Skip to main content
Cancer Medicine logoLink to Cancer Medicine
. 2024 Mar 8;13(4):e6865. doi: 10.1002/cam4.6865

Radiation enteropathy‐related depression: A neglectable course of disease by gut bacterial dysbiosis

Xinliang Liu 1, Ying Li 1, Meichen Gu 1, Tiankai Xu 1, Chuanlei Wang 2,, Pengyu Chang 1,
PMCID: PMC10923036  PMID: 38457257

Abstract

Radiation enteropathy (RE) is common in patients treated with radiotherapy for pelvic–abdominal cancers. Accumulating data indicate that gut commensal bacteria determine intestinal radiosensitivity. Radiotherapy can result in gut bacterial dysbiosis. Gut bacterial dysbiosis contributes to the pathogenesis of RE. Mild to moderate depressive symptoms can be observed in patients with RE in clinical settings; however, the rate of these symptoms has not been reported. Studies have demonstrated that gut bacterial dysbiosis induces depression. In the state of comorbidity, RE and depression may be understood as local and abscopal manifestations of gut bacterial disorders. The ability of comorbid depression to worsen inflammatory bowel disease (IBD) has long been demonstrated and is associated with dysfunction of cholinergic neural anti‐inflammatory pathways. There is a lack of direct evidence for RE comorbid with depression. It is widely accepted that RE shares similar pathophysiologic mechanisms with IBD. Therefore, we may be able to draw on the findings of the relationship between IBD and depression. This review will explore the relationship between gut bacteria, RE, and depression in light of the available evidence and indicate a method for investigating the mechanisms of RE combined with depression. We will also describe new developments in the treatment of RE with probiotics, prebiotics, and fecal microbial transplantation.

Keywords: depression, dysbiosis, gut bacteria, HPA, radiation enteropathy

1. INTRODUCTION

According to evidence‐based medicine (EBM) data, 52% of cancer patients require radiotherapy (RT) during their treatment course. 1 Radiation‐induced tissue injury remains a major challenge when performing RT, especially in tissues that are highly sensitive to ionizing radiation (such as the intestinal epithelium). 2 , 3 Statistically, the incidence of radiation enteropathy (RE) differs among various tumor types treated with RT, with 80%–90% observed in carcinoma of the cervix, 60% observed in endometrial cancer, and 50% observed in carcinoma of the vulva. 4 The clinical manifestations of RE typically include acute diarrhea, blood in the stool, chronic intestinal fibrosis, strictures, perforations, and fistulas, among other effects. 5

The pathogenesis of RE is complicated and typically includes intestinal stem cell apoptosis, endothelial injury, oxidative stress, and inflammation. 6 In fact, gut bacterial dysbiosis can greatly contribute to these processes. When RE occurs, bacterial overgrowth becomes a characteristic feature. 7 McLaughlin and Matsuzawa discovered that germ‐free (GF) mice outlived normal mice after abdominal irradiation in 1964, which elicited the study of gut microbiota in RE. 8 , 9 , 10 As research progressed, the relationship between the gut microbiota and intestinal radiosensitivity became clearer. 10 RT results in gut bacterial dysbiosis. 7 Gut bacterial dysbiosis can correspondingly increase intestinal radiosensitivity, thus worsening RE. 11 First, gut bacterial dysbiosis has pro‐inflammatory properties and can increase the levels of inflammatory factors. 12 Second, gut bacterial dysbiosis can degrade the mucus layer and disrupt the phospholipid bilayer of enterocytes to cause damage to the intestinal mucosa. 13 The translocation of gut microbiota caused by intestinal epithelial damage exacerbates the intestinal inflammatory response, which correspondingly disrupts the tight junctions between intestinal epithelial cells. 14 , 15 Finally, gut bacterial dysbiosis has the ability to cause oxidative stress. 11 These three processes affect and promote each other, thus forming a feedback loop. In reality, RE is essentially a systemic disease that is mainly due to abnormal metabolism of substances elicited by gut bacterial dysbiosis. Therefore, the restoration of gut bacterial dysbiosis to interrupt this feedback pathway is one of the therapeutic modalities for RE.

Depression is a group of affective psychiatric disorders with a main clinical feature characterized by a marked and persistent depressed mood that is inconsistent with the situation. The pathogenesis of depression is complex, including the monoaminergic transmitter deficiency hypothesis, the neuroinflammatory hypothesis, and HPA axis abnormalities. Recently, several studies have found that gut bacteria play an integral role in the development of depression. Gut bacterial dysbiosis, which is represented by a decline in Firmicutes (such as Lactobacillus) and an increase in Bacteroidetes and Proteobacteria, occurs in depressed patients. 16 The transplantation of feces from depressed mice to healthy mice has been shown to cause depressive behavior in the latter group of mice. 16 The metabolites of the intestinal flora and the composition of bacteria are key points of this idea. 16

Depression is often comorbid with a variety of disorders, such as constipation, irritable bowel syndrome, and inflammatory bowel disease (IBD). 17 Rome IV considered functional gastrointestinal disease to be a disorder of abnormal gut–brain interaction. 18 It has been reported that 25.2% of patients with IBD present with depressive symptoms. 19 It can be observed in clinical settings that mild to moderate depressive symptoms are present in RE, and its incidence has not yet been reported. Passive mood increases the risk of recurrence and progression of IBD. 20 The mechanisms underlying this comorbid relationship are related to abnormalities in the gut–brain axis, including inflammation and autonomic disorders. 21 In a mouse model of depression, impaired vagus nerve activity increased vulnerability to IBD. 22 In fact, cholinergic nerves have an anti‐inflammatory role in regulating innate immune cells, which is also known as the “cholinergic anti‐inflammatory pathway”. 23 After the central nervous system receives the inflammatory signal, it transmits the message to the vagus nerve, thus causing the release of Ach from the vagus nerve endings, which activates the alpha 7 subunit of the nicotinic acetylcholine receptor (α7nAchR) on the membrane of the immune cells and inhibits the synthesis and release of inflammatory factors (such as IL‐6 and TNF‐α) by regulating signaling pathways, such as NF‐kB and JAK/STAT3. 24 In addition, as early as 1988, a study reported that intravenous injections of choline‐chlorine (a precursor of acetylcholine) prior to radiotherapy improved survival in mice. 25 Another recent study also confirmed the ability of choline to alleviate doxorubicin‐induced cardiotoxicity by correcting oxidative stress and inflammation. 26

In recent years, with the development of high‐throughput sequencing technology, it has been found that gut bacteria are a key point in regulating the gut–brain axis; thus, the concept of the “microbiota‐gut‐brain axis” has been proposed. 27 Gut bacteria interact with the brain through neural, humoral, and immune pathways. The “microbiota‐gut‐brain axis” information exchange is bidirectional. Indeed, in a comorbid state, RE and depression may be understood as being local and distant manifestations of gut bacterial dysbiosis. RE and depression share some similar pathogenetic processes, such as the action of LPS on TLR4, which activates both locally infiltrating macrophages and microglia in the central nervous system, thus exerting pro‐inflammatory effects. 28 In addition, RE interacts with depression. However, direct evidence is lacking, but we may be able to draw on studies of the relationship between IBD and depression. This review will explore the relationship between gut bacteria, RE, and depression in the context of the available evidence and provide directions for studying the mechanisms of RE combined with depression. We will also describe new developments in the treatment of RE with probiotics, prebiotics, and fecal microbial transplantation.

2. GUT COMMENSAL BACTERIA AND RE PATHOGENESIS

2.1. Gut commensal bacteria impact intestinal radiosensitivity

The gut microbiota and its metabolites play a considerable role in modulating the growth and function of gut‐associated lymphoid tissues (GALT) and systemic immune systems. 29 , 30 , 31 Preclinical studies have shown that impaired development of lymphoid tissue and regulatory T cells (Treg) were observed in the gut of GF mice. 32 Nevertheless, all of these abnormalities are reversed after a few weeks of inoculation of GF mice with commensal bacteria. 33

Intriguingly, subclinical studies have shown that GF mice are more radiation resistant than common mice, 10 and the mechanism may involve the prominent disability of the intestinal immune response. McLaughlin and Matsuzawa found that GF mice survived significantly longer than common mice after whole‐body X‐ray irradiation. 9 , 10 Although Anderson's subsequent studies demonstrated inconsistent results, 34 several studies in recent years have confirmed Matsuzawa's viewpoint. For example, one study found that GF mice exhibited less apoptosis of endothelial cells when treated with gamma radiation compared to normal mice. 35 Mechanistically, gut microbiota can downregulate fasting‐induced adipose factor (Fiaf), which is also known as angiopoietin‐like protein 4 (a secreted protein of intestinal epithelial cell origin), which has the ability to promote angiogenesis and the maintenance of mucosal barrier integrity. 35 , 36 Similar to the germ‐free mouse assay, Zhao et al. created a near GF mouse intestinal state via pretreatment with antibiotics prior to radiotherapy and found that antibiotic pretreatment reduced the activation of the LPS/TLR4/MyD88/NF‐κB p65 and TGF‐β1/Smad‐3 signaling pathways to attenuate inflammation and reduce intestinal wall fibrosis, respectively, which ultimately mitigated RE. 37 In summary, the gut microbiota can affect intestinal radiosensitivity by modulating host immunity.

