1
|
Dong Y, Zhang Y, Xu F, Zou K. Extensive genomic characterization, pre-clinical probiotic evaluation, and safety analysis of Bifidobacterium longum subsp. longum BL21 isolated from infant feces. Microb Pathog 2024; 197:107100. [PMID: 39505088 DOI: 10.1016/j.micpath.2024.107100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 10/03/2024] [Accepted: 11/01/2024] [Indexed: 11/08/2024]
Abstract
OBJECTIVE This study aimed to evaluate the safety and probiotic properties of Bifidobacterium longum subsp. longum BL21 isolated from infant feces for use as a commercial probiotic strain. METHODS Whole-genome sequencing; physiological and biochemical assessments; enzymatic assays; metabolite, antibiotic sensitivity, cell adhesion and cytotoxicity, and tolerance tests; and a 14-day oral toxicity study were conducted. RESULTS BL21 exhibited genetic integrity, and its genome lacked genes related to antibiotic resistance or virulence. It was found to be non-pathogenic, had efficient carbohydrate metabolism and mucin degradation ability, and was free from biogenic amines. It also showed susceptibility to antibiotics, strong cell adhesion, and resilience to adverse conditions. The aforementioned results confirm that BL21 is a functional probiotic strain with genetic stability, enzymatic capabilities, and non-pathogenic properties that mean it is safe for oral consumption, demonstrating that it is a promising candidate for probiotic applications. CONCLUSION The study demonstrates that BL21 is a genetically stable, non-pathogenic probiotic strain with metabolic potential. The strain lacks virulence and antibiotic resistance genes, and its resilience to gastrointestinal conditions, as well as the results of the 14-day oral toxicity study, suggest that BL21 is safe for oral consumption. However, further long-term studies and clinical trials are needed to confirm its safety and efficacy for therapeutic use.
Collapse
Affiliation(s)
- Yao Dong
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yinan Zhang
- Shanghai Institute of Quality Inspection and Technical Research, Shanghai, 200233, China
| | - Fei Xu
- College of Food Science and Technology, Henan University of Technology, Zhengzhou, China
| | - Kang Zou
- Germline Stem Cells and Microenvironment Lab, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China; Stem Cell Research and Translation Center, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
2
|
Wang X, Liu M, Xia W. Causal Relationship Between Sjögren's Syndrome and Gut Microbiota: A Two-Sample Mendelian Randomization Study. Biomedicines 2024; 12:2378. [PMID: 39457690 PMCID: PMC11505323 DOI: 10.3390/biomedicines12102378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/13/2024] [Accepted: 10/15/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Gut microbiota have been previously reported to be related to a variety of immune diseases. However, the causal connection between Sjögren's syndrome (SS) and gut microbiota has yet to be clarified. Methods: We employed a two-sample Mendelian randomization (MR) analysis to evaluate the causal connection between gut microbiota and SS, utilizing summary statistics from genome-wide association studies (GWASs) obtained from the MiBioGen and FinnGen consortia. The inverse variance weighted (IVW) approach represents the primary method of Mendelian randomization (MR) analysis. Sensitivity analysis was used to eliminate instrumental variables heterogeneity and horizontal pleiotropy. In addition, we performed an analysis using independent GWAS summary statistics for SS from the European Bioinformatics Institute (EBI) dataset for further verify our results. Results: IVW results demonstrated that the phylum Lentisphaerae (OR = 0.79, 95% CI: 0.63-0.99, p = 0.037), class Deltaproteobacteria (OR = 0.67, 95% CI: 0.47-0.96, p = 0.030), family Porphyromonadaceae (OR = 0.60, 95% CI: 0.38-0.94, p = 0.026), genus Eubacterium coprostanoligenes group (OR = 0.61, 95% CI: 0.4-0.93, p = 0.021), genus Blautia (OR = 0.62, 95% CI: 0.43-0.90, p = 0.012), genus Butyricicoccus (OR = 0.61, 95% CI: 0.42-0.90, p = 0.012), genus Escherichia.Shigella (OR = 0.7, 95% CI: 0.49-0.99, p = 0.045) and genus Subdoligranulum (OR = 0.61, 95% CI: 0.44-0.86, p = 0.005) exhibited protective effects on SS. Relevant heterogeneity of horizontal pleiotropy or instrumental variables was not detected. Furthermore, repeating our results with an independent cohort provided by the EBI dataset, only the genus Eubacterium coprostanoligenes group remained significantly associated with the protective effect on SS (OR = 0.41, 95% CI: 0.18-0.91, p = 0.029). Two-step MR analysis further revealed that genus Eubacterium coprostanoligenes group exerts its protective effect by reducing CXCL6 levels in SS (OR, 0.87; 95% CI = 0.76-0.99, p = 0.033). Conclusions: Our study using two-sample MR analysis identified a causal association between multiple genera and SS. A two-step MR result calculated that genus Eubacterium coprostanoligenes group mediated its protective effect by reducing CXCL6 levels in SS. However, the datasets available from the MiBioGen and FinnGen consortia do not provide sufficient information or comprehensive demographic data for subgroup analyses. Additional validation using various omics technologies is necessary to comprehend the development of SS in the intricate interplay between genes and the environment over a period of time.
Collapse
Affiliation(s)
- Xinrun Wang
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China;
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China;
| | - Minghui Liu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China;
- Department of Urology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Weiping Xia
- Department of Critical Care Medicine, Xiangya Hospital, Central South University, Changsha 410008, China;
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China;
| |
Collapse
|
3
|
Fujiwara A, Fujimoto S, Ishikawa R, Tanaka A. Virtual reality training for radiation safety in cardiac catheterization laboratories - an integrated study. RADIATION PROTECTION DOSIMETRY 2024; 200:1462-1469. [PMID: 39244378 DOI: 10.1093/rpd/ncae187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 07/25/2024] [Accepted: 08/21/2024] [Indexed: 09/09/2024]
Abstract
The advent of fluoroscopically guided cardiology procedures has greatly improved patient outcomes but has also increased occupational radiation exposure for healthcare professionals, leading to adverse health effects such as radiation-induced cataracts, alopecia, and cancer. This emphasizes the need for effective radiation safety training. Traditional training methods, often based on passive learning, fail to simulate the dynamic catheterization laboratory environment adequately. Virtual Reality (VR) offers a promising alternative by providing immersive, interactive experiences that mimic real-world scenarios without the risks of actual radiation exposure. Our study aims to assess the effectiveness of VR-based radiation safety training compared to traditional methods. We conducted a prospective cohort study involving 48 healthcare professionals in a catheterization lab setting. Participants underwent a 1-hour self-directed VR training session using Virtual Medical Coaching's RadSafe VR software, which simulates real-world clinical scenarios. Pre- and post-intervention radiation dose levels were measured using personal dosimeters at the eye, chest, and pelvis. Knowledge and skills were assessed through tests, and feedback was gathered through surveys and interviews. Statistical analysis revealed significant reductions in radiation exposure across all professional groups after VR training. For cardiologists, the eye dose dropped by 21.88% (from 2.88 mSv to 2.25 mSv), the chest dose decreased by 21.65% (from 4.11 mSv to 3.22 mSv), and the pelvis dose went down by 21.84% (from 2.06 mSv to 1.61 mSv). Perioperative nurses experienced similar reductions, with eye doses decreasing by 14.74% (from 1.56 mSv to 1.33 mSv), chest doses by 26.92% (from 2.6 mSv to 1.9 mSv), and pelvis doses by 26.92% (from 1.3 mSv to 0.95 mSv). Radiographers saw their eye doses reduced by 18.95% (from 0.95 mSv to 0.77 mSv), chest doses by 42.11% (from 1.9 mSv to 1.1 mSv), and pelvis doses by 27.63% (from 0.76 mSv to 0.55 mSv).Participants reported enhanced engagement, improved understanding of radiation safety, and a preference for VR over traditional methods. A cost analysis also demonstrated the economic advantages of VR training, with significant savings in staff time and rental costs compared to traditional methods. Our findings suggest that VR is a highly effective and cost-efficient training tool for radiation safety in healthcare, offering significant benefits over traditional training approaches.
Collapse
Affiliation(s)
- Asahi Fujiwara
- Fujita Health University Hospital, Physics, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Sota Fujimoto
- Fujita Health University Hospital, Physics, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Ren Ishikawa
- Fujita Health University Hospital, Physics, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| | - Aoi Tanaka
- Fujita Health University Hospital, Physics, 1-98 Dengakugakubo, Kutsukake-cho, Toyoake, Aichi 470-1192, Japan
| |
Collapse
|
4
|
Ju T, Song Z, Qin D, Cheng J, Li T, Hu G, Fu S. Neohesperidin Attenuates DSS-Induced Ulcerative Colitis by Inhibiting Inflammation, Reducing Intestinal Barrier Damage, and Modulating Intestinal Flora Composition. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:20419-20431. [PMID: 39249130 DOI: 10.1021/acs.jafc.4c04433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/10/2024]
Abstract
Flavonoid natural products are emerging as a promising approach for treating Ulcerative Colitis (UC) due to their natural origin and minimal toxicity. This study investigates the effects of Neohesperidin (NEO), a natural flavonoid, on Dextran Sodium Sulfate (DSS)-induced UC in mice, focusing on the underlying molecular mechanisms. Early intervention with NEO (25 and 50 mg/kg) mitigated colon shortening, restored damaged barrier proteins, and significantly reduced the inflammatory cytokine levels. Moreover, NEO inhibited the MAPK/NF-κB signaling pathway and enhanced the levels of intestinal barrier proteins (Claudin-3 and ZO-1). Additionally, NEO increased beneficial intestinal probiotics (S24-7 and Lactobacillaceae) while reducing harmful bacteria (Erysipelotrichi, Enterobacteriaceae). Fecal microbial transplantation (FMT) results demonstrated that NEO (50 mg/kg) markedly improved UC symptoms. In conclusion, early NEO intervention may alleviate DSS-induced UC by inhibiting inflammatory responses, preserving intestinal barrier integrity and modulating gut microbiota.
Collapse
Affiliation(s)
- Tianyuan Ju
- State Key Laboratory for diagnosis and treatment of Sever Zoonotic Infectious Diseases, Key Laboratory for Zoonsis Research of the Ministry of Education, Institute of Zoonsis and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Zheyu Song
- Department of Gastrointestinal, Colorectal and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130031, China
| | - Di Qin
- State Key Laboratory for diagnosis and treatment of Sever Zoonotic Infectious Diseases, Key Laboratory for Zoonsis Research of the Ministry of Education, Institute of Zoonsis and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Ji Cheng
- State Key Laboratory for diagnosis and treatment of Sever Zoonotic Infectious Diseases, Key Laboratory for Zoonsis Research of the Ministry of Education, Institute of Zoonsis and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Tong Li
- State Key Laboratory for diagnosis and treatment of Sever Zoonotic Infectious Diseases, Key Laboratory for Zoonsis Research of the Ministry of Education, Institute of Zoonsis and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Guiqiu Hu
- State Key Laboratory for diagnosis and treatment of Sever Zoonotic Infectious Diseases, Key Laboratory for Zoonsis Research of the Ministry of Education, Institute of Zoonsis and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Shoupeng Fu
- State Key Laboratory for diagnosis and treatment of Sever Zoonotic Infectious Diseases, Key Laboratory for Zoonsis Research of the Ministry of Education, Institute of Zoonsis and College of Veterinary Medicine, Jilin University, Changchun 130062, China
| |
Collapse
|
5
|
Acharya M, Venkidesh BS, Mumbrekar KD. Bacterial supplementation in mitigation of radiation-induced gastrointestinal damage. Life Sci 2024; 353:122921. [PMID: 39032692 DOI: 10.1016/j.lfs.2024.122921] [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: 04/27/2024] [Revised: 07/08/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Pelvic irradiation, a crucial treatment for pelvic malignancies, is associated with the risk of gastrointestinal (GI) damage due to the high proliferation rate of epithelial cells. The radiosensitive gastrointestinal tract acts as a dose-limiting organ. High doses of ionizing radiation can cause inflammation and rupture of mucosal barriers and can also lead to intestinal fibrosis. Intestinal damage can cause acute to chronic complications, reducing patients' quality of life. The gut microbiota plays a vital role in maintaining gut health, and any changes in the gut microbial composition can worsen damage, emphasizing the importance of therapies that target and sustain the gut microbiota during radiotherapy. One potential strategy to prevent radiation-induced GI damage is to use bacterial supplements. Research suggests that probiotic supplementation may alleviate radiation-induced gastrointestinal damage, maintaining intestinal morphology and decreasing epithelial injury in cancer patients. The observed protective effects occur through various mechanisms, including antioxidant activities, modulation of the immune response, and preservation of gut barrier function. To optimize probiotic therapies, it is imperative to elucidate these mechanisms. The efficiency of probiotics as radioprotectors is highly dependent on the time and dose of administration, and their interaction with the host immune system is a key facet of their therapeutic potential. This review explores the potential benefits of bacterial supplementation in mitigating radiation-induced GI damage and the underlying mechanism. This highlights the need for further research to establish standardized protocols and refine probiotic supplementation strategies, underscoring the potential for enhancing therapeutic outcomes in patients undergoing pelvic radiotherapy.
