Next Article in Journal
The Cano-eMLST Program: An Approach for the Calculation of Canonical Extended Multi-Locus Sequence Typing, Making Comparison of Genetic Differences Among Bunches of Bacterial Strains
Previous Article in Journal
Transcriptomic Responses to Thermal Stress and Varied Phosphorus Conditions in Fugacium kawagutii
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Food Supplements to Mitigate Detrimental Effects of Pelvic Radiotherapy

1
Interdisciplinary Biosciences Group, Belgian Nuclear Research Centre (SCK•CEN), Boeretang 200, 2400 Mol, Belgium
2
Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020 Antwerp, Belgium
3
Department of Biotechnology, University of Ghent, Coupure Links 653, 9000 Ghent, Belgium
*
Author to whom correspondence should be addressed.
Microorganisms 2019, 7(4), 97; https://doi.org/10.3390/microorganisms7040097
Submission received: 26 February 2019 / Revised: 21 March 2019 / Accepted: 28 March 2019 / Published: 3 April 2019
(This article belongs to the Section Gut Microbiota)

Abstract

:
Pelvic radiotherapy has been frequently reported to cause acute and late onset gastrointestinal (GI) toxicities associated with significant morbidity and mortality. Although the underlying mechanisms of pelvic radiation-induced GI toxicity are poorly understood, they are known to involve a complex interplay between all cell types comprising the intestinal wall. Furthermore, increasing evidence states that the human gut microbiome plays a role in the development of radiation-induced health damaging effects. Gut microbial dysbiosis leads to diarrhea and fatigue in half of the patients. As a result, reinforcement of the microbiome has become a hot topic in various medical disciplines. To counteract GI radiotoxicities, apart from traditional pharmacological compounds, adjuvant therapies are being developed including food supplements like vitamins, prebiotics, and probiotics. Despite the easy, cheap, safe, and feasible approach to protect patients against acute radiation-induced toxicity, clinical trials have yielded contradictory results. In this review, a detailed overview is given of the various clinical, intestinal manifestations after pelvic irradiation as well as the role of the gut microbiome herein. Furthermore, whilst discussing possible strategies to prevent these symptoms, food supplements are presented as auspicious, prophylactic, and therapeutic options to mitigate acute pelvic radiation-induced GI injury by exploring their molecular mechanisms of action.

Graphical Abstract

1. Local Radiotherapy as Pelvic Cancer Treatment

Pelvic cancers are among the most frequently diagnosed cancers worldwide [1], and pelvic radiotherapy is often an essential part of the multimodal therapeutic approaches [2,3]. To comfort the patients during their treatment, conventional fractionation is favored. Fractions are limited to 1.8–2 Gy/day, administered during five consecutive days. A full course may take up to 7 weeks or longer, which results in 50 Gy cumulated dose on average. Although it is widely used, a majority of pelvic cancer survivors suffers from a wide diversity of complications that collectively have been termed pelvic radiation disease [4].

2. Gastrointestinal Complications Associated with Pelvic Radiotherapy

Gastrointestinal (GI) complications may develop as result of the inevitable exposure of healthy intestinal tissue with an increased risk for the terminal ileum, colon, or rectum [5]. As a result, patients undergoing pelvic radiotherapy suffer from a significantly impaired quality of life, which adds an extra burden on top of the cost of health care.
In general, radiation-induced GI toxicity is labelled as acute or chronic clinical manifestations, both having a different pathogenesis. Acute radiotoxicity symptoms (nausea, diarrhea, abdominal pain, and fatigue) develop within the course of treatment or a period up to 90 days thereafter [5,6]. Nausea typically occurs relatively early, whilst diarrhea and abdominal pain become problematic 2–3 weeks after the start of radiotherapy. Although early events are frequently reported, they are generally considered reversible. Nevertheless, these symptoms may affect the patient significantly so that treatment interruption or original treatment plan adaptations may be required, which compromise the efficacy of tumor control.
Symptoms of chronic radiotoxicity (altered intestinal transit, malabsorption [7], and dysmotility, which may progress to intestinal obstruction, fistula formation or intestinal perforation) appear after a latency period of 90 days to many years post-irradiation. They are mainly mediated by complex chronic processes including vascular sclerosis and transmural fibrosis [8]. Delayed events are less commonly reported but also less likely to reverse. Of note, acute and chronic radiation toxicities are not independent events, underlined by the consequential late effect theory saying that late injury is more likely to develop when severe acute toxicity exists [9,10,11]. Overall, radiation-induced GI toxicity is a progressive condition with a substantial long-term morbidity and mortality.

2.1. Mechanisms of Pelvic Radiotherapy-Induced Effects to the Healthy Intestine

Although radiation-induced damage to the healthy intestinal tissue is a dynamic and progressive process, it is artificially divided into mucosal breakdown and subsequently induced inflammation as reviewed by François A. et al. [12]. In the following sections, we provide an overview of the underlying mechanisms of pelvic radiotherapy and its activated processes, highlighting the actions most relevant to discuss interference possibilities by food supplements.

2.1.1. Pelvic Radiotherapy-Induced Breakdown of Mucosal Homeostasis

The healthy intestine is a self-renewing tissue every 3–5 days throughout life [13,14]. This high turnover rate is sustained by a sub-population of stem cells that reside at the crypt base [15]. The resulting daughter cells exit the crypt and move to the amplifying transit compartment where they become precursor or progenitor cells. Subsequently, the cells migrate along the crypt/villus axis and differentiate into three different cell lineages, i.e., epithelial bordering cells, goblet cells, and entero-endocrine cells. The fourth cell lineage of Paneth cells moves back into the crypts [16].
Ionizing radiation mainly interacts with water molecules generating reactive oxygen species (ROS), which are considered as the most reactive molecules and thus capable of damaging DNA and other macromolecules (Figure 1) [17,18,19]. This may result in phenotypic modifications, alterations in downstream biological pathways, cell cycle arrest, and/or cell death by apoptosis, necrosis or mitotic catastrophe [19].
The sensitivity of any particular cell type to ionizing radiation is directly proportional to its mitotic rate and thus indirectly proportional to the extent of its differentiation [20]. Therefore, cells lining the GI tract are particularly vulnerable to the damaging effects of ionizing radiation, with stem cells being most radiosensitive. Within the intestine, two functionally distinct stem cells are distinguished, including Lgr5+ and Bmi1+ cells [21]. Lgr5+ stem cells are highly proliferating thereby maturing into epithelial cells whilst migrating towards the villus top [21,22]. These are highly sensitive to radiation and—within hours—quantitatively ablated by such procedures (Figure 1). The first two days following irradiation, an apoptotic phase, characterized by continuous crypt loss, shrinkage in crypt size, and shortening of the villi during this phase is apparent [23]. In contrast, Bmi1+ stem cells located at higher positions in the crypts are considered as a stem cell reservoir comprising less mitotically active and are thus more radioresistant cells [21]. Interestingly, Bmi1+ stem cells are activated two days after radiation-induced crypt apoptosis [23]. During this regeneration phase, Bmi1+ stem cells dramatically proliferate to repopulate intestinal crypts and villi [24]. The resulting, regenerated crypts are enlarged in size whilst the overall number of crypts decreases [25]. Eventually, during the normalization phase, the size of the crypts and the length of the villi are restored to pre-irradiation conditions [25].
After initial damage of Lgr5+ stem cells residing in the intestinal crypts, a tissue scaring process is initiated. Consequential late effects involve fibroblasts, which are stimulated upon exposure to radiation, which then enhance proliferation and collagen deposition, eventually resulting in tissue fibrosis [11]. Moreover, ionizing radiation is thought to induce a chronic, self-maintained scaring process driving fibrosis. However, during the acute phase, mucosal breakdown is induced through reduced expression of tight junctions and epithelial cell death compromising villous integrity, which leads to the development of inflammation, as described in the following section (Figure 1) [26].

2.1.2. Pelvic Radiotherapy-Induced Inflammation

Ionizing radiation induces an inflammatory profile in healthy intestinal tissue. Due to disruption of the mucosal barrier, bacterial translocation is enabled, thereby triggering inflammation and immune cell recruitment (Figure 1) [12]. In addition, damaged vascular endothelium creates a proinflammatory, prothrombotic, and antifibrinolytic phenotype, which further stimulates increased secretion of cytokines, chemokines, and growth factors [27]. In parallel, the increased expression of adhesion molecules including VCAM-1, ICAM-1, PECAM-1, and selectins E and P are favored [28]. This proinflammatory state activates resident macrophages and enhances early extravasation of polymorphonuclear neutrophils (Figure 1) [29]. The presence of these neutrophils in tissues typically indicates an acute inflammatory profile, and is a hallmark of irradiated tissues [20,30]. Moreover, polymorphonuclear neutrophils exhibit microbicidal activity in case of bacterial translocation after vascular and mucosal breakdown [12]. ROS produced by the so-called neutrophils’ “respiratory burst” are crucial in the first control action of GI infection and inflammation. However, excessive and sustained ROS production damages neighboring healthy cells and tissues, and participates in the progression and the chronicity of radiation injury to the intestinal wall. The primary inflammatory state is then amplified through the additional recruitment and transmigration of monocytes, as well as the activation of resident mast cells (Figure 1) [12]. Both will stimulate the production of proinflammatory and profibrosing mediators such as IL-1β, IL-6, TGF-β, and TNFα (Figure 1) [12]. Importantly, the innate immune system recognizes motifs of bacteria, so called pathogen-associated molecular patterns (PAMPs), through pattern recognition receptors (PRRs) [31]. In addition, damage-associated molecular patterns (DAMPs) consisting of components of collectively dead/dying cells and damaged tissues can also activate PRRs. Among the different PRRs, toll-like receptors (TLRs) were the first family discovered and are best studied in their structure and activation mechanism. After exposure to ionizing radiation, TLRs present on the surface of multiple cell types including immune cells and intestinal epithelial cells (IEC) are reported to play an important role in tissue homeostasis and repair [32]. Of note, the gut microbiome was reported to influence the immune response development through blockage of specific immune checkpoints, which impacts cancer immunotherapy success [33]. The innate immune response characterized by macrophages, neutrophils, and mast cells is supported in a complementary way by the adaptive immune response regulated by B and T lymphocytes.
In general, the adaptive immune response is activated after a few days and necessitates the presentation of a specific antigen by specialized antigen-presenting cells such as dendritic cells (DCs) to B and T lymphocytes. As the rapid innate response develops within minutes after tissue irradiation exposure, micro-environmental changes favor the maturation of DCs. Antigen presentation then stimulates naive CD4+ T cells to differentiate into different T cell subsets such as Th1, Th2, Th17, or Treg cells [34]. Each of them shows a specific profile of cytokine expression assuming various redundant or opposed roles in tissue immune responses [35]. Overall, the continuing immune imbalance may play a significant role in the chronicity of radiation lesions [12].
In conclusion, the response of healthy intestinal tissue to radiation is mediated partly by cell death as well as activation of a strong oxidative and immune-modulated inflammatory component within all different tissue compartments (Figure 1). However, the precise roles of the different resident and attracted immune cells are still vague, as well as the contributions of the innate and adaptive immune responses.

2.2. Pelvic Radiotherapy-Induced Effects on Gut Microbiota

Recently, microbiome research began to boom due to the development of 16S ribosomal RNA gene-sequence-based metagenetic methods resulting in major advantages in characterizing microbial populations [36]. The presence of 1013 bacterial cells in a healthy adult shows the high complexity and diversity of human microbiota [37]. Starting from the nose and mouth up until the rectum, microbiota differ in number and composition, whilst being highly susceptible to influences including the diet. Specifically, anaerobic bacteria represented by the genera Bacteroides, Eubacterium, Bifidobacterium, Peptostreptococcus, Ruminococcus, Clostridium, and Propionibacterium were characterized as the predominant (>109 Colony Forming Units (CFU)/g)) microbiota of the human gut [38]. Multiple functions have been attributed to the gut microbiota including roles in the development of intestinal structure and morphology, metabolism like vitamin production and fermentation, neuroendocrine signaling through the gut-brain axis, as well as protection through immune system development and homeostasis [39,40]. Immune functions are generally based on the interaction of commensal bacteria with TLRs and subsequent NFκB signaling pathway activation. Furthermore, it has been suggested that microbiota have a crucial role in immune tolerance induction since germ-free animals lack this ability with regard to oral tolerance [41,42]. Despite incomplete knowledge regarding the underlying mechanisms, the mucosal microbiota are capable of limiting growth and even kill certain transient microbial pathogens that enter their habitat [43,44,45,46,47].
Changes in the gut microbial ecosystem are associated with dysbiosis, which can be defined in various ways, but generally refers to changes in microbial compositions that cause a drastic imbalance between beneficial and potentially pathogenic bacteria, which renders the gut more vulnerable to pathogenic insults [48]. Moreover, an imbalanced microbiome is characterized by changes in its functionality and metabolic activity, or changes in their local distributions along the GI tract. As a consequence, recent insights have linked gut microbiome dysbiosis with various disease states of which inflammatory bowel disease and cancer are well-known examples [49,50].
Radiation has also been defined as a stressor of the GI microbial ecosystem. Radiation-induced gut microbial dysbiosis was hypothesized to negatively contribute to pelvic radiation disease in cancer patients with associated mucositis, diarrhea, systemic inflammatory response, and fatigue [51,52]. This key role of gut microbiota in radiotherapy-induced GI toxicity has been known for decades. For instance, germ-free mice are known to be resistant to radiation-induced enteritis [53,54,55,56]. In addition, post-radiotherapy changes in gut microbial diversity have been described [57,58]. Additional clinical studies have shown that cancer patients exposed to radiation therapy exhibit marked alterations in gut microbiota composition [51,59,60]. Most frequently, a decrease in potentially protecting Bifidobacterium, Clostridium cluster XIVa, and Faecalibacterium prausnitzii were observed, together with an increase of Enterobacteriaceae and Bacteroides, which might contribute to the development of mucositis, and diarrhea in particular [60]. In addition, patients developing post-radiotherapy diarrhea compared to no diarrhea patients were characterized by an increase in Actinobacteria and Bacilli, as well as a decrease in Clostridia [57].
Notably, gut microbiota are not only affected by radiotherapy, but as importantly, the pre-exposure microbial profile is associated with the susceptibility of developing post-radiotherapy diarrhea [51]. For instance, a pilot study revealed that gut microbial diversity richness, and the Firmicutes/Bacteroidetes ratio were significantly altered prior to pelvic radiotherapy in patients who later developed diarrhea [51]. Unfortunately, the exact microbial biomarkers (specific taxa or biodiversity markers), which could predict this susceptibility have not yet been defined due to the highly individualized and fluctuating microbiome profiles, complex interactions between the host, clinical and environmental factors, as well as the heterogeneity of reported microbiome studies. In addition to biomarker discovery, increasing knowledge about important microbiome features will also allow the exploration of personalized radiotherapy delivery methods.
All this knowledge highlights the importance of re-establishing bacterial homeostasis after pelvic radiation, as well as prevention of dysbiosis induced by radiotherapy. One favorable way consists of the use of natural food supplements, being commonly used because of their long-standing positive association with health. These interventions will be discussed further in this review.

3. Nutritional Interventions for Pelvic Radiotherapy-Induced Side Effects

To maintain a positive balance between the quality of tumor control by pelvic radiotherapy and the damage posed to healthy surrounding intestinal tissue, different strategies—technical and biological—have been developed. Here, we will focus on nutritional interventions including vitamins, prebiotics, and probiotics.