2.2. Ionizing irradiation induces gut bacterial dysbiosis

Perturbations of gut bacteria, which is also known as gut bacterial dysbiosis, have been proven to be associated with various diseases, such as mental disorders, metabolic syndromes such as obesity, and autoimmune diseases (see Figure 1). 38 , 39 , 40 Unfortunately, a multitude of trials have shown that RT induces changes in the composition and function of gut microbiota, 12 , 41 , 42 , 43 with reduced bacterial diversity 44 and altered abundance, 13 , 45 such as a decrease in the number of probiotics and an increase in the abundance of opportunistic pathogenic bacteria. 46 Depending on the type of viability and energy metabolism under different oxygen partial pressure conditions, the abundance of specialized anaerobes decreases, whereas parthenogenetic anaerobes, such as Enterobacteriaceae, are upregulated. 47 An early observational study, which included a total of 39 patients undergoing radical pelvic radiotherapy, showed that nearly 25% of patients developed bacterial overgrowth in the small intestine. 7 Another study showed that pelvic RT resulted in a 10% reduction in Firmicutes phyla but a 3% increase in Fusobacterium phyla. 45 Notably, there are no agreed diagnostic criteria for postradiotherapy dysbiosis. Several studies have shown that the degree of gut bacterial dysbiosis is related to the duration and site of irradiation. However, there is a question as to if the degree of dysbiosis is correlated with the irradiation dose. This question merits further investigation.

FIGURE 1.

FIGURE 1

Diseases associated with gut bacterial dysbiosis. Gut bacterial dysbiosis is associated with various diseases, such as mental disorders, metabolic syndromes, and autoimmune diseases.

The intestinal mucosa can secrete antimicrobial substances (also known as defensins) to prevent the overgrowth of pathogenic bacteria. 48 However, RT causes damage to the intestinal epithelium, which indirectly causes dysbiosis. 6 Second, different flora have different radiosensitivities to ionizing irradiation, 49 which may be due to disparities in the content of oxygen radical‐scavenging enzymes in different bacteria. 50 In addition, several studies have shown that the shaping of gut microbiota by RT is affected by differences in sex and time of treatment. 51 , 52

2.3. Intestinal radiosensitivity is increased by gut bacterial dysbiosis

Under normal physiologic conditions, gut commensal bacteria are able to form a “biological barrier” to prevent pathogenic bacteria from colonizing the intestinal epithelium, promote the expression of tight junctions in the intestinal epithelium and the formation of a mucus layer to maintain the integrity of the intestinal epithelial barrier, and increase the expression of anti‐inflammatory genes (such as IL‐10). 53 However, radiation‐induced gut bacterial dysbiosis is potentially pathogenic, which may increase intestinal susceptibility to irradiation injury 44 and promote the progression of RE (see Figure 2).

FIGURE 2.

FIGURE 2

Positive feedback between gut bacterial dysbiosis and radiation enteropathy (RE). Ionizing irradiation induces gut bacterial dysbiosis. Irradiation‐induced dysbiosis is pathogenic, which increases intestinal radiosensitivity and can aggravate RE, constituting vicious positive feedback.

Regarding the mechanism by which radiation‐induced gut bacterial dysbiosis enhances intestinal radiosensitivity, gut bacterial dysbiosis has potential pro‐inflammatory properties. Radiotherapy‐induced intestinal inflammation can alter the composition of the intestinal flora, thus promoting pro‐inflammatory bacteria and inhibiting the growth of anti‐inflammatory bacteria. 54 For example, Faecalibacterium prausnitzii was significantly reduced in a colitis model. 55 Gerassy‐Vainberg et al. 12 used an in vitro coculture model of epithelial cells and gut microbiota, in which gut microbiota from irradiated or unirradiated mice were cocultured with epithelial cells in a microaerobic environment. The results showed that the irradiated dysregulated flora significantly upregulated TNF‐α (p < 0.05) and IL‐1β (p < 0.01) levels in epithelial cells. This finding also demonstrates that FMT can transmit susceptibility to RE and provides evidence that radiation‐induced gut bacterial dysbiosis aggravates RE. 12 Similarly, in an in vitro monolayer epithelial cell coculture model, Wang et al. found that radiation‐induced dysbiosis can stimulate the release of inflammatory factors such as TNF‐α and IL‐1β. 56 In addition, the overgrowth of the flora causes reactive aggregation of TH17 cells with pro‐inflammatory properties in the intestine. 57 Given that the RE phenotype can be transferred through FMT, these experimental results show that gut bacterial dysbiosis may not merely be a result of RE. Instead, these data once again suggest that the gut microbiota may be crucial in the pathogenesis of RE. 58 Inflammatory signaling pathways between gut bacteria and RE are associated with Toll‐like receptors (TLRs) (see Table 1). Pathogen‐associated molecular patterns (PAMPs) from gut bacteria can be recognized by TLRs, which are one of the most important pattern recognition receptors, on the surface of the body's natural immune cells (such as dendritic cells and macrophages).

TABLE 1.

Summary of TLRs related to RE.

Receptor Ligand Effectivity
TLR2 59 Lactobacillus rhamnosus GG Improvement of crypt survival rate
TLR4 60 LPS, peptidoglycan Upregulation of inflammatory factor expression
TLR5 61 Salmonella flagellin Radioprotective activity
TLR9 62 Cytosine‐phosphate‐guanine (CpG) dinucleotides Radioprotective activity

Note: TLR3, TLR7, and TLR8 recognize viral RNAs and are thus not listed.

Second, gut bacterial dysbiosis impairs the intestinal epithelial barrier (IEB). The intestinal mucus layer, the main functional component of which is mucin‐2 (MUC2), is not only involved in constituting the first line of immune defense in the gastrointestinal tract but is also a source of energy for certain commensal bacteria. 63 , 64 Studies have shown that gut bacterial dysbiosis interferes with mucus layer formation to increase the contact of gut microbiota with immune cells in the intestinal wall. 13 In fact, the degradation of mucin significantly increases the risk of developing Crohn's disease. 65 In addition, radiation‐induced gut bacterial dysbiosis and the disturbance of intestinal epithelial lipid metabolism are related. 66 The intestinal epithelial cell membrane, which consists of a lipid bilayer, has an indispensable role in maintaining the integrity of the intestinal epithelial barrier. 67 A recent multiomics study showed that glycerophospholipid metabolism is most correlated with RE and that there is a significant functional correlation between gut microbiota dysbiosis and changes in intestinal epithelial lipid metabolites, such as Bacteroides‐diglyceride (DG) (r = +0.99) and Escherichia‐Shigella‐triglyceride (TG) (r = +0.99). 66 However, there are limitations to this multiomics correlation analysis, which refers to the fact that only a correlation between dysbiosis and lipid metabolites can be demonstrated (not a causal relationship). 66 Notably, irradiation‐induced dysregulated flora‐induced IL‐1β can disrupt the tight junctions of the intestinal epithelium and aggravate mucosal damage in mice. 14 , 15 In addition, a study by Tian et al. found that the waterfall release of TNF‐α is capable of leading to intense oxidative stress and activation of the caspase‐3 pathway, which subsequently induces apoptosis of gut mucosal cells. 68

Finally, gut bacterial dysbiosis can contribute to oxidative stress. 11 Oxidative stress is the production of more pro‐oxidative substances, such as reactive oxygen species (ROS), compared to their antioxidative counterparts. 69 Oxidative stress can be directly induced by the gut microbiota, such as hydroxyl radicals produced by Enterococcus faecalis and the release of ROS from intestinal epithelial cells (IECs) promoted by Peptostreptococcus anaerobius and Escherichia coli. 69 Conversely, oxidative stress can destabilize homeostasis of the intestinal microbial environment. Notably, inflammatory factors secondary to gut bacterial dysbiosis, such as IL‐1β, IL‐6, and TNF‐α, can promote the aggregation of neutrophils to release ROS (reviewed by Zhang 70 ).

Gut bacterial dysbiosis can affect intestinal radiosensitivity through the abovementioned pathways, and there may be interactions between the different pathways. Taken together, irradiation‐induced dysbiosis is pathogenic, which correspondingly increases intestinal radiosensitivity. 54

3. ASSOCIATION BETWEEN DEPRESSION AND GUT MICROBIOTA

Neufeld found that GF mice had less depressive behavior than specific pathogen‐free (SPF) mice, 71 thus suggesting a nonnegligible role of the gut microbiota in depression. As with RE, there is gut bacterial dysbiosis in depression. 16 , 72 For example, Bifidobacteria, Dialister, and Coprococcus spp. were significantly reduced, whereas segmented filamentous bacteria (SFB) were increased. 73 Table 2 below depicts a summary of the changes in gut microbiota in depressed patients.

TABLE 2.

Alterations in the gut microbiota of depression.

Refs. Findings
Enriched in cases Reduced in cases Enriched in controls
74 Phyla: Bacteroidetes, Proteobacteria, and Actinobacteria; Genera: Enterobacteriaceae and Alistipes Phyla: Firmicutes; Genera: Faecalibacterium NA
73 NA Genera: Coprococcus and Dialister NA
75 Phylum: Bacteroidetes, proteobacteria, and Fusobacteria NA Phylum: Firmicutes and Actinobacteria
76 Eggerthella, Atopobium, and Bifidobacterium Faecalibacterium NA
77 NA Bifidobacterium, Lactobacillus NA
78 Janthinobacterium (blood) Neisseria (blood) NA

Note: Longitudinal heading Ref. [79].