Collapse
Affiliation(s)
- Meghana Acharya
- Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Babu Santhi Venkidesh
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India
| | - Kamalesh Dattaram Mumbrekar
- Department of Radiation Biology & Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal 576104, India.
| |
Collapse
|
6
|
Xie LW, Lu HY, Tang LF, Tang FL, Zhu RQ, Wang DF, Cai S, Tian Y, Li M. Probiotic Consortia Protect the Intestine Against Radiation Injury by Improving Intestinal Epithelial Homeostasis. Int J Radiat Oncol Biol Phys 2024; 120:189-204. [PMID: 38485099 DOI: 10.1016/j.ijrobp.2024.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 02/25/2024] [Accepted: 03/02/2024] [Indexed: 04/14/2024]
Abstract
PURPOSE Radiation-induced intestinal injury (RIII) commonly occur during abdominal-pelvic cancer radiation therapy; however, no effective prophylactic or therapeutic agents are available to manage RIII currently. This study aimed to clarify the potential of probiotic consortium supplementation in alleviating RIII. METHODS AND MATERIALS Male C57BL/6J mice were orally administered a probiotic mixture comprising Bifidobacterium longum BL21, Lactobacillus paracasei LC86, and Lactobacillus plantarum Lp90 for 30 days before exposure to 13 Gy of whole abdominal irradiation. The survival rates, clinical scores, and histologic changes in the intestines of mice were assessed. The impacts of probiotic consortium treatment on intestinal stem cell proliferation, differentiation, and epithelial barrier function; oxidative stress; and inflammatory cytokines were evaluated. A comprehensive examination of the gut microbiota composition was conducted through 16S rRNA sequencing, while changes in metabolites were identified using liquid chromatography-mass spectrometry. RESULTS The probiotic consortium alleviated RIII, as reflected by increased survival rates, improved clinical scores, and mitigated mucosal injury. The probiotic consortium treatment exhibited enhanced therapeutic effects at the histologic level compared with individual probiotic strains, although there was no corresponding improvement in survival rates and colon length. Moreover, the probiotic consortium stimulated intestinal stem cell proliferation and differentiation, enhanced the integrity of the intestinal epithelial barrier, and regulated redox imbalance and inflammatory responses in irradiated mice. Notably, the treatment induced a restructuring of the gut microbiota composition, particularly enriching short-chain fatty acid-producing bacteria. Metabolomic analysis revealed distinctive metabolic changes associated with the probiotic consortium, including elevated levels of anti-inflammatory and antiradiation metabolites. CONCLUSIONS The probiotic consortium attenuated RIII by modulating the gut microbiota and metabolites, improving inflammatory symptoms, and regulating oxidative stress. These findings provide new insights into the maintenance of intestinal health with probiotic consortium supplementation and will facilitate the development of probiotic-based therapeutic strategies for RIII in clinical practice.
Collapse
Affiliation(s)
- Li-Wei Xie
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, China; Institute of Radiotherapy and Oncology, Soochow University, Suzhou, China
| | - Hai-Yan Lu
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, China; Institute of Radiotherapy and Oncology, Soochow University, Suzhou, China
| | - Lin-Feng Tang
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, China; State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Feng-Ling Tang
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, China; Institute of Radiotherapy and Oncology, Soochow University, Suzhou, China
| | - Rui-Qiu Zhu
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, China; Institute of Radiotherapy and Oncology, Soochow University, Suzhou, China
| | - Di-Fan Wang
- Medical College of Soochow University, Suzhou, China
| | - Shang Cai
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, China; Institute of Radiotherapy and Oncology, Soochow University, Suzhou, China
| | - Ye Tian
- Department of Radiotherapy and Oncology, Second Affiliated Hospital of Soochow University, Suzhou, China; Institute of Radiotherapy and Oncology, Soochow University, Suzhou, China.
| | - Ming Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China.
| |
Collapse
|
7
|
Zhu X, Li Y, Tian X, Jing Y, Wang Z, Yue L, Li J, Wu L, Zhou X, Yu Z, Zhang Y, Guan F, Yang M, Zhang B. REGγ Mitigates Radiation-Induced Enteritis by Preserving Mucin Secretion and Sustaining Microbiome Homeostasis. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:975-988. [PMID: 38423356 DOI: 10.1016/j.ajpath.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/02/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024]
Abstract
Radiation-induced enteritis, a significant concern in abdominal radiation therapy, is associated closely with gut microbiota dysbiosis. The mucus layer plays a pivotal role in preventing the translocation of commensal and pathogenic microbes. Although significant expression of REGγ in intestinal epithelial cells is well established, its role in modulating the mucus layer and gut microbiota remains unknown. The current study revealed notable changes in gut microorganisms and metabolites in irradiated mice lacking REGγ, as compared to wild-type mice. Concomitant with gut microbiota dysbiosis, REGγ deficiency facilitated the infiltration of neutrophils and macrophages, thereby exacerbating intestinal inflammation after irradiation. Furthermore, fluorescence in situ hybridization assays unveiled an augmented proximity of bacteria to intestinal epithelial cells in REGγ knockout mice after irradiation. Mechanistically, deficiency of REGγ led to diminished goblet cell populations and reduced expression of key goblet cell markers, Muc2 and Tff3, observed in both murine models, minigut organoid systems and human intestinal goblet cells, indicating the intrinsic role of REGγ within goblet cells. Interestingly, although administration of broad-spectrum antibiotics did not alter the goblet cell numbers or mucin 2 (MUC2) secretion, it effectively attenuated inflammation levels in the ileum of irradiated REGγ absent mice, bringing them down to the wild-type levels. Collectively, these findings highlight the contribution of REGγ in counteracting radiation-triggered microbial imbalances and cell-autonomous regulation of mucin secretion.
Collapse
Affiliation(s)
- Xiangzhan Zhu
- Institute of Pediatric Medicine, Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China; School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ya Li
- Henan Key Laboratory of Rehabilitation Medicine, Henan Joint International Research Laboratory of Chronic Liver Injury, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xue Tian
- Department of Respiratory and Critical Care Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiming Jing
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China
| | - Zimeng Wang
- Department of Pharmacology and Cancer, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Lingling Yue
- Institute of Pediatric Medicine, Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Jianhui Li
- Department of Pathology, Xuchang Central Hospital Affiliated to Henan University of Science and Technology, Xuchang, China
| | - Ling Wu
- Department of Medical Imaging, Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Xinkui Zhou
- Institute of Pediatric Medicine, Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Zhidan Yu
- Institute of Pediatric Medicine, Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Yaodong Zhang
- Institute of Pediatric Medicine, Henan Children's Hospital, Zhengzhou Children's Hospital, Children's Hospital Affiliated of Zhengzhou University, Zhengzhou, China
| | - Fangxia Guan
- School of Life Sciences, Zhengzhou University, Zhengzhou, China
| | - Minglei Yang
- Department of Orthopedic Oncology, The Second Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Bianhong Zhang
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences, School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
8
|
Deyang T, Baig MAI, Dolkar P, Hediyal TA, Rathipriya AG, Bhaskaran M, PandiPerumal SR, Monaghan TM, Mahalakshmi AM, Chidambaram SB. Sleep apnoea, gut dysbiosis and cognitive dysfunction. FEBS J 2024; 291:2519-2544. [PMID: 37712936 DOI: 10.1111/febs.16960] [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: 05/26/2023] [Revised: 08/14/2023] [Accepted: 09/13/2023] [Indexed: 09/16/2023]
Abstract
Sleep disorders are becoming increasingly common, and their distinct effects on physical and mental health require elaborate investigation. Gut dysbiosis (GD) has been reported in sleep-related disorders, but sleep apnoea is of particular significance because of its higher prevalence and chronicity. Cumulative evidence has suggested a link between sleep apnoea and GD. This review highlights the gut-brain communication axis that is mediated via commensal microbes and various microbiota-derived metabolites (e.g. short-chain fatty acids, lipopolysaccharide and trimethyl amine N-oxide), neurotransmitters (e.g. γ-aminobutyric acid, serotonin, glutamate and dopamine), immune cells and inflammatory mediators, as well as the vagus nerve and hypothalamic-pituitary-adrenal axis. This review also discusses the pathological role underpinning GD and altered gut bacterial populations in sleep apnoea and its related comorbid conditions, particularly cognitive dysfunction. In addition, the review examines the preclinical and clinical evidence, which suggests that prebiotics and probiotics may potentially be beneficial in sleep apnoea and its comorbidities through restoration of eubiosis or gut microbial homeostasis that regulates neural, metabolic and immune responses, as well as physiological barrier integrity via the gut-brain axis.
Collapse
Affiliation(s)
- Tenzin Deyang
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Md Awaise Iqbal Baig
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Phurbu Dolkar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Tousif Ahmed Hediyal
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | | | - Mahendran Bhaskaran
- College of Pharmacy and Pharmaceutical Sciences, Frederic and Mary Wolf Center, University of Toledo Health Science Campus, OH, USA
| | - Seithikuruppu R PandiPerumal
- Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
- Division of Research and Development, Lovely Professional University, Phagwara, India
| | - Tanya M Monaghan
- National Institute for Health Research Nottingham Biomedical Research Centre, University of Nottingham, UK
- Nottingham Digestive Diseases Centre, School of Medicine, University of Nottingham, UK
| | - Arehally M Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
- SIG-Brain, Behaviour and Cognitive Neurosciences Research (BBRC), JSS Academy of Higher Education & Research, Mysuru, India
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
- SIG-Brain, Behaviour and Cognitive Neurosciences Research (BBRC), JSS Academy of Higher Education & Research, Mysuru, India
| |
Collapse
|
9
|
Zheng C, Niu M, Kong Y, Liu X, Li J, Gong X, Ren X, Hong C, Yin M, Wang L. Oral administration of probiotic spore ghosts for efficient attenuation of radiation-induced intestinal injury. J Nanobiotechnology 2024; 22:303. [PMID: 38822376 PMCID: PMC11140926 DOI: 10.1186/s12951-024-02572-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 05/20/2024] [Indexed: 06/03/2024] Open
Abstract
Radiation-induced intestinal injury is the most common side effect during radiotherapy of abdominal or pelvic solid tumors, significantly impacting patients' quality of life and even resulting in poor prognosis. Until now, oral application of conventional formulations for intestinal radioprotection remains challenging with no preferred method available to mitigate radiation toxicity in small intestine. Our previous study revealed that nanomaterials derived from spore coat of probiotics exhibit superior anti-inflammatory effect and even prevent the progression of cancer. The aim of this work is to determine the radioprotective effect of spore coat (denoted as spore ghosts, SGs) from three clinically approved probiotics (B.coagulans, B.subtilis and B.licheniformis). All the three SGs exhibit outstanding reactive oxygen species (ROS) scavenging ability and excellent anti-inflammatory effect. Moreover, these SGs can reverse the balance of intestinal flora by inhibiting harmful bacteria and increasing the abundance of Lactobacillus. Consequently, administration of SGs significantly reduce radiation-induced intestinal injury by alleviating diarrhea, preventing X-ray induced apoptosis of small intestinal epithelial cells and promoting restoration of barrier integrity in a prophylactic study. Notably, SGs markedly improve weight gain and survival of mice received total abdominal X-ray radiation. This work may provide promising radioprotectants for efficiently attenuating radiation-induced gastrointestinal syndrome and promote the development of new intestinal predilection.
