development and Therapy Response. Cancers 2020, 12, 1406. [Google Scholar] [CrossRef]
  83. Silva, M.; Brunner, V.; Tschurtschenthaler, M. Microbiota and Colorectal Cancer: From Gut to Bedside. Front. Pharmacol. 2021, 12, 760280. [Google Scholar] [CrossRef]
  84. Rahman, M.M.; Islam, M.R.; Shohag, S.; Ahasan, M.T.; Sarkar, N.; Khan, H.; Hasan, A.M.; Cavalu, S.; Rauf, A. Microbiome in cancer: Role in carcinogenesis and impact in therapeutic strategies. Biomed. Pharmacother. 2022, 149, 112898. [Google Scholar] [CrossRef] [PubMed]
  85. Wong, C.C.; Yu, J. Gut microbiota in colorectal cancer development and therapy. Nat. Rev. Clin. Oncol. 2023, 20, 429–452. [Google Scholar] [CrossRef] [PubMed]
  86. Feng, J.; Gong, Z.; Sun, Z.; Li, J.; Xu, N.; Thorne, R.F.; Zhang, X.D.; Liu, X.; Liu, G. Microbiome and metabolic features of tissues and feces reveal diagnostic biomarkers for colorectal cancer. Front. Microbiol. 2023, 14, 1034325. [Google Scholar] [CrossRef]
  87. Zwezerijnen-Jiwa, F.H.; Sivov, H.; Paizs, P.; Zafeiropoulou, K.; Kinross, J. A systematic review of microbiome-derived biomarkers for early colorectal cancer detection. Neoplasia 2023, 36, 100868. [Google Scholar] [CrossRef]
  88. Saber, S.; Yahya, G.; Gobba, N.A.; Sharaf, H.; Alshaman, R.; Alattar, A.; Amin, N.A.; El-Shedody, R.; Aboutouk, F.H.; Abd El-Galeel, Y.; et al. The Supportive Role of NSC328382, a P2X7R Antagonist, in Enhancing the Inhibitory Effect of CRID3 on NLRP3 Inflammasome Activation in Rats with Dextran Sodium Sulfate-Induced Colitis. J. Inflamm. Res. 2021, 14, 3443–3463. [Google Scholar] [CrossRef]
  89. Saber, S.; Abd El-Fattah, E.E.; Yahya, G.; Gobba, N.A.; Maghmomeh, A.O.; Khodir, A.E.; Mourad, A.A.E.; Saad, A.S.; Mohammed, H.G.; Nouh, N.A.; et al. A Novel Combination Therapy Using Rosuvastatin and Lactobacillus Combats Dextran Sodium Sulfate-Induced Colitis in High-Fat Diet-Fed Rats by Targeting the TXNIP/NLRP3 Interaction and Influencing Gut Microbiome Composition. Pharmaceuticals 2021, 14, 341. [Google Scholar] [CrossRef]
  90. Džidić-Krivić, A.; Kusturica, J.; Sher, E.K.; Selak, N.; Osmančević, N.; Karahmet Farhat, E.; Sher, F. Effects of intestinal flora on pharmacokinetics and pharmacodynamics of drugs. Drug. Metab. Rev. 2023, 55, 126–139. [Google Scholar] [CrossRef]
  91. LaCourse, K.D.; Zepeda-Rivera, M.; Kempchinsky, A.G.; Baryiames, A.; Minot, S.S.; Johnston, C.D.; Bullman, S. The cancer chemotherapeutic 5-fluorouracil is a potent Fusobacterium nucleatum inhibitor and its activity is modified by intratumoral microbiota. Cell. Rep. 2022, 41, 111625. [Google Scholar] [CrossRef] [PubMed]
  92. Thorakkattu, P.; Khanashyam, A.C.; Shah, K.; Babu, K.S.; Mundanat, A.S.; Deliephan, A.; Deokar, G.S.; Santivarangkna, C.; Nirmal, N.P. Postbiotics: Current Trends in Food and Pharmaceutical Industry. Foods 2022, 11, 3094. [Google Scholar] [CrossRef]
  93. Mani-López, E.; Arrioja-Bretón, D.; López-Malo, A. The impacts of antimicrobial and antifungal activity of cell-free supernatants from lactic acid bacteria in vitro and foods. Compr. Rev. Food Sci. Food Saf. 2022, 21, 604–641. [Google Scholar] [CrossRef] [PubMed]
  94. Żółkiewicz, J.; Marzec, A.; Ruszczyński, M.; Feleszko, W. Postbiotics—A Step Beyond Pre- and Probiotics. Nutrients 2020, 12, 2189. [Google Scholar] [CrossRef]
  95. Scarpellini, E.; Rinninella, E.; Basilico, M.; Colomier, E.; Rasetti, C.; Larussa, T.; Santori, P.; Abenavoli, L. From Pre- and Probiotics to Post-Biotics: A Narrative Review. Int. J. Environ. Res. Public Health 2022, 19, 37. [Google Scholar] [CrossRef]
  96. Nataraj, B.H.; Ali, S.A.; Behare, P.V.; Yadav, H. Postbiotics-parabiotics: The new horizons in microbial biotherapy and functional foods. Microb. Cell. Fact. 2020, 19, 168. [Google Scholar] [CrossRef]
  97. Sadeghi-Aliabadi, H.; Mohammadi, F.; Fazeli, H.; Mirlohi, M. Effects of Lactobacillus plantarum A7 with probiotic potential on colon cancer and normal cells proliferation in comparison with a commercial strain. Iran. J. Basic. Med. Sci. 2014, 17, 815–819. [Google Scholar] [CrossRef]
  98. Kahouli, I.; Malhotra, M.; Alaoui-Jamali, M.A.; Prakash, S. In-Vitro Characterization of the Anti-Cancer Activity of the Probiotic Bacterium Lactobacillus Fermentum NCIMB 5221 and Potential against Colorectal Cancer Cells. Cancer Sci. Ther. 2015, 7, 7. [Google Scholar] [CrossRef]
  99. Kahouli, I.; Handiri, N.R.; Malhotra, M.; Riahi, A.; Alaoui-Jamali, M.; Prakash, S. Characterization of L. Reuteri NCIMB 701359 probiotic features for potential use as a colorectal cancer biotherapeutic by identifying fatty acid profile and anti-proliferative action against colorectal cancer cells. Drug Des. 2016, 5, 1–11. [Google Scholar] [CrossRef]
  100. Tiptiri-Kourpeti, A.; Spyridopoulou, K.; Santarmaki, V.; Aindelis, G.; Tompoulidou, E.; Lamprianidou, E.E.; Saxami, G.; Ypsilantis, P.; Lampri, E.S.; Simopoulos, C.; et al. Lactobacillus casei Exerts Anti-Proliferative Effects Accompanied by Apoptotic Cell Death and Up-Regulation of TRAIL in Colon Carcinoma Cells. PLoS ONE 2016, 11, e0147960. [Google Scholar] [CrossRef]
  101. Chen, Z.-Y.; Hsieh, Y.-M.; Huang, C.-C.; Tsai, C.-C. Inhibitory Effects of Probiotic Lactobacillus on the Growth of Human Colonic Carcinoma Cell Line HT-29. Molecules 2017, 22, 107. [Google Scholar] [CrossRef] [Green Version]
  102. Chuah, L.O.; Foo, H.L.; Loh, T.C.; Mohammed Alitheen, N.B.; Yeap, S.K.; Abdul Mutalib, N.E.; Abdul Rahim, R.; Yusoff, K. Postbiotic metabolites produced by Lactobacillus plantarum strains exert selective cytotoxicity effects on cancer cells. BMC Complement. Altern. Med. 2019, 19, 114. [Google Scholar] [CrossRef] [Green Version]
  103. Doublier, S.; Cirrincione, S.; Scardaci, R.; Botta, C.; Lamberti, C.; Giuseppe, F.D.; Angelucci, S.; Rantsiou, K.; Cocolin, L.; Pessione, E. Putative probiotics decrease cell viability and enhance chemotherapy effectiveness in human cancer cells: Role of butyrate and secreted proteins. Microbiol. Res. 2022, 260, 127012. [Google Scholar] [CrossRef]
  104. Faghfoori, Z.; Pourghassem Gargari, B.; Saber, A.; Seyyedi, M.; Fazelian, S.; Khosroushahi, A.Y. Prophylactic effects of secretion metabolites of dairy lactobacilli through downregulation of ErbB-2 and ErbB-3 genes on colon cancer cells. Eur. J. Cancer Prev. 2020, 29, 201–209. [Google Scholar] [CrossRef]
  105. Sharma, M.; Chandel, D.; Shukla, G. Antigenotoxicity and Cytotoxic Potentials of Metabiotics Extracted from Isolated Probiotic, Lactobacillus rhamnosus MD 14 on Caco-2 and HT-29 Human Colon Cancer Cells. Nutr. Cancer 2020, 72, 110–119. [Google Scholar] [CrossRef]
  106. Adiyoga, R.; Arief, I.I.; Budiman, C.; Abidin, Z. In vitro anticancer potentials of Lactobacillus plantarum IIA-1A5 and Lactobacillus acidophilus IIA-2B4 extracts against WiDr human colon cancer cell line. Food Sci. Technol. 2022, 42, e87221. [Google Scholar] [CrossRef]
  107. Elham, N.; Naheed, M.; Elahe, M.; Hossein, M.M.; Majid, T. Selective Cytotoxic effect of Probiotic, Paraprobiotic and Postbiotics of L. casei strains against Colorectal Cancer Cells: In vitro studies. Braz. J. Pharm. Sci. 2022, 58, e19400. [Google Scholar] [CrossRef]
  108. Ma, E.L.; Choi, Y.J.; Choi, J.; Pothoulakis, C.; Rhee, S.H.; Im, E. The anticancer effect of probiotic Bacillus polyfermenticus on human colon cancer cells is mediated through ErbB2 and ErbB3 inhibition. Int. J. Cancer 2010, 127, 780–790. [Google Scholar] [CrossRef] [Green Version]
  109. Lee, N.-K.; Son, S.-H.; Jeon, E.B.; Jung, G.H.; Lee, J.-Y.; Paik, H.-D. The prophylactic effect of probiotic Bacillus polyfermenticus KU3 against cancer cells. J. Funct. Foods 2015, 14, 513–518. [Google Scholar] [CrossRef]