3.1. General Improvements in Gut Health

Vitamins Vitamins are dietary micronutrients acting as cofactors for numerous enzymes essential for health. Among those, vitamins E, A, C, B6, B9, and B12 have been reported as potent GI radioprotectors, and will therefore be discussed here [61].
First, vitamin E is an essential nutrient for all animal species with many biological functions of which its antioxidative property is considered most important [62]. For instance, exogenous provision of vitamin E was reported to attenuate acute mountain sickness characterized by compromised intestinal integrity, with physical improvements of mountaineers at high altitudes [63,64,65].
Vitamin A derivatives are known for their pleiotropic effects on human health and to modulate several biological processes including intestinal immune and barrier functions [66,67,68]. Interestingly, supplementation has been shown to reduce infant mortality due to diarrhea in endemic areas of vitamin A deficiency [69].
Vitamin C (ascorbic acid) is broadly used as antioxidant and essential for normal cell function, growth and development and thus indispensable for human health and well-being [70,71]. For instance, vitamin C impacts wound healing processes through stimulation of collagen biosynthesis and early resolution of inflammation and tissue remodeling [72]. In view of this data, pre-operative vitamin C supplementation was shown to improve colorectal anastomotic healing and biochemical parameters in malnourished rats [73].
Finally, vitamin B6 (pyridoxine), B9 (folate), and B12 (cobalamin) are provided by food or retrieved from commensal bacteria that produce them in the gut. Generally, vitamin B6 is involved in various homeostatic processes in health and disease, including host immune responses [74]. Since their bioavailability is dependent on the correct function of the GI tract, vitamins B9/B12 deficiencies are associated with GI diseases including inflammatory bowel disease. Vitamin B9 supplementation on chemically-induced intestinal stress has yielded contradictory results [75,76].
Prebiotics In 2017, the International Scientific Association of Probiotics and Prebiotics (ISAPP) updated the definition and scope of prebiotics to “a substrate that is selectively utilized by host microorganisms conferring a health benefit” [77].
Initially, oligosaccharides were considered as the main prebiotic source, with fructans (fructooligosaccharides (FOS) and inulin) and galactans (galactooligosaccharides (GOS)) in particular. Their effects on gut microbiota have been extensively investigated. For instance, preclinical studies uniformly reported changes in specific gut microorganisms of diet-induced obese and diabetic animals following prebiotic supplementation. Specifically, a decreased Firmicutes/Bacteroidetes ratio as well as the proportion of Tenericutes, Cyanobacteria, and Verrucomicrobia was observed. Moreover, this impacted several important metabolites implicated in metabolic disorders [78,79]. This effect was confirmed in obese women [80]. In addition to their metabolic influence, dietary FOS and GOS were reported to affect fecal microbiota by increasing Bifidobacteria abundance, which alleviated functional constipation in constipated subjects [81,82,83].
Another class of candidate prebiotics is polyphenolic phyco- and phytochemicals including anthocyanins, flavonoids, tannins, and lignins amongst others, which are highly enriched in cyanobacteria, plants, and herbs [84]. Most of them (95–99%) are not absorbed in the small intestine and reach the colon where they are subjected to microbial biotransformations producing bioactive metabolites [85]. Overall, these polyphenol-derived metabolites are possibly responsible for health benefits due to their ability to modulate intestinal bacterial populations [86]. This two-way interaction between an individual’s gut microbiota and polyphenols to determine health effects is well-established [87,88,89]. For instance, the prebiotic potential of cocoa flavanols was assessed by a cross-over intervention study in healthy volunteers, which reported increased Bifidobacteria and Lactobacilli, and decreased Clostridia counts [90]. Other studies reported the gut microbiota shaping potential of polyphenols and enriched food, which further promote health benefits related to inflammation and obesity [78,91,92,93].
Probiotics Recently, ISAPP strengthened the FAO/WHO definition of probiotics: “live microorganisms that, when administered in adequate amounts, confer a health benefit on the host” [94]. The concept of probiotics is becoming increasingly popular, as evidenced by rapidly expanding research highlighting its beneficial effects, with lactic acid bacteria from the genera Lactobacillus and Bifidobacterium being the major representatives. General intestinal health effects including intestinal barrier improvements, inhibition of (opportunistic) pathogens or stimulation of beneficial microorganisms, and reduction of intestinal inflammation profiles were documented, in healthy and diseased animals and humans. A sample of in vitro and in vivo research is summarized in Table 1.
In addition to general intestinal health support, there is also convincing evidence supporting the efficacy of probiotics in the treatment of acute diarrhea, especially in children with rotavirus infection, likely attributed to the stimulation of IgA-specific antibody secreting cells [95,96]. Frequently studied probiotics in that field are L. rhamnosus GG and Saccharomyces boulardii. Guarino, Guandalini, and Lo Vecchio (2015) reviewed the modern perspective on the potential use of probiotic agents in the management of infectious diseases, with special emphasis on diarrheic symptoms [97].

3.2. Preclinical Evidence for Reduction of Radiation-Induced GI Toxicity

Since food supplements have been reported to improve general gut health as well as diarrheal diseases, it is of interest to investigate whether they can preserve surrounding, healthy tissues during radiotherapy and reduce radiation-induced GI toxicities. Both preclinical and clinical studies exploring their radioprotective properties are discussed in the following sections (summarized in Table 2 and Table 3).
Vitamins Since ionizing radiation produces free radicals, antioxidative vitamins have been explored for their radioprotective potential [118]. For instance, dietary vitamin E (alpha tocopherol) pre-treatment preserved general small intestinal morphology of rodents against high doses of radiation [119,120,121]. Notably, the starting point of diet supplementation matters as lethality was only mitigated when antioxidant vitamin E supplementation was started 24 h after 8 Gy whole-body irradiation [122].
Pyridoxamine, one of the natural forms of vitamin B6, was shown to prevent apoptosis in small intestinal epithelium of mice exposed to 4 or 8 Gy whole body irradiation [123]. Preclinical, radioprotective effects of vitamins B9/B12 on the GI tract have not yet been reported.
Radioprotection by vitamin A is questionable. For instance, vitamin A had no effect on clinical parameters and could not protect mice against 7–10 Gy whole body irradiation-induced changes in intestinal sugar transport [124]. However, a combined cocktail of antioxidant vitamins A, C, and E before irradiation protected the absorptive function of the small intestine when tested in an identical experimental setup by the same research group [125].
Improved survival was reported with oral vitamin C pre-treatment, either by preventing small intestinal apoptosis in mice exposed to 14 Gy whole-body irradiation [126] or by reducing inflammatory cytokines and free radical metabolites after 7–8 Gy whole-body irradiation of mice [127]. In addition, vitamin C protected rats form radiation-induced liver damage through activation of the antioxidant defense system including glutathione-s-transferase (GST), superoxide dismutase (SOD) and catalase (CAT) [128].
Prebiotics Whilst oligosaccharides have not yet been tested for their radioprotective applications in preclinical settings, several flavonoids have been identified as potent radioprotectants [129,130]. For instance, radioprotective capacities of bioflavonoid propolis were seen in mice exposed to 9 Gy whole-body irradiation. This effect was confirmed by diminished primary DNA damage in leukocytes as well as delayed onset of mortality [131,132].
Interestingly, prebiotics can be delivered locally by probiotic bacteria. For instance, it was suggested that L. reuteri strain 121 produces prebiotic FOS and is therefore able to stimulate beneficial intestinal microbiota [133]. In addition, increased fecal abundance of butyrate-producing bacteria was reported to be associated with increased resistance against respiratory viral infections [134]. Alternatively, cyanobacterium Arthrospira platensis products (Spirulina) are rich in vitamins A, B6, B9, E, and C as well as polyphenolic phycochemicals, and have been shown to protect murine bone marrow cells exposed to γ rays [135].
ProbioticsTable 2 provides an overview of preclinical studies investigating the effects of probiotic supplementation on radiation-induced GI toxicities. Of interest, although many health benefits of probiotics appear to be strain-specific [136], little variation is seen in the types of bacteria applied as probiotics in radiation research, with predominant use of Lactobacilli. This implies that Lactobacilli could have some core properties, which mediate protection against radiation-induced GI complications. To illustrate, preventive effects towards radiation-induced GI toxicities were seen with Microflorana®-F, an active probiotic mixture composed of L. acidophilus, L. helveticus, and Bifidobacterium spp., which improved both overall survival as well as bacterial contamination, even after abdominal exposure to 20 Gy [137]. In addition, improvements of small intestinal morphology were observed with probiotic L. acidophilus (strain not mentioned) supplementation in rats locally irradiated with ≤15 Gy [138]. Furthermore, reduced epithelial apoptosis and improved crypt survival was demonstrated upon probiotic L. rhamnosus GG ATCC 53103 supplementation without causing drastic shifts in bacterial compositions, following 12 Gy whole-body γ radiation [139]. Even more interesting, these effects of L. rhamnosus GG ATCC 53103-mediated small intestinal radioprotection extended to fractionated dosing regimens [140]. Besides preventive applications, therapeutic effects of L. delbrueckii subspecies bulgaricus B3 strain were observed in rats following a dose of 11 Gy directed to the abdominal-pelvic area [141,142]. Finally, L. plantarum 299v was shown to reduce GI injury and inflammation in rats exposed to 10 Gy lower abdominal radiation and even improved colonic anastomotic healing [143].

3.3. Clinical Evidence for Reduction of Radiation-Induced GI Toxicity

Vitamins Due to preclinical success and overall safety, vitamins may be investigated for their radioprotective potential in clinical settings. A pilot study investigating vitamin E and C supplementation in prostatic or gynecologic cancer patients exposed to radiotherapy was shown to improve diarrhea and other symptoms, despite rectal pain. Furthermore, continuation of this supplementation up to 1 year resulted in sustained improvements of the symptoms [144]. To our knowledge, there have been no studies performed on the sole effect of vitamin A, B6, B9, or B12 in clinical settings of pelvic radiotherapy.
Prebiotics A recent, placebo-controlled clinical trial investigated oligosaccharide supplementation in patients with gynecological cancers who received radiotherapy with a cumulated dose of 52.2 Gy after surgery [145]. Although radiotherapy is well-reported to cause a decrease in fecal Lactobacillus and Bifidobacterium counts, administration of this prebiotic supplement was able to improve the recovery of both genera afterwards.
Although most of the data provide preclinical evidence, human clinical studies investigating polyphenolic phyco- and phytochemicals are scarce and often controversial, partly due to inter-individual variability in responsiveness.
ProbioticsTable 3 summarizes evidence, which exists on probiotic applications in cancer patients exposed to radiotherapy, with a focus on the reduction of diarrhea and viral gastroenteritis [146]. Meanwhile, a meta-analysis and several randomized clinical trials have studied a plurality of probiotics and their therapeutic and preventive effects in radiotherapy-exposed patients [146]. Most of these trials primarily focused on radiotherapy-induced diarrhea and its effects on lower parts of the GI tract [147]. For instance, Delia et al. (2002, 2007) organized multiple clinical trials investigating a high-potency probiotic preparation VSL#3® composed of four strains of Lactobacilli (L. paracasei BP07, L. plantarum BP06, L. acidophilus BA05, and L. delbruekii subsp. bulgaricus BD08), three strains of Bifidobacteria (B. longum BL03, B. breve BB02, and B. infantis BI04), and Streptococcus salivarius subsp. thermophiles BT01. Results suggested the effectiveness of VSL#3® in preventing the occurrence of diarrhea in patients submitted to radiotherapy with direct and indirect improvements of their quality of life, as well as a good tolerance [148,149]. Other probiotics were also reported to have various beneficial effects on radiotherapy-induced diarrheic complications [150,151,152,153,154,155,156,157].
All this evidence as listed in Table 3 suggests that probiotics may have a beneficial role in prevention of radiation-induced diarrheic symptoms. However, most studies could only report modest effects, without affecting all diarrheic parameters including stool frequency, consistency, bowel movements, and the needs for anti-diarrheal medications simultaneously. In addition, no overall improvements in all study participants were reported suggesting inter-individual variability for probiotics’ success. Moreover, limited underlying modes of action or objective observations of protection against radiation-induced GI injuries have been described. Of note, the absence of serious adverse events reported in clinical trial reports should be interpreted with caution, especially since systemic infections associated with specific probiotic intake have been described elsewhere. For example, cases reporting sepsis or endocarditis and bacteremia following L. rhamnosus GG and L. casei, supplementation were recorded by Doron and Snydman (2015) [158]. Bifidobacteria are rarely associated with negative effects, yet bacterial sepsis and cholangitis were reported due to Bacillus subtilis intake [159]. Overall predisposing risk factors include immunosuppression, prior hospitalization, severe underlying comorbidities, previous antibiotic therapy, prior surgical interventions, critically ill patient status, and central venous catheters. Another study showed an increased risk of mortality associated with enteral administration of a multispecies probiotic preparation to prevent infectious complications in patients with predicted severe acute pancreatitis [160]. Therefore, cautious intake of probiotics is encouraged, especially in critically ill patients or in those at risk of non-occlusive mesenteric ischemia.

3.4. Mechanisms of Radioprotection

There is only limited knowledge on the underlying mechanisms by which food supplements elicit their radio-protective effects. In the case of probiotics, general modes of action for Lactobacillus have been reviewed by Lebeer et al. [136,161]. In summary, its probiotic application assumes that the mechanisms underlying health-promoting capacities include pathogen inhibition and restoration of microbial homeostasis through microbe-microbe interactions, enhancement of epithelial barrier function, and modulation of immune responses. However, given the complexity of these three main functions, more experimental work is still required.

3.4.1. Antimicrobial Capacities

Vitamins Interestingly, vitamin A has been associated with antiviral efficacy towards murine norovirus, acting through a significant increase in relative abundance of Lactobacillus sp. [162,163]. In addition, vitamin B6 was shown to eliminate Salmonella Typhimurium from the gut [164]. However, to our knowledge, antimicrobial actions for vitamins E, C, B9, and B12 have not yet been reported.
Prebiotics Although prebiotics are—according to their definition—selectively used by host microorganisms to confer a health benefit, no direct antimicrobial actions have been reported. However, prebiotic research showed stimulated mucus production by Goblet cells and competitive interference with enteropathogens [165,166,167,168].
Probiotics Probiotics were suggested to beneficially affect the host through direct effects on microbiota. Traditionally, probiotics—like commensal bacteria—can exert anti-pathogenic activities through nutrient competition, production of antimicrobials and/or competitive exclusion. For instance, metabolic cross-feeding is extremely interesting when Lactobacilli-produced lactic acid is converted into butyric acid by butyrate-producing colonic bacteria since butyrate exerts intestinal health-promoting capacities [169,170,171,172,173,174]. In addition to the general antimicrobial metabolite lactic acid, many probiotics may also produce bacteriocins, which are strain-specific peptides with direct antimicrobial activity either through membrane permeabilization, pH lowering, and/or capture of elements essential for microbial growth [175,176,177,178,179,180,181,182]. Furthermore, some L. reuteri strains produce additional antimicrobial reuterin [183]. Recently, oral administration of probiotic L. casei CRL 431 and L. paracasei CNCM I-1518 was described to increase Paneth cells as well as intestinal antimicrobial activity [184]. As Paneth cells contribute to small intestinal remodeling following whole-body γ irradiation, they should be explored in further detail for probiotic-induced radio-protective evidence [185]. Interestingly, antibacterial activity of probiotic L. rhamnosus GG and L. acidophilus LA towards Salmonella Typhimurium C5 was observed, both in vitro and in germ-free mice, without affecting the normal gut microbiota [186,187]. Besides nutrient competition, probiotics may compete for binding to host mucosa and thereby exclude intestinal pathogens and thus prevent bacterial translocation [188]. Intermicrobial binding or co-aggregation is thought to be another possible mechanism by which clearance of pathogens during mucus flushing is enhanced [189,190]. Whether these intermicrobial interactions by probiotics are able to reduce infections in vivo, remains to be largely determined.

3.4.2. Barrier-Enhancing Capacities

Vitamins Antioxidative capacities of vitamin C were reported to significantly reduce the complaints of breast cancer patients one year post-surgery [191]. In parallel, antioxidative actions of vitamin E as well as vitamins B6 and B9/B12 were shown by reduced oxidative stress when rats’ intestinal barrier was subjected to different stressors [64,123,192].
Barrier protection by vitamin A was proposed by enhanced mucin synthesis [67,168]. In addition, vitamin A might regulate and maintain the intestinal epithelial barrier through IL-22 [193]. Specifically, retinoic acid, the most biological form of vitamin A, was reported to promote tolerance and reduce inflammation in rodent colitis models [194,195]. Other studies have described vitamin A as a stimulating agent for migration and proliferation of IECs whilst balancing the gut microbiome [193,196].
A combined cocktail of vitamins A, C, and E before irradiation was reported to reduce oxidative stress in small intestinal crypt cells, which then protected absorptive transport of sugars, amino acids, bile acids, and peptides [124,197]. Still, many gaps remain in our understanding of vitamin interactions with gut microbiota, mucosal immunity, and intestinal barriers, which are today’s research subjects.
Prebiotics Similar to vitamins, prebiotics are studied for their antioxidant capacities to delay or prevent radiation-induced oxidative stress and consequent mucosal damage. Despite limited knowledge about prebiotic oligosaccharide antioxidant actions, FOS antioxidative and hepato-protective effects were observed in mice that were chronically exposed to oxidative stress mimicking accelerated ageing [198]. Furthermore, supplementation of FOS to constipated nursing-home elderly residents reduced lipid peroxidation levels, likely mediated by colonic bacteria in humans [199]. On the contrary, a large number of phytochemicals were reported to possess antioxidant properties by averting the continuation of the free radical chain reaction upon contact [200]. In humans, a phytonutrient mix tested as an ageing defense mechanism in UV radiation-induced skin damage was evaluated positively as it provided antioxidant protection and cellular repair as well as modulated inflammation [201].
Although the role of prebiotic oligosaccharides in barrier enhancement through tight junctions is rather disputable [202,203,204], in vitro research could show induced transcription and assembly of tight junction proteins following incubation with the most abundant flavonoid quercetin [205,206]. Unfortunately, their applications as protectants of the intestinal mucosal barrier in radiation-induced GI toxicity have not yet been investigated.
Probiotics A possible way by which probiotics may exert radioprotection is through intestinal barrier enhancing modalities. For instance, safe and natural antioxidant additives that delay or prevent radiation-induced oxidative stress and consequent damage are of high interest. Probiotic antioxidant actions can be obtained through their metal ion chelating capacities [207,208]. Besides this, exopolysaccharides (EPS) of probiotic lactic acid bacteria have shown promising biological activities including antioxidant, antitumor, anti-biofilm formation by pathogens, and immunomodulation [209,210,211,212,213,214,215]. Furthermore, potential roles in stress resistance, adhesion, colonization, and host-bacteria interactions have been suggested [216,217,218]. Animal studies investigating the effects of feeding EPS-producing Lactobacilli demonstrated gut health benefits in part through altered gut microbiota composition and functioning, as well as favored antioxidant activities [219,220]. Another defense approach involves stimulation of the complex inherent antioxidant enzymatic systems, including SOD, CAT GST, glutathione peroxidase (GSH-Px) and glutathione reductase (GR), which are present in both the host and probiotic bacteria. Despite promising, preliminary results, therapeutic application of antioxidant enzymes is limited due to its short circulatory half-life, which restricts its bioavailability. Consequently, probiotics are now being exploited for their capacity of local antioxidant enzyme delivery. In the past, engineered L. casei BL23 strains producing SOD and CAT were shown to reduce or even prevent intestinal pathologies caused by ROS [221,222]. Recent comparative genome analysis suggests that also endogenous enzymatic activities could be used, with SOD being specific for L. paracasei strains and CAT for L. casei strain [223]. Similar beneficial observations of specific probiotic strains carrying antioxidant enzymes may also apply for cases of radiation-induced oxidative stress in the intestine. Alternatively, food supplements can also stimulate the host’s defense system through efficient activation of antioxidant actors, which was proven effective for a variety of probiotic bacteria tested in vivo [224,225,226,227,228,229,230]. On the other hand, probiotics can also produce diverse, non-enzymatic antioxidant metabolites such as glutathione (GSH), short chain fatty acids (SCFAs), and vitamin B9 [104,161]. This is highly interesting as radiation induces GSH depletion whilst maintaining ROS levels. This then increases the host’s susceptibility to oxidative tissue damage and hinders recovery of intestinal mucosa [231,232,233]. Unfortunately, it is difficult to increase circulating glutathione to a clinically beneficial amount through oral administration [234] and the applicability of GSH precursors (e.g., N- acetylcysteine) is limited by their toxic side effects [235,236]. To our knowledge, no studies in the context of intestinal radiation injury have been performed investigating probiotics that locally produce radio-protective GSH. Another feature shared by many probiotics is the ability of locally producing SCFAs of which butyrate has received particular attention for its beneficial effects on intestinal health [169,237]. For instance, butyrate was reported to stimulate a variety of colonic mucosal functions such as inhibition of inflammation and carcinogenesis, re-establishment of its defense barrier and decreasing oxidative stress whilst positively balancing gut microbiota [170,171,172,238]. For these reasons, lactate- and butyrate-producing bacteria are extremely interesting in the context of intestinal radiation injury, despite conflicting data obtained by randomized trials organized so far [173,174]. In addition, folate-producing Bifidobacterium strains tested in animal and human trials confirmed folate production and absorption upon intake, as well as enhanced fecal folate levels, without further investigations of their intestinal effects [239,240].
Radiation also negatively affects the expression of tight junctions-related proteins in an intestinal segment specific fashion contributing to the impairment of intestinal barrier integrity [26,241,242,243]. Both commensal bacteria and probiotics have been shown to enhance and restore intestinal barrier integrity in vitro and in vivo through tight junction regulation [244,245,246,247,248,249,250,251]. Specifically, probiotic L. plantarum WCFS1-mediated activation of TLR2 was reported to improve the intestinal barrier through changes in tight junction expression in human tissue [111]. A final intestinal barrier protection mechanism involves the mucus layer that is affected by external beam irradiation [252,253]. Adherence to IECs and mucosal surfaces has been suggested to be an important qualification for colonization of bacteria along the GI tract providing a competitive advantage in this ecosystem [161,254]. As such, probiotics can also minimize biofilm formation in which pathogens can aggregate [255]. Researchers have shown mucus production to be stimulated by Goblet cells as well as competitive interference with enteropathogens through the use of probiotics [256,257,258,259,260,261]. However, their applications as protectants of the intestinal mucosal barrier in radiation-induced GI toxicity have not yet been widely investigated.

