Next Article in Journal
Legionella in the City: Unveiling Legionella pneumophila in Hillbrow’s High-Rise Water Systems
Previous Article in Journal
Phytochemicals from Euclea natalensis Modulate Th17 Differentiation, HIV Latency, and Comorbid Pathways: A Systems Pharmacology and Thermodynamic Profiling Approach
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Gut Microbiota and Its Metabolites in Mitigating Radiation Damage

1
Key Laboratory of Biological Resource and Ecological Environment of Chinese Education Ministry, College of Life Sciences, Sichuan University, Chengdu 610064, China
2
Core Laboratory, School of Medicine, Sichuan Provincial People’s Hospital Affiliated to University of Electronic Science and Technology of China, Chengdu 610072, China
3
Irradiation Preservation and Effect Key Laboratory of Sichuan Province, Sichuan Institute of Atomic Energy, Chengdu 610101, China
*
Authors to whom correspondence should be addressed.
Microorganisms 2025, 13(9), 2151; https://doi.org/10.3390/microorganisms13092151
Submission received: 10 August 2025 / Revised: 6 September 2025 / Accepted: 10 September 2025 / Published: 15 September 2025

Abstract

With the widespread use of ionizing radiation (IR) in medical and industrial settings, irradiation has become increasingly common, posing significant risks to human health. Among the various organs affected, the gut is particularly sensitive to radiation-induced damage, leading to conditions such as radiation-induced intestinal damage (RIID). Recent studies have emphasized the critical role of gut microbiota and its metabolites in mitigating radiation-induced injury. This review discusses the effects of IR on the mammalian and human gut microbiota. We examine the dynamics of gut microbiota composition during and after irradiation, and emphasize the protective role of the gut flora and the metabolites in the pathophysiological mechanisms exhibited during radiation injury. In addition, this article investigates how specific metabolites, such as short-chain fatty acids and indole derivatives, may contribute to the mitigation of inflammation and promotion of gut barrier integrity. In addition, various therapeutic strategies based on modulating the gut microbiota, such as probiotics, antibiotics, and fecal microbiota transplantation, are discussed to understand their potential to prevent or mitigate RIID. Understanding the interactions between IR, gut microbiota and their metabolites provides new avenues for developing innovative therapeutic approaches to improve patient outcomes during and after radiotherapy. Future research directions could focus on optimizing microbiota-based therapies and exploring the role of diet and lifestyle in enhancing intestinal health during irradiation.

1. Introduction

With the widespread use of ionizing radiation (IR) in research, industry, homeland security, and modern medicine, it has become virtually unavoidable for humans to be exposed to low doses of artificial IR on a daily basis [1]. IR is increasingly used in medicine, which is one of the largest sources of IR exposure as a diagnostic and therapeutic modality [2]. Most hospitals around the world utilize radiation for medical diagnosis and radiotherapy. Although this technology has brought benefits to the treatment of human diseases, it can also cause some damage to living organisms and side effects, which can lead to the serious ionization of biological macromolecules or chromosomal aberrations and mutations [3,4,5]. Different organs have different sensitivities to radiation and therefore the biological effects of radiation on different organs vary [6]. The gut tract, particularly gut epithelial cells and crypt stem cells, is radiation-sensitive and the biological effects as well as protection from radiation have been extensively investigated. Radiation-induced intestinal damage (RIID) is one of the most common diseases following radiation exposure [7].
The gut microbiota is inseparably associated with gut health. The gut microbiota forms a symbiotic association with the host, mainly through its metabolites, maintaining a dynamic balance between the pathological and physiological processes of the host. But any alteration to this balance leads to ecological disorders [8,9]. Studies have shown dynamic changes in the composition of the gut microbiota during and after radiotherapy for disease [10]. Irradiation therapy leads to an increase/decrease in the abundance of some specific gut microorganisms, which may induce abnormal conditions/health states [11]. Gastrointestinal (GI) complications, such as inflammatory bowel disease (IBD), have also been demonstrated to be correlated with radiation-induced disruption of the gut microbial homeostasis [12,13], whereas some microbiota will provide protection to the host from high doses of radiation by promoting hematopoietic proliferation and reducing cell death.
A number of technological and biological strategies have been developed to prevent and mitigate radiation damage to the gut, such as prophylactic surgical techniques, endoscopic treatments, topical treatments, anti-inflammatory drugs, and biological, chemical, and pharmacological therapies [14]. However, these methods also have certain limitations and risks [15]. In recent years, the relationship between radiation and the gut microbiota, as well as the relationship between the gut microbiota, its metabolites and RIID, has attracted the attention of researchers. The treatment of radiation enteritis has progressively focused on remodeling the microbiota or restoring its integrity [16,17]. Microbiota modulation may be an effective way to reduce radiation-induced GI syndromes [18].
Therefore, elucidating the link between IR, the gut microbiota and its metabolites is essential for the development of radiation injury-related preventive therapies. In this review, we will discuss the effects of IR on the gut microbiota of mammals and humans and the link between the gut microbiota and ionizing radiation-induced injuries. We hope to emphasize the protective role of the gut flora and their metabolites in the pathophysiological mechanisms manifested during radiation injury. The advantages and efficacy of modulating the gut microbiota are also discussed, and relevant therapies based on the gut flora and its metabolites are outlined. Finally, we outline the future directions of this approach for the prevention and treatment of radiation-induced injury. This study aims to clarify the potential of the gut flora and its metabolites and to explore new avenues of research and preventive therapeutic strategies for GI injuries induced by radiation exposure.

2. The Role of the Gut Microbiota in Human Health

The human gut is a remarkably complex and diverse microecosystem, home to trillions of microorganisms that play essential roles in maintaining host health [19,20]. This intricate ecosystem is composed of various microbes, including bacteria, viruses, fungi, and archaea [21].
The gut microbiota plays an important role in material metabolism, biological barriers, immune regulation and host defense [22,23,24,25]. The microbes in the GI tract assist in breaking down complex carbohydrates, fats, bile salts, and fat-soluble vitamins. They also utilize a diverse array of enzymes that the human body alone cannot produce. This microbial activity not only facilitates the efficient extraction of energy from food but also contributes to the synthesis of essential nutrients, such as short-chain fatty acids, which play a vital role in maintaining intestinal health [21,26]. In addition to their metabolic functions, gut microbes are deeply involved in regulating multiple metabolic pathways within the host. They produce metabolites that can influence distant organs, establishing a physiological link between the gut and other key systems, including the liver, muscles, and brain [27]. This gut–organ axis underscores the significance of the microbiota in overall health, as alterations in microbial composition can have far-reaching consequences. For instance, changes in gut microbiota can modulate immune responses, enhancing or impairing the body’s ability to fight off infections and other diseases.
Recent studies have highlighted the profound impact of gut microbiota on host health, particularly in relation to dysbiosis—a state of microbial imbalance [28]. Dysbiosis has been associated with a wide range of diseases, including metabolic disorders, inflammatory conditions, and even certain cancers [29,30]. The disruption of the normal microbial community can lead to altered intestinal metabolites, which may directly damage the intestinal lining or indirectly impair gut function by disrupting communication between the gut and other organs. In the context of colorectal cancer, research has shown that germ-free animal models with antibiotic-induced dysbiosis (GF-AOMS) exhibited an increased abundance of Enterobacteriaceae and secondary bile acids like taurodeoxycholic acid (TDCA). These changes were linked to the activation of the MAPK/ERK pathway and impairment of the intestinal barrier, ultimately compromising the integrity of the colonic epithelium [31].
Moreover, the microbial transformation of certain substances has emerged as a key metabolic marker in the progression of various diseases [32]. As research into the gut microbiota and its metabolites continues to deepen, there is growing interest in the potential of manipulating the microbial community to prevent or treat disease [27,33,34]. By targeting specific microbes or modulating the production of particular metabolites, it may be possible to restore balance within the gut ecosystem and improve host health outcomes. In summary, the gut microbiota is an indispensable component of human physiology, with its influence extending far beyond the GI tract. Understanding the dynamics of this microbial ecosystem and its interactions with the host is crucial for developing innovative strategies to prevent and treat diseases associated with dysbiosis. As scientific knowledge in this field expands, the potential for microbiota-based therapies continues to offer promising avenues for improving health and combating a range of conditions.

3. Radiation-Induced Intestinal Damage

The gut is both a highly radiosensitive and an important dose-limiting organ. So radiation therapy for tumors of the abdominal, pelvic, and colorectal systems will typically develop RIID, leading to side effects that include vomiting, weight loss, anorexia, dehydration, diarrhea, and infections [35,36,37,38]. RIID is categorized as either acute or chronic. Acute RIID is mainly characterized by crypt cell apoptosis, which leads to usually reversible rupture of the epithelial barrier and subsequent inflammation [39]. Chronic RIID is delayed, which leads to loss of GI function and is associated with vascular sclerosis and fibrosis of the GI wall [40,41,42].

3.1. Cellular Damage and Inflammation

IR causes damage at the cellular level mainly by inducing DNA breaks, leading to cell death or dysfunction [39]. The intestinal epithelium consists of rapidly dividing cells that line the intestines and provide an important barrier against harmful substances entering the body [43]. Due to the high proliferation rate of intestinal epithelial cells, it is particularly vulnerable to radiation, and within the intestinal crypt stem cells, there exists a “hypersensitive subpopulation” that is more vulnerable to damage than proliferating cells or even lymphocytes [40,44]. When exposed to IR, the integrity of the intestinal barrier is compromised, leading to an increase in intestinal permeability, which can make it easier for pathogens and toxins to pass through the intestinal wall and trigger an inflammatory response [45,46].
The inflammatory response induced by radiation injury leads to the upregulation of various pro-inflammatory cytokines, including tumor necrosis factor-alpha (TNF-alpha), interleukin-6 (IL-6), and interleukin-1β (IL-1β), which play a key role in amplifying the inflammatory response and recruiting immune cells to the site of injury [40,47,48,49]. However, this response, while part of the body’s attempt to repair tissue, can exacerbate damage if it becomes chronic or excessive, leading to tissue inflammation and disruption of the mucosal barrier [50,51]. The persistence of these inflammatory mediators not only leads to further epithelial cell death and dysfunction, it also leads to disruption of the extracellular matrix and intercellular tight junctions, thus exacerbating barrier dysfunction [52].

3.2. Vascular Injury with Hypoxia and Fibrosis

IR severely damages the microvascular system within the intestinal tissues, injuring endothelial cells and destroying the integrity of the vascular barrier, leading to increased vascular permeability and reduced blood flow [53]. This leads to a hypoxic environment where tissues are deprived of essential oxygen and nutrients. This hypoxia not only exacerbates cellular stress, but also triggers a pro-inflammatory response [54]. The inflammatory environment recruits a variety of immune cells to the site of injury, releasing cytokines and other mediators that further propagate tissue damage, which further leads to increased vascular permeability and vasodilation [55,56]. Over time, ongoing inflammation leads to fibrosis, which can cause the intestinal wall to harden, disrupting its normal function and leading to long-term complications. The interplay between endothelial cell damage, decreased blood perfusion, hypoxia, and subsequent inflammation and fibrosis creates a vicious cycle that ultimately exacerbates damage to the intestinal wall following radiation exposure [40,57,58].