  110. Nami, Y.; Haghshenas, B.; Haghshenas, M.; Abdullah, N.; Yari Khosroushahi, A. The Prophylactic Effect of Probiotic Enterococcus lactis IW5 against Different Human Cancer Cells. Front. Microbiol. 2015, 6, 1317. [Google Scholar] [CrossRef] [Green Version]
  111. Bahmani, S.; Azarpira, N.; Moazamian, E. Anti-colon cancer activity of Bifidobacterium metabolites on colon cancer cell line SW742. Turk. J. Gastroenterol. 2019, 30, 835–842. [Google Scholar] [CrossRef]
  112. Alan, Y.; Savcı, A.; Koçpınar, E.F.; Ertaş, M. Postbiotic metabolites, antioxidant and anticancer activities of probiotic Leuconostoc pseudomesenteroides strains in natural pickles. Arch. Microbiol. 2022, 204, 571. [Google Scholar] [CrossRef]
  113. Dikeocha, I.J.; Al-Kabsi, A.M.; Chiu, H.-T.; Alshawsh, M.A. Faecalibacterium prausnitzii Ameliorates Colorectal Tumorigenesis and Suppresses Proliferation of HCT116 Colorectal Cancer Cells. Biomedicines 2022, 10, 1128. [Google Scholar] [CrossRef]
  114. Dikeocha, I.J.; Al-Kabsi, A.M.; Ahmeda, A.F.; Mathai, M.; Alshawsh, M.A. Investigation into the Potential Role of Propionibacterium freudenreichii in Prevention of Colorectal Cancer and Its Effects on the Diversity of Gut Microbiota in Rats. Int. J. Mol. Sci. 2023, 24, 8080. [Google Scholar] [CrossRef]
  115. Wan, Y.; Xin, Y.; Zhang, C.; Wu, D.; Ding, D.; Tang, L.; Owusu, L.; Bai, J.; Li, W. Fermentation supernatants of Lactobacillus delbrueckii inhibit growth of human colon cancer cells and induce apoptosis through a caspase 3-dependent pathway. Oncol. Lett. 2014, 7, 1738–1742. [Google Scholar] [CrossRef] [Green Version]
  116. Madempudi, R.S.; Kalle, A.M. Antiproliferative Effects of Bacillus coagulans Unique IS2 in Colon Cancer Cells. Nutr. Cancer 2017, 69, 1062–1068. [Google Scholar] [CrossRef]
  117. Guo, Y.; Zhang, T.; Gao, J.; Jiang, X.; Tao, M.; Zeng, X.; Wu, Z.; Pan, D. Lactobacillus acidophilus CICC 6074 inhibits growth and induces apoptosis in colorectal cancer cells in vitro and in HT-29 cells induced-mouse model. J. Funct. Foods 2020, 75, 104290. [Google Scholar] [CrossRef]
  118. Dehghani, N.; Tafvizi, F.; Jafari, P. Cell cycle arrest and anti-cancer potential of probiotic Lactobacillus rhamnosus against HT-29 cancer cells. Bioimpacts. 2021, 11, 245–252. [Google Scholar] [CrossRef]
  119. Kim, Y.; Kim, H.J.; Ji, K. The Proliferation Inhibitory Effect of Postbiotics Prepared from Probiotics with Antioxidant Activity against HT-29 Cells. Appl. Sci. 2022, 12, 12519. [Google Scholar] [CrossRef]
  120. Nowak, A.; Zakłos-Szyda, M.; Rosicka-Kaczmarek, J.; Motyl, I. Anticancer Potential of Post-Fermentation Media and Cell Extracts of Probiotic Strains: An In Vitro Study. Cancers 2022, 14, 1853. [Google Scholar] [CrossRef]
  121. Baghbani-Arani, F.; Asgary, V.; Hashemi, A. Cell-free extracts of Lactobacillus acidophilus and Lactobacillus delbrueckii display antiproliferative and antioxidant activities against HT-29 cell line. Nutr. Cancer 2020, 72, 1390–1399. [Google Scholar] [CrossRef]
  122. Amin, M.; Navidifar, T.; Saeb, S.; Barzegari, E.; Jamalan, M. Tumor-targeted induction of intrinsic apoptosis in colon cancer cells by Lactobacillus plantarum and Lactobacillus rhamnosus strains. Mol. Biol. Rep. 2023, 50, 5345–5354. [Google Scholar] [CrossRef]
  123. Jan, G.; Belzacq, A.S.; Haouzi, D.; Rouault, A.; Métivier, D.; Kroemer, G.; Brenner, C. Propionibacteria induce apoptosis of colorectal carcinoma cells via short-chain fatty acids acting on mitochondria. Cell. Death Differ. 2002, 9, 179–188. [Google Scholar] [CrossRef]
  124. Kim, Y.; Lee, D.; Kim, D.; Cho, J.; Yang, J.; Chung, M.; Kim, K.; Ha, N. Inhibition of proliferation in colon cancer cell lines and harmful enzyme activity of colon bacteria by Bifidobacterium adolescentis SPM0212. Arch. Pharm. Res. 2008, 31, 468–473. [Google Scholar] [CrossRef]
  125. Sharma, P.; Kaur, S.; Kaur, R.; Kaur, M.; Kaur, S. Proteinaceous Secretory Metabolites of Probiotic Human Commensal Enterococcus hirae 20c, E. faecium 12a and L12b as Antiproliferative Agents Against Cancer Cell Lines. Front. Microbiol. 2018, 9, 948. [Google Scholar] [CrossRef] [Green Version]
  126. Srikham, K.; Thirabunyanon, M. Bioprophylactic potential of novel human colostrum probiotics via apoptotic induction of colon cancer cells and cell immune activation. Biomed. Pharmacother. 2022, 149, 112871. [Google Scholar] [CrossRef]
  127. Barigela, A.; Bhukya, B. Probiotic Pediococcus acidilactici strain from tomato pickle displays anti-cancer activity and alleviates gut inflammation in-vitro. 3 Biotech 2021, 11, 23. [Google Scholar] [CrossRef]
  128. Pahumunto, N.; Teanpaisan, R. Anti-cancer Properties of Potential Probiotics and Their Cell-free Supernatants for the Prevention of Colorectal Cancer: An In Vitro Study. Probiotics Antimicrob. Proteins. 2022. [Google Scholar] [CrossRef]
  129. Isono, A.; Katsuno, T.; Sato, T.; Nakagawa, T.; Kato, Y.; Sato, N.; Seo, G.; Suzuki, Y.; Saito, Y. Clostridium butyricum TO-A culture supernatant downregulates TLR4 in human colonic epithelial cells. Dig. Dis. Sci. 2007, 52, 2963–2971. [Google Scholar] [CrossRef]
  130. Escamilla, J.; Lane, M.A.; Maitin, V. Cell-free supernatants from probiotic Lactobacillus casei and Lactobacillus rhamnosus GG decrease colon cancer cell invasion in vitro. Nutr. Cancer 2012, 64, 871–878. [Google Scholar] [CrossRef]
  131. Nouri, Z.; Karami, F.; Neyazi, N.; Modarressi, M.H.; Karimi, R.; Khorramizadeh, M.R.; Taheri, B.; Motevaseli, E. Dual Anti-Metastatic and Anti-Proliferative Activity Assessment of Two Probiotics on HeLa and HT-29 Cell Lines. Cell. J. 2016, 18, 127–134. [Google Scholar] [CrossRef]
  132. An, J.; Ha, E.M. Lactobacillus-derived metabolites enhance the antitumor activity of 5-FU and inhibit metastatic behavior in 5-FU-resistant colorectal cancer cells by regulating claudin-1 expression. J. Microbiol. 2020, 58, 967–977. [Google Scholar] [CrossRef]
  133. Maghsood, F.; Johari, B.; Rohani, M.; Madanchi, H.; Saltanatpour, Z.; Kadivar, M. Anti-proliferative and Anti-metastatic Potential of High Molecular Weight Secretory Molecules from Probiotic Lactobacillus Reuteri Cell-Free Supernatant Against Human Colon Cancer Stem-Like Cells (HT29-ShE). Int. J. Pept. Res. Ther. 2020, 26, 2619–2631. [Google Scholar] [CrossRef]
  134. Yue, Y.C.; Yang, B.Y.; Lu, J.; Zhang, S.-W.; Liu, L.; Nassar, K.; Xu, X.-X.; Pang, X.-Y.; Lv, J.-P. Metabolite secretions of Lactobacillus plantarum YYC-3 may inhibit colon cancer cell metastasis by suppressing the VEGF-MMP2/9 signaling pathway. Microb. Cell. Fact. 2020, 19, 213. [Google Scholar] [CrossRef]
  135. Tegopoulos, K.; Stergiou, O.S.; Kiousi, D.E.; Tsifintaris, M.; Koletsou, E.; Papageorgiou, A.C.; Argyri, A.A.; Chorianopoulos, N.; Galanis, A.; Kolovos, P. Genomic and Phylogenetic Analysis of Lactiplantibacillus plantarum L125, and Evaluation of Its Anti-Proliferative and Cytotoxic Activity in Cancer Cells. Biomedicines 2021, 9, 1718. [Google Scholar] [CrossRef]
  136. Salemi, R.; Vivarelli, S.; Ricci, D.; Scillato, M.; Santagati, M.; Gattuso, G.; Falzone, L.; Libra, M. Lactobacillus rhamnosus GG cell-free supernatant as a novel anti-cancer adjuvant. J. Transl. Med. 2023, 21, 195. [Google Scholar] [CrossRef]
  137. Lee, J.; Lee, J.E.; Kim, S.; Kang, D.; Yoo, H.M. Evaluating Cell Death Using Cell-Free Supernatant of Probiotics in Three-Dimensional Spheroid Cultures of Colorectal Cancer Cells. J. Vis. Exp. 2020, 160, e61285. [Google Scholar] [CrossRef]
  138. Vallino, L.; Garavaglia, B.; Visciglia, A.; Amoruso, A.; Pane, M.; Ferraresi, A.; Isidoro, C. Cell-free Lactiplantibacillus plantarum OC01 supernatant suppresses IL-6-induced proliferation and invasion of human colorectal cancer cells: Effect on β-Catenin degradation and induction of autophagy. J. Tradit. Complement. Med. 2023, 13, 193–206. [Google Scholar] [CrossRef]
