1
|
Wu Q, Li Q, Zhang R, Cui Y, Zou J, Zhang J, Ma C, Han D, Peng Y. Research progress of radiation esophagitis: A narrative review. Medicine (Baltimore) 2025; 104:e42273. [PMID: 40355243 PMCID: PMC12074111 DOI: 10.1097/md.0000000000042273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 04/11/2025] [Indexed: 05/14/2025] Open
Abstract
Radiation esophagitis (RE) is an adverse reaction to radiotherapy for thoracic malignancies and a major factor limiting the dosage for thoracic tumors. In cases of RE, mild conditions may involve only discomfort in the throat, severe cases can present with intense pain and significant difficulty swallowing, and may lead to interruptions in radiation therapy, affecting the treatment outcome. However, much of the current research focuses on how to diagnose and treat the disease and needs to address the prevention of RE. In this review, several online databases, including PubMed, Web of Science, Google Scholar, the China National Knowledge Infrastructure, and the Chinese Medical Journal Database, were searched to collect relevant published studies. This review provides updated information on the pathogenesis, diagnosis, risk factors, development of predictive models, prevention, and treatment of RE, aiming to serve as a reference for its diagnosis, treatment, and prevention.
Collapse
Affiliation(s)
- Qiang Wu
- Shandong Second Medical University, Weifang, Shandong Province, China
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Qiuxia Li
- Department of Journal Center, Shandong Provincal Hospital Affiliated to Shandong First Medical University, Jinan, Shandong Province, China
| | - Ruidan Zhang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Yongbin Cui
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Jingmin Zou
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Jianbo Zhang
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Changsheng Ma
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Dali Han
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Yiru Peng
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| |
Collapse
|
2
|
Abdeen SK, Mastandrea I, Stinchcombe N, Puschhof J, Elinav E. Diet-microbiome interactions in cancer. Cancer Cell 2025; 43:680-707. [PMID: 40185096 DOI: 10.1016/j.ccell.2025.03.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2025] [Revised: 02/26/2025] [Accepted: 03/11/2025] [Indexed: 04/07/2025]
Abstract
Diet impacts cancer in diverse manners. Multiple nutritional effects on tumors are mediated by dietary modulation of commensals, residing in mucosal surfaces and possibly also within the tumor microenvironment. Mechanistically understanding such diet-microbiome-host interactions may enable to develop precision nutritional interventions impacting cancer development, dissemination, and treatment responses. However, data-driven nutritional strategies integrating diet-microbiome interactions are infrequently incorporated into cancer prevention and treatment schemes. Herein, we discuss how dietary composition affects cancer-related processes through alterations exerted by specific nutrients and complex foods on the microbiome. We highlight how dietary timing, including time-restricted feeding, impacts microbial function in modulating cancer and its therapy. We review existing and experimental nutritional approaches aimed at enhancing microbiome-mediated cancer treatment responsiveness while minimizing adverse effects, and address challenges and prospects in integrating diet-microbiome interactions into precision oncology. Collectively, mechanistically understanding diet-microbiome-host interactomes may enable to achieve a personalized and microbiome-informed optimization of nutritional cancer interventions.
Collapse
Affiliation(s)
- Suhaib K Abdeen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Nina Stinchcombe
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany; Junior Research Group Epithelium Microbiome Interactions, DKFZ, Heidelberg, Germany
| | - Jens Puschhof
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, Heidelberg, Germany; Junior Research Group Epithelium Microbiome Interactions, DKFZ, Heidelberg, Germany.
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany.
| |
Collapse
|
3
|
Ohsawa M, Nishi H, Emi M, Yoshikawa T, Hamai Y, Ibuki Y, Kurokawa T, Hirohata R, Kitasaki N, Kawada-Matsuo M, Komatsuzawa H, Kawaguchi H, Okada M. Impact of Fusobacterium nucleatum in the treatment of cancer, including radiotherapy and its future potential in esophageal cancer. JOURNAL OF RADIATION RESEARCH 2024; 65:i126-i134. [PMID: 39679879 DOI: 10.1093/jrr/rrae061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 05/09/2024] [Indexed: 12/17/2024]
Abstract
Despite advances in multimodality therapy, including surgery, chemotherapy, radiation therapy and chemoradiation, the fatality rate for esophageal cancer remains high. Specifically, Fusobacterium nucleatum, due to its aggregation capacity, has shown a tendency to form biofilms. The biofilm-forming capabilities of microbial communities are of utmost importance in the context of cancer treatment, as they have been shown to drive significant losses in the efficaciousness of various cancer treatments. Therefore, elucidating the dynamics of F. nucleatum will be important for the development of effective treatments for esophageal cancer. Therefore, this review summarizes the current knowledge of F. nucleatum, its involvement in cancer and its impact on chemotherapy and radiation therapy. In conclusion, further research on the role of F. nucleatum is essential for the continued advancement of the treatment of esophageal cancer and patient care.
