1
|
Balasubramanian D, Agraharam G, Girigoswami A, Girigoswami K. Multiple radiations and its effect on biological system - a review on in vitro and in vivo mechanisms. Ann Med 2025; 57:2486595. [PMID: 40219761 PMCID: PMC11995768 DOI: 10.1080/07853890.2025.2486595] [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: 08/19/2023] [Revised: 01/07/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
PURPOSE We are exposed to different types of radiation from natural sources or for biomedical diagnostic and therapeutic purposes at different doses or times. The dose, duration, and number of exposures can cause multiple effects both in vivo and in vitro. Several researchers have explored the effects of ionizing and non-ionizing radiation in cell lines and animal models. Macromolecules, such as DNA, RNA, and proteins, are the primary targets of damage and can lead to several diseases, including cancer and even cell death. Chronic low-dose exposure of cells to radiation can cause alterations in gene expression and can be deleterious to the fate of the cells. We aim to discuss the implications of multiple radiations on different biological systems, including how nanotechnology can facilitate the effects of radiation in therapeutics. CONCLUSION In this review, we discuss the in vitro and in vivo changes that occur due to exposure to different types of radiation used in diagnosis, therapeutics, and other means, such as radiation equipment operators and patients being exposed. The effects of ionizing and non-ionizing radiation have been discussed separately. We have also mentioned in detail about the human-caused accidents of Hiroshima and Chernobyl in this article. The application of nanotechnology in facilitating the effects of radiation in the therapy and management of radioresistance of cells has also been discussed. The radio resistance and method to improve the radiosensitivity have also been mentioned. This review article can reflect the recent developments in the various uses of ionizing and non-ionizing radiation in biomedical field and will open up new avenues to utilize radiation in a more prudent way. The role of nanotechnology in reducing the harmful effects of radiation is also discussed.
Collapse
Affiliation(s)
- Deepika Balasubramanian
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chennai, India
| | - Gopikrishna Agraharam
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chennai, India
| | - Agnishwar Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chennai, India
| | - Koyeli Girigoswami
- Medical Bionanotechnology, Faculty of Allied Health Sciences, Chettinad Hospital and Research Institute, Chettinad Academy of Research and Education, Kelambakkam, Chennai, India
| |
Collapse
|
2
|
Katona BW, Shukla A, Hu W, Nyul T, Dudzik C, Arvanitis A, Clay D, Dungan M, Weber M, Tu V, Hao F, Gan S, Chau L, Buchner AM, Falk GW, Jaffe DL, Ginsberg G, Palmer SN, Zhan X, Patterson AD, Bittinger K, Ni J. Microbiota and metabolite-based prediction tool for colonic polyposis with and without a known genetic driver. Gut Microbes 2025; 17:2474141. [PMID: 40069167 PMCID: PMC11913376 DOI: 10.1080/19490976.2025.2474141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/22/2025] [Accepted: 02/25/2025] [Indexed: 03/19/2025] Open
Abstract
Despite extensive investigations into the microbiome and metabolome changes associated with colon polyps and colorectal cancer (CRC), the microbiome and metabolome profiles of individuals with colonic polyposis, including those with (Gene-pos) and without (Gene-neg) a known genetic driver, remain comparatively unexplored. Using colon biopsies, polyps, and stool from patients with Gene-pos adenomatous polyposis (N = 9), Gene-neg adenomatous polyposis (N = 18), and serrated polyposis syndrome (SPS, N = 11), we demonstrated through 16S rRNA sequencing that the mucosa-associated microbiota in individuals with colonic polyposis is representative of the microbiota associated with small polyps, and that both Gene-pos and SPS cohorts exhibit differential microbiota populations relative to Gene-neg polyposis cohorts. Furthermore, we used these differential microbiota taxa to perform linear discriminant analysis to differentiate Gene-neg subjects from Gene-pos and from SPS subjects with an accuracy of 89% and 93% respectively. Stool metabolites were quantified via 1H NMR, revealing an increase in alanine in SPS subjects relative to non-polyposis subjects, and Partial Least Squares Discriminant Analysis (PLS-DA) analysis indicated that the proportion of leucine to tyrosine in fecal samples may be predictive of SPS. Use of these microbial and metabolomic signatures may allow for better diagnostric and risk-stratification tools for colonic polyposis patients and their families as well as promote development of microbiome-targeted approaches for polyp prevention.
Collapse
Affiliation(s)
- Bryson W Katona
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Ashutosh Shukla
- Division of Digestive & Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Weiming Hu
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Thomas Nyul
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Christina Dudzik
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Alex Arvanitis
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Daniel Clay
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Michaela Dungan
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Marina Weber
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Vincent Tu
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Fuhua Hao
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA, USA
| | - Shuheng Gan
- Peter O'Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Lillian Chau
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Anna M Buchner
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gary W Falk
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David L Jaffe
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Gregory Ginsberg
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Suzette N Palmer
- Peter O'Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaowei Zhan
- Peter O'Donnell Jr. School of Public Health, Quantitative Biomedical Research Center, Center for the Genetics and Host Defense, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Andrew D Patterson
- Department of Veterinary and Biomedical Sciences, Center for Molecular Toxicology and Carcinogenesis, Penn State University, University Park, PA, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Josephine Ni
- Division of Digestive & Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
3
|
Liu X, Zhao J, Liu J, Huang Y, Deng W, Yan L, Cui M, Pan X, Xiao H, Liu X. Association of α-Klotho with anti-aging effects of Ganoderma lucidum in animal models. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119597. [PMID: 40057146 DOI: 10.1016/j.jep.2025.119597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/15/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aging is a complex, universal process characterized by structural and functional decline across multiple organs. Ganoderma lucidum (G. lucidum), a renowned traditional Chinese medicinal fungus, has long been recognized for its anti-aging properties. However, the underlying mechanisms remain incompletely understood. AIM OF THE STUDY This study aimed to investigate the anti-aging effects of G. lucidum and its underlying mechanisms. MATERIALS AND METHODS We investigated the anti-aging effects of G. lucidum sporoderm-broken spore powder (Gl-SBSP) on Caenorhabditis elegans (C. elegans) lifespan and aging across multiple organs using natural aging, D-galactose (D-gal)-induced aging, and radiation-induced premature senescence mouse models. In C. elegans, we assessed lifespan, reproductive capacity, body length, pharyngeal pumping, body bends, fat and lipofuscin levels, as well as reactive oxygen species (ROS) accumulation. In mice, histopathological staining, complete blood counts, and enzyme-linked immunosorbent assay (ELISA) were used to evaluate tissue damage, while quantitative real-time PCR (RT-qPCR) was employed to access small intestine barrier integrity. Western blot (WB) and immunohistochemistry (IHC) were utilized to analyze the distribution of alpha Klotho (α-Klotho) in the kidney, blood, and urine. RESULTS Gl-SBSP significantly extended C. elegans lifespan, improved reproductive capacity and mobility, and reduced lipofuscin and ROS levels. In naturally aged mice, Gl-SBSP enhanced physical appearance and performance. Additionally, Gl-SBSP alleviated aging-related structural and functional decline in multiple organs, including the colon, spleen, kidneys, liver, and small intestine, across all aging models. Biochemical analyses revealed that Gl-SBSP increased transmembrane α-Klotho (mα-Klotho) and soluble α-Klotho (sα-Klotho) levels in kidney tissue and elevated sα-Klotho levels in serum and urine. CONCLUSION This study is the first to demonstrate that G. lucidum exerts α-Klotho-associated anti-aging effects in animal models, highlighting its potential as an anti-aging intervention.
Collapse
Affiliation(s)
- Xiaojing Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jiamin Zhao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Jia Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Yan Huang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Wei Deng
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Luwen Yan
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, 238 Baidi Road, Tianjin, 300192, China
| | - Xinhua Pan
- Jiangxi Xiankelai Biotechnology Co., Ltd., 10 Shacheng Road, Jiujiang, 332000, China
| | - Huiwen Xiao
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China
| | - Xingzhong Liu
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Molecular Microbiology and Technology of the Ministry of Education, Department of Microbiology, College of Life Sciences, Nankai University, 94 Weijin Road, Tianjin, 300071, China.
| |
Collapse
|
4
|
Zhang C, Liu B, Cui Z, Wu K, Huang H, Wang Y, Ma X, Tan B. Effects of Magnolia officinalis extract on the growth performance and immune function of weaned piglets. Porcine Health Manag 2025; 11:16. [PMID: 40181480 PMCID: PMC11969803 DOI: 10.1186/s40813-025-00430-z] [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: 09/30/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Magnolia officinalis is a medicinal herb known for its pharmacological properties and as a potential natural feed additive. We aimed to assess the effects of dietary Magnolia officinalis extract (MOE) on the growth performance and immune function of piglets, and explored the potential of MOE as a natural alternative to antibiotics for piglet nutrition during weaning. RESULTS Compared with the basal diet group (CK), the MOE diet significantly increased average daily feed intake and reduced diarrhea incidence and serum interleukin-6 (IL-6) levels. Compared with 0.1% MOE group, the 0.05% MOE group had lower diarrhea rates, eosinophils (EOS) count, EOS' percentage, and serum interleukin-4 levels. Compared with CK, 0.05% MOE supplementation in the diet could reduce the diarrhea incidence and the thymus index by elevating the levels of transforming growth factor-β (TGF-β) and interleukin-10 (IL-10) in the serum, jejunum, and ileum. Compared with the basal diet group, 0.05% MOE supplementation upregulated the mRNA expressions of IL-10 and TGF-β1 in the jejunum and ileum (P < 0.05) and those of IL-10, interleukin-1β (IL-1β), and interferon-γ (IFN-γ) in the thymus (P < 0.05). Moreover, 0.05% MOE increased the levels of butyric, isobutyric, isovaleric, and valeric acids in the colon. CONCLUSIONS MOE supplementation could modulate the immune status of animals, lower production costs, and contribute to more sustainable and ethical pig farming practices by promoting healthier growth and reducing disease susceptibility. Our findings offer a sustainable solution to antibiotic use in animal farming, addressing concerns about antibiotic resistance and food safety.
Collapse
Affiliation(s)
- Chen Zhang
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, 230031, China.
- Yuelushan Laboratory, Changsha, 410128, China.
- Institute of Yunnan Circular Agricultural Industry, Pu'er, 665000, China.
| | - Bifan Liu
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
- Institute of Yunnan Circular Agricultural Industry, Pu'er, 665000, China
| | - Zhijuan Cui
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
| | - Kunfu Wu
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
| | - Haibo Huang
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
- Anhui Provincial Key Laboratory of Livestock and Poultry Product Safety, Institute of Animal Husbandry and Veterinary Medicine, Anhui Academy of Agricultural Sciences, Hefei, 230031, China
- Yuelushan Laboratory, Changsha, 410128, China
- Institute of Yunnan Circular Agricultural Industry, Pu'er, 665000, China
| | - Yongliang Wang
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
- Institute of Yunnan Circular Agricultural Industry, Pu'er, 665000, China
| | - Xiaokang Ma
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China
- Yuelushan Laboratory, Changsha, 410128, China
| | - Bi'e Tan
- Key Laboratory for Quality Regulation of Livestock and Poultry Products of Hunan Province, College of Animal Science and Technology, Hunan Agricultural University, Changsha, 410128, China.
- Yuelushan Laboratory, Changsha, 410128, China.
- Institute of Yunnan Circular Agricultural Industry, Pu'er, 665000, China.
| |
Collapse
|
5
|
Jia H, Xie Y, Yi L, Cheng W, Song G, Shi W, Zhu J, Zhao S. Comparative Analysis of Short-Chain Fatty Acids and the Immune Barrier in Cecum of Dahe Pigs and Dahe Black Pigs. Animals (Basel) 2025; 15:920. [PMID: 40218314 PMCID: PMC11987949 DOI: 10.3390/ani15070920] [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: 02/07/2025] [Revised: 03/17/2025] [Accepted: 03/18/2025] [Indexed: 04/14/2025] Open
Abstract
The intestinal immune barrier is a developed and complex immune system, and there is a fine synergy between it and the induced immune response. Short-chain fatty acids (SCFAs) are the main metabolites of intestinal microbial fermentation. In the cecum of pigs, SCFAs not only provide energy for the host but also participate in regulating the function of the intestinal immune system. The purpose of this study was to explore the mechanism of SCFAs in the regulation of immune gene expression in porcine cecum. SCFAs content and mRNA expression levels of immune genes in cecum were detected, and Gene Ontology (GO) function annotation, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis, Protein-Protein Interaction Networks (PPI) network construction, key gene identification, and correlation analysis were performed. The results showed that the content of SCFAs in the cecum of Dahe black pigs (DHB) was lower than that of Dahe pigs (DH). There were significant differences in mRNA expression of some immune genes between the two groups. GO functional annotation found terms related to cytokine activity and protein heterodimerization activity; the KEGG pathway was enriched in several pathways related to intestinal immunity. The PPI network identified Interleukin-6 (IL-6), Interleukin-8 (IL-8), Interleukin-10 (IL-10), Interleukin-17A (IL-17A), and Interleukin-18 (IL-18) as key proteins. The correlation analysis showed that acetic acid and valerate were closely related to the immune response. In this study, the differences in cecal short-chain fatty acids and the immune barrier between Dahe pigs and Dahe black pigs were compared, which provided a theoretical basis for improving the intestinal immunity of pigs.