  139. Saxami, G.; Karapetsas, A.; Lamprianidou, E.; Kotsianidis, I.; Chlichlia, A.; Tassou, C.; Zoumpourlis, V.; Galanis, A. Two potential probiotic lactobacillus strains isolated from olive microbiota exhibit adhesion and anti-proliferative effects in cancer cell lines. J. Funct. Foods 2016, 24, 461–471. [Google Scholar] [CrossRef]
  140. Dong, Y.; Zhu, J.; Zhang, M.; Ge, S.; Zhao, L. Probiotic Lactobacillus salivarius Ren prevent dimethylhydrazine-induced colorectal cancer through protein kinase B inhibition. Appl. Microbiol. Biotechnol. 2020, 104, 7377–7389. [Google Scholar] [CrossRef]
  141. An, J.; Ha, E.M. Combination Therapy of Lactobacillus plantarum Supernatant and 5-Fluouracil Increases Chemosensitivity in Colorectal Cancer Cells. J. Microbiol. Biotechnol. 2016, 26, 1490–1503. [Google Scholar] [CrossRef]
  142. Kim, H.J.; An, J.; Ha, E.M. Lactobacillus plantarum-derived metabolites sensitize the tumor-suppressive effects of butyrate by regulating the functional expression of SMCT1 in 5-FU-resistant colorectal cancer cells. J. Microbiol. 2022, 60, 100–117. [Google Scholar] [CrossRef]
  143. Jeong, S.; Kim, Y.; Park, S.; Lee, D.; Lee, J.; Hlaing, S.P.; Yoo, J.-W.; Rhee, S.H.; Im, E. Lactobacillus plantarum Metabolites Elicit Anticancer Effects by Inhibiting Autophagy-Related Responses. Molecules 2023, 28, 1890. [Google Scholar] [CrossRef]
  144. Saber, A.; Alipour, B.; Faghfoori, Z.; Mousavi Jam, A.; Yari Khosroushahi, A. Secretion metabolites of probiotic yeast, Pichia kudriavzevii AS-12, induces apoptosis pathways in human colorectal cancer cell lines. Nutr. Res. 2017, 41, 36–46. [Google Scholar] [CrossRef]
  145. Nag, D.; Goel, A.; Padwad, Y.; Singh, D. In Vitro Characterisation Revealed Himalayan Dairy Kluyveromyces marxianus PCH397 as Potential Probiotic with Therapeutic Properties. Probiotics Antimicrob. Proteins. 2023, 15, 761–773. [Google Scholar] [CrossRef]
  146. Jurášková, D.; Ribeiro, S.C.; Silva, C.C.G. Exopolysaccharides Produced by Lactic Acid Bacteria: From Biosynthesis to Health-Promoting Properties. Foods 2022, 11, 156. [Google Scholar] [CrossRef]
  147. Oerlemans, M.M.P.; Akkerman, R.; Ferrari, M.; Walvoort, M.T.C.; de Vos, P. Benefits of bacteria-derived exopolysaccharides on gastrointestinal microbiota, immunity and health. J. Funct. Foods 2021, 76, 104289. [Google Scholar] [CrossRef]
  148. Tang, H.; Huang, W.; Yao, Y.F. The metabolites of lactic acid bacteria: Classification, biosynthesis and modulation of gut microbiota. Microb. Cell. 2023, 10, 49–62. [Google Scholar] [CrossRef]
  149. Zhou, Y.; Cui, Y.; Qu, X. Exopolysaccharides of lactic acid bacteria: Structure, bioactivity and associations: A review. Carbohydr. Polym. 2019, 207, 317–332. [Google Scholar] [CrossRef]
  150. Angelin, J.; Kavitha, M. Exopolysaccharides from probiotic bacteria and their health potential. J. Biol. Macromol. 2020, 162, 853–865. [Google Scholar] [CrossRef]
  151. Wu, J.; Zhang, Y.; Ye, L.; Wang, C. The anti-cancer effects and mechanisms of lactic acid bacteria exopolysaccharides in vitro: A review. Carbohydr. Polym. 2021, 253, 117308. [Google Scholar] [CrossRef]
  152. Oleksy, M.; Klewicka, E. Exopolysaccharides produced by Lactobacillus sp.: Biosynthesis and applications. Crit. Rev. Food Sci. Nutr. 2018, 58, 450–462. [Google Scholar] [CrossRef]
  153. Haroun, B.M.; Refaat, B.M.; El- Menoufy, H.A.; Amin, H.A.; El-Waseif, A.A. Structure Analysis and Antitumor Activity of the Exopolysaccharide from Probiotic Lactobacillus plantarum NRRL B-4496 In vitro and In vivo. J. Appl. Sci. Res. 2013, 9, 425–434. [Google Scholar]
  154. Wang, K.; Li, W.; Rui, X.; Chen, X.; Jiang, M.; Dong, M. Characterization of a novel exopolysaccharide with antitumor activity from Lactobacillus plantarum 70810. Int. J. Biol. Macromol. 2014, 63, 133–139. [Google Scholar] [CrossRef]
  155. Li, W.; Tang, W.; Ji, J.; Xia, X.; Rui, X.; Chen, X.; Jiang, M.; Zhou, J.; Dong, M. Characterization of a novel polysaccharide with anti-colon cancer activity from Lactobacillus helveticus MB2-1. Carbohydr. Res. 2015, 411, 6–14. [Google Scholar] [CrossRef]
  156. Wang, J.; Zhao, X.; Yang, Y.; Zhao, A.; Yang, Z. Characterization and bioactivities of an exopolysaccharide produced by Lactobacillus plantarum YW32. Int. J. Biol. Macromol. 2015, 74, 119–126. [Google Scholar] [CrossRef]
  157. Liu, Z.; Zhang, Z.; Qiu, L.; Zhang, F.; Xu, X.; Wei, H.; Tao, X. Characterization and bioactivities of the exopolysaccharide from a probiotic strain of Lactobacillus plantarum WLPL04. J. Dairy Sci. 2017, 100, 6895–6905. [Google Scholar] [CrossRef]
  158. Ayyash, M.; Abu-Jdayil, B.; Itsaranuwat, P.; Almazrouei, N.; Galiwango, E.; Esposito, G.; Hunashal, Y.; Hamed, F.; Najjar, Z. Exopolysaccharide produced by the potential probiotic Lactococcus garvieae C47: Structural characteristics, rheological properties, bioactivities and impact on fermented camel milk. Food Chem. 2020, 333, 127418. [Google Scholar] [CrossRef]
  159. Ayyash, M.; Abu-Jdayil, B.; Olaimat, A.; Esposito, G.; Itsaranuwat, P.; Osaili, T.; Obaid, R.; Kizhakkayil, J.; Liu, S.Q. Physicochemical, bioactive and rheological properties of an exopolysaccharide produced by a probiotic Pediococcus pentosaceus M41. Carbohydr. Polym. 2020, 229, 115462. [Google Scholar] [CrossRef]
  160. Kumar, R.; Bansal, P.; Singh, J.; Dhanda, S. Purification, partial structural characterization and health benefits of exopolysaccharides from potential probiotic Pediococcus acidilactici NCDC 252. Process. Biochem. 2020, 99, 79–86. [Google Scholar] [CrossRef]
  161. Di, W.; Zhang, L.; Wang, S.; Yi, H.; Han, X.; Fan, R.; Zhang, Y. Physicochemical characterization and antitumour activity of exopolysaccharides produced by Lactobacillus casei SB27 from yak milk. Carbohydr. Polym. 2017, 171, 307–315. [Google Scholar] [CrossRef]
  162. Rajoka, M.S.R.; Mehwish, H.M.; Fang, H.; Padhiar, A.A.; Zeng, X.; Khurshid, M.; He, Z.; Zhao, L. Characterization and anti-tumor activity of exopolysaccharide produced by Lactobacillus kefiri isolated from Chinese kefir grains. J. Funct. Foods. 2019, 63, 103588. [Google Scholar] [CrossRef]
  163. Tukenmez, U.; Aktas, B.; Aslim, B.; Yavuz, S. The relationship between the structural characteristics of lactobacilli-EPS and its ability to induce apoptosis in colon cancer cells in vitro. Sci. Rep. 2019, 9, 8268. [Google Scholar] [CrossRef] [Green Version]
  164. Sun, M.; Liu, W.; Song, Y.; Tuo, Y.; Mu, G.; Ma, F. The Effects of Lactobacillus plantarum-12 Crude Exopolysaccharides on the Cell Proliferation and Apoptosis of Human Colon Cancer (HT-29) Cells. Probiotics Antimicrob. Proteins. 2021, 13, 413–421. [Google Scholar] [CrossRef]
  165. Khalil, M.A.; Sonbol, F.I.; Al-Madboly, L.A.; Aboshady, T.A.; Alqurashi, A.S.; Ali, S.S. Exploring the Therapeutic Potentials of Exopolysaccharides Derived from Lactic Acid Bacteria and Bifidobacteria: Antioxidant, Antitumor, and Periodontal Regeneration. Front. Microbiol. 2022, 13, 803688. [Google Scholar] [CrossRef]
  166. Zhou, X.; Hong, T.; Yu, Q.; Nie, S.; Gong, D.; Xiong, T.; Xie, M. Exopolysaccharides from Lactobacillus plantarum NCU116 induce c-Jun dependent Fas/Fasl-mediated apoptosis via TLR2 in mouse intestinal epithelial cancer cells. Sci. Rep. 2017, 7, 14247. [Google Scholar] [CrossRef] [Green Version]
  167. Di, W.; Zhang, L.; Yi, H.; Han, X.; Zhang, Y.; Xin, L. Exopolysaccharides produced by Lactobacillus strains suppress HT-29 cell growth via induction of G0/G1 cell cycle arrest and apoptosis. Oncol. Lett. 2018, 16, 3577–3586. [Google Scholar] [CrossRef] [Green Version]
