Targeting gut microbiota and metabolites in cancer radiotherapy.

  • Abstract
  • References
  • Similar Papers
Abstract
Translate article icon Translate Article Star icon
Take notes icon Take Notes

Radiotherapy (RT) is a cornerstone in cancer treatment, but often causes radiation-induced injury. Accumulating evidence points to the gut microbiota in modulating immune functions and maintaining intestinal integrity to impact RT efficacy. This review examines the current understanding of intestinal flora and their metabolites within the context of RT. We outlined the current research applications in how microbiota-targeted strategies such as probiotics, prebiotics, dietary interventions, and faecal microbiota transplantation could restore microbial balance, reduce toxicity, and improve patient prognosis. Microbial byproducts such as short-chain fatty acids, bile acids and tryptophan exhibit protective effects against radiation damage, supporting immune modulation and enhancing tumour radiosensitivity. These microbial products underscore the potential of gut microbiota-targeted therapies as adjunctive treatments in RT, with implications for reducing toxicity and personalizing cancer care. All these strategies targeting gut microbiota and metabolites potentially aim to develop innovative therapies that boost RT effectiveness while minimizing side effects, and finally revolutionizing personalized cancer treatment. KEY POINTS: RT alters gut microbiota composition and contributes to intestinal injury and systemic toxicity. Gut microbiota regulate mucosal integrity, immune responses and therapeutic outcomes of RT. Microbial metabolites, including SCFAs, BAs and tryptophan derivatives, protect against radiation injury and enhance tumour radiosensitivity. Microbiota-targeted interventions (e.g. probiotics, prebiotics, dietary strategies, FMT) show promise for reducing RT-related toxicity and improving patient prognosis.

ReferencesShowing 10 of 181 papers
  • Open Access Icon
  • PDF Download Icon
  • Cite Count Icon 21
  • 10.3389/fmicb.2020.01450
Gut Microbiota Metabolite Fights Against Dietary Polysorbate 80-Aggravated Radiation Enteritis.
  • Jun 26, 2020
  • Frontiers in Microbiology
  • Yuan Li + 9 more

  • 10.1136/gutjnl-2023-iddf.98
IDDF2023-ABS-0240 High-fat diet promotes radiation resistance in rectal cancer through inducing gut dysbiosis and increasing bile acid metabolites
  • Jun 1, 2023
  • Gut
  • Lihong Liu + 2 more

  • Cite Count Icon 10
  • 10.1007/s10388-023-01004-0
Gut microbiome can predict chemoradiotherapy efficacy in patients with esophageal squamous cell carcinoma.
  • Apr 22, 2023
  • Esophagus
  • Takuma Sasaki + 13 more

  • Cite Count Icon 47
  • 10.1002/adhm.202201671
Bioinspired and Inflammation-Modulatory Glycopeptide Hydrogels for Radiation-Induced Chronic Skin Injury Repair.
  • Oct 13, 2022
  • Advanced Healthcare Materials
  • Zujian Feng + 11 more

  • Open Access Icon
  • PDF Download Icon
  • Cite Count Icon 40
  • 10.1186/s12915-020-00836-x
Association of Bacteroides acidifaciens relative abundance with high-fibre diet-associated radiosensitisation
  • Aug 19, 2020
  • BMC Biology
  • Chee Kin Then + 4 more

  • Open Access Icon
  • Cite Count Icon 1893
  • 10.1016/j.ccell.2023.02.016
The evolving tumor microenvironment: From cancer initiation to metastatic outgrowth.
  • Mar 1, 2023
  • Cancer Cell
  • Karin E De Visser + 1 more

  • Open Access Icon
  • Cite Count Icon 73
  • 10.1016/j.ccell.2022.11.013
Gut microbiota-mediated nucleotide synthesis attenuates the response to neoadjuvant chemoradiotherapy in rectal cancer.
  • Jan 1, 2023
  • Cancer Cell
  • Huajing Teng + 19 more