Collapse
Affiliation(s)
- Manato Ohsawa
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Hiromi Nishi
- Department of General Dentistry, Hiroshima University Hospital, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Manabu Emi
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Toru Yoshikawa
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Yoichi Hamai
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Yuta Ibuki
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Tomoaki Kurokawa
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Ryosuke Hirohata
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Nao Kitasaki
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Miki Kawada-Matsuo
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Hitoshi Komatsuzawa
- Department of Bacteriology, Hiroshima University Graduate School of Biomedical and Health Sciences, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Hiroyuki Kawaguchi
- Department of General Dentistry, Hiroshima University Hospital, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| | - Morihito Okada
- Department of Surgical Oncology, Hiroshima University, 1-2-3-Kasumi, Minami-ku, Hiroshima City, Hiroshima 734-0037, Japan
| |
Collapse
|
4
|
Wang J, Qiu D, Dong X, Liu Y, Chen J. Efficacy and safety of QingReJieDu therapy in preventing acute radiation esophagitis: A systematic review and meta-analysis. Medicine (Baltimore) 2024; 103:e40779. [PMID: 39654193 PMCID: PMC11630928 DOI: 10.1097/md.0000000000040779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 11/11/2024] [Accepted: 11/14/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Acute radiation esophagitis (ARE) is a frequently observed complication of radiotherapy for malignant tumors. QingReJieDu (QRJD) therapy is commonly used for the prevention and treatment of ARE. There is a lack of evidence regarding the effectiveness and safety of traditional Chinese medicine in preventing ARE. Therefore, the objective of this study was to comprehensively evaluate the efficacy and safety of QRJD therapy in preventing ARE. METHODS Systematic searches were conducted across 8 databases from their inception to December 30, 2023. This study included randomized controlled trials that assessed the effectiveness of QRJD therapy in preventing ARE. The methodological quality of the included studies was evaluated using the Cochrane collaboration tool. Data analysis was conducted using RevMan 5.3 and Stata 15. RESULTS Eleven randomized controlled studies involving 754 participants were included in this analysis. The results demonstrated that QRJD therapy effectively prevented the occurrence of ARE (relative risk = 0.84, 95% CI: 0.80-0.89, P < .001), as well as the development of grade ≥ 3 ARE (relative risk = 0.41, 95% CI: 0.28-0.59, P < .001). Furthermore, QRJD therapy delayed the time of the first episode of ARE (weighted mean difference = 5.67, 95% CI: 5.33-6.02, P < .001). No significant increase in adverse events was observed with the QRJD therapy. CONCLUSION Our results indicate that QRJD therapy has a significant preventive effect on ARE in clinical practice. However, it is important to note that these findings should be confirmed through more rigorously designed, multicenter, large-sample, and large-scale randomized controlled trials.
Collapse
Affiliation(s)
- Jian Wang
- The Third Clinical Medical College of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Dan Qiu
- Department of Medical Oncology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, Zhejiang, China
| | | | - Yangxin Liu
- Changsha Medical University, Changsha, China
| | - Jianxin Chen
- Department of Medical Oncology, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, Zhejiang, China
| |
Collapse
|
5
|
Li X, Xu B, Yang H, Zhu Z. Gut Microbiota, Human Blood Metabolites, and Esophageal Cancer: A Mendelian Randomization Study. Genes (Basel) 2024; 15:729. [PMID: 38927665 PMCID: PMC11203100 DOI: 10.3390/genes15060729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/23/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Unbalances in the gut microbiota have been proposed as a possible cause of esophageal cancer (ESCA), yet the exact causal relationship remains unclear. PURPOSE To investigate the potential causal relationship between the gut microbiota and ESCA with Mendelian randomization (MR) analysis. METHODS Genome-wide association studies (GWASs) of 207 gut microbial taxa (5 phyla, 10 classes, 13 orders, 26 families, 48 genera, and 105 species) and 205 gut microbiota metabolic pathways conducted by the Dutch Microbiome Project (DMP) and a FinnGen cohort GWAS of esophageal cancer specified the summary statistics. To investigate the possibility of a mediation effect between the gut microbiota and ESCA, mediation MR analyses were performed for 1091 blood metabolites and 309 metabolite ratios. RESULTS MR analysis indicated that the relative abundance of 10 gut microbial taxa was associated with ESCA but all the 12 gut microbiota metabolic pathways with ESCA indicated no statistically significant association existing. Two blood metabolites and a metabolite ratio were discovered to be mediating factors in the pathway from gut microbiota to ESCA. CONCLUSION This research indicated the potential mediating effects of blood metabolites and offered genetic evidence in favor of a causal correlation between gut microbiota and ESCA.