  168. El-Deeb, N.M.; Yassin, A.M.; Al-Madboly, L.A.; El-Hawiet, A. A novel purified Lactobacillus acidophilus 20079 exopolysaccharide, LA-EPS-20079, molecularly regulates both apoptotic and NF-κB inflammatory pathways in human colon cancer. Microb. Cell. Fact. 2018, 17, 29. [Google Scholar] [CrossRef] [Green Version]
  169. Mojibi, P.; Tafvizi, F.; Bikhof Torbati, M. Cell-bound Exopolysaccharide Extract from Indigenous Probiotic Bacteria Induce Apoptosis in HT-29 cell-line. Iran. J. Pathol. 2019, 14, 41–51. [Google Scholar] [CrossRef] [Green Version]
  170. Wei, Y.; Li, F.; Li, L.; Huang, L.; Li, Q. Genetic and Biochemical Characterization of an Exopolysaccharide With in vitro Antitumoral Activity Produced by Lactobacillus fermentum YL-11. Front. Microbiol. 2019, 10, 2898. [Google Scholar] [CrossRef]
  171. Deepak, V.; Ramachandran, S.; Balahmar, R.M.; Pandian, S.R.; Sivasubramaniam, S.D.; Nellaiah, H.; Sundar, K. In vitro evaluation of anticancer properties of exopolysaccharides from Lactobacillus acidophilus in colon cancer cell lines. In Vitro Cell. Dev. Biol. Anim. 2016, 52, 163–173. [Google Scholar] [CrossRef]
  172. Deepak, V.; Ram Kumar Pandian, S.; Sivasubramaniam, S.D.; Nellaiah, H.; Sundar, K. Optimization of anticancer exopolysaccharide production from probiotic Lactobacillus acidophilus by response surface methodology. Prep. Biochem. Biotechnol. 2016, 46, 288–297. [Google Scholar] [CrossRef]
  173. Kim, Y.; Oh, S.; Yun, H.S.; Oh, S.; Kim, S.H. Cell-bound exopolysaccharide from probiotic bacteria induces autophagic cell death of tumour cells. Lett. Appl. Microbiol. 2010, 51, 123–130. [Google Scholar] [CrossRef]
  174. Liu, C.T.; Chu, F.J.; Chou, C.C.; Yu, R.C. Antiproliferative and anticytotoxic effects of cell fractions and exopolysaccharides from Lactobacillus casei 01. Mutat. Res. 2011, 721, 157–162. [Google Scholar] [CrossRef]
  175. Saadat, Y.R.; Khosroushahi, A.Y.; Movassaghpour, A.A.; Talebi, M.; Gargari, B.P. Modulatory role of exopolysaccharides of Kluyveromyces marxianus and Pichia kudriavzevii as probiotic yeasts from dairy products in human colon cancer cells. J. Funct. Foods 2020, 64, 103675. [Google Scholar] [CrossRef]
  176. Daba, G.M.; Elnahas, M.O.; Elkhateeb, W.A. Beyond biopreservatives, bacteriocins biotechnological applications: History, current status, and promising potentials. Biocatal. Agric. Biotechnol. 2022, 39, 102248. [Google Scholar] [CrossRef]
  177. Teng, K.; Huang, F.; Liu, Y.; Wang, Y.; Xia, T.; Yun, F.; Zhong, J. Food and gut originated bacteriocins involved in gut microbe-host interactions. Crit. Rev. Microbiol. 2022, 49, 515–527. [Google Scholar] [CrossRef]
  178. Kaur, S.; Kaur, S. Bacteriocins as Potential Anticancer Agents. Front. Pharmacol. 2015, 6, 272. [Google Scholar] [CrossRef] [Green Version]
  179. Daba, G.M.; Elkhateeb, W.A. Bacteriocins of lactic acid bacteria as biotechnological tools in food and pharmaceuticals: Current applications and future prospects. Biocatal. Agric. Biotechnol. 2020, 28, 101750. [Google Scholar] [CrossRef]
  180. Molujin, A.M.; Abbasiliasi, S.; Nurdin, A.; Lee, P.-C.; Gansau, J.A.; Jawan, R. Bacteriocins as Potential Therapeutic Approaches in the Treatment of Various Cancers: A Review of In Vitro Studies. Cancers 2022, 14, 4758. [Google Scholar] [CrossRef]
  181. Lawrence, G.W.; McCarthy, N.; Walsh, C.J.; Kunyoshi, T.M.; Lawton, E.M.; O’Connor, P.M.; Begley, M.; Cotter, P.D.; Guinane, C.M. Effect of a bacteriocin-producing Streptococcus salivarius on the pathogen Fusobacterium nucleatum in a model of the human distal colon. Gut Microbes. 2022, 14, 2100203. [Google Scholar] [CrossRef]
  182. Dreyer, L.; Smith, C.; Deane, S.M.; Dicks, L.M.T.; van Staden, A.D. Migration of Bacteriocins Across Gastrointestinal Epithelial and Vascular Endothelial Cells, as Determined Using In Vitro Simulations. Sci. Rep. 2019, 9, 11481. [Google Scholar] [CrossRef] [Green Version]
  183. Dicks, L.M.T.; Dreyer, L.; Smith, C.; van Staden, A.D. A Review: The Fate of Bacteriocins in the Human Gastro-Intestinal Tract: Do They Cross the Gut-Blood Barrier? Front. Microbiol. 2018, 9, 2297. [Google Scholar] [CrossRef] [Green Version]
  184. Dobrzyńska, I.; Szachowicz-Petelska, B.; Sulkowski, S.; Figaszewski, Z. Changes in electric charge and phospholipids composition in human colorectal cancer cells. Mol. Cell. Biochem. 2005, 276, 113–119. [Google Scholar] [CrossRef]
  185. Broughton, L.J.; Crow, C.; Maraveyas, A.; Madden, L.A. Duramycin-induced calcium release in cancer cells. Anticancer Drugs 2016, 27, 173–182. [Google Scholar] [CrossRef]
  186. Datta, M.; Rajeev, A.; Chattopadhyay, I. Application of antimicrobial peptides as next-generation therapeutics in the biomedical world. Biotechnol. Genet. Eng. Rev. 2023, 1–39. [Google Scholar] [CrossRef]
  187. Goh, K.S.; Ng, Z.J.; Halim, M.; Oslan, S.N.; Oslan, S.N.H.; Tan, J.S. A Comprehensive Review on the Anticancer Potential of Bacteriocin: Preclinical and Clinical Studies. Int. J. Pept. Res. Ther. 2022, 28, 75. [Google Scholar] [CrossRef]
  188. Piper, C.; Hill, C.; Cotter, P.D.; Ross, R.P. Bioengineering of a Nisin A-producing Lactococcus lactis to create isogenic strains producing the natural variants Nisin F, Q and Z. Microb. Biotechnol. 2011, 4, 375–382. [Google Scholar] [CrossRef] [Green Version]
  189. Maher, S.; McClean, S. Investigation of the cytotoxicity of eukaryotic and prokaryotic antimicrobial peptides in intestinal epithelial cells in vitro. Biochem. Pharmacol. 2006, 71, 1289–1298. [Google Scholar] [CrossRef]
  190. Joo, N.E.; Ritchie, K.; Kamarajan, P.; Miao, D.; Kapila, Y.L. Nisin, an apoptogenic bacteriocin and food preservative, attenuates HNSCC tumorigenesis via CHAC1. Cancer Med. 2012, 1, 295–305. [Google Scholar] [CrossRef]
  191. Khazaei Monfared, Y.; Mahmoudian, M.; Cecone, C.; Caldera, F.; Zakeri-Milani, P.; Matencio, A.; Trotta, F. Stabilization and Anticancer Enhancing Activity of the Peptide Nisin by Cyclodextrin-Based Nanosponges against Colon and Breast Cancer Cells. Polymers 2022, 14, 594. [Google Scholar] [CrossRef]
  192. Soltani, S.; Zirah, S.; Rebuffat, S.; Couture, F.; Boutin, Y.; Biron, E.; Subirade, M.; Fliss, I. Gastrointestinal Stability and Cytotoxicity of Bacteriocins From Gram-Positive and Gram-Negative Bacteria: A Comparative in vitro Study. Front. Microbiol. 2022, 12, 780355. [Google Scholar] [CrossRef]
  193. Hosseini, S.S.; Hajikhani, B.; Faghihloo, E.; Goudarzi, H. Increased expression of caspase genes in colorectal cancer cell line by nisin. Arch. Clin. Infect. Dis. 2020, 15, e97734. [Google Scholar] [CrossRef] [Green Version]
  194. Ahmadi, S.; Ghollasi, M.; Hosseini, H.M. The apoptotic impact of nisin as a potent bacteriocin on the colon cancer cells. Microb. Pathog. 2017, 111, 193–197. [Google Scholar] [CrossRef]
  195. Hosseini, S.S.; Goudarzi, H.; Ghalavand, Z.; Hajikhani, B.; Rafeieiatani, Z.; Hakemi-Vala, M. Anti-proliferative effects of cell wall, cytoplasmic extract of Lactococcus lactis and nisin through down-regulation of cyclin D1 on SW480 colorectal cancer cell line. Iran. J. Microbiol. 2020, 12, 424–430. [Google Scholar] [CrossRef]
  196. Norouzi, Z.; Salimi, A.; Halabian, R.; Fahimi, H. Nisin, a potent bacteriocin and anti-bacterial peptide, attenuates expression of metastatic genes in colorectal cancer cell lines. Microb. Pathog. 2018, 123, 183–189. [Google Scholar] [CrossRef]
  197. Kamarajan, P.; Hayami, T.; Matte, B.; Liu, Y.; Danciu, T.; Ramamoorthy, A.; Worden, F.; Kapila, S.; Kapila, Y. Nisin ZP, a Bacteriocin and Food Preservative, Inhibits Head and Neck Cancer Tumorigenesis and Prolongs Survival. PLoS ONE 2015, 10, e0131008. [Google Scholar] [CrossRef] [Green Version]
  198. Ankaiah, D.; Esakkiraj, P.; Perumal, V.; Ayyanna, R.; Venkatesan, A. Probiotic characterization of Enterococcus faecium por1: Cloning, over expression of Enterocin-a and evaluation of antibacterial, anticancer properties. J. Funct. Foods 2017, 38, 280–292. [Google Scholar] [CrossRef]