Collapse
Affiliation(s)
- Huijin Jia
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Yuxiao Xie
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
- College of Biology and Agriculture, Zunyi Normal University, Zunyi 563006, China
| | - Lanlan Yi
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Wenjie Cheng
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Guangyao Song
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Wenzhe Shi
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Junhong Zhu
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| | - Sumei Zhao
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming 650201, China
| |
Collapse
|
6
|
Huang H, Zhao T, Ma W. Omega-3 polyunsaturated fatty acids attenuates cognitive impairment via the gut-brain axis in diabetes-associated cognitive dysfunction rats. Brain Behav Immun 2025; 127:147-169. [PMID: 40068791 DOI: 10.1016/j.bbi.2025.03.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 02/11/2025] [Accepted: 03/06/2025] [Indexed: 03/17/2025] Open
Abstract
Diabetes-related cognitive dysfunction (DACD) is a comorbidity of type 2 diabetes that has a negative effect on patients' quality of life. Research has indicated that disruption of the gut microbiota (GM) may be linked to dementia with altered cognitive performance. Conversely, omega-3 polyunsaturated fatty acids (n-3 PUFAs) may reverse DACD. The present study aimed to assess the effects of an n-3 PUFA intervention and fecal microbiota transplantation (FMT) on high-fat and streptozotocin-induced DACD model rats. In DACD rats, n-3 PUFA treatment restored fasting blood glucose (FBG) levels and cognitive function, increased the expression of anti-inflammatory cytokines and downregulated the expression of proinflammatory cytokines in the cortex and colon. Additionally, the expression of the postsynaptic density protein-95 mRNA and protein varied with n-3 PUFA treatment. Treatment with n-3 PUFAs also increased the expression of tight junction proteins. Beneficial and short-chain fatty acid-producing bacteria were more abundant when rats were exposed to n-3 PUFAs. After FMT from the rats with DACD symptoms that were improved by the n-3 PUFA dietary intervention into another batch of DACD rats, we observed recovery in recipient DACD rats. These results indicated that the alleviation of DACD symptoms by n-3 PUFAs was attributed to gut microbiota remodeling.
Collapse
Affiliation(s)
- Hongying Huang
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China; Nanchang Institute of Disease Control and Prevention, China Railway Nanchang Bureau Group Co., Ltd., Nanchang, 330003, People's Republic of China
| | - Tong Zhao
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China
| | - Weiwei Ma
- School of Public Health, Capital Medical University, Beijing 100069, People's Republic of China.
| |
Collapse
|
7
|
Wu J, Ji K, Kang G, Zhang M, Wang J, Wang L, Gao M, Jia X, Lu X, Wang Y, Gao X, Guo Y, Zhu Z, Wang Q, Zhao Z, Liu Q, Huang H. Butyrate-engineered yeast activates Nppa and Sgcg genes and reduces radiation-induced heart damage via the gut-heart axis. Pharmacol Res 2025; 213:107642. [PMID: 39909125 DOI: 10.1016/j.phrs.2025.107642] [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: 12/23/2024] [Revised: 01/26/2025] [Accepted: 02/02/2025] [Indexed: 02/07/2025]
Abstract
Radiotherapy is a method of treating cancer through radiation aimed at killing cancer cells or inhibiting their growth. However, radiotherapy has numerous side effects because it kills tumors while causing damage to normal cells or tissues. The literature shows that radiation can cause damage to heart tissue. This study found that engineered yeast that produced butyrate can maintain small intestinal barrier function by recovering GPR109A to reduce intestinal damage caused by abdominal irradiation in mice. We unexpectedly found that engineered yeast could mitigate irradiation-induced heart damage via the gut-heart axis. Mechanistically, engineered yeast enhanced taurine and nicotinamide metabolism by increasing the relative abundance of Akkermansia and Lachnospiraceae_NK4A136; then, yeast modulated cardiac function by activating the Sgcg and Nppa genes to attenuate cardiac damage induced by abdominal irradiation. Finally, we confirmed that engineered yeast mitigated cardiac damage caused by total body irradiation, which protected other vital organs through the intestinal tract. This study has a profound impact on cancer treatment, the emergence of engineered yeast will alleviate radiotherapy side effects and benefit patients.
Collapse
Affiliation(s)
- Jiahao Wu
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Kaihua Ji
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Guangbo Kang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Manman Zhang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Jigang Wang
- Department of Pathology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - Lina Wang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Mengxue Gao
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Xiaoxiao Jia
- Department of Anatomy, Shandong Second Medical University, Weifang 261053, China
| | - Xinran Lu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Yan Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China
| | - Xinran Gao
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Yufei Guo
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Zhixin Zhu
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Qinghua Wang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China
| | - Zhenyu Zhao
- NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin 300134, China
| | - Qiang Liu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin Institutes of Health Science, State Key Laboratory of Advanced Medical Materials and Devices, Tianjin 300192, China; School of Population Medicine and Public Health, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100730, China.
| | - He Huang
- School of Chemical Engineering and Technology, State Key Laboratory of Synthetic Biology, Tianjin Key Laboratory of Biological and Pharmaceutical Engineering, Tianjin University, Tianjin 300350, China.
| |
Collapse
|
8
|
Yang P, Bai H, Yan T, Xu X, Tang X, Song B, Liu Y, Lu Y, Liu P, Tu W, Shi Y, Zhang S. Integrative multi-omics analysis of radionuclide-induced intestinal injury reveals the radioprotective role of L-citrulline through histone H3-mediated Cxcl3. J Transl Med 2025; 23:180. [PMID: 39953550 PMCID: PMC11829450 DOI: 10.1186/s12967-025-06197-9] [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] [Received: 09/19/2024] [Accepted: 02/03/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND The widespread application of nuclear technology has markedly heightened the risk of extensive, uncontrolled exposure to radiation. Nevertheless, in contrast to external irradiation, the biological impacts and countermeasures against internal irradiation from radionuclides remain inadequately characterized. METHODS Mice were administered yttrium-90 (Y90) carbon microspheres via gavage at different dosages (0-5.0 mCi) to establish a radionuclides exposure model. A multi-omics analysis was employed to access alterations in gut microbiota, fecal and colonic metabolites profiles, and intestinal mRNA expression post-irradiation. The function of significant metabolite was validated at both cellular levels and organismal levels. Additionally, ChIP-Seq and RNA-Seq techniques were utilized to investigate the molecular mechanism underlying the actions of key metabolite. RESULTS Exposure to Y90 resulted in intestinal damage and hematological impairment. Multi-omics analysis revealed significant alternations of gut microbiota, fecal metabolites, colonic metabolites, and intestinal mRNA expression following internal radiation exposure. Notably, L-citrulline was identified as a metabolite with changes observed in both fecal and colonic tissues, demonstrating radioprotective properties in vitro and in vivo. Mechanistically, L-citrulline facilitated the citrullination of histone H3 at the 17th site (H3Cit17), and multiple mRNAs including C-X-C motif chemokine ligand 3 (Cxcl3), were transcriptionally regulated by H3Cit17 post L-citrulline treatment. Furthermore, Cxcl3 conferred protective effects for intestinal epithelial cells against ionizing radiation. CONCLUSIONS The research offers critical perspectives on the intestinal and gut microbiota's reaction to radionuclides exposure. It underscores the promise of L-citrulline as a radioprotective compound, which may have substantial ramifications for the formulation of strategies to mitigate radiation exposure.
Collapse
Affiliation(s)
- Ping Yang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Hao Bai
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Tao Yan
- The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Xiaopeng Xu
- Department of Gastroenterology, the Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, 214400, China
| | - Xiaoyou Tang
- Medical College of Tibet University, Lasa, 850000, China
| | - Bin Song
- West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yulan Liu
- The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Yunyun Lu
- Radiochemical Laboratory, Reactor Operation and Application Research Sub-Institute, Nuclear Power Institute of China, Chengdu, 610200, China
| | - Pengfei Liu
- Department of Gastroenterology, the Jiangyin Clinical College of Xuzhou Medical University, Jiangyin, 214400, China
| | - Wenling Tu
- The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Yuhong Shi
- The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China
| | - Shuyu Zhang
- Laboratory of Radiation Medicine, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China.
- The Second Affiliated Hospital of Chengdu Medical College, Nuclear Industry 416 Hospital, Chengdu, 610051, China.
- Medical College of Tibet University, Lasa, 850000, China.
- West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
- NHC Key Laboratory of Nuclear Technology Medical Transformation (Mianyang Central Hospital), Mianyang, 621099, China.
| |
Collapse
|
9
|
Rong Y, Zhu M, Wang N, Zhang F, Liu T. Photodynamic therapy with a novel photosensitizer inhibits DSS-induced ulcerative colitis in rats via the NF-κB signaling pathway. Front Pharmacol 2025; 15:1539363. [PMID: 39845801 PMCID: PMC11750845 DOI: 10.3389/fphar.2024.1539363] [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: 12/04/2024] [Accepted: 12/23/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Ulcerative colitis (UC) is an inflammatory bowel disease characterized by inflammation and ulceration of the digestive tract. Methods Photodynamic therapy (PDT) with a novel photosensitizer LD4 was used to treat UC rat models to explore the therapeutic effect and mechanism of LD4-PDT on UC. 16S ribosomal RNA was used to detect the composition of Gut microbiota. Results Our findings indicate that LD4-PDT could protect the integrity of the colonic mucosa, alleviate the inflammatory response and promote the healing of colonic mucosa. Mechanism studies demonstrated that LD4-PDT could inhibit the NF-κB signaling pathway, downregulated the expression of the inflammatory factors' tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6) and myeloperoxidase (MPO), increased the contents of glutathione (GSH) and superoxide dismutase (SOD) and decreased the content of malondialdehyde (MDA). Additionally, analysis of gut microbiota revealed that LD4-PDT treatment could decrease the abundance of the Proteobacteria phylum in fecal samples, while no significant differences were observed in the Firmicutes, Bacteroidetes, or Actinobacteria phyla among the three groups using 16S rRNA analysis. Discussion In summary, our data suggested that LD4-PDT could inhibit DSS-induced UC in rats via the NF-κB signaling pathway, indicating its potential as a novel photosensitizer for the treatment of UC.
Collapse
Affiliation(s)
- Yumei Rong
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
- Tianjin Key Laboratory of Biomedical Material, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Minghui Zhu
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Nan Wang
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Feiyu Zhang
- The Third Central Hospital of Tianjin, Tianjin, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
- Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Tianjun Liu
- Tianjin Key Laboratory of Biomedical Material, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
10
|
Nogacka AM, García A, G de Los Reyes-Gavilán C, Arboleya S, Gueimonde M. In vitro assessment of horse-isolated strains of Lactobacillus acidophilus and Ligilactobacillus equi species for fecal microbiota modulation in horses. J Equine Vet Sci 2024; 145:105341. [PMID: 39742928 DOI: 10.1016/j.jevs.2024.105341] [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: 09/30/2024] [Revised: 11/24/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Horses are hindgut fermenters that harbor a complex intestinal microbiota (IM) which provides key enzymes aiding in the breakdown of complex carbohydrates present in their herbivorous diet. Therefore, these animals are deeply dependent on their IM for digestion and nutrition. Consequently, IM imbalances may result in alteration of fermentation patterns with impact on the animal health and the risk of disease. In this context, strategies for assisting the maintenance of a healthy IM in horses are of interest. However, there is limited research concerning the use of probiotics to improve hindgut fermentation and diet digestibility, with very few studies focusing on the use of lactobacilli strains from equine origin. Herein, we conducted independent fecal batch fermentations, using feces from "Asturcón" horses as inocula, added individually with four different lactobacilli strains (two strains of Lactobacillus acidophilus and two of Ligilactobacillus equi) isolated from this same horse breed. The impact on the gut microbiota composition was assessed by 16S rRNA gene profiling and the metabolic activity (production of short-chain fatty acids) by gas chromatography. The functionality of the lactobacilli strain was determined by monitoring in real-time gas production and determining changes in pH along incubation. L. acidophilus IPLA20127, promoted an increase in IM diversity and in the relative abundance of Lactobacillus genus, as well as higher butyric and valeric acid levels. This strain shows potential as probiotic supplement, without triggering acidification, nor promoting an increase of gas production or abrupt IM changes in our experimental model.
Collapse
Affiliation(s)
- A M Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33011 Oviedo, Asturias, Spain
| | - A García
- Valgrande Asturcones. 33315 Bedriñana, Villaviciosa, Asturias, Spain
| | - C G de Los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33011 Oviedo, Asturias, Spain
| | - S Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33011 Oviedo, Asturias, Spain
| | - M Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33011 Oviedo, Asturias, Spain.
| |
Collapse
|
11
|
Han JM, Mwiti G, Yeom SJ, Lim J, Kim WS, Lim S, Lim ST, Byun EB. Radiation-Resistant Bacteria Deinococcus radiodurans-Derived Extracellular Vesicles as Potential Radioprotectors. Adv Healthc Mater 2024:e2403192. [PMID: 39676336 DOI: 10.1002/adhm.202403192] [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: 08/25/2024] [Revised: 11/26/2024] [Indexed: 12/17/2024]
Abstract
The increasing use of radiation presents a risk of radiation exposure, making the development of radioprotectors necessary. In the previous study, it is investigated that Deinococcus radiodurans (R1-EVs) exert the antioxidative properties. However, the radioprotective activity of R1-EVs remains unclear. In the present study, the protective effects of R1-EVs against total body irradiation (TBI)-induced acute radiation syndrome (ARS) are investigated. To assess R1-EVs' radioprotective efficacy, ARS is induced in mice with 8 Gy of TBI, and protection against hematopoietic (H)- and gastrointestinal (GI)-ARS is evaluated. The survival rate of irradiated mice group decreases substantially after irradiation. In contrast, pretreatment with R1-EVs increases the survival rates of the mice. The administration of R1-EVs provides effective protection against radiation-induced death of bone marrow cells and splenocytes by scavenging reactive oxygen species (ROS). Additionally, R1-EVs protect both intestinal stem and epithelial cells from radiation-induced apoptosis. R1-EVs stimulate the production of short-chain fatty acids in the gastrointestinal tract, suppress proinflammatory cytokines, and increase regulatory T cells in pretreated mice versus the irradiation-only group. Proteomic analysis shows that the R1-EV proteome is significantly enriched with proteins involved in oxidative stress response. These findings highlight R1-EVs as potent radioprotectors with applications against radiation damage and ROS-mediated diseases.