3.3. Chronic Radiation-Induced Intestinal Damage

While most epithelial and microvascular injuries occur during the acute phase of irradiation, unresolved epithelial barrier defects coupled with progressive vascular injury and fibrosis can evolve into chronic radiation-induced intestinal damage (RIID). Clinically, chronic RIID typically manifests after a latency of ≥90 days—often months to years—following radiotherapy, presenting with persistent diarrhea, malabsorption, abdominal pain, rectal bleeding, strictures, fistulae, and even obstruction, with substantial detriments to quality of life [59,60]. Notably, ~90% of patients experience permanent changes in bowel habits, and 30–66% of pelvic cancer survivors report chronic gastrointestinal symptoms, underscoring the magnitude of the problem [40,61,62].
Pathobiologically, chronic RIID is characterized by sustained endothelial dysfunction, vascular sclerosis, tissue hypoperfusion/hypoxia, and progressive fibrosis, culminating in mural thickening and mechanical stiffness [63,64]. Repetitive or fractionated irradiation reduces mucosal perfusion and oxygen delivery, perpetuates oxidative stress, and activates fibroblasts to drive excessive extracellular-matrix deposition and collagen remodeling; these events are reinforced by long-term immune-cell infiltration and persistent up-regulation of TNF-α, IL-6, and profibrotic mediators such as TGF-β, creating a self-amplifying loop that differentiates chronic from (potentially reversible) acute injury [65,66,67].
The natural history of chronic RIID reflects this multicellular cascade: (i) vascular rarefaction and barrier failure sustain ischemia and low-grade inflammation [65]; (ii) persistent myofibroblasts and extracellular matrix remodeling consolidate strictures and stiffness [68]; and (iii) attrition of the intestinal stem-cell niche limits crypt regeneration, predisposing to epithelial atrophy and ulceration [69]. Because multiple bowel segments can lie within the radiation field, symptom clusters often arise from more than a single physiological driver and may progress despite apparent resolution of early acute toxicity.

4. Interaction Between Gut Microbiota, Its Metabolites and Radiological Bowel Injury

4.1. Changes in Gut Microbiota and Its Metabolites After Radiotherapy

In a healthy state, the composition and species of the gut microbiota remain in a relatively stable state, and the main phyla of the gut microbiota are Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria, and Fusobacteria, of which the proportion of Firmicutes and Bacteroidetes is largest [70,71]. IR causes significant changes in the gut flora of organisms, and high doses of radiation directly kill microbial cells in the gut (Table 1), thereby reducing overall microbial diversity and abundance [7,72,73].
Analysis of changes in gut microbial communities based on high-throughput sequencing techniques (marker gene sequencing and whole-genome shotgun metagenomics) revealed that changes in gut flora and their metabolites were not constant after radiation, which may be related to radiation duration, radiation level, radiation type and experimental subjects [52,80,81]. The following section focuses on the effects of IR on the intestinal flora of mammals and humans (Table 2).
As can be seen from Table 1, the ratio of Bacteroidetes to Firmicutes decreased in most cases, while the number of Helicobacter and Clostridium increased, while the metabolites that showed significant differences were mainly related to short-chain fatty acids, lipids and amino acids [12,72,81,82,83,84,85]. And we found that most of the previous studies have focused on higher doses of acute radiation therapy, probably due to equipment limitations and other reasons. However, radiation therapy is a longer cycle, and there are few studies on low-dose, long-term radiation therapy, which is a weak point in the field of radiation research and deserves more in-depth studies in the future.

4.2. The Role of Gut Microbiota and Its Metabolites in Radiation-Induced Intestinal Damage

The interactions between IR, host response and the gut microbiota are complex, and the exact underlying mechanisms are still under investigation [86]. Intestinal flora and their metabolites may attenuate radiation-induced injury and reduce pro-inflammatory responses to prevent or minimize radiation-induced injury, such as short-chain fatty acids, indole derivatives, and other intestinal microbiota metabolites, and have long-term radioprotective effects [13,82].

4.2.1. Short-Chain Fatty Acids (SCFAs)

SCFAs such as acetate (C2), propionate (C3), and butyrate (C4) are metabolites of intestinal microorganisms that act as signaling molecules to strengthen the intestinal barrier and modulate immunomodulatory functions [52].
The Toll-like receptor (TLR) affects the activation of the nuclear factor-κB (NF-κB) pathway, which, together with the secretion of pro-inflammatory cytokines (IL-1β, TNF-α, and IL-6), ultimately leads to the development of a pro-inflammatory response in ionizing radiation-induced epithelial injury [87,88]. SCFAs can inhibit the secretion of the pro-inflammatory cytokine IL-6 by inhibiting the NF-κB signaling pathway, and C4 can stimulate the expression of anti-inflammatory cytokine IL-10 through the homologous pathway GPR109A in IECs, suggesting that SCFAs can effectively alleviate radiation-induced intestinal inflammatory responses [85,89,90].
C4 has been shown to support the integrity of the intestinal barrier, promote the differentiation of colonic regulatory T cells, and exert anti-inflammatory effects [91,92]. C4 promotes gene activation of epithelial integrity-maintaining Glut1 (Slc2a1), Pgk1, and multidrug resistance protein 1 (MDR1), which maintain epithelial integrity, and increases the integrity of small intestinal villi and the number of cup cells, which attenuates RIID by facilitating the assembly of tight junctions and enhancing mucus secretion [93,94,95]. Butyrate and propionate promote anti-inflammatory pathways by inhibiting histone deacetylases (HDACs), resulting in increased histone acetylation and altered gene expression, an epigenetic modulation that is critical for maintaining a balanced immune response and attenuating radiation-induced inflammation [96,97].

4.2.2. Indoles and Their Derivatives

Indole or indole derivatives produced by tryptophan metabolism in Escherichia coli and other intestinal microbiota are signaling molecules in the intestinal tract that have a variety of protective effects on intestinal epithelial cells and may limit infrared-induced intestinal inflammation [98,99]. Indole derivatives such as indole-3-carbinol (I3C) and indole-3-propionic acid (IPA) contribute to the maintenance of intestinal homeostasis and can interact with the aryl hydrocarbon receptor (AhR), the activation of which plays a crucial role in modulating immune responses, promoting epithelial barrier function, and preventing inflammatory damage [100].
Indole and its derivatives can enhance the integrity of the intestinal barrier, thereby mitigating RIID [101,102,103,104]. IR damages rapidly dividing intestinal epithelial cells, leading to increased permeability, weakened mucus layer and impaired barrier function. Indole helps to strengthen the tight junctions between epithelial cells, thereby decreasing permeability, preventing harmful substances from entering the bloodstream from the intestinal lumen, and avoiding the exacerbation of inflammatory reactions [101,105]. At the same time, indoles and their derivatives have potent anti-inflammatory properties. They inhibit the expression of pro-inflammatory cytokines and chemokines, thereby alleviating intestinal inflammation. Also, they promote the differentiation of regulatory T cells (Tregs) and the production of anti-inflammatory cytokines, such as IL-10, through the activation of AhR [101,106,107]. This shift to an anti-inflammatory state helps reduce the chronic inflammation associated with radiation enteropathy. Indole promotes the proliferation and differentiation of intestinal stem cells and helps the intestinal mucosa to recover after radiation damage, thus alleviating the depletion of intestinal stem cells caused by IR, which is essential for stem cell renewal and repair of the epithelial lining [108,109].
IPA protects against gastrointestinal toxicity by preserving the intestinal bacterial conformation and small intestinal protein profile of radiation-challenged hosts while activating intestinal PXR/ACBP signaling, preventing radiation-associated hematopoietic syndromes and gastrointestinal syndromes without accelerating tumor growth [102]. Recent studies have shown that I3A enhances the proliferation and differentiation of Lgr5 intestinal stem cells, maintains intestinal barrier integrity, and attenuates mucosal damage. As an important tryptophan metabolite, I3A promotes proliferation of intestinal epithelial cells through the AhR/IL-10/Wnt signaling pathway and up-regulates the abundance of probiotics for the treatment of radiation-induced enteropathy [103]. Meanwhile, I3A effectively ameliorates radiation-induced enteropathy by accelerating the recovery of peripheral blood cells, promoting the recovery of bone marrow cells, and enhancing functional hematopoietic stem and progenitor cells (HSPCs). I3A effectively improves radiation-induced hematopoietic injury by accelerating peripheral blood cell recovery and enhancing functional hematopoietic stem and progenitor cell (HSPC) regeneration. Meanwhile, it can inhibit the production of intracellular reactive oxygen species (ROS) and thus inhibit the apoptosis of HSPCs induced by radiation exposure [110].

4.2.3. Other Intestinal Microbiota Metabolites

In addition to short-chain fatty acids and indoles and their derivatives, a number of intestinal microbial metabolites can likewise mitigate IRRD. The gut flora transforms ellagitanninin in pomegranates, strawberries, and walnuts into urolithin A (UroA), which reduces radiation damage to the gut by decreasing p53 expression, inhibiting caspase 8 and caspase 3 overexpression, and remodeling the gut microbiota to improve the maintenance of gut homeostasis and regeneration. PGF2α activates the FP/MAPK/NF-κB axis to promote cell proliferation and inhibit apoptosis with radiation stress, counteracts infrared-induced alveolar structural damage and collagen accumulation, and reduces post-irradiation pro-inflammatory factors (TGF-β1, IL-1β, and TNF-α), preventing and mitigating IR injury [111]. l-Histidine and its secondary metabolite imidazole propionate (ImP) activate different signaling pathways while reducing the expression of GSDMD, TNF-α and NF-κB increased by irradiation. They also play the function of radioprotection by inhibiting pyroptosis to resist radiation-induced damage [112]. Other metabolites such as tryptophan, quinolinic acid, polyamines, and bile acids can similarly provide protection to tissues by enhancing intestinal epithelial barrier function, modulating immune responses, thus preventing and alleviating RIID.

5. Therapeutics Based on Gut Microbiota and Its Metabolites

Because of medical exposures and radiological and nuclear public health emergencies, the development of safer and more effective protection strategies to prevent and treat IR damage to humans has become critical. The following section describes the safety and efficacy of widely used strategies for protection against radiation toxicity: probiotics and bacterial supplements, antibiotics and fecal microbiota transplantation and so on (Figure 1).