  199. Ankaiah, D.; Palanichamy, E.; Antonyraj, C.B.; Ayyanna, R.; Perumal, V.; Ahamed, S.I.B.; Arul, V. Cloning, overexpression, purification of bacteriocin enterocin-B and structural analysis, interaction determination of enterocin-A, B against pathogenic bacteria and human cancer cells. Int. J. Biol. Macromol. 2018, 116, 502–512. [Google Scholar] [CrossRef]
  200. Sharma, P.; Kaur, S.; Chadha, B.S.; Kaur, R.; Kaur, M.; Kaur, S. Anticancer and antimicrobial potential of enterocin 12a from Enterococcus faecium. BMC Microbiol. 2021, 21, 39. [Google Scholar] [CrossRef]
  201. Patra, S.; Sahu, N.; Saxena, S.; Pradhan, B.; Nayak, S.K.; Roychowdhury, A. Effects of Probiotics at the Interface of Metabolism and Immunity to Prevent Colorectal Cancer-Associated Gut Inflammation: A Systematic Network and Meta-Analysis With Molecular Docking Studies. Front. Microbiol. 2022, 13, 878297. [Google Scholar] [CrossRef] [PubMed]
  202. Villarante, K.I.; Elegado, F.B.; Iwatani, S.; Zendo, T.; Sonomoto, K.; de Guzman, E.E. Purification, characterization and in vitro cytotoxicity of the bacteriocin from Pediococcus acidilactici K2a2-3 against human colon adenocarcinoma (HT29) and human cervical carcinoma (HeLa) cells. World J. Microbiol. Biotechnol. 2011, 27, 975–980. [Google Scholar] [CrossRef]
  203. Buss, G.P.; Wilson, C.M. Exploring the cytotoxic mechanisms of Pediocin PA-1 towards HeLa and HT29 cells by comparison to known bacteriocins: Microcin E492, enterocin heterodimer and Divercin V41. PLoS ONE 2021, 16, e0251951. [Google Scholar] [CrossRef]
  204. Wang, H.; Jin, J.; Pang, X.; Bian, Z.; Zhu, J.; Hao, Y.; Zhang, H.; Xie, Y. Plantaricin BM-1 decreases viability of SW480 human colorectal cancer cells by inducing caspase-dependent apoptosis. Front. Microbiol. 2023, 13, 1103600. [Google Scholar] [CrossRef]
  205. De Giani, A.; Bovio, F.; Forcella, M.; Fusi, P.; Sello, G.; Di Gennaro, P. Identification of bacteriocin-like compound from Lactobacillus plantarum with antimicrobial activity and effects on normal and cancerogenic human intestinal cells. AMB Express 2019, 9, 88. [Google Scholar] [CrossRef] [Green Version]
  206. Al-Fakharany, O.M.; Aziz, A.A.A.; El-Banna, T.E.-S.; Sonbol, F.I. Immunomodulatory and Anticancer Activities of Enterocin Oe-342 Produced by Enterococcus Feacalis Isolated from Stool. J. Clin. Cell. Immunol. 2018, 9, 558. [Google Scholar] [CrossRef]
  207. Dan, A.K.; Manna, A.; Ghosh, S.; Sikdar, S.; Sahu, R.; Parhi, P.K.; Parida, S. Molecular mechanisms of the lipopeptides from Bacillus subtilis in the apoptosis of cancer cells—A review on its Current Status in different cancer cell lines. Adv. Cancer Biol. Metastasis 2021, 3, 100019. [Google Scholar] [CrossRef]
  208. Baindara, P.; Mandal, S.M. Bacteria and bacterial anticancer agents as a promising alternative for cancer therapeutics. Biochimie 2020, 177, 164–189. [Google Scholar] [CrossRef]
  209. Wu, Y.S.; Ngai, S.C.; Goh, B.H.; Chan, K.G.; Lee, L.H.; Chuah, L.H. Anticancer Activities of Surfactin and Potential Application of Nanotechnology Assisted Surfactin Delivery. Front. Pharmacol. 2017, 8, 761. [Google Scholar] [CrossRef] [Green Version]
  210. Sivapathasekaran, C.; Das, P.; Mukherjee, S.; Saravanakumar, J.; Mandal, M.; Sen, R. Marine Bacterium Derived Lipopeptides: Characterization and Cytotoxic Activity Against Cancer Cell Lines. Int. J. Pept. Res. Ther. 2010, 16, 215–222. [Google Scholar] [CrossRef]
  211. Kim, S.Y.; Kim, J.Y.; Kim, S.H.; Bae, H.J.; Yi, H.; Yoon, S.H.; Koo, B.S.; Kwon, M.; Cho, J.Y.; Lee, C.E.; et al. Surfactin from Bacillus subtilis displays anti-proliferative effect via apoptosis induction, cell cycle arrest and survival signaling suppression. FEBS Lett. 2007, 581, 865–871. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Zhao, H.; Xu, X.; Lei, S.; Shao, D.; Jiang, C.; Shi, J.; Zhang, Y.; Liu, L.; Lei, S.; Sun, H.; et al. Iturin A-like lipopeptides from Bacillus subtilis trigger apoptosis, paraptosis, and autophagy in Caco-2 cells. J. Cell. Physiol. 2019, 234, 6414–6427. [Google Scholar] [CrossRef] [PubMed]
  213. Cheng, W.; Feng, Y.Q.; Ren, J.; Jing, D.; Wang, C. Anti-tumor role of Bacillus subtilis fmbJ-derived fengycin on human colon cancer HT29 cell line. Neoplasma 2016, 63, 215–222. [Google Scholar] [CrossRef] [Green Version]
  214. Soleimanpour, S.; Hasanian, S.M.; Avan, A.; Yaghoubi, A.; Khazaei, M. Bacteriotherapy in gastrointestinal cancer. Life Sci. 2020, 254, 117754. [Google Scholar] [CrossRef]
  215. Ebrahimzadeh, S.; Ahangari, H.; Soleimanian, A.; Hosseini, K.; Ebrahimi, V.; Ghasemnejad, T.; Soofiyani, S.R.; Tarhriz, V.; Eyvazi, S. Colorectal cancer treatment using bacteria: Focus on molecular mechanisms. BMC Microbiol. 2021, 21, 218. [Google Scholar] [CrossRef]
  216. Karpiński, T.M.; Adamczak, A. Anticancer Activity of Bacterial Proteins and Peptides. Pharmaceutics 2018, 10, 54. [Google Scholar] [CrossRef] [Green Version]
  217. Zhang, H.L.; Hua, H.M.; Pei, Y.H.; Yao, X.S. Three new cytotoxic cyclic acylpeptides from marine Bacillus sp. Chem. Pharm. Bull. 2004, 52, 1029–1030. [Google Scholar] [CrossRef] [Green Version]
  218. Rodrigues, G.; Silva, G.G.O.; Buccini, D.F.; Duque, H.M.; Dias, S.C.; Franco, O.L. Bacterial Proteinaceous Compounds With Multiple Activities Toward Cancers and Microbial Infection. Front. Microbiol. 2019, 10, 1690. [Google Scholar] [CrossRef] [Green Version]
  219. Chauhan, S.; Dhawan, D.K.; Saini, A.; Preet, S. Antimicrobial peptides against colorectal cancer-a focused review. Pharmacol. Res. 2021, 167, 105529. [Google Scholar] [CrossRef]
  220. He, J.F.; Jin, D.X.; Luo, X.G.; Zhang, T.C. LHH1, a novel antimicrobial peptide with anti-cancer cell activity identified from Lactobacillus casei HZ1. AMB Express 2020, 10, 204. [Google Scholar] [CrossRef] [PubMed]
  221. Tsai, T.L.; Li, A.C.; Chen, Y.C.; Liao, Y.S.; Lin, T.H. Antimicrobial peptide m2163 or m2386 identified from Lactobacillus casei ATCC 334 can trigger apoptosis in the human colorectal cancer cell line SW480. Tumour Biol. 2015, 36, 3775–3789. [Google Scholar] [CrossRef] [PubMed]
  222. Chen, Y.C.; Tsai, T.L.; Ye, X.H.; Lin, T.H. Anti-proliferative effect on a colon adenocarcinoma cell line exerted by a membrane disrupting antimicrobial peptide KL15. Cancer Biol. Ther. 2015, 16, 1172–1183. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  223. Konishi, H.; Fujiya, M.; Tanaka, H.; Ueno, N.; Moriichi, K.; Sasajima, J.; Ikuta, K.; Akutsu, H.; Tanabe, H.; Kohgo, Y. Probiotic-derived ferrichrome inhibits colon cancer progression via JNK-mediated apoptosis. Nat. Commun. 2016, 7, 12365. [Google Scholar] [CrossRef]
  224. An, B.C.; Hong, S.; Park, H.J.; Kim, B.-K.; Ahn, J.Y.; Ryu, Y.; An, J.H.; Chung, M.J. Anti-Colorectal Cancer Effects of Probiotic-Derived p8 Protein. Genes 2019, 10, 624. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. An, B.C.; Ahn, J.Y.; Kwon, D.; Kwak, S.H.; Heo, J.Y.; Kim, S.; Ryu, Y.; Chung, M.J. Anti-Cancer Roles of Probiotic-Derived P8 Protein in Colorectal Cancer Cell Line DLD-1. Int. J. Mol. Sci. 2023, 24, 9857. [Google Scholar] [CrossRef] [PubMed]
  226. Ju, X.; Wu, X.; Chen, Y.; Cui, S.; Cai, Z.; Zhao, L.; Hao, Y.; Zhou, F.; Chen, F.; Yu, Z.; et al. Mucin Binding Protein of Lactobacillus casei Inhibits HT-29 Colorectal Cancer Cell Proliferation. Nutrients 2023, 15, 2314. [Google Scholar] [CrossRef]
  227. Cong, J.; Zhou, P.; Zhang, R. Intestinal Microbiota-Derived Short Chain Fatty Acids in Host Health and Disease. Nutrients 2022, 14, 1977. [Google Scholar] [CrossRef]