Collapse
Affiliation(s)
- Cuixia Zheng
- Translational medicine Center, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Mengya Niu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Yueyue Kong
- Xinjiang Aksu First People's Hospital, Akesu, 843000, China
| | - Xinxin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
- Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471009, China
| | - Junxiu Li
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xunwei Gong
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Xinyuan Ren
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Chen Hong
- Translational medicine Center, Huaihe Hospital of Henan University, Kaifeng, 475000, China
| | - Menghao Yin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Lei Wang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China.
- Pingyuan Lab, Henan Normal University, Xinxiang, 453007, China.
- Luoyang Central Hospital Affiliated to Zhengzhou University, Luoyang, 471009, China.
| |
Collapse
|
10
|
Chuandong Z, Hu J, Li J, Wu Y, Wu C, Lai G, Shen H, Wu F, Tao C, Liu S, Zhang W, Shao H. Distribution and roles of Ligilactobacillus murinus in hosts. Microbiol Res 2024; 282:127648. [PMID: 38367479 DOI: 10.1016/j.micres.2024.127648] [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: 08/30/2023] [Revised: 10/26/2023] [Accepted: 02/10/2024] [Indexed: 02/19/2024]
Abstract
Ligilactobacillus murinus, a member of the Ligilactobacillus genus, holds significant potential as a probiotic. While research on Ligilactobacillus murinus has been relatively limited compared to well-studied probiotic lactic acid bacteria such as Limosilactobacillus reuteri and Lactobacillus gasseri, a mounting body of evidence highlights its extensive involvement in host intestinal metabolism and immune activities. Moreover, its abundance exhibits a close correlation with intestinal health. Notably, beyond the intestinal context, Ligilactobacillus murinus is gaining recognition for its contributions to metabolism and regulation in the oral cavity, lungs, and vagina. As such, Ligilactobacillus murinus emerges as a potential probiotic candidate with a pivotal role in supporting host well-being. This review delves into studies elucidating the multifaceted roles of Ligilactobacillus murinus. It also examines its medicinal potential and associated challenges, underscoring the imperative to delve deeper into unraveling the mechanisms of its actions and exploring its health applications.
Collapse
Affiliation(s)
- Zhou Chuandong
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Jicong Hu
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Jiawen Li
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Yuting Wu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Chan Wu
- School of Clinical Medicine, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Guanxi Lai
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Han Shen
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Fenglin Wu
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Changli Tao
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Song Liu
- School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China
| | - Wenfeng Zhang
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China.
| | - Hongwei Shao
- School of Life Science and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
11
|
Giridhar P, Pradhan S, Dokania S, Venkatesulu B, Sarode R, Welsh JS. Microbiome and Abdominopelvic Radiotherapy Related Chronic Enteritis: A Microbiome-based Mechanistic Role of Probiotics and Antibiotics. Am J Clin Oncol 2024; 47:246-252. [PMID: 38193365 DOI: 10.1097/coc.0000000000001082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2024]
Abstract
Chronic diarrhea and abdominal pain after radiotherapy continue to be a problem in cancer survivors. Gut microbiomes are essential for preventing intestinal inflammation, maintaining intestinal integrity, maintaining enterohepatic circulation, regulating bile acid metabolism, and absorption of nutrients, including fat-soluble vitamins. Gut microbiome dysbiosis is expected to cause inflammation, bile acid malabsorption, malnutrition, and associated symptoms. Postradiotherapy, Firmicutes and Bacteroidetes phylum are significantly decreased while Fusobacteria and other unclassified bacteria are increased. Available evidence suggests harmful bacteria Veillonella, Erysipelotrichaceae, and Ruminococcus are sensitive to Metronidazole or Ciprofloxacin. Beneficial bacteria lactobacillus and Bifidobacterium are relatively resistant to metronidazole. We hypothesize and provide an evidence-based review that short-course targeted antibiotics followed by specific probiotics may lead to alleviation of radiation enteritis.
Collapse
Affiliation(s)
| | | | | | - Bhanuprasad Venkatesulu
- Department of Radiation Oncology, Loyola University Chicago, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Maywood
- Department of Radiation Oncology, MPMMCC/HBCH Varanasi Edward Hines Veteran Affairs Hospital, Chicago, IL
| | - Rahul Sarode
- Department of Microbiology, Mahamana Pandit Madanmohan Malaviya Cancer Centre/Homi Bhabha Cancer hospital, Tata Memorial Centre, Varanasi, India
| | - James S Welsh
- Department of Radiation Oncology, Loyola University Chicago, Stritch School of Medicine, Cardinal Bernardin Cancer Center, Maywood
- Department of Radiation Oncology, MPMMCC/HBCH Varanasi Edward Hines Veteran Affairs Hospital, Chicago, IL
| |
Collapse
|
12
|
Lee SU, Jang BS, Na YR, Lee SH, Han S, Chang JH, Kim HJ. Effect of Lactobacillus Rhamnosus GG for Regulation of Inflammatory Response in Radiation-Induced Enteritis. Probiotics Antimicrob Proteins 2024; 16:636-648. [PMID: 37072632 DOI: 10.1007/s12602-023-10071-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/22/2023] [Indexed: 04/20/2023]
Abstract
The purpose of this study was to investigate the role of Lactobacillus rhamnosus GG (LGG) probiotics in radiation enteritis using in vivo mice. A total of 40 mice were randomly assigned to four groups: control, probiotics, radiotherapy (RT), and RT + probiotics. For the group of probiotics, 0.2 mL of solution that contained 1.0 × 108 colony-forming units (CFU) of LGG was used and orally administered daily until sacrifice. For RT, a single dose of 14 Gy was administered using a 6 mega-voltage photon beam to the abdominopelvic area. Mice were sacrifice at day 4 (S1) and day 7 (S2) after RT. Their jejunum, colon, and stool were collected. A multiplex cytokine assay and 16 s ribosomal RNA amplicon sequencing were then performed. Regarding cytokine concentrations in tissues, pro-inflammatory cytokines, such as tumor necrosis factor-α, interleukin-6 and monocyte chemotactic protein-1, showed significantly decreased protein levels in colon tissues of the RT + probiotics group than in the RT alone group (all p < 0.05). As for comparing microbial abundance through alpha-diversity and beta-diversity, no significant differences were observed between the RT + probiotics and RT alone groups, except for an increase in alpha-diversity in the stool of the RT + probiotics group. Upon analysis of differential microbes based on treatment, the dominance of anti-inflammatory-related microbes, such as Porphyromonadaceae, Bacteroides acidifaciens, and Ruminococcus, was observed in the jejunum, colon, and stool of the RT + probiotics group. With regard to predicted metabolic pathway abundances, the pathways associated with anti-inflammatory processes, such as biosynthesis of pyrimidine nucleotides, peptidoglycans, tryptophan, adenosylcobalamin, and propionate, were differentially identified in the RT + probiotics group compared to the RT alone group. Protective effects of probiotics on radiation enteritis were potentially derived from dominant anti-inflammation-related microbes and metabolites.
Collapse
Affiliation(s)
- Sung Uk Lee
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, South Korea
- Proton Therapy Center, National Cancer Center, Goyang, South Korea
| | - Bum-Sup Jang
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, South Korea
- Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea
| | - Yi Rang Na
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Sun Hwa Lee
- Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea
| | - Sunwoo Han
- Transdisciplinary Department of Medicine and Advanced Technology, Seoul National University Hospital, Seoul, South Korea
| | - Ji Hyun Chang
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea.
| | - Hak Jae Kim
- Department of Radiation Oncology, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Radiation Oncology, Seoul National University Hospital, Seoul, South Korea.
- Cancer Research Institute, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
13
|
Kong Q, Chen X, Liu Y, Ali F, Idrees A, Ataya FS, Shang Z, Li K. Sodium acetate and sodium butyrate attenuate diarrhea in yak calves by regulating gut microbiota and metabolites. Heliyon 2024; 10:e26564. [PMID: 38439875 PMCID: PMC10909669 DOI: 10.1016/j.heliyon.2024.e26564] [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: 07/01/2023] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 03/06/2024] Open
Abstract
Diarrhea is a severe issue in calves that causes fertility problems and economic issues worldwide. Sodium acetate/sodium butyrate (SA/SB) alleviates diarrhea in mice; however, little information is available about the preventive effect of SA/SB on diarrheic yak calves living on the Tibet plateau. Yak calves (n = 19) of age ≥4 months and weight 37 ± 2 Kg were randomly divided into control (C, n = 10) and supplement groups (S, n = 9). Yaks belonging to the supplement group were given sodium butyrate (10 g/kg) and sodium acetate (5 g/kg) for 28 days, along with normal feed, seasonal grasses, pasture, and water. The blood and fecal samples from yak calves were collected for assessment of antioxidant capacity, inflammatory cytokines, microbiome, and short-chain fatty acids (SCFAs) concentration analysis. Results of this study revealed that a lower diarrhea rate, higher weight, and net weight gain were recorded in yaks belonging to group S supplemented with SA/SB. Similarly, increased antioxidant capacity with higher levels of T-AOC, SOD, and GSH-px and decreased inflammatory reactions by decreasing both TNF-α and IL-1β concentrations were recorded in yaks of group S. The concentration of SCFAs was significantly higher (p < 0.05) in yaks from group S than group C. Microbiome analysis revealed that 8 phyla and 54 genera were significantly different (p < 0.05) in both yak groups, with increased probiotics (Akkermansia, Oscillospira), SCFAs producing genera (Oscillospira, ASF356, Anaerosporobacter and Phascolarctobacterium), and decreased inflammatory related genus (Flavonifractor, Fournierella) and harmful bacteria (Oscillibacter, Achromobacter) in group S. In conclusion, the results demonstrated that SA and SB could decrease diarrhea rates in yak calves on the plateau via increasing antioxidant ability and SCFAs, while decreasing inflammatory responses in yaks by moderating gut microbiota. The current results provide new insights for the prevention and treatment of diarrhea in yaks.
Collapse
Affiliation(s)
- Qinghui Kong
- Key Laboratory of Clinical Veterinary Medicine in Tibet, Tibet Agriculture and Animal Husbandry College, Linzhi, 860000, Tibet, China
| | - Xiushuang Chen
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yang Liu
- Institute of Animal Husbandry and Veterinary Medicine, Tibet Academy of Agricultural and Animal Husbandry Sciences, Lhasa, 850000, China
| | - Farah Ali
- Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, 63100, Pakistan
| | - Asif Idrees
- KBCMA, College of Veterinary and Animal Sciences, Narowal, Pakistan
| | - Farid Shokry Ataya
- Department of Biochemistry, College of Science, King Saud University, PO Box 2455, Riyadh, 11451, Saudi Arabia
| | - Zhenda Shang
- Key Laboratory of Clinical Veterinary Medicine in Tibet, Tibet Agriculture and Animal Husbandry College, Linzhi, 860000, Tibet, China
| | - Kun Li
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| |
Collapse
|
14
|
Thandar M, Yang X, Zhu Y, Zhang X, Chen Z, Huang S, Chi P. Dysbiosis of gut microbiota and metabolites is associated with radiation-induced colorectal fibrosis and is restored by adipose-derived mesenchymal stem cell therapy. Life Sci 2024; 341:122502. [PMID: 38350495 DOI: 10.1016/j.lfs.2024.122502] [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: 11/22/2023] [Revised: 02/01/2024] [Accepted: 02/08/2024] [Indexed: 02/15/2024]
Abstract
AIMS This study aimed to investigate the effects of adipose-derived mesenchymal stem cells (ADSCs) on radiation-induced colorectal fibrosis (RICF) along with the associated dysbiosis of gut microbiota and metabolites. MAIN METHODS Fecal microbiota were assessed through 16S rRNA gene sequencing, and the fecal metabolome was characterized using liquid chromatography-mass spectrometry and gas chromatography-mass spectrometry. The correlation between microbiota and metabolome data was explored. KEY FINDINGS ADSC injection demonstrated a significant restoration of radiation-induced intestinal damage in vivo. At the phylum level, irradiated rats exhibited an increase in Bacteroidota and Campilobacterota, and a decrease in Firmicutes and Desulfobacterota, contrasting with the ADSC treatment group. Metabolomic analysis revealed 72 differently expressed metabolites (DEMs) from gas chromatography-mass spectrometry and 284 DEMs from liquid chromatography-mass spectrometry in the radiation group compared to the blank group. In the ADSC treatment group versus the radiation group, 36 DEMs from gas chromatography-mass spectrometry and 341 DEMs from liquid chromatography-mass spectrometry were identified. KEGG enrichment analysis implicated pathways such as steroid hormone biosynthesis, gap junction, primary bile acid biosynthesis, citrate cycle, cAMP signaling pathway, and alanine, aspartate, and glutamate metabolism during RICF progression and after treated with ADSCs. Correlation analysis highlighted the role of ADSCs in modulating the metabolic process of Camelledionol in fecal Bacteroides. SIGNIFICANCE These findings underscore the potential of ADSCs in reversing dysbiosis and restoring normal colonic flora in the context of RICF, offering valuable insights for therapeutic interventions targeting radiation-induced complications.