5.1. Advantages of Gut Microbes and Their Metabolites in Mitigating Radiation Damage

Many technological and biological strategies are currently being developed to prevent and mitigate radiation damage to the gut, such as prophylactic surgical techniques, endoscopic treatments, topical treatments, anti-inflammatory drugs, and biological, chemical, and pharmacological therapies [14]. However, all have certain limitations and drawbacks, such as the possibility of compromising mucosal healing with potential side effects, or leading to microflora dysbiosis or barrier dysfunction, and even high morbidity and mortality associated with treatment options such as surgery [113]. As mentioned earlier in the paper, dysregulation of the ecological niche of the gut flora plays an important role in the severity of radiation-induced gastrointestinal tract damage. While radiation duration, radiation level and type of radiation all affect changes in the gut flora, a symptomatic approach to restoring the optimal microbial composition may be more effective. The intestinal flora and its metabolites have been shown to have long-term radioprotective effects, which are more prominent in maintaining and restoring the integrity of the intestinal barrier, anti-inflammatory properties and restoration of the microbial homeostasis than traditional pharmacological methods. The traditional pharmacological methods are always associated with adverse effects after prolonged use, suggesting that intestinal flora and its metabolites might be beneficial even in the presence of intestinal inflammation, meeting the criteria of an ideal therapeutic agent for radiological protection.

5.2. Probiotics and Bacterial Supplementation

Probiotics are a group of active microorganisms that colonize the gut, restore the balance of the gut microbiota, and support overall health by modulating the immune response and maintaining the integrity of the gut, mainly including Lactobacillus, Clostridium casei, Bifidobacterium, Actinomyces and Lactobacillus acidophilus [114,115,116].
Lactobacillus species and their components primarily influence the immune response by facilitating the exchange of immune signals between the gastrointestinal tract and other organs [117]. Lactobacillus reuteri, known as a second-generation probiotic, can stabilize the number and capacity of Lgr5 intestinal crypt stem cells and protect intestinal microvascular endothelial cells by releasing IL-22, which directly inhibits the growth of intestinal pathogens. Pepoyan et al. found that Lactobacillus rhamnosus Vahe and Lactobacillus delbrueckii IAHAHI were the most promising radioprotective probiotics at radiation doses less than 20 Gy, and that they mitigated radiation damage by affecting leukocyte and glucose levels, and significantly reduced the symptoms of radiation-induced diarrhea [118]. Lactobacillus rhamnosus GG contribute to the protection and repair of the intestinal mucosa from radiation-induced damage through the release of lipoteichoic acid, macrophage activation and the migration of mesenchymal stem cells, and together with Bifidobacterium longum, produces SCFAs and enhances epithelial repair [119,120]. Meanwhile, standard doses of Lactobacillus acidophilus LAC-361 and Bifidobacterium longum BB-536 reduce grade 2, 3, and 4 diarrhea due to radiation enteritis in surgical patients [121].
A new generation of probiotics, including bacterial strains such as Akermansia muciniphila and Faecalibacterium prausnitzii, which are naturally occurring in the human gut but have not traditionally been used as probiotics, maintain intestinal barrier function and have anti-inflammatory properties [122,123,124]. Studies in animal models have shown that supplementation with these bacteria reduces intestinal permeability and attenuates inflammation following radiation exposure [125,126].
Combinations of probiotics have been used safely and extensively in the treatment of gastrointestinal disorders. The mixtures of prebiotics (nondigestible food ingredients that selectively stimulate the growth or activity of beneficial gut microorganisms, such as oligosaccharides, inulin, and resistant starches) and probiotics (oligogalactose, L. acidophilus, and Lactobacillus casei) have shown a positive effect in repairing damage from IR and in protecting patients with radiation-induced diarrhoea, and probiotic complexes have also been shown to play a positive role in acute RIID [37,127].

5.3. Antibiotics

Antibiotics, which treat individuals by killing and inhibiting pathogenic bacteria in their bodies, have a major impact on the restoration of the gut microbiota and have long been used to control infections in patients with compromised immune systems [128,129,130]. Some researchers have linked the antimicrobial properties of antibiotics to intestinal bacterial infections following radiation, suggesting that eliminating intestinal microbes with antibiotics may eliminate radiation effects [9,131]. Zhao et al. conducted a study on the specific mechanisms by which antibiotics reduce RIID, and their results showed that both antibiotic cocktail and metronidazole were beneficial in rebalancing the dysbiosis of the gut microbiota after radiation [132,133]. The antibiotic cocktail effectively mitigated radiation-induced microbiota disruption and intestinal damage by inhibiting the TLR4/MyD88/NF-κB signaling pathway, decreasing LPS levels, modulating macrophage polarization, and regulating multiple cytokines [133]. Studies have shown that administration of ciprofloxacin, an antibiotic targeting Gram-negative bacteria, reduces systemic bacterial translocation and improves survival after radiation exposure in mice, suggesting that some antibiotics can selectively target pathogenic bacteria without severely damaging beneficial microorganisms [134,135]. These findings suggest that antibiotic therapy has good results in targeting RIID.
Although antibiotics are essential for the treatment of ionizing radiation-induced dysbiosis and the prevention of bacterial infections, they may also have an indirect effect on the composition of the gut microbiota in different individuals and may instead promote an inflammatory response [136,137,138]. Exposure of wild-type mice to antibiotics and their subsequent monitoring for more than 700 days found that early exposure to antibiotics disrupted the development of the gut microbiota, leading to an increased inflammatory response and a shorter lifespan [139]. Notably, both epidemiologic studies and experimental data suggest that the effects of antibiotics may accumulate in the body. In the long run it may lead to the development of drug resistance. The elimination of commensal bacteria plays a key role in maintaining intestinal integrity, regulating immune responses, and nutrient metabolism. Ultimately, antibiotics can severely alter the structure of the intestinal microbiota, which can adversely affect intellectual performance and health [140,141,142,143]. On the other hand, antibiotic therapy may cause bacterial infections resulting from damage to the intestinal lining, and this damage can impair the intestinal barrier function and allow pathogenic bacteria to invade the bloodstream [144,145].
In conclusion, antibiotics are effective against intestinal bacterial infections caused by abdominal radiation, but they should be applied in a standardized manner in order to avoid the emergence of a resistance crisis.

5.4. Fecal Microbiota Transplantation

Fecal microbiota transplantation (FMT) involves extracting functional bacteria from the feces of healthy individuals through procedures such as centrifugal filtration and transplanting them into the gastrointestinal tract of a patient to restore balance to the dysbiotic intestinal microbiota for therapeutic purposes [146,147]. FMT has attracted attention as a potential treatment for a wide range of disorders related to dysbiosis of the intestinal flora [148,149,150]. Generally, FMT is used for the treatment of recurrent Clostridioides difficile infections. However, it has also been reported to be effective in ulcerative colitis and irritable bowel syndrome, where it improves the intestinal microbiota and reduces intestinal inflammation, thus potentially mitigating RIID.
Xiao et al. observed higher levels of microbial-derived IPA in the feces of irradiated mice subjected to FMT, which reduced chronic inflammation, lowered the risk of hematopoietic organ damage and bone marrow suppression, and improved gastrointestinal function and epithelial integrity [102]. Wang et al. found FMT to be an effective treatment for chronic radiculitis, and patients experienced significant relief of clinical symptoms after two courses of FMT, which demonstrated the short- and long-term efficacy of the therapy, while the shift from pathogenic to beneficial bacteria with relief of clinical symptoms illustrated the therapeutic potential of FMT. A study showed that FMT increased survival and peripheral white blood cell counts and improved gastrointestinal function and intestinal epithelial integrity in irradiated mice [151]. Meanwhile, FMT preserved intestinal bacterial composition in a sex-specific manner and preserved the mRNA and long-stranded non-coding RNA expression profiles of the host small intestine. Meanwhile, FMT promoted angiogenesis but did not accelerate the proliferation and growth of cancer cells in vivo [151].
All of the above studies suggest that FMT is a promising treatment for modifying microbial communities. However, there are risks associated with the procedure, including the potential spread of infectious agents and the lack of standardized protocols for donor selection, screening and fecal disposal [152,153]. In addition, the long-term effects of FMT are not yet fully understood, and more research is needed to determine its safety and efficacy in different patient populations, including radiation-injured patients.

5.5. Other Protection Strategies

The host and diet can control and influence the growth of the gut flora, which has been established as a method of preventing and mitigating IR-induced injury by remodeling IR-induced dysbiosis of the gut bacterial ecology. Natural herbs and their active ingredients can inhibit radiation-induced tissue damage and promote repair. And its mechanism of action is multi-targeted, multi-layered and, most importantly, with minimal side effects [154,155]. It has been shown that baicalin have radioprotective properties and can substantially rebalance radiation-induced flora dysbiosis [156]. Quercetin may mitigate IR injury by modulating the gut microbiota and reducing inflammatory cytokine levels [157]. Strategies such as sanguinarine and phycocyanin can reduce inflammatory cytokine levels by reducing LPS-producing bacteria or blocking the TLR4/Myd88/NF-B pathway [158].
These therapies provide good concepts for protection and repair of radiation damage (Table 3). However, their safety and the stability of potential therapeutic effects are uncertain and therefore need to be further explored in future studies.

6. Conclusions and Future Directions

RIID is a complex and challenging condition that has a significant impact on patients undergoing radiotherapy. Understanding the mechanisms, risk factors, and clinical manifestations of RIID is essential for developing effective management and prevention strategies. IR causes radiation enteritis by disrupting the immune system, disrupting the balance of the intestinal microbiota, and damaging the intestinal and vascular endothelium. The gut flora plays a crucial role in the etiology and treatment of radiation-induced injuries. As irradiation promotes the proliferation of harmful gut microbes, increasing the risk of intestinal diseases, beneficial bacteria that naturally inhabit the gut can directly affect the intestinal epithelium or modulate the intestinal immune system, thereby providing radiation protection. This review emphasizes the key role and mechanisms of the gut microbiota and its metabolites, as well as the complex interactions between the microbiota and radiation potency and injury, providing promising therapeutic avenues—microbiota-based therapies such as probiotics, FMT, antibiotics, and others—which have been discussed as a potential strategy for controlling radiation-induced injury. However, at the same time, RIID can be further exacerbated by reductions in probiotic populations and expression of their metabolites, etc. Despite the challenges, the potential benefits of microbiota-based therapies are substantial. Future research should focus on elucidating the specific mechanisms by which the gut microbiota influences radiation injury, optimizing therapeutic strategies. By deepening our understanding of the gut microbiota and its metabolites, we can develop innovative therapeutic approaches to prevent and manage radiation-induced injury, thereby improving patient outcomes and quality of life.

Author Contributions

Conceptualization, H.Z. and X.Y.; methodology, H.Z.; software, H.Z. and X.Y.; validation, H.Z. and X.Y.; formal analysis, H.S. and Y.C.; investigation, H.Z., H.S. and X.Y.; data curation, Y.Z., S.Y. and J.Z. (Jian Zhang); writing—original draft preparation, H.Z.; writing—review and editing, M.C. and J.Z. (Jian Zhao); visualization, C.H. (Chaoyi Han) and Q.C.; supervision, N.Z., C.H. (Changyi Huang) and J.W.; project administration, M.C. and J.Z. (Jian Zhao). All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Central Guidance on Local Science and Technology Development Fund of Sichuan Province, No. 2024ZYD0194; Open Project Program of Panxi Crops Research and Utilization Key Laboratory of Sichuan Province, No. SZKF202309; Science and Technology Plan Project of Chengdu Longquanyi District, No. 2024LQRD0045.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study.