  • Open Access Icon
  • Cite Count Icon 842
  • 10.1080/19490976.2020.1848158
Gut Bacteroides species in health and disease
  • Jan 1, 2021
  • Gut Microbes
  • Hassan Zafar + 1 more

  • Cite Count Icon 9
  • 10.1016/j.ijrobp.2024.03.003
Probiotic Consortia Protect the Intestine Against Radiation Injury by Improving Intestinal Epithelial Homeostasis
  • Mar 12, 2024
  • International Journal of Radiation Oncology, Biology, Physics
  • Li-Wei Xie + 8 more

  • Cite Count Icon 12
  • 10.1016/j.ijrobp.2022.07.032
Gut Microbiome Is Associated With the Response to Chemoradiotherapy in Patients With Non-small Cell Lung Cancer
  • Jul 26, 2022
  • International Journal of Radiation Oncology*Biology*Physics
  • Bo Qiu + 13 more

Similar Papers
  • Research Article
  • Cite Count Icon 6
  • 10.4103/1673-5374.335149
Can we promote neural regeneration through microbiota-targeted strategies? Introducing the new concept of neurobiotics.
  • Jan 1, 2022
  • Neural Regeneration Research
  • Stefano Geuna + 2 more

Can we promote neural regeneration through microbiota-targeted strategies? Introducing the new concept of neurobiotics.

  • Research Article
  • 10.1007/s00210-025-04069-z
The current findings on the gut-liver axis and the molecular basis of NAFLD/NASH associated with gut microbiome dysbiosis.
  • Apr 9, 2025
  • Naunyn-Schmiedeberg's archives of pharmacology
  • Seema Sharma + 2 more

Recent research has highlighted the complex relationship between gut microbiota, metabolic pathways, and nonalcoholic fatty liver disease (NAFLD) progression. Gut dysbiosis, commonly observed in NAFLD patients, impairs intestinal permeability, leading to the translocation of bacterial products like lipopolysaccharides, short-chain fatty acids, and ethanol to the liver. These microbiome-associated mechanisms contribute to intestinal and hepatic inflammation, potentially advancing NAFLD to NASH. Dietary habits, particularly those rich in saturated fats and fructose, can modify the microbiome composition, leading to dysbiosis and fatty liver development. Metabolomic approaches have identified unique profiles in NASH patients, with specific metabolites like ethanol linked to disease progression. While bariatric surgery has shown promise in preventing NAFLD progression, the role of gut microbiome and metabolites in this improvement remains to be proven. Understanding these microbiome-related pathways may provide new diagnostic and therapeutic targets for NAFLD and NASH. A comprehensive review of current literature was conducted using multiple medical research databases, including PubMed, Scopus, Web of Science, Embase, Cochrane Library, ClinicalTrials.gov, ScienceDirect, Medline, ProQuest, and Google Scholar. The review focused on studies that examine the relationship between gut microbiota composition, metabolic pathways, and NAFLD progression. Key areas of interest included microbial dysbiosis, endotoxin production, and the influence of diet on gut microbiota. The analysis revealed that gut dysbiosis contributes to NAFLD through several mechanisms, diet significantly influences gut microbiota composition, which in turn affects liver function through the gut-liver axis. High-fat diets can lead to dysbiosis, altering microbial metabolic activities and promoting liver inflammation. Specifically, gut microbiota-mediated generation of saturated fatty acids, such as palmitic acid, can activate liver macrophages and increase TNF-α expression, contributing to NASH development. Different dietary components, including cholesterol, fiber, fat, and carbohydrates, can modulate the gut microbiome and influence NAFLD progression. This gut-liver axis plays a crucial role in maintaining immune homeostasis, with the liver responding to gut-derived bacteria by activating innate and adaptive immune responses. Microbial metabolites, such as bile acids, tryptophan catabolites, and branched-chain amino acids, regulate adipose tissue and intestinal homeostasis, contributing to NASH pathogenesis. Additionally, the microbiome of NASH patients shows an elevated capacity for alcohol production, suggesting similarities between alcoholic steatohepatitis and NASH. These findings indicate that targeting the gut microbiota may be a promising approach for NASH treatment and prevention. Recent research highlights the potential of targeting gut microbiota for managing nonalcoholic fatty liver disease (NAFLD). The gut-liver axis plays a crucial role in NAFLD pathophysiology, with dysbiosis contributing to disease progression. Various therapeutic approaches aimed at modulating gut microbiota have shown promise, including probiotics, prebiotics, synbiotics, fecal microbiota transplantation, and dietary interventions. Probiotics have demonstrated efficacy in human randomized controlled trials, while other interventions require further investigation in clinical settings. These microbiota-targeted therapies may improve NAFLD outcomes through multiple mechanisms, such as reducing inflammation and enhancing metabolic function. Although lifestyle modifications remain the primary recommendation for NAFLD management, microbiota-focused interventions offer a promising alternative for patients struggling to achieve weight loss targets.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 20
  • 10.3390/metabo14010074
Gut Microbiota-Gut Metabolites and Clostridioides difficile Infection: Approaching Sustainable Solutions for Therapy.
  • Jan 22, 2024
  • Metabolites
  • Bijay Gurung + 2 more