One of the main traits of depression is abnormal immunologic activity. The immune system regulates neurologic growth and development and maintains normal physiologic functions (e.g., the ability of mast cells to synthesize nerve growth factors and the impaired memory and learning abilities of mice lacking T cells). 80 However, autoimmune diseases exhibit a higher risk of depression; for example, a previous study reported of a 43% increase of depression in Graves' disease. 81 A meta‐analysis of longitudinal studies also found that elevated inflammatory markers (including C‐reactive protein [CRP] and IL‐6) were significantly associated with an increased risk of subsequent depressive symptoms, thus lending credence to the inflammation‐to‐depression causal link. 82

The gut microbiota can regulate inflammation to influence depression. The study by Grigoleit demonstrated that LPS increases depressed mood due to excessive release of pro‐inflammatory cytokines. 83 Moreover, Beurel et al. found that SFB induces depression‐like behavior. 72 Specifically, SFB is able to secrete autoinducer‐2 (AI‐2, which is a quorum‐sensing molecule) to promote T helper 17 (Th17) cell proliferation and aggregation in the hippocampus. Additionally, a previous study confirmed the antidepressant effect of Bifidobacteria infantis. 84 B. infantis reduced the release of pro‐inflammatory cytokines such as IL‐6, IFN‐γ, and TNF‐α and decreased the IFN‐γ:IL‐10 ratio in Sprague–Dawley rats receiving immune stimulation. 84 Additionally, the B. infantis group had significantly higher concentrations of tryptophan and kynurenic acid (Kyna) in the plasma than in the control group. 84 Wong et al. also showed that it is feasible for the gut microbiota to influence depression by modulating inflammasome signaling. 85 Recently, Luo et al. found that the depletion of gut bacteria after irradiation reduces the synthesis of brain pro‐inflammatory factors. 86 In addition, TLRs are distributed in neurons and glial cells, and their mediated signaling pathways are involved in the development of the nervous system and the process of disease development (reviewed by Okun). 87 For example, the expression of TLR4 mRNA and the synthesis level of downstream proteins are significantly increased in patients with depression. 88

Several studies have observed differences in the gut microbiota metabolites in depressed patients compared to healthy individuals. For example, the metabolomics of depressed mice was significantly disordered in the blood and feces, with enhanced carbohydrate metabolism and decreased glutamate‐ and phenylalanine‐related metabolites being observed. 16 Valles‐Colomer found a positive correlation between 3,4‐dihydroxyphenylacetic acid levels and mental health, as well as increased levels of both GABA and glutamate, in patients with depression. 73 In fact, the gut microbiota can synthesize neuroactive metabolites associated with depression and influence its development. The serotonin concentration in the serum of conventional mice was 2.8 times (p = 1.27 × 10−10) higher than that of GF mice. 89 Higuchi reported that Lactobacillus sp. Strain E1 was able to decarboxylate glutamate to form gamma‐aminobutyric acid (GABA). 90 In addition, the gut microbiota can alter the permeability of the blood–brain barrier (BBB). 91

In addition to the blood pathway, gut bacterial metabolites can act indirectly on the central nervous system via the autonomic nervous system. Butyrate, which is a metabolite of fermentable carbohydrates, is able to activate vagal neurons and their projecting nucleus medial nucleus tractus solitaries. 92 In fact, the vagus nerve is the primary pathway by which gut bacteria communicate information to the brain. The Lactobacillus strain regulates the expression of GABA (Aα2) mRNA in different regions of the brain, thereby improving depression‐related behaviors. 93 However, this neurochemical and behavioral effect disappeared after dissecting the vagus nerve. 93 Moreover, Wang et al. found that subdiaphragmatic vagotomy was able to block the depressive phenotype induced by Lactobacillus intestinalis and Lactobacillus reuteri. 94

Stressors have been shown to induce depressive‐like behaviors through mechanisms including the aberrant activation of inflammation and HPA hyperactivity. 95 In fact, gut bacterial dysbiosis is an integral part of the abovementioned process, which is related to the “leaky gut” hypothesis. 96 , 97 Restraint stress leads to hyperactivity of the HPA axis, which is manifested by elevated plasma corticosterone concentrations. 98 The hyperactive HPA causes an increase in intestinal permeability and the consequent translocation of gut bacteria. 99 Blood‐entered LPS activates microglial TLR 4 in the brain, thus inducing depressive‐like behaviors by eliciting excessive inflammatory responses. 96 When cognitive–behavioral therapy was administered, patients with major depressive disorder had a significant decrease in TLR4 mRNA and protein, which was accompanied by a significant improvement in clinical symptoms. 100 There is a bidirectional regulation between gut bacteria and the HPA axis. Following the administration of restraint stress, GF mice exhibited a hyperactive HPA axis characterized by higher plasma ACTH and corticosterone levels compared to SPF mice. 101 Meanwhile, the colonization of Bifidobacterium infantis into GF mice reversed this result. 101 Second, stressor exposure disrupts the composition of gut bacteria, such as increasing colonization by the pathogens Citrobacter rodentium and Enterobacteriaceae, as well as decreasing the genus Bacteroides, thus resulting in the upregulation of inflammatory factors. 102 , 103 The mechanism of this effect is associated with a decrease in alpha‐defensin secretion by PAN cells and an increase in apoptosis of CD45+CD90+ cells, which produce protective IL‐22. 104 , 105 Bailey et al. discovered that antibiotic‐treated mice exposed to a social stressor did not exhibit an increase in IL‐6 or MCP‐1. 106 This implies that the stressor affects the immune system by altering the intestinal flora composition. 106

In summary, the three pathways described above interact with each other. 27 Stimulation of the vagus nerve increased the expression of corticotropin‐releasing factor mRNA in the hypothalamus and plasma levels of corticosterone and corticosterone by upregulating IL‐1 beta expression in the brain. 107

4. GUT BACTERIAL DYSBIOSIS: A PATHOPHYSIOLOGIC LINK BETWEEN DEPRESSION AND RE

In the state of comorbidity, RE and depression may be understood as being local and distant representations of gut bacterial dysbiosis (see Figure 3). Gut bacterial dysbiosis can cause changes in intestinal permeability, thus allowing the gut microbiota to enter the bloodstream. The nuclear transcription factors NF‐kB and the NLRP3 inflammasome are activated as a result of the action of PAMPs from translocated gut microbiota, such as LPS and flagellin, on pattern recognition receptors (PRRs) on intrinsic immune cells. The fact that inflammatory factors such as IL‐1, IL‐6, and TNF‐ɑ are increased in both depression and RE is easily understood. 108 However, there is little direct evidence on whether RE affects depression through inflammation; however, we reasoned this mechanism by examining existing studies of depression in IBD. Bercik et al. have shown that chronic enteritis can induce anxiety‐like behavior, which is a process mediated by pro‐inflammatory cytokines. 109 One notable aspect of this study is that supplementation with Bifidobacterium longum was able to reverse the abnormal behavior. Yang et al. found that intraperitoneal injection of Fast Green FCF inhibited the LPS/TLR4/Myd88/NF‐κB signaling pathway, which attenuated depressive‐like behavior. 110 Recently, Hu et al. found that oral administration of quercetin to a radiation‐induced brain injury (RIBI) mouse model resulted in improved anxiety, behavioral activity, and memory, as well as decreased TNF‐α and IL‐6 levels, along with reduced intestinal epithelial damage. 111 These effects disappeared when antibiotics were administered to RIBI mice, thus suggesting that quercetin acts by regulating gut bacteria. 111

FIGURE 3.

FIGURE 3

The pathophysiologic link between depression and radiation enteropathy (RE) through gut bacterial dysbiosis. In the state of comorbidity, RE and depression may be understood as being local and distant representations of gut bacterial dysbiosis. The mechanisms underlying this comorbid relationship are related to abnormalities in the gut–brain axis, including inflammation, HPA axis hyperactivity and autonomic disorders.

AlSharari et al. found that in DSS‐induced colitis, α7 knockout mice had more severe colitis and higher levels of TNF‐α than wild‐type mice. Choline supplementation (α7nAChR receptor agonist) alleviated DSS‐induced colitis. 112 Recently, Li et al. elucidated the mechanisms involved from the perspective of gut bacteria, thus confirming that choline mediates intestinal immune tolerance through the activation of α7nAChR. 113 Specifically, Enterobacter ludwigii is able to produce choline, which promotes DC synthesis and the release of retinoic acid (RA) and transforming growth factor beta (TGF‐β). Moreover, RA and TGF‐β are able to induce the differentiation of naïve CD4+ T cells into Treg cells rather than pro‐inflammatory Th1 or Th17 cells. 113 Choline levels are significantly decreased in intestinal tissue and serum samples from IBD patients. 114 , 115 Indeed, cholinergic anti‐inflammatory pathways play an important role in depression‐induced intestinal inflammation. 116 Ghia et al. demonstrated impaired parasympathetic function and colitis in depressed mice. 22 Antidepressants were able to prevent or treat depression‐associated intestinal inflammation, but the protective effect was completely lost when the vagus nerve was dissociated. 22 Another study by these researchers showed that depression can cause chronic to recurrent depression. 21 Macrophages in blood samples from depressed mice showed abnormal pro‐inflammatory activity, including increased secretion of pro‐inflammatory cytokines. 21 The mechanism for this effect is that the vagus nerve has an abnormal anti‐inflammatory pathway mediated through α7nAChR. 21 After a systematic search, no studies have been reported on choline changes in RE. 25 However, the abovementioned studies may be able to indicate the manner of researching the mechanism of the link between RE and depression.