3.4.3. Immunomodulatory Capacities

Vitamins Although vitamins mainly exert radioprotective potential through antioxidant mechanisms, they have been shown to modulate the immune response, likely mediated by TLRs [194]. For instance, survival of 10 to 12 Gy whole-body irradiated mice was enhanced by vitamin E inhibiting pro-inflammatory cytokines IL-1β and -6, and reducing apoptosis in IEC [262]. In addition, vitamins B6 and B9 were shown to ameliorate the severity of murine colitis through reduction of Salmonella Typhimurium loads or augmentation of cellular methylation, respectively [263].
Prebiotics Generally, little is known about immunomodulatory capacities of prebiotics in humans, and even less is reported in clinical radio-therapeutic settings. However, some preclinical studies showed possible immunosuppressive roles in cases of inflammatory bowel diseases [91,203,264]. The natural bioflavonoid propolis revealed successful, protective effects when mice were exposed to 9 Gy of γ rays, through an augmented activity of multiple immune cells [131,132].
Probiotics Finally, current probiotic research is extremely interested in its ability to influence the immune response evoked by radiation. To this regard, probiotic interventions with Lactobacilli and/or Bifidobacteria have been shown to influence immune responses, likely mediated by TLRs [139,140,265,266,267,268,269]. Applications during radiotherapy showed, for example, that diarrhea could be prevented by live L. acidophilus plus B. bifidum (Infloran®), likely because of an improved immune status of the patients involved [152].

4. Discussion

With cancer survival rates improving over recent years, more patients are frequently suffering from treatment-induced side effects. For instance, cancer patients have reported GI symptoms due to pelvic radiotherapy, which induces injuries in healthy surrounding tissues. Unfortunately, our understanding of the biological complexities that underlie GI toxicity of healthy intestinal tissues initiated by pelvic radiotherapy is still limited. In all, there is still a long way ahead to provide patients with actual, effective prevention or treatment.
Current advances in linking the pathobiology of radiation-induced GI symptoms with gut microbial dysbiosis have encouraged research on food supplements as promising actors for prevention and/or treatment of these symptoms. The scientific basis for studies applying dietary interventions in humans can be found in a large number of animal experiments that have identified potential pathophysiological mechanisms in the small and large intestine. For instance, interventional, preclinical trials have shown that some of these mechanisms can be blocked, leading to a significant reduction in radiation-induced GI damage.
Given their safe character, food supplements like probiotics, prebiotics, and vitamins are very attractive to investigate for their potential in modulating the intestinal response to pelvic irradiation. In addition, research has shown evidence for their barrier enhancing, immunomodulatory and intermicrobial activity. Initial attempts in applying food supplements appear safe in the context of radiation-induced diarrhea, although the patient’s health status (e.g., immunocompromised patients) can affect this response. In addition to these negative complications of food supplement use, there is the issue of high inter-individual variability for its success. In future scenarios, patients may be screened to characterize their microbiome profile to subscribe the most appropriate food supplement, or a symbiotic combination, to the patient.
Altogether, future research investigating the effects of radiation on the GI tract and the microbiome is warranted. First, it is important to stress that applied single fraction radiotherapy does not adequately reproduce the conditions of fractionated radiotherapy as used in patients. Therefore, the acute induced GI toxicity studied in experimental models may not fully represent clinical outcomes of patients. In addition, findings obtained from animal studies remain to be translated to diagnostic, prophylactic, or therapeutic treatments for humans. One caveat concerns a potential different radiosensitivity between animals and humans. Another hurdle is that microbiota members differ not only among host species but also between individual host organisms, and fluctuate in time. In addition, diet and different treatments (e.g., chemotherapy, surgery) are important confounding factors that differentiate and modulate the composition of the gut microbiota in an individual over time. Furthermore, it remains unclear whether an altered microbiome associated with a disease in humans causes, contributes to or merely is a consequence of the disease state. Hence, it will be a major challenge to explore strategies to translate the experimental data into a clinical therapy. To make further progress within the field of food supplement research, a number of basic issues still needs to be addressed, including optimal strain characterization together with quality control, dose optimization, clear definition of the desired effects, as well as (combined) formulation. Then, study endpoints—other than diarrhea—have to be defined accurately and a clinically meaningful study duration described. In parallel, regulatory aspects have to be clarified and appropriate guidelines for the evaluation of food supplements, whether as food or drug, need to be developed. Although the road of developing a good food supplement for radiotherapy patients seems long, promising evidence gathered from (pre)clinical studies should be a scientist’s driving force.

Author Contributions

C.S. wrote the manuscript with input from all the authors. M.V. and F.M conceived the original idea.

Funding

This research received no external funding.