Clostridioides difficile (C. difficile) infection (CDI) is the most common hospital-acquired infection. With the combination of a high rate of antibiotic resistance and recurrence, it has proven to be a debilitating public health threat. Current treatments for CDI include antibiotics and fecal microbiota transplantation, which contribute to recurrent CDIs and potential risks. Therefore, there is an ongoing need to develop new preventative treatment strategies for CDI. Notably, gut microbiota dysbiosis is the primary risk factor for CDI and provides a promising target for developing novel CDI therapy approaches. Along with gut microbiota dysbiosis, a reduction in important gut metabolites like secondary bile acids and short-chain fatty acids (SCFAs) were also seen in patients suffering from CDI. In this review study, we investigated the roles and mechanisms of gut microbiota and gut microbiota-derived gut metabolites, especially secondary bile acids and SCFAs in CDI pathogenesis. Moreover, specific signatures of gut microbiota and gut metabolites, as well as different factors that can modulate the gut microbiota, were also discussed, indicating that gut microbiota modulators like probiotics and prebiotics can be a potential therapeutic strategy for CDI as they can help restore gut microbiota and produce gut metabolites necessary for a healthy gut. The understanding of the associations between gut microbiota-gut metabolites and CDI will allow for developing precise and sustainable approaches, distinct from antibiotics and fecal transplant, for mitigating CDI and other gut microbiota dysbiosis-related diseases.

  • Research Article
  • Cite Count Icon 82
  • 10.1038/s12276-021-00627-6
Gut microbiota restoration through fecal microbiota transplantation: a new atopic dermatitis therapy
  • May 1, 2021
  • Experimental & Molecular Medicine
  • Jong-Hwa Kim + 2 more

The pathogenesis of atopic dermatitis (AD) involves complex factors, including gut microbiota and immune modulation, which remain poorly understood. The aim of this study was to restore gut microbiota via fecal microbiota transplantation (FMT) to ameliorate AD in mice. FMT was performed using stool from donor mice. The gut microbiota was characterized via 16S rRNA sequencing and analyzed using Quantitative Insights into Microbial Ecology 2 with the DADA2 plugin. Gut metabolite levels were determined by measuring fecal short-chain fatty acid (SCFA) contents. AD-induced allergic responses were evaluated by analyzing blood parameters (IgE levels and eosinophil percentage, eosinophil count, basophil percentage, and monocyte percentage), the levels of Th1 and Th2 cytokines, dermatitis score, and the number of mast cells in the ileum and skin tissues. Calprotectin level was measured to assess gut inflammation after FMT. FMT resulted in the restoration of gut microbiota to the donor state and increases in the levels of SCFAs as gut metabolites. In addition, FMT restored the Th1/Th2 balance, modulated Tregs through gut microbiota, and reduced IgE levels and the numbers of mast cells, eosinophils, and basophils. FMT is associated with restoration of gut microbiota and immunologic balance (Th1/Th2) along with suppression of AD-induced allergic responses and is thus a potential new therapy for AD.