Collapse
Affiliation(s)
- Mya Thandar
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Department of Colorectal Surgery, Fuzhou, Fujian Province 350001, China
| | - Xiaojie Yang
- Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Department of Colorectal Surgery, Fuzhou, Fujian Province 350001, China; Department of Thoracic Surgery, Third Affiliated Hospital of Chongqing Medical University, Chongqing 401100, China
| | - Yuanchang Zhu
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Department of Colorectal Surgery, Fuzhou, Fujian Province 350001, China
| | - Xueying Zhang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Department of Colorectal Surgery, Fuzhou, Fujian Province 350001, China
| | - Zhifen Chen
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Training Center of Minimally Invasive Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China.
| | - Shenghui Huang
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Training Center of Minimally Invasive Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China.
| | - Pan Chi
- Department of Colorectal Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China; Key Laboratory of Ministry of Education for Gastrointestinal Cancer, Fujian Medical University, Department of Colorectal Surgery, Fuzhou, Fujian Province 350001, China; Training Center of Minimally Invasive Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province 350001, China.
| |
Collapse
|
15
|
Wang W, Cui B, Nie Y, Sun L, Zhang F. Radiation injury and gut microbiota-based treatment. Protein Cell 2024; 15:83-97. [PMID: 37470727 PMCID: PMC10833463 DOI: 10.1093/procel/pwad044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 07/10/2023] [Indexed: 07/21/2023] Open
Abstract
The exposure to either medical sources or accidental radiation can cause varying degrees of radiation injury (RI). RI is a common disease involving multiple human body parts and organs, yet effective treatments are currently limited. Accumulating evidence suggests gut microbiota are closely associated with the development and prevention of various RI. This article summarizes 10 common types of RI and their possible mechanisms. It also highlights the changes and potential microbiota-based treatments for RI, including probiotics, metabolites, and microbiota transplantation. Additionally, a 5P-Framework is proposed to provide a comprehensive strategy for managing RI.
Collapse
Affiliation(s)
- Weihong Wang
- Department of Microbiota Medicine and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
- Department of Microbiotherapy, Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Bota Cui
- Department of Microbiota Medicine and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
- Department of Microbiotherapy, Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
| | - Yongzhan Nie
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi’an 710032, China
- National Clinical Research Center for Digestive Diseases, Xi’an 710032, China
| | - Lijuan Sun
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Northwest University, Xi’an 710069, China
| | - Faming Zhang
- Department of Microbiota Medicine and Medical Center for Digestive Diseases, The Second Affiliated Hospital of Nanjing Medical University, Nanjing 210011, China
- Department of Microbiotherapy, Sir Run Run Hospital, Nanjing Medical University, Nanjing 211166, China
- National Clinical Research Center for Digestive Diseases, Xi’an 710032, China
| |
Collapse
|
16
|
Xie LW, Cai S, Lu HY, Tang FL, Zhu RQ, 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. [PMID: 38706205 PMCID: PMC11086037 DOI: 10.1080/19490976.2024.2347722] [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: 10/01/2023] [Accepted: 04/22/2024] [Indexed: 05/07/2024] Open
Abstract
The intestine is prone to radiation damage in patients undergoing radiotherapy for pelvic tumors. However, there are currently no effective drugs available for the prevention or treatment of radiation-induced enteropathy (RIE). In this study, we aimed at investigating the impact of indole-3-carboxaldehyde (I3A) derived from the intestinal microbiota on RIE. Intestinal organoids were isolated and cultivated for screening radioprotective tryptophan metabolites. A RIE model was established using 13 Gy whole-abdominal irradiation in male C57BL/6J mice. After oral administration of I3A, its radioprotective ability was assessed through the observation of survival rates, clinical scores, and pathological analysis. Intestinal stem cell survival and changes in the intestinal barrier were observed through immunofluorescence and immunohistochemistry. Subsequently, the radioprotective mechanisms of I3A was investigated through 16S rRNA and transcriptome sequencing, respectively. Finally, human colon cancer cells and organoids were cultured to assess the influence of I3A on tumor radiotherapy. I3A exhibited the most potent radioprotective effect on intestinal organoids. Oral administration of I3A treatment significantly increased the survival rate in irradiated mice, improved clinical and histological scores, mitigated mucosal damage, enhanced the proliferation and differentiation of Lgr5+ intestinal stem cells, and maintained intestinal barrier integrity. Furthermore, I3A enhanced the abundance of probiotics, and activated the AhR/IL-10/Wnt signaling pathway to promote intestinal epithelial proliferation. As a crucial tryptophan metabolite, I3A promotes intestinal epithelial cell proliferation through the AhR/IL-10/Wnt signaling pathway and upregulates the abundance of probiotics to treat RIE. Microbiota-derived I3A demonstrates potential clinical application value for the treatment of RIE.
Collapse
Affiliation(s)
- Li-Wei Xie
- Suzhou Key Laboratory for Radiation Oncology, Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Shang Cai
- Suzhou Key Laboratory for Radiation Oncology, Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Hai-Yan Lu
- Suzhou Key Laboratory for Radiation Oncology, Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Feng-Ling Tang
- Suzhou Key Laboratory for Radiation Oncology, Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Rui-Qiu Zhu
- Suzhou Key Laboratory for Radiation Oncology, Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ye Tian
- Suzhou Key Laboratory for Radiation Oncology, Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ming Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University; Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| |
Collapse
|
17
|
Chen Y, Wang X, Ye Y, Ren Q. Gut microbiota in cancer: insights on microbial metabolites and therapeutic strategies. Med Oncol 2023; 41:25. [PMID: 38129370 DOI: 10.1007/s12032-023-02249-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Accepted: 11/11/2023] [Indexed: 12/23/2023]
Abstract
In recent years, the role of gut microbiota in cancer treatment has attracted substantial attention. It is now well established that gut microbiota and its metabolites significantly contribute to the incidence, treatment, and prognosis of various cancers. This review provides a comprehensive review on the pivotal role of gut microbiota and their metabolites in cancer initiation and progression. Furthermore, it evaluates the impact of gut microbiota on the efficacy and associated side effects of anticancer therapies, including radiotherapy, chemotherapy, and immunotherapy, thus emphasizing the clinical importance of gut microbiota reconstitution in cancer treatment.
Collapse
Affiliation(s)
- Yalan Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Xibin Wang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Yuwei Ye
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China
- Gansu Province Clinical Research Center for Digestive Diseases, Lanzhou University, Lanzhou, 730000, Gansu Province, China
| | - Qian Ren
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
- Department of Gastroenterology, The First Hospital of Lanzhou University, Lanzhou, 730000, Gansu Province, China.
- Gansu Province Clinical Research Center for Digestive Diseases, Lanzhou University, Lanzhou, 730000, Gansu Province, China.
| |
Collapse
|
18
|
Zhao H, Li W, Zhou X, Pan L, Feng Y, Gao P, Ji J, Zhang H, Zhao K, Wang C, Lu Z. C-X-C Motif Chemokine Ligand 1 Promotes Colitis by Modulating the Gut Microbiota. J Innate Immun 2023; 16:33-44. [PMID: 38071977 PMCID: PMC10776137 DOI: 10.1159/000535637] [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: 07/17/2023] [Accepted: 12/01/2023] [Indexed: 01/11/2024] Open
Abstract
INTRODUCTION C-X-C motif chemokine ligand 1 (CXCL1) is a potent neutrophil chemoattractant that plays a pivotal role in recruiting neutrophils during inflammatory conditions. This study explored the role of CXCL1 in modulating the gut microbiota, influencing neutrophil infiltration, and contributing to the development of colitis. METHODS We employed quantitative PCR to assess CXCL1 expression in colon samples. A mouse model of dextran sulfate sodium (DSS)-induced colitis was utilized to explore the progression of colitis in wild-type (WT) and CXCL1-deficient (CXCL1-/-) mice. RESULTS Colitis attenuation was evident in CXCL1-/- mice. Significant alterations were observed in the gut microbiome, as revealed by 16S rRNA gene sequencing. Furthermore, CXCL1-/- mice exhibited reduced gut permeability and diminished endotoxin levels in peripheral blood following DSS treatment compared to WT mice. In response to DSS treatment, WT mice showed a clear increase in neutrophil infiltration, while CXCL1-/- mice exhibited lower levels of infiltration. Fecal microbiota transplantation (FMT) using stools from CXCL1-/- mice alleviated DSS-induced colitis. Interestingly, FMT from patients with colitis increased CXCL1 and Ly6G expression in the colons of gut-sterilized mice. Clinical data analysis revealed elevated CXCL1 and CD15 expression in patients with colitis, with a positive correlation between the severity of colitis and the expression of CXCL1 and CD15. CONCLUSION These findings shed light on the pivotal role of CXCL1 in promoting colitis by modulating the gut microbiota.
Collapse
Affiliation(s)
- Hang Zhao
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Wenhua Li
- Department of Gastroenterology, Jiading Branch of Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Zhou
- Department of Gastroenterology, Jintan Hospital, Jiangsu University, Changzhou, China
| | - Liang Pan
- Department of Gastroenterology, Jintan Hospital, Jiangsu University, Changzhou, China
| | - Yun Feng
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Pingyu Gao
- Pathology Center, Jintan Hospital, Jiangsu University, Zhenjiang, China
| | - Jie Ji
- Clinical Laboratory, Jintan Hospital, Jiangsu University, Zhenjiang, China
| | - Huanyan Zhang
- Clinical Laboratory, Jintan Hospital, Jiangsu University, Zhenjiang, China
| | - Kai Zhao
- Department of Gastroenterology, Jintan Hospital, Jiangsu University, Changzhou, China
| | - Chi Wang
- Department of Precision Mechanical Engineering, Shanghai University, Shanghai, China
| | - Zhanjun Lu
- Department of Gastroenterology, Jintan Hospital, Jiangsu University, Changzhou, China
- Department of Gastroenterology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
19
|
Li W, Cheng F, Zhang J, Li C, Yu D, Simayijiang H, Liu H, Li S, Yan J. Changes in Gut Microbiota and Metabolites in Papillary Thyroid Carcinoma Patients Following Radioactive Iodine Therapy. Int J Gen Med 2023; 16:4453-4464. [PMID: 37808207 PMCID: PMC10557971 DOI: 10.2147/ijgm.s433433] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 09/28/2023] [Indexed: 10/10/2023] Open
Abstract
Purpose Radioactive iodine therapy is administered through oral route, which is accumulated and absorbed in the intestine. However, its effects on the intestine remain unclear. In this study, we investigated the changes in the gut microbiota and metabolites following radioactive iodine therapy. Patients and Methods A total of 76 stool samples from the same 38 patients were collected at the start of radioactive iodine therapy and three days following the therapy. Stool microbiota and metabolites were detected using 16S rRNA gene sequencing and liquid chromatography-mass spectrometry. Results Enterobacteriales, Enterobacteriaceae and Escherichia-Shigella were elevated in most patients (27/38) following the therapy. The levels of 2-hydroxyundec-7-enoylcarnitine were significantly lower, whereas those of 5-dehydroavenasterol, butylisopropylamine, and salsoline-1-carboxylate were higher following the therapy. The relative abundance of Escherichia-Shigella was negatively correlated with 2-hydroxyundec-7-enoylcarnitine level (r2 = -0.661, P = 0.009). Functional pathways were predicted to be involved in amino acid and lipid metabolism following the therapy. Particularly, phenylalanine, linoleic acid, sphingolipid, purine, and alpha-linolenic acid metabolism were the main metabolic pathways. Conclusion Gut microbiota was disturbed following radioactive iodine therapy, with increased Escherichia-Shigella. Processes associated with energy production seems to be impacted following the therapy, with significantly decreased 2-hydroxyundec-7-enoylcarnitine level. Meanwhile, some metabolites and functional pathways may have a positive effect on intestinal homeostasis, and may be related to the repair and promotion of gut recovery following the therapy. This study provides a basic foundation to explore how radioactive iodine affects gut microbiota and metabolites, and how gut function is regulated in response to radioactive iodine therapy.