Acknowledgments

C.S. was supported by the Belgian Nuclear Research Centre, SCK•CEN, through a PhD AWM Grant.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jemal, A.; Bray, F.; Center, M.M.; Ferlay, J.; Ward, E.; Forman, D. Global cancer statistics. CA Cancer J. Clin. 2011, 61, 69–90. [Google Scholar] [CrossRef] [PubMed]
  2. Haddock, M.G.; Sloan, J.A.; Bollinger, J.W.; Soori, G.; Steen, P.D.; Martenson, J.A. Patient assessment of bowel function during and after pelvic radiotherapy: Results of a prospective phase III North Central Cancer Treatment Group clinical trial. J. Clin. Oncol. 2007, 25, 1255–1259. [Google Scholar] [CrossRef] [PubMed]
  3. Hogan, N.M.; Kerin, M.J.; Joyce, M.R. Gastrointestinal complications of pelvic radiotherapy: Medical and surgical management strategies. Curr. Probl. Surg. 2013, 50, 395–407. [Google Scholar] [CrossRef]
  4. Andreyev, H.J.N.; Wotherspoon, A.; Denham, J.W.; Hauer-Jensen, M. Defining pelvic-radiation disease for the survivorship era. Lancet Oncol. 2010, 11, 310–312. [Google Scholar] [CrossRef]
  5. Hauer-Jensen, M.; Denham, J.W.; Andreyev, H.J.N. Radiation enteropathy–pathogenesis, treatment and prevention. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 470–479. [Google Scholar] [CrossRef] [PubMed]
  6. Ahlberg, K.; Ekman, T.; Gaston-Johansson, F. The experience of fatigue, other symptoms and global quality of life during radiotherapy for uterine cancer. Int. J. Nurs. Stud. 2005, 42, 377–386. [Google Scholar] [CrossRef] [PubMed]
  7. Husebye, E.; Hauer-Jensen, M.; Kjorstad, K.; Skar, V. Severe late radiation enteropathy is characterized by impaired motility of proximal small intestine. Dig. Dis. Sci. 1994, 39, 2341–2349. [Google Scholar] [CrossRef]
  8. Denham, J.W.; Hauer-Jensen, M. The radiotherapeutic injury—A complex “wound”. Radiother. Oncol. 2002, 63, 129–145. [Google Scholar] [CrossRef]
  9. Peters, L.J.; Ang, K.K.; Thames, H.D.J. Accelerated fractionation in the radiation treatment of head and neck cancer. A critical comparison of different strategies. Acta Oncol. 1988, 27, 185–194. [Google Scholar] [CrossRef]
  10. Bourne, R.G.; Kearsley, J.H.; Grove, W.D.; Roberts, S.J. The relationship between early and late gastrointestinal complications of radiation therapy for carcinoma of the cervix. Int. J. Radiat. Oncol. Biol. Phys. 1983, 9, 1445–1450. [Google Scholar] [CrossRef]
  11. Dörr, W.; Hendry, J.H. Consequential late effects in normal tissues. Radiother. Oncol. 2001, 61, 223–231. [Google Scholar] [CrossRef]
  12. François, A.; Milliat, F.; Guipaud, O.; Benderitter, M. Inflammation and immunity in radiation damage to the gut mucosa. BioMed Res. Int. 2013, 2013, 123241. [Google Scholar] [CrossRef] [PubMed]
  13. Simons, B.D.; Clevers, H. Stem cell self-renewal in intestinal crypt. Exp. Cell Res. 2011, 317, 2719–2724. [Google Scholar] [CrossRef] [PubMed]
  14. Darwich, A.S.; Aslam, U.; Ashcroft, D.M.; Rostami-Hodjegan, A. Meta-analysis of the turnover of intestinal epithelia in preclinical animal species and humans. Drug Metab. Dispos. 2014, 42, 2016–2022. [Google Scholar] [CrossRef]
  15. Umar, S. Intestinal stem cells. Curr. Gastroenterol. Rep. 2010, 12, 340–348. [Google Scholar] [CrossRef] [PubMed]
  16. Bjerknes, M.; Cheng, H. The stem-cell zone of the small intestinal epithelium. I. Evidence from Paneth cells in the adult mouse. Am. J. Anat. 1981, 160, 51–63. [Google Scholar] [CrossRef]
  17. Banerjeea, S.; Aykin-Burnsa, N.; Kragera, K.J.; Shaha, S.K.; Melnykb, S.; Hauer-Jensena, M.; Pawar, S.A. Loss of C/EBPδ enhances IR-induced cell death by promoting oxidative stress and mitochondrial dysfunction. Free Radic. Biol. Med. 2016, 99, 296–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  18. Mikkelsen, R.B.; Wardman, P. Biological chemistry of reactive oxygen and nitrogen and radiation-induced signal transduction mechanisms. Oncogene 2003, 22, 5734–5754. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Desouky, O.; Ding, N.; Zhou, G. Targeted and non-targeted effects of ionizing radiation. J. Radiat. Res. Appl. Sci. 2015, 8, 247–254. [Google Scholar] [CrossRef] [Green Version]
  20. Singh, V.K.; Newman, V.L.; Romaine, P.L.; Hauer-Jensen, M.; Pollard, H.B. Use of biomarkers for assessing radiation injury and efficacy of countermeasures. Expert Rev. Mol. Diagn. 2016, 16, 65–81. [Google Scholar] [CrossRef]
  21. Yan, K.; Chia, L.; Li, X.; Ootani, A.; Su, J.; Lee, J.; Su, N.; Luo, Y.; Heilshorn, S.; Amieva, M.; et al. The intestinal stem cell markers Bmi1 and Lgr5 identify two functionally distinct populations. Proc. Natl. Acad. Sci. USA 2012, 109, 466–471. [Google Scholar] [CrossRef]
  22. Schepers, A.G.; Vries, R.; Van Den Born, M.; Van De Wetering, M.; Clevers, H. Lgr5 intestinal stem cells have high telomerase activity and randomly segregate their chromosomes. EMBO J. 2011, 30, 1104–1109. [Google Scholar] [CrossRef] [Green Version]
  23. Kim, C.; Yang, V.W.; Bialkowska, A.B. The Role of Intestinal Stem Cells in Epithelial Regeneration Following Radiation-Induced Gut Injury. Curr. Stem Cell Rep. 2017, 3, 320–332. [Google Scholar] [CrossRef] [Green Version]
  24. Barker, N. Adult intestinal stem cells: Critical drivers of epithelial homeostasis and regeneration. Nat. Rev. Mol. Cell Biol. 2013, 15, 19. [Google Scholar] [CrossRef] [PubMed]
  25. Booth, C.; Tudor, G.; Tudor, J.; Katz, B.P.; MacVittie, T.J. Acute gastrointestinal syndrome in high-dose irradiated mice. Health Phys. 2012, 103, 383–399. [Google Scholar] [CrossRef] [PubMed]
  26. Shukla, P.K.; Gangwar, R.; Manda, B.; Meena, A.S.; Yadav, N.; Szabo, E.; Balogh, A.; Lee, S.C.; Tigyi, G.; Rao, R. Rapid disruption of intestinal epithelial tight junction and barrier dysfunction by ionizing radiation in mouse colon in vivo: Protection by N-acetyl-l-cysteine. Am. J. Physiol. Gastrointest. Liver Physiol. 2016, 310, G705–G715. [Google Scholar] [CrossRef]
  27. Wang, J.; Boerma, M.; Fu, Q.; Hauer-Jensen, M. Significance of endothelial dysfunction in the pathogenesis of early and delayed radiation enteropathy. World J. Gastroenterol. 2007, 13, 3047–3055. [Google Scholar] [CrossRef] [PubMed]
  28. Mollà, M.; Gironella, M.; Miquel, R.; Tovar, V.; Engel, P.; Biete, A.; Piqué, J.M.; Panés, J. Relative roles of ICAM-1 and VCAM-1 in the pathogenesis of experimental radiation-induced intestinal inflammation. Int. J. Radiat. Oncol. Biol. Phys. 2003, 57, 264–273. [Google Scholar] [CrossRef]
  29. Mollà, M.; Panés, J. Radiation-induced intestinal inflammation. World J. Gastroenterol. 2007, 13, 3043–3046. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Kim, N.; Luster, A. The role of tissue resident cells in neutrophil recruitment. Trends Immunol. 2015, 36, 547–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Gómez-Llorente, C.; Muñoz, S.; Gil, A. Role of Toll-like receptors in the development of immunotolerance mediated by probiotics. Proc. Nutr. Soc. 2010, 69, 381–389. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Chen, H.; Wang, Z.D.; Chen, M.S.; Zhang, X.Q.; Shen, L.P.; Zhang, J.X.; Chen, Y. Activation of Toll-like receptors by intestinal microflora reduces radiation-induced DNA damage in mice. Mutat. Res. Genet. Toxicol. Environ. Mutagen 2014, 774, 22–28. [Google Scholar] [CrossRef] [PubMed]
  33. Vétizou, M.; Pitt, J.M.; Daillère, R.; Lepage, P.; Flament, C.; Rusakiewicz, S.; Routy, B.; Maria, P.; Duong, C.P.M.; Poirier-colame, V.; et al. Anticancer immunotherapy by CTLA-4 blockade relies on the gut microbiota. Science 2016, 350, 1079–1084. [Google Scholar] [CrossRef] [PubMed]
  34. Zhu, J.; Paul, W.E. Heterogeneity and plasticity of T helper cells. Cell Res. 2010, 20, 4–12. [Google Scholar] [CrossRef]
  35. Schaue, D.; McBride, W.H. T lymphocytes and normal tissue responses to radiation. Front. Oncol. 2012, 2, 119. [Google Scholar] [CrossRef]
  36. Zoetendal, E.G.; Akkermans, A.D.; De Vos, W.M. Temperature gradient gel electrophoresis analysis of 16S rRNA from human fecal samples reveals stable and host-specific communities of active bacteria. Appl. Environ. Microbiol. 1998, 64, 3854–3859. [Google Scholar] [PubMed]
  37. Sender, R.; Fuchs, S.; Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol. 2016, 14, e1002533. [Google Scholar] [CrossRef]
  38. Harmsen, H.J.M.; Raangs, G.C.; He, T.; Degener, J.E.; Welling, G.W. Extensive set of 16S rRNA-based probes for detection of bacteria in human feces. Appl. Environ. Microbiol. 2002, 68, 2982–2990. [Google Scholar] [CrossRef]
  39. Vyas, U.; Ranganathan, N. Probiotics, prebiotics, and synbiotics: Gut and beyond. Gastroenterol. Res. Pract. 2012, 2012, 872716. [Google Scholar] [CrossRef]
  40. Heiss, C.N.; Olofsson, L.E. The role of the gut microbiota in development, function and disorders of the central nervous system and the enteric nervous system. J. Neuroendocrinol. 2019, e12684. [Google Scholar] [CrossRef]
  41. Mowat, A.M. To respond or not to respond—A personal perspective of intestinal tolerance. Nat. Rev. Immunol. 2018, 18, 405–415. [Google Scholar] [CrossRef] [PubMed]
  42. Tordesillas, L.; Berin, M.C. Mechanisms of Oral Tolerance. Clin. Rev. Allergy Immunol. 2018, 55, 107–117. [Google Scholar] [CrossRef] [PubMed]
  43. Bengmark, S. Ecological control of the gastrointestinal tract. The role of probiotic flora. Gut 1998, 42, 2–7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Vanderhoof, J.A.; Young, R.J. Use of probiotics in childhood gastrointestinal disorders. J. Pediatr. Gastroenterol. Nutr. 1998, 27, 323–332. [Google Scholar] [CrossRef]
  45. Holzapfel, W.H.; Haberer, P.; Snel, J.; Schillinger, U.; Huis in’t Veld, J.H. Overview of gut flora and probiotics. Int. J. Food Microbiol. 1998, 41, 85–101. [Google Scholar] [CrossRef]
  46. Vandenbergh, P. Lactic acid bacteria, their metabolic prodcuts and interference with microbial growth. FEMS Microbiol. Rev. 1993, 12, 221–238. [Google Scholar] [CrossRef]
  47. Davidson, J.N.; Hirsh, D.C. Bacterial competition as a means of preventing neonatal diarrhea in pigs. Infect. Immun. 1976, 13, 1773–1774. [Google Scholar] [PubMed]
  48. DeGruttola, A.K.; Low, D.; Mizoguchi, A.; Mizoguchi, E. Current understanding of dysbiosis in disease in human and animal models. Inflamm. Bowel Dis. 2016, 22, 1137–1150. [Google Scholar] [CrossRef] [Green Version]
  49. Ghavami, S.B.; Rostami, E.; Sephay, A.A.; Shahrokh, S.; Balaii, H.; Aghdaei, H.A.; Zali, M.R. Alterations of the human gut Methanobrevibacter smithii as a biomarker for inflammatory bowel diseases. Microb. Pathog. 2018, 117, 285–289. [Google Scholar] [CrossRef] [PubMed]
  50. Ferreira, R.M.; Pereira-Marques, J.; Pinto-Ribeiro, I.; Costa, J.L.; Carneiro, F.; Machado, J.C.; Figueiredo, C. Gastric microbial community profiling reveals a dysbiotic cancer-associated microbiota. Gut 2018, 67, 226–236. [Google Scholar] [CrossRef]
  51. Wang, A.; Ling, Z.; Yang, Z.; Kiela, P.R.; Wang, T.; Wang, C.; Cao, L.; Geng, F.; Shen, M.; Ran, X.; et al. Gut Microbial Dysbiosis May Predict Diarrhea and Fatigue in Patients Undergoing Pelvic Cancer Radiotherapy: A Pilot Study. PLoS ONE 2015, 10, e0126312. [Google Scholar] [CrossRef] [PubMed]
  52. Gerassy-Vainberg, S.; Blatt, A.; Danin-Poleg, Y.; Gershovich, K.; Sabo, E.; Nevelsky, A.; Daniel, S.; Dahan, A.; Ziv, O.; Dheer, R.; et al. Radiation induces proinflammatory dysbiosis: Transmission of inflammatory susceptibility by host cytokine induction. Gut 2018, 67, 97–107. [Google Scholar] [CrossRef] [PubMed]
  53. Crawford, P.A.; Gordon, J.I. Microbial regulation of intestinal radiosensitivity. Proc. Natl. Acad. Sci. USA 2005, 102, 13254–13259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Wilson, B.R. Survival studies of whole-body X-irradiated germfree (axenic) mice. Radiat. Res. 1963, 20, 477–483. [Google Scholar] [CrossRef] [PubMed]
  55. McLauglin, M.M.; Dacquisto, M.P.; Jacobus, D.P.; Horowitz, R.E. Effects of the germfree state on responses of mice to whole-body irradiation. Radiat. Res. 1964, 23, 333–349. [Google Scholar] [CrossRef]
  56. Jervis, H.R.; McLaughlin, M.M.; Johnson, M.C. Effect of neutron-gamma radiation on the morphology of the mucosa of the small intestine of germfree and conventional mice. Radiat. Res. 1971, 45, 613–628. [Google Scholar] [CrossRef]
  57. Manichanh, C.; Varela, E.; Martinez, C.; Antolin, M.; Llopis, M.; Dore, J.; Giralt, J.; Guarner, F.; Malagelada, J.-R. The gut microbiota predispose to the pathophysiology of acute postradiotherapy diarrhea. Am. J. Gastroenterol. 2008, 103, 1754–1761. [Google Scholar] [CrossRef]
  58. Kim, Y.S.; Kim, J.; Park, S.-J. High-throughput 16S rRNA gene sequencing reveals alterations of mouse intestinal microbiota after radiotherapy. Anaerobe 2015, 33, 1–7. [Google Scholar] [CrossRef]
  59. Nam, Y.-D.; Kim, H.J.; Seo, J.-G.; Kang, S.W.; Bae, J.-W. Impact of Pelvic Radiotherapy on Gut Microbiota of Gynecological Cancer Patients Revealed by Massive Pyrosequencing. PLoS ONE 2013, 8, e82659. [Google Scholar] [CrossRef] [PubMed]
  60. Touchefeu, Y.; Montassier, E.; Nieman, K.; Gastinne, T.; Potel, G.; Bruley Des Varannes, S.; Le Vacon, F.; De La Cochetière, M.F. Systematic review: The role of the gut microbiota in chemotherapy- or radiation-induced gastrointestinal mucositis—Current evidence and potential clinical applications. Aliment. Pharmacol. Ther. 2014, 40, 409–421. [Google Scholar] [CrossRef] [PubMed]
  61. Paul, P.; Unnikrishnan, M.K.; Nagappa, A.N. Phytochemicals as radioprotective agents—A Review. Indian J. Nat. Prod. Resour. 2011, 2, 137–150. [Google Scholar]
  62. Traber, M.G.; Atkinson, J. Vitamin E, antioxidant and nothing more. Free Radic. Biol. Med. 2007, 43, 4–15. [Google Scholar] [CrossRef]
  63. Bailey, D.M.; Davies, B. Acute mountain sickness; prophylactic benefits of antioxidant vitamin supplementation at high altitude. High Alt. Med. Biol. 2001, 2, 21–29. [Google Scholar] [CrossRef] [PubMed]
  64. Bailey, D.M.; Bartsch, P.; Knauth, M.; Baumgartner, R.W. Emerging concepts in acute mountain sickness and high-altitude cerebral edema: From the molecular to the morphological. Cell Mol. Life Sci. 2009, 66, 3583–3594. [Google Scholar] [CrossRef]
  65. Xu, C.; Sun, R.; Qiao, X.; Xu, C.; Shang, X.; Niu, W.; Chao, Y. Effect of Vitamin E Supplementation on Intestinal Barrier Function in Rats Exposed to High Altitude Hypoxia Environment. Korean J. Physiol. Pharmacol. 2014, 18, 313. [Google Scholar] [CrossRef] [PubMed]
  66. Czarnewski, P.; Das, S.; Parigi, S.M.; Villablanca, E.J. Retinoic acid and its role in modulating intestinal innate immunity. Nutrients 2017, 9, 68. [Google Scholar] [CrossRef]
  67. Yousefi, B.; Azizzadeh, F. The histopathalogical effects of retinoic acid on the tissues. Pakistan J. Biol. Sci. 2010, 13, 927–936. [Google Scholar] [CrossRef]
  68. De Medeiros, P.H.Q.S.; Pinto, D.V.; de Almeida, J.Z.; Rêgo, J.M.C.; Rodrigues, F.A.P.; Lima, A.Â.M.; Bolick, D.T.; Guerrant, R.L.; Oriá, R.B. Modulation of intestinal immune and barrier functions by vitamin A: Implications for current understanding of malnutrition and enteric infections in children. Nutrients 2018, 10, 1128. [Google Scholar] [CrossRef] [PubMed]
  69. Tielsch, J.M.; Rahmathullah, L.; Thulasiraj, R.D.; Katz, J.; Coles, C.; Sheeladevi, S.; John, R.; Prakash, K. Newborn vitamin A dosing reduces the case fatality but not incidence of common childhood morbidities in South India. J. Nutr. 2007, 137, 2470–2474. [Google Scholar] [CrossRef]
  70. Lester Packer, J.F. Vitamin C in Health and Disease; CRC Press: New York, NY, USA, 1997; pp. 28–30. [Google Scholar]
  71. Fischer, N.; Seo, E.J.; Efferth, T. Prevention from radiation damage by natural products. Phytomedicine 2018, 47, 192–200. [Google Scholar] [CrossRef]