Collapse
Affiliation(s)
- Jeong Moo Han
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
- Department of Microbiology and Immunology, College of Medicine, Seoul National University, Seoul, 03080, South Korea
- Institute for Data Innovation in Science, Seoul National University, Seoul, 08826, South Korea
| | - Godfrey Mwiti
- Department of Microbiology and Immunology, The University of Melbourne, Melbourne, 3000, Australia
- Department of Food and Nutrition, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Seo-Joon Yeom
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Integrative Food, Bioscience and Biotechnology, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jaeyoon Lim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Food and Nutrition, Chungnam National University, Daejeon, 34134, Republic of Korea
| | - Woo Sik Kim
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| | - Sangyong Lim
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
- Department of Radiation Science, University of Science and Technology, Daejeon, 01812, Republic of Korea
| | - Seung-Taik Lim
- Department of Biotechnology, College of Life Science and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Eui-Baek Byun
- Advanced Radiation Technology Institute, Korea Atomic Energy Research Institute, Jeongeup-si, Jeollabuk-do, 56212, Republic of Korea
| |
Collapse
|
12
|
Dong Y, Dong J, Xiao H, Li Y, Wang B, Zhang S, Cui M. A gut microbial metabolite cocktail fights against obesity through modulating the gut microbiota and hepatic leptin signaling. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:9356-9367. [PMID: 39030978 DOI: 10.1002/jsfa.13758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 06/09/2024] [Accepted: 06/26/2024] [Indexed: 07/22/2024]
Abstract
BACKGROUND Excessive body weight and obesity elevate the risk of chronic non-communicable diseases. The judicious application of the gut microbiome, encompassing both microorganisms and their derived compounds, holds considerable promise in the treatment of obesity. RESULTS In this study, we showed that a cocktail of gut microbiota-derived metabolites, comprising indole 3-propionic acid (IPA), sodium butyrate (SB) and valeric acid (VA), alleviated various symptoms of obesity in both male and female mice subjected to a high-fat diet (HFD). The 16S ribosomal RNA (rRNA) sequencing revealed that administering the cocktail via oral gavage retained the gut microbiota composition in obese mice. Fecal microbiota transplantation using cocktail-treated mice as donors mitigated the obesity phenotype of HFD-fed mice. Transcriptomic sequencing analysis showed that the cocktail preserved the gene expression profile of hepatic tissues in obese mice, especially up-regulated the expression level of leptin receptor. Gene delivery via in vivo fluid dynamics further validated that the anti-obesity efficacy of the cocktail was dependent on leptin signaling at least partly. The cocktail also inhibited the expression of appetite stimulators in hypothalamus. Together, the metabolite cocktail combated adiposity by retaining the gut microbiota configuration and activating the hepatic leptin signaling pathway. CONCLUSIONS Our findings provide a sophisticated regulatory network between the gut microbiome and host, and highlight a cocktail of gut microbiota-derived metabolites, including IPA, SB, and VA, might be a prospective intervention for anti-obesity in a preclinical setting. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yanxi Dong
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Jiali Dong
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Huiwen Xiao
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin, China
| | - Yuan Li
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Bin Wang
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Shuqin Zhang
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| | - Ming Cui
- Institute of Radiation Medicine, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China
| |
Collapse
|
13
|
Thapa R, Magar AT, Shrestha J, Panth N, Idrees S, Sadaf T, Bashyal S, Elwakil BH, Sugandhi VV, Rojekar S, Nikhate R, Gupta G, Singh SK, Dua K, Hansbro PM, Paudel KR. Influence of gut and lung dysbiosis on lung cancer progression and their modulation as promising therapeutic targets: a comprehensive review. MedComm (Beijing) 2024; 5:e70018. [PMID: 39584048 PMCID: PMC11586092 DOI: 10.1002/mco2.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Lung cancer (LC) continues to pose the highest mortality and exhibits a common prevalence among all types of cancer. The genetic interaction between human eukaryotes and microbial cells plays a vital role in orchestrating every physiological activity of the host. The dynamic crosstalk between gut and lung microbiomes and the gut-lung axis communication network has been widely accepted as promising factors influencing LC progression. The advent of the 16s rDNA sequencing technique has opened new horizons for elucidating the lung microbiome and its potential pathophysiological role in LC and other infectious lung diseases using a molecular approach. Numerous studies have reported the direct involvement of the host microbiome in lung tumorigenesis processes and their impact on current treatment strategies such as radiotherapy, chemotherapy, or immunotherapy. The genetic and metabolomic cross-interaction, microbiome-dependent host immune modulation, and the close association between microbiota composition and treatment outcomes strongly suggest that designing microbiome-based treatment strategies and investigating new molecules targeting the common holobiome could offer potential alternatives to develop effective therapeutic principles for LC treatment. This review aims to highlight the interaction between the host and microbiome in LC progression and the possibility of manipulating altered microbiome ecology as therapeutic targets.
Collapse
Affiliation(s)
- Rajan Thapa
- Department of Pharmacy, Universal college of medical sciencesTribhuvan UniversityBhairahawaRupendehiNepal
| | - Anjana Thapa Magar
- Department of MedicineKathmandu Medical College Teaching Hospital, SinamangalKathmanduNepal
| | - Jesus Shrestha
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Nisha Panth
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Sobia Idrees
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Tayyaba Sadaf
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Saroj Bashyal
- Department of Pharmacy, Manmohan Memorial Institute of Health SciencesTribhuvan University, SoalteemodeKathmanduNepal
| | - Bassma H. Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences TechnologyPharos University in AlexandriaAlexandriaEgypt
| | - Vrashabh V. Sugandhi
- Department of pharmaceutical sciences, College of Pharmacy & Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Satish Rojekar
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ram Nikhate
- Department of PharmaceuticsDattakala Shikshan Sanstha, Dattakala college of pharmacy (Affiliated to Savitribai Phule Pune universityPuneMaharashtraIndia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Centre of Medical and Bio‐allied Health Sciences ResearchAjman UniversityAjmanUAE
| | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraIndia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| |
Collapse
|
14
|
Zhai S, Gao Y, Jiang Y, Li Y, Fan Q, Tie S, Wu Y, Gu S. Weizmannia coagulans BC99 affects valeric acid production via regulating gut microbiota to ameliorate inflammation and oxidative stress responses in Helicobacter pylori mice. J Food Sci 2024; 89:9985-10002. [PMID: 39556495 DOI: 10.1111/1750-3841.17514] [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: 08/13/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 11/20/2024]
Abstract
Helicobacter pylori is a highly prevalent pathogen in human gastric mucosa epithelial cells with strong colonization ability. Weizmannia coagulans is a kind of active microorganism that is beneficial to the improvement of host gut microbiota balance and can prevent and treat intestinal diseases. We investigated the beneficial effects of W. coagulans BC99 in H. pylori infected mice and measured inflammation response, oxidative stress, and gut microbiota. Results showed that BC99 could alleviate the gastric inflammation, inhibit the increasing of inflammation parameters endotoxin, interleukin-10, transforming growth factor-β, and interferon-γ and oxidative stress myeloperoxidase and malondialdehyde, promote the levels of superoxide dismutase and catalase. Furthermore, 16S rRNA gene sequencing analysis revealed that BC99 reversed the change of gut microbiota by reducing the abundance of Olsenella, Candidatus_Saccharimonas, Monoglobus, and increasing the abundance of Tyzzerella. Meanwhile, BC99 caused elevated levels of Ligilactobacillus and Lactobacillus. In view of the beneficial effect of BC99 on the content of short-chain fatty acid, valeric acid with sodium valerate interfered with H. pylori infection in mice found that valeric acid had a good restorative effect of H. pylori infection relating inflammation and oxidative stress responses. These results suggest that W. coagulans BC99 can be used as a potential probiotic to prevent and treat H. pylori infection by regulating the inflammation, oxidative stress, and gut microbiota.
Collapse
Affiliation(s)
- Shirui Zhai
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Yinyin Gao
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Yiru Jiang
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Yuwan Li
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- National Demonstration Center for Experimental Food Processing and Safety Education, Luoyang, China
| | - Qiuxia Fan
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
| | - Shanshan Tie
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- National Demonstration Center for Experimental Food Processing and Safety Education, Luoyang, China
| | - Ying Wu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Henan Engineering Research Center of Food Microbiology, Luoyang, China
| | - Shaobin Gu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, China
- Henan Engineering Research Center of Food Microbiology, Luoyang, China
- National Demonstration Center for Experimental Food Processing and Safety Education, Luoyang, China
| |
Collapse
|
15
|
Li Y, Chen Y, Liao Z, Liu Y, Liu C, Yang W, Bai J, Huang X, Hao Y, Liu S, Liu Y. WenTongGanPi decoction alleviates diarrhea-predominant irritable bowel syndrome by improving intestinal barrier. JOURNAL OF ETHNOPHARMACOLOGY 2024; 334:118544. [PMID: 39013542 DOI: 10.1016/j.jep.2024.118544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/24/2024] [Accepted: 07/08/2024] [Indexed: 07/18/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE WenTongGanPi Decoction (WTGPD) is a representative medical practice of the Fuyang School of Traditional Chinese Medicine (TCM), which originated from the classical Lu's Guizhi method. WTGPD places emphasis on the balance and functionality of yang qi, and is effective in treating TCM symptoms related to liver qi stagnation and spleen yang deficiency. In TCM, diarrhea-predominant irritable bowel syndrome (IBS-D) is often diagnosed as liver depression and spleen deficiency, and the use of WTGPD has shown significant therapeutic effect. However, the underlying mechanism of WTGPD treating IBS-D remains unclear. AIM OF THE STUDY To explore the effect and mechanism of WTGPD in the treatment of IBS-D. MATERIALS AND METHODS An IBS-D model with liver depression and spleen deficiency was constructed by chronic immobilization stress stimulation and sennae folium aqueous gavage. The impact of WTGPD on IBS-D rats was evaluated through measurements of body weight, fecal water content, and abdominal withdrawal reflex (AWR). Intestinal permeability was assessed using hematoxylin-eosin (HE), alcian blue-periodic acid schiff (AB-PAS), immunofluorescence (IF) staining, and quantitative real-time PCR (qRT-PCR). The components of WTGPD were analyzed using UPLC-Q-TOF-MS. The underlying mechanisms were investigated through network pharmacology, transcriptomics sequencing, western blot (WB), molecular docking, and 16S rRNA sequencing. RESULTS WTGPD treatment effectively alleviated diarrhea and abnormal pain in IBS-D rats (P < 0.05). It enhanced the intestinal barrier function by improving colonic structure and increasing the expression of tight junction proteins (P < 0.05). A total of 155 components were identified in WTGPD. Both network pharmacology and transcriptomics sequencing analysis highlighted MAPK as the key signaling pathway in WTGPD's anti-IBS-D effect. The WB results showed a significant decrease in p-p38, p-ERK and p-JNK expression after WTGPD treatment (P < 0.0001). Guanosine, adenosine and hesperetin in WTGPD may be involved in regulating the phosphorylation of p38, ERK and JNK. Additionally, WTGPD significantly enhanced microbial diversity and increased the production of colonic valeric acid in IBS-D rats (P < 0.01). CONCLUSION In conclusion, our findings suggest that WTGPD can effectively alleviate IBS-D and improve intestinal barrier likely via inhibiting MAPK signal pathway and improving micobial dysbiosis.
Collapse
Affiliation(s)
- Yaoguang Li
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China
| | - Yangyang Chen
- College of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China
| | - Zhengyue Liao
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China
| | - Yixin Liu
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China
| | - Chenhao Liu
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China
| | - Wenjing Yang
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China
| | - Jing Bai
- College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China
| | - Xinggui Huang
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China
| | - Yule Hao
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China
| | - Sijing Liu
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China; College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China.
| | - Yi Liu
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611103, China.
| |
Collapse
|
16
|
Ma C, Xu X, Qin S, Zhou J. Screening of biomarkers in acute radiation enteritis based on microbiome and clustering methods. BMC Microbiol 2024; 24:463. [PMID: 39516773 PMCID: PMC11545530 DOI: 10.1186/s12866-024-03620-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Radiation enteritis (RE) is a common complication of radiotherapy for abdominal and pelvic tumors, adversely affecting treatment outcomes and patients' quality of life. Gut microbiome alterations may contribute to RE development, but the underlying pathogenic factors are not fully understood. This study aimed to characterize the intestinal microbial changes associated with RE and severe acute radiation enteritis (SARE) and to identify predictive biomarkers. METHODS We enrolled 50 cervical cancer patients undergoing radiotherapy and 15 healthy women (controls). Stool samples were collected at the baseline and during weeks 2, 4, and 6 of radiotherapy, and then analyzed using 16 S rDNA sequencing and bioinformatics. RESULTS Although the Bacteroidetes/Firmicutes (B/F) ratio was higher in patients with RE or SARE, it alone could not predict these conditions. Three enterotypes were identified based on dominant genera: Blautia (enterotype 1), Escherichia-Shigella (enterotype 2), and Faecalibacterium (enterotype 3). A decrease in Blautia and an increase in Escherichia-Shigella and Faecalibacterium were correlated with RE and SARE. Univariate logistic regression revealed that the Faecalibacterium enterotype at the baseline was associated with a 4.4-fold higher risk of developing SARE (odds ratio 5.400; P = 0.017). The Escherichia-Shigella enterotype was also linked to increased SARE incidence. CONCLUSION These findings suggest that while single bacterial genera or the B/F ratio are insufficient predictors, enterotype classification may serve as a potential biomarker for predicting SARE in patients undergoing radiotherapy.