5. MODIFICATION OF GUT MICROBIOTA AND RE MANAGEMENT

In the early 1900s, Russian embryologist Elie Metchnikoff proposed the therapeutic concept that changing the gut microbiota would prolong life. 117 Mitra et al. found that the severity of radiation‐induced intestinal toxicity was negatively correlated with gut microbiota diversity. 118 Therefore, researchers have attempted to modulate the composition of the patient's gut microbiota to prevent or alleviate clinical symptoms of RE, 119 such as RID. 120

5.1. Fecal microbiota transplantation (FMT)

FMT is the transfer of fecal bacteria from a healthy donor to a patient for the treatment of intestinal and extraintestinal diseases via injections of liquid flora or oral administration of flora capsules. 121 In the late fourth century AD, the use of fecal transplants for the treatment of diseases was documented in the “Zhou Hou Bei Ji Fang” written by the Chinese scholar Ge Hong. 122 In 1988, a clinical trial demonstrated that live Lactobacillus acidophilus cultures could be used to prevent RID. 123 Animal studies have reported of improved intestinal function and epithelial integrity, as well as significant increases in survival and body weight, in mice that received FMT after total body irradiation (TBI). 124 qPCR assays further confirmed that FMT upregulated small intestinal Vegf mRNA expression levels and promoted angiogenesis without accelerating tumor growth. 124 Chen et al. found that FMT attenuated radiotherapy‐related toxicity by maintaining PGF2α levels. 125 On November 30, 2022, the FDA (U.S. Food and Drug Administration) approved the first fecal bacteria product (Rebyota) for the prevention of Clostridium difficile infection (CDI) recurrence in people 18 years and older, which was a significant discovery (News from FDA). More indications are likely to be approved in the future.

5.2. Probiotics and prebiotics

The effect of probiotics on RE has long been proven. Chitapanarux et al. conducted a prospective, randomized controlled clinical trial (n = 63) that investigated the role of L. acidophilus and Bifidobacterium bifidum in reducing the incidence of acute RID and found a reduction in the incidence and severity of diarrhea, an increase in stool consistency, a significant reduction in antidiarrheal medication use, and no adverse effects in the probiotic group. 126 Another large double‐blind, placebo‐controlled clinical trial showed that the highly effective probiotic agent VSL#3 (with probiotic lactobacilli as the main ingredient) could reduce the incidence and severity of diarrhea and the use of the antidiarrheal drug loperamide. 127 A summary of clinical studies on the treatment of RE from a probiotic perspective is shown in Tables 3 and 4.

TABLE 3.

Summary of published clinical studies on probiotics for radiation enteropathy.

Author, Year Probiotics/Prebiotics Type of cancer Radiation dose Major findings
Giralt 2008 128 Lactobacillus Cervical carcinoma, endometrial adenocarcinoma 45–50 Gy Significantly improved stool consistency, but did not reduce the incidence of diarrhea (Grade 2–5)
Chitapanarux 2010 126 Lactobacillus acidophilus + Bifidobacterium bifidum Cervical cancer 200 cGy ↓ The incidence of diarrhea and the need for anti‐diarrheal medication
Demers 2014 129 Bifilact Pelvic cancer 40–50.4 Gy ↓ The incidence of diarrhea (Grade 2–4)
Garcia‐Peris 2016 130 Inulin, fructo‐oligosaccharide Gynecologic cancer NA ↑ The consistency of stools
Sasidharan 2019 131 Resistant starch Cervical cancer 50 Gy No significant gain
Delia 2007 127 VSL#3 Sigmoid, rectal, or cervical cancer 60–70 Gy ↓ Incidence and severity of RID, number of bowel movements per day
Linn 2019 132 Lactobacillus Cervical cancer 50 Gy ↓ Incidence and severity of RID
Urbancsek 2001 120 Lactobacillus rhamnosus Abdominopelvic tumor 50 Gy ↓ Number of bowel movements

TABLE 4.

Summary of ongoing clinical studies on probiotics for radiation enteropathy (date from https://clinicaltrials.gov).

NCT number Study designs Phases Start date Interventions Outcome measures Enrollment
NCT03978949 Randomized, double, parallel assignment III 2019 Bacillus licheniformis Grade 2 or more acute intestinal toxicities 248
NCT05406882 Randomized, open label, crossover assignment II/III 2022 Bifidobacterium and Lactobacillus, Glutamine Incidence of radiation proctitis 176
NCT03742596 Non‐randomized, quadruple, parallel assignment II/III 2018 Lactobacillus, Bifidobacteria The level of immunoglobulin (Ig) A 40
NCT03516461 Non‐randomized, open label, parallel assignment NA 2018 FMT Change of toxicity grade 30
NCT05032027 Randomized, quadruple, intervention model: Parallel assignment NA 2021 Probiotics Grade 3 enteritis 40

As gene editing technology becomes more accurate and affordable (especially with the development of CRISPR technology), engineered probiotics are becoming more feasible and widespread. 133 Compared with conventional probiotics, engineered probiotics have better specificity and safety. 133 The anti‐inflammatory cytokine IL‐22 fosters repair and healing of the intestinal mucosa. 134 IL‐22‐producing recombinant next‐generation probiotics (NGPs), which are also known as live biotherapeutic products (LBPs), 135 have been shown to increase 30‐day survival in C57BL/6 mice undergoing TBI by up to 85%. 136

Probiotics are defined as substrates that can be selectively utilized by host microorganisms and provide a benefit to the host. 137 Some studies have confirmed that prebiotics have the ability to promote the proliferation and replication of probiotics and consequently improve quality of life in abdominopelvic tumor patients. 131 , 138 A randomized, double‐blind, placebo‐controlled clinical trial in gynecologic oncology patients treated with postoperative abdominal radiotherapy included 38 patients, with the experimental group given inulin and fructo‐oligosaccharide and the control group receiving maltodextrin. The results of this study showed that prebiotics administered in the experimental group restored the number of corresponding probiotics more quickly, reduced the number of days of watery stools (p = 0.07), and improved the quality of life of patients undergoing abdominal radiotherapy. 130

5.3. Antibiotics

The overproliferation and translocation of gut bacteria is one of the characteristics of RE. Therefore, antibiotics are one of the options for the treatment of RE‐related infections. 139 The Chinese consensus on the diagnosis and treatment of radiation proctitis (2018) states that antibiotics can be given to patients with RE who have uncomfortable symptoms, such as abdominal bloating and diarrhea due to bacterial overproliferation; moreover, the course of treatment is usually 7–10 days (Grade of recommendation: 1B). 140 The efficacy of different antibiotics in the treatment of bacteremia secondary to RE was systematically described earlier by Brook et al. 140 The palliative effect of metronidazole in chronic RE was confirmed in a study by Cavcić et al. 141 Recently, Cui et al. found that the survival rate of the antibiotic‐treated group was significantly higher than that of the blank control group in mice after 6.5 Gy irradiation. 142

When using antibiotics, their safety should not be ignored, especially their effects on antitumor efficacy. Some antibiotics can directly exert anticancer effects through the following mechanisms: antiproliferative, proapoptotic, and anti‐epithelial mesenchymal transition (EMT) mechanisms (reviewed by Gao). 143 For example, vancomycin (an antibiotic that acts primarily on Gram‐positive bacteria and that has antimicrobial action limited to the gut) enhances RT‐induced antitumor immune responses and tumor growth inhibition. However, Gopalakrishnan et al. found that the administration of antibiotics before and after immunotherapy shortened progression‐free survival and overall survival in patients with stage IV tumors. In addition, vancomycin has been reported to impair intestinal IL‐17A and GM‐CSF‐mediated antifungal immune responses, thereby increasing the risk of fungal infections. Brook et al. found that an increase in lactose‐fermenting bacteria was associated with septicemia mortality, which was associated with the use of broad‐spectrum anti‐anaerobic agents. 144 This study demonstrates the need for precision antimicrobial therapy.

Finally, antibiotics are effective and relatively specific in killing intestinal flora. Therefore, antibiotics serve as a widely used experimental intervention, both in basic and clinical trials related to RE.