  72. Mohammed, B.M.; Fisher, B.J.; Kraskauskas, D.; Farkas, D.; Brophy, D.F.; Fowler, A.A.; Natarajan, R. Vitamin C: A novel regulator of neutrophil extracellular trap formation. Nutrients 2013, 5, 3131–3151. [Google Scholar] [CrossRef]
  73. Danielski, L.G.; Walczewski, E.; de Jesus, C.R.; Florentino, D.; Giustina ADella Goldim, M.P.; Kanis, L.A.; Pereira, G.W.; Pereira, V.D.; Felisberto, F.; et al. Preoperative vitamin C supplementation improves colorectal anastomotic healing and biochemical parameters in malnourished rats. Int. J. Colorectal. Dis. 2016, 31, 1759–1766. [Google Scholar] [CrossRef]
  74. Rall, L.C.; Meydani, S.N. Vitamin B6 and immune competence. Nutr. Rev. 1993, 51, 217–225. [Google Scholar] [CrossRef] [PubMed]
  75. Al-Numair, K.S.; Waly, M.I.; Ali, A.; Essa, M.M.; Farhat, M.F.; Alsaif, M.A. Dietary folate protects against azoxymethane-induced aberrant crypt foci development and oxidative stress in rat colon. Exp. Biol. Med. 2011, 236, 1005–1011. [Google Scholar] [CrossRef] [PubMed]
  76. MacFarlane, A.J.; Behan, N.A.; Matias, F.M.G.; Green, J.; Caldwell, D.; Brooks, S.P.J. Dietary folate does not significantly affect the intestinal microbiome, inflammation or tumorigenesis in azoxymethane-dextran sodium sulphate-treated mice. Br. J. Nutr. 2013, 109, 630–638. [Google Scholar] [CrossRef] [PubMed]
  77. Gibson, G.R.; Hutkins, R.; Sanders, M.E.; Prescott, S.L.; Reimer, R.A.; Salminen, S.J.; Scott, K.; Stanton, C.; Swanson, K.S.; Cani, P.D.; et al. Expert consensus document: The International Scientific Association for Probiotics and Prebiotics (ISAPP) consensus statement on the definition and scope of prebiotics. Nat. Rev. Gastroenterol. Hepatol. 2017, 14, 491–502. [Google Scholar] [CrossRef]
  78. Everard, A.; Lazarevic, V.; Gaïa, N.; Johansson, M.; Ståhlman, M.; Backhed, F.; Delzenne, N.M.; Schrenzel, J.; François, P.; Cani, P.D. Microbiome of prebiotic-treated mice reveals novel targets involved in host response during obesity. ISME J. 2014, 8, 2116–2130. [Google Scholar] [CrossRef]
  79. Paul, H.A.; Bomhof, M.R.; Vogel, H.J.; Reimer, R.A. Diet-induced changes in maternal gut microbiota and metabolomic profiles influence programming of offspring obesity risk in rats. Sci. Rep. 2016, 6, 1–14. [Google Scholar] [CrossRef] [PubMed]
  80. Dewulf, E.M.; Cani, P.D.; Claus, S.P.; Fuentes, S.; Puylaert, P.G.B.; Neyrinck, A.M.; Bindels, L.B.; De Vos, W.M.; Gibson, G.R.; Thissen, J.P.; et al. Insight into the prebiotic concept: Lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut 2013, 62, 1112–1121. [Google Scholar] [CrossRef]
  81. Vandeputte, D.; Falony, G.; Vieira-Silva, S.; Wang, J.; Sailer, M.; Theis, S.; Verbeke, K.; Raes, J. Prebiotic inulin-type fructans induce specific changes in the human gut microbiota. Gut 2017, 66, 1968–1974. [Google Scholar] [CrossRef] [Green Version]
  82. Li, T.; Lu, X.; Yang, X. Stachyose-enriched alpha-galacto-oligosaccharides regulate gut microbiota and relieve constipation in mice. J. Agric. Food Chem. 2013, 61, 11825–11831. [Google Scholar] [CrossRef] [PubMed]
  83. Li, T.; Lu, X.; Yang, X. Evaluation of clinical safety and beneficial effects of stachyose-enriched α-galacto-oligosaccharides on gut microbiota and bowel function in humans. Food Funct. 2017, 8, 262–269. [Google Scholar] [CrossRef]
  84. CJagetia, G. Radioprotective Potential of Plants and Herbs against the Effects of Ionizing Radiation. J. Clin. Biochem. Nutr. 2007, 40, 74–81. [Google Scholar] [CrossRef] [Green Version]
  85. Cardona, F.; Andrés-Lacueva, C.; Tulipani, S.; Tinahones, F.J.; Queipo-Ortuño, M.I. Benefits of polyphenols on gut microbiota and implications in human health. J. Nutr. Biochem. 2013, 24, 1415–1422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Espín, J.C.; González-Sarrías, A.; Tomás-Barberán, F.A. The gut microbiota: A key factor in the therapeutic effects of (poly)phenols. Biochem. Pharmacol. 2017, 139, 82–93. [Google Scholar] [CrossRef]
  87. Tomas-Barberan, F.A.; Selma, M.V.; Espin, J.C. Interactions of gut microbiota with dietary polyphenols and consequences to human health. Curr. Opin. Clin. Nutr. Metab. Care 2016, 19, 471–476. [Google Scholar] [CrossRef]
  88. Anhe, F.F.; Varin, T.V.; Le Barz, M.; Desjardins, Y.; Levy, E.; Roy, D.; Marette, A. Gut Microbiota Dysbiosis in Obesity-Linked Metabolic Diseases and Prebiotic Potential of Polyphenol-Rich Extracts. Curr. Obes. Rep. 2015, 4, 389–400. [Google Scholar] [CrossRef]
  89. Selma, M.V.; Espin, J.C.; Tomas-Barberan, F.A. Interaction between phenolics and gut microbiota: Role in human health. J. Agric. Food Chem. 2009, 57, 6485–6501. [Google Scholar] [CrossRef]
  90. Tzounis, X.; Rodriguez-Mateos, A.; Vulevic, J.; Gibson, G.R.; Kwik-Uribe, C.; Spencer, J.P.E. Prebiotic evaluation of cocoa-derived flavanols in healthy humans by using a randomized, controlled, double-blind, crossover intervention study. Am. J. Clin. Nutr. 2011, 93, 62–72. [Google Scholar] [CrossRef] [PubMed]
  91. Zhang, Z.; Wu, X.; Cao, S.; Wang, L.; Wang, D.; Yang, H.; Feng, Y.; Wang, S.; Li, L. Caffeic acid ameliorates colitis in association with increased Akkermansia population in the gut microbiota of mice. Oncotarget 2016, 7, 31790–31799. [Google Scholar] [CrossRef]
  92. Song, H.; Chu, Q.; Yan, F.; Yang, Y.; Han, W.; Zheng, X. Red pitaya betacyanins protects from diet-induced obesity, liver steatosis and insulin resistance in association with modulation of gut microbiota in mice. J. Gastroenterol. Hepatol. 2016, 31, 1462–1469. [Google Scholar] [CrossRef]
  93. Moreno-Indias, I.; Sanchez-Alcoholado, L.; Perez-Martinez, P.; Andres-Lacueva, C.; Cardona, F.; Tinahones, F.; Queipo-Ortuno, M.I. Red wine polyphenols modulate fecal microbiota and reduce markers of the metabolic syndrome in obese patients. Food Funct. 2016, 7, 1775–1787. [Google Scholar] [CrossRef] [Green Version]
  94. Hill, C.; Guarner, F.; Reid, G.; Gibson, G.R.; Merenstein, D.J.; Pot, B.; Morelli, L.; Canani, R.B.; Flint, H.J.; Salminen, S.; et al. Expert consensus document. The International Scientific Association for Probiotics and Prebiotics consensus statement on the scope and appropriate use of the term probiotic. Nat. Rev. Gastroenterol. Hepatol. 2014, 11, 506–514. [Google Scholar] [CrossRef] [PubMed]
  95. Kaila, M.; Isolauri, E.; Soppi, E.; Virtanen, E.; Laine, S.; Arvilommi, H. Enhancement of the circulating antibody secreting cell response in human diarrhea by a human Lactobacillus strain. Pediatr. Res. 1992, 32, 141–144. [Google Scholar] [CrossRef] [PubMed]
  96. Kaila, M.; Isolauri, E.; Saxelin, M.; Arvilommi, H.; Vesikari, T. Viable versus inactivated Lactobacillus strain GG in acute rotavirus diarrhoea. Arch. Dis. Child. 1995, 72, 51–53. [Google Scholar] [CrossRef] [PubMed]
  97. Guarino, A.; Guandalini, S.; Lo Vecchio, A. Probiotics for Prevention and Treatment of Diarrhea. J. Clin. Gastroenterol. 2015, 49 (Suppl. 1), S37–S45. [Google Scholar] [CrossRef]
  98. Conway, P.L.; Gorbach, S.L.; Goldin, B.R. Survival of lactic acid bacteria in the human stomach and adhesion to intestinal cells. J. Dairy Sci. 1987, 70, 1–12. [Google Scholar] [CrossRef]
  99. Yan, F. Probiotic Bacterium Prevents Cytokine-induced Apoptosis in Intestinal Epithelial Cells. J. Biol. Chem. 2002, 277, 50959–50965. [Google Scholar] [CrossRef]
  100. Johnson-Henry, K.C.; Donato, K.A.; Shen-Tu, G.; Gordanpour, M.; Sherman, P.M. Lactobacillus rhamnosus Strain GG Prevents Enterohemorrhagic Escherichia coli O157:H7-Induced Changes in Epithelial Barrier Function. Infect. Immun. 2008, 76, 1340–1348. [Google Scholar] [CrossRef]
  101. Zhao, H.; Zhao, C.; Dong, Y.; Zhang, M.; Wang, Y.; Li, F.; Li, X.; McClain, C.; Yang, S.; Feng, W. Inhibition of miR122a by Lactobacillus rhamnosus GG culture supernatant increases intestinal occludin expression and protects mice from alcoholic liver disease. Toxicol. Lett. 2015, 234, 194–200. [Google Scholar] [CrossRef]
  102. Orlando, A.; Linsalata, M.; Notarnicola, M.; Tutino, V.; Russo, F. Lactobacillus GG restoration of the gliadin induced epithelial barrier disruption: The role of cellular polyamines. BMC Microbiol. 2014, 14, 19. [Google Scholar] [CrossRef]
  103. Kumar, A.; Wu, H.; Collier-Hyams, L.S.; Hansen, J.M.; Li, T.; Yamoah, K.; Pan, Z.-Q.; Jones, D.P.; Neish, A.S. Commensal bacteria modulate cullin-dependent signaling via generation of reactive oxygen species. EMBO J. 2007, 26, 4457–4466. [Google Scholar] [CrossRef] [Green Version]
  104. Lin, P.W.; Myers, L.E.S.; Ray, L.; Song, S.-C.; Nasr, T.R.; Berardinelli, A.J.; Kundu, K.; Murthy, N.; Hansen, J.M.; Neish, A.S. Lactobacillus rhamnosus blocks inflammatory signaling in vivo via reactive oxygen species generation. Free Radic. Biol. Med. 2009, 47, 1205–1211. [Google Scholar] [CrossRef] [PubMed]
  105. Vong, L.; Lorentz, R.J.; Assa, A.; Glogauer, M.; Sherman, P.M. Probiotic Lactobacillus rhamnosus inhibits the formation of neutrophil extracellular traps. J. Immunol. 2014, 192, 1870–1877. [Google Scholar] [CrossRef]
  106. Pieścik-Lech, M.; Urbańska, M.; Szajewska, H. Lactobacillus GG (LGG) and smectite versus LGG alone for acute gastroenteritis: A double-blind, randomized controlled trial. Eur. J. Pediatr. 2013, 172, 247–253. [Google Scholar] [CrossRef]
  107. Pant, A.R.; Graham, S.M.; Allen, S.J.; Harikul, S.; Sabchareon, A.; Cuevas, L.; Hart, C.A. Lactobacillus GG and acute diarrhoea in young children in the tropics. J. Trop. Pediatr. 1996, 42, 162–165. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  108. Raza, S.; Graham, S.M.; Allen, S.J.; Sultana, S.; Cuevas, L.; Hart, C.A. Lactobacillus GG promotes recovery from acute nonbloody diarrhea in Pakistan. Pediatr. Infect. Dis. J. 1995, 14, 107–111. [Google Scholar] [CrossRef] [PubMed]
  109. Shornikova, A.V.; Isolauri, E.; Burkanova, L.; Lukovnikova, S.; Vesikari, T. A trial in the Karelian Republic of oral rehydration and Lactobacillus GG for treatment of acute diarrhoea. Acta Paediatr. 1997, 86, 460–465. [Google Scholar] [CrossRef] [PubMed]
  110. Kawahara, T.; Makizaki, Y.; Oikawa, Y.; Tanaka, Y.; Maeda, A.; Shimakawa, M.; Komoto, S.; Moriguchi, K.; Ohno, H.; Taniguchi, K. Oral administration of Bifidobacterium bifidum G9-1 alleviates rotavirus gastroenteritis through regulation of intestinal homeostasis by inducing mucosal protective factors. PLoS ONE 2017, 12, e0173979. [Google Scholar] [CrossRef]
  111. Karczewski, J.; Troost, F.J.; Konings, I.; Dekker, J.; Kleerebezem, M.; Brummer, R.-J.M.; Wells, J.M. Regulation of human epithelial tight junction proteins by Lactobacillus plantarum in vivo and protective effects on the epithelial barrier. Am. J. Physiol. Gastrointest. Liver Physiol. 2010, 298, G851–G859. [Google Scholar] [CrossRef]
  112. Yang, F.; Wang, A.; Zeng, X.; Hou, C.; Liu, H.; Qiao, S. Lactobacillus reuteri I5007 modulates tight junction protein expression in IPEC-J2 cells with LPS stimulation and in newborn piglets under normal conditions. BMC Microbiol. 2015, 15, 32. [Google Scholar] [CrossRef] [PubMed]
  113. Tien, M.-T.; Girardin, S.E.; Regnault, B.; Le Bourhis, L.; Dillies, M.-A.; Coppee, J.-Y.; Bourdet-Sicard, R.; Sansonetti, P.J.; Pedron, T. Anti-inflammatory effect of Lactobacillus casei on Shigella-infected human intestinal epithelial cells. J. Immunol. 2006, 176, 1228–1237. [Google Scholar] [CrossRef] [PubMed]
  114. Eun, C.S.; Kim, Y.S.; Han, D.S.; Choi, J.H.; Lee, A.R.; Park, Y.K. Lactobacillus casei prevents impaired barrier function in intestinal epithelial cells. APMIS 2011, 119, 49–56. [Google Scholar] [CrossRef]
  115. Toumi, R.; Abdelouhab, K.; Rafa, H.; Soufli, I.; Raissi-Kerboua, D.; Djeraba, Z.; Touil-Boukoffa, C. Beneficial role of the probiotic mixture Ultrabiotique on maintaining the integrity of intestinal mucosal barrier in DSS-induced experimental colitis. Immunopharmacol. Immunotoxicol. 2013, 35, 403–409. [Google Scholar] [CrossRef]
  116. Park, J.-S.; Choi, J.; Jhun, J.; Kwon, J.Y.; Lee, B.-I.; Yang, C.W.; Park, S.-H.; Cho, M.-L. Lactobacillus acidophilus Improves Intestinal Inflammation in an Acute Colitis Mouse Model by Regulation of Th17 and Treg Cell Balance and Fibrosis Development. J. Med. Food 2018, 21, 215–224. [Google Scholar] [CrossRef]
  117. Pearce, J.L.; Hamilton, J.R. Controlled trial of orally administered Lactobacilli in acute infantile diarrhea. J. Pediatr. 1974, 84, 261–262. [Google Scholar] [CrossRef]
  118. Haton, C.; Francois, A.; Vandamme, M.; Wysocki, J.; Griffiths, N.M.; Benderitter, M. Imbalance of the antioxidant network of mouse small intestinal mucosa after radiation exposure. Radiat. Res. 2007, 167, 445–453. [Google Scholar] [CrossRef]
  119. Felemovicius, I.; Bonsack, M.E.; Baptista, M.L.; Delaney, J.P. Intestinal radioprotection by vitamin E (alpha-tocopherol). Ann. Surg. 1995, 222, 504–510. [Google Scholar] [CrossRef] [PubMed]
  120. Kumar, K.S.; Srinivasan, V.; Toles, R.; Jobe, L.; Seed, T.M. Nutritional approaches to radioprotection: Vitamin E. Mil. Med. 2002, 167 (Suppl. 2), 57–59. [Google Scholar] [PubMed]
  121. Singh, P.K.; Wise, S.Y.; Ducey, E.J.; Fatanmi, O.O.; Elliott, T.B.; Singh, V.K. alpha-Tocopherol succinate protects mice against radiation-induced gastrointestinal injury. Radiat. Res. 2012, 177, 133–145. [Google Scholar] [CrossRef]
  122. Brown, S.L.; Kolozsvary, A.; Liu, J.; Jenrow, K.A.; Ryu, S.; Kim, J.H. Antioxidant diet supplementation starting 24 hours after exposure reduces radiation lethality. Radiat. Res. 2010, 173, 462–468. [Google Scholar] [CrossRef] [PubMed]
  123. Thotala, D.; Chetyrkin, S.; Hudson, B.; Hallahan, D.; Voziyan, P.; Yazlovitskaya, E. Pyridoxamine protects intestinal epithelium from ionizing radiation-induced apoptosis. Free Radic. Biol. Med. 2009, 47, 779–785. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Roche, M.; Neti, P.V.S.V.; Kemp, F.W.; Agrawal, A.; Attanasio, A.; Douard, V.; Muduli, A.; Azzam, E.I.; Norkus, E.; Brimacombe, M.; et al. Radiation-induced reductions in transporter mRNA levels parallel reductions in intestinal sugar transport. Am. J. Physiol. Regul. Integr. Comp. Physiol. 2010, 298, R173–R182. [Google Scholar] [CrossRef]
  125. Roche, M.; Kemp, F.W.; Agrawal, A.; Attanasio, A.; Neti, P.V.S.V.; Howell, R.W.; Ferraris, R.P. Marked changes in endogenous antioxidant expression precede vitamin A, C and E-protectable, radiation-induced reductions in small intestinal nutrient transport. Free Radic. Biol. Med. 2011, 50, 55–65. [Google Scholar] [CrossRef]
  126. Yamamoto, T.; Kinoshita, M.; Shinomiya, N.; Hiroi, S.; Sugasawa, H.; Matsushita, Y.; Majima, T.; Saitoh, D.; Seki, S. Pretreatment with Ascorbic Acid Prevents Lethal Gastrointestinal Syndrome in Mice Receiving a Massive Amount of Radiation. J. Radiat. Res. 2010, 51, 145–156. [Google Scholar] [CrossRef] [Green Version]
  127. Sato, T.; Kinoshita, M.; Yamamoto, T.; Ito, M.; Nishida, T.; Takeuchi, M.; Saitoh, D.; Seki, S.; Mukai, Y. Treatment of irradiated mice with high-dose ascorbic acid reduced lethality. PLoS ONE 2015, 10, e0117020. [Google Scholar] [CrossRef] [PubMed]
  128. Adaramoye, O.; Ogungbenro, B.; Anyaegbu, O.; Fafunso, M. Protective effects of extracts of Vernonia amygdalina, Hibiscus sabdariffa and vitamin C against radiation-induced liver damage in rats. J. Radiat. Res. 2008, 49, 123–131. [Google Scholar] [CrossRef]
  129. Shimoi, K.; Masuda, S.; Furugori, M.; Esaki, S.; Kinae, N. Radioprotective effect of antioxidative flavonoids in gamma-ray irradiated mice. Carcinogenesis 1994, 15, 2669–2672. [Google Scholar] [CrossRef] [PubMed]
  130. Shimoi, K.; Masuda, S.; Shen, B.; Furugori, M.; Kinae, N. Radioprotective effects of antioxidative plant flavonoids in mice. Mutat. Res. 1996, 350, 153–161. [Google Scholar] [CrossRef]