Collapse
Affiliation(s)
- Chenying Ma
- Department of Radiation Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Xiaoting Xu
- Department of Radiation Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Songbing Qin
- Department of Radiation Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Juying Zhou
- Department of Radiation Oncology, the First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, 215123, China.
| |
Collapse
|
17
|
Vasseur M, Lepers R, Langevin N, Julliand S, Grimm P. Fibrolytic efficiency of the large intestine microbiota may benefit running speed in French trotters: A pilot study. Physiol Rep 2024; 12:e70110. [PMID: 39533164 PMCID: PMC11557442 DOI: 10.14814/phy2.70110] [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: 09/26/2024] [Revised: 10/17/2024] [Accepted: 10/17/2024] [Indexed: 11/16/2024] Open
Abstract
This pilot study sought to explore the contribution of the large intestine microbiota to energy metabolism and exercise performance through its ability to degrade fibers into short-chain fatty acids (SCFAs). To investigate this, a correlational study was carried out on athlete horses under the same management conditions. Fecal microbiota diversity and composition, fibrolytic efficiency and SCFAs were analyzed. An incremental running test was carried out to estimate the maximal running speed (MRS) of the horses, and blood samples were taken to measure energy metabolism parameters. MRS was positively correlated with the efficiency of the fecal microbiota in degrading cellulose in vitro (r = 0.51; p = 0.02). The abundance of fibrolytic bacterial taxa was not associated with MRS, but functional inference analysis revealed a positive association between MRS and pathways potentially related to fibrolytic activity (r = 0.54; p = 0.07 and r = 0.56; p = 0.05 for butyrate metabolism and thiamine metabolism, respectively). In contrast, the metabolic pathway of starch degradation appeared negatively associated with MRS (r = -0.55; p = 0.06). The present findings suggest a potential contribution of the large intestine microbiota and dietary fibers digestion to exercise capacity in equine athletes.
Collapse
Affiliation(s)
- Maximilien Vasseur
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR 1093, Cognition Action et Plasticité Sensorimotrice (CAPS), Faculty of Sport SciencesUniversité de BourgogneDijonFrance
- Lab to FieldDijonFrance
| | - Romuald Lepers
- Institut National de la Santé et de la Recherche Médicale, Inserm UMR 1093, Cognition Action et Plasticité Sensorimotrice (CAPS), Faculty of Sport SciencesUniversité de BourgogneDijonFrance
| | | | | | | |
Collapse
|
18
|
Wang X, Wang P, Li Y, Guo H, Wang R, Liu S, Qiu J, Wang X, Hao Y, Zhao Y, Liao H, Zou Z, Thinwa J, Liu R. Procyanidin C1 Modulates the Microbiome to Increase FOXO1 Signaling and Valeric Acid Levels to Protect the Mucosal Barrier in Inflammatory Bowel Disease. ENGINEERING 2024; 42:108-120. [DOI: 10.1016/j.eng.2023.10.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
19
|
Chen SM, Guo BJ, Feng AQ, Wang XL, Zhang SL, Miao CY. Pathways regulating intestinal stem cells and potential therapeutic targets for radiation enteropathy. MOLECULAR BIOMEDICINE 2024; 5:46. [PMID: 39388072 PMCID: PMC11467144 DOI: 10.1186/s43556-024-00211-0] [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: 07/22/2024] [Accepted: 09/19/2024] [Indexed: 10/12/2024] Open
Abstract
Radiotherapy is a pivotal intervention for cancer patients, significantly impacting their treatment outcomes and survival prospects. Nevertheless, in the course of treating those with abdominal, pelvic, or retroperitoneal malignant tumors, the procedure inadvertently exposes adjacent intestinal tissues to radiation, posing risks of radiation-induced enteropathy upon reaching threshold doses. Stem cells within the intestinal crypts, through their controlled proliferation and differentiation, support the critical functions of the intestinal epithelium, ensuring efficient nutrient absorption while upholding its protective barrier properties. Intestinal stem cells (ISCs) regulation is intricately orchestrated by diverse signaling pathways, among which are the WNT, BMP, NOTCH, EGF, Hippo, Hedgehog and NF-κB, each contributing to the complex control of these cells' behavior. Complementing these pathways are additional regulators such as nutrient metabolic states, and the intestinal microbiota, all of which contribute to the fine-tuning of ISCs behavior in the intestinal crypts. It is the harmonious interplay among these signaling cascades and modulating elements that preserves the homeostasis of intestinal epithelial cells (IECs), thereby ensuring the gut's overall health and function. This review delves into the molecular underpinnings of how stem cells respond in the context of radiation enteropathy, aiming to illuminate potential biological targets for therapeutic intervention. Furthermore, we have compiled a summary of several current treatment methodologies. By unraveling these mechanisms and treatment methods, we aspire to furnish a roadmap for the development of novel therapeutics, advancing our capabilities in mitigating radiation-induced intestinal damage.
Collapse
Affiliation(s)
- Si-Min Chen
- Department of Pharmacology, Second Military Medical University/Naval Medical University, 325 Guo He Road, Shanghai, 200433, China
| | - Bing-Jie Guo
- Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - An-Qiang Feng
- Department of Digestive Disease, Xuzhou Central Hospital, Xuzhou, China
| | - Xue-Lian Wang
- School of Medicine, Shanghai University, Shanghai, China
| | - Sai-Long Zhang
- Department of Pharmacology, Second Military Medical University/Naval Medical University, 325 Guo He Road, Shanghai, 200433, China.
| | - Chao-Yu Miao
- Department of Pharmacology, Second Military Medical University/Naval Medical University, 325 Guo He Road, Shanghai, 200433, China.
| |
Collapse
|
20
|
Dai N, Yang X, Pan P, Zhang G, Sheng K, Wang J, Liang X, Wang Y. Bacillus paralicheniformis, an acetate-producing probiotic, alleviates ulcerative colitis via protecting the intestinal barrier and regulating the NLRP3 inflammasome. Microbiol Res 2024; 287:127856. [PMID: 39079268 DOI: 10.1016/j.micres.2024.127856] [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: 05/28/2024] [Revised: 07/11/2024] [Accepted: 07/22/2024] [Indexed: 08/22/2024]
Abstract
Ulcerative colitis (UC) presents a challenging scenario in digestive health, characterized by recurrent inflammation that is often hard to manage. Bacteria capable of producing short-chain fatty acids (SCFAs) play a pivotal role in mitigating UC symptoms, rendering them promising candidates for probiotic therapy. In this investigation, we assessed the impact of Bacillus paralicheniformis HMPM220325 on dextran sodium sulfate (DSS)-induced UC in mice. Genomic analysis of the strain revealed the presence of protease genes associated with acetate and butyrate synthesis, with acetic acid detected in its fermentation broth. Administration of B. paralicheniformis HMPM220325 to UC mice ameliorated pathological manifestations of the condition and restored intestinal barrier function. Furthermore, B. paralicheniformis HMPM220325 suppressed the activation of the NLRP3 inflammasome signaling pathway and modulated the composition of the intestinal microbiota. These findings shed significant light on the potential of B. paralicheniformis as a probiotic candidate, offering a novel avenue for the prevention and therapeutic intervention of colitis.
Collapse
Affiliation(s)
- Nini Dai
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Xinting Yang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Peilong Pan
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Guanghui Zhang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Kangliang Sheng
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Jingmin Wang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China
| | - Xiao Liang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China.
| | - Yongzhong Wang
- School of Life Sciences, Anhui University, Hefei, China; Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China; Anhui Province Joint Construction Discipline Key Laboratory of Nanobody Technology, Hefei, China.
| |
Collapse
|
21
|
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
|
22
|
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
|
23
|
Teng Y, Li J, Guo J, Yan C, Wang A, Xia X. Alginate oligosaccharide improves 5-fluorouracil-induced intestinal mucositis by enhancing intestinal barrier and modulating intestinal levels of butyrate and isovalerate. Int J Biol Macromol 2024; 276:133699. [PMID: 38972652 DOI: 10.1016/j.ijbiomac.2024.133699] [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: 01/16/2024] [Revised: 07/03/2024] [Accepted: 07/04/2024] [Indexed: 07/09/2024]
Abstract
Chemotherapy-induced mucositis (CIM) is the typical side effect of chemotherapy. This study investigates the potential of alginate oligosaccharide (AOS) in ameliorating CIM induced by 5-fluorouracil (5-FU) in a murine model and its underlying mechanisms. AOS effectively mitigated body weight loss and histopathological damage, modulated inflammatory cytokines and attenuated the oxidative stress. AOS restored intestinal barrier integrity through enhancing expression of tight junction proteins via MLCK signaling pathway. AOS alleviated intestinal mucosal damage by inhibiting TLR4/MyD88/NF-κB signaling pathway, downregulating the pro-apoptotic protein Bax and upregulating the anti-apoptotic protein Bcl-2. Moreover, AOS significantly enriched intestinal Akkermansiaceae and increased the production of short-chain fatty acids (SCFAs), most notably butyrate and isovalerate. Pre-treatment with butyrate and isovalerate also alleviated 5-FU-induced CIM. In conclusion, AOS effectively mitigated CIM through strenghthening intestinal barrier, attenuating inflammation, and modulating gut microbiota and intestianl levels of butyrate and isovalerate. These finding indicate that AOS could be potentially utilized as a supplemental strategy for prevention or mitigation of CIM.
Collapse
Affiliation(s)
- Yue Teng
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Jiahui Li
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Jian Guo
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Chunhong Yan
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Ailing Wang
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China
| | - Xiaodong Xia
- Dalian Jinshiwan Laboratory, Dalian, Liaoning 116034, China; State Key Laboratory of Marine Food Processing and Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian, Liaoning 116034, China.
| |
Collapse
|
24
|
Yan Z, Li Y, Xia T, Wang K, Liao Z, Zhang L, Wang Y, Shen P, Bai Z, Wang N, Zhou W, Ni Z, Dou Y, Gao Y. Revitalizing gut health: Liangxue guyuan yishen decoction promotes akkermansia muciniphila -induced intestinal stem cell recovery post-radiation in mice. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155888. [PMID: 39084128 DOI: 10.1016/j.phymed.2024.155888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 06/28/2024] [Accepted: 07/15/2024] [Indexed: 08/02/2024]
Abstract
BACKGROUND The efficacy of Liangxue Guyuan Yishen Decoction (LGYD), a traditional Chinese medicine, has been scientifically proven in the treatment of radiation-induced intestinal injury (RIII) and preservation of intestinal integrity and function following high-dose radiation exposure. However, further investigation is required to comprehensively elucidate the precise mechanisms underlying the therapeutic effects of LGYD in order to provide potential pharmaceutical options for radiation protection. PURPOSE This study aims to elucidate the potential mechanism through which LGYD exerts its therapeutic effects on RIII by modulating the gut microbiota (GM). METHODS 16 s rRNA analysis was employed to assess the impact of varying doses of whole body irradiation (WBI) on GM in order to establish an appropriate model for this study. The effects of LGYD on GM and SCFA were evaluated using 16 s rRNA and Quantification of SCFA. UHPLC-QE-MS was utilized to identify the active components in LGYD as well as LGYD drug containing serum (LGYD-DS). Subsequently, immunofluorescence and immunohistochemical staining were conducted to validate the influence of LGYD and/or characteristic microbiota on RIII recovery in vivo. The effects of LGYD-DS, characteristic flora, and SCFA on intestinal stem cell (ISC) were assessed by measuring organoid surface area in intestinal organoid model. RESULTS The species composition and abundance of GM were significantly influenced by whole-body irradiation with a dose of 8.5 Gy, which was used as in vivo model. LGYD significantly improves the survival rate and promotes recovery from RIII. Additionally, LGYD exhibited a notable increase in the abundance of Akkermansia muciniphila (AKK) and levels of SCFA, particularly isobutyric acid. LGYD-DS consisted of seven main components derived from herbs of LGYD. In vivo experiments indicated that both LGYD and AKK substantially enhanced the survival rate after radiation and facilitated the recovery process for intestinal structure and function. In the organoid model, treatment with LGYD-DS, AKK supernatant or isobutyric acid significantly increased organoid surface area. CONCLUSIONS LGYD has the potential to enhance RIII by promoting the restoration of intestinal stem cell, which is closely associated with the upregulation of AKK abundance and production of SCFA, particularly isobutyric acid.
Collapse
Affiliation(s)
- Ziqiao Yan
- Department of Traditional Chinese Medicine, the Sixth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China; Chinese PLA Medical School, Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China; Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Yangshuo Li
- Department of Traditional Chinese Gynecology, The First Affiliated Hospital of Naval Medical University, Shanghai, PR China
| | - Tiantian Xia
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China; Medical College of Qinghai University, Xining, PR China
| | - Kaili Wang
- Department of Traditional Chinese Medicine, the Sixth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China
| | - Zebin Liao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Liangliang Zhang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, PR China
| | - Yuguo Wang
- Department of Traditional Chinese Medicine, the Sixth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China
| | - Pan Shen
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Zhijie Bai
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Ningning Wang
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Wei Zhou
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China
| | - Zhexin Ni
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China.
| | - Yongqi Dou
- Department of Traditional Chinese Medicine, the Sixth Medical Center of Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China; Chinese PLA Medical School, Chinese People's Liberation Army (PLA) General Hospital, Beijing, PR China.
| | - Yue Gao
- Department of Pharmaceutical Sciences, Beijing Institute of Radiation Medicine, Beijing, PR China; Medical College of Qinghai University, Xining, PR China; School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou, PR China; State Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, PR China.
| |
Collapse
|
25
|
Lu L, Li F, Gao Y, Kang S, Li J, Guo J. Microbiome in radiotherapy: an emerging approach to enhance treatment efficacy and reduce tissue injury. Mol Med 2024; 30:105. [PMID: 39030525 PMCID: PMC11264922 DOI: 10.1186/s10020-024-00873-0] [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: 11/06/2023] [Accepted: 07/08/2024] [Indexed: 07/21/2024] Open
Abstract
Radiotherapy is a widely used cancer treatment that utilizes powerful radiation to destroy cancer cells and shrink tumors. While radiation can be beneficial, it can also harm the healthy tissues surrounding the tumor. Recent research indicates that the microbiota, the collection of microorganisms in our body, may play a role in influencing the effectiveness and side effects of radiation therapy. Studies have shown that specific species of bacteria living in the stomach can influence the immune system's response to radiation, potentially increasing the effectiveness of treatment. Additionally, the microbiota may contribute to adverse effects like radiation-induced diarrhea. A potential strategy to enhance radiotherapy outcomes and capitalize on the microbiome involves using probiotics. Probiotics are living microorganisms that offer health benefits when consumed in sufficient quantities. Several studies have indicated that probiotics have the potential to alter the composition of the gut microbiota, resulting in an enhanced immune response to radiation therapy and consequently improving the efficacy of the treatment. It is important to note that radiation can disrupt the natural balance of gut bacteria, resulting in increased intestinal permeability and inflammatory conditions. These disruptions can lead to adverse effects such as diarrhea and damage to the intestinal lining. The emerging field of radiotherapy microbiome research offers a promising avenue for optimizing cancer treatment outcomes. This paper aims to provide an overview of the human microbiome and its role in augmenting radiation effectiveness while minimizing damage.