Acknowledgments

The successful completion of this study would not have been possible without the support and collaboration of all authors. Special thanks are extended to the School of Life Sciences at Sichuan University and Sichuan Provincial People’s Hospital for their generous assistance.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Dartnell, L.R. Ionizing radiation and life. Astrobiology 2011, 6, 551–582. [Google Scholar] [CrossRef] [PubMed]
  2. Masjedi, H.; Zare, M.H.; Keshavarz Siahpoush, N.; Razavi-Ratki, S.K.; Alavi, F.; Shabani, M. European trends in radiology: Investigating factors affecting the number of examinations and the effective dose. La Radiol. Med. 2020, 125, 296–305. [Google Scholar] [CrossRef] [PubMed]
  3. Christensen, D.M.; Livingston, G.K.; Sugarman, S.L.; Parillo, S.J.; Glassman, E.S. Management of ionizing radiation injuries and illnesses, part 3: Radiobiology and health effects of ionizing radiation. J. Osteopath. Med. 2014, 114, 556–565. [Google Scholar] [CrossRef] [PubMed]
  4. Saleh, H.M.; Hassan, A.I. Editorial: Ionizing radiation and reproductive health. Front. Public Health 2023, 11, 1147934. [Google Scholar] [CrossRef]
  5. Szatkowska, M.; Krupa, R. Regulation of dna damage response and homologous recombination repair by microRNA in human cells exposed to ionizing radiation. Cancers 2020, 12, 1838. [Google Scholar] [CrossRef]
  6. Talapko, J.; Talapko, D.; Katalinić, D.; Kotris, I.; Erić, I.; Belić, D.; Mihaljević, M.V.; Vasilj, A.; Erić, S.; Flam, J.; et al. Health effects of ionizing radiation on the human body. Medicina 2024, 60, 653. [Google Scholar] [CrossRef]
  7. Kumagai, T.; Rahman, F.; Smith, A.M. The microbiome and radiation induced-bowel injury: Evidence for potential mechanistic role in disease pathogenesis. Nutrients 2018, 10, 1405. [Google Scholar] [CrossRef]
  8. Di Vincenzo, F.; Del Gaudio, A.; Petito, V.; Lopetuso, L.R.; Scaldaferri, F. Gut microbiota, intestinal permeability, and systemic inflammation: A narrative review. Intern. Emerg. Med. 2024, 19, 275–293. [Google Scholar] [CrossRef]
  9. Xin, J.-Y.; Wang, J.; Ding, Q.-Q.; Chen, W.; Xu, X.-K.; Wei, X.-T.; Lv, Y.-H.; Wei, Y.-P.; Feng, Y.; Zu, X.-P. Potential role of gut microbiota and its metabolites in radiation-induced intestinal damage. Ecotoxicol. Environ. Saf. 2022, 248, 114341. [Google Scholar] [CrossRef]
  10. Sokol, H.; Adolph, T.E. The microbiota: An underestimated actor in radiation-induced lesions? Gut 2018, 67, 1–2. [Google Scholar] [CrossRef]
  11. Jameus, A.; Dougherty, J.; Narendrula, R.; Levert, D.; Valiquette, M.; Pirkkanen, J.; Lalonde, C.; Bonin, P.; Gagnon, J.D.; Appanna, V.D.; et al. Acute impacts of ionizing radiation exposure on the gastrointestinal tract and gut microbiome in mice. Int. J. Mol. Sci. 2024, 25, 3339. [Google Scholar] [CrossRef]
  12. Kim, Y.S.; Kim, J.; Park, S. High-throughput 16s rRNA gene sequencing reveals alterations of mouse intestinal microbiota after radiotherapy. Anaerobe 2015, 33, 1–7. [Google Scholar] [CrossRef]
  13. Yu, Y.; Lin, X.; Feng, F.; Wei, Y.; Wei, S.; Gong, Y.; Guo, C.; Wang, Q.; Shuai, P.; Wang, T.; et al. Gut microbiota and ionizing radiation-induced damage: Is there a link? Environ. Res. 2023, 229, 115947. [Google Scholar] [CrossRef]
  14. Shadad, A.K.; Sullivan, F.J.; Martin, J.D.; Egan, L.J. Gastrointestinal radiation injury: Prevention and treatment. World J. Gastroenterol. 2013, 2, 199–208. [Google Scholar] [CrossRef]
  15. Wang, K.; Tepper, J.E. Radiation therapy-associated toxicity: Etiology, management, and prevention. CA Cancer J. Clin. 2021, 71, 437–454. [Google Scholar] [CrossRef]
  16. Amit, U.; Facciabene, A.; Ben-Josef, E. Radiation therapy and the microbiome; More than a gut feeling. Cancer J. 2023, 29, 84–88. [Google Scholar] [CrossRef]
  17. Zhang, Y.; Dong, Y.; Lu, P.; Wang, X.; Li, W.; Dong, H.; Fan, S.; Li, D. Gut metabolite urolithin a mitigates ionizing radiation-induced intestinal damage. J. Cell Mol. Med. 2021, 25, 10306–10312. [Google Scholar] [CrossRef] [PubMed]
  18. Chen, G.; Han, Y.; Zhang, H.; Tu, W.; Zhang, S. Radiotherapy-induced digestive injury: Diagnosis, treatment and mechanisms. Front. Oncol. 2021, 11, 757973. [Google Scholar] [CrossRef] [PubMed]
  19. Bäckhed, F.; Ley, R.E.; Sonnenburg, J.L.; Peterson, D.A.; Gordon, J.I. Host-bacterial mutualism in the human intestine. Science 2005, 307, 1915–1920. [Google Scholar] [CrossRef] [PubMed]
  20. Proctor, L.M.; Creasy, H.H.; Fettweis, J.M.; Lloyd-Price, J.; Mahurkar, A.; Zhou, W. The integrative human microbiome project. Nature 2019, 569, 641–648. [Google Scholar]
  21. Adak, A.; Khan, M.R. An insight into gut microbiota and its functionalities. Cell Mol. Life Sci. 2019, 76, 473–493. [Google Scholar] [CrossRef] [PubMed]
  22. Cani, P.D. Human gut microbiome: Hopes, threats and promises. Gut 2018, 67, 1716–1725. [Google Scholar] [CrossRef] [PubMed]
  23. Mowat, A.M.; Agace, W.W. Regional specialization within the intestinal immune system. Nat. Rev. Immunol. 2014, 14, 667–685. [Google Scholar] [CrossRef]
  24. O’Neill, L.A.J.; Golenbock, D.; Bowie, A.G. The history of toll-like receptors—Redefining innate immunity. Nat. Rev. Immunol. 2013, 13, 453–460. [Google Scholar] [CrossRef]
  25. Wilson, I.D.; Nicholson, J.K. Gut microbiome interactions with drug metabolism, efficacy, and toxicity. Transl. Res. 2017, 179, 204–222. [Google Scholar] [CrossRef]
  26. Bergman, E.N. Energy contributions of volatile fatty acids from the gastrointestinal tract in various species. Physiol. Rev. 1990, 70, 567–590. [Google Scholar] [CrossRef]
  27. Nicholson, J.K.; Holmes, E.; Kinross, J.; Burcelin, R.; Gibson, G.; Jia, W.; Pettersson, S. Host-gut microbiota metabolic interactions. Science 2012, 336, 1262–1267. [Google Scholar] [CrossRef]
  28. Chen, L.; Wang, D.; Garmaeva, S.; Kurilshikov, A.; Vich Vila, A.; Gacesa, R.; Sinha, T.; Segal, E.; Weersma, R.; Wijmenga, C.; et al. The long-term genetic stability and individual specificity of the human gut microbiome. Cell 2021, 184, 2302–2315. [Google Scholar] [CrossRef]
  29. Song, W.; Yue, Y.; Zhang, Q. Imbalance of gut microbiota is involved in the development of chronic obstructive pulmonary disease: A review. Biomed. Pharmacother. 2023, 165, 115150. [Google Scholar] [CrossRef]
  30. Teramoto, Y.; Akagawa, S.; Hori, S.; Tsuji, S.; Higasa, K.; Kaneko, K. Dysbiosis of the gut microbiota as a susceptibility factor for kawasaki disease. Front. Immunol. 2023, 14, 1268453. [Google Scholar] [CrossRef]
  31. Bai, X.; Wei, H.; Liu, W.; Coker, O.O.; Gou, H.; Liu, C.; Zhao, L.; Li, C.; Zhou, Y.; Wang, G.; et al. Cigarette smoke promotes colorectal cancer through modulation of gut microbiota and related metabolites. Gut 2022, 71, 2439–2450. [Google Scholar] [CrossRef] [PubMed]
  32. Schugar, R.C.; Shih, D.M.; Warrier, M.; Helsley, R.N.; Burrows, A.; Ferguson, D.; Brown, A.; Gromovsky, A.; Heine, M.; Chatterjee, A.; et al. The tmao-producing enzyme flavin-containing monooxygenase 3 regulates obesity and the beiging of white adipose tissue. Cell Rep. 2017, 20, 279. [Google Scholar] [CrossRef] [PubMed]
  33. Lou, M.; Cao, A.; Jin, C.; Mi, K.; Xiong, X.; Zeng, Z.; Pan, X.; Qie, J.; Qiu, S.; Niu, Y.; et al. Deviated and early unsustainable stunted development of gut microbiota in children with autism spectrum disorder. Gut 2022, 71, 1588–1599. [Google Scholar] [CrossRef] [PubMed]
  34. Zhang, X.; Coker, O.O.; Chu, E.S.; Fu, K.; Lau, H.C.H.; Wang, Y.; Chan, A.; Wei, H.; Yang, X.; Sung, J.; et al. Dietary cholesterol drives fatty liver-associated liver cancer by modulating gut microbiota and metabolites. Gut 2021, 70, 761–774. [Google Scholar] [CrossRef]
  35. Eltahawy, N.A.; Elsonbaty, S.M.; Abunour, S.; Zahran, W.E. Erratum to: Synergistic effect of aluminum and ionizing radiation upon ultrastructure, oxidative stress and apoptotic alterations in paneth cells of rat intestine. Environ. Sci. Pollut. Res. Int. 2017, 24, 15860. [Google Scholar] [CrossRef]
  36. Kim, B.H.; Jung, H.; Seo, S.H.; Shin, H.; Kwon, J.; Suh, J.M. Synergistic actions of fgf2 and bone marrow transplantation mitigate radiation-induced intestinal injury. Cell Death Dis. 2018, 9, 383. [Google Scholar] [CrossRef]
  37. 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. [Google Scholar] [CrossRef]
  38. Suman, S.; Kumar, S.; Moon, B.; Fornace, A.J.; Kallakury, B.V.S.; Datta, K. Increased transgenerational intestinal tumorigenesis in offspring of ionizing radiation exposed parent apc1638n/+ mice. J. Cancer 2017, 8, 1769–1773. [Google Scholar] [CrossRef]
  39. Classen, J.; Belka, C.; Paulsen, F.; Budach, W.; Hoffmann, W.; Bamberg, M. Radiation-induced gastrointestinal toxicity. Pathophysiology, approaches to treatment and prophylaxis. Strahlenther. Onkol. 1998, 174 (Suppl. S3), 82–84. [Google Scholar]