5.4. Other

In recent years, a number of new radiation protection agents have been explored in basic research. According to a rodent study, the green tea polyphenol (−)‐epigallocatechin‐3‐gallate (EGCG) was found to protect against RE by increasing the ratio of Firmicutes/Bacteroidetes and the abundance of probiotics, thereby reverting gut dysbiosis. 145 Zhang et al. found that urolithin A (UroA) improved intestinal cell regeneration, which was primarily accomplished through the restoration of the intestinal bacterial profile. 146 Li et al. found that butyrate attenuated RE via the activation of G protein‐coupled receptors (GPCRs) and the restoration of the intestinal bacterial composition. 147 Another study by Li et al. found that 30% calorie‐restricted dietary preconditioning ameliorated radiation enterotoxicity, which was associated with a reduced abundance of pro‐inflammatory bacteria and increased numbers of short‐chain fatty acid‐producing bacteria. 148 Recently, Wang et al. found that orally administered carbon nanoparticles can remove ROS, prevent gut bacterial dysbiosis, and reduce the apoptosis of intestinal epithelial cells to play a role in radiation protection. 149

6. CONCLUSION

Gut commensal bacteria determine intestinal radiosensitivity. Ionizing irradiation induces gut bacterial dysbiosis, which increases intestinal radiosensitivity and induces depression. Patients with RE combined with depression tend to have a poorer prognosis. In the state of comorbidity, RE and depression may be understood as being local and abscopal manifestations of gut bacterial disorders. The microbiota–gut–brain axis is a potential mechanism. Gut bacteria is a potential target for the treatment of RE. Current or pending problems in the field include patients with RE with comorbid depression who are not properly identified and valued. Both preclinical and clinical evidence lacks consistent, high‐quality conclusions about the therapeutic effects of gut microbiota and metabolites. Complete control of confounding factors is not easy because the gut microbiota is susceptible to a variety of factors. In addition, most studies on gut microbiota and disease have been cross‐sectional studies, yet this only yields correlations and lacks longitudinal studies. Moreover, most of the studies have been conducted in animals, and there is still much work to be done to translate the results of animal tests to complex humans. There are several future research directions that can be enacted. First, the characterization of the gut microbiota before radiotherapy can be used to assess and predict gastrointestinal syndromes after radiotherapy, which can correspondingly be stratified by risk factors and screened for high‐risk groups, thus helping clinicians in developing individualized treatment plans. In the intestine, in addition to bacteria, fungi, viruses, archaea, and other microorganisms interact with each other and constitute an intricate grid. Therefore, the study of RE cannot be limited to the bacterial level. At present, the study of intestinal flora is still in the initial stage, and with the introduction of more new and efficient research methods (such as Mendelian randomization analysis and artificial intelligence algorithms), it will greatly facilitate the development of this field.

AUTHOR CONTRIBUTIONS

Xinliang Liu: Writing – original draft (lead). Ying Li: Formal analysis (supporting). Meichen Gu: Formal analysis (supporting). Tiankai Xu: Supervision. Chuanlei Wang: Methodology (equal). Pengyu Chang: Conceptualization (equal); funding acquisition (equal); writing – review and editing (equal).

FUNDING INFORMATION

This article was supported by National Natural Science Foundation of China under grant number: 82272738, 81874254; Scientific and Technological Developing Scheme Foundation of Jilin Province under grant number: 20200201400JC; and Special Foundation to health professionals of Jilin Province under grant number: JLSWSRCZX2020‐00931. The Figure 3 in this review are made by Figdraw (www.figdraw.cosssm).

CONFLICT OF INTEREST STATEMENT

The authors declare that there is no conflict of interest. This article has been read and approved for publication in the journal by all authors.

Liu X, Li Y, Gu M, Xu T, Wang C, Chang P. Radiation enteropathy‐related depression: A neglectable course of disease by gut bacterial dysbiosis. Cancer Med. 2024;13:e6865. doi: 10.1002/cam4.6865

Contributor Information

Chuanlei Wang, Email: wang5cl@jlu.edu.cn.

Pengyu Chang, Email: changpengyu@jlu.edu.cn.

DATA AVAILABILITY STATEMENT

Not applicable.