  131. Benkovic, V.; Horvat Knezevic, A.; Dikic, D.; Lisicic, D.; Orsolic, N.; Basic, I.; Kosalec, I.; Kopjar, N. Radioprotective effects of propolis and quercetin in γ-irradiated mice evaluated by the alkaline comet assay. Phytomedicine 2008, 15, 851–858. [Google Scholar] [CrossRef]
  132. Orsolić, N.; Benković, V.; Horvat-Knezević, A.; Kopjar, N.; Kosalec, I.; Bakmaz, M.; Mihaljević, Z.; Bendelja, K.; Basić, I. Assessment by survival analysis of the radioprotective properties of propolis and its polyphenolic compounds. Biol. Pharm. Bull. 2007, 30, 946–951. [Google Scholar] [CrossRef] [PubMed]
  133. Van Hijum, S.A.F.T.; van Geel-Schutten, G.H.; Rahaoui, H.; van der Maarel, M.J.E.C.; Dijkhuizen, L. Characterization of a novel fructosyltransferase from Lactobacillus reuteri that synthesizes high-molecular-weight inulin and inulin oligosaccharides. Appl. Environ. Microbiol. 2002, 68, 4390–4398. [Google Scholar] [CrossRef]
  134. Haak, B.W.; Littmann, E.R.; Chaubard, J.-L.; Pickard, A.J.; Fontana, E.; Adhi, F.; Gyaltshen, Y.; Ling, L.; Morjaria, S.M.; Peled, J.U.; et al. Impact of gut colonization with butyrate-producing microbiota on respiratory viral infection following allo-HCT. Blood 2018, 131, 2978–2986. [Google Scholar] [CrossRef] [PubMed]
  135. Qishen, P.; Guo, B.J.; Kolman, A. Radioprotective effect of extract from Spirulina platensis in mouse bone marrow cells studied by using the micronucleus test. Toxicol. Lett. 1989, 48, 165–169. [Google Scholar] [CrossRef]
  136. Sanders, M.E.; Benson, A.; Lebeer, S.; Merenstein, D.J.; Klaenhammer, T.R. Shared mechanisms among probiotic taxa: Implications for general probiotic claims. Curr. Opin. Biotechnol. 2018, 49, 207–216. [Google Scholar] [CrossRef]
  137. Seal, M.; Naito, Y.; Barreto, R.; Lorenzetti, A.; Safran, P.; Marotta, F. Experimental radiotherapy-induced enteritis: A probiotic interventional study. J. Dig. Dis. 2007, 8, 143–147. [Google Scholar] [CrossRef]
  138. Ki, Y.; Kim, W.; Cho, H.; Ahn, K.; Choi, Y.; Kim, D. The effect of probiotics for preventing radiation-induced morphological changes in intestinal mucosa of rats. J. Korean Med. Sci. 2014, 29, 1372–1378. [Google Scholar] [CrossRef] [PubMed]
  139. Ciorba, M.A.; Riehl, T.E.; Rao, M.S.; Moon, C.; Ee, X.; Nava, G.M.; Walker, M.R.; Marinshaw, J.M.; Stappenbeck, T.S.; Stenson, W.F. Lactobacillus probiotic protects intestinal epithelium from radiation injury in a TLR-2/cyclo-oxygenase-2-dependent manner. Gut 2012, 61, 829–838. [Google Scholar] [CrossRef]
  140. Riehl, T.E.; Alvarado, D.; Ee, X.; Zuckerman, A.; Foster, L.; Kapoor, V.; Thotala, D.; Ciorba, M.A.; Stenson, W. Lactobacillus rhamnosus GG protects the intestinal epithelium from radiation injury through release of lipoteichoic acid, macrophage activation and the migration of mesenchymal stem cells. Gut 2018. [Google Scholar] [CrossRef]
  141. Demirer, S.; Aydintug, S.; Aslim, B.; Kepenekci, I.; Sengül, N.; Evirgen, O.; Gerceker, D.; Andrieu, M.N.; Ulusoy, C.; Karahüseyinoglu, S. Effects of probiotics on radiation-induced intestinal injury in rats. Nutrition 2006, 22, 179–186. [Google Scholar] [CrossRef]
  142. Demirer, S.; Ulusu, N.N.; Aslim, B.; Kepenekci, I.; Ulusoy, C.; Andrieu, M.N.; Erkek, B.; Aydintug, S. Protective effects of Lactobacillus delbrueckii subsp bulgaricus B3 on intestinal enzyme activities after abdominal irradiation in rats. Nutr. Res. 2007, 27, 300–305. [Google Scholar] [CrossRef]
  143. Liu, Q.; Nobaek, S.; Adawi, D.; Mao, Y.; Wang, M.; Molin, G.; Ekelund, M.; Jeppsson, B. Administration of Lactobacillus plantarum 299v reduces side-effects of external radiation on colon anastomotic healing in an experimental model. Colorectal Dis. 2001, 3, 245–252. [Google Scholar] [CrossRef] [PubMed]
  144. Kennedy, M.; Bruninga, K.; Mutlu, E.A.; Losurdo, J.; Choudhary, S.; Keshavarzian, A. Successful and sustained treatment of chronic radiation proctitis with antioxidant vitamins E and C. Am. J. Gastroenterol. 2001, 96, 1080. [Google Scholar] [CrossRef]
  145. García-Peris, P.; Velasco, C.; Lozano, M.A.; Moreno, Y.; Paron, L.; de la Cuerda, C.; Bretón, I.; Camblor, M.; García-Hernández, J.; Guarner, F.; et al. Effect of a mixture of inulin and fructo-oligosaccharide on Lactobacillus and Bifidobacterium intestinal microbiota of patients receiving radiotherapy: A randomised, double-blind, placebo-controlled trial. Nutr. Hosp. 2012, 27, 1908–1915. [Google Scholar]
  146. 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]
  147. Fuccio, L.; Frazzoni, L.; Guido, A. Prevention of pelvic radiation disease. World J. Gastrointest. Pharmacol. Ther. 2015, 6, 1–9. [Google Scholar] [CrossRef]
  148. Delia, P.; Sansotta, G.; Donato, V.; Messina, G.; Frosina, P.; Pergolizzi, S.; De Renzis, C. Prophylaxis of diarrhoea in patients submitted to radiotherapeutic treatment on pelvic district: Personal experience. Dig. Liver Dis. 2002, 34 (Suppl. 2), S84–S86. [Google Scholar] [CrossRef]
  149. Delia, P.; Sansotta, G.; Donato, V.; Frosina, P.; Messina, G.; De Renzis, C.; Famularo, G. Use of probiotics for prevention of radiation-induced diarrhea. World J. Gastroenterol. 2007, 13, 912–915. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  150. Salminen, E.; Elomaa, I.; Minkkinen, J.; Vapaatalo, H.; Salminen, S. Preservation of intestinal integrity during radiotherapy using live Lactobacillus acidophilus cultures. Clin. Radiol. 1988, 39, 435–437. [Google Scholar] [CrossRef]
  151. Urbancsek, H.; Kazar, T.; Mezes, I.; Neumann, K. Results of a double-blind, randomized study to evaluate the efficacy and safety of Antibiophilus in patients with radiation-induced diarrhoea. Eur. J. Gastroenterol. Hepatol. 2001, 13, 391–396. [Google Scholar] [CrossRef] [PubMed]
  152. Chitapanarux, I.; Chitapanarux, T.; Traisathit, P.; Kudumpee, S.; Tharavichitkul, E. Randomized controlled trial of live lactobacillus acidophilus plus bifidobacterium bifidum in prophylaxis of diarrhea during radiotherapy in cervical cancer patients. Radiat. Oncol. 2010, 5, 31. [Google Scholar] [CrossRef] [PubMed]
  153. Österlund, P.; Ruotsalainen, T.; Korpela, R.; Saxelin, M.; Ollus, A.; Valta, P.; Kouri, M.; Elomaa, I.; Joensuu, H. Lactobacillus supplementation for diarrhoea related to chemotherapy of colorectal cancer: A randomised study. Br. J. Cancer 2007, 97, 1028. [Google Scholar] [CrossRef]
  154. Germain, I.; Desjardins, J.; Demers, M.; Dagnault, A. Phase III Study: Impact of Probiotics on Diarrhea in Patients Treated with Pelvic Radiation. Int. J. Radiat. Oncol. 2011, 81, S667–S668. [Google Scholar] [CrossRef]
  155. Demers, M.; Dagnault, A.; Desjardins, J. A randomized double-blind controlled trial: Impact of probiotics on diarrhea in patients treated with pelvic radiation. Clin. Nutr. 2014, 33, 761–767. [Google Scholar] [CrossRef]
  156. Giralt, J.; Regadera, J.P.; Verges, R.; Romero, J.; de la Fuente, I.; Biete, A.; Villoria, J.; Cobo, J.M.; Guarner, F. Effects of probiotic Lactobacillus casei DN-114 001 in prevention of radiation-induced diarrhea: Results from multicenter, randomized, placebo-controlled nutritional trial. Int. J. Radiat. Oncol. Biol. Phys. 2008, 71, 1213–1219. [Google Scholar] [CrossRef]
  157. Timko, J. Probiotics as prevention of radiation-induced diarrhoea. J. Radiother. Pract. 2010, 9, 201–208. [Google Scholar] [CrossRef]
  158. Doron, S.; Snydman, D.R. Risk and safety of probiotics. Clin. Infect. Dis. 2015, 60 (Suppl. 2), S129–S134. [Google Scholar] [CrossRef]
  159. Oggioni, M.R.; Pozzi, G.; Valensin, P.E.; Galieni, P.; Bigazzi, C. Recurrent septicemia in an immunocompromised patient due to probiotic strains of Bacillus subtilis. J. Clin. Microbiol. 1998, 36, 325–326. [Google Scholar]
  160. Besselink, M.G.; van Santvoort, H.C.; Buskens, E.; Boermeester, M.A.; van Goor, H.; Timmerman, H.M.; Nieuwenhuijs, V.B.; Bollen, T.L.; van Ramshorst, B.; Witteman, B.J.; et al. Probiotic prophylaxis in predicted severe acute pancreatitis: A randomised, double-blind, placebo-controlled trial. Lancet 2008, 371, 651–659. [Google Scholar] [CrossRef]
  161. Lebeer, S.; Vanderleyden, J.; De Keersmaecker, S.C.J. Genes and Molecules of Lactobacilli Supporting Probiotic Action. Microbiol. Mol. Biol. Rev. 2008, 72, 728–764. [Google Scholar] [CrossRef] [PubMed]
  162. Lee, H.; Ko, G. New perspectives regarding the antiviral effect of vitamin A on norovirus using modulation of gut microbiota. Gut Microbes 2017, 8, 616–620. [Google Scholar] [CrossRef] [PubMed]
  163. Lee, H.; Ko, G.P. Antiviral effect of Vitamin A on norovirus infection via modulation of the gut microbiome. Sci. Rep. 2016, 6, 1–9. [Google Scholar] [CrossRef]
  164. Miki, T.; Goto, R.; Fujimoto, M.; Okada, N.; Hardt, W.D. The Bactericidal Lectin RegIIIβ Prolongs Gut Colonization and Enteropathy in the Streptomycin Mouse Model for Salmonella Diarrhea. Cell Host Microbe 2017, 21, 195–207. [Google Scholar] [CrossRef] [PubMed]
  165. Kanjan, P.; Hongpattarakere, T. Prebiotic efficacy and mechanism of inulin combined with inulin-degrading Lactobacillus paracasei I321 in competition with Salmonella. Carbohydr. Polym. 2017, 169, 236–244. [Google Scholar] [CrossRef]
  166. Hino, S.; Takemura, N.; Sonoyama, K.; Morita, A.; Kawagishi, H.; Aoe, S.; Morita, T. Small Intestinal Goblet Cell Proliferation Induced by Ingestion of Soluble and Insoluble Dietary Fiber Is Characterized by An Increase in Sialylated Mucins in Rats. J. Nutr. 2012, 142, 1429–1436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  167. Ishii, K.; Kono, H.; Hosomura, N.; Tsuchiya, M.; Ohgiku, M.; Tanaka, N.; Fujii, H. Medium-chain triglycerides enhance mucous secretion and cell proliferation in the rat. J. Gastroenterol. 2009, 44, 204–211. [Google Scholar] [CrossRef]
  168. Moran, E.T. Nutrients central to maintaining intestinal absorptive efficiency and barrier integrity with fowl. Poult. Sci. 2017, 96, 1348–1363. [Google Scholar] [CrossRef]
  169. Guilloteau, P.; Martin, L.; Eeckhaut, V.; Ducatelle, R.; Zabielski, R.; Van Immerseel, F. From the gut to the peripheral tissues: The multiple effects of butyrate. Nutr. Res. Rev. 2010, 23, 366–384. [Google Scholar] [CrossRef]
  170. Hamer, H.M.; Jonkers, D.; Venema, K.; Vanhoutvin, S.; Troost, F.J.; Brummer, R.-J. Review article: The role of butyrate on colonic function. Aliment Pharmacol. Ther. 2008, 27, 104–119. [Google Scholar] [CrossRef]
  171. Tan, J.; McKenzie, C.; Potamitis, M.; Thorburn, A.N.; Mackay, C.R.; Macia, L. The Role of Short-Chain Fatty Acids in Health and Disease. Adv. Immunol. 2014, 121, 91–119. [Google Scholar]
  172. Huang, C.; Song, P.; Fan, P.; Hou, C.; Thacker, P.; Ma, X. Dietary Sodium Butyrate Decreases Postweaning Diarrhea by Modulating Intestinal Permeability and Changing the Bacterial Communities in Weaned Piglets. J. Nutr. 2015, 145, 2774–2780. [Google Scholar] [CrossRef] [Green Version]
  173. Vernia, P.; Fracasso, P.L.; Casale, V.; Villotti, G.; Marcheggiano, A.; Stigliano, V.; Pinnaro, P.; Bagnardi, V.; Caprilli, R. Topical butyrate for acute radiation proctitis: Randomised, crossover trial. Lancet 2000, 356, 1232–1235. [Google Scholar] [CrossRef]
  174. Maggio, A.; Magli, A.; Rancati, T.; Fiorino, C.; Valvo, F.; Fellin, G.; Ricardi, U.; Munoz, F.; Cosentino, D.; Cazzaniga, L.F.; et al. Daily Sodium Butyrate Enema for the Prevention of Radiation Proctitis in Prostate Cancer Patients Undergoing Radical Radiation Therapy: Results of a Multicenter Randomized Placebo-Controlled Dose-Finding Phase 2 Study. Int. J. Radiat. Oncol. Biol. Phys. 2018, 89, 518–524. [Google Scholar] [CrossRef]
  175. Buffie, C.G.; Pamer, E.G. Microbiota-mediated colonization resistance against intestinal pathogens. Nat. Rev. Immunol. 2013, 13, 790–801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. De Keersmaecker, S.C.J.; Verhoeven, T.L.A.; Desair, J.; Marchal, K.; Vanderleyden, J.; Nagy, I. Strong antimicrobial activity of Lactobacillus rhamnosus GG against Salmonella typhimurium is due to accumulation of lactic acid. FEMS Microbiol. Lett. 2006, 259, 89–96. [Google Scholar] [CrossRef] [PubMed]
  177. Durant, J.A.; Corrier, D.E.; Ricke, S.C. Short-chain volatile fatty acids modulate the expression of the hilA and invF genes of Salmonella typhimurium. J. Food Prot. 2000, 63, 573–578. [Google Scholar] [CrossRef] [PubMed]
  178. Alakomi, H.; Skyttä, E.; Saarela, M.; Helander, I.M. Lactic Acid Permeabilizes Gram-Negative Bacteria by Disrupting the Outer Membrane Lactic Acid Permeabilizes Gram-Negative Bacteria by Disrupting the Outer Membrane. Appl. Environ. Microbiol. 2005, 66, 2000–2005. [Google Scholar]
  179. Presser, K.A.; Ratkowsky, D.A.; Ross, T. Modelling the growth rate of Escherichia coli as a function of pH and lactic acid concentration. Appl. Environ. Microbiol. 1997, 63, 2355–2360. [Google Scholar] [PubMed]
  180. Doron, S.; Gorbach, S.L. Probiotics: Their role in the treatment and prevention of disease. Expert Rev. Anti Infect. Ther. 2006, 4, 261–275. [Google Scholar] [CrossRef]
  181. Eijsink, V.G.H.; Axelsson, L.; Diep, D.B.; Havarstein, L.S.; Holo, H.; Nes, I.F. Production of class II bacteriocins by lactic acid bacteria; an example of biological warfare and communication. Antonie van Leeuwenhoek 2002, 81, 639–654. [Google Scholar] [CrossRef] [PubMed]
  182. Alvarez-Olmos, M.I.; Oberhelman, R.A. Probiotic agents and infectious diseases: A modern perspective on a traditional therapy. Clin. Infect. Dis. 2001, 32, 1567–1576. [Google Scholar] [CrossRef] [PubMed]
  183. Zhang, D.; Li, R.; Li, J. Lactobacillus reuteri ATCC 55730 and L22 display probiotic potential in vitro and protect against Salmonella-induced pullorum disease in a chick model of infection. Res. Vet. Sci. 2012, 93, 366–373. [Google Scholar] [CrossRef]
  184. Cazorla, S.I.; Maldonado-Galdeano, C.; Weill, R.; De Paula, J.; Perdigon, G.D.V. Oral Administration of Probiotics Increases Paneth Cells and Intestinal Antimicrobial Activity. Front. Microbiol. 2018, 9, 736. [Google Scholar] [CrossRef]
  185. Gorbunov, N.V.; Garrison, B.R.; Kiang, J.G. Response of crypt paneth cells in the small intestine following total-body gamma-irradiation. Int. J. Immunopathol. Pharmacol. 2010, 23, 1111–1123. [Google Scholar] [CrossRef] [PubMed]
  186. Hudault, S.; Lievin, V.; Bernet-Camard, M.F.; Servin, A.L. Antagonistic activity exerted in vitro and in vivo by Lactobacillus casei (strain GG) against Salmonella typhimurium C5 infection. Appl. Environ. Microbiol. 1997, 63, 513–518. [Google Scholar]
  187. Bernet-Camard, M.F.; Lievin, V.; Brassart, D.; Neeser, J.R.; Servin, A.L.; Hudault, S. The human Lactobacillus acidophilus strain LA1 secretes a nonbacteriocin antibacterial substance(s) active in vitro and in vivo. Appl. Environ. Microbiol. 1997, 63, 2747–2753. [Google Scholar] [PubMed]
  188. Le Bouguenec, C. Adhesins and invasins of pathogenic Escherichia coli. Int. J. Med. Microbiol. 2005, 295, 471–478. [Google Scholar] [CrossRef] [PubMed]
  189. Schachtsiek, M.; Hammes, W.P.; Hertel, C. Characterization of Lactobacillus coryniformis DSM 20001 T Surface Protein Cpf Mediating Coaggregation with and Aggregation among Pathogens Characterization of Lactobacillus coryniformis DSM 20001 T Surface Protein Cpf Mediating Coaggregatio. Appl. Environ. Microbiol. 2004, 70, 7078–7085. [Google Scholar] [CrossRef]
  190. Bergonzelli, G.E.; Granato, D.; Pridmore, R.D.; Marvin-Guy, L.F.; Donnicola, D. GroEL of Lactobacillus johnsoni La1 (NCC533) Is cell surface Associated: Potential Role in Interactions with the Host and the Gastric Pathogen Helicobacter pylori. Infect. Immun. 2006, 74, 425–434. [Google Scholar] [CrossRef]
  191. Vollbracht, C.; Schneider, B.; Leendert, V.; Weiss, G.; Auerbach, L.; Beuth, J. Intravenous Vitamin C Administration Improves Quality of Life in Breast Cancer Patients during Chemo-/Radiotherapy and Aftercare: Results of a Retrospective, Multicentre, Epidemiological Cohort Study in Germany. In Vivo 2011, 25, 983–990. [Google Scholar]