  40. Lu, L.; Li, W.; Chen, L.; Su, Q.; Wang, Y.; Guo, Z.; Lu, Y.; Liu, B.; Qin, S. Radiation-induced intestinal damage: Latest molecular and clinical developments. Future Oncol. 2019, 15, 4105–4118. [Google Scholar] [CrossRef]
  41. Mutsumi, M.; Masahiro, N.; Kazuko, S.; Kumio, O.; Toshiyuki, N.; Ichiro, S. Basic fibroblast growth factor suppresses radiation-induced apoptosis and tp53 pathway in rat small intestine. Radiat. Res. 2010, 174, 52–61. [Google Scholar] [CrossRef]
  42. Takemura, N.; Kurashima, Y.; Mori, Y.; Okada, K.; Ogino, T.; Osawa, H.; Matsuno, H.; Aayam, L.; Kaneto, S.; Park, E.; et al. Eosinophil depletion suppresses radiation-induced small intestinal fibrosis. Sci. Transl. Med. 2018, 10, eaan333. [Google Scholar] [CrossRef]
  43. Allaire, J.M.; Crowley, S.M.; Law, H.T.; Chang, S.; Ko, H.; Vallance, B.A. The intestinal epithelium: Central coordinator of mucosal immunity. Trends Immunol. 2018, 39, 677–696. [Google Scholar] [CrossRef]
  44. POTTENCS Extreme sensitivity of some intestinal crypt cells to x and γ irradiation. Nature 1977, 269, 518–521. [CrossRef]
  45. Shadad, A.K.; Sullivan, F.J.; Martin, J.D.; Egan, L.J. Gastrointestinal radiation injury: Symptoms, risk factors and mechanisms. World J. Gastroenterol. 2013, 2, 185–198. [Google Scholar] [CrossRef]
  46. Chen, G.; Yu, Z.; Zhang, Y.; Liu, S.; Chen, C.; Zhang, S. Radiation-induced gastric injury during radiotherapy: Molecular mechanisms and clinical treatment. J. Radiat. Res. 2023, 64, 870–879. [Google Scholar] [CrossRef]
  47. Gulati, K.; Guhathakurta, S.; Joshi, J.C.; Rai, N.; Ray, A. Cytokines and their role in health and disease: A brief overview. MOJ Immunol. 2016, 4, 00121. [Google Scholar] [CrossRef]
  48. Han, C.; Godfrey, V.; Liu, Z.; Han, Y.; Liu, L.; Peng, H.; Weichselbaum, R.; Zaki, H.; Fu, Y. The aim2 and nlrp3 inflammasomes trigger il-1–mediated antitumor effects during radiation. Sci. Immunol. 2021, 6, eabc6998. [Google Scholar] [CrossRef]
  49. Linard, C.; Marquette, C.; Mathieu, J.; Pennequin, A.; Clarençon, D.; Mathé, D. Acute induction of inflammatory cytokine expression after γ-irradiation in the rat: Effect of an NF-κb inhibitor. Int. J. Radiat. Oncol. Biol. Phys. 2004, 58, 427–434. [Google Scholar] [CrossRef]
  50. Damm, R.; Pech, M.; Haag, F.; Cavalli, P.; Gylstorff, S.; Omari, J.; Seidensticker, R.; Ricke, J.; Seidensticker, M.; Relja, B. TNF-α indicates radiation-induced liver injury after interstitial high dose-rate brachytherapy. In Vivo 2022, 36, 2265. [Google Scholar] [CrossRef]
  51. Kwong, J.; Chan, F.L.; Wong, K.; Birrer, M.J.; Archibald, K.M.; Balkwill, F.R.; Berkowitz, R.; Mok, S. Inflammatory cytokine tumor necrosis factor α confers precancerous phenotype in an organoid model of normal human ovarian surface epithelial cells. Neoplasia 2009, 11, 529–541. [Google Scholar] [CrossRef]
  52. Kim, M.H.; Kang, S.G.; Park, J.H.; Yanagisawa, M.; Kim, C.H. Short-chain fatty acids activate gpr41 and gpr43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology 2013, 145, 396–406. [Google Scholar] [CrossRef]
  53. Baker, D.G.; Krochak, R.J. The response of the microvascular system to radiation: A review. Cancer Investig. 1989, 7, 287–294. [Google Scholar] [CrossRef]
  54. Wijerathne, H.; Langston, J.C.; Yang, Q.; Sun, S.; Miyamoto, C.; Kilpatrick, L.E.; Kiani, M. Mechanisms of radiation-induced endothelium damage: Emerging models and technologies. Radiother. Oncol. 2021, 158, 21–32. [Google Scholar] [CrossRef]
  55. Hekim, N.; Cetin, Z.; Nikitaki, Z.; Cort, A.; Saygili, E.I. Radiation triggering immune response and inflammation. Cancer Lett. 2015, 368, 156–163. [Google Scholar] [CrossRef]
  56. Medzhitov, R. Origin and physiological roles of inflammation. Nature 2008, 454, 428–435. [Google Scholar] [CrossRef]
  57. Carr, N.D.; Pullen, B.R.; Hasleton, P.S.; Schofield, P.F. Microvascular studies in human radiation bowel disease. Gut 1984, 25, 448. [Google Scholar] [CrossRef]
  58. Zhao, W.; Robbins, M.E.C. Inflammation and chronic oxidative stress in radiation-induced late normal tissue injury: Therapeutic implications. Curr. Med. Chem. 2009, 2, 130–143. [Google Scholar] [CrossRef]
  59. Olopade, F.A.; Norman, A.; Blake, P.; Dearnaley, D.P.; Harrington, K.J.; Khoo, V.; Tait, D.; Hackett, C.; Andreyev, H. A modified inflammatory bowel disease questionnaire and the vaizey incontinence questionnaire are simple ways to identify patients with significant gastrointestinal symptoms after pelvic radiotherapy. Br. J. Cancer 2005, 92, 1663–1670. [Google Scholar] [CrossRef]
  60. Dalsania, R.M.; Shah, K.P.; Stotsky-Himelfarb, E.; Hoffe, S.; Willingham, F.F. Management of long-term toxicity from pelvic radiation therapy. Am. Soc. Clin. Oncol. Educ. Book 2021, 41, 1–11. [Google Scholar] [CrossRef]
  61. Yeh, M.; Chang, Y.; Tsai, Y.; Chen, S.; Huang, T.; Chiu, J.; Chang, H. Bone marrow derived macrophages fuse with intestine stromal cells and contribute to chronic fibrosis after radiation. Radiother. Oncol. 2016, 119, 250–258. [Google Scholar] [CrossRef] [PubMed]
  62. Berntsson, H.; Thien, A.; Hind, D.; Stewart, L.; Mahzabin, M.; Tung, W.S.; Bradburn, M.; Kurien, M. Interventions for managing late gastrointestinal symptoms following pelvic radiotherapy: A systematic review and meta-analysis. Clin. Oncol. (R. Coll. Radiol.) 2024, 36, 318–334. [Google Scholar] [CrossRef] [PubMed]
  63. Fan, J.; Lin, B.; Fan, M.; Niu, T.; Gao, F.; Tan, B.; Du, X. Research progress on the mechanism of radiation enteritis. Front. Oncol. 2022, 12, 888962. [Google Scholar] [CrossRef] [PubMed]
  64. Hu, Z.; Zhang, J.; Li, H.; Wang, X.; Zhang, G.; Cui, H.; Nian, J. Research progress on the hallmarks of radiation-induced intestinal injury: Mechanisms, biomarkers and therapeutic targets. Arch. Biochem. Biophys. 2025, 772, 110562. [Google Scholar] [CrossRef]
  65. 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]
  66. Shukla, P.K.; Gangwar, R.; Manda, B.; Meena, A.S.; Yadav, N.; Szabo, E.; Balogh, A.; Lee, S.; 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]
  67. Zheng, M.; Liu, Z.; He, Y. Radiation-induced fibrosis: Mechanisms and therapeutic strategies from an immune microenvironment perspective. Immunology 2024, 172, 533–546. [Google Scholar] [CrossRef]
  68. Chen, S.; Guo, B.; Feng, A.; Wang, X.; Zhang, S.; Miao, C. Pathways regulating intestinal stem cells and potential therapeutic targets for radiation enteropathy. Mol. Biomed. 2024, 5, 46. [Google Scholar] [CrossRef]
  69. 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, 485–496. [Google Scholar] [CrossRef]
  70. Stojanov, S.; Berlec, A.; Štrukelj, B. The influence of probiotics on the firmicutes/bacteroidetes ratio in the treatment of obesity and inflammatory bowel disease. Microorganisms 2020, 8, 1715. [Google Scholar] [CrossRef]
  71. Fernandes, A.; Oliveira, A.; Soares, R.; Barata, P. The effects of ionizing radiation on gut microbiota, a systematic review. Nutrients 2021, 13, 3025. [Google Scholar] [CrossRef]
  72. 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, e126312. [Google Scholar] [CrossRef]
  73. Jeong, S.; Marks, B.P.; Ryser, E.T.; Moosekian, S.R. Inactivation of Escherichia coli O157:H7 on lettuce, using low-energy x-ray irradiation. J. Food Prot. 2010, 73, 547–551. [Google Scholar] [CrossRef]
  74. Clavero, M.R.; Monk, J.D.; Beuchat, L.R.; Doyle, M.P.; Brackett, R.E. Inactivation of Escherichia coli O157:H7, Salmonellae, and Campylobacter jejuni in raw ground beef by gamma irradiation. Appl. Environ. Microbiol. 1994, 60, 2069–2075. [Google Scholar] [CrossRef]
  75. Schopf, S.; Gotzmann, G.; Dietze, M.; Gerschke, S.; Kenner, L.; König, U. Investigations into the suitability of bacterial suspensions as biological indicators for low-energy electron irradiation. Front. Immunol. 2022, 13, 814767. [Google Scholar] [CrossRef]
  76. Pribil, W.; Gehringer, P.; Eschweiler, H.; Cabaj, A.; Haider, T.; Sommer, R. Assessment of Bacillus subtilis spores as a possible bioindicator for evaluation of the microbicidal efficacy of radiation processing of water. Water Environ. Res. 2007, 79, 720–724. [Google Scholar] [CrossRef]
  77. Gosiewski, T.; Mróz, T.; Ochońska, D.; Pabian, W.; Bulanda, M.; Brzychczy-Wloch, M. A study of the effects of therapeutic doses of ionizing radiation in vitro on Lactobacillus isolates originating from the vagina—A pilot study. BMC Microbiol. 2016, 16, 99. [Google Scholar] [CrossRef]
  78. Porfiri, L.; Burtscher, J.; Kangethe, R.T.; Verhovsek, D.; Cattoli, G.; Domig, K.J.; Wijewardana, V. Irradiated non-replicative lactic acid bacteria preserve metabolic activity while exhibiting diverse immune modulation. Front. Vet. Sci. 2022, 9, 859124. [Google Scholar] [CrossRef]