REFERENCES

  • 1. Delaney G, Jacob S, Featherstone C, Barton M. The role of radiotherapy in cancer treatment: estimating optimal utilization from a review of evidence‐based clinical guidelines. Cancer. 2005;104(6):1129‐1137. [DOI] [PubMed] [Google Scholar]
  • 2. Somosy Z, Horváth G, Telbisz Á, Réz G, Pálfia Z. Morphological aspects of ionizing radiation response of small intestine. Micron. 2002;33(2):167‐178. [DOI] [PubMed] [Google Scholar]
  • 3. Li Y, Dong J, Xiao H, et al. Gut commensal derived‐valeric acid protects against radiation injuries. Gut Microbes. 2020;11(4):789‐806. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4. Kim YJ, Yu J, Park SP, Lee SH, Kim YS. Prevention of radiotherapy induced enteropathy by probiotics (PREP): protocol for a double‐blind randomized placebo‐controlled trial. BMC Cancer. 2021;21(1):1032. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Kumagai T, Rahman F, Smith AM. The microbiome and radiation induced‐bowel injury: evidence for potential mechanistic role in disease pathogenesis. Nutrients. 2018;10(10):1405. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Milliat F, François A, Tamarat R, Benderitter M. Role of endothelium in radiation‐induced normal tissue damages. Ann Cardiol Angeiol (Paris). 2008;57(3):139‐148. [DOI] [PubMed] [Google Scholar]
  • 7. Wedlake L, Thomas K, McGough C, Andreyev HJN. Small bowel bacterial overgrowth and lactose intolerance during radical pelvic radiotherapy: an observational study. Eur J Cancer. 2008;44(15):2212‐2217. [DOI] [PubMed] [Google Scholar]
  • 8. Zhang S, Wang Q, Zhou C, et al. Colorectal cancer, radiotherapy and gut microbiota. Chin J Cancer Res. 2019;31(1):212‐222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Matsuzawa T. Survival time in germfree mice after lethal whole body X‐irradiation. Tohoku J Exp Med. 1965;85:257‐263. [DOI] [PubMed] [Google Scholar]
  • 10. McLaughlin MM, Dacquisto MP, Jacobus DP, Horowitz RE. Effects of the germfree state on responses of mice to whole‐body irradiation. Radiat Res. 1964;23:333‐349. [PubMed] [Google Scholar]
  • 11. van Vliet MJ, Harmsen HJM, de Bont ESJM, Tissing WJE. The role of intestinal microbiota in the development and severity of chemotherapy‐induced mucositis. PLoS Pathog. 2010;6(5):e1000879. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Gerassy‐Vainberg S, Blatt A, Danin‐Poleg Y, et al. Radiation induces proinflammatory dysbiosis: transmission of inflammatory susceptibility by host cytokine induction. Gut. 2018;67(1):97‐107. [DOI] [PubMed] [Google Scholar]
  • 13. Bai J, Barandouzi ZA, Rowcliffe C, Meador R, Tsementzi D, Bruner DW. Gut microbiome and its associations with acute and chronic gastrointestinal toxicities in cancer patients with pelvic radiation therapy: a systematic review. Front Oncol. 2021;11:5237. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Kanarek N, Grivennikov SI, Leshets M, et al. Critical role for IL‐1β in DNA damage‐induced mucositis. Proc Natl Acad Sci U S A. 2014;111(6):E702‐E711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Al‐Sadi R, Ye D, Dokladny K, Ma TY. Mechanism of IL‐1beta‐induced increase in intestinal epithelial tight junction permeability. J Immunol. 2008;180(8):5653‐5661. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16. Zheng P, Zeng B, Zhou C, et al. Gut microbiome remodeling induces depressive‐like behaviors through a pathway mediated by the host's metabolism. Mol Psychiatry. 2016;21(6):786‐796. [DOI] [PubMed] [Google Scholar]
  • 17. Ballou S, Katon J, Singh P, et al. Chronic diarrhea and constipation are more common in depressed individuals. Clin Gastroenterol Hepatol. 2019;17(13):2696‐2703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Drossman DA, Hasler WL. Rome IV‐functional GI disorders: disorders of gut‐brain interaction. Gastroenterology. 2016;150(6):1257‐1261. [DOI] [PubMed] [Google Scholar]
  • 19. Gilbert A, Drinkwater K, McParland L, et al. UK national cohort of anal cancer treated with intensity‐modulated radiotherapy: one‐year oncological and patient‐reported outcomes. Eur J Cancer. 2020;128:7‐16. [DOI] [PubMed] [Google Scholar]
  • 20. Moulton CD, Pavlidis P, Norton C, et al. Depressive symptoms in inflammatory bowel disease: an extraintestinal manifestation of inflammation? Clin Exp Immunol. 2019;197(3):308‐318. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Ghia JE, Blennerhassett P, Deng Y, Verdu EF, Khan WI, Collins SM. Reactivation of inflammatory bowel disease in a mouse model of depression. Gastroenterology. 2009;136(7):2280‐2288.e1‐4. [DOI] [PubMed] [Google Scholar]
  • 22. Ghia JE, Blennerhassett P, Collins SM. Impaired parasympathetic function increases susceptibility to inflammatory bowel disease in a mouse model of depression. J Clin Invest. 2008;118(6):2209‐2218. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Van Der Zanden EP, Boeckxstaens GE, de Jonge WJ. The vagus nerve as a modulator of intestinal inflammation. Neurogastroenterol Motil. 2009;21(1):6‐17. [DOI] [PubMed] [Google Scholar]
  • 24. Wang H, Yu M, Ochani M, et al. Nicotinic acetylcholine receptor alpha7 subunit is an essential regulator of inflammation. Nature. 2003;421(6921):384‐388. [DOI] [PubMed] [Google Scholar]
  • 25. Strazhevskaia NB, Struchkov VA. Effect of choline on the DNA supramolecular complex in rats and their survival following gamma irradiation. Radiobiologiia. 1988;28(2):199‐200. [PubMed] [Google Scholar]
  • 26. Guo F, Wang Y, Wang J, et al. Choline protects the heart from doxorubicin‐induced cardiotoxicity through vagal activation and Nrf2/HO‐1 pathway. Oxid Med Cell Longev. 2022;2022:4740931. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Cryan JF, O'Riordan KJ, Cowan CSM, et al. The microbiota‐gut‐brain axis. Physiol Rev. 2019;99(4):1877‐2013. [DOI] [PubMed] [Google Scholar]
  • 28. Hines DJ, Choi HB, Hines RM, Phillips AG, MacVicar BA. Prevention of LPS‐induced microglia activation, cytokine production and sickness behavior with TLR4 receptor interfering peptides. PLoS ONE. 2013;8(3):e60388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29. Littman DR, Pamer EG. Role of the commensal microbiota in normal and pathogenic host immune responses. Cell Host Microbe. 2011;10(4):311‐323. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Chung H, Pamp SJ, Hill JA, et al. Gut immune maturation depends on colonization with a host‐specific microbiota. Cell. 2012;149(7):1578‐1593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Al‐Qadami G, Van Sebille Y, Le H, Bowen J. Gut microbiota: implications for radiotherapy response and radiotherapy‐induced mucositis. Expert Rev Gastroenterol Hepatol. 2019;13(5):485‐496. [DOI] [PubMed] [Google Scholar]
  • 32. Strauch UG, Obermeier F, Grunwald N, et al. Influence of intestinal bacteria on induction of regulatory T cells: lessons from a transfer model of colitis. Gut. 2005;54(11):1546‐1552. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Macpherson AJ, Harris NL. Interactions between commensal intestinal bacteria and the immune system. Nat Rev Immunol. 2004;4(6):478‐485. [DOI] [PubMed] [Google Scholar]
  • 34. Anderson RE, Scaletti JV, Howarth JL. Radiation‐induced life shortening in germfree mice. Exp Gerontol. 1972;7(5):289‐301. [DOI] [PubMed] [Google Scholar]
  • 35. Crawford PA, Gordon JI. Microbial regulation of intestinal radiosensitivity. Proc Natl Acad Sci U S A. 2005;102(37):13254‐13259. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Santulli G. Angiopoietin‐like proteins: a comprehensive look. Front Endocrinol (Lausanne). 2014;5:4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Zhao Z, Cheng W, Qu W, Shao G, Liu S. Antibiotic alleviates radiation‐induced intestinal injury by remodeling microbiota, reducing inflammation, and inhibiting fibrosis. ACS Omega. 2020;5(6):2967‐2977. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38. Järbrink‐Sehgal E, Andreasson A. The gut microbiota and mental health in adults. Curr Opin Neurobiol. 2020;62:102‐114. [DOI] [PubMed] [Google Scholar]
  • 39. Pennisi E. Meet the psychobiome. Science. 2020;368(6491):570‐573. [DOI] [PubMed] [Google Scholar]
  • 40. Zhang X, Zhang D, Jia H, et al. The oral and gut microbiomes are perturbed in rheumatoid arthritis and partly normalized after treatment. Nat Med. 2015;21(8):895‐905. [DOI] [PubMed] [Google Scholar]
  • 41. Wang A, Ling Z, Yang Z, et al. Gut microbial dysbiosis may predict diarrhea and fatigue in patients undergoing pelvic cancer radiotherapy: a pilot study. PLoS ONE. 2015;10(5):e0126312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42. Kent TH, Osborne JW, Wende CM. Intestinal flora in whole‐body and intestinal x‐irradiated rats. Radiat Res. 1968;35(3):635‐651. [PubMed] [Google Scholar]
  • 43. Vincent JG, Veomett RC, Riley RF. Relation of the indigenous flora of the small intestine of the rat to post‐irradiation bacteremia. J Bacteriol. 1955;69(1):38‐44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44. Wang L, Wang X, Zhang G, et al. The impact of pelvic radiotherapy on the gut microbiome and its role in radiation‐induced diarrhoea: a systematic review. Radiat Oncol. 2021;16(1):187. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Nam YD, Kim HJ, Seo JG, Kang SW, Bae JW. Impact of pelvic radiotherapy on gut microbiota of gynecological cancer patients revealed by massive pyrosequencing. PLoS ONE. 2013;8(12):e82659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. DeGruttola AK, Low D, Mizoguchi A, Mizoguchi E. Current understanding of dysbiosis in disease in human and animal models. Inflamm Bowel Dis. 2016;22(5):1137‐1150. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Hai‐yan H, Li M, Xiao X. Correlation between gut microecology and development of radiation enteritis. J Chongqing Med Univ. 2020;45(9):1319‐1322. [Google Scholar]
  • 48. Otte JM, Kiehne K, Herzig KH. Antimicrobial peptides in innate immunity of the human intestine. J Gastroenterol. 2003;38(8):717‐726. [DOI] [PubMed] [Google Scholar]
  • 49. Witkin EM. Inherited differences in sensitivity to radiation in Escherichia coli . Proc Natl Acad Sci U S A. 1946;32(3):59‐68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50. Yamanouchi K, Tsujiguchi T, Sakamoto Y, Ito K. Short‐term follow‐up of intestinal flora in radiation‐exposed mice. J Radiat Res. 2019;60(3):328‐332. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51. Cosar R, Ozen A, Tastekin E, et al. Does gender difference effect radiation‐induced lung toxicity? An experimental study by genetic and histopathological predictors. Radiat Res. 2022;197(3):280‐288. [DOI] [PubMed] [Google Scholar]
  • 52. Chan S, Rowbottom L, McDonald R, et al. Does the time of radiotherapy affect treatment outcomes? A review of the literature. Clin Oncol (R Coll Radiol). 2017;29(4):231‐238. [DOI] [PubMed] [Google Scholar]