  • Research Article
  • 10.3389/fmicb.2025.1630823
The role of gut microbiota in diarrhea and its alleviation through microbiota-targeted interventions
  • Oct 28, 2025
  • Frontiers in Microbiology
  • Rui Tian + 11 more

Diarrhea is a common gastrointestinal disease and closely related to the balance of the gut microbiota (GM). In turn, dysregulation of the GM can affect the onset and progression of diarrhea through regulating the metabolism, intestinal immune function, intestinal barrier function and changes in the brain-gut axis of host. Although increasing evidence suggests that GM is associated with gastrointestinal homeostasis and disease, the underlying mechanisms are not fully understood. GM disorder was often accompanied by diarrhea patients and animals, and the diarrhea caused by GM imbalance mainly involved the effects on short chain fatty acids (SCFAs), bile acids (BAs), intestinal barrier, immune system, and brain-gut microbiota axis (BGMA). In addition, intervening in the GM (probiotics, fecal microbiota transplantation and bacteriophage therapy) has been shown to be an effective way to alleviate diarrhea. In this review, the mechanism of diarrhea occurrence, probiotics, fecal microbiota transplantation and bacteriophage therapy intervene in diarrhea by regulating GM from basic and clinical research were summarized and discussed. We aim to provide the latest reference for studying the mechanism of treating diarrhea from the perspective of GM, and provide data support for clinical treatment of diarrhea.

  • Research Article
  • Cite Count Icon 6
  • 10.1093/ecco-jcc/jjad212.0305
P175 HucMSC-Exo ameliorate experimental colitis via modulating gut microbiota and metabolites
  • Jan 24, 2024
  • Journal of Crohn's and Colitis
  • C Wang + 8 more

Background Gut microbiota and metabolites have been shown to influence the development of inflammatory bowel disease (IBD). Exosomes derived from human umbilical cord mesenchymal stem cells (HucMSC-Exo) have been identified as a promising biological therapy for the treatment of IBD. Previous studies have described that HucMSC-Exo could modulate gut microbiota. However, the mechanism remains unclear. Here, we explored the role of gut microbiota and metabolites in HucMSC-Exo-mediated amelioration of experimental colitis. Methods Dextran sulfate sodium (DSS) was used to induce colitis. The role of HucMSC-Exo were assessed in colitis mice. Fecal microbiota transplantation (FMT) and sterile fecal filtrate gavage were employed to evaluate the effect of gut microbiota and metabolites. Gut microbiota and metabolites were analyzed through 16S rRNA sequencing and metabolomic profiling. The proportion of CD4+ T cells were phenotyped by multicolour flow cytometry. Results HucMSC-Exo treatment alleviated colon inflammation by regulating flora composition, significantly up-regulated the levels of bacteria such as Bacteroides, Parabacteroides distasonis, and Tannerellaceae. Meanwhile, HucMSC-Exo transformed metabolite short-chain fatty acid (SCFA) profiles, particularly increased butyrate level. Additionally, HucMSC-Exo selectively up-regulated the frequencies of regulatory T (Treg) cells as well as down-regulated the ratio of T helper type 17 (Th17) cells in colonic lamina propria to maintain intestinal immune homeostasis and repaired the colonic mucus barrier. FMT and sterile fecal filtrate gavage were conducted to confirm the above mechanism. Microbiota from HucMSC-Exo-treated mice alleviated the colitis over microbiota from DSS-treated mice. Sterile fecal filtrate from HucMSC-Exo-treated mice and butyrate induced similar beneficial outcomes, such as improved the Th17/Treg balance and repaired the colonic mucus barrier. Collectively, HucMSC-Exo ameliorate colitis by directly modulating gut microbiota and metabolites. Conclusion HucMSC-Exo ameliorated experimental colitis via directly modulating gut microbiota and metabolite butyrate, which could regulate CD4+T cells homeostasis and repair colonic mucus barrier. The findings demonstrated the HucMSC-Exo as a potential gut microbiota and metabolites modulator to treat IBD.