  79. Hazards, E.P.O.B. Scientific opinion on the efficacy and microbiological safety of irradiation of food. EFSA J. 2011, 9, 2103. [Google Scholar] [CrossRef]
  80. Bharti, R.; Grimm, D.G. Current challenges and best-practice protocols for microbiome analysis. Brief. Bioinform. 2021, 22, 178–193. [Google Scholar] [CrossRef]
  81. Goudarzi, M.; Mak, T.D.; Jacobs, J.P.; Moon, B.; Strawn, S.J.; Braun, J.; Brenner, D.; Fornace, A.; Li, H. An integrated multi-omic approach to assess radiation injury on the host-microbiome axis. Radiat. Res. 2016, 186, 219–234. [Google Scholar] [CrossRef]
  82. Guo, H.; Chou, W.; Lai, Y.; Liang, K.; Tam, J.W.; Brickey, W.J.; Chen, L.; Montgomery, N.; Li, X.; Bohannon, M.; et al. Multi-omics analyses of radiation survivors identify radioprotective microbes and metabolites. Science 2020, 370, eaay9097. [Google Scholar] [CrossRef]
  83. Li, Y.; Yan, H.; Zhang, Y.; Li, Q.; Yu, L.; Li, Q.; Liu, C.; Xie, Y.; Chen, K.; Ye, F.; et al. Alterations of the gut microbiome composition and lipid metabolic profile in radiation enteritis. Front. Cell. Infect. Microbiol. 2020, 10, 541178. [Google Scholar] [CrossRef]
  84. Zhu, S.; Liang, J.; Zhu, F.; Zhang, X.; Xu, M.; Zhao, K.; Zeng, L.; Xu, K. The effects of myeloablative or non-myeloablative total body irradiations on intestinal tract in mice. Biosci. Rep. 2021, 41, BSR20202993. [Google Scholar] [CrossRef]
  85. Fernandes, A.; Oliveira, A.; Guedes, C.; Fernandes, R.; Soares, R.; Barata, P. Ionizing radiation from radiopharmaceuticals and the human gut microbiota: An ex vivo approach. Int. J. Mol. Sci. 2022, 23, 10809. [Google Scholar] [CrossRef]
  86. Acharya, M.; Venkidesh, B.S.; Mumbrekar, K.D. Bacterial supplementation in mitigation of radiation-induced gastrointestinal damage. Life Sci. 2024, 353, 122921. [Google Scholar] [CrossRef]
  87. van Vliet, M.J.; Harmsen, H.J.M.; de Bont, E.S.J.M.; Tissing, W.J.E. The role of intestinal microbiota in the development and severity of chemotherapy-induced mucositis. PLoS Pathog. 2010, 6, e1000879. [Google Scholar] [CrossRef]
  88. Wardill, H.R.; Gibson, R.J.; Van Sebille, Y.Z.A.; Secombe, K.R.; Coller, J.K.; White, I.A.; Manavis, J.; Hutchinson, M.; Staikopoulos, V.; Logan, R.; et al. Irinotecan-induced gastrointestinal dysfunction and pain are mediated by common tlr4-dependent mechanisms. Mol. Cancer Ther. 2016, 15, 1376–1386. [Google Scholar] [CrossRef]
  89. Chang, P.V.; Hao, L.; Offermanns, S.; Medzhitov, R. The microbial metabolite butyrate regulates intestinal macrophage function via histone deacetylase inhibition. Proc. Natl. Acad. Sci. USA 2014, 111, 2247–2252. [Google Scholar] [CrossRef]
  90. Singh, N.; Gurav, A.; Sivaprakasam, S.; Brady, E.; Padia, R.; Shi, H.; Thangaraju, M.; Prasad, P.; Manicassamy, S.; Munn, D.; et al. Activation of gpr109a, receptor for niacin and the commensal metabolite butyrate, suppresses colonic inflammation and carcinogenesis. Immunity 2014, 40, 128–139. [Google Scholar] [CrossRef]
  91. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef]
  92. Zhang, L.S.; Davies, S.S. Microbial metabolism of dietary components to bioactive metabolites: Opportunities for new therapeutic interventions. Genome Med. 2016, 8, 46. [Google Scholar] [CrossRef]
  93. Bach Knudsen, K.E.; Lærke, H.N.; Hedemann, M.S.; Nielsen, T.S.; Ingerslev, A.K.; Gundelund Nielsen, D.S.; Theil, P.; Purup, S.; Hald, S.; Schioldan, A.; et al. Impact of diet-modulated butyrate production on intestinal barrier function and inflammation. Nutrients 2018, 10, 1499. [Google Scholar] [CrossRef]
  94. Dupraz, L.; Magniez, A.; Rolhion, N.; Richard, M.L.; Da Costa, G.; Touch, S.; Mayeur, C.; Planchais, J.; Agus, A.; Danne, C.; et al. Gut microbiota-derived short-chain fatty acids regulate IL-17 production by mouse and human intestinal γδ t cells. Cell Rep. 2021, 36, 109332. [Google Scholar] [CrossRef]
  95. Qiu, P.; Ishimoto, T.; Fu, L.; Zhang, J.; Zhang, Z.; Liu, Y. The gut microbiota in inflammatory bowel disease. Front. Cell. Infect. Microbiol. 2022, 12, 733992. [Google Scholar] [CrossRef]
  96. Furusawa, Y.; Obata, Y.; Fukuda, S.; Endo, T.A.; Nakato, G.; Takahashi, D.; Nakanishi, Y.; Uetake, C.; Kato, K.; Kato, T.; et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory t cells. Nature 2013, 504, 446–450. [Google Scholar] [CrossRef]
  97. Mirzaei, R.; Afaghi, A.; Babakhani, S.; Sohrabi, M.R.; Hosseini-Fard, S.R.; Babolhavaeji, K.; Akbari, S.K.; Yousefimashouf, R.; Karampoor, S. Role of microbiota-derived short-chain fatty acids in cancer development and prevention. Biomed. Pharmacother. 2021, 139, 111619. [Google Scholar] [CrossRef]
  98. Bansal, T.; Alaniz, R.C.; Wood, T.K.; Jayaraman, A. The bacterial signal indole increases epithelial-cell tight-junction resistance and attenuates indicators of inflammation. Proc. Natl. Acad. Sci. USA 2010, 107, 228–233. [Google Scholar] [CrossRef]
  99. Roager, H.M.; Licht, T.R. Microbial tryptophan catabolites in health and disease. Nat. Commun. 2018, 9, 3294. [Google Scholar] [CrossRef]
  100. Wlodarska, M.; Luo, C.; Kolde, R.; DHennezel, E.; Annand, J.W.; Heim, C.E.; Krastel, P.; Schmitt, E.K.; Omar, A.S.; Creasey, E.A.; et al. Indoleacrylic acid produced by commensal Peptostreptococcus species suppresses inflammation. Cell Host Microbe 2017, 22, 25–37. [Google Scholar] [CrossRef]
  101. Zelante, T.; Iannitti, R.G.; Cunha, C.; De Luca, A.; Giovannini, G.; Pieraccini, G.; Zecchi, R.; D’Angelo, C.; Massi-Benedetti, C.; Fallarino, F.; et al. Tryptophan catabolites from microbiota engage aryl hydrocarbon receptor and balance mucosal reactivity via interleukin-22. Immunity 2013, 39, 372–385. [Google Scholar] [CrossRef]
  102. Xiao, H.; Cui, M.; Li, Y.; Dong, J.; Zhang, S.; Zhu, C.; Jiang, M.; Zhu, T.; Wang, B.; Wang, H.; et al. Gut microbiota-derived indole 3-propionic acid protects against radiation toxicity via retaining acyl-coa-binding protein. Microbiome 2020, 8, 69. [Google Scholar] [CrossRef]
  103. Xie, L.; Cai, S.; Lu, H.; Tang, F.; Zhu, R.; Tian, Y.; Li, M. Microbiota-derived i3a protects the intestine against radiation injury by activating ahr/il-10/wnt signaling and enhancing the abundance of probiotics. Gut Microbes 2024, 16, 2347722. [Google Scholar] [CrossRef]
  104. Lu, L.; Jiang, M.; Zhu, C.; He, J.; Fan, S. Amelioration of whole abdominal irradiation-induced intestinal injury in mice with 3,3′-diindolylmethane (dim). Free Radic. Biol. Med. 2019, 130, 244–255. [Google Scholar] [CrossRef]
  105. Scott, S.A.; Fu, J.; Chang, P.V. Microbial tryptophan metabolites regulate gut barrier function via the aryl hydrocarbon receptor. Proc. Natl. Acad. Sci. USA 2020, 117, 19376–19387. [Google Scholar] [CrossRef]
  106. Lamas, B.; Natividad, J.M.; Sokol, H. Aryl hydrocarbon receptor and intestinal immunity. Mucosal. Immunol. 2018, 11, 1024–1038. [Google Scholar] [CrossRef]
  107. Murray, I.A.; Patterson, A.D.; Perdew, G.H. Aryl hydrocarbon receptor ligands in cancer: Friend and foe. Nat. Rev. Cancer 2014, 14, 801–814. [Google Scholar] [CrossRef]
  108. Leibowitz, B.J.; Zhao, G.; Wei, L.; Ruan, H.; Epperly, M.; Chen, L.; Lu, X.; Greenberger, J.S.; Zhang, L.; Yu, J. Interferon β drives intestinal regeneration after radiation. Sci. Adv. 2021, 7, eabi5253. [Google Scholar] [CrossRef]
  109. Swimm, A.; Giver, C.R.; DeFilipp, Z.; Rangaraju, S.; Sharma, A.; Ulezko Antonova, A.; Sonowal, R.; Capaldo, C.; Powell, D.; Qayed, M.; et al. Indoles derived from intestinal microbiota act via type i interferon signaling to limit graft-versus-host disease. Blood 2018, 132, 2506–2519. [Google Scholar] [CrossRef]
  110. Guan, D.; Yang, Y.; Pang, M.; Liu, X.; Li, Y.; Huang, P.; Shang, H.; Wei, H.; Ye, Z. Indole-3-carboxaldehyde ameliorates ionizing radiation-induced hematopoietic injury by enhancing hematopoietic stem and progenitor cell quiescence. Mol. Cell Biochem. 2024, 479, 313–323. [Google Scholar]
  111. Chen, Z.; Xiao, H.; Dong, J.; Li, Y.; Wang, B.; Fan, S.; Cui, M. microbiota-derived pgf2α fights against radiation-induced lung toxicity through the mapk/nf-κb pathway. Antioxidants 2021, 11, 65. [Google Scholar] [CrossRef]
  112. Chen, Z.; Wang, B.; Dong, J.; Li, Y.; Zhang, S.; Zeng, X.; Xiao, H.; Fan, S.; Cui, M. Gut microbiota-derived l-histidine/imidazole propionate axis fights against the radiation-induced cardiopulmonary injury. Int. J. Mol. Sci. 2021, 22, 11436. [Google Scholar] [CrossRef]
  113. Araujo, I.K.; Muñoz-Guglielmetti, D.; Mollà, M. Radiation-induced damage in the lower gastrointestinal tract: Clinical presentation, diagnostic tests and treatment options. Best Pract. Res. Clin. Gastroenterol. 2020, 48–49, 101707. [Google Scholar] [CrossRef]