Collapse
Affiliation(s)
- Lina Lu
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China.
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China.
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China.
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China.
| | - Fengxiao Li
- Department of Pharmacy, the Affiliated Hospital of Qingdao University, Qingdao, China
| | | | - Shuhe Kang
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| | - Jia Li
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| | - Jinwang Guo
- School of Chemical Engineering, Northwest Minzu University, No.1, Northwest New Village, Lanzhou, Gansu, 730030, China
- Key Laboratory of Environment-Friendly Composite Materials of the State Ethnic Affairs Commission, Lanzhou, Gansu, China
- Gansu Provincial Biomass Function Composites Engineering Research Center, Lanzhou, Gansu, China
- Key Laboratory for Utility of Environment-Friendly Composite Materials and Biomass in, University of Gansu Province, Lanzhou, Gansu, China
| |
Collapse
|
26
|
Sasaki M, Suaini NHA, Afghani J, Heye KN, O'Mahony L, Venter C, Lauener R, Frei R, Roduit C. Systematic review of the association between short-chain fatty acids and allergic diseases. Allergy 2024; 79:1789-1811. [PMID: 38391245 DOI: 10.1111/all.16065] [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: 09/26/2023] [Revised: 01/23/2024] [Accepted: 02/09/2024] [Indexed: 02/24/2024]
Abstract
We performed a systematic review to investigate the current evidence on the association between allergic diseases and short chain fatty acids (SCFAs), which are microbially produced and suggested as one mechanism on how gut microbiome affects the risk of allergic diseases. Medline, Embase and Web of Science were searched from data inception until September 2022. We identified 37 papers, of which 17 investigated prenatal or early childhood SCFAs and the development of allergic diseases in childhood, and 20 assessed SCFAs in patients with pre-existing allergic diseases. Study design, study populations, outcome definition, analysis method and reporting of the results varied between papers. Overall, there was some evidence showing that the three main SCFAs (acetate, propionate and butyrate) in the first few years of life had a protective effect against allergic diseases, especially for atopic dermatitis, wheeze or asthma and IgE-mediated food allergy in childhood. The association between each SCFA and allergic disease appeared to be different by disease and the age of assessment. Further research that can determine the potentially timing specific effect of each SCFA will be useful to investigate how SCFAs can be used in treatment or in prevention against allergic diseases.
Collapse
Affiliation(s)
- Mari Sasaki
- University Children's Hospital Zürich, Zürich, Switzerland
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, University of Bern, Bern, Switzerland
| | - Noor H A Suaini
- Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jamie Afghani
- Environmental Medicine Faculty of Medicine, University of Augsburg, Augsburg, Germany
- ZIEL-Institute for Food and Health, Technical University of Munich, Freising, Germany
- Institute of Environmental Medicine, Environmental Health Centre, Helmholtz Munich - German Research Centre for Environmental Health (GmbH), Neuherberg, Germany
| | - Kristina N Heye
- Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
| | - Liam O'Mahony
- Department of Medicine, University College Cork, National University of Ireland, Cork, Ireland
- APC Microbiome Ireland, University College Cork, National University of Ireland, Cork, Ireland
- School of Microbiology, University College Cork, National University of Ireland, Cork, Ireland
| | - Carina Venter
- Pediatric Allergy and Immunology, University of Colorado/Childrens Hospital Colorado, Aurora, Colorado, USA
| | - Roger Lauener
- Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| | - Remo Frei
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, University of Bern, Bern, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
- Department of Biomedical Research, University of Bern, Bern, Switzerland
| | - Caroline Roduit
- University Children's Hospital Zürich, Zürich, Switzerland
- Division of Respiratory Medicine and Allergology, Department of Paediatrics, Inselspital, University of Bern, Bern, Switzerland
- Children's Hospital of Eastern Switzerland, St. Gallen, Switzerland
- Christine Kühne Center for Allergy Research and Education (CK-CARE), Davos, Switzerland
| |
Collapse
|
27
|
Muradás TC, Freitas RDS, Gonçalves JIB, Xavier FAC, Marinowic DR. Potential antitumor effects of short-chain fatty acids in breast cancer models. Am J Cancer Res 2024; 14:1999-2019. [PMID: 38859825 PMCID: PMC11162650 DOI: 10.62347/etuq6763] [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: 10/30/2023] [Accepted: 03/13/2024] [Indexed: 06/12/2024] Open
Abstract
The effects of short-chain fatty acids (SCFAs) have been explored against cancer due to the crosstalk between gut microbiota alterations and the immune system as a crucial role in cancer development. We evaluated the SCFAs effects in both in vitro and in vivo breast cancer models. In vitro, the SCFAs displayed contrasting effects on viability index, according to the evaluation of breast cancer cells with different phenotypes, human MCF-7, SK-BR-3, MDA-MD-231, or the mouse 4T1 lineage. Acetate displayed minimal effects at concentrations up to 100 mM. Alternatively, propionate increases or reduces cell viability depending on the concentration. Butyrate and valerate showed consistent time- and concentration-dependent effects on the viability of human or mouse breast cancer cells. The selective FFA2 4-CMTB or FFA3 AR420626 receptor agonists failed to overtake the SCFA actions, except by modest inhibitory effects on MDA-MB-231 and 4T1 cell viability. The FFA2 CATPB or FFA3 and β-hydroxybutyrate receptor antagonists lacked significant activity on human cell lines, although CATPB reduced 4T1 cell viability. Butyrate significantly affected cell morphology, clonogenicity, and migration, according to the evaluation of MDA-MB-231 and 4T1 cells. A preliminary examination of in vivo oral effects of butyrate, propionate, or valerate, dosed in prophylactic or therapeutic regimens, on several parameters evaluated in an orthotopic breast cancer model showed a reduction of lung metastasis in post-tumor induction butyrate-treated mice. Overall, the present results indicate that in vitro effects of SCFAs did not rely on FFA2 or FFA3 receptor activation, and they were not mirrored in vivo, at least at the tested conditions. Overall, the present results indicate potential in vitro inhibitory effects of SCFAs in breast cancer, independent of FFA2 or FFA3 receptor activation, and, in the metastatic breast cancer model, the butyrate-dosed therapeutic regimen reduced the number of lung metastases.
Collapse
Affiliation(s)
- Thaís C Muradás
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do SulPorto Alegre, RS, Brazil
| | - Raquel DS Freitas
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do SulPorto Alegre, RS, Brazil
| | - João IB Gonçalves
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do SulPorto Alegre, RS, Brazil
- Brain Institute of Rio Grande do SulPorto Alegre, RS, Brazil
| | - Fernando AC Xavier
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do SulPorto Alegre, RS, Brazil
- Brain Institute of Rio Grande do SulPorto Alegre, RS, Brazil
| | - Daniel R Marinowic
- Programa de Pós-graduação em Medicina e Ciências da Saúde, Escola de Medicina, Pontifícia Universidade Católica do Rio Grande do SulPorto Alegre, RS, Brazil
- Brain Institute of Rio Grande do SulPorto Alegre, RS, Brazil
| |
Collapse
|
28
|
Horseman T, Rittase WB, Slaven JE, Bradfield DT, Frank AM, Anderson JA, Hays EC, Ott AC, Thomas AE, Huppmann AR, Lee SH, Burmeister DM, Day RM. Ferroptosis, Inflammation, and Microbiome Alterations in the Intestine in the Göttingen Minipig Model of Hematopoietic-Acute Radiation Syndrome. Int J Mol Sci 2024; 25:4535. [PMID: 38674120 PMCID: PMC11050692 DOI: 10.3390/ijms25084535] [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: 03/06/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Hematopoietic acute radiation syndrome (H-ARS) involves injury to multiple organ systems following total body irradiation (TBI). Our laboratory demonstrated that captopril, an angiotensin-converting enzyme inhibitor, mitigates H-ARS in Göttingen minipigs, with improved survival and hematopoietic recovery, as well as the suppression of acute inflammation. However, the effects of captopril on the gastrointestinal (GI) system after TBI are not well known. We used a Göttingen minipig H-ARS model to investigate captopril's effects on the GI following TBI (60Co 1.79 or 1.80 Gy, 0.42-0.48 Gy/min), with endpoints at 6 or 35 days. The vehicle or captopril (0.96 mg/kg) was administered orally twice daily for 12 days, starting 4 h post-irradiation. Ilea were harvested for histological, protein, and RNA analyses. TBI increased congestion and mucosa erosion and hemorrhage, which were modulated by captopril. GPX-4 and SLC7A11 were downregulated post-irradiation, consistent with ferroptosis at 6 and 35 days post-irradiation in all groups. Interestingly, p21/waf1 increased at 6 days in vehicle-treated but not captopril-treated animals. An RT-qPCR analysis showed that radiation increased the gene expression of inflammatory cytokines IL1B, TNFA, CCL2, IL18, and CXCL8, and the inflammasome component NLRP3. Captopril suppressed radiation-induced IL1B and TNFA. Rectal microbiome analysis showed that 1 day of captopril treatment with radiation decreased overall diversity, with increased Proteobacteria phyla and Escherichia genera. By 6 days, captopril increased the relative abundance of Enterococcus, previously associated with improved H-ARS survival in mice. Our data suggest that captopril mitigates senescence, some inflammation, and microbiome alterations, but not ferroptosis markers in the intestine following TBI.
Collapse
Affiliation(s)
- Timothy Horseman
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (T.H.); (D.M.B.)
| | - W. Bradley Rittase
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (W.B.R.); (J.E.S.); (D.T.B.)
| | - John E. Slaven
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (W.B.R.); (J.E.S.); (D.T.B.)
| | - Dmitry T. Bradfield
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (W.B.R.); (J.E.S.); (D.T.B.)
| | - Andrew M. Frank
- Department of Anatomy, Physiology and Genetics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA;
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD 20817, USA
| | - Joseph A. Anderson
- Comparative Pathology Division, Department of Laboratory Animal Resources, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Evelyn C. Hays
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (W.B.R.); (J.E.S.); (D.T.B.)
| | - Andrew C. Ott
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (W.B.R.); (J.E.S.); (D.T.B.)
| | - Anjali E. Thomas
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (W.B.R.); (J.E.S.); (D.T.B.)
| | - Alison R. Huppmann
- Department of Biomedical Sciences, University of South Carolina School of Medicine, Greenville, SC 29605, USA;
| | - Sang-Ho Lee
- Pathology Department, Research Services, Naval Medical Research Center, Silver Spring, MD 20910, USA;
| | - David M. Burmeister
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (T.H.); (D.M.B.)
| | - Regina M. Day
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD 20814, USA; (W.B.R.); (J.E.S.); (D.T.B.)
| |
Collapse
|
29
|
Liu M, Zhang Y, Liu J, Xiang C, Lu Q, Lu H, Yang T, Wang X, Zhang Q, Fan C, Feng C, Zou D, Li H, Tang W. Revisiting the Role of Valeric Acid in Manipulating Ulcerative Colitis. Inflamm Bowel Dis 2024; 30:617-628. [PMID: 38206334 DOI: 10.1093/ibd/izad187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Ulcerative colitis (UC) is characterized by a complicated interaction between mucosal inflammation, epithelial dysfunction, abnormal activation of innate immune responses, and gut microbiota dysbiosis. Though valeric acid (VA), one type of short-chain fatty acids (SCFAs), has been identified in other inflammatory disorders and cancer development, the pathological role of VA and underlying mechanism of VA in UC remain under further investigation. METHODS Studies of human clinical specimens and experimental colitis models were conducted to confirm the pathological manifestations of the level of SCFAs from human fecal samples and murine colonic homogenates. Valeric acid-intervened murine colitis and a macrophage adoptive transfer were applied to identify the underlying mechanisms. RESULTS In line with gut microbiota dysfunction in UC, alteration of SCFAs from gut microbes were identified in human UC patients and dextran sodium sulfate -induced murine colitis models. Notably, VA was consistently negatively related to the disease severity of UC, the population of monocytes, and the level of interluekin-6. Moreover, VA treatment showed direct suppressive effects on lipopolysaccharides (LPS)-activated human peripheral blood mononuclear cells and murine macrophages in the dependent manner of upregulation of GPR41 and GPR43. Therapeutically, replenishment of VA or adoptive transfer with VA-modulated macrophages showed resistance to dextran sodium sulfate-driven murine colitis though modulating the production of inflammatory cytokine interleukin-6. CONCLUSIONS In summary, the research uncovered the pathological role of VA in modulating the activation of macrophages in UC and suggested that VA might be a potential effective agent for UC patients.