  228. Ramos Meyers, G.; Samouda, H.; Bohn, T. Short Chain Fatty Acid Metabolism in Relation to Gut Microbiota and Genetic Variability. Nutrients 2022, 14, 5361. [Google Scholar] [CrossRef]
  229. Gomes, S.; Rodrigues, A.C.; Pazienza, V.; Preto, A. Modulation of the Tumor Microenvironment by Microbiota-Derived Short-Chain Fatty Acids: Impact in Colorectal Cancer Therapy. Int. J. Mol. Sci. 2023, 24, 5069. [Google Scholar] [CrossRef]
  230. Fusco, W.; Lorenzo, M.B.; Cintoni, M.; Porcari, S.; Rinninella, E.; Kaitsas, F.; Lener, E.; Mele, M.C.; Gasbarrini, A.; Collado, M.C.; et al. Short-Chain Fatty-Acid-Producing Bacteria: Key Components of the Human Gut Microbiota. Nutrients 2023, 15, 2211. [Google Scholar] [CrossRef]
  231. Xiong, R.-G.; Zhou, D.-D.; Wu, S.-X.; Huang, S.-Y.; Saimaiti, A.; Yang, Z.-J.; Shang, A.; Zhao, C.-N.; Gan, R.-Y.; Li, H.-B. Health Benefits and Side Effects of Short-Chain Fatty Acids. Foods 2022, 11, 2863. [Google Scholar] [CrossRef]
  232. Carretta, M.D.; Quiroga, J.; López, R.; Hidalgo, M.A.; Burgos, R.A. Participation of Short-Chain Fatty Acids and Their Receptors in Gut Inflammation and Colon Cancer. Front. Physiol. 2021, 12, 662739. [Google Scholar] [CrossRef] [PubMed]
  233. Tian, Y.; Xu, Q.; Sun, L.; Ye, Y.; Ji, G. Short-chain fatty acids administration is protective in colitis-associated colorectal cancer development. J. Nutr. Biochem. 2018, 57, 103–109. [Google Scholar] [CrossRef]
  234. Pan, P.; Oshima, K.; Huang, Y.W.; Agle, K.A.; Drobyski, W.R.; Chen, X.; Zhang, J.; Yearsley, M.M.; Yu, J.; Wang, L.S. Loss of FFAR2 promotes colon cancer by epigenetic dysregulation of inflammation suppressors. Int. J. Cancer 2018, 143, 886–896. [Google Scholar] [CrossRef] [PubMed]
  235. Kim, M.; Friesen, L.; Park, J.; Kim, H.M.; Kim, C.H. Microbial metabolites, short-chain fatty acids, restrain tissue bacterial load, chronic inflammation, and associated cancer in the colon of mice. Eur. J. Immunol. 2018, 48, 1235–1247. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  236. van der Beek, C.M.; Dejong, C.H.C.; Troost, F.J.; Masclee, A.A.M.; Lenaerts, K. Role of short-chain fatty acids in colonic inflammation, carcinogenesis, and mucosal protection and healing. Nutr. Rev. 2017, 75, 286–305. [Google Scholar] [CrossRef] [Green Version]
  237. Wang, G.; Yu, Y.; Wang, Y.Z.; Wang, J.J.; Guan, R.; Sun, Y.; Shi, F.; Gao, J.; Fu, X.L. Role of SCFAs in gut microbiome and glycolysis for colorectal cancer therapy. J. Cell. Physiol. 2019, 234, 17023–17049. [Google Scholar] [CrossRef]
  238. Liu, P.; Wang, Y.; Yang, G.; Zhang, Q.; Meng, L.; Xin, Y.; Jiang, X. The role of short-chain fatty acids in intestinal barrier function, inflammation, oxidative stress, and colonic carcinogenesis. Pharmacol. Res. 2021, 165, 105420. [Google Scholar] [CrossRef]
  239. Alvandi, E.; Wong, W.K.M.; Joglekar, M.V.; Spring, K.J.; Hardikar, A.A. Short-chain fatty acid concentrations in the incidence and risk-stratification of colorectal cancer: A systematic review and meta-analysis. BMC Med. 2022, 20, 323. [Google Scholar] [CrossRef]
  240. Ohara, T.; Mori, T. Antiproliferative Effects of Short-chain Fatty Acids on Human Colorectal Cancer Cells via Gene Expression Inhibition. Anticancer Res. 2019, 39, 4659–4666. [Google Scholar] [CrossRef] [Green Version]
  241. Gomes, S.; Baltazar, F.; Silva, E.; Preto, A. Microbiota-Derived Short-Chain Fatty Acids: New Road in Colorectal Cancer Therapy. Pharmaceutics 2022, 14, 2359. [Google Scholar] [CrossRef] [PubMed]
  242. Chen, Y.; Chen, Y.-X. Microbiota-Associated Metabolites and Related Immunoregulation in Colorectal Cancer. Cancers 2021, 13, 4054. [Google Scholar] [CrossRef] [PubMed]
  243. Yao, Y.; Cai, X.; Fei, W.; Ye, Y.; Zhao, M.; Zheng, C. The role of short-chain fatty acids in immunity, inflammation and metabolism. Crit. Rev. Food Sci. Nutr. 2022, 62, 1–12. [Google Scholar] [CrossRef] [PubMed]
  244. Mirzaei, R.; Dehkhodaie, E.; Bouzari, B.; Rahimi, M.; Gholestani, A.; Hosseini-Fard, S.R.; Keyvani, H.; Teimoori, A.; Karampoor, S. Dual role of microbiota-derived short-chain fatty acids on host and pathogen. Biomed. Pharmacother. 2022, 145, 112352. [Google Scholar] [CrossRef]
  245. Wu, X.; Wu, Y.; He, L.; Wu, L.; Wang, X.; Liu, Z. Effects of the intestinal microbial metabolite butyrate on the development of colorectal cancer. J. Cancer 2018, 9, 2510–2517. [Google Scholar] [CrossRef]
  246. Chen, J.; Zhao, K.N.; Vitetta, L. Effects of Intestinal Microbial–Elaborated Butyrate on Oncogenic Signaling Pathways. Nutrients 2019, 11, 1026. [Google Scholar] [CrossRef] [Green Version]
  247. Gheorghe, A.S.; Negru, S.M.; Preda, M.; Mihăilă, R.I.; Komporaly, I.A.; Dumitrescu, E.A.; Lungulescu, C.V.; Kajanto, L.A.; Georgescu, B.; Radu, E.A.; et al. Biochemical and Metabolical Pathways Associated with Microbiota-Derived Butyrate in Colorectal Cancer and Omega-3 Fatty Acids Implications: A Narrative Review. Nutrients 2022, 14, 1152. [Google Scholar] [CrossRef]
  248. Garavaglia, B.; Vallino, L.; Ferraresi, A.; Esposito, A.; Salwa, A.; Vidoni, C.; Gentilli, S.; Isidoro, C. Butyrate Inhibits Colorectal Cancer Cell Proliferation through Autophagy Degradation of β-Catenin Regardless of APC and β-Catenin Mutational Status. Biomedicines 2022, 10, 1131. [Google Scholar] [CrossRef]
  249. Wang, L.; Shannar, A.A.F.; Wu, R.; Chou, P.; Sarwar, M.S.; Kuo, H.C.; Peter, R.M.; Wang, Y.; Su, X.; Kong, A.N. Butyrate Drives Metabolic Rewiring and Epigenetic Reprogramming in Human Colon Cancer Cells. Mol. Nutr. Food Res. 2022, 66, e2200028. [Google Scholar] [CrossRef]
  250. Xiao, T.; Wu, S.; Yan, C.; Zhao, C.; Jin, H.; Yan, N.; Xu, J.; Wu, Y.; Li, C.; Shao, Q.; et al. Butyrate upregulates the TLR4 expression and the phosphorylation of MAPKs and NK-κB in colon cancer cell in vitro. Oncol. Lett. 2018, 16, 4439–4447. [Google Scholar] [CrossRef] [Green Version]
  251. Bian, Z.; Sun, X.; Liu, L.; Qin, Y.; Zhang, Q.; Liu, H.; Mao, L.; Sun, S. Sodium Butyrate Induces CRC Cell Ferroptosis via the CD44/SLC7A11 Pathway and Exhibits a Synergistic Therapeutic Effect with Erastin. Cancers 2023, 15, 423. [Google Scholar] [CrossRef]
  252. Korsten, S.G.P.J.; Vromans, H.; Garssen, J.; Willemsen, L.E.M. Butyrate Protects Barrier Integrity and Suppresses Immune Activation in a Caco-2/PBMC Co-Culture Model While HDAC Inhibition Mimics Butyrate in Restoring Cytokine-Induced Barrier Disruption. Nutrients 2023, 15, 2760. [Google Scholar] [CrossRef] [PubMed]
  253. Huang, C.; Deng, W.; Xu, H.Z.; Zhou, C.; Zhang, F.; Chen, J.; Bao, Q.; Zhou, X.; Liu, M.; Li, J.; et al. Short-chain fatty acids reprogram metabolic profiles with the induction of reactive oxygen species production in human colorectal adenocarcinoma cells. Comput. Struct. Biotechnol. J. 2023, 21, 1606–1620. [Google Scholar] [CrossRef] [PubMed]
  254. Marques, C.; Oliveira, C.S.F.; Alves, S.; Chaves, S.R.; Coutinho, O.P.; Côrte-Real, M.; Preto, A. Acetate-induced apoptosis in colorectal carcinoma cells involves lysosomal membrane permeabilization and cathepsin D release. Cell. Death Dis. 2013, 4, e507. [Google Scholar] [CrossRef] [Green Version]
  255. Sahuri-Arisoylu, M.; Mould, R.R.; Shinjyo, N.; Bligh, S.W.A.; Nunn, A.V.W.; Guy, G.W.; Thomas, E.L.; Bell, J.D. Acetate Induces Growth Arrest in Colon Cancer Cells Through Modulation of Mitochondrial Function. Front. Nutr. 2021, 8, 588466. [Google Scholar] [CrossRef] [PubMed]
  256. Ryu, T.Y.; Kim, K.; Son, M.Y.; Min, J.K.; Kim, J.; Han, T.S.; Kim, D.S.; Cho, H.S. Downregulation of PRMT1, a histone arginine methyltransferase, by sodium propionate induces cell apoptosis in colon cancer. Oncol. Rep. 2019, 41, 1691–1699. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  257. Ryu, T.Y.; Kim, K.; Han, T.S.; Lee, M.O.; Lee, J.; Choi, J.; Jung, K.B.; Jeong, E.J.; An, D.M.; Jung, C.R.; et al. Human gut-microbiome-derived propionate coordinates proteasomal degradation via HECTD2 upregulation to target EHMT2 in colorectal cancer. ISME J. 2022, 16, 1205–1221. [Google Scholar] [CrossRef]