  114. Deng, S.; Pei, C.; Cai, K.; Huang, W.; Xiao, X.; Zhang, X.; Liang, R.; Chen, Y.; Xie, Z.; Li, P.; et al. Lactobacillus acidophilus and its metabolite ursodeoxycholic acid ameliorate ulcerative colitis by promoting treg differentiation and inhibiting m1 macrophage polarization. Front. Microbiol. 2024, 15, 1302998. [Google Scholar] [CrossRef]
  115. 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. 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]
  116. Oelschlaeger, T.A. Mechanisms of probiotic actions—A review. Int. J. Med. Microbiol. 2010, 300, 57–62. [Google Scholar] [CrossRef]
  117. Wang, W.; Cui, B.; Nie, Y.; Sun, L.; Zhang, F. Radiation injury and gut microbiota-based treatment. Protein Cell 2024, 15, 83–97. [Google Scholar] [CrossRef]
  118. Pepoyan, A.Z.; Manvelyan, A.M.; Balayan, M.H.; McCabe, G.; Tsaturyan, V.V.; Melnikov, V.G.; Chikindas, M.L.; Weeks, R.; Karlyshev, A.V. The effectiveness of potential probiotics Lactobacillus rhamnosus Vahe and Lactobacillus delbrueckii IAHAHI in irradiated rats depends on the nutritional stage of the host. Probiotics Antimicrob. Proteins 2020, 12, 1439–1450. [Google Scholar] [CrossRef]
  119. Ma, X.; Shin, Y.; Jang, H.; Joo, M.; Yoo, J.; Kim, D. Lactobacillus rhamnosus and Bifidobacterium longum alleviate colitis and cognitive impairment in mice by regulating IFN-γ to IL-10 and TNF-α to IL-10 expression ratios. Sci. Rep. 2021, 11, 20659. [Google Scholar] [CrossRef]
  120. Riehl, T.E.; Alvarado, D.; Ee, X.; Zuckerman, A.; Foster, L.; Kapoor, V.; Thotala, K.; Ciorba, M.A.; Stenson, W.F. 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 2019, 68, 1003. [Google Scholar] [CrossRef]
  121. Jian, Y.; Zhang, D.; Liu, M.; Wang, Y.; Xu, Z. The impact of gut microbiota on radiation-induced enteritis. Front. Cell. Infect. Microbiol. 2021, 11, 586392. [Google Scholar] [CrossRef]
  122. Cani, P.D.; de Vos, W.M. Next-generation beneficial microbes: The case of Akkermansia muciniphila. Front. Microbiol. 2017, 8, 1765. [Google Scholar] [CrossRef]
  123. Derrien, M.; van Hylckama Vlieg, J.E.T. Fate, activity, and impact of ingested bacteria within the human gut microbiota. Trends Microbiol. 2015, 23, 354–366. [Google Scholar] [CrossRef]
  124. Reunanen, J.; Kainulainen, V.; Huuskonen, L.; Ottman, N.; Belzer, C.; Huhtinen, H.; Vos, W.M.; Satokari, R. Akkermansia muciniphila adheres to enterocytes and strengthens the integrity of the epithelial cell layer. Appl. Environ. Microbiol. 2015, 81, 3655–3662. [Google Scholar] [CrossRef]
  125. Mao, X.; Ma, J.; Jiao, C.; Tang, N.; Zhao, X.; Wang, D.; Zhang, Y.; Ye, Z.; Xu, C.; Jiang, J.; et al. Faecalibacterium prausnitzii attenuates dss-induced colitis by inhibiting the colonization and pathogenicity of Candida albicans. Mol. Nutr. Food Res. 2021, 65, 2100433. [Google Scholar] [CrossRef]
  126. Zheng, M.; Han, R.; Yuan, Y.; Xing, Y.; Zhang, W.; Sun, Z.; Liu, Y.; Li, J.; Mao, T. The role of Akkermansia muciniphila in inflammatory bowel disease: Current knowledge and perspectives. Front. Immunol. 2023, 13, 1089600. [Google Scholar] [CrossRef]
  127. Xie, P.; Luo, M.; Deng, X.; Fan, J.; Xiong, L. Outcome-specific efficacy of different probiotic strains and mixtures in irritable bowel syndrome: A systematic review and network meta-analysis. Nutrients 2023, 15, 3856. [Google Scholar] [CrossRef]
  128. Atchade, A.M.; Williams, J.L.; Mermelstein, L.; Nemesure, B. Unraveling the complexities of early-onset colorectal cancer: A perspective on dietary and microbial influences. Front. Public Health 2024, 12, 1370108. [Google Scholar] [CrossRef]
  129. Palleja, A.; Mikkelsen, K.H.; Forslund, S.K.; Kashani, A.; Allin, K.H.; Nielsen, T.; Hansen, T.H.; Liang, S.; Feng, Q.; Zhang, C.; et al. Recovery of gut microbiota of healthy adults following antibiotic exposure. Nat. Microbiol. 2018, 3, 1255–1265. [Google Scholar] [CrossRef]
  130. Suez, J.; Elinav, E. The path towards microbiome-based metabolite treatment. Nat. Microbiol. 2017, 2, 17075. [Google Scholar] [CrossRef]
  131. Hutchings, M.I.; Truman, A.W.; Wilkinson, B. Antibiotics: Past, present and future. Curr. Opin. Microbiol. 2019, 51, 72–80. [Google Scholar] [CrossRef]
  132. Strati, F.; Pujolassos, M.; Burrello, C.; Giuffrè, M.R.; Lattanzi, G.; Caprioli, F.; Troisi, J.; Facciotti, F. Antibiotic-associated dysbiosis affects the ability of the gut microbiota to control intestinal inflammation upon fecal microbiota transplantation in experimental colitis models. Microbiome 2021, 9, 39. [Google Scholar] [CrossRef]
  133. 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, 2967–2977. [Google Scholar] [CrossRef]
  134. Horseman, T.S.; Frank, A.M.; Cannon, G.; Zhai, M.; Olson, M.G.; Lin, B.; Hull, L.; Xiao, M.; Kiang, J.; Burmeister, D.M. Effects of combined ciprofloxacin and neulasta therapy on intestinal pathology and gut microbiota after high-dose irradiation in mice. Front. Public Health 2024, 12, 1365161. [Google Scholar] [CrossRef]
  135. Kiang, J.G.; Fukumoto, R. Ciprofloxacin increases survival after ionizing irradiation combined injury: γ-h2ax formation, cytokine/chemokine, and red blood cells. Health Phys. 2014, 106, 720–726. [Google Scholar] [CrossRef]
  136. Lavelle, A.; Hoffmann, T.W.; Pham, H.; Langella, P.; Guédon, E.; Sokol, H. Baseline microbiota composition modulates antibiotic-mediated effects on the gut microbiota and host. Microbiome 2019, 7, 111. [Google Scholar] [CrossRef]
  137. Rashidi, A.; Ebadi, M.; Rehman, T.U.; Elhusseini, H.; Nalluri, H.; Kaiser, T.; Holtan, S.G.; Khoruts, A.; Weisdorf, D.J.; Staley, C. Gut microbiota response to antibiotics is personalized and depends on baseline microbiota. Microbiome 2021, 9, 211. [Google Scholar] [CrossRef]
  138. Uribe-Herranz, M.; Bittinger, K.; Rafail, S.; Guedan, S.; Pierini, S.; Tanes, C.; Ganetsky, A.; Morgan, M.; Gill, A.; Tanyi, J.; et al. Gut microbiota modulates adoptive cell therapy via cd8α dendritic cells and IL-12. JCI Insight 2018, 3, e94952. [Google Scholar] [CrossRef]
  139. Lynn, M.A.; Eden, G.; Ryan, F.J.; Bensalem, J.; Wang, X.; Blake, S.J.; Choo, J.M.; Chern, Y.T.; Sribnaia, A.; James, J.; et al. The composition of the gut microbiota following early-life antibiotic exposure affects host health and longevity in later life. Cell Rep. 2021, 36, 109564. [Google Scholar] [CrossRef]
  140. Blaser, M.J. Antibiotic use and its consequences for the normal microbiome. Science 2016, 352, 544–545. [Google Scholar] [CrossRef]
  141. Jernberg, C.; Löfmark, S.; Edlund, C.; Jansson, J.K. Long-term impacts of antibiotic exposure on the human intestinal microbiota. Microbiology 2010, 156, 3216–3223. [Google Scholar] [CrossRef]
  142. Leclercq, S.; Mian, F.M.; Stanisz, A.M.; Bindels, L.B.; Cambier, E.; Ben-Amram, H.; Koren, O.; Forsythe, P.; Bienenstock, J. Low-dose penicillin in early life induces long-term changes in murine gut microbiota, brain cytokines and behavior. Nat. Commun. 2017, 8, 15062. [Google Scholar] [CrossRef]
  143. Zhang, Y.; Liang, H.; Wang, Y.; Cheng, R.; Pu, F.; Yang, Y.; Li, J.; Wu, S.; Shen, X.; He, F. Heat-inactivated Lacticaseibacillus paracasei n1115 alleviates the damage due to brain function caused by long-term antibiotic cocktail exposure in mice. BMC Neurosci. 2022, 23, 38. [Google Scholar] [CrossRef] [PubMed]
  144. Kim, C.G.; Koh, J.; Shin, S.; Shin, J.; Hong, M.; Chung, H.C.; Rha, S.Y.; Kim, H.S.; Lee, C.; Lee, J.H.; et al. Prior antibiotic administration disrupts anti-pd-1 responses in advanced gastric cancer by altering the gut microbiome and systemic immune response. Cell Rep. Med. 2023, 4, 101251. [Google Scholar] [CrossRef] [PubMed]
  145. Ma, C.; Han, M.; Heinrich, B.; Fu, Q.; Zhang, Q.; Sandhu, M.; Agdashian, D.; Terabe, M.; Berzofsky, J.A.; Fako, V.; et al. Gut microbiome–mediated bile acid metabolism regulates liver cancer via nkt cells. Science 2018, 360, eaan5931. [Google Scholar] [CrossRef]
  146. Cammarota, G.; Ianiro, G.; Tilg, H.; Rajilić-Stojanović, M.; Kump, P.; Satokari, R.; Sokol, H.; Arkkila, P.; Pintus, C.; Hart, A.; et al. European consensus conference on faecal microbiota transplantation in clinical practice. Gut 2017, 66, 569. [Google Scholar] [CrossRef]
  147. Khanna, S.; Kraft, C.S. Fecal microbiota transplantation: Tales of caution. Clin. Infect. Dis. 2021, 72, e881–e882. [Google Scholar] [CrossRef]
  148. Boicean, A.; Ichim, C.; Todor, S.B.; Anderco, P.; Popa, M.L. The importance of microbiota and fecal microbiota transplantation in pancreatic disorders. Diagnostics 2024, 14, 861. [Google Scholar] [CrossRef]