Collapse
Affiliation(s)
- Xiuzhi Li
- State Key Laboratory of Oncology in South China, Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China;
| | - Bingchen Xu
- State Key Laboratory of Oncology in South China, Department of Minimally Invasive Intervention, Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China;
| | - Han Yang
- State Key Laboratory of Oncology in South China, Department of Thoracic Surgery, Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China;
| | - Zhihua Zhu
- State Key Laboratory of Oncology in South China, Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangdong Provincial Clinical Research Center for Cancer, Collaborative Innovation Center for Cancer Medicine, Guangzhou 510060, China
| |
Collapse
|
6
|
He Y, Li XY, Hu AQ, Qian D. Salivary microbiome is associated with the response to chemoradiotherapy in initially inoperable patients with esophageal squamous cell carcinoma. J Oral Microbiol 2024; 16:2359887. [PMID: 38813524 PMCID: PMC11134033 DOI: 10.1080/20002297.2024.2359887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 05/20/2024] [Indexed: 05/31/2024] Open
Abstract
Background The salivary microbiome may interact with chemoradiotherapy through dynamic changes in microbial composition and systemic immunity. We aimed to explore the association between the salivary microbiome and response to chemoradiotherapy in initially inoperable patients with local advanced esophageal squamous cell carcinoma (LAESCC). Methods Salivary and peripheral blood samples were collected before and after chemoradiotherapy. The microbiome and metabolic pathways were analyzed by 16S ribosomal RNA sequencing and liquid chromatography tandem mass spectrometry/Mass spectrometry analyses. Results The salivary microbiome exhibited characteristic variations between patients and healthy controls. A significant correlation was found between Prevotella_salivae, Saccharibacteria_TM7_G3_bacterium_HMT_351, and Veillonellaceae_G1_bacterium_HMT_129 and pathological complete response (pCR) in initially inoperable patients who underwent surgery. The PICRUSt suggested that immune diseases and cell motility were different in tumor compared to normal groups. KEGG enrichment analysis showed enriched lipid metabolism, signal transduction, and membrane transport in the tumor group. CD3+CD8 T cells, IL6, IL10, and IFNγ exhibited an increasing trend during the treatment process of chemoradiotherapy. Conclusions Our study demonstrated that variations in specific saliva taxa associated with host immunomodulatory cells and cytokines could be promising for early efficacy prediction of chemoradiotherapy in initially inoperable patients with LAESCC.
Collapse
Affiliation(s)
- Yuan He
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Xiao-Yang Li
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - An-Qi Hu
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Dong Qian
- Department of Radiation Oncology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
7
|
Zhang Z, Zhang G, Huang Z, Shi Y, Wang D. Application of Mendelian randomization to assess host gene-gut microbiota correlations in patients with esophageal cancer. Front Microbiol 2023; 14:1309596. [PMID: 38179450 PMCID: PMC10764629 DOI: 10.3389/fmicb.2023.1309596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Accepted: 12/06/2023] [Indexed: 01/06/2024] Open
Abstract
Background Increasing evidence suggests that esophageal cancer (ESCA) may be correlated with gut flora. However, their causal connection remains unclear. This study aimed to evaluate potential causal linkages and gene-gut microbiome associations between the gut microbiota and ESCA using Mendelian randomization (MR). Methods We analyzed the data using genome-wide association studies. The exposure factor and outcome variable were the gut microbiota and ESCA, respectively. The MR-Egger method, weighted median, inverse-variance weighted method, heterogeneity test, sensitivity analysis, and multiplicity analysis were used for the MR analysis. And it was validated using an external dataset. Further meta-analysis was performed to validate the robustness of this relationship. Finally, we annotated single nucleotide polymorphisms in the gut microbiota that were causally associated with ESCA to explore possible host gene-gut microbiota correlations in patients with ESCA. Results We identified four species with potential associations with ESCA. Three of these species had a negative causal relationship with ESCA (odds ratio (OR): 0.961; 95% confidence interval (CI): 0.923-0.971; p = 0.047 for Romboutsia; OR: 0.972; 95% CI: 0.921-0.961; p = 0.018 for Lachnospira; OR: 0.948; 95% CI: 0.912-0.970; p = 0.032 for Eubacterium). A positive causal relationship was observed between one bacterial group and ESCA (OR: 1.105; 95% CI: 1.010-1.072; p = 0.018 for Veillonella). External datasets show the same trend. This is further supported by meta-analysis. None of the data showed pleiotropy, and leave-one-out analysis indicated the reliability of these findings. The gut microbiomes of patients with ESCA may correlate with the 19 identified genes. Conclusion Our data indicate a potential causal link between these four gut bacteria and ESCA and identify a correlation between host genes and gut microbiota in ESCA, offering novel therapeutic options.