  258. Liu, C.; Zheng, J.; Ou, X.; Han, Y. Anti-cancer Substances and Safety of Lactic Acid Bacteria in Clinical Treatment. Front. Microbiol. 2021, 12, 722052. [Google Scholar] [CrossRef]
  259. Soltani, S.; Hammami, R.; Cotter, P.D.; Rebuffat, S.; Said, L.B.; Gaudreau, H.; Bédard, F.; Biron, E.; Drider, D.; Fliss, I. Bacteriocins as a new generation of antimicrobials: Toxicity aspects and regulations. FEMS Microbiol. Rev. 2021, 45, fuaa039. [Google Scholar] [CrossRef]
  260. Huang, F.; Teng, K.; Liu, Y.; Cao, Y.; Wang, T.; Ma, C.; Zhang, J.; Zhong, J. Bacteriocins: Potential for Human Health. Oxid. Med. Cell. Longev. 2021, 2021, 5518825. [Google Scholar] [CrossRef]
  261. Zou, J.; Jiang, H.; Cheng, H.; Fang, J.; Huang, G. Strategies for screening, purification and characterization of bacteriocins. Int. J. Biol. Macromol. 2018, 117, 781–789. [Google Scholar] [CrossRef] [PubMed]
  262. Benítez-Chao, D.F.; León-Buitimea, A.; Lerma-Escalera, J.A.; Morones-Ramírez, J.R. Bacteriocins: An Overview of Antimicrobial, Toxicity, and Biosafety Assessment by in vivo Models. Front. Microbiol. 2021, 12, 630695. [Google Scholar] [CrossRef] [PubMed]
  263. Heilbronner, S.; Krismer, B.; Brötz-Oesterhelt, H.; Peschel, A. The microbiome-shaping roles of bacteriocins. Nat. Rev. Microbiol. 2021, 19, 726–739. [Google Scholar] [CrossRef] [PubMed]
  264. Flynn, J.; Ryan, A.; Hudson, S.P. Pre-formulation and delivery strategies for the development of bacteriocins as next generation antibiotics. Eur. J. Pharm. Biopharm. 2021, 165, 149–163. [Google Scholar] [CrossRef] [PubMed]
  265. Varas, M.A.; Muñoz-Montecinos, C.; Kallens, V.; Simon, V.; Allende, M.L.; Marcoleta, A.E.; Lagos, R. Exploiting Zebrafish Xenografts for Testing the in vivo Antitumorigenic Activity of Microcin E492 Against Human Colorectal Cancer Cells. Front. Microbiol. 2020, 11, 405. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  266. Rana, K.; Sharma, R.; Preet, S. Augmented therapeutic efficacy of 5-fluorouracil in conjunction with lantibiotic nisin against skin cancer. Biochem. Biophys. Res. Commun. 2019, 520, 551–559. [Google Scholar] [CrossRef]
  267. Rahbar Saadat, Y.; Yari Khosroushahi, A.; Pourghassem Gargari, B. A comprehensive review of anticancer, immunomodulatory and health beneficial effects of the lactic acid bacteria exopolysaccharides. Carbohydr. Polym. 2019, 217, 79–89. [Google Scholar] [CrossRef]
  268. Salimi, F.; Farrokh, P. Recent advances in the biological activities of microbial exopolysaccharides. World J. Microbiol. Biotechnol. 2023, 39, 213. [Google Scholar] [CrossRef]
  269. Daba, G.M.; Elnahas, M.O.; Elkhateeb, W.A. Contributions of exopolysaccharides from lactic acid bacteria as biotechnological tools in food, pharmaceutical, and medical applications. Int. J. Biol. Macromol. 2021, 173, 79–89. [Google Scholar] [CrossRef]
  270. Muninathan, C.; Guruchandran, S.; Kalyan, A.J.V.; Ganesan, N.D. Microbial exopolysaccharides: Role in functional food engineering and gut-health management. Int. J. Food Sci. Technol. 2022, 57, 27–34. [Google Scholar] [CrossRef]
  271. Pourjafar, H.; Ansari, F.; Sadeghi, A.; Samakkhah, S.A.; Jafari, S.M. Functional and health-promoting properties of probiotics’ exopolysaccharides; isolation, characterization, and applications in the food industry. Crit. Rev. Food Sci. Nutr. 2022, 1–32. [Google Scholar] [CrossRef]
  272. Chen, Y.; Zhang, M.; Ren, F. A Role of Exopolysaccharide Produced by Streptococcus thermophilus in the Intestinal Inflammation and Mucosal Barrier in Caco-2 Monolayer and Dextran Sulphate Sodium-Induced Experimental Murine Colitis. Molecules 2019, 24, 513. [Google Scholar] [CrossRef] [Green Version]
  273. Kuang, J.H.; Huang, Y.Y.; Hu, J.S.; Yu, J.J.; Zhou, Q.Y.; Liu, D.M. Exopolysaccharides from Bacillus amyloliquefaciens DMBA-K4 Ameliorate Dextran Sodium Sulfate-Induced Colitis via Gut Microbiota Modulation. J. Funct. Foods 2020, 75, 104212. [Google Scholar] [CrossRef]
  274. Ma, F.; Song, Y.; Sun, M.; Wang, A.; Jiang, S.; Mu, G.; Tuo, Y. Exopolysaccharide Produced by Lactiplantibacillus plantarum-12 Alleviates Intestinal Inflammation and Colon Cancer Symptoms by Modulating the Gut Microbiome and Metabolites of C57BL/6 Mice Treated by Azoxymethane/Dextran Sulfate Sodium Salt. Foods 2021, 10, 3060. [Google Scholar] [CrossRef]
  275. Chung, K.S.; Shin, J.S.; Lee, J.H.; Park, S.E.; Han, H.S.; Rhee, Y.K.; Cho, C.W.; Hong, H.D.; Lee, K.T. Protective effect of exopolysaccharide fraction from Bacillus subtilis against dextran sulfate sodium-induced colitis through maintenance of intestinal barrier and suppression of inflammatory responses. Int. J. Biol. Macromol. 2021, 178, 363–372. [Google Scholar] [CrossRef] [PubMed]
  276. Deepak, V.; Sundar, W.A.; Pandian, S.R.K.; Sivasubramaniam, S.D.; Hariharan, N.; Sundar, K. Exopolysaccharides from Lactobacillus acidophilus modulates the antioxidant status of 1,2-dimethyl hydrazine-induced colon cancer rat model. 3 Biotech 2021, 11, 225. [Google Scholar] [CrossRef]
  277. Li, F.; Jiao, X.; Zhao, J.; Liao, X.; Wei, Y.; Li, Q. Antitumor mechanisms of an exopolysaccharide from Lactobacillus fermentum on HT-29 cells and HT-29 tumor-bearing mice. Int. J. Biol. Macromol. 2022, 209, 552–562. [Google Scholar] [CrossRef] [PubMed]
  278. Mahmoud, M.G.; Selim, M.S.; Mohamed, S.S.; Hassan, A.I.; Abdal-Aziz, S.A. Study of the chemical structure of exopolysaccharide produced from streptomycete and its effect as an attenuate for antineoplastic drug 5-fluorouracil that induced gastrointestinal toxicity in rats. Anim. Biotechnol. 2020, 31, 397–412. [Google Scholar] [CrossRef] [PubMed]
  279. Thananimit, S.; Pahumunto, N.; Teanpaisan, R. Characterization of Short Chain Fatty Acids Produced by Selected Potential Probiotic Lactobacillus Strains. Biomolecules 2022, 12, 1829. [Google Scholar] [CrossRef]
  280. Al-Qadami, G.H.; Secombe, K.R.; Subramaniam, C.B.; Wardill, H.R.; Bowen, J.M. Gut Microbiota-Derived Short-Chain Fatty Acids: Impact on Cancer Treatment Response and Toxicities. Microorganisms 2022, 10, 2048. [Google Scholar] [CrossRef]
  281. Rauf, A.; Khalil, A.A.; Rahman, U.U.; Khalid, A.; Naz, S.; Shariati, M.A.; Rebezov, M.; Urtecho, E.Z.; de Albuquerque, R.D.D.G.; Anwar, S.; et al. Recent advances in the therapeutic application of short-chain fatty acids (SCFAs): An updated review. Crit. Rev. Food Sci. Nutr. 2022, 62, 6034–6054. [Google Scholar] [CrossRef]
  282. Kang, J.; Sun, M.; Chang, Y.; Chen, H.; Zhang, J.; Liang, X.; Xiao, T. Butyrate ameliorates colorectal cancer through regulating intestinal microecological disorders. Anticancer Drugs 2023, 34, 227–237. [Google Scholar] [CrossRef] [PubMed]
  283. Shuwen, H.; Yangyanqiu, W.; Jian, C.; Boyang, H.; Gong, C.; Jing, Z. Synergistic effect of sodium butyrate and oxaliplatin on colorectal cancer. Transl. Oncol. 2023, 27, 101598. [Google Scholar] [CrossRef] [PubMed]
  284. Hou, H.; Chen, D.; Zhang, K.; Zhang, W.; Liu, T.; Wang, S.; Dai, X.; Wang, B.; Zhong, W.; Cao, H. Gut microbiota-derived short-chain fatty acids and colorectal cancer: Ready for clinical translation? Cancer Lett. 2022, 526, 225–235. [Google Scholar] [CrossRef]
  285. Singh, N.K.; Beckett, J.M.; Kalpurath, K.; Ishaq, M.; Ahmad, T.; Eri, R.D. Synbiotics as Supplemental Therapy for the Alleviation of Chemotherapy-Associated Symptoms in Patients with Solid Tumours. Nutrients 2023, 15, 1759. [Google Scholar] [CrossRef]
Figure 1. Diagrammatic representation of probiotic-derived bioactive compounds with antiproliferative properties against CRC.