  • Research Article
  • Cite Count Icon 14
  • 10.1097/mco.0000000000001028
Microbiome and its impact on fetal and neonatal brain development: current opinion in pediatrics.
  • Mar 12, 2024
  • Current opinion in clinical nutrition and metabolic care
  • Nina M Frerichs + 2 more

Emerging evidence suggests that the gut microbiota and its metabolites regulate neurodevelopment and cognitive functioning via a bi-directional communication system known as the microbiota-gut-brain axis (MGBA). The MGBA influences brain development and function via the hypothalamic-pituitary axis, the vagal nerve, immune signaling, bacterial production of neurotransmitters, and microbial metabolites like short-chain fatty acids, tryptophan derivatives, and bile acids. Animal studies show fetal neurodevelopment is mediated by maternal microbiota derivatives, immune activation, and diet. Furthermore, manipulation of the microbiota during critical windows of development, like antibiotic exposure and fecal microbiota transplantation, can affect cognitive functioning and behavior in mice. Evidence from human studies, particularly in preterm infants, also suggests that a disrupted gut microbiota colonization may negatively affect neurodevelopment. Early microbial signatures were linked to favorable and adverse neurodevelopmental outcomes. The link between the gut microbiota and the brain is evident. Future studies, including experimental studies, larger participant cohort studies with longitudinal analyses of microbes, their metabolites, and neurotransmitters, and randomized controlled trials are warranted to further elucidate the mechanisms of the MGBA. Identification of early, predictive microbial markers could pave the way for the development of novel early microbiota-based intervention strategies, such as targeted probiotics, and vaginal or fecal microbiota transplantation, aimed at improving infant neurodevelopment.

  • Supplementary Content
  • Cite Count Icon 24
  • 10.3389/fendo.2022.994930
Roles of gut microbiota and metabolites in overweight and obesity of children
  • Sep 8, 2022
  • Frontiers in Endocrinology
  • Shengan Zhang + 1 more

The prevalence of overweight and obesity in children and adolescents is an increasing public health problem. Pediatric overweight and obesity result from multiple factors, including genetic background, diet, and lifestyle. In addition, the gut microbiota and their metabolites play crucial roles in the progression of overweight and obesity of children. Therefore, we reviewed the roles of gut microbiota in overweight/obese children. The relationship between pediatric overweight/obesity and gut metabolites, such as short-chain fatty acids, medium-chain fatty acids, amino acids, amines, and bile acids, are also summarized. Targeting gut microbiota and metabolites might be a promising strategy for interventions aimed at reducing pediatric overweight/obesity.

  • PDF Download Icon
  • Research Article
  • Cite Count Icon 65
  • 10.3390/microorganisms7090338
Microbiota, Microbial Metabolites, and Barrier Function in A Patient with Anorexia Nervosa after Fecal Microbiota Transplantation.
  • Sep 10, 2019
  • Microorganisms
  • Petra Prochazkova + 13 more