Collapse
Affiliation(s)
- Wanting Li
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Feng Cheng
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Jun Zhang
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Caihong Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Daijing Yu
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Halimureti Simayijiang
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| | - Haiyan Liu
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Sijin Li
- Department of Nuclear Medicine, First Hospital of Shanxi Medical University, Taiyuan, People’s Republic of China
| | - Jiangwei Yan
- Shanxi Key Laboratory of Forensic Medicine, Shanxi Medical University, Jinzhong, People’s Republic of China
| |
Collapse
|
20
|
Shukla PK, Rao RG, Meena AS, Giorgianni F, Lee SC, Raju P, Shashikanth N, Shekhar C, Beranova S, Balazs L, Tigyi G, Gosain A, Rao R. Paneth cell dysfunction in radiation injury and radio-mitigation by human α-defensin 5. Front Immunol 2023; 14:1174140. [PMID: 37638013 PMCID: PMC10448521 DOI: 10.3389/fimmu.2023.1174140] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023] Open
Abstract
Introduction The mechanism underlying radiation-induced gut microbiota dysbiosis is undefined. This study examined the effect of radiation on the intestinal Paneth cell α-defensin expression and its impact on microbiota composition and mucosal tissue injury and evaluated the radio-mitigative effect of human α-defensin 5 (HD5). Methods Adult mice were subjected to total body irradiation, and Paneth cell α-defensin expression was evaluated by measuring α-defensin mRNA by RT-PCR and α-defensin peptide levels by mass spectrometry. Vascular-to-luminal flux of FITC-inulin was measured to evaluate intestinal mucosal permeability and endotoxemia by measuring plasma lipopolysaccharide. HD5 was administered in a liquid diet 24 hours before or after irradiation. Gut microbiota was analyzed by 16S rRNA sequencing. Intestinal epithelial junctions were analyzed by immunofluorescence confocal microscopy and mucosal inflammatory response by cytokine expression. Systemic inflammation was evaluated by measuring plasma cytokine levels. Results Ionizing radiation reduced the Paneth cell α-defensin expression and depleted α-defensin peptides in the intestinal lumen. α-Defensin down-regulation was associated with the time-dependent alteration of gut microbiota composition, increased gut permeability, and endotoxemia. Administration of human α-defensin 5 (HD5) in the diet 24 hours before irradiation (prophylactic) significantly blocked radiation-induced gut microbiota dysbiosis, disruption of intestinal epithelial tight junction and adherens junction, mucosal barrier dysfunction, and mucosal inflammatory response. HD5, administered 24 hours after irradiation (treatment), reversed radiation-induced microbiota dysbiosis, tight junction and adherens junction disruption, and barrier dysfunction. Furthermore, HD5 treatment also prevents and reverses radiation-induced endotoxemia and systemic inflammation. Conclusion These data demonstrate that radiation induces Paneth cell dysfunction in the intestine, and HD5 feeding prevents and mitigates radiation-induced intestinal mucosal injury, endotoxemia, and systemic inflammation.
Collapse
Affiliation(s)
- Pradeep K. Shukla
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Roshan G. Rao
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Avtar S. Meena
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Francesco Giorgianni
- College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sue Chin Lee
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Preeti Raju
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Nitesh Shashikanth
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Chandra Shekhar
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Sarka Beranova
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Louisa Balazs
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Gabor Tigyi
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - Ankush Gosain
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| | - RadhaKrishna Rao
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN, United States
| |
Collapse
|
21
|
Guan B, Li D, Meng A. Development of radiation countermeasure agents for acute radiation syndromes. Animal Model Exp Med 2023; 6:329-336. [PMID: 37642199 PMCID: PMC10486342 DOI: 10.1002/ame2.12339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Accepted: 07/18/2023] [Indexed: 08/31/2023] Open
Abstract
The risk of internal and external exposure to ionizing radiation (IR) has increased alongside the development and implementation of nuclear technology. Therefore, serious security issues have emerged globally, and there has been an increase in the number of studies focusing on radiological prevention and medical countermeasures. Radioprotective drugs are particularly important components of emergency medical preparedness strategies for the clinical management of IR-induced injuries. However, a few drugs have been approved to date to treat such injuries, and the related mechanisms are not entirely understood. Thus, the aim of the present review was to provide a brief overview of the World Health Organization's updated list of essential medicines for 2023 for the proper management of national stockpiles and the treatment of radiological emergencies. This review also discusses the types of radiation-induced health injuries and the related mechanisms, as well as the development of various radioprotective agents, including Chinese herbal medicines, for which significant survival benefits have been demonstrated in animal models of acute radiation syndrome.
Collapse
Affiliation(s)
- Bowen Guan
- National Human Diseases Animal Model Resource Center, NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal Sciences Chinese Academy of Medical Sciences (CAMS), Peking Union Medical College (PUMC), National Center of Technology Innovation for Animal ModelBeijingChina
| | - Deguan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear MedicineInstitute of Radiation Medicine, Chinese Academy of Medical Science, Peking Union Medical CollegeTianjinChina
| | - Aimin Meng
- National Human Diseases Animal Model Resource Center, NHC Key Laboratory of Human Disease Comparative Medicine, Beijing Engineering Research Center for Experimental Animal Models of Human Critical DiseasesInstitute of Laboratory Animal Sciences Chinese Academy of Medical Sciences (CAMS), Peking Union Medical College (PUMC), National Center of Technology Innovation for Animal ModelBeijingChina
| |
Collapse
|
22
|
Shi YZ, Tao QF, Qin HY, Li Y, Zheng H. Causal relationship between gut microbiota and urticaria: a bidirectional two-sample mendelian randomization study. Front Microbiol 2023; 14:1189484. [PMID: 37426010 PMCID: PMC10324650 DOI: 10.3389/fmicb.2023.1189484] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Accepted: 06/08/2023] [Indexed: 07/11/2023] Open
Abstract
Background Cumulative evidence showed an association between gut microbiota and urticaria, but the causal relationship between them is unclear. We aimed to verify whether there is a causal relationship between the composition of gut microbiota and urticaria and explore whether the causal effect was bidirectional. Methods We obtained genome-wide association studies (GWAS) summary data of 211 gut microbiota and urticaria from the most extensive available GWAS database. A bidirectional two-sample mendelian randomization (MR) study was used to test the causal relationship between the gut microbiota and urticaria. The MR analysis was primarily performed with the inverse variance weighted (IVW) method, and MR-Egger, weighted median (WM), and MR-PRESSO were performed as sensitivity analyses. Results The Phylum Verrucomicrobia (OR 1.27, 95%CI 1.01 to 1.61; p = 0.04), Genus Defluviitaleaceae UCG011 (OR 1.29, 95%CI 1.04 to 1.59; p = 0.02), and Genus Coprococcus 3 (OR 1.44, 95%CI 1.02 to 2.05; p = 0.04) was a risk effect against urticaria. And Order Burkholderiales (OR 0.68, 95%CI 0.49 to 0.99; p = 0.04) and Genus Eubacterium xylanophilum group (OR 0.78, 95%CI 0.62 to 0.99; p = 0.04) were negatively associated with urticaria, suggesting a protective effect. At the same time, urticaria had a positively causal effect on gut microbiota (Genus Eubacterium coprostanoligenes group) (OR 1.08, 95%CI 1.01 to 1.16; p = 0.02). These findings showed no influence by heterogeneity or horizontal pleiotropy. Moreover, most sensitivity analyses showed results consistent with those of IVW analysis. Conclusion Our MR study confirmed the potential causal relationship between gut microbiota and urticaria, and the causal effect was bidirectional. Nevertheless, these findings warrant further examination owing to the unclear mechanisms.
Collapse
|
23
|
Zhang D, He J, Cui J, Wang R, Tang Z, Yu H, Zhou M. Oral Microalgae-Nano Integrated System against Radiation-Induced Injury. ACS NANO 2023; 17:10560-10576. [PMID: 37253200 DOI: 10.1021/acsnano.3c01502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The increasing applications of ionizing radiation in society raise the risk of radiation-induced intestinal and whole-body injury. Astaxanthin is a powerful antioxidant to reduce the reactive oxygen generated from radiation and the subsequent damage. However, the oral administration of astaxanthin remains challenging owing to its low solubility and poor bioavailability. Herein, we facilely construct an orally used microalgae-nano integrated system (SP@ASXnano) against radiation-induced intestinal and whole-body injury, combining natural microalgae Spirulina platensis (SP) with astaxanthin nanoparticles (ASXnano). SP and ASXnano show complementation in drug delivery to improve distribution in the intestine and blood. SP displays limited gastric drug loss, prolonged intestinal retention, constant ASXnano release, and progressive degradation. ASXnano improves drug solubility, gastric stability, cell uptake, and intestinal absorption. SP and ASXnano have synergy in many aspects such as anti-inflammation, microbiota protection, and fecal short-chain fatty acid up-regulation. In addition, the system is ensured with biosafety for long-term administration. The system organically combines the properties of microalgae and nanoparticles, which was expected to expand the medical application of SP as a versatile drug delivery platform.
Collapse
Affiliation(s)
- Dongxiao Zhang
- Department of Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jian He
- Department of Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jiarong Cui
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Ruoxi Wang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zhe Tang
- Department of Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Hongyu Yu
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Min Zhou
- Department of Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| |
Collapse
|
24
|
Yi Y, Lu W, Shen L, Wu Y, Zhang Z. The gut microbiota as a booster for radiotherapy: novel insights into radio-protection and radiation injury. Exp Hematol Oncol 2023; 12:48. [PMID: 37218007 DOI: 10.1186/s40164-023-00410-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Accepted: 05/04/2023] [Indexed: 05/24/2023] Open
Abstract
Approximately 60-80% of cancer patients treated with abdominopelvic radiotherapy suffer post-radiotherapy toxicities including radiation enteropathy and myelosuppression. Effective preventive and therapeutic strategies are lacking for such radiation injury. The gut microbiota holds high investigational value for deepening our understanding of the pathogenesis of radiation injury, especially radiation enteropathy which resembles inflammatory bowel disease pathophysiology and for facilitating personalized medicine by providing safer therapies tailored for cancer patients. Preclinical and clinical data consistently support that gut microbiota components including lactate-producers, SCFA-producers, indole compound-producers and Akkermansia impose intestinal and hematopoietic radio-protection. These features serve as potential predictive biomarkers for radiation injury, together with the microbial diversity which robustly predicts milder post-radiotherapy toxicities in multiple types of cancer. The accordingly developed manipulation strategies including selective microbiota transplantation, probiotics, purified functional metabolites and ligands to microbe-host interactive pathways are promising radio-protectors and radio-mitigators that merit extensive validation in clinical trials. With massive mechanistic investigations and pilot clinical trials reinforcing its translational value the gut microbiota may boost the prediction, prevention and mitigation of radiation injury. In this review, we summarize the state-of-the-art landmark researches related with radio-protection to provide illuminating insights for oncologists, gastroenterologists and laboratory scientists interested in this overlooked complexed disorder.
Collapse
Affiliation(s)
- Yuxi Yi
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Weiqing Lu
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China
| | - Lijun Shen
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China.
| | - Yang Wu
- Key Laboratory of Medical Molecular Virology (MOE/NHC/CAMS), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Zhen Zhang
- Department of Radiation Oncology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
- Shanghai Clinical Research Center for Radiation Oncology, Shanghai, China.