  192. Padmanabhan, S.; Waly, M.I.; Taranikanti, V.; Guizani, N.; Ali, A.; Rahman, M.S.; Al-Attabi, Z.; Al-Malky, R.N.; Al-Maskari, S.N.M.; Al-Ruqaishi, B.R.S.; et al. Folate/Vitamin B12 Supplementation Combats Oxidative Stress-Associated Carcinogenesis in a Rat Model of Colon Cancer. Nutr. Cancer 2018. [Google Scholar] [CrossRef] [PubMed]
  193. Wang, Y.; Mumm, J.B.; Herbst, R.; Kolbeck, R.; Wang, Y. IL-22 Increases Permeability of Intestinal Epithelial Tight Junctions by Enhancing Claudin-2 Expression. J. Immunol. 2017, 199, 3316–3325. [Google Scholar] [CrossRef] [PubMed]
  194. Nguyen, V.; Pearson, K.; Kim, J.-H.; Kamdar, K.; DePaolo, R.W. Retinoic acid can exacerbate T cell intrinsic TLR2 activation to promote tolerance. PLoS ONE 2015, 10, e0118875. [Google Scholar] [CrossRef] [PubMed]
  195. Reifen, R.; Levy, E.; Berkovich, Z.; Tirosh, O. Vitamin A exerts its antiinflammatory activities in colitis through preservation of mitochondrial activity. Nutrition 2015, 31, 1402–1407. [Google Scholar] [CrossRef] [PubMed]
  196. Cha, H.-R.; Chang, S.-Y.; Chang, J.-H.; Kim, J.-O.; Yang, J.-Y.; Kim, C.-H.; Kweon, M.-N. Downregulation of Th17 Cells in the Small Intestine by Disruption of Gut Flora in the Absence of Retinoic Acid. J. Immunol. 2010, 184, 6799–6806. [Google Scholar] [CrossRef] [Green Version]
  197. Roche, M.; Neti, P.V.S.V.; Kemp, F.W.; Azzam, E.I.; Ferraris, R.P.; Howell, R.W. High Levels of Dietary Supplement Vitamins A, C and E are Absorbed in the Small Intestine and Protect Nutrient Transport Against Chronic Gamma Irradiation. Radiat. Res. 2015, 184, 470–481. [Google Scholar] [CrossRef] [Green Version]
  198. Chen, H.L.; Wang, C.H.; Kuo, Y.W.; Tsai, C.H. Antioxidative and hepatoprotective effects of fructo-oligosaccharide in d-galactose-treated Balb/cJ mice. Br. J. Nutr. 2011, 105, 805–809. [Google Scholar] [CrossRef]
  199. Yen, C.H.; Kuo, Y.W.; Tseng, Y.H.; Lee, M.C.; Chen, H.L. Beneficial effects of fructo-oligosaccharides supplementation on fecal bifidobacteria and index of peroxidation status in constipated nursing-home residents-A placebo-controlled, diet-controlled trial. Nutrition 2011, 27, 323–328. [Google Scholar] [CrossRef]
  200. Lee, M.T.; Lin, W.C.; Yu, B.; Lee, T.T. Antioxidant capacity of phytochemicals and their potential effects on oxidative status in animals—A review. Asian-Australas. J. Anim. Sci. 2017, 30, 299–308. [Google Scholar] [CrossRef]
  201. Wood, S.M.; Mastaloudis, A.F.; Hester, S.N.; Gray, R.; Kern, D.; Namkoong, J.; Draelos, Z.D. Protective effects of a novel nutritional and phytonutrient blend on ultraviolet radiation-induced skin damage and inflammatory response through aging defense mechanisms. J. Cosmet. Dermatol. 2017, 16, 491–499. [Google Scholar] [CrossRef]
  202. Wang, W.; Li, Z.; Han, Q.; Guo, Y.; Zhang, B.; D’Inca, R. Dietary live yeast and mannan-oligosaccharide supplementation attenuate intestinal inflammation and barrier dysfunction induced by Escherichia coli in broilers. Br. J. Nutr. 2016, 116, 1878–1888. [Google Scholar] [CrossRef] [PubMed]
  203. Akbari, P.; Braber, S.; Alizadeh, A.; Verheijden, K.A.; Schoterman, M.H.; Kraneveld, A.D.; Garssen, J.; Fink-Gremmels, J. Galacto-oligosaccharides Protect the Intestinal Barrier by Maintaining the Tight Junction Network and Modulating the Inflammatory Responses after a Challenge with the Mycotoxin Deoxynivalenol in Human Caco-2 Cell Monolayers and B6C3F1 Mice. J. Nutr. 2015, 145, 1604–1613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  204. Christensen, E.G.; Licht, T.R.; Leser, T.D.; Bahl, M.I. Dietary Xylo-oligosaccharide stimulates intestinal bifidobacteria and lactobacilli but has limited effect on intestinal integrity in rats. BMC Res. Notes 2014, 7, 1–14. [Google Scholar] [CrossRef]
  205. Amasheh, M.; Schlichter, S.; Amasheh, S.; Mankertz, J.; Zeitz, M.; Fromm, M.; Schulzke, J.D. Quercetin enhances epithelial barrier function and increases claudin-4 expression in Caco-2 cells. J. Nutr. 2008, 138, 1067–1073. [Google Scholar] [CrossRef] [PubMed]
  206. Suzuki, T.; Hara, H. Quercetin enhances intestinal barrier function through the assembly of zonula occludens-2, occludin, and claudin-1 and the expression of claudin-4 in Caco-2 cells. J. Nutr. 2009, 139, 965–974. [Google Scholar] [CrossRef]
  207. Lin, M.; Yen, C. Antioxidative ability of pactic acid bacteria. J. Agric. Food Chem. 1999, 47, 1460–1466. [Google Scholar] [CrossRef]
  208. Lee, J.; Hwang, K.-T.; Heo, M.-S.; Lee, J.-H.; Park, K.-Y. Resistance of Lactobacillus plantarum KCTC 3099 from Kimchi to oxidative stress. J. Med. Food 2005, 8, 299–304. [Google Scholar] [CrossRef]
  209. Li, S.; Huang, R.; Shah, N.P.; Tao, X.; Xiong, Y.; Wei, H. Antioxidant and antibacterial activities of exopolysaccharides from Bifidobacterium bifidum WBIN03 and Lactobacillus plantarum R315. J. Dairy Sci. 2014, 97, 7334–7343. [Google Scholar] [CrossRef] [PubMed]
  210. 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] [PubMed]
  211. Li, S.; Shah, N.P. Antioxidant and antibacterial activities of sulphated polysaccharides from Pleurotus eryngii and Streptococcus thermophilus ASCC 1275. Food Chem. 2014, 165, 262–270. [Google Scholar] [CrossRef] [PubMed]
  212. Li, W.; Ji, J.; Rui, X.; Yu, J.; Tang, W.; Chen, X.; Jiang, M.; Dong, M. Production of exopolysaccharides by Lactobacillus helveticus MB2-1 and its functional characteristics in vitro. LWT Food Sci. Technol. 2014, 59, 732–739. [Google Scholar] [CrossRef]
  213. London, L.E.E.; Kumar, A.H.S.; Wall, R.; Casey, P.G.; Sullivan, O.O.Õ.; Shanahan, F.; Hill, C.; Cotter, P.D.; Fitzgerald, G.F.; Ross, R.P.; et al. Exopolysaccharide-Producing Probiotic Lactobacilli Reduce Serum Cholesterol and Modify Enteric Microbiota in ApoE deficient mice. J. Nutr. 2014, 144, 1956–1962. [Google Scholar] [CrossRef] [PubMed]
  214. Wu, M.H.; Pan, T.M.; Wu, Y.J.; Chang, S.J.; Chang, M.S.; Hu, C.Y. Exopolysaccharide activities from probiotic Bifidobacterium: Immunomodulatory effects (on J774A.1 macrophages) and antimicrobial properties. Int. J. Food Microbiol. 2010, 144, 104–110. [Google Scholar] [CrossRef]
  215. Ballal, S.A.; Veiga, P.; Fenn, K.; Michaud, M.; Kim, J.H.; Gallini, C.A.; Glickman, J.N.; Quéré, G.; Garault, P.; Béal, C.; et al. Host lysozyme-mediated lysis of Lactococcus lactis facilitates delivery of colitis-attenuating superoxide dismutase to inflamed colons. Proc. Natl. Acad. Sci. USA 2015, 112, 7803–7808. [Google Scholar] [CrossRef]
  216. Fanning, S.; Hall, L.J.; Cronin, M.; Zomer, A.; MacSharry, J.; Goulding, D.; Motherway, M.O.; Shanahan, F.; Nally, K.; Dougan, G.; et al. Bifidobacterial surface-exopolysaccharide facilitates commensal-host interaction through immune modulation and pathogen protection. Proc. Natl. Acad. Sci. USA 2012, 109, 2108–2113. [Google Scholar] [CrossRef] [Green Version]
  217. Dertli, E.; Colquhoun, I.J.; Gunning, A.P.; Bongaerts, R.J.; Le Gall, G.; Bonev, B.B.; Mayer, M.J.; Narbad, A. Structure and biosynthesis of two exopolysaccharides produced by Lactobacillus johnsonii FI9785. J. Biol. Chem. 2013, 288, 31938–31951. [Google Scholar] [CrossRef]
  218. Lee, I.-C.; Caggianiello, G.; van Swam, I.I.; Taverne, N.; Meijerink, M.; Bron, P.A.; Spano, G.; Kleerebezem, M. Strain-Specific Features of Extracellular Polysaccharides and Their Impact on Lactobacillus plantarum-Host Interactions. Appl. Environ. Microbiol. 2016, 82, 3959–3970. [Google Scholar] [CrossRef] [PubMed]
  219. Le, M.H.A.; Galle, S.; Yang, Y.; Landero, J.L.; Beltranena, E.; Gänzle, M.G.; Zijlstra, R.T. Effects of feeding fermented wheat with Lactobacillus reuteri on gut morphology, intestinal fermentation, nutrient digestibility, and growth performance in weaned pigs. J. Anim. Sci. 2016, 94, 4677–4687. [Google Scholar] [CrossRef]
  220. Zhang, J.; Zhao, X.; Jiang, Y.; Zhao, W.; Guo, T.; Cao, Y.; Teng, J.; Hao, X.; Zhao, J.; Yang, Z. Antioxidant status and gut microbiota change in an aging mouse model as influenced by exopolysaccharide produced by Lactobacillus plantarum YW11 isolated from Tibetan kefir. J. Dairy Sci. 2017, 100, 6025–6041. [Google Scholar] [CrossRef]
  221. LeBlanc, J.G.; del Carmen, S.; Miyoshi, A.; Azevedo, V.; Sesma, F.; Langella, P.; Bermudez-Humaran, L.G.; Watterlot, L.; Perdigon, G.; de Moreno de LeBlanc, A. Use of superoxide dismutase and catalase producing lactic acid bacteria in TNBS induced Crohn’s disease in mice. J. Biotechnol. 2011, 151, 287–293. [Google Scholar] [CrossRef] [PubMed]
  222. Spyropoulos, B.G.; Misiakos, E.P.; Fotiadis, C.; Stoidis, C.N. Antioxidant properties of probiotics and their protective effects in the pathogenesis of radiation-induced enteritis and colitis. Dig. Dis. Sci. 2011, 56, 285–294. [Google Scholar] [CrossRef] [PubMed]
  223. Wuyts, S.; Wittouck, S.; De Boeck, I.; Allonsius, C.N.; Pasolli, E.; Segata, N.; Lebeer, S. Large-Scale Phylogenomics of the Lactobacillus casei Group Highlights Taxonomic Inconsistencies and Reveals Novel Clade-Associated Features. mSystems 2017, 2, e00061-17. [Google Scholar] [CrossRef]
  224. Wang, J.; Ji, H.F.; Wang, S.X.; Zhang, D.Y.; Liu, H.; Shan, D.C.; Wang, Y.M. Lactobacillus plantarum ZLP001: In vitro assessment of antioxidant capacity and effect on growth performance and antioxidant status in weaning piglets. Asian-Australas. J. Anim. Sci. 2012, 25, 1153–1158. [Google Scholar] [CrossRef]
  225. Dowarah, R.; Verma, A.K.; Agarwal, N.; Patel, B.H.M.; Singh, P. Effect of swine based probiotic on performance, diarrhoea scores, intestinal microbiota and gut health of grower-finisher crossbred pigs. Livest. Sci. 2017, 195, 74–79. [Google Scholar] [CrossRef]
  226. Aluwong, T.; Kawu, M.; Raji, M.; Dzenda, T.; Govwang, F.; Sinkalu, V.; Ayo, J. Effect of Yeast Probiotic on Growth, Antioxidant Enzyme Activities and Malondialdehyde Concentration of Broiler Chickens. Antioxidants 2013, 2, 326–339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  227. Klayraung, S.; Okonogi, S. Antibacterial and Antioxidant Activities of Acid and Bile Resistant Strains of Lactobacillus fermentum Isolated from Miang. Braz. J. Microbiol. 2009, 40, 757–766. [Google Scholar] [CrossRef] [PubMed]
  228. Uskova, M.A.; Kravchenko, L.V. Antioxidant properties of lactic acid bacteria—Probiotic and yogurt strains. Vopr. Pitan. 2009, 78, 18–23. [Google Scholar]
  229. Capcarova, M.; Weiss, J.; Hrncar, C.; Kolesarova, A.; Pal, G. Effect of Lactobacillus fermentum and Enterococcus faecium strains on internal milieu, antioxidant status and body weight of broiler chickens. J. Anim. Physiol. Anim. Nutr. 2010, 94, e215–e224. [Google Scholar] [CrossRef] [PubMed]
  230. Ejtahed, H.S.; Mohtadi-Nia, J.; Homayouni-Rad, A.; Niafar, M.; Asghari-Jafarabadi, M.; Mofid, V. Probiotic yogurt improves antioxidant status in type 2 diabetic patients. Nutrition 2012, 28, 539–543. [Google Scholar] [CrossRef] [PubMed]
  231. Buffinton, G.D.; Doe, W.F. Depleted mucosal antioxidant defences in inflammatory bowel disease. Free Radic. Biol. Med. 1995, 19, 911–918. [Google Scholar] [CrossRef]
  232. Loguercio, C.; D’Argenio, G.; Delle Cave, M.; Cosenza, V.; Della Valle, N.; Mazzacca, G.; Del Vecchio Blanco, C. Glutathione supplementation improves oxidative damage in experimental colitis. Dig. Liver Dis. 2003, 35, 635–641. [Google Scholar] [CrossRef]
  233. Lutgendorff, F.; Nijmeijer, R.M.; Sandstrom, P.A.; Trulsson, L.M.; Magnusson, K.-E.; Timmerman, H.M.; van Minnen, L.P.; Rijkers, G.T.; Gooszen, H.G.; Akkermans, L.M.A.; et al. Probiotics prevent intestinal barrier dysfunction in acute pancreatitis in rats via induction of ileal mucosal glutathione biosynthesis. PLoS ONE 2009, 4, e4512. [Google Scholar] [CrossRef] [PubMed]
  234. Witschi, A.; Reddy, S.; Stofer, B.; Lauterburg, B.H. The systemic availability of oral glutathione. Eur. J. Clin. Pharmacol. 1992, 43, 667–669. [Google Scholar] [CrossRef]
  235. Mansour, H.H.; Hafez, H.F.; Fahmy, N.M.; Hanafi, N. Protective effect of N-acetylcysteine against radiation induced DNA damage and hepatic toxicity in rats. Biochem. Pharmacol. 2008, 75, 773–780. [Google Scholar] [CrossRef]
  236. Wu, W.; Abraham, L.; Ogony, J.; Matthews, R.; Goldstein, G.; Ercal, N. Effects of N-acetylcysteine amide (NACA), a thiol antioxidant on radiation-induced cytotoxicity in Chinese hamster ovary cells. Life Sci. 2008, 82, 1122–1130. [Google Scholar] [CrossRef]
  237. Kau, A.L.; Ahern, P.P.; Griffin, N.W.; Goodman, A.L.; Gordon, J.I. Human nutrition, the gut microbiome, and immune system: Envisioning the future. Nature 2011, 474, 327–336. [Google Scholar] [CrossRef] [PubMed]
  238. Ma, N.; Wu, Y.; Xie, F.; Du, K.; Wang, Y.; Shi, L.; Ji, L.; Liu, T.; Ma, X. Dimethyl fumarate reduces the risk of mycotoxins via improving intestinal barrier and microbiota. Oncotarget 2017, 8, 44625–44638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  239. Pompei, A.; Cordisco, L.; Amaretti, A.; Zanoni, S.; Raimondi, S.; Matteuzzi, D.; Rossi, M. Administration of folate-producing Bifidobacteria enhances folate status in Wistar rats. J. Nutr. 2007, 137, 2742–2746. [Google Scholar] [CrossRef] [PubMed]
  240. Strozzi, G.P.; Mogna, L. Quantification of folic acid in human feces after administration of Bifidobacterium probiotic strains. J. Clin. Gastroenterol. 2008, 42 (Suppl. 3), S179–S184. [Google Scholar] [CrossRef]
  241. Garg, S.; Zheng, J.; Wang, J.; Authier, S.; Pouliot, M.; Hauer-Jensen, M. Segmental Differences in Radiation-Induced Alterations of Tight Junction-Related Proteins in Non-Human Primate Jejunum, Ileum and Colon. Radiat. Res. 2016, 185, 50–59. [Google Scholar] [CrossRef]
  242. Kiang, J.G.; Fukumoto, R.; Gorbunov, N.V. Lipid Peroxidation After Ionizing Irradiation Leads to Apoptosis and Autophagy. In Lipid Peroxidation; InTech: Rijeka, Croatia, 2012; pp. 261–278. [Google Scholar]
  243. Farquhar, M.G.; Palade, G.E. Junctional complexes in various epithelia. J. Cell Biol. 1963, 17, 375–412. [Google Scholar] [CrossRef] [PubMed]
  244. De Santis, S.; Cavalcanti, E.; Mastronardi, M.; Jirillo, E.; Chieppa, M. Nutritional Keys for Intestinal Barrier Modulation. Front. Immunol. 2015, 6, 612. [Google Scholar] [CrossRef]
  245. Ulluwishewa, D.; Anderson, R.C.; McNabb, W.C.; Moughan, P.J.; Wells, J.M.; Roy, N.C. Regulation of Tight Junction Permeability by Intestinal Bacteria and Dietary Components. J. Nutr. 2011, 141, 769–776. [Google Scholar] [CrossRef] [Green Version]
  246. La Fata, G.; Weber, P.; Mohajeri, M.H. Probiotics and the Gut Immune System: Indirect Regulation. Probiotics Antimicrob. Proteins 2018, 10, 11–21. [Google Scholar] [CrossRef]
  247. Anderson, R.C.; Cookson, A.L.; McNabb, W.C.; Park, Z.; McCann, M.J.; Kelly, W.J.; Roy, N.C. Lactobacillus plantarum MB452 enhances the function of the intestinal barrier by increasing the expression levels of genes involved in tight junction formation. BMC Microbiol. 2010, 10, 316. [Google Scholar] [CrossRef] [PubMed]
  248. Anderson, R.C.; Cookson, A.L.; McNabb, W.C.; Kelly, W.J.; Roy, N.C. Lactobacillus plantarum DSM 2648 is a potential probiotic that enhances intestinal barrier function. FEMS Microbiol. Lett. 2010, 309, 184–192. [Google Scholar] [CrossRef] [PubMed]
  249. Qin, H.; Zhang, Z.; Hang, X.; Jiang, Y.L. plantarum prevents Enteroinvasive Escherichia coli-induced tight junction proteins changes in intestinal epithelial cells. BMC Microbiol. 2009, 9, 63. [Google Scholar] [CrossRef]
  250. Trivedi, K.; Barrett, K.E.; Silvia, C.R.-L. Probiotic inhibition of the entry of enteroinvasive E. coli into, human intestinal epithelial cells involves both Rho-dependent and -independent pathways. Gastroenterology 2003, 124, A106. [Google Scholar] [CrossRef]