The change in the gut microbiome and microbial metabolites in a patient suffering from severe and enduring anorexia nervosa (AN) and diagnosed with small intestinal bacterial overgrowth syndrome (SIBO) was investigated. Microbial gut dysbiosis is associated with both AN and SIBO, and therefore gut microbiome changes by serial fecal microbiota transplantation (FMT) is a possible therapeutic modality. This study assessed the effects of FMT on gut barrier function, microbiota composition, and the levels of bacterial metabolic products. The patient treatment with FMT led to the improvement of gut barrier function, which was altered prior to FMT. Very low bacterial alpha diversity, a lack of beneficial bacteria, together with a great abundance of fungal species were observed in the patient stool sample before FMT. After FMT, both bacterial species richness and gut microbiome evenness increased in the patient, while the fungal alpha diversity decreased. The total short-chain fatty acids (SCFAs) levels (molecules presenting an important source of energy for epithelial gut cells) gradually increased after FMT. Contrarily, one of the most abundant intestinal neurotransmitters, serotonin, tended to decrease throughout the observation period. Overall, gut microbial dysbiosis improvement after FMT was considered. However, there were no signs of patient clinical improvement. The need for an in-depth analysis of the donor´s stool and correct selection pre-FMT is evident.

  • Front Matter
  • Cite Count Icon 32
  • 10.1053/j.gastro.2015.05.030
Fecal Microbiota Transplantation for Ulcerative Colitis: Not Just Yet
  • May 26, 2015
  • Gastroenterology
  • Ari M Grinspan + 1 more

Fecal Microbiota Transplantation for Ulcerative Colitis: Not Just Yet

  • Research Article
  • Cite Count Icon 1
  • 10.3390/biomedicines13020526
Microbiota and Radiotherapy: Unlocking the Potential for Improved Gastrointestinal Cancer Treatment.
  • Feb 19, 2025
  • Biomedicines
  • Damir Vučinić + 3 more

Radiotherapy (RT) is one of the major cornerstones in managing gastrointestinal (GI) cancers. However, several side effects, such as intestinal inflammation, mucosal injury, and dysbiosis, often compromise this. The gut microbiota increasingly attracts much interest as an essential modulator of RT effects influencing immune responses and tissue repair. Through short-chain fatty acids such as butyrate, representatives of certain bacterial species play a crucial role under normal conditions, keeping the mucosal integrity intact and reducing oxidative stress-mediated damage. Dysbiosis, a state where diminished microbial diversity and increased pathogenic species in the microbiota are seen, amplifies RT-induced toxicity in patients. Clinical investigations highlight that microbiota-targeted interventions, including probiotics, prebiotics, and fecal microbiota transplantation, hold the means to augment RT efficacy and lessen toxicity. Increased microflora diversity and specific microbial profiles have yielded serious patient improvements. Advanced RT methods use stereotactic body radiotherapy combined with microbiota modulation as a promising technique to shield healthy tissue and maximize immune-mediated antitumor effects. Additionally, there is an implication in tumor behavior regulated by the intratumoral microbiota regarding the response to radiotherapy. Notably, the modulation of gut and tumor microbiota provides an avenue to optimize RT benefits in GI cancers, underscoring the importance of personalized therapy.

  • Research Article
  • Cite Count Icon 2
  • 10.1128/msystems.00665-24
The role of gut microbiome and its metabolites in pancreatitis.
  • Oct 22, 2024
  • mSystems
  • Letian Pan + 4 more

Gut microbiome plays a vital role in the intestinal ecosystem and has close association with metabolites. Due to the development of metabolomics and microbiomics, recent studies have observed that alteration of either the gut microbiome or metabolites may have effects on the progression of pancreatitis. Several new treatments based on the gut microbiome or metabolites have been studied extensively in recent years. Gut microbes, such as Bifidobacterium, Akkermansia, and Lactobacillus, and metabolites, such as short-chain fatty acids, bile acids, vitamin, hydrogen sulfide, and alcohol, have different effects on pancreatitis. Some preliminary studies about new intervention measures were based on the gut microbiome and metabolites such as diet, prebiotic, herbal medicine, and fecal microbiota transplantation. This review aims to summarize the recent advances about the gut microbiome, metabolites, and pancreatitis in order to determine the potential beneficial role of the gut microbiome and metabolites in pancreatitis.