Collapse
Affiliation(s)
- Moting Liu
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yao Zhang
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Jia Liu
- Institutional Technology Service Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Caigui Xiang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qiukai Lu
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huimin Lu
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Yang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xiaohan Wang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Qingli Zhang
- Institutional Technology Service Center, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Chen Fan
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Chunlan Feng
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Duowu Zou
- Department of Gastroenterology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Heng Li
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Wei Tang
- Laboratory of Anti-inflammation and Immunopharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
- School of Pharmacy, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
30
|
Chen R, Li K, Wang Y, Song L, Wang R, Fan W, Zhao N, Zou W, Yang Z, Yan J. Valeric acid reduction by chitosan oligosaccharide induces autophagy in a Parkinson's disease mouse model. J Drug Target 2024; 32:423-432. [PMID: 38315456 DOI: 10.1080/1061186x.2024.2315468] [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: 10/18/2023] [Accepted: 01/30/2024] [Indexed: 02/07/2024]
Abstract
Parkinson's disease (PD) is a central nervous system disease with the highest disability and mortality rate worldwide, and it is caused by a variety of factors. The most common medications for PD have side effects with limited therapeutic outcomes. Many studies have reported that chitosan oligosaccharide (COS) crossed blood-brain barrier to achieve a neuroprotective effect in PD. However, the role of COS in PD remains unclear. The present study demonstrated that COS increased dopaminergic neurons in the substantia nigra (SN) and ameliorated dyskinesia in a PD mouse model. Moreover, COS reduced gut microbial diversity and faecal short-chain fatty acids. Valeric acid supplementation enhanced the inflammatory response in the colon and SN, and it reversed COS - suppressed dopamine neurons damage. Autophagy was involved in COS modulating inflammation through valeric acid. These results suggest that COS reduces bacterial metabolites - valeric acid, which diminishes inflammation via activating autophagy, ultimately alleviating PD.
Collapse
Affiliation(s)
- Rongsha Chen
- Center Laboratory of the Second Hospital affiliated, Kunming Medical University, Kunming, Yunnan, China
| | - Ke Li
- Center Laboratory of the Second Hospital affiliated, Kunming Medical University, Kunming, Yunnan, China
| | - Yinying Wang
- Center Laboratory of the Second Hospital affiliated, Kunming Medical University, Kunming, Yunnan, China
- Yunnan Provincial Key Laboratory of Molecular Biology for Sino medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Liyun Song
- Yunnan Provincial Key Laboratory of Molecular Biology for Sino medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Ruohua Wang
- Center Laboratory of the Second Hospital affiliated, Kunming Medical University, Kunming, Yunnan, China
| | - Wenhui Fan
- Center Laboratory of the Second Hospital affiliated, Kunming Medical University, Kunming, Yunnan, China
| | - Ninghui Zhao
- Neurosurgery department of the Second Hospital affiliated, Kunming Medical University, Kunming, Yunnan, China
| | - Wei Zou
- School of Public Health, Kunming Medical University, Kunming, Yunnan, China
| | - Zhongshan Yang
- Yunnan Provincial Key Laboratory of Molecular Biology for Sino medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Jinyuan Yan
- Center Laboratory of the Second Hospital affiliated, Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
31
|
Dong J, Wang B, Xiao Y, Liu J, Wang Q, Xiao H, Jin Y, Liu Z, Chen Z, Li Y, Fan S, Li Y, Cui M. Roseburia intestinalis sensitizes colorectal cancer to radiotherapy through the butyrate/OR51E1/RALB axis. Cell Rep 2024; 43:113846. [PMID: 38412097 DOI: 10.1016/j.celrep.2024.113846] [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: 05/22/2023] [Revised: 10/30/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
The radioresistant signature of colorectal cancer (CRC) hampers the clinical utility of radiotherapy. Here, we find that fecal microbiota transplantation (FMT) potentiates the tumoricidal effects of radiation and degrades the intertwined adverse events in azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced CRC mice. FMT cumulates Roseburia intestinalis (R. intestinalis) in the gastrointestinal tract. Oral gavage of R. intestinalis assembles at the CRC site and synthetizes butyrate, sensitizing CRC to radiation and alleviating intestinal toxicity in primary and CRC hepatic metastasis mouse models. R. intestinalis-derived butyrate activates OR51E1, a G-protein-coupled receptor overexpressing in patients with rectal cancer, facilitating radiogenic autophagy in CRC cells. OR51E1 shows a positive correlation with RALB in clinical rectal cancer tissues and CRC mouse model. Blockage of OR51E1/RALB signaling restrains butyrate-elicited autophagy in irradiated CRC cells. Our findings highlight that the gut commensal bacteria R. intestinalis motivates radiation-induced autophagy to accelerate CRC cell death through the butyrate/OR51E1/RALB axis and provide a promising radiosensitizer for CRC in a pre-clinical setting.
Collapse
Affiliation(s)
- Jiali Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Bin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yunong Xiao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Jia Liu
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qi Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Huiwen Xiao
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuxiao Jin
- Department of Anesthesiology, Changshu No. 2 People's Hospital, Changshu, Jiangsu Province 215501, China
| | - Zhihong Liu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215004, China
| | - Zhiyuan Chen
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yiliang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
32
|
Song I, Yang J, Saito M, Hartanto T, Nakayama Y, Ichinohe T, Fukuda S. Prebiotic inulin ameliorates SARS-CoV-2 infection in hamsters by modulating the gut microbiome. NPJ Sci Food 2024; 8:18. [PMID: 38485724 PMCID: PMC10940623 DOI: 10.1038/s41538-024-00248-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 01/16/2024] [Indexed: 03/18/2024] Open
Abstract
Current treatment options for COVID-19 are limited, with many antivirals and immunomodulators restricted to the most severe cases and preventative care limited to vaccination. As the SARS-CoV-2 virus and its increasing variants threaten to become a permanent fixture of our lives, this new reality necessitates the development of cost-effective and accessible treatment options for COVID-19. Studies have shown that there are correlations between the gut microbiome and severity of COVID-19, especially with regards to production of physiologically beneficial short-chain fatty acids (SCFAs) by gut microbes. In this study, we used a Syrian hamster model to study how dietary consumption of the prebiotic inulin affected morbidity and mortality resulting from SARS-CoV-2 infection. After two weeks of observation, we discovered that inulin supplementation attenuated morbid weight loss and increased survival rate in hamster subjects. An analysis of microbiome community structure showed significant alterations in 15 genera. Notably, there were also small increases in fecal DCA and a significant increase in serum DCA, perhaps highlighting a role for this secondary bile acid in conferring protection against SARS-CoV-2. In light of these results, inulin and other prebiotics are promising targets for future investigation as preventative treatment options for COVID-19.
Collapse
Affiliation(s)
- Isaiah Song
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Jiayue Yang
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan
| | - Misa Saito
- Metagen, Inc., Tsuruoka, Yamagata, Japan
| | | | | | - Takeshi Ichinohe
- Division of Viral Infection, Department of Infectious Disease Control, International Research Center for Infectious Diseases, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Shinji Fukuda
- Institute for Advanced Biosciences, Keio University, Tsuruoka, Yamagata, Japan.
- Metagen, Inc., Tsuruoka, Yamagata, Japan.
- Gut Environmental Design Group, Kanagawa Institute of Industrial Science and Technology, Kawasaki, Kanagawa, Japan.
- Transborder Medical Research Center, University of Tsukuba, Tsukuba, Ibaraki, Japan.
- Laboratory for Regenerative Microbiology, Juntendo University Graduate School of Medicine, Tokyo, Japan.
| |
Collapse
|
33
|
Wu L, Chen L, Li H, Wang Y, Xu K, Chen W, Zhang A, Wang Y, Shi C. Nocardia rubra cell-wall skeleton mitigates whole abdominal irradiation-induced intestinal injury via regulating macrophage function. BURNS & TRAUMA 2024; 12:tkad045. [PMID: 38444637 PMCID: PMC10914217 DOI: 10.1093/burnst/tkad045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 04/13/2023] [Accepted: 08/16/2023] [Indexed: 03/07/2024]
Abstract
Background Ionizing radiation (IR)-induced intestinal injury is a major side effect and dose-limiting toxicity in patients receiving radiotherapy. There is an urgent need to identify an effective and safe radioprotectant to reduce radiation-induced intestinal injury. Immunoregulation is considered an effective strategy against IR-induced injury. The purpose of this article was to investigate the protective effect of Nocardia rubra cell wall skeleton (Nr-CWS), an immunomodulator, on radiation-induced intestinal damage and to explore its potential mechanism. Methods C57BL/6 J male mice exposed to 12 Gy whole abdominal irradiation (WAI) were examined for survival rate, morphology and function of the intestine and spleen, as well as the gut microbiota, to comprehensively evaluate the therapeutic effects of Nr-CWS on radiation-induced intestinal and splenetic injury. To further elucidate the underlying mechanisms of Nr-CWS-mediated intestinal protection, macrophages were depleted by clodronate liposomes to determine whether Nr-CWS-induced radioprotection is macrophage dependent, and the function of peritoneal macrophages stimulated by Nr-CWS was detected in vitro. Results Our data showed that Nr-CWS promoted the recovery of intestinal barrier function, enhanced leucine-rich repeat-containing G protein-coupled receptor 5+ intestinal stem cell survival and the regeneration of intestinal epithelial cells, maintained intestinal flora homeostasis, protected spleen morphology and function, and improved the outcome of mice exposed to 12 Gy WAI. Mechanistic studies indicated that Nr-CWS recruited macrophages to reduce WAI-induced intestinal damage. Moreover, macrophage depletion by clodronate liposomes blocked Nr-CWS-induced radioprotection. In vitro, we found that Nr-CWS activated the nuclear factor kappa-B signaling pathway and promoted the phagocytosis and migration ability of peritoneal macrophages. Conclusions Our study suggests the therapeutic effect of Nr-CWS on radiation-induced intestinal injury, and provides possible therapeutic strategy and potential preventive and therapeutic drugs to alleviate it.
Collapse
Affiliation(s)
- Lingling Wu
- Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Long Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Huijuan Li
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Yawei Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Kexin Xu
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), 400038, Chongqing, China
- College of Biological Engineering, Chongqing University 400044, Chongqing, China
| | - Wanchao Chen
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Aihua Zhang
- Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025, China
| | - Yu Wang
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| | - Chunmeng Shi
- Department of Toxicology, School of Public Health, Guizhou Medical University, Guiyang, 550025, China
- State Key Laboratory of Trauma and Chemical Poisoning, Third Military Medical University (Army Medical University), 400038, Chongqing, China
| |
Collapse
|
34
|
Barman M, Gio-Batta M, Andrieux L, Stråvik M, Saalman R, Fristedt R, Rabe H, Sandin A, Wold AE, Sandberg AS. Short-chain fatty acids (SCFA) in infants' plasma and corresponding mother's milk and plasma in relation to subsequent sensitisation and atopic disease. EBioMedicine 2024; 101:104999. [PMID: 38340558 PMCID: PMC10869761 DOI: 10.1016/j.ebiom.2024.104999] [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: 07/02/2023] [Revised: 01/19/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Short-chain fatty acids (SCFAs) in intestinal contents may influence immune function, while less is known about SCFAs in blood plasma. The aims were to investigate the relation between infants' and maternal plasma SCFAs, as well as SCFAs in mother's milk, and relate SCFA concentrations in infant plasma to subsequent sensitisation and atopic disease. METHODS Infant plasma (N = 148) and corresponding mother's milk and plasma were collected four months postpartum. Nine SCFA (formic, acetic, propionic, isobutyric, butyric, succinic, valeric, isovaleric, and caproic acid) were analysed by UPLC-MS. At 12 months of age, atopic disease was diagnosed by a pediatric allergologist, and sensitisation was measured by skin prick test. All families participated in the Swedish birth cohort NICE (Nutritional impact on Immunological maturation during Childhood in relation to the Environment). FINDINGS Infants with sensitisation, atopic eczema, or food allergy had significantly lower concentrations of five, three, and two SCFAs, respectively, in plasma at four months. Logistic regressions models showed significant negative associations between formic, succinic, and caproic acid and sensitisation [ORadj (95% CI) per SD: 0.41 (0.19-0.91); 0.19 (0.05-0.75); 0.25 (0.09-0.66)], and between acetic acid and atopic eczema [0.42 (0.18-0.95)], after adjusting for maternal allergy. Infants' and maternal plasma SCFA concentrations correlated strongly, while milk SCFA concentrations were unrelated to both. Butyric and caproic acid concentrations were enriched around 100-fold, and iso-butyric and valeric acid around 3-5-fold in mother's milk, while other SCFAs were less prevalent in milk than in plasma. INTERPRETATION Butyric and caproic acid might be actively transported into breast milk to meet the needs of the infant, although mechanistic studies are needed to confirm this. The negative associations between certain SCFAs on sensitisation and atopic disease adds to prior evidence regarding their immunoregulatory potential. FUNDING Swedish Research Council (Nr. 2013-3145, 2019-0137 and 2023-02217 to A-S.S.), Swedish Research Council for Health, Working Life and Welfare FORTE, Nr 2018-00485 to A.W.), The Swedish Asthma and Allergy Association's Research Fund (2020-0020 to A.S.).
Collapse
Affiliation(s)
- Malin Barman
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden.
| | - Monica Gio-Batta
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Léna Andrieux
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden; Département de Biologie, École Normale Supérieure de Lyon, Université Claude Bernard Lyon 1, 69342 Lyon Cedex 07, France
| | - Mia Stråvik
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Robert Saalman
- Institute of Clinical Sciences, Department of Pediatrics, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Rikard Fristedt
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden
| | - Hardis Rabe
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Anna Sandin
- Department of Clinical Science, Pediatrics, Sunderby Research Unit, Umeå University, Umeå 901 87, Sweden
| | - Agnes E Wold
- Institute of Biomedicine, Department of Infectious Diseases, Sahlgrenska Academy, University of Gothenburg, Gothenburg 405 30, Sweden
| | - Ann-Sofie Sandberg
- Department of Life Sciences, Food and Nutrition Science, Chalmers University of Technology, Gothenburg 412 96, Sweden
| |
Collapse
|
35
|
Gracia MI, Vazquez P, Ibáñez-Pernía Y, Pos J, Tawde S. Performance Evaluation of a Novel Combination of Four- and Five-Carbon [Butyric and Valeric] Short-Chain Fatty Acid Glyceride Esters in Broilers. Animals (Basel) 2024; 14:617. [PMID: 38396585 PMCID: PMC10885893 DOI: 10.3390/ani14040617] [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: 01/09/2024] [Revised: 02/08/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024] Open
Abstract
A novel combination of Butyric and Valeric acid glycerol esters with oregano oil in a dry powder form was evaluated for performance improvements in broilers. The dosing regimen (500 g/Ton feed in starter and grower; 250 g/Ton in finisher feed) was considered low compared to conventional practices using non-esterified Butyric and Valeric short-chain fatty acids (SCFA). Six trials were conducted at various trial facilities in Italy, United Kingdom, Spain, and Poland. Supplemented broilers weighed significantly more than the control birds at 28 days of age (+3.4%; 1459 g vs. 1412 g; p = 0.0006) and at 42 days of age (+2.5%; 2834 g vs. 2763 g; p = 0.0030). Supplementation significantly reduced mortality from 1.9% to 0.8% during the finisher phase (from 29 to 42 days of age); however, average mortality was 3.2% for the whole 42-day growth period and was not affected. Further, supplemented broilers grew more (66.4 vs. 64.5 g/day; p = 0.0005), ate more feed (104.7 vs. 103.1 g/day; p = 0.0473), converted feed significantly more efficiently (1.58 vs. 1.60; p = 0.0072), leading to better EPEF value (410 vs. 389; p = 0.0006) than the control broilers. Meta-analysed trial performance data for novel SCFA formulations such as these are not commonly available, and serve to facilitate efficacy determination from an end-user perspective. The use of short- and medium-chain fatty acid esters in optimal low-dose combinations to reliably augment gut health and performance appears promising in commercial broiler production, and may lead to further improvements in industry practices and reduced antibiotic use.