  251. Shi, Y.; Zhao, X.; Zhao, J.; Zhang, H.; Zhai, Q.; Narbad, A.; Chen, W. A mixture of Lactobacillus species isolated from traditional fermented foods promote recovery from antibiotic-induced intestinal disruption in mice. J. Appl. Microbiol. 2018, 124, 842–854. [Google Scholar] [CrossRef]
  252. Cornick, S.; Tawiah, A.; Chadee, K. Roles and regulation of the mucus barrier in the gut. Tissue barriers. 2015, 3, e982426. [Google Scholar] [CrossRef] [Green Version]
  253. Jang, H.; Park, S.; Lee, J.; Myung, J.K.; Jang, W.S.; Lee, S.J.; Myung, H.; Lee, C.; Kim, H.; Lee, S.S.; et al. Rebamipide alleviates radiation-induced colitis through improvement of goblet cell differentiation in mice. J. Gastroenterol. Hepatol. 2018, 33, 878–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  254. Celebioglu, H.; Svensson, B. Dietary Nutrients, Proteomes, and Adhesion of Probiotic Lactobacilli to Mucin and Host Epithelial Cells. Microorganisms 2018, 6, 90. [Google Scholar] [CrossRef] [PubMed]
  255. Kaur, S.; Sharma, P.; Kalia, N.; Singh, J.; Kaur, S. Anti-biofilm Properties of the Fecal Probiotic Lactobacilli Against Vibrio spp. Front. Cell Infect. Microbiol. 2018, 8, 120. [Google Scholar] [CrossRef] [PubMed]
  256. Mack, D.R.; Michail, S.; Wei, S.; McDougall, L.; Hollingsworth, M.A. Probiotics inhibit enteropathogenic E. coli adherence in vitro by inducing intestinal mucin gene expression. Am. J. Physiol. 1999, 276, G941–G950. [Google Scholar]
  257. Hafez, M.M. Upregulation of Intestinal Mucin Expression by the Probiotic Bacterium E. coli Nissle 1917. Probiotics Antimicrob. Proteins 2012, 4, 67–77. [Google Scholar] [CrossRef] [PubMed]
  258. Mattar, A.F.; Teitelbaum, D.H.; Drongowski, R.A.; Yongyi, F.; Harmon, C.M.; Coran, A.G. Probiotics up-regulate MUC-2 mucin gene expression in a Caco-2 cell-culture model. Pediatr. Surg. Int. 2002, 18, 586–590. [Google Scholar] [PubMed]
  259. Caballero-Franco, C.; Keller, K.; De Simone, C.; Chadee, K. The VSL#3 probiotic formula induces mucin gene expression and secretion in colonic epithelial cells. Am. J. Physiol. Gastrointest. Liver Physiol. 2007, 292, G315–G322. [Google Scholar]
  260. Tareb, R.; Bernardeau, M.; Gueguen, M.; Vernoux, J.P. In vitro characterization of aggregation and adhesion properties of viable and heat-killed forms of two probiotic Lactobacillus strains and interaction with foodborne zoonotic bacteria, especially Campylobacter jejuni. J. Med. Microbiol. 2013, 62, 637–649. [Google Scholar] [CrossRef]
  261. Jiang, H.; Przybyszewski, J.; Mitra, D.; Becker, C.; Brehm-Stecher, B.; Tentinger, A.; MacDonald, R.S. Soy Protein Diet, but Not Lactobacillus rhamnosus GG, Decreases Mucin-1, Trefoil Factor-3, and Tumor Necrosis Factor- in Colon of Dextran Sodium Sulfate-Treated C57BL/6 Mice. J. Nutr. 2011, 141, 1239–1246. [Google Scholar] [CrossRef] [PubMed]
  262. Li, X.H.; Ghosh, S.P.; Ha, C.T.; Fu, D.; Elliott, T.B.; Bolduc, D.L.; Villa, V.; Whitnall, M.H.; Landauer, M.R.; Xiao, M. Delta-tocotrienol protects mice from radiation-induced gastrointestinal injury. Radiat. Res. 2013, 180, 649–657. [Google Scholar] [CrossRef] [PubMed]
  263. Kominsky, D.J.; Keely, S.; Macmanus, C.F.; Glover, L.E.; Scully, M.; Collins, C.B.; Bowers, B.E.; Campbell, E.L.; Colgan, P. An Endogenously Anti-Inflammatory Role for Methylation in Mucosal Inflammation Identified through Metabolite Profiling. J. Immunol. 2011, 186, 6505–6514. [Google Scholar] [CrossRef] [Green Version]
  264. Sanguri, S.; Gupta, D. Mannan oligosaccharide requires functional ETC and TLR for biological radiation protection to normal cells. BMC Cell Biol. 2018, 19, 9. [Google Scholar] [CrossRef] [PubMed]
  265. Kim, S.O.; Sheikh, H.I.; Ha, S.D.; Martins, A.; Reid, G. G-CSF-mediated inhibition of JNK is a key mechanism for Lactobacillus rhamnosus-induced suppression of TNF production in macrophages. Cell Microbiol. 2006, 8, 1958–1971. [Google Scholar] [CrossRef]
  266. Roessler, A.; Friedrich, U.; Vogelsang, H.; Bauer, A.; Kaatz, M.; Hipler, U.C.; Schmidt, I.; Jahreis, G. The immune system in healthy adults and patients with atopic dermatitis seems to be affected differently by a probiotic intervention. Clin. Exp. Allergy 2008, 38, 93–102. [Google Scholar] [PubMed]
  267. Haller, D.; Serrant, P.; Granato, D.; Schiffrin, E.J.; Blum, S. Activation of human NK cells by staphylococci and lactobacilli requires cell contact-dependent costimulation by autologous monocytes. Clin. Diagn. Lab. Immunol. 2002, 9, 649–657. [Google Scholar] [CrossRef] [PubMed]
  268. Smits, H.H.; Engering, A.; Van Der Kleij, D.; De Jong, E.C.; Schipper, K.; Van Capel, T.M.M.; Zaat, B.A.J.; Yazdanbakhsh, M.; Wierenga, E.A.; Van Kooyk, Y.; et al. Selective probiotic bacteria induce IL-10-producing regulatory T cells in vitro by modulating dendritic cell function through dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin. J. Allergy Clin. Immunol. 2005, 115, 1260–1267. [Google Scholar] [CrossRef] [PubMed]
  269. Jeon, S.G.; Kayama, H.; Ueda, Y.; Takahashi, T.; Asahara, T.; Tsuji, H.; Tsuji, N.M.; Kiyono, H.; Ma, J.S.; Kusu, T.; et al. Probiotic Bifidobacterium breve induces IL-10-producing Tr1 cells in the colon. PLoS Pathog. 2012, 8, 1–15. [Google Scholar] [CrossRef]
Figure 1. Scheme of radiotherapy inducing interactions between epithelial and endothelial radiation injuries in the gastrointestinal tract resulting in acute events and late tissue fibrosis.
Figure 1. Scheme of radiotherapy inducing interactions between epithelial and endothelial radiation injuries in the gastrointestinal tract resulting in acute events and late tissue fibrosis.
Microorganisms 07 00097 g001
Table 1. Overview of experimental studies on probiotic strategies for improving general gastrointestinal functioning.
Table 1. Overview of experimental studies on probiotic strategies for improving general gastrointestinal functioning.
ProbioticGut Improving EffectType of StudyReferences
Individual L. acidophilus strains ADH or N2, or L. bulgaricus (strain not mentioned), or Streptococcus thermophiles (strain not mentioned)Survival in human stomach and adhesion to IECs In vitro
In vivo (healthy pigs and humans)
[98]
L. rhamnosus GG ATCC 53103Prevention of cytokine-induced apoptosis in IECs In vitro[99]
Prevention of E. coli-induced changes in epithelial barrier functionIn vitro[100]
Restoration of intestinal integrity of murine ileum through occludin expression In vivo (mice with alcoholic liver disease)[101]
Prevention of increased intestinal paracellular permeability in Caco-2 cells;
Restoration of tight junction proteins such as ZO-1 , claudin-1, and occludin
In vitro[102]
Induction of inflammatory tolerance of the intestinal mucosaIn vitro
In vivo (healthy mice)
[103,104]
Local dampening of innate immune responses with desensitization towards luminal antigensIn vitro [105]
Effective treatment of acute gastroenteritisIn vivo (children with acute gastroenteritis)[106]
Faster recovery of acute non-bloody diarrheaIn vivo (children with acute diarrhea)[107,108,109]
B. bifidum G9-1Induction of mucosal protective factors including MUC2-4 , TGFβ1 and TFF3 ;
Alleviation of diarrhea partly through protection against human rotavirus induced lesions
In vivo (mice pups with rotavirus gastroenteritis)[110]
L. plantarum WCFS1Restoration of tight junction proteins ZO-1 and occludinIn vitro
In vivo (healthy humans)
[111]
L. reuteri I5007Restoration of tight junction proteins claudin-1, occluding, and ZO-1 In vitro
In vivo (healthy piglets)
[112]
L. casei DN-114 001Prevention of transcription of numerous pro-inflammatory genes encoding cytokines, chemokines and adherence molecules In vitro[113]
Restoration of tight junction protein ZO-1 in Caco-2 cellsIn vitro[114]
Ultrabiotique®
(L. acidophilus + B. lactis + L. plantarum + B. breve)
Improvement of clinical symptoms and histological alterations;
Down regulation of nitric oxide production by peritoneal macrophages;
Enhancement of mucus production with modification of microflora
In vivo (mice with colitis)[115]
L. acidophilus (strain not mentioned)Overall attenuation of the severity of DSS -induced colitis, specifically by suppressing
pro-inflammatory cytokines
In vitro
In vivo (mice with colitis)
[116]
Mixture of Streptococcus thermophilus + L. acidophilus + L. bulgaricus (strains not mentioned)No improvement of acute diarrhea In vivo (children with acute diarrhea)[117]
IEC = intestinal epithelial cell; ZO-1 = zonula occludens-1; MUC2 = mucin 2; TGFβ1 = transforming growth factor 1; TFF3 = trefoil factor 3; DSS = dextran sulfate sodium.
Table 2. Overview of preclinical studies on probiotic strategies for improving radiation-induced GI toxicity in tumor-free rodents.
Table 2. Overview of preclinical studies on probiotic strategies for improving radiation-induced GI toxicity in tumor-free rodents.
ProbioticRadiation DoseMethod of SupplementationDuration of SupplementationResults of SupplementationReferences
Microflorana®-F
(L. acidophilus + L. helveticus + Bifidobacterium spp)
Abdominal X irradiation with
1 × 20 Gy
Oral gavage of 1 mL of probiotic solution three times dailyStarted seven days before the irradiation procedure and maintained until 14 days thereafterImproved overall survival;
Improved endotoxin levels;
Reduced incidence of bacterial contamination
[137]
L. acidophilus (strain not mentioned)Abdominal-pelvic irradiation with either 1 × 10, 15 or 20 GyOral gavage of 2 mL of probiotic solution (108 CFU ) Started six days before irradiation and maintained until three days thereafterImproved morphology of the small intestine after 10 or 15 Gy;
No improvements of jejunal villi height when irradiated with 15 Gy;
No improvements when irradiated with 20 Gy
[138]
L. rhamnosus GG
ATCC 53103
Whole body γ irradiation with
1 × 12 Gy
Oral gavage of probiotic solution (5 × 10⁷ CFU ), dailyThree consecutive days before irradiation Reduced epithelial apoptosis particularly at crypt bases;
Improved crypt survival;
No detectable shifts in bacterial compositions
[139]
Oral gavage or intraperitoneal injection of lipoteichoic acid
(5 mg/kg), a radioprotective agent in L. rhamnosus GG, daily
Three consecutive days before irradiationImproved small intestinal crypt survival[140]
Total abdominal X irradiation with
7 or 8 × 4 Gy
Intraperitoneal injection of lipoteichoic acid (5 mg/kg), a radioprotective agent in L. rhamnosus GG, dailyOne hour before each fractionated radiation doseImproved post-radiation weight recovery and survival
L. delbrueckii subsp. Bulgaricus B3 strainAbdominal-pelvic γ irradiation with
1 × 11 Gy
Oral gavage of 2 mL of probiotic solution
(1010 CFU /mL), daily
Seven consecutive days after irradiationReduced scores for inflammation and vascularity;
Accelerated healing;
Decreased bacterial translocation;
Reduced diarrhea
[141,142]
L. plantarum 299vLower abdominal X irradiation with 2 × 10 GyOral gavage of probiotic solution (2 × 109 CFU ),
twice daily
Started one day after irradiation and was continued throughout the experiment for a maximum of 15 days, except for the operation dayIncreased collagen content;
Decreased mucosal myeloperoxidase activity
[143]
CFU = colony-forming units.
Table 3. Overview of clinical studies on probiotic strategies for treating radiation induced gastrointestinal toxicity in cancer patients.
Table 3. Overview of clinical studies on probiotic strategies for treating radiation induced gastrointestinal toxicity in cancer patients.
ProbioticSummary of StudyMethod of SupplementationDuration of SupplementationResults References
L. acidophilus NCDO 1748Patients (n = 24) with gynecological tumors, 80 Gy A formulated drink
(≥2 × 109 CFU ) and 6.5% lactulose as bacterial substrate, once daily
Started five days prior to radiotherapy, daily throughout the radiotherapy period including the interval, and continued for 10 days thereafterReduced diarrhea[150]
L. casei DN-114 001Patients (n = 85) with gynecological tumors, 45–50 Gy with weekly cisplatin treatment (40 mg/m2)A formulated drink
(108 CFU /g), three times daily
Started one week prior to radiotherapyImproved stool consistency;
No significant differences in need for rescue anti-diarrheal therapy, neither diarrhea severity
[156]
Infloran®
(L. acidophilus and
B. bifidum)
Patients (n = 63) with locally advanced cervical cancer receiving 50 Gy with additional brachytherapy of four times 7 Gy with weekly cisplatin during radiotherapy procedureTwo oral capsules (2 × 109 CFU /g of each bacteria), twice dailyStarted 7 days prior to radiotherapy and maintained during radiotherapyReduced severity of diarrhea;Reduced need of rescue anti-diarrheal therapy;
Improved stool consistency
[152]
VSL#3®
(Streptococcus thermophilus BT01, B. breve BB02, B. longum BL03, B. infantis BI04, L. acidophilus BA05, L. plantarum BP06, L. paracasei BP07, L. delbrueckii subsp. bulgaricus BD08)
Patients (n = 190) with pelvic tumors, 60–70 Gy A formulation (450 × 109 CFU /g), three times dailyStarted on the first day of radiotherapy, for 6 to 7 consecutive weeks of therapyReduced number of patients suffering from radiation induced toxicity;
Reduced severity of toxicity
[148,149]
Patients (n = 239) with postoperative cervical, sigmoid or rectal tumors, 60–70 GyImproved number of bowel movements;
Delayed need for additional anti-diarrheal therapy
Bifilact®
(L. acidophilus LAC-361 and B. longum BB-536)
Patients (n = 246) with rectal, cervical, endometrial or prostatic cancers that had radiotherapy with or without surgery or chemotherapyOral capsules (1.3 × 1012 CFU ), twice daily or three times dailyStarted on the first day of radiotherapy and maintained up until the last day of radiotherapyReduced severity of diarrhea with standard dosing[154,155]
“5” Strain Dophilus®
(L. rhamnosus HA-111, B. Breve HA-129, L. acidophilus HA-122, B. longum HA-135, L. Casei HA-108)
Patients (n = 42) with abdominal-pelvic cancers who received post-operative radiotherapy or radiotherapy with chemotherapy, 50–67 GyOral capsules (6 × 1012 CFU ), twice dailyStarted on the first day of radiotherapy and maintained up until the last day of radiotherapyReduced incidence and severity of diarrhea [157]
Antibiophilus®
(L. rhamnosus LCR 35)
Patients (n = 206) with several lower abdominal and pelvic tumors, 50 GyOral capsules (1.5 × 109 CFU ), plus lactulose as bacterial substrate, three times dailyStarted in case of diarrhea and maintained up to one week, depending on the response of the diarrhea Improved number of bowel movements;
Improved fecal consistency;
[151]
Gefiluss®
(L. rhamnosus GG ATCC 53103)
Patients (n = 39) with colorectal cancers receiving 45–50.4 Gy and 24 weeks of 5-FU chemotherapyOral capsules (1–2 × 1010 CFU ), twice dailyStarted at the start of adjuvant 5-FU chemotherapy and maintained for 24 weeksReduced severity of diarrhea;
Less abdominal discomfort reported;
Lower need for hospital care;
Reduced need for chemotherapy dose adjustments due to bowel toxicity
[153]
CFU = colony-forming units.

Share and Cite

MDPI and ACS Style

Segers, C.; Verslegers, M.; Baatout, S.; Leys, N.; Lebeer, S.; Mastroleo, F. Food Supplements to Mitigate Detrimental Effects of Pelvic Radiotherapy. Microorganisms 2019, 7, 97. https://doi.org/10.3390/microorganisms7040097

AMA Style

Segers C, Verslegers M, Baatout S, Leys N, Lebeer S, Mastroleo F. Food Supplements to Mitigate Detrimental Effects of Pelvic Radiotherapy. Microorganisms. 2019; 7(4):97. https://doi.org/10.3390/microorganisms7040097

Chicago/Turabian Style

Segers, Charlotte, Mieke Verslegers, Sarah Baatout, Natalie Leys, Sarah Lebeer, and Felice Mastroleo. 2019. "Food Supplements to Mitigate Detrimental Effects of Pelvic Radiotherapy" Microorganisms 7, no. 4: 97. https://doi.org/10.3390/microorganisms7040097

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