Figure 1. Diagrammatic representation of probiotic-derived bioactive compounds with antiproliferative properties against CRC.
Microorganisms 11 01898 g001
Figure 2. Cytotoxic effects of bacteriocins and nonribosomal bacterial lipopeptides.
Figure 2. Cytotoxic effects of bacteriocins and nonribosomal bacterial lipopeptides.
Microorganisms 11 01898 g002
Figure 3. SCFAs effects on CRC cells: histone deacetylases (HDACs) inhibition and cell surface G-protein-coupled receptors (GPR) activation, immunomodulation, apoptosis.
Figure 3. SCFAs effects on CRC cells: histone deacetylases (HDACs) inhibition and cell surface G-protein-coupled receptors (GPR) activation, immunomodulation, apoptosis.
Microorganisms 11 01898 g003
Table 2. In vitro effects of exopolysaccharides on CRC cells.
Table 2. In vitro effects of exopolysaccharides on CRC cells.
Probiotic StrainCRC Cell LineEffect/Mode of ActionReference
Lactobacillus spp.
L. acidophilus 10307Caco-2dose-dependent anticancer activity (in both
normoxic and hypoxic conditions), ↑ PPARG,
EPO under normoxia
[172]
HCT-15,
Caco-2
↓ cell proliferation, reduction of membrane integrity,
antioxidative properties (↑ HMOX1),
VEGF and HIF1A, ↑ TIMP3 and HIF2A, ↑ PAI-1 gene
[171]
L. acidophilus 606HT-29activation of autophagic cell death via
Beclin-1, GRP78, and Bak induction
[173]
L. acidophilus DSMZ 20079Caco-2↓ cell proliferation, cell cycle arrest (G0/G1),
morphological changes related to apoptosis
(shrinkage, membrane blebbing),
NF-κB inflammatory pathway inactivation
[168]
L. brevis LB63HT-29time-dependent antiproliferative effect, apoptosis induction (↑ Bax, caspase-3, -9/↓ Bcl-2 and survivin)[163]
L. brevis TD4HT-29dose and time-dependent cytotoxic activity,
apoptosis induction (↑ DNA fragmentation)
[169]
L. casei 01HT-29dose-dependent antiproliferative effect,
reduction of pro-mutagen’s 4-NQO cytotoxicity
[174]
L. casei SB27HT-29↓ cell proliferation, apoptotic morphological changes,
BAD, BAX, CASP3, CASP8
[161]
L. casei strains
(K11, M5, SB27, and X12)
HT-29dose-dependent antiproliferative effects,
cell cycle arrest (G0/G1), apoptotic bodies
formation, ↑ caspase-3
[167]
L. delbrueckii ssp. bulgaricus B3HT-29time-dependent antiproliferative effect, apoptosis induction (↑ Bax, caspase-3, - 9/↓ Bcl-2 and survivin)[163]
L. delbrueckii ssp. bulgaricus DSM 20080Caco-2antioxidative and antitumor properties,
apoptosis induction (↑ BAX, CASP3, CASP8, p53/
BCL2, MCL1, Vimentin)
[165]
L. fermentum YL-11HT-29,
Caco-2
dose-dependent antitumor effect,
nuclear condensation related to apoptosis
[170]
L. helveticus MB2-1Caco-2dose and time-dependent anticancer effect[155]
L. kefiri MSR101HT-29dose-dependent anticancer activity, apoptosis induction (↑ cyt c, Bax, Bad, and caspase-3, -8, -9)[162]
L. paracasei TD3HT-29dose and time-dependent cytotoxic activity,
apoptosis induction (↑ DNA fragmentation)
[169]
L. plantarum-12HT-29↓ cell proliferation, ↑ ROS production, intrinsic apoptotic pathway (↑ Bax, caspase-3, -8, -9/↓ Bcl-2), PCNA inhibition in dose-dependent manner[164]
L. plantarum 70810HT-29dose and time-dependent antitumor effect[154]
L. plantarum GD2HT-29time-dependent antiproliferative effect, apoptosis induction (↑ Bax, caspase-3, -9/↓ Bcl-2 and survivin)[163]
L. plantarum NCU116CT26↓ cell proliferation, ↑ TLR2, c-Jun dependent Fas/FasL-mediated apoptotic pathway[166]
L. plantarum NRRL B- 4496HCT 116,
Caco-2
dose-dependent antitumor activity[153]
L. plantarum WLPL04HT-29dose and time-dependent antitumor effect,
inhibition of E. coli adhesion to HT-29 cells
[157]
L. plantarum YW32HT-29dose and time-dependent anticancer activity[156]
L. rhamnosus E9HT-29time-dependent antiproliferative effect, apoptosis induction (↑ Bax, caspase-3, -9/↓ Bcl-2 and survivin)[163]
Others
Lactococcus garvieae C47Caco-2antioxidant and antitumor activity[158]
Pediococcus acidilactici
NCDC 252
HCT 116dose-dependent antiproliferative activity[160]
Pediococcus pentosaceus M41Caco-2antioxidant and antitumor activity[159]
Yeasts
Kluyveromyces marxianus,
Pichia kudriavzevii
SW-480, HT-29, HCT 116↓ cell proliferation, suppression of AKT-1, JAK-1 and mTOR pathways, apoptosis induction (↓ BCL2/↑ BAX, CASP3, CASP8)[175]
↑: increase or up-regulation, ↓: decrease or down-regulation. Colon cancer cell lines: Caco-2, HCT-15, HT-29, CT26 (mouse epithelial colorectal cell line), HCT 116, SW-480. PPARG: Peroxisome proliferator-activated receptor-gamma gene, EPO: Erythropoietin gene, HMOX1: Hemeoxygenase-1 gene, VEGF: Vascular endothelial growth factor gene, HIF: Hypoxia-inducible factor gene, TIMP3: Tissue inhibitor of metalloproteinase-3 gene, PAI-1: Plasminogen activator inhibitor-1, GRP78: G-protein coupled receptor, NF-κB: Nuclear factor-κB, 4-NQO: 4-nitroquinoline 1-oxide, CASP: caspase gene, MCL1: Myeloid leukemia 1 gene, cyt c: cytochrome c, ROS: Reactive oxygen species, PCNA: Proliferating cell nuclear antigen, TLR2: Toll-like receptor 2, c-Jun: transcription factor, FasL: Fas ligand, AKT-1: AKT serine/threonine kinase 1, JAK-1: Janus kinase, mTOR: mammalian target of rapamycin.
Table 3. In vitro effects of bacteriocins on CRC cells.
Table 3. In vitro effects of bacteriocins on CRC cells.
BacteriocinCRC Cell LineEffect/Mode of ActionReference
Duramycin
(Streptomyces sp.)
Caco-2,
HCT 116,
LoVo
detection of PE on cell surface,
dose- and time-dependent Ca2+ release
[185]
Enterocin 12a
(Enterococcus faecium 12a)
HCT-15dose-dependent antiproliferative activity,
morphological changes related to apoptosis
[200]
Enterocin-A
(Enterococcus faecium por1)
HT-29,
Caco-2
dose-dependent cytotoxic effect, morphological
changes related to apoptosis, cell cycle arrest (G1)
[198]
Heterodimer Enterocin-A + B
(Enterococcus faecium)
HT-29improved cytotoxicity compared to enterocin-B alone,
apoptosis related morphological changes
[199]
Enterocin OE-342
(Enterococcus faecalis OE-342)
HCT 116dose-dependent cytotoxic effect, immunomodulatory
activity, cell cycle arrest (G2/M), morphological
changes related to apoptosis
[206]
Nisin A
(Lactococcus lactis subsp. lactis)
Caco-2,
HT-29
↓ cell proliferation,
loss of plasma membrane integrity
[189]
SW-480dose-dependent cytotoxic effect, intrinsic apoptotic
pathway (↑ Bax/Bcl-2 ratio)
[194]
LS 180, HT-29,
SW48, Caco-2
↓ cell proliferation, anti-metastatic effects
(↓CEA, CEAM6, MMP2F, and MMP9F)
[196]
SW-480dose-dependent cytotoxic effect,
BAX/BCL2 ratio, ↑CASP3, CASP9
[193]
dose-dependent cytotoxic effect, ↓ CCND1[195]
Pediocin PA-1
(Pediococcus acidilactici K2a2-3)
HT-29↓ cell proliferation[202]
Plantaricin BM-1
(Lactobacillus plantarum BM-1)
SW-480,
Caco-2,
HCT 116
dose-dependent cytotoxic effect, morphological
changes related to apoptosis, caspase-dependent apoptosis pathway (PARP-1 cleavage, dysregulation
of TNF, NF-κB, and MAPK signaling pathways)
[204]
Plantaricin P1053
(Lactobacillus plantarum PBS067)
E705dose-dependent cytotoxic effect[205]
↑: increase or up-regulation, ↓: decrease or down-regulation. Colon cancer cell lines: Caco-2, HCT 116, LoVo, HCT-15, HT-29, SW-480, LS 180, SW48, E705. PE: Phosphatidylethanolamine, CEA: Carcinoembryonic antigen gene, CEAM6: Carcinoembryonic cell adhesion molecule 6 gene, MMP2F: Matrix metalloproteinase-2F gene, MMP9F: Matrix metalloproteinase-9F gene, CASP: caspase gene, CCND1: cyclin D1 gene, PARP-1: Poly (ADP-ribose) polymerase-1, TNF: Tumor necrosis factor, NF-κB: Nuclear factor-κB, MAPK: Mitogen-activated protein kinase.
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

Thoda, C.; Touraki, M. Probiotic-Derived Bioactive Compounds in Colorectal Cancer Treatment. Microorganisms 2023, 11, 1898. https://doi.org/10.3390/microorganisms11081898

AMA Style

Thoda C, Touraki M. Probiotic-Derived Bioactive Compounds in Colorectal Cancer Treatment. Microorganisms. 2023; 11(8):1898. https://doi.org/10.3390/microorganisms11081898

Chicago/Turabian Style

Thoda, Christina, and Maria Touraki. 2023. "Probiotic-Derived Bioactive Compounds in Colorectal Cancer Treatment" Microorganisms 11, no. 8: 1898. https://doi.org/10.3390/microorganisms11081898

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