Collapse
Affiliation(s)
- Marta I. Gracia
- Imasde Agroalimentaria, S.L. C/Nápoles 3, 28224 Pozuelo de Alarcón, Spain; (P.V.); (Y.I.-P.)
| | - Patricia Vazquez
- Imasde Agroalimentaria, S.L. C/Nápoles 3, 28224 Pozuelo de Alarcón, Spain; (P.V.); (Y.I.-P.)
| | - Yolanda Ibáñez-Pernía
- Imasde Agroalimentaria, S.L. C/Nápoles 3, 28224 Pozuelo de Alarcón, Spain; (P.V.); (Y.I.-P.)
| | - Jeroen Pos
- Perstorp Animal Nutrition, Perstorp Waspik BV, 5165 NH Waspik, The Netherlands; (J.P.); (S.T.)
| | - Snehal Tawde
- Perstorp Animal Nutrition, Perstorp Waspik BV, 5165 NH Waspik, The Netherlands; (J.P.); (S.T.)
| |
Collapse
|
36
|
Liu X, Li Y, Gu M, Xu T, Wang C, Chang P. Radiation enteropathy-related depression: A neglectable course of disease by gut bacterial dysbiosis. Cancer Med 2024; 13:e6865. [PMID: 38457257 PMCID: PMC10923036 DOI: 10.1002/cam4.6865] [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: 07/10/2023] [Revised: 12/08/2023] [Accepted: 12/12/2023] [Indexed: 03/10/2024] Open
Abstract
Radiation enteropathy (RE) is common in patients treated with radiotherapy for pelvic-abdominal cancers. Accumulating data indicate that gut commensal bacteria determine intestinal radiosensitivity. Radiotherapy can result in gut bacterial dysbiosis. Gut bacterial dysbiosis contributes to the pathogenesis of RE. Mild to moderate depressive symptoms can be observed in patients with RE in clinical settings; however, the rate of these symptoms has not been reported. Studies have demonstrated that gut bacterial dysbiosis induces depression. In the state of comorbidity, RE and depression may be understood as local and abscopal manifestations of gut bacterial disorders. The ability of comorbid depression to worsen inflammatory bowel disease (IBD) has long been demonstrated and is associated with dysfunction of cholinergic neural anti-inflammatory pathways. There is a lack of direct evidence for RE comorbid with depression. It is widely accepted that RE shares similar pathophysiologic mechanisms with IBD. Therefore, we may be able to draw on the findings of the relationship between IBD and depression. This review will explore the relationship between gut bacteria, RE, and depression in light of the available evidence and indicate a method for investigating the mechanisms of RE combined with depression. We will also describe new developments in the treatment of RE with probiotics, prebiotics, and fecal microbial transplantation.
Collapse
Affiliation(s)
- Xinliang Liu
- Department of Radiation Oncology and TherapyThe First Hospital of Jilin UniversityChangchunChina
| | - Ying Li
- Department of Radiation Oncology and TherapyThe First Hospital of Jilin UniversityChangchunChina
| | - Meichen Gu
- Department of Radiation Oncology and TherapyThe First Hospital of Jilin UniversityChangchunChina
| | - Tiankai Xu
- Department of Radiation Oncology and TherapyThe First Hospital of Jilin UniversityChangchunChina
| | - Chuanlei Wang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery CenterThe First Hospital of Jilin UniversityChangchunChina
| | - Pengyu Chang
- Department of Radiation Oncology and TherapyThe First Hospital of Jilin UniversityChangchunChina
| |
Collapse
|
37
|
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
|
38
|
Zhang H, Dong M, Zheng J, Yang Y, He J, Liu T, Wei H. Fecal bacteria-free filtrate transplantation is proved as an effective way for the recovery of radiation-induced individuals in mice. Front Cell Infect Microbiol 2024; 13:1343752. [PMID: 38357210 PMCID: PMC10864540 DOI: 10.3389/fcimb.2023.1343752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 12/27/2023] [Indexed: 02/16/2024] Open
Abstract
Background Ionizing radiation can cause intestinal microecological dysbiosis, resulting in changes in the composition and function of gut microbiota. Altered gut microbiota is closely related to the development and progression of radiation-induced intestinal damage. Although microbiota-oriented therapeutic options such as fecal microbiota transplantation (FMT) have shown some efficacy in treating radiation toxicity, safety concerns endure. Therefore, fecal bacteria-free filtrate transplantation (FFT), which has the potential to become a possible alternative therapy, is well worth investigating. Herein, we performed FFT in a mouse model of radiation exposure and monitored its effects on radiation damage phenotypes, gut microbiota, and metabolomic profiles to assess the effectiveness of FFT as an alternative therapy to FMT safety concerns. Results FFT treatment conferred radioprotection against radiation-induced toxicity, representing as better intestinal integrity, robust proinflammatory and anti-inflammatory cytokines homeostasis, and accompanied by significant shifts in gut microbiome. The bacterial compartment of recipients following FFT was characterized by an enrichment of radioprotective microorganisms (members of family Lachnospiraceae). Furthermore, metabolome data revealed increased levels of microbially generated short-chain fatty acids (SCFAs) in the feces of FFT mice. Conclusions FFT improves radiation-induced intestinal microecological dysbiosis by reshaping intestinal mucosal barrier function, gut microbiota configurations, and host metabolic profiles, highlighting FFT regimen as a promising safe alternative therapy for FMT is effective in the treatment of radiation intestinal injury.
Collapse
Affiliation(s)
- Hang Zhang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Technology, College of Animal Medicine, Huazhong Agricultural University, Wuhan, China
| | - Miaomiao Dong
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Technology, College of Animal Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jixia Zheng
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Technology, College of Animal Medicine, Huazhong Agricultural University, Wuhan, China
| | - Yapeng Yang
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Technology, College of Animal Medicine, Huazhong Agricultural University, Wuhan, China
| | - Jinhui He
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Technology, College of Animal Medicine, Huazhong Agricultural University, Wuhan, China
| | - Tianhao Liu
- Department of Gastroenterology, Affiliated Hospital of Jiangnan University, Wuxi, China
| | - Hong Wei
- State Key Laboratory of Agricultural Microbiology, College of Animal Science and Technology, College of Animal Medicine, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
39
|
Russo E, Bellando-Randone S, Carboni D, Fioretto BS, Romano E, Baldi S, El Aoufy K, Ramazzotti M, Rosa I, Lepri G, Di Gloria L, Pallecchi M, Bruni C, Melchiorre D, Guiducci S, Manetti M, Bartolucci GL, Matucci-Cerinic M, Amedei A. The differential crosstalk of the skin-gut microbiome axis as a new emerging actor in systemic sclerosis. Rheumatology (Oxford) 2024; 63:226-234. [PMID: 37154625 DOI: 10.1093/rheumatology/kead208] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 03/29/2023] [Accepted: 04/23/2023] [Indexed: 05/10/2023] Open
Abstract
OBJECTIVES We characterized the microbiota in SSc, focusing on the skin-oral-gut axis and the serum and faecal free fatty acid (FFA) profile. METHODS Twenty-five SSc patients with ACA or anti-Scl70 autoantibodies were enrolled. The microbiota of faecal, saliva and superficial epidermal samples was assessed through next-generation sequencing analysis. GC-MS was used to quantify faecal and serum FFAs. Gastrointestinal symptoms were investigated with the University of California Los Angeles Scleroderma Clinical Trial Consortium Gastrointestinal Tract Instrument (UCLA GIT-2.0) questionnaire. RESULTS The ACA+ and anti-Scl70+ groups displayed different cutaneous and faecal microbiota profiles. The classes of cutaneous Sphingobacteriia and Alphaproteobacteria, the faecal phylum Lentisphaerae, the levels of the classes Lentisphaeria and Opitutae, and the genus NA-Acidaminococcaceae were significantly higher in faecal samples from the ACA+ patients than in samples from the anti-Scl70+ patients. The cutaneous Sphingobacteria and the faecal Lentisphaerae were significantly correlated (rho = 0.42; P = 0.03). A significant increase in faecal propionic acid was observed in ACA+ patients. Moreover, all levels of faecal medium-chain FFAs and hexanoic acids were significantly higher in the ACA+ group than in the anti-Scl70+ group (P < 0.05 and P < 0.001, respectively). In the ACA+ group, the analysis of the serum FFA levels showed an increasing trend in valeric acid. CONCLUSION Different microbiota signatures and FFA profiles were found for the two groups of patients. Despite being in different body districts, the cutaneous Sphingobacteria and faecal Lentisphaerae appear interdependent.
Collapse
Affiliation(s)
- Edda Russo
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Silvia Bellando-Randone
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Division of Rheumatology, Scleroderma Unit, AOU Careggi, Florence, Italy
| | - Davide Carboni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Division of Rheumatology, Scleroderma Unit, AOU Careggi, Florence, Italy
| | | | - Eloisa Romano
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Simone Baldi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Khadija El Aoufy
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Matteo Ramazzotti
- Department of Biomedical, Experimental and Clinical Science "Mario Serio", University of Florence, Florence, Italy
| | - Irene Rosa
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Gemma Lepri
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Division of Rheumatology, Scleroderma Unit, AOU Careggi, Florence, Italy
| | - Leandro Di Gloria
- Department of Biomedical, Experimental and Clinical Science "Mario Serio", University of Florence, Florence, Italy
| | - Marco Pallecchi
- Department of Neurosciences, Psychology, Drug Research and Child Health, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Florence, Italy
| | - Cosimo Bruni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Division of Rheumatology, Scleroderma Unit, AOU Careggi, Florence, Italy
- Rheumatology, University Hospital of Zürich, Zürich, Switzerland
| | - Daniela Melchiorre
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Serena Guiducci
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mirko Manetti
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Gian Luca Bartolucci
- Department of Neurosciences, Psychology, Drug Research and Child Health, Section of Pharmaceutical and Nutraceutical Sciences, University of Florence, Florence, Italy
| | - Marco Matucci-Cerinic
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Division of Rheumatology, Scleroderma Unit, AOU Careggi, Florence, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases, San Raffaele Hospital, Milan, Italy
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| |
Collapse
|
40
|
Han L, Li Q, Du M, Mao X. Bovine milk osteopontin improved intestinal health of pregnant rats fed a high-fat diet through improving bile acid metabolism. J Dairy Sci 2024; 107:24-39. [PMID: 37690710 DOI: 10.3168/jds.2023-23802] [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: 05/27/2023] [Accepted: 07/31/2023] [Indexed: 09/12/2023]
Abstract
The main purpose of the current study was to investigate the ameliorative effects of bovine milk osteopontin (bmOPN) on the gut dysfunction of pregnant rats fed a high-fat diet (HFD). Bovine milk osteopontin was supplemented at a dose of 6 mg/kg body weight. Bovine milk osteopontin supplementation during pregnancy reduced colonic inflammation of HFD dams, and it also increased the colonic expression of ZO-1 and claudin-4 of HFD dams. Bovine milk osteopontin significantly enriched the relative abundance of Bacteroidetes, whereas it decreased Proteobacteria, Helicobacteraceae, and Desulfovibrionaceae in feces of HFD dams. The levels of isobutyric acid and pentanoic acid in the HFD + bmOPN group were higher than that of the HFD group. Functional predication analysis of microbial genomes revealed that bmOPN supplementation to HFD pregnancies changed 4 Kyoto Encyclopedia of Genes and Genomes pathways including bile acid biosynthesis. Further, bmOPN enriched hepatic taurochenodeoxycholic acid and tauroursodeoxycholic acid plus taurohyodeoxycholic acid in the gut of HFD maternal rats. Our findings suggested that bmOPN improved the gut health of HFD pregnant rats partially through modulating bile acid biosynthesis.