  • Supplementary Content
  • 10.3389/fimmu.2025.1617459
The gut microbiota in osteoporosis: dual roles and therapeutic prospects
  • Sep 1, 2025
  • Frontiers in Immunology
  • Xingwen Xie + 4 more

Recent advances in bone biology have underscored the essential role of the gut microbiota in maintaining skeletal homeostasis. Gut-derived metabolites, particularly short chain fatty acids and tryptophan derivatives, influence bone metabolism through modulation of immune signaling, inflammation, and endocrine networks. Emerging evidence indicates that these effects are context dependent and dose dependent, rather than uniformly beneficial or detrimental. For instance, butyrate and lipopolysaccharide exhibit biphasic effects on both osteogenesis and osteoclastogenesis, contingent on concentration, immune status, and the local microenvironment. Microbiota-targeted strategies such as probiotics, prebiotics, and fecal microbiota transplantation are under active investigation as innovative interventions for osteoporosis in both preclinical and clinical contexts. However, substantial knowledge gaps persist, including inconsistent therapeutic outcomes, limited mechanistic insight into host–microbiota interactions, and the absence of standardized microbial intervention protocols. In addition, safety concerns related to FMT, particularly in immunocompromised elderly populations, emphasize the need for rigorous donor screening, extended follow-up periods, and personalized risk and benefit assessment models. To advance the field, future studies should incorporate multi-omics platforms and precision medicine tools to identify key microbial targets and enhance therapeutic efficacy. This review consolidates current evidence and proposes a conceptual framework to clarify the context-specific roles of the gut microbiota in bone remodeling. A deeper mechanistic understanding will be crucial for translating microbiota-based strategies into safe and effective treatments for metabolic bone disorders.

  • Abstract
  • Cite Count Icon 4
  • 10.1016/j.ijrobp.2021.07.812
Gut Bacteria Erysipelatoclostridium and Its Related Metabolite Ptilosteroid A Could Predict Grade 2 Radiation-Induced Intestinal Injury During Pelvic Radiotherapy
  • Oct 22, 2021
  • International Journal of Radiation Oncology*Biology*Physics
  • S Cai + 5 more

Gut Bacteria Erysipelatoclostridium and Its Related Metabolite Ptilosteroid A Could Predict Grade 2 Radiation-Induced Intestinal Injury During Pelvic Radiotherapy

  • Research Article
  • Cite Count Icon 2
  • 10.3390/sclerosis3010008
Microbial Influences on Amyotrophic Lateral Sclerosis: The Gut–Brain Axis and Therapeutic Potential of Microbiota Modulation
  • Mar 5, 2025
  • Sclerosis
  • Victòria Ayala + 8 more

Background/Objectives: Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by the progressive degeneration of motor neurons. The gut microbiota, a community of microorganisms in the digestive tract, has recently been implicated in ALS pathogenesis through its influence on neuroinflammation and metabolic pathways. This review explores the potential role of digestive microbiota and its metabolites in ALS progression and investigates therapeutic approaches targeting gut microbiota. Methods: A comprehensive review of the current literature was conducted to assess the relationship between gut microbiota composition, microbial metabolites, and ALS progression in patients. We searched for published reports on microbiota composition, microbial metabolites, and ALS, emphasizing the complex interplay between dysbiosis, neuroinflammation, and systemic metabolism. Special emphasis was placed on studies exploring short-chain fatty acids (SCFAs), bacterial amyloids (curli-like factors), and neurotoxins such as β-methylamino-L-alanine (BMAA). The role of the liver–gut axis was evaluated as well. The potential changes in microbiota would sustain the rationale for therapeutic strategies such as probiotics, prebiotics, fecal microbiota transplantation (FMT), and dietary interventions. Results: ALS patients exhibit gut dysbiosis, characterized by reduced SCFA-producing bacteria and an increase in potentially pathogenic genera. Of note, different studies do not agree on common patterns of microbiota being linked to ALS, supporting the need for further, more extensive studies. Dysbiosis sometimes correlates with systemic inflammation and disrupted liver function, amplifying neuroinflammatory responses. Key microbial metabolites, including SCFAs, bacterial amyloids, and BMAA, may exacerbate motor neuron degeneration by promoting protein misfolding, oxidative stress, and neuroinflammation. Emerging therapeutic strategies, including probiotics and FMT, show potential in restoring microbial balance, although clinical data in ALS patients remain limited. Conclusions: The gut microbiota could modulate neuroinflammation and systemic metabolism in ALS. Microbiota-targeted therapies, such as probiotics and dietary interventions, represent promising avenues for mitigating disease progression. Further research is required to validate these interventions through large-scale, longitudinal studies and to develop personalized microbiota-based treatments tailored to individual ALS phenotypes.