- Shanghai Key Laboratory of Radiation Oncology, Shanghai, China.
| |
Collapse
|
25
|
Feng L, Bao T, Bai L, Mu X, Ta N, Bao M, Li Y, Zhang J, Fu M, Chen Y. Mongolian medicine formulae Ruda-6 alleviates indomethacin-induced gastric ulcer by regulating gut microbiome and serum metabolomics in rats. JOURNAL OF ETHNOPHARMACOLOGY 2023; 314:116545. [PMID: 37196816 DOI: 10.1016/j.jep.2023.116545] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 01/16/2023] [Accepted: 04/21/2023] [Indexed: 05/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ruda-6 (RD-6), a typical traditional Mongolian medicine formulae consisting of 6 herbs, has been traditionally used in treating gastric disorders. Even though it has been shown to protect against gastric ulcers (GU) in animal models, the gut microbiome and serum metabololite-related mechanisms that prevent GU are not well understood. AIM OF THE STUDY This study was conducted to evaluate the gastroprotective mechanism of RD-6 associated with the alteration of the gut microbiome and serum metabolic profiles in GU rats. MATERIALS AND METHODS RD-6 (0.27, 1.35 and 2.7 g/kg) or ranitidine (40 mg/kg) were orally administered in rats for three weeks before the induction of gastric ulcer using indomethacin (30 mg/kg, single oral dose). The gastric ulcer index, ulcer area, H&E staining, and the levels of TNF-α, iNOS, MPO and MDA were quantified to evaluate the ulcer inhibitory effects of RD-6. Then, 16S rRNA gene sequencing combined with LC-MS metabolic profiling was performed to investigate the effect of RD-6 on the gut microbiota and serum metabolites in rats. Moreover, a spearman analysis was used to calculate the correlation coefficient between the different microbiota and the metabolites. RESULTS RD-6 inhibited the gastric lesion damage caused by indomethacin in rats, decreased the ulcer index by 50.29% (p < 0.05), reduced the levels of TNF-α, iNOS, MDA and MPO in gastric tissue. Additionally, RD-6 reshaped the diversity and microbial composition, and reversed the reduced bacteria including [Eubacterium]_xylanophilum group, Sellimonas, Desulfovibrio, and UCG-009, and the increased bacteria Aquamicrobium caused by indomethacin induction. Furthermore, RD-6 regulated the levels of metabolites including amino acids and organic acids, and these affected metabolites were involved in taurine and hypotaurine metabolism and tryptophan metabolism. Spearman analysis revealed that the perturbed gut microbiota were closely related to the changes in differential serum metabolites. CONCLUSION In view of the 16S rRNA gene sequencing and LC-MS metabolic results, the present study suggests the mechanism of RD-6 ameliorating GU via modulating intestinal microbiota and their metabolites.
Collapse
Affiliation(s)
- Lan Feng
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Terigele Bao
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China
| | - Laxinamujila Bai
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China
| | - Xiyele Mu
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China
| | - Na Ta
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China
| | - Minglan Bao
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China
| | - Yonghui Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Junqing Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China
| | - Minghai Fu
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China; Key Laboratory of Tropical Translational Medicine of Ministry of Education, Hainan Provincial Key Laboratory for Research and Development of Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, China.
| | - Yongsheng Chen
- NMPA Key Laboratory for Quality Control of Traditional Chinese Medicine (Mongolian Medicine), School of Mongolian Medicine, Inner Mongolia Minzu University, Tongliao, China.
| |
Collapse
|
26
|
Liu Y, Wang C, Liu R, Zhao M, Ding X, Zhang T, He R, Zhu S, Dong X, Xie J, Gu Z, Zhao Y. Adhesive Ergothioneine Hyaluronate Gel Protects against Radiation Gastroenteritis by Alleviating Apoptosis, Inflammation, and Gut Microbiota Dysbiosis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:19833-19846. [PMID: 37052616 DOI: 10.1021/acsami.2c23142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Radiation gastroenteritis represents one of the most prevalent and hazardous complications of abdominopelvic radiotherapy, which not only severely reduces patients' life quality but also restricts radiotherapy efficacy. However, there is currently no clinically available oral radioprotector for this threatening disease due to its complex pathogenesis and the harsh gastrointestinal environment. To this end, this study developed a facile but effective oral radioprotector, ergothioneine hyaluronate (EGT@HA) gel, protecting against radiation gastroenteritis by synergistically regulating oxidative stress, inflammation, and gut microbiota. In vitro and cellular experiments verified the chemical stability and free radical scavenging ability of EGT and its favorable cellular radioprotective efficacy by inhibiting intracellular reactive oxidative species (ROS) generation, DNA damage, mitochondrial damage, and apoptosis. At the in vivo level, EGT@HA with prolonged gastrointestinal residence mitigated radiation-induced gastrointestinal tissue injury, apoptosis, neutrophil infiltration, and gut flora dysbiosis. For the first time, this work investigated the protective effects of EGT@HA gel on radiation gastroenteritis, which not only hastens the advancement of the novel gastrointestinal radioprotector but also provides a valuable gastrointestinal radioprotection paradigm by synergistically modulating oxidative stress, inflammation, and gut microbiota disturbance.
Collapse
Affiliation(s)
- Yaping Liu
- The First Affiliated Hospital of University of Science and Technology of China, Hefei 230001, Anhui, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Chengyan Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Ruixue Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Maoru Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Xuefeng Ding
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Tingjun Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Rendong He
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Shuang Zhu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Xinghua Dong
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
| | - Jiani Xie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- China School of Food and Biological Engineering, Chengdu University, Chengdu 610106, Sichuan, China
| | - Zhanjun Gu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, Institute of High Energy Physics and National Center for Nanoscience and Technology, Chinese Academy of Sciences, Beijing 100049, China
- College of Materials Science and Optoelectronic Technology, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
27
|
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: 8] [Impact Index Per Article: 8.0] [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
|
28
|
Yang Q, Qin B, Hou W, Qin H, Yin F. Pathogenesis and therapy of radiation enteritis with gut microbiota. Front Pharmacol 2023; 14:1116558. [PMID: 37063268 PMCID: PMC10102376 DOI: 10.3389/fphar.2023.1116558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 03/07/2023] [Indexed: 04/03/2023] Open
Abstract
Radiotherapy is widely used in clinic due to its good effect for cancer treatment. But radiotherapy of malignant tumors in the abdomen and pelvis is easy to cause radiation enteritis complications. Gastrointestinal tract contains numerous microbes, most of which are mutualistic relationship with the host. Abdominal radiation results in gut microbiota dysbiosis. Microbial therapy can directly target gut microbiota to reverse microbiota dysbiosis, hence relieving intestinal inflammation. In this review, we mainly summarized pathogenesis and novel therapy of the radiation-induced intestinal injury with gut microbiota dysbiosis and envision the opportunities and challenges of radiation enteritis therapy.
Collapse
Affiliation(s)
- Qilin Yang
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- School of Clinical Medicine of Nanjing Medical University, Nanjing, China
| | - Bingzhi Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
| | - Weiliang Hou
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- Shanghai Cancer Institute, Renji Hospital School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| | - Huanlong Qin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| | - Fang Yin
- Research Institute of Intestinal Diseases, Shanghai Tenth People’s Hospital, Tongji University, Shanghai, China
- *Correspondence: Weiliang Hou, ; Huanlong Qin, ; Fang Yin,
| |
Collapse
|
29
|
Zheng Z, Hu Y, Tang J, Xu W, Zhu W, Zhang W. The implication of gut microbiota in recovery from gastrointestinal surgery. Front Cell Infect Microbiol 2023; 13:1110787. [PMID: 36926517 PMCID: PMC10011459 DOI: 10.3389/fcimb.2023.1110787] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 02/15/2023] [Indexed: 03/08/2023] Open
Abstract
Recovery from gastrointestinal (GI) surgery is often interrupted by the unpredictable occurrence of postoperative complications, including infections, anastomotic leak, GI dysmotility, malabsorption, cancer development, and cancer recurrence, in which the implication of gut microbiota is beginning to emerge. Gut microbiota can be imbalanced before surgery due to the underlying disease and its treatment. The immediate preparations for GI surgery, including fasting, mechanical bowel cleaning, and antibiotic intervention, disrupt gut microbiota. Surgical removal of GI segments also perturbs gut microbiota due to GI tract reconstruction and epithelial barrier destruction. In return, the altered gut microbiota contributes to the occurrence of postoperative complications. Therefore, understanding how to balance the gut microbiota during the perioperative period is important for surgeons. We aim to overview the current knowledge to investigate the role of gut microbiota in recovery from GI surgery, focusing on the crosstalk between gut microbiota and host in the pathogenesis of postoperative complications. A comprehensive understanding of the postoperative response of the GI tract to the altered gut microbiota provides valuable cues for surgeons to preserve the beneficial functions and suppress the adverse effects of gut microbiota, which will help to enhance recovery from GI surgery.
Collapse
Affiliation(s)
| | | | | | | | | | - Wei Zhang
- Department of General Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
30
|
Yuan H, Gui R, Wang Z, Fang F, Zhao H. Gut microbiota: A novel and potential target for radioimmunotherapy in colorectal cancer. Front Immunol 2023; 14:1128774. [PMID: 36798129 PMCID: PMC9927011 DOI: 10.3389/fimmu.2023.1128774] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Accepted: 01/17/2023] [Indexed: 02/04/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers, with a high mortality rate, and is a major burden on human health worldwide. Gut microbiota regulate human immunity and metabolism through producing numerous metabolites, which act as signaling molecules and substrates for metabolic reactions in various biological processes. The importance of host-gut microbiota interactions in immunometabolic mechanisms in CRC is increasingly recognized, and interest in modulating the microbiota to improve patient's response to therapy has been raising. However, the specific mechanisms by which gut microbiota interact with immunotherapy and radiotherapy remain incongruent. Here we review recent advances and discuss the feasibility of gut microbiota as a regulatory target to enhance the immunogenicity of CRC, improve the radiosensitivity of colorectal tumor cells and ameliorate complications such as radiotoxicity. Currently, great breakthroughs in the treatment of non-small cell lung cancer and others have been achieved by radioimmunotherapy, but radioimmunotherapy alone has not been effective in CRC patients. By summarizing the recent preclinical and clinical evidence and considering regulatory roles played by microflora in the gut, such as anti-tumor immunity, we discuss the potential of targeting gut microbiota to enhance the efficacy of radioimmunotherapy in CRC and expect this review can provide references and fresh ideas for the clinical application of this novel strategy.
Collapse
Affiliation(s)
- Hanghang Yuan
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China,National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Ruirui Gui
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China,National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Zhicheng Wang
- National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China
| | - Fang Fang
- National Health Commission (NHC) Key laboratory of Radiobiology, School of Public Health, Jilin University, Changchun, China,*Correspondence: Fang Fang, ; Hongguang Zhao,
| | - Hongguang Zhao
- Department of Nuclear Medicine, The First Hospital of Jilin University, Changchun, China,*Correspondence: Fang Fang, ; Hongguang Zhao,
| |
Collapse
|
31
|
Cook JA, Sowers AL, Choudhuri R, Gadisetti C, Edmondson EF, Gohain S, Krishna MC, Mitchell JB. The effect of modulation of gut microbiome profile on radiation-induced carcinogenesis and survival. JOURNAL OF RADIATION RESEARCH 2023; 64:24-32. [PMID: 36253079 PMCID: PMC9855309 DOI: 10.1093/jrr/rrac062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 08/04/2022] [Indexed: 06/16/2023]
Abstract
Non-lethal doses of ionizing radiation (IR) delivered to humans because of terrorist events, nuclear accidents or radiotherapy can result in carcinogenesis. Means of protecting against carcinogenesis are lacking. We questioned the role of the gut microbiome in IR-induced carcinogenesis. The gut microbiome was modulated by administering broad spectrum antibiotics (Ab) in the drinking water. Mice were given Ab 3 weeks before and 3 weeks after 3 Gy total body irradiation (TBI) or for 6 weeks one month after TBI. Three weeks of Ab treatment resulted in a 98% reduction in total 16S rRNA counts for 4 out of 6 of the phylum groups detected. However, 3 more weeks of Ab treatment (6 weeks total) saw an expansion in the phylum groups Proteobacteria and Actinobacteria. The Ab treatment altered the bacteria diversity in the gut, and shortened the lifespan when Ab were administered before and after TBI. Mortality studies indicated that the adverse Ab lifespan effects were due to a decrease in the time in which solid tumors started to appear and not to any changes in hematopoietic or benign tumors. In contrast, when Ab were administered one month after TBI, lifespan was unchanged compared to the control TBI group. Use of broad-spectrum antibiotics to simulate the germ-free condition did not afford an advantage on carcinogenesis or lifespan.