Collapse
Affiliation(s)
- Lihua Han
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Qiqi Li
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Min Du
- Department of Animal Sciences, Washington State University, Pullman, WA 99163
| | - Xueying Mao
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
41
|
Hao W, Luo Q, Tomic I, Quan W, Hartmann T, Menger MD, Fassbender K, Liu Y. Modulation of Alzheimer's disease brain pathology in mice by gut bacterial depletion: the role of IL-17a. Gut Microbes 2024; 16:2363014. [PMID: 38904096 PMCID: PMC11195493 DOI: 10.1080/19490976.2024.2363014] [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: 12/07/2023] [Accepted: 05/29/2024] [Indexed: 06/22/2024] Open
Abstract
Gut bacteria regulate brain pathology of Alzheimer's disease (AD) patients and animal models; however, the underlying mechanism remains unclear. In this study, 3-month-old APP-transgenic female mice with and without knock-out of Il-17a gene were treated with antibiotics-supplemented or normal drinking water for 2 months. The antibiotic treatment eradicated almost all intestinal bacteria, which led to a reduction in Il-17a-expressing CD4-positive T lymphocytes in the spleen and gut, and to a decrease in bacterial DNA in brain tissue. Depletion of gut bacteria inhibited inflammatory activation in both brain tissue and microglia, lowered cerebral Aβ levels, and promoted transcription of Arc gene in the brain of APP-transgenic mice, all of which effects were abolished by deficiency of Il-17a. As possible mechanisms regulating Aβ pathology, depletion of gut bacteria inhibited β-secretase activity and increased the expression of Abcb1 and Lrp1 in the brain or at the blood-brain barrier, which were also reversed by the absence of Il-17a. Interestingly, a crossbreeding experiment between APP-transgenic mice and Il-17a knockout mice further showed that deficiency of Il-17a had already increased Abcb1 and Lrp1 expression at the blood-brain barrier. Thus, depletion of gut bacteria attenuates inflammatory activation and amyloid pathology in APP-transgenic mice via Il-17a-involved signaling pathways. Our study contributes to a better understanding of the gut-brain axis in AD pathophysiology and highlights the therapeutic potential of Il-17a inhibition or specific depletion of gut bacteria that stimulate the development of Il-17a-expressing T cells.
Collapse
Affiliation(s)
- Wenlin Hao
- Department of Neurology, Saarland University, Homburg/Saar, Germany
- German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
| | - Qinghua Luo
- Department of Neurology, Saarland University, Homburg/Saar, Germany
- German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
- Department of Neurology, The second affiliated hospital of Nanchang University, Nanchang, China
| | - Inge Tomic
- Department of Neurology, Saarland University, Homburg/Saar, Germany
- German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
| | - Wenqiang Quan
- Department of Neurology, Saarland University, Homburg/Saar, Germany
- German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
- Department of Clinical Laboratory, Tongji Hospital, Tongji University Medical School, Shanghai, China
| | - Tobias Hartmann
- German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
- Department of Experimental Neurology, Saarland University, Homburg/Saar, Germany
| | - Michael D. Menger
- Department of Experimental Surgery, Saarland University, Homburg/Saar, Germany
| | - Klaus Fassbender
- Department of Neurology, Saarland University, Homburg/Saar, Germany
- German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
| | - Yang Liu
- Department of Neurology, Saarland University, Homburg/Saar, Germany
- German Institute for Dementia Prevention (DIDP), Saarland University, Homburg/Saar, Germany
| |
Collapse
|
42
|
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
|
43
|
Xiao X, Guo K, Liu J, Liu Y, Yang C, Xu Y, Deng B. The Effect of Sodium Alginate-Coated Nano-Zinc Oxide on the Growth Performance, Serum Indexes and Fecal Microbial Structure of Weaned Piglets. Animals (Basel) 2023; 14:146. [PMID: 38200877 PMCID: PMC10778004 DOI: 10.3390/ani14010146] [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: 11/22/2023] [Revised: 12/27/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
High dose of zinc oxide (ZnO) could improve growth performance and alleviate disease status, whereas it caused serious environmental pollution and bacterial resistance. This study was to investigate whether low doses of sodium alginate-coated nano zinc oxide (saZnO), a new type of zinc resource, could serve as a potential alternative to pharmacological doses of traditional ZnO in weaned piglets. A total of 144 crossbred piglets were randomly allocated into three groups, including a basal diet without the addition of Zn (CON), a basal diet with 1600 mg Zn/kg from traditional ZnO (ZnO), and a basal diet with 500 mg Zn/kg from saZnO (saZnO). The experiment lasted for 28 days. The results showed that supplementing with ZnO and saZnO for 14 and 28 days significantly improved body weight (BW) and average daily gain (ADG) (p < 0.01) and markedly reduced the feed intake-to-gain ratio (F/G) (p < 0.05) and diarrhea rate. In addition, dietary ZnO and saZnO significantly increased the activities of the total antioxidant capacity (T-AOC) and alkaline phosphatase (ALP) (p < 0.01). Supplementing with saZnO also promoted the levels of superoxide dismutase (SOD), IgM and copper- and zinc-containing superoxide dismutase (Cu/Zn-SOD) in serum (p < 0.05), whereas a ZnO addition decreased the concentration of malondialdehyde (MDA) (p < 0.05), indicating the beneficial effect of Zn on antioxidant and immune functions. Piglets fed the ZnO diet showed higher serum Zn accumulations than those fed the CON and saZnO diets at d 28 (p < 0.01), and supplementing with ZnO and saZnO markedly contributed to Zn excretion in feces, especially in the ZnO diet (p < 0.01). Additionally, piglets fed the saZnO diet had greater valeric acid concentrations (p < 0.05) in their feces, while other short chain fatty acids (SCFAs) were not affected by different treatments (p > 0.05). Microbial alpha diversity was reduced in the saZnO group compared with the CON group (p < 0.05), while an obvious separation of microbial composition, the marker of beta diversity, was shown among the three groups (p < 0.05). At the genus level, six genera, including Clostridium_sensu_stricto_1, Terrisporobacter, f_Muribaculaceae, Subdoligranulum and Intestinibacter, were pronouncedly increased in the ZnO and saZnO groups (p < 0.05); another nine species were dramatically downregulated, such as f_Lachnospiraceae, f_Prevotellaceae, f_Butyricicoccaceae and f_Ruminococcaceae (p < 0.05). Finally, a functional analysis indicated that altered microbes significantly changed the "Metabolism" pathway (p < 0.05). These findings suggested that saZnO could act as a feasible substitute for ZnO to reduce Zn emission and enhance growth performance, antioxidant and immune functions, and to adjust the structure of gut microbiota in piglets.
Collapse
Affiliation(s)
- Xiao Xiao
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China; (X.X.); (K.G.); (Y.X.)
| | - Kai Guo
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China; (X.X.); (K.G.); (Y.X.)
| | - Jinsong Liu
- Zhejiang Vegamax Biotechnology Co., Ltd., Anji, Huzhou 313300, China; (J.L.); (Y.L.); (C.Y.)
| | - Yulan Liu
- Zhejiang Vegamax Biotechnology Co., Ltd., Anji, Huzhou 313300, China; (J.L.); (Y.L.); (C.Y.)
| | - Caimei Yang
- Zhejiang Vegamax Biotechnology Co., Ltd., Anji, Huzhou 313300, China; (J.L.); (Y.L.); (C.Y.)
| | - Yinglei Xu
- Key Laboratory of Applied Technology on Green-Eco-Healthy Animal Husbandry of Zhejiang Province, Zhejiang Provincial Engineering Laboratory for Animal Health and Internet Technology, College of Animal Science and Technology, College of Veterinary Medicine, Zhejiang Agricultural and Forestry University, Hangzhou 311300, China; (X.X.); (K.G.); (Y.X.)
| | - Bo Deng
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| |
Collapse
|
44
|
Lu Q, Liang Y, Tian S, Jin J, Zhao Y, Fan H. Radiation-Induced Intestinal Injury: Injury Mechanism and Potential Treatment Strategies. TOXICS 2023; 11:1011. [PMID: 38133412 PMCID: PMC10747544 DOI: 10.3390/toxics11121011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023]
Abstract
Radiation-induced intestinal injury (RIII) is one of the most common intestinal complications caused by radiotherapy for pelvic and abdominal tumors and it seriously affects the quality of life of patients. However, the treatment of acute RIII is essentially symptomatic and nutritional support treatment and an ideal means of prevention and treatment is lacking. Researchers have conducted studies at the cellular and animal levels and found that some chemical or biological agents have good therapeutic effects on RIII and may be used as potential candidates for clinical treatment. This article reviews the injury mechanism and potential treatment strategies based on cellular and animal experiments to provide new ideas for the diagnosis and treatment of RIII in clinical settings.
Collapse
Affiliation(s)
- Qianying Lu
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yangfan Liang
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Sijia Tian
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Jie Jin
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Yanmei Zhao
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| | - Haojun Fan
- Institute of Disaster and Emergency Medicine, Tianjin University, Tianjin 300072, China; (Q.L.); (Y.L.); (S.T.); (J.J.)
- Tianjin Key Laboratory of Disaster Medicine Technology, Tianjin 300072, China
| |
Collapse
|
45
|
Chen Z, Guan D, Wang Z, Li X, Dong S, Huang J, Zhou W. Microbiota in cancer: molecular mechanisms and therapeutic interventions. MedComm (Beijing) 2023; 4:e417. [PMID: 37937304 PMCID: PMC10626288 DOI: 10.1002/mco2.417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 10/04/2023] [Accepted: 10/12/2023] [Indexed: 11/09/2023] Open
Abstract
The diverse bacterial populations within the symbiotic microbiota play a pivotal role in both health and disease. Microbiota modulates critical aspects of tumor biology including cell proliferation, invasion, and metastasis. This regulation occurs through mechanisms like enhancing genomic damage, hindering gene repair, activating aberrant cell signaling pathways, influencing tumor cell metabolism, promoting revascularization, and remodeling the tumor immune microenvironment. These microbiota-mediated effects significantly impact overall survival and the recurrence of tumors after surgery by affecting the efficacy of chemoradiotherapy. Moreover, leveraging the microbiota for the development of biovectors, probiotics, prebiotics, and synbiotics, in addition to utilizing antibiotics, dietary adjustments, defensins, oncolytic virotherapy, and fecal microbiota transplantation, offers promising alternatives for cancer treatment. Nonetheless, due to the extensive and diverse nature of the microbiota, along with tumor heterogeneity, the molecular mechanisms underlying the role of microbiota in cancer remain a subject of intense debate. In this context, we refocus on various cancers, delving into the molecular signaling pathways associated with the microbiota and its derivatives, the reshaping of the tumor microenvironmental matrix, and the impact on tolerance to tumor treatments such as chemotherapy and radiotherapy. This exploration aims to shed light on novel perspectives and potential applications in the field.
Collapse
Affiliation(s)
- Zhou Chen
- The First Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The First Hospital of Lanzhou UniversityLanzhouGansuChina
| | - Defeng Guan
- The First Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The First Hospital of Lanzhou UniversityLanzhouGansuChina
| | - Zhengfeng Wang
- The First Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The First Hospital of Lanzhou UniversityLanzhouGansuChina
| | - Xin Li
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The Department of General SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Shi Dong
- The Second Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The Department of General SurgeryLanzhou University Second HospitalLanzhouGansuChina
| | - Junjun Huang
- The First Hospital of Lanzhou UniversityLanzhouGansuChina
| | - Wence Zhou
- The First Clinical Medical CollegeLanzhou UniversityLanzhouGansuChina
- The Department of General SurgeryLanzhou University Second HospitalLanzhouGansuChina
| |
Collapse
|
46
|
Yuan T, Zhang J, Zhao Y, Guo Y, Fan S. Single-cell RNA sequencing of intestinal crypts reveals vital events in damage repair and the double-edged sword effect of the Wnt3/β-catenin pathway in irradiated mice. Redox Biol 2023; 68:102942. [PMID: 37918127 PMCID: PMC10638071 DOI: 10.1016/j.redox.2023.102942] [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: 09/11/2023] [Revised: 10/13/2023] [Accepted: 10/19/2023] [Indexed: 11/04/2023] Open
Abstract
In this study, we executed single-cell RNA sequencing of intestinal crypts. We analyzed the differentially expressed genes (DEGs) at different time points (the first, third, and fifth days) after 13 Gy and 15 Gy abdominal body radiation (ABR) exposure and then executed gene ontology (GO) enrichment analysis, RNA velocity analysis, cell communication analysis, and ligand‒receptor interaction analysis to explore the vital events in damage repair and the multiple effects of the Wnt3/β-catenin pathway on irradiated mice. Results from bioinformatics analysis were confirmed by a series of biological experiments. Results showed that the antibacterial response is a vital event during the damage response process after 13 Gy ABR exposure; ionizing radiation (IR) induced high heterogeneity in the transient amplification (TA) cluster, which may differentiate into mature cells and stem cells in irradiated small intestine (SI) crypts. Conducting an enrichment analysis of the DEGs between mice exposed to 13 Gy and 15 Gy ABR, we concluded that the Wnt3/β-catenin and MIF-CD74/CD44 signaling pathways may contribute to 15 Gy ABR-induced mouse death. Wnt3/β-catenin promotes the recovery of irradiated SI stem/progenitor cells, which may trigger macrophage migration inhibitory factor (MIF) release to further repair IR-induced SI injury; however, with the increase in radiation dose, activation of CD44 on macrophages provides the receptor for MIF signal transduction, initiating the inflammatory cascade response and ultimately causing a cytokine release syndrome. In contrast to previous research, we confirmed that inhibition of the Wnt3/β-catenin pathway or blockade of CD44 on the second day after 15 Gy ABR may significantly protect against ABR-induced death. This study indicates that the Wnt3/β-catenin pathway plays multiple roles in damage repair after IR exposure; we also propose a novel point that the interaction between intestinal crypt stem cells (ISCs) and macrophages through the MIF-CD74/CD44 axis may exacerbate SI damage in irradiated mice.
Collapse
Affiliation(s)
- Tong Yuan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, 300192, People's Republic of China
| | - Junling Zhang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, 300192, People's Republic of China.
| | - Yue Zhao
- Annoroad Gene Technology (Beijing) Co. Ltd, Beijing, 100176, People's Republic of China
| | - Yuying Guo
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, 300192, People's Republic of China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Peking Union Medical College and Chinese Academy of Medical Science, Tianjin, 300192, People's Republic of China.
| |
Collapse
|
47
|
Van Dingenen L, Segers C, Wouters S, Mysara M, Leys N, Kumar-Singh S, Malhotra-Kumar S, Van Houdt R. Dissecting the role of the gut microbiome and fecal microbiota transplantation in radio- and immunotherapy treatment of colorectal cancer. Front Cell Infect Microbiol 2023