More from: Clinical and translational medicine
  • Research Article
  • 10.1002/ctm2.70513
Altered nicotinamide adenine dinucleotide metabolism drives cartilage degeneration and osteoarthritis.
  • Nov 1, 2025
  • Clinical and translational medicine
  • Xiaoxin Wu + 12 more

  • Discussion
  • 10.1002/ctm2.70519
DNA damage response pathway is a promising therapeutic target for extrachromosomal DNA‐positive tumours
  • Nov 1, 2025
  • Clinical and Translational Medicine
  • Congcong Tian + 1 more

  • Journal Issue
  • 10.1002/ctm2.v15.11
  • Nov 1, 2025
  • Clinical and Translational Medicine

  • Research Article
  • 10.1002/ctm2.70524
Progression significance for low-level aberrant B-lymphoblasts in chronic myeloid leukaemia patients.
  • Nov 1, 2025
  • Clinical and translational medicine
  • Haoren Wang + 12 more

  • Research Article
  • 10.1002/ctm2.70523
CLINICAL AND TRANSLATIONAL MEDICINE
  • Nov 1, 2025
  • Clinical and Translational Medicine

  • Discussion
  • 10.1002/ctm2.70515
Making the impossible possible: Baby KJ and the road map to personalized gene-editing care.
  • Nov 1, 2025
  • Clinical and translational medicine
  • Vanessa Almendro + 1 more

  • Supplementary Content
  • 10.1002/ctm2.70499
Exploring the mechanisms of mutual influence between lactylation and macrophage polarization in the context of disease
  • Nov 1, 2025
  • Clinical and Translational Medicine
  • Houhua Guo + 5 more

  • Supplementary Content
  • 10.1002/ctm2.70512
Clinical application of single‐cell RNA sequencing in disease and therapy
  • Oct 31, 2025
  • Clinical and Translational Medicine
  • Aisha Shigna Nadukkandy + 3 more

  • Research Article
  • 10.1002/ctm2.70514
Advances in gene therapy for Lafora disease: Intravenous recombinant adeno‐associated virus‐mediated delivery of EPM2A and EPM2B genes
  • Oct 30, 2025
  • Clinical and Translational Medicine
  • Luis Zafra‐Puerta + 8 more

  • Research Article
  • 10.1002/ctm2.70510
Liraglutide upregulates the Cftr gene and regulates the mucus transcriptome profile in Brunner's glands in mice
  • Oct 27, 2025
  • Clinical and Translational Medicine
  • Louise Marie Voetmann + 10 more

Save Icon
Up Arrow
Open/Close
  • Ask R Discovery Star icon
  • Chat PDF Star icon

AI summaries and top papers from 250M+ research sources.

Search IconWhat is the difference between bacteria and viruses?
Open In New Tab Icon
Search IconWhat is the function of the immune system?
Open In New Tab Icon
Search IconCan diabetes be passed down from one generation to the next?
Open In New Tab Icon