Collapse
Affiliation(s)
- John A Cook
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892, USA
| | - Anastasia L Sowers
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892, USA
| | - Rajani Choudhuri
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892, USA
| | | | - Elijah F Edmondson
- Molecular Histopathology Laboratory, Frederick National Laboratory for Cancer Research, Frederick, MD 21702, USA
| | - Sangeeta Gohain
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892, USA
| | - Murali C Krishna
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892, USA
| | - James B Mitchell
- Radiation Biology Branch, Center for Cancer Research, National Cancer Institute, Bethesda MD 20892, USA
| |
Collapse
|
32
|
Nista EC, Del Gaudio A, Del Vecchio LE, Mezza T, Pignataro G, Piccioni A, Gasbarrini A, Franceschi F, Candelli M. Pancreatic Cancer Resistance to Treatment: The Role of Microbiota. Biomedicines 2023; 11:biomedicines11010157. [PMID: 36672664 PMCID: PMC9856157 DOI: 10.3390/biomedicines11010157] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/30/2022] [Accepted: 01/04/2023] [Indexed: 01/11/2023] Open
Abstract
Pancreatic cancer (PC) is an aggressive malignancy and the fourth leading cause of cancer death in the United States and Europe. It is estimated that PC will be the second leading cause of cancer death by 2030. In addition to late diagnosis, treatment resistance is a major cause of shortened survival in pancreatic cancer. In this context, there is growing evidence that microbes play a regulatory role, particularly in therapy resistance and in creating a microenvironment in the tumor, that favors cancer progression. The presence of certain bacteria belonging to the gamma-proteobacteria or mycoplasmas appears to be associated with both pharmacokinetic and pharmacodynamic changes. Recent evidence suggests that the microbiota may also play a role in resistance mechanisms to immunotherapy and radiotherapy. However, the interactions between microbiota and therapy are bilateral and modulate therapy tolerance. Future perspectives are increasingly focused on elucidating the role of the microbiota in tumorigenesis and processes of therapy resistance, and a better understanding of these mechanisms may provide important opportunities to improve survival in these patients.
Collapse
Affiliation(s)
- Enrico Celestino Nista
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Angelo Del Gaudio
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Livio Enrico Del Vecchio
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Teresa Mezza
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giulia Pignataro
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Andrea Piccioni
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Antonio Gasbarrini
- Medical and Surgical Science Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Francesco Franceschi
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Marcello Candelli
- Emergency Medicine Department, Fondazione Policlinico Universitario Agostino Gemelli—IRCCS, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
- Correspondence: ; Tel.: +0039-063-0153-188
| |
Collapse
|
33
|
Hua Q, Zhang H, Xu R, Tian C, Gao T, Yuan Y, Han Y, Li Y, Qi C, Zhong F, Ma A. Lacticaseibacillus casei ATCC334 Ameliorates Radiation-Induced Intestinal Injury in Rats by Targeting Microbes and Metabolites. Mol Nutr Food Res 2023; 67:e2200337. [PMID: 36408889 DOI: 10.1002/mnfr.202200337] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/13/2022] [Indexed: 11/22/2022]
Abstract
SCOPE Gastrointestinal side effects are frequently observed in patients receiving medical radiation therapy. As Lacticaseibacillus casei ATCC334 potentially affects microbial ecosystem, the study hypothesizes that it may improve radiation-induced intestinal injury in rats by modulating the "gut microbiota-metabolite-barrier axis." METHODS AND RESULTS Rats are fed one of three or no doses of L. casei ATCC334 for 7 days and then expose to a single dose of 9 Gy X-ray total abdominal irradiation. Supplementation with L. casei ATCC334 promote the proliferation of intestinal stem cells (ISCs), increase the expression of tight junction proteins, reduce intestinal permeability, and protect intestinal barrier integrity. Moreover, 16S rRNA sequencing show that medium and high doses of L. casei ATCC334 inhibit the growth of Escherichia/Shigella and favor Akkermansia proliferation. L. casei ATCC334 intervention reprogram the metabolic profile and inhibit putrescine production but promote alpha-linolenic acid (ALA) production. Notably, a decrease in putrescine and an increase in ALA are significantly correlated with the proliferation of ISCs and enhanced intestinal barrier function following L. casei ATCC334 intervention. CONCLUSION These results highlight that medium and high doses of L. casei ATCC334 alleviate radiation-induced intestinal damage by enhancing the mucosal barrier and remodeling the gut microbiota structure and metabolic activity.
Collapse
Affiliation(s)
- Qinglian Hua
- School of Public health, Qingdao University, Qingdao, 266071, China
| | - Haowen Zhang
- Institute of Nutrition & Health, Qingdao University, Qingdao, 266071, China
| | - Rui Xu
- Nanan District Center for Disease Control and Prevention, Chongqing, 400000, China
| | | | - Tianlin Gao
- School of Public health, Qingdao University, Qingdao, 266071, China
| | - Yanlei Yuan
- School of Public health, Qingdao University, Qingdao, 266071, China
| | - Yaling Han
- School of Public health, Qingdao University, Qingdao, 266071, China
| | - Yue Li
- School of Public health, Qingdao University, Qingdao, 266071, China
| | - Ce Qi
- Institute of Nutrition & Health, Qingdao University, Qingdao, 266071, China
| | - Feng Zhong
- School of Public health, Qingdao University, Qingdao, 266071, China
| | - Aiguo Ma
- Institute of Nutrition & Health, Qingdao University, Qingdao, 266071, China
| |
Collapse
|
34
|
Gut microbiota: a potential target for improved cancer therapy. J Cancer Res Clin Oncol 2023; 149:541-552. [PMID: 36550389 DOI: 10.1007/s00432-022-04546-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/16/2022] [Indexed: 12/24/2022]
Abstract
Drug resistance and toxicity are major challenges observed during cancer treatment. In recent years, gut microbiota has been found to be strongly associated with the efficacy, toxicity, and side effects of chemotherapy, radiotherapy, and immunotherapy. Both preclinical studies and clinical trials have demonstrated the potential of microbiota modulation for cancer treatment. The human gut microbiota has exciting prospects for developing biomarkers to predict the outcome of cancer treatment. Moreover, multiple approaches can alter the gut microbiota composition, including faecal microbiota transplantation (FMT), probiotics, antibiotics (ATB), and diet. We describe the mechanisms by which the gut microbiota influences the efficacy and toxicity of cancer therapy, disease-related biomarkers, and methods to target the gut microbiota to improve outcomes. The purpose of this review is to provide new ideas for optimising cancer therapy by providing up-to-date information on the relationship between gut microbiota and cancer therapy, and hopes to find new targets for cancer treatment from human microbiota.
Collapse
|
35
|
Sgro M, Iacono G, Yamakawa GR, Kodila ZN, Marsland BJ, Mychasiuk R. Age matters: Microbiome depletion prior to repeat mild traumatic brain injury differentially alters microbial composition and function in adolescent and adult rats. PLoS One 2022; 17:e0278259. [PMID: 36449469 PMCID: PMC9710846 DOI: 10.1371/journal.pone.0278259] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 11/13/2022] [Indexed: 12/02/2022] Open
Abstract
Dysregulation of the gut microbiome has been shown to perpetuate neuroinflammation, alter intestinal permeability, and modify repetitive mild traumatic brain injury (RmTBI)-induced deficits. However, there have been no investigations regarding the comparative effects that the microbiome may have on RmTBI in adolescents and adults. Therefore, we examined the influence of microbiome depletion prior to RmTBI on microbial composition and metabolome, in adolescent and adult Sprague Dawley rats. Rats were randomly assigned to standard or antibiotic drinking water for 14 days, and to subsequent sham or RmTBIs. The gut microbiome composition and metabolome were analysed at baseline, 1 day after the first mTBI, and at euthanasia (11 days following the third mTBI). At euthanasia, intestinal samples were also collected to quantify tight junction protein (TJP1 and occludin) expression. Adolescents were significantly more susceptible to microbiome depletion via antibiotic administration which increased pro-inflammatory composition and metabolites. Furthermore, RmTBI induced a transient increase in 'beneficial bacteria' (Lachnospiraceae and Faecalibaculum) in only adolescents that may indicate compensatory action in response to the injury. Finally, microbiome depletion prior to RmTBI generated a microbiome composition and metabolome that exemplified a potentially chronic pathogenic and inflammatory state as demonstrated by increased Clostridium innocuum and Erysipelatoclostridium and reductions in Bacteroides and Clostridium Sensu Stricto. Results highlight that adolescents are more vulnerable to RmTBI compared to adults and dysbiosis prior to injury may exacerbate secondary inflammatory cascades.
Collapse
Affiliation(s)
- Marissa Sgro
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Giulia Iacono
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Glenn R. Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Zoe N. Kodila
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Benjamin J. Marsland
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
36
|
Wang L, Wang S, Zhang Q, He C, Fu C, Wei Q. The role of the gut microbiota in health and cardiovascular diseases. MOLECULAR BIOMEDICINE 2022; 3:30. [PMID: 36219347 PMCID: PMC9554112 DOI: 10.1186/s43556-022-00091-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/05/2022] [Indexed: 11/17/2022] Open
Abstract
The gut microbiota is critical to human health, such as digesting nutrients, forming the intestinal epithelial barrier, regulating immune function, producing vitamins and hormones, and producing metabolites to interact with the host. Meanwhile, increasing evidence indicates that the gut microbiota has a strong correlation with the occurrence, progression and treatment of cardiovascular diseases (CVDs). In patients with CVDs and corresponding risk factors, the composition and ratio of gut microbiota have significant differences compared with their healthy counterparts. Therefore, gut microbiota dysbiosis, gut microbiota-generated metabolites, and the related signaling pathway may serve as explanations for some of the mechanisms about the occurrence and development of CVDs. Several studies have also demonstrated that many traditional and latest therapeutic treatments of CVDs are associated with the gut microbiota and its generated metabolites and related signaling pathways. Given that information, we summarized the latest advances in the current research regarding the effect of gut microbiota on health, the main cardiovascular risk factors, and CVDs, highlighted the roles and mechanisms of several metabolites, and introduced corresponding promising treatments for CVDs regarding the gut microbiota. Therefore, this review mainly focuses on exploring the role of gut microbiota related metabolites and their therapeutic potential in CVDs, which may eventually provide better solutions in the development of therapeutic treatment as well as the prevention of CVDs.
Collapse
Affiliation(s)
- Lu Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Shiqi Wang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Qing Zhang
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chengqi He
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| | - Chenying Fu
- grid.412901.f0000 0004 1770 1022National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,grid.412901.f0000 0004 1770 1022Aging and Geriatric Mechanism Laboratory, West China Hospital, Sichuan University, Chengdu, People’s Republic of China
| | - Quan Wei
- grid.412901.f0000 0004 1770 1022Rehabilitation Medicine Center and Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, People’s Republic of China ,Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, People’s Republic of China
| |
Collapse
|
37
|
Biţă A, Scorei IR, Bălşeanu TA, Ciocîlteu MV, Bejenaru C, Radu A, Bejenaru LE, Rău G, Mogoşanu GD, Neamţu J, Benner SA. New Insights into Boron Essentiality in Humans and Animals. Int J Mol Sci 2022; 23:ijms23169147. [PMID: 36012416 PMCID: PMC9409115 DOI: 10.3390/ijms23169147] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 08/12/2022] [Accepted: 08/14/2022] [Indexed: 11/19/2022] Open
Abstract
Boron (B) is considered a prebiotic chemical element with a role in both the origin and evolution of life, as well as an essential micronutrient for some bacteria, plants, fungi, and algae. B has beneficial effects on the biological functions of humans and animals, such as reproduction, growth, calcium metabolism, bone formation, energy metabolism, immunity, and brain function. Naturally organic B (NOB) species may become promising novel prebiotic candidates. NOB-containing compounds have been shown to be essential for the symbiosis between organisms from different kingdoms. New insights into the key role of NOB species in the symbiosis between human/animal hosts and their microbiota will influence the use of natural B-based colon-targeting nutraceuticals. The mechanism of action (MoA) of NOB species is related to the B signaling molecule (autoinducer-2-borate (AI-2B)) as well as the fortification of the colonic mucus gel layer with NOB species from B-rich prebiotic diets. Both the microbiota and the colonic mucus gel layer can become NOB targets. This paper reviews the evidence supporting the essentiality of the NOB species in the symbiosis between the microbiota and the human/animal hosts, with the stated aim of highlighting the MoA and targets of these species.