Collapse
Affiliation(s)
- Zhenhu Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Guodong Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Zhulan Huang
- Department of Ultrasound Medicine, Longgang District Maternity and Child Healthcare Hospital of Shenzhen City, Shenzhen, Guangdong, China
| | - Yamin Shi
- Department of Foreign Languages, Shandong University of Finance and Economics, Jinan, China
| | - Dong Wang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China
| |
Collapse
|
8
|
Ye X, Yu F, Zhou J, Zhao C, Wu J, Ni X. Analysis of the gut microbiota in children with gastroesophageal reflux disease using metagenomics and metabolomics. Front Cell Infect Microbiol 2023; 13:1267192. [PMID: 37900308 PMCID: PMC10613033 DOI: 10.3389/fcimb.2023.1267192] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 09/19/2023] [Indexed: 10/31/2023] Open
Abstract
Background There is no direct evidence of gut microbiota disturbance in children with gastroesophageal reflux disease (GERD). This study aimed to provide direct evidence and a comprehensive understanding of gut microbiota disturbance in children with GERD through combined metagenomic and metabolomic analysis. Methods 30 children with GERD and 30 healthy controls (HCs) were continuously enrolled, and the demographic and clinical characteristics of the subjects were collected. First, 16S rRNA sequencing was used to evaluate differences in the gut microbiota between children with GERD and HC group, and 10 children with GERD and 10 children in the HC group were selected for metagenomic analysis. Nontargeted metabolomic analysis was performed using liquid chromatography/mass spectrometry (LC/MS), and metagenomic and metabolomic data were analyzed together. Results There were significant differences in the gut microbiota diversity and composition between children with GERD and HCs. The dominant bacteria in children with GERD were Proteobacteria and Bacteroidota. At the species level, the top three core bacterial groups were Bacteroides stercoris, Bacteroides vulgatus and Alistipes putredinis. The main differential pathways were identified to be related to energy, amino acid, vitamin, carbohydrate and lipid metabolism. LC/MS detected 288 different metabolites in the positive and negative ion modes between children with GERD and HCs, which were mainly involved in arachidonic acid (AA), tyrosine, glutathione and caffeine metabolism. Conclusion This study provides new evidence of the pathogenesis of GERD. There are significant differences in the gut microbiota, metabolites and metabolic pathways between HCs and children with GERD, and the differences in metabolites are related to specific changes in bacterial abundance. In the future, GERD may be treated by targeting specific bacteria related to AA metabolism.
Collapse
Affiliation(s)
- Xiaolin Ye
- Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Feihong Yu
- Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jin Zhou
- Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Chunna Zhao
- Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jie Wu
- Department of Gastroenterology, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Xin Ni
- National Center for Pediatric Cancer Surveillance, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| |
Collapse
|
9
|
Yu Y, Lin X, Feng F, Wei Y, Wei S, Gong Y, Guo C, Wang Q, Shuai P, Wang T, Qin H, Li G, Yi L. Gut microbiota and ionizing radiation-induced damage: Is there a link? ENVIRONMENTAL RESEARCH 2023; 229:115947. [PMID: 37080277 DOI: 10.1016/j.envres.2023.115947] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 04/14/2023] [Accepted: 04/17/2023] [Indexed: 05/03/2023]
Abstract
According to observational findings, ionizing radiation (IR) triggers dysbiosis of the intestinal microbiota, affecting the structural composition, function, and species of the gut microbiome and its metabolites. These modifications can further exacerbate IR-induced damage and amplify proinflammatory immune responses. Conversely, commensal bacteria and favorable metabolites can remodel the IR-disturbed gut microbial structure, promote a balance between anti-inflammatory and proinflammatory mechanisms in the body, and mitigate IR toxicity. The discovery of effective and safe remedies to prevent and treat radiation-induced injuries is vitally needed because of the proliferation of radiation toxicity threats produced by recent radiological public health disasters and increasing medical exposures. This review examines how the gut microbiota and its metabolites are linked to the processes of IR-induced harm. We highlight protective measures based on interventions with gut microbes to optimize the distress caused by IR damage to human health. We offer prospects for research in emerging and promising areas targeting the prevention and treatment of IR-induced damage.
Collapse
Affiliation(s)
- Yueqiu Yu
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Xiang Lin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Feiyang Feng
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yuanyun Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Shuang Wei
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Yaqi Gong
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Caimao Guo
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Qingyu Wang
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Peimeng Shuai
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Tiantian Wang
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Hui Qin
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Guoqing Li
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Lan Yi
- Institute of Cytology and Genetics, The Hengyang Key Laboratory of Cellular Stress Biology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|