  • 53. Zhang Z, Shao S, Zhang Y, et al. Xiaoyaosan slows cancer progression and ameliorates gut dysbiosis in mice with chronic restraint stress and colorectal cancer xenografts. Biomed Pharmacother. 2020;132:110916. [DOI] [PubMed] [Google Scholar]
  • 54. Poonacha KNT, Villa TG, Notario V. The interplay among radiation therapy, antibiotics and the microbiota: impact on cancer treatment outcomes. Antibiotics (Basel). 2022;11(3):331. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55. Sokol H, Pigneur B, Watterlot L, et al. Faecalibacterium prausnitzii is an anti‐inflammatory commensal bacterium identified by gut microbiota analysis of Crohn disease patients. Proc Natl Acad Sci U S A. 2008;105(43):16731‐16736. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56. Wang Z, Wang Q, Wang X, et al. Gut microbial dysbiosis is associated with development and progression of radiation enteritis during pelvic radiotherapy. J Cell Mol Med. 2019;23(5):3747‐3756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57. Shao L, Li M, Zhang B, Chang P. Bacterial dysbiosis incites Th17 cell revolt in irradiated gut. Biomed Pharmacother. 2020;131:110674. [DOI] [PubMed] [Google Scholar]
  • 58. Le Chatelier E, Nielsen T, Qin J, et al. Richness of human gut microbiome correlates with metabolic markers. Nature. 2013;500(7464):541‐546. [DOI] [PubMed] [Google Scholar]
  • 59. Ciorba MA, Riehl TE, Rao MS, et al. Lactobacillus probiotic protects intestinal epithelium from radiation injury in a TLR‐2/cyclo‐oxygenase‐2‐dependent manner. Gut. 2012;61(6):829‐838. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60. Riehl T, Cohn S, Tessner T, Schloemann S, Stenson WF. Lipopolysaccharide is radioprotective in the mouse intestine through a prostaglandin‐mediated mechanism. Gastroenterology. 2000;118(6):1106‐1116. [DOI] [PubMed] [Google Scholar]
  • 61. Burdelya LG, Krivokrysenko VI, Tallant TC, et al. An agonist of toll‐like receptor 5 has radioprotective activity in mouse and primate models. Science. 2008;320(5873):226‐230. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Saha S, Bhanja P, Liu L, et al. TLR9 agonist protects mice from radiation‐induced gastrointestinal syndrome. PLoS ONE. 2012;7(1):e29357. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Paone P, Cani PD. Mucus barrier, mucins and gut microbiota: the expected slimy partners? Gut. 2020;69(12):2232‐2243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Wlodarska M, Luo C, Kolde R, et al. Indoleacrylic acid produced by commensal peptostreptococcus species suppresses inflammation. Cell Host Microbe. 2017;22(1):25‐37.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65. Joossens M, Huys G, Cnockaert M, et al. Dysbiosis of the faecal microbiota in patients with Crohn's disease and their unaffected relatives. Gut. 2011;60(5):631‐637. [DOI] [PubMed] [Google Scholar]
  • 66. Li Y, Yan H, Zhang Y, et al. Alterations of the gut microbiome composition and lipid metabolic profile in radiation enteritis. Front Cell Infect Microbiol. 2020;10:541178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67. Trimble WS, Grinstein S. Barriers to the free diffusion of proteins and lipids in the plasma membrane. J Cell Biol. 2015;208(3):259‐271. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68. Tian R, Tan JT, Wang RL, Xie H, Qian YB, Yu KL. The role of intestinal mucosa oxidative stress in gut barrier dysfunction of severe acute pancreatitis. Eur Rev Med Pharmacol Sci. 2013;17(3):349‐355. [PubMed] [Google Scholar]
  • 69. Cheng Y, Ling Z, Li L. The intestinal microbiota and colorectal cancer. Front Immunol. 2020;11:615056. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70. Zhang Y, Zhang B, Dong L, Chang P. Potential of Omega‐3 polyunsaturated fatty acids in managing chemotherapy‐ or radiotherapy‐related intestinal microbial dysbiosis. Adv Nutr. 2019;10(1):133‐147. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Neufeld KM, Kang N, Bienenstock J, Foster JA. Reduced anxiety‐like behavior and central neurochemical change in germ‐free mice. Neurogastroenterol Motil. 2011;23(3):255‐264.e119. [DOI] [PubMed] [Google Scholar]
  • 72. Medina‐Rodriguez EM, Madorma D, O'Connor G, et al. Identification of a signaling mechanism by which the microbiome regulates Th17 cell‐mediated depressive‐like behaviors in mice. Am J Psychiatry. 2020;177(10):974‐990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73. Valles‐Colomer M, Falony G, Darzi Y, et al. The neuroactive potential of the human gut microbiota in quality of life and depression. Nat Microbiol. 2019;4(4):623‐632. [DOI] [PubMed] [Google Scholar]
  • 74. Jiang H, Ling Z, Zhang Y, et al. Altered fecal microbiota composition in patients with major depressive disorder. Brain Behav Immun. 2015;48:186‐194. [DOI] [PubMed] [Google Scholar]
  • 75. Chen YH, Xue F, Yu SF, et al. Gut microbiota dysbiosis in depressed women: the association of symptom severity and microbiota function. J Affect Disord. 2021;282:391‐400. [DOI] [PubMed] [Google Scholar]
  • 76. Knudsen JK, Bundgaard‐Nielsen C, Hjerrild S, Nielsen RE, Leutscher P, Sørensen S. Gut microbiota variations in patients diagnosed with major depressive disorder—a systematic review. Brain Behav. 2021;11(7):e02177. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77. Aizawa E, Tsuji H, Asahara T, et al. Possible association of Bifidobacterium and Lactobacillus in the gut microbiota of patients with major depressive disorder. J Affect Disord. 2016;202:254‐257. [DOI] [PubMed] [Google Scholar]
  • 78. Ciocan D, Cassard AM, Becquemont L, et al. Blood microbiota and metabolomic signature of major depression before and after antidepressant treatment: a prospective case‐control study. J Psychiatry Neurosci. 2021;46(3):E358‐E368. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Bhatt AP, Redinbo MR, Bultman SJ. The role of the microbiome in cancer development and therapy. CA Cancer J Clin. 2017;67(4):326‐344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80. Bienenstock J, Forsythe P, Karimi K, Kunze W. Neuroimmune aspects of food intake. Int Dairy J. 2010;20(4):253‐258. [Google Scholar]
  • 81. Leone M, Kuja‐Halkola R, Leval A, et al. Genetic and environmental contribution to the co‐occurrence of endocrine‐metabolic disorders and depression: a nationwide Swedish study of siblings. Am J Psychiatry. 2022;179(11):824‐832. [DOI] [PubMed] [Google Scholar]
  • 82. Valkanova V, Ebmeier KP, Allan CL. CRP, IL‐6 and depression: a systematic review and meta‐analysis of longitudinal studies. J Affect Disord. 2013;150(3):736‐744. [DOI] [PubMed] [Google Scholar]
  • 83. Grigoleit JS, Kullmann JS, Wolf OT, et al. Dose‐dependent effects of endotoxin on neurobehavioral functions in humans. PLoS ONE. 2011;6(12):e28330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Desbonnet L, Garrett L, Clarke G, Bienenstock J, Dinan TG. The probiotic Bifidobacteria infantis: an assessment of potential antidepressant properties in the rat. J Psychiatr Res. 2008;43(2):164‐174. [DOI] [PubMed] [Google Scholar]
  • 85. Wong ML, Inserra A, Lewis MD, et al. Inflammasome signaling affects anxiety‐ and depressive‐like behavior and gut microbiome composition. Mol Psychiatry. 2016;21(6):797‐805. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86. Luo N, Zhu W, Li X, et al. Impact of gut microbiota on radiation‐associated cognitive dysfunction and neuroinflammation in mice. Radiat Res. 2022;197(4):350‐364. [DOI] [PubMed] [Google Scholar]
  • 87. Okun E, Griffioen KJ, Mattson MP. Toll‐like receptor signaling in neural plasticity and disease. Trends Neurosci. 2011;34(5):269‐281. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Hung YY, Kang HY, Huang KW, Huang TL. Association between toll‐like receptors expression and major depressive disorder. Psychiatry Res. 2014;220(1–2):283‐286. [DOI] [PubMed] [Google Scholar]
  • 89. Wikoff WR, Anfora AT, Liu J, et al. Metabolomics analysis reveals large effects of gut microflora on mammalian blood metabolites. Proc Natl Acad Sci U S A. 2009;106(10):3698‐3703. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Higuchi T, Hayashi H, Abe K. Exchange of glutamate and gamma‐aminobutyrate in a lactobacillus strain. J Bacteriol. 1997;179(10):3362‐3364. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91. Braniste V, al‐Asmakh M, Kowal C, et al. The gut microbiota influences blood‐brain barrier permeability in mice. Sci Transl Med. 2014;6(263):263ra158. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92. Goswami C, Iwasaki Y, Yada T. Short‐chain fatty acids suppress food intake by activating vagal afferent neurons. J Nutr Biochem. 2018;57:130‐135. [DOI] [PubMed] [Google Scholar]
  • 93. Bravo JA, Forsythe P, Chew MV, et al. Ingestion of lactobacillus strain regulates emotional behavior and central GABA receptor expression in a mouse via the vagus nerve. Proc Natl Acad Sci U S A. 2011;108(38):16050‐16055. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94. Wang S, Ishima T, Zhang J, et al. Ingestion of lactobacillus intestinalis and lactobacillus reuteri causes depression‐ and anhedonia‐like phenotypes in antibiotic‐treated mice via the vagus nerve. J Neuroinflammation. 2020;17(1):241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Liu RT, Alloy LB. Stress generation in depression: a systematic review of the empirical literature and recommendations for future study. Clin Psychol Rev. 2010;30(5):582‐593. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96. Gárate I, García‐Bueno B, Madrigal JLM, et al. Origin and consequences of brain toll‐like receptor 4 pathway stimulation in an experimental model of depression. J Neuroinflammation. 2011;8:151. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Maes M, Kubera M, Leunis JC. The gut‐brain barrier in major depression: intestinal mucosal dysfunction with an increased translocation of LPS from gram negative enterobacteria (leaky gut) plays a role in the inflammatory pathophysiology of depression. Neuro Endocrinol Lett. 2008;29(1):117‐124. [PubMed] [Google Scholar]
  • 98. Hamieh AM, Mallaret G, Meleine M, et al. Toll‐like receptor 5 knock‐out mice exhibit a specific low level of anxiety. Brain Behav Immun. 2021;93:226‐237. [DOI] [PubMed] [Google Scholar]
  • 99. Bailey MT, Engler H, Sheridan JF. Stress induces the translocation of cutaneous and gastrointestinal microflora to secondary lymphoid organs of C57BL/6 mice. J Neuroimmunol. 2006;171(1–2):29‐37. [DOI] [PubMed] [Google Scholar]
  • 100. Kéri S, Szabó C, Kelemen O. Expression of toll‐like receptors in peripheral blood mononuclear cells and response to cognitive‐behavioral therapy in major depressive disorder. Brain Behav Immun. 2014;40:235‐243. [DOI] [PubMed] [Google Scholar]