Collapse
Affiliation(s)
- Andrei Biţă
- Department of Biochemistry, BioBoron Research Institute, S.C. Natural Research S.R.L., 31B Dunării Street, 207465 Podari, Romania
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Ion Romulus Scorei
- Department of Biochemistry, BioBoron Research Institute, S.C. Natural Research S.R.L., 31B Dunării Street, 207465 Podari, Romania
- Correspondence: ; Tel.: +40-351-407-543
| | - Tudor Adrian Bălşeanu
- Department of Physiology, Faculty of Medicine, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Maria Viorica Ciocîlteu
- Department of Analytical Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Cornelia Bejenaru
- Department of Pharmaceutical Botany, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Antonia Radu
- Department of Pharmaceutical Botany, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Ludovic Everard Bejenaru
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Gabriela Rău
- Department of Organic Chemistry, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - George Dan Mogoşanu
- Department of Pharmacognosy & Phytotherapy, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Johny Neamţu
- Department of Physics, Faculty of Pharmacy, University of Medicine and Pharmacy of Craiova, 2 Petru Rareş Street, 200349 Craiova, Romania
| | - Steven A. Benner
- Foundation for Applied Molecular Evolution (FfAME), 13709 Progress Avenue, Room N112, Alachua, FL 32615, USA
| |
Collapse
|
38
|
Dong Y, Liao W, Tang J, Fei T, Gai Z, Han M. Bifidobacterium BLa80 mitigates colitis by altering gut microbiota and alleviating inflammation. AMB Express 2022; 12:67. [PMID: 35670877 PMCID: PMC9174416 DOI: 10.1186/s13568-022-01411-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 05/27/2022] [Indexed: 12/18/2022] Open
Abstract
This study was conducted to explore the therapeutic effect of the probiotic Bifidobacterium animalis subsp. lactis BLa80 on inflammatory bowel disease. A model of ulcerative colitis (UC) was induced in C57BL/6 mice by administering of 2.5% dextran sulphate sodium (DSS) for 8 days. After developing UC, some mice were treated via intragastric administration of BLa80 at a dose of 109 colony-forming units to assess the preventive effects of BLa80 on DSS-induced UC. Compared with non-treated UC model mice, BLa80-treated mice had reduced colon shortening and improvements in colonic tissue structure. Treatment with BLa80 also decreased the serum concentrations of the proinflammatory cytokines tumor necrosis factor-alpha (TNF-α), interleukin (IL) 6 and IL-17 in mice. 16S rRNA gene sequencing revealed that BLa80 increased gut microbial diversity in mice and modulated UC-associated imbalances in the gut microbiota. BLa80 selectively promoted the growth of beneficial bacteria, including Romboutsia and Adlercreutzia, the abundances of which were negatively correlated with concentration of cellular inflammatory factors. In summary, the study results demonstrated that pretreatment with B. lactis BLa80 reduced intestinal inflammation and altered the gut microbiota, implying that BLa80 is a promising probiotic strain with potential therapeutic function in UC.
Collapse
Affiliation(s)
- Yao Dong
- Department of Research and Development, Wecare-Bio Probiotics (Suzhou) Co., Ltd., Wujiang Bridge Road, 1033, Suzhou, Jiangsu, China
| | - Wenyan Liao
- State Key Laboratory of Dairy Biotechnology, Technology Center Bright Dairy & Food Co., Ltd., Shanghai, 200436, China
| | - Jing Tang
- Shanghai Business School, 2271# Zhongshanxilu Road, Shanghai, 200235, China
| | - Teng Fei
- Department of Research and Development, Wecare-Bio Probiotics (Suzhou) Co., Ltd., Wujiang Bridge Road, 1033, Suzhou, Jiangsu, China
| | - Zhonghui Gai
- Department of Research and Development, Wecare-Bio Probiotics (Suzhou) Co., Ltd., Wujiang Bridge Road, 1033, Suzhou, Jiangsu, China
| | - Mei Han
- Shanghai Business School, 2271# Zhongshanxilu Road, Shanghai, 200235, China.
| |
Collapse
|
39
|
Rengachar P, Bhatt AN, Polavarapu S, Veeramani S, Krishnan A, Sadananda M, Das UN. Gamma-Linolenic Acid (GLA) Protects against Ionizing Radiation-Induced Damage: An In Vitro and In Vivo Study. Biomolecules 2022; 12:797. [PMID: 35740923 PMCID: PMC9221136 DOI: 10.3390/biom12060797] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 05/31/2022] [Accepted: 06/01/2022] [Indexed: 02/04/2023] Open
Abstract
Radiation is pro-inflammatory in nature in view of its ability to induce the generation of reactive oxygen species (ROS), cytokines, chemokines, and growth factors with associated inflammatory cells. Cells are efficient in repairing radiation-induced DNA damage; however, exactly how this happens is not clear. In the present study, GLA reduced DNA damage (as evidenced by micronuclei formation) and enhanced metabolic viability, which led to an increase in the number of surviving RAW 264.7 cells in vitro by reducing ROS generation, and restoring the activities of desaturases, COX-1, COX-2, and 5-LOX enzymes, TNF-α/TGF-β, NF-kB/IkB, and Bcl-2/Bax ratios, and iNOS, AIM-2, and caspases 1 and 3, to near normal. These in vitro beneficial actions were confirmed by in vivo studies, which revealed that the survival of female C57BL/6J mice exposed to lethal radiation (survival~20%) is significantly enhanced (to ~80%) by GLA treatment by restoring altered levels of duodenal HMGB1, IL-6, TNF-α, and IL-10 concentrations, as well as the expression of NF-kB, IkB, Bcl-2, Bax, delta-6-desaturase, COX-2, and 5-LOX genes, and pro- and anti-oxidant enzymes (SOD, catalase, glutathione), to near normal. These in vitro and in vivo studies suggest that GLA protects cells/tissues from lethal doses of radiation by producing appropriate changes in inflammation and its resolution in a timely fashion.
Collapse
Affiliation(s)
- Poorani Rengachar
- BioScience Research Centre, Department of Medicine, GVP Medical College and Hospital, Visakhapatnam 530048, India; (P.R.); (S.P.)
- Department of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi 110054, India;
| | - Anant Narayan Bhatt
- Department of Radiation Biosciences, Institute of Nuclear Medicine and Allied Sciences, DRDO, Delhi 110054, India;
| | - Sailaja Polavarapu
- BioScience Research Centre, Department of Medicine, GVP Medical College and Hospital, Visakhapatnam 530048, India; (P.R.); (S.P.)
| | - Senthil Veeramani
- Quality Assurance Laboratory, Ship Building Centre, Vishakhapatnam 530014, India;
| | - Anand Krishnan
- Department of Radiotherapy, Queen’s NRI Hospital, Vishakhapatnam 530013, India;
| | - Monika Sadananda
- Department of Biosciences, Mangalore University, Mangalore 574199, India;
| | - Undurti N. Das
- BioScience Research Centre, Department of Medicine, GVP Medical College and Hospital, Visakhapatnam 530048, India; (P.R.); (S.P.)
- Department of Biosciences, Mangalore University, Mangalore 574199, India;
- UND Life Sciences, 2221 NW 5th St., Battle Ground, WA 98604, USA
- Department of Biotechnology, Indian Institute of Technology, Sangareddy 502284, India
- Department of Medicine, Sri Ramachandra Medical College and Research Institute, Chennai 600116, India
| |
Collapse
|
40
|
Obrador E, Salvador-Palmer R, Villaescusa JI, Gallego E, Pellicer B, Estrela JM, Montoro A. Nuclear and Radiological Emergencies: Biological Effects, Countermeasures and Biodosimetry. Antioxidants (Basel) 2022; 11:1098. [PMID: 35739995 PMCID: PMC9219873 DOI: 10.3390/antiox11061098] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 05/26/2022] [Accepted: 05/27/2022] [Indexed: 11/17/2022] Open
Abstract
Atomic and radiological crises can be caused by accidents, military activities, terrorist assaults involving atomic installations, the explosion of nuclear devices, or the utilization of concealed radiation exposure devices. Direct damage is caused when radiation interacts directly with cellular components. Indirect effects are mainly caused by the generation of reactive oxygen species due to radiolysis of water molecules. Acute and persistent oxidative stress associates to radiation-induced biological damages. Biological impacts of atomic radiation exposure can be deterministic (in a period range a posteriori of the event and because of destructive tissue/organ harm) or stochastic (irregular, for example cell mutation related pathologies and heritable infections). Potential countermeasures according to a specific scenario require considering basic issues, e.g., the type of radiation, people directly affected and first responders, range of doses received and whether the exposure or contamination has affected the total body or is partial. This review focuses on available medical countermeasures (radioprotectors, radiomitigators, radionuclide scavengers), biodosimetry (biological and biophysical techniques that can be quantitatively correlated with the magnitude of the radiation dose received), and strategies to implement the response to an accidental radiation exposure. In the case of large-scale atomic or radiological events, the most ideal choice for triage, dose assessment and victim classification, is the utilization of global biodosimetry networks, in combination with the automation of strategies based on modular platforms.
Collapse
Affiliation(s)
- Elena Obrador
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Rosario Salvador-Palmer
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Juan I. Villaescusa
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain; (J.I.V.); (A.M.)
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| | - Eduardo Gallego
- Energy Engineering Department, School of Industrial Engineering, Polytechnic University of Madrid, 28040 Madrid, Spain;
| | - Blanca Pellicer
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - José M. Estrela
- Department of Physiology, Faculty of Medicine and Odontology, University of Valencia, 46010 Valencia, Spain; (R.S.-P.); (B.P.); (J.M.E.)
| | - Alegría Montoro
- Service of Radiological Protection, Clinical Area of Medical Image, La Fe University Hospital, 46026 Valencia, Spain; (J.I.V.); (A.M.)
- Biomedical Imaging Research Group GIBI230, Health Research Institute (IISLaFe), La Fe University Hospital, 46026 Valencia, Spain
| |
Collapse
|
41
|
Cai S, Xie LW, Xu JY, Zhou H, Yang C, Tang LF, Tian Y, Li M. (-)-Epigallocatechin-3-Gallate (EGCG) Modulates the Composition of the Gut Microbiota to Protect Against Radiation-Induced Intestinal Injury in Mice. Front Oncol 2022; 12:848107. [PMID: 35480105 PMCID: PMC9036363 DOI: 10.3389/fonc.2022.848107] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022] Open
Abstract
The high radiosensitivity of the intestinal epithelium limits the outcomes of radiotherapy against abdominal malignancies, which results in poor prognosis. Currently, no effective prophylactic or therapeutic strategy is available to mitigate radiation toxicity in the intestine. Our previous study revealed that the green tea polyphenol (-)-epigallocatechin-3-gallate (EGCG) attenuates radiation-induced intestinal injury (RIII). The aim of the present study was to determine the effect of EGCG on the intestinal flora of irradiated mice. EGCG administration reduced radiation-induced intestinal mucosal injury, and significantly increased the number of Lgr5+ intestinal stem cells (ISCs) and Ki67+ crypt cells. In addition, EGCG reversed radiation-induced gut dysbiosis, restored the Firmicutes/Bacteroidetes ratio, and increased the abundance of beneficial bacteria. Our findings provide novel insight into EGCG-mediated remission of RIII, revealing that EGCG could be a potential modulator of gut microbiota to prevent and treat RIII.
Collapse
Affiliation(s)
- Shang Cai
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Radiotherapy and Oncology, Soochow University, Suzhou, China
| | - Li-Wei Xie
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Radiotherapy and Oncology, Soochow University, Suzhou, China
| | - Jia-Yu Xu
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Radiotherapy and Oncology, Soochow University, Suzhou, China
| | - Hao Zhou
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Chao Yang
- Department of Nucleus Radiation-related Injury Treatment, Chinese People's Liberation Army Rocket Force Characteristic Medical Center, Beijing, China
| | - Lin-Feng Tang
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
| | - Ye Tian
- Department of Radiotherapy and Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Radiotherapy and Oncology, Soochow University, Suzhou, China
- *Correspondence: Ye Tian, ; Ming Li,
| | - Ming Li
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Medical College of Soochow University, Collaborative Innovation Center of Radiation Medicine of Jiangsu Higher Education Institutions, Soochow University, Suzhou, China
- *Correspondence: Ye Tian, ; Ming Li,
| |
Collapse
|