  149. Quraishi, M.N.; Widlak, M.; Bhala, N.; Moore, D.; Price, M.; Sharma, N.; Iqbal, T.H. Systematic review with meta-analysis: The efficacy of faecal microbiota transplantation for the treatment of recurrent and refractory Clostridium difficile infection. Aliment. Pharmacol. Ther. 2017, 46, 479–493. [Google Scholar] [CrossRef]
  150. Vendrik, K.E.W.; Ooijevaar, R.E.; de Jong, P.R.C.; Laman, J.D.; van Oosten, B.W.; van Hilten, J.J.; Ducarmon, Q.R.; Keller, J.J.; Kuijper, E.J.; Contarino, M.F. Fecal microbiota transplantation in neurological disorders. Front. Cell. Infect. Microbiol. 2020, 10, 98. [Google Scholar] [CrossRef]
  151. Cui, M.; Xiao, H.; Li, Y.; Zhou, L.; Zhao, S.; Luo, D.; Zheng, Q.; Dong, J.; Zhao, Y.; Zhang, X.; et al. Faecal microbiota transplantation protects against radiation-induced toxicity. EMBO Mol. Med. 2017, 9, 448–461. [Google Scholar] [CrossRef] [PubMed]
  152. Fischer, M.; Sipe, B.W.; Rogers, N.A.; Cook, G.K.; Robb, B.W.; Vuppalanchi, R.; Rex, D.K. Faecal microbiota transplantation plus selected use of vancomycin for severe-complicated Clostridium difficile infection: Description of a protocol with high success rate. Aliment. Pharmacol. Ther. 2015, 42, 470–476. [Google Scholar] [CrossRef] [PubMed]
  153. Lu, L.; Li, F.; Gao, Y.; Kang, S.; Li, J.; Guo, J. Microbiome in radiotherapy: An emerging approach to enhance treatment efficacy and reduce tissue injury. Mol. Med. 2024, 30, 105. [Google Scholar] [CrossRef] [PubMed]
  154. Wang, Y.; Gao, Y.; Feng, J.; Dou, Y. Effect of modified xijiao dihuang decoction (加味犀角地黄汤) on intestinal flora and th17/treg in rats with radiation enteritis. Chin. J. Integr. Med. 2021, 27, 198–205. [Google Scholar] [CrossRef]
  155. Zhang, X.; Chen, X.; Wang, L.; He, C.; Shi, Z.; Fu, Q.; Xu, W.; Zhang, S.; Hu, S. Review of the efficacy and mechanisms of traditional Chinese medicines as a therapeutic option for ionizing radiation induced damage. Front. Pharmacol. 2021, 12, 617559. [Google Scholar] [CrossRef]
  156. Huang, S.; Huang, Y.; Lin, W.; Wang, L.; Yang, Y.; Li, P.; Xiao, L.; Chen, Y.; Chu, Q.; Yuan, X. Sitagliptin alleviates radiation-induced intestinal injury by activating nrf2-antioxidant axis, mitigating nlrp3 inf–lammasome activation, and reversing gut microbiota disorder. Oxid. Med. Cell Longev. 2022, 2022, 2586305. [Google Scholar] [CrossRef]
  157. Hu, J.; Jiao, W.; Tang, Z.; Wang, C.; Li, Q.; Wei, M.; Song, S.; Du, L.; Jin, Y. Quercetin inclusion complex gels ameliorate radiation-induced brain injury by regulating gut microbiota. Biomed. Pharmacother. 2023, 158, 114142. [Google Scholar] [CrossRef]
  158. Gu, J.; Zhao, L.; Chen, Y.; Guo, Y.; Sun, Y.; Guo, Q.; Duan, G.X.; Li, C.; Tang, Z.B.; Zhang, Z.X.; et al. Preventive effect of sanguinarine on intestinal injury in mice exposed to whole abdominal irradiation. Biomed. Pharmacother. 2022, 146, 112496. [Google Scholar] [CrossRef]
Figure 1. Therapeutics based on gut microbiota and its metabolites intervention.
Figure 1. Therapeutics based on gut microbiota and its metabolites intervention.
Microorganisms 13 02151 g001
Table 1. Radiosensitivity of representative gut-associated bacteria to ionizing radiation.
Table 1. Radiosensitivity of representative gut-associated bacteria to ionizing radiation.
Bacterial SpeciesRadiation Type & EnergySensitivity Index (D10 kGy)References
E. coliLow-energy X-ray, 70 kV0.040–0.078[74]
γ0.24–0.35[75]
Bacillus spp.e-beam/γ0.31–0.37[76]
Low-energy X-ray3.3[77]
Lactobacillus spp.IR at clinical RT dosesrelatively resistant[78]
^60Co γ0.526[79]
Clostridium perfringensγ (Direct X-ray D10 data limited)0.40–0.80[80]
Table 2. Changes in the gut microbiota and its metabolites after radiotherapy.
Table 2. Changes in the gut microbiota and its metabolites after radiotherapy.
Radiation TypeRadiation LevelRadiation TimeObjectsBiospecimenAnalytical TechniquesChanges in the Gut Microbiota and Its MetabolitesReferences
total body radiation8.2–9.2 Gy290 daysC57BL/6 miceFeces16 S rRNA gene sequencingLachnospiraceae ↑, Enterococcaceae ↑;
short-chain fatty acids (acetate, butyrate, and propionate) ↑, I3A and KYNA ↑
[82]
X-ray5, 12 Gy30 daysC57BL/6J miceFeces16 S rRNA gene sequencingLactobacillaceae ↑, Staphylococcaceae
Lachnospiraceae ↓, Ruminococcaceae ↓, Clostridiaceae ↓;
glyceric acid ↓, homogentisic acid ↓, glutaconic acid ↓ and pipecolic acid ↓
hippuric acid ↑, taurine ↑, urobilinogen ↑
[83]
pelvic radiotherapy1.8–2.0 Gy/dayfive times a week during a 5 week period11 cancer patientsBlood16 S rRNA gene sequencing Firmicutes ↑, Bacteroidetes[72]
60Co γ-ray8 GysingleC57BL/6 miceThe small and large intestinal contents16 S rRNA gene sequencingAlistipes ↑, Lactobacillus ↑, Akkermansia
Barnesiella ↓, Prevotella ↓, Bacteroides ↓, Oscillibacter ↓, Pseudoflavonifractor ↓, Mucispirillum
[12]
X-ray18 GySingleC57BL/6 J miceIntestinal biopsy
feces
16 S rDNA sequencing
UHPLC-MS
Bacteroidetes ↓, Firmicutes ↓;
Significant changes in CerG1, CerG3GNac1, cPA, DG, dMePE, FA, ganglioside GM3, LdMePE, LPC, LPE, LPG, LPI, PC, PE, PG, PI, SM, So and the acute phase is the opposite of the chronic phase
[83]
60Co γ-ray4 Gy (non-myeloablative)/8 Gy (myeloablative)SingleBALB/c miceCecal contents16 S rRNA gene sequencing Bacteroidaceae
Ruminococcaceae ↓, Lachnospiraceae ↓, Lactobacillaceae ↓, Defluviitaleaceae ↓, Peptococcaceae ↓, Christensenellaceae
acetic acid ↓, valeric acid ↓
[84]
223Ra, 99mTc 2 healthy volunteersFeces16 S rRNA gene sequencing Firmicutes ↑, Proteobacteria
Bacteroidetes ↓, Actinobacteria
[85]
↑ Increase; ↓ Decrease.
Table 3. Microbiota-based and metabolite-targeted therapeutics for RIID.
Table 3. Microbiota-based and metabolite-targeted therapeutics for RIID.
Intervention TypeCategoryMechanism of ActionEfficacy and Outcomes
Probiotics and Bacterial Supplementation TherapyTraditional ProbioticsRegulate immune signal transduction Stabilize crypt stem cells to protect intestinal microvascular endothelial cells Modulates leukocyte counts and blood glucose levels
Activates macrophages, and promotes mesenchymal stem cell migration
Significantly reduces radiation-associated diarrhea incidence, promotes intestinal mucosal repair and regeneration
Next-Generation ProbioticsMaintains intestinal barrier integrity and exerts anti-inflammatory activityReduce intestinal permeability and alleviate inflammatory responses
Probiotic-Prebiotic Complex FormulationsRepair tissue damage and selectively promote the proliferation and metabolic activity of beneficial gut microbiotaEffectively repairs IR-induced injury, protects patients with radiation-induced diarrhea, and demonstrates clear improvement in acute RIID
Antibiotic TherapyBroad-spectrum and Targeted AntibioticsReduces LPS levels
Modulates macrophage polarization and cytokine networks
Improves intestinal dysbiosis
Effectively control intestinal bacterial infections, alleviate dysbiosis and intestinal tissue damage
FMTFecal Microbiota Transplantation TechnologyRestore gut microbiota balance
Increase IPA production and promote angiogenesis
Regulate host small intestine mRNA and long non-coding RNA expression profiles
Reduces chronic inflammatory responses and hematopoietic organ damage risk, improves gastrointestinal function and epithelial integrity
Alleviates clinical symptoms of chronic radiation enteritis
Other Protective StrategiesNatural Herbal Active ComponentsRebalances radiation-induced dysbiosis
Reduces inflammatory cytokine levels
Inhibits inflammatory cascade reactions
Exhibits definite radioprotective activity and mitigates IR-induced tissue damage
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zhu, H.; Yan, X.; Shi, H.; Chen, Y.; Huang, C.; Zhou, Y.; Yan, S.; Zhang, N.; Wang, J.; Zhang, J.; et al. The Role of Gut Microbiota and Its Metabolites in Mitigating Radiation Damage. Microorganisms 2025, 13, 2151. https://doi.org/10.3390/microorganisms13092151

AMA Style

Zhu H, Yan X, Shi H, Chen Y, Huang C, Zhou Y, Yan S, Zhang N, Wang J, Zhang J, et al. The Role of Gut Microbiota and Its Metabolites in Mitigating Radiation Damage. Microorganisms. 2025; 13(9):2151. https://doi.org/10.3390/microorganisms13092151

Chicago/Turabian Style

Zhu, Hansheng, Xin Yan, Hao Shi, Yiping Chen, Changyi Huang, Yue Zhou, Shiying Yan, Nan Zhang, Jia Wang, Jian Zhang, and et al. 2025. "The Role of Gut Microbiota and Its Metabolites in Mitigating Radiation Damage" Microorganisms 13, no. 9: 2151. https://doi.org/10.3390/microorganisms13092151

APA Style

Zhu, H., Yan, X., Shi, H., Chen, Y., Huang, C., Zhou, Y., Yan, S., Zhang, N., Wang, J., Zhang, J., Han, C., Chen, Q., Zhao, J., & Cao, M. (2025). The Role of Gut Microbiota and Its Metabolites in Mitigating Radiation Damage. Microorganisms, 13(9), 2151. https://doi.org/10.3390/microorganisms13092151

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