  • 101. Sudo N, Chida Y, Aiba Y, et al. Postnatal microbial colonization programs the hypothalamic‐pituitary‐adrenal system for stress response in mice. J Physiol. 2004;558(Pt 1):263‐275. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102. Bailey MT, Dowd SE, Parry NMA, Galley JD, Schauer DB, Lyte M. Stressor exposure disrupts commensal microbial populations in the intestines and leads to increased colonization by Citrobacter rodentium . Infect Immun. 2010;78(4):1509‐1519. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103. Galley JD, Yu Z, Kumar P, Dowd SE, Lyte M, Bailey MT. The structures of the colonic mucosa‐associated and luminal microbial communities are distinct and differentially affected by a prolonged murine stressor. Gut Microbes. 2014;5(6):748‐760. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 104. Suzuki K, Nakamura K, Shimizu Y, et al. Decrease of α‐defensin impairs intestinal metabolite homeostasis via dysbiosis in mouse chronic social defeat stress model. Sci Rep. 2021;11(1):9915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Shaler CR, Parco AA, Elhenawy W, et al. Psychological stress impairs IL22‐driven protective gut mucosal immunity against colonising pathobionts. Nat Commun. 2021;12(1):6664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Bailey MT, Dowd SE, Galley JD, Hufnagle AR, Allen RG, Lyte M. Exposure to a social stressor alters the structure of the intestinal microbiota: implications for stressor‐induced immunomodulation. Brain Behav Immun. 2011;25(3):397‐407. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107. Hosoi T, Okuma Y, Nomura Y. Electrical stimulation of afferent vagus nerve induces IL‐1beta expression in the brain and activates HPA axis. Am J Physiol Regul Integr Comp Physiol. 2000;279(1):R141‐R147. [DOI] [PubMed] [Google Scholar]
  • 108. Lopresti AL, Maker GL, Hood SD, Drummond PD. A review of peripheral biomarkers in major depression: the potential of inflammatory and oxidative stress biomarkers. Prog Neuropsychopharmacol Biol Psychiatry. 2014;48:102‐111. [DOI] [PubMed] [Google Scholar]
  • 109. Bercik P, Verdu EF, Foster JA, et al. Chronic gastrointestinal inflammation induces anxiety‐like behavior and alters central nervous system biochemistry in mice. Gastroenterology. 2010;139(6):2102‐2112.e1. [DOI] [PubMed] [Google Scholar]
  • 110. Yang J, Liu R, Lu F, et al. Fast green FCF attenuates lipopolysaccharide‐induced depressive‐like behavior and downregulates TLR4/Myd88/NF‐κB signal pathway in the mouse hippocampus. Front Pharmacol. 2019;10:501. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Hu J, Jiao W, Tang Z, et al. Quercetin inclusion complex gels ameliorate radiation‐induced brain injury by regulating gut microbiota. Biomed Pharmacother. 2023;158:114142. [DOI] [PubMed] [Google Scholar]
  • 112. AlSharari SD, Bagdas D, Akbarali HI, et al. Sex differences and drug dose influence the role of the α7 nicotinic acetylcholine receptor in the mouse dextran sodium sulfate‐induced colitis model. Nicotine Tob Res. 2017;19(4):460‐468. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113. Li Q, Sun X, Yu K, et al. Enterobacter ludwigii protects DSS‐induced colitis through choline‐mediated immune tolerance. Cell Rep. 2022;40(9):111308. [DOI] [PubMed] [Google Scholar]
  • 114. Dawiskiba T, Deja S, Mulak A, et al. Serum and urine metabolomic fingerprinting in diagnostics of inflammatory bowel diseases. World J Gastroenterol. 2014;20(1):163‐174. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115. Balasubramanian K, Kumar S, Singh RR, et al. Metabolism of the colonic mucosa in patients with inflammatory bowel diseases: an in vitro proton magnetic resonance spectroscopy study. Magn Reson Imaging. 2009;27(1):79‐86. [DOI] [PubMed] [Google Scholar]
  • 116. Ghia JE, Blennerhassett P, Kumar–Ondiveeran H, Verdu EF, Collins SM. The vagus nerve: a tonic inhibitory influence associated with inflammatory bowel disease in a murine model. Gastroenterology. 2006;131(4):1122‐1130. [DOI] [PubMed] [Google Scholar]
  • 117. Podolsky SH. Metchnikoff and the microbiome. Lancet. 2012;380(9856):1810‐1811. [DOI] [PubMed] [Google Scholar]
  • 118. Mitra A, Grossman Biegert GW, Delgado AY, et al. Microbial diversity and composition is associated with patient‐reported toxicity during chemoradiation therapy for cervical cancer. Int J Radiat Oncol Biol Phys. 2020;107(1):163‐171. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119. Reis Ferreira M, Andreyev HJN, Mohammed K, et al. Microbiota‐ and radiotherapy‐induced gastrointestinal side‐effects (MARS) study: a large pilot study of the microbiome in acute and late‐radiation enteropathy. Clin Cancer Res. 2019;25(21):6487‐6500. [DOI] [PubMed] [Google Scholar]
  • 120. 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(4):391‐396. [DOI] [PubMed] [Google Scholar]
  • 121. Staley C, Khoruts A, Sadowsky MJ. Contemporary applications of fecal microbiota transplantation to treat intestinal diseases in humans. Arch Med Res. 2017;48(8):766‐773. [DOI] [PubMed] [Google Scholar]
  • 122. Stripling J, Rodriguez M. Current evidence in delivery and therapeutic uses of fecal microbiota transplantation in human diseases‐Clostridium difficile disease and beyond. Am J Med Sci. 2018;356(5):424‐432. [DOI] [PubMed] [Google Scholar]
  • 123. 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(4):435‐437. [DOI] [PubMed] [Google Scholar]
  • 124. Cui M, Xiao H, Li Y, et al. Faecal microbiota transplantation protects against radiation‐induced toxicity. EMBO mol Med. 2017;9(4):448‐461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 125. Chen ZY, Xiao HW, Dong JL, et al. Gut microbiota‐derived PGF2α fights against radiation‐induced lung toxicity through the MAPK/NF‐κB pathway. Antioxidants (Basel). 2021;11(1):65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Chitapanarux I, Chitapanarux T, Traisathit P, Kudumpee S, Tharavichitkul E, Lorvidhaya V. 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Delia P, Sansotta G, Donato V, et al. Use of probiotics for prevention of radiation‐induced diarrhea. World J Gastroenterol. 2007;13(6):912‐915. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Giralt J, Regadera JP, Verges R, et al. 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(4):1213‐1219. [DOI] [PubMed] [Google Scholar]
  • 129. 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(5):761‐767. [DOI] [PubMed] [Google Scholar]
  • 130. Garcia‐Peris P, Velasco C, Hernandez M, et al. Effect of inulin and fructo‐oligosaccharide on the prevention of acute radiation enteritis in patients with gynecological cancer and impact on quality‐of‐life: a randomized, double‐blind, placebo‐controlled trial. Eur J Clin Nutr. 2016;70(2):170‐174. [DOI] [PubMed] [Google Scholar]
  • 131. Sasidharan BK, Ramadass B, Viswanathan PN, et al. A phase 2 randomized controlled trial of oral resistant starch supplements in the prevention of acute radiation proctitis in patients treated for cervical cancer. J Cancer Res Ther. 2019;15(6):1383‐1391. [DOI] [PubMed] [Google Scholar]
  • 132. Linn YH, Thu KK, Win NHH. Effect of probiotics for the prevention of acute radiation‐induced diarrhoea among cervical cancer patients: a randomized double‐blind placebo‐controlled study. Probiotics Antimicrob Proteins. 2019;11(2):638‐647. [DOI] [PubMed] [Google Scholar]
  • 133. Ma J, Lyu Y, Liu X, et al. Engineered probiotics. Microb Cell Fact. 2022;21(1):72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134. Zha JM, Li HS, Lin Q, et al. Interleukin 22 expands transit‐amplifying cells while depleting Lgr5(+) stem cells via inhibition of Wnt and notch signaling. Cell Mol Gastroenterol Hepatol. 2019;7(2):255‐274. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. O'Toole PW, Marchesi JR, Hill C. Next‐generation probiotics: the spectrum from probiotics to live biotherapeutics. Nat Microbiol. 2017;2:17057. [DOI] [PubMed] [Google Scholar]
  • 136. Zhang X, Fisher R, Hou W, et al. Second‐generation probiotics producing IL‐22 increase survival of mice after total body irradiation. In Vivo. 2020;34(1):39‐50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137. Gibson GR, Hutkins R, Sanders ME, 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(8):491‐502. [DOI] [PubMed] [Google Scholar]
  • 138. Rosli D, Shahar S, Manaf ZA, et al. Randomized controlled trial on the effect of partially hydrolyzed guar gum supplementation on diarrhea frequency and gut microbiome count among pelvic radiation patients. J Parenter Enteral Nutr. 2021;45(2):277‐286. [DOI] [PubMed] [Google Scholar]
  • 139. Livstone EM, Hersh T, Spiro HM, Floch MH. The gastrointestinal microflora of irradiated mice. II. Effect of oral antibiotic administration on the colonic flora and survival of adult mice. Yale J Biol Med. 1970;42(6):448‐454. [PMC free article] [PubMed] [Google Scholar]
  • 140. Brook I, Elliott TB, Ledney GD, Shoemaker MO, Knudson GB. Management of postirradiation infection: lessons learned from animal models. Mil Med. 2004;169(3):194‐197. [DOI] [PubMed] [Google Scholar]
  • 141. Cavcić J, Turcić J, Martinac P, et al. Metronidazole in the treatment of chronic radiation proctitis: clinical trial. Croat Med J. 2000;41(3):314‐318. [PubMed] [Google Scholar]
  • 142. Xaioqi S, Hongyan W. Research progress on the interaction between gut microbiota and radiation enteritis. J Int Oncol. 2023;50(4):244‐247. [Google Scholar]
  • 143. Gao Y, Shang Q, Li W, et al. Antibiotics for cancer treatment: a double‐edged sword. J Cancer. 2020;11(17):5135‐5149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 144. Brook I, Ledney GD. Effect of antimicrobial therapy on the gastrointestinal bacterial flora, infection and mortality in mice exposed to different doses of irradiation. J Antimicrob Chemother. 1994;33(1):63‐72. [DOI] [PubMed] [Google Scholar]
  • 145. Cai S, Xie LW, Xu JY, et al. (−)‐Epigallocatechin‐3‐gallate (EGCG) modulates the composition of the gut microbiota to protect against radiation‐induced intestinal injury in mice. Front Oncol. 2022;12:848107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 146. Zhang Y, Dong Y, Lu P, et al. Gut metabolite urolithin a mitigates ionizing radiation‐induced intestinal damage. J Cell Mol Med. 2021;25(21):10306‐10312. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147. Li Y, Xiao H, Dong J, et al. Gut microbiota metabolite fights against dietary polysorbate 80‐aggravated radiation enteritis. Front Microbiol. 2020;11:1450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 148. Li Y, Dong J, Xiao H, et al. Caloric restriction alleviates radiation injuries in a sex‐dependent fashion. FASEB J. 2021;35(8):e21787. [DOI] [PubMed] [Google Scholar]
  • 149. Wang C, Xie J, Dong X, et al. Clinically approved carbon nanoparticles with oral administration for intestinal radioprotection via protecting the small intestinal crypt stem cells and maintaining the balance of intestinal flora. Small. 2020;16(16):e1906915. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Not applicable.


Articles from Cancer Medicine are provided here courtesy of Wiley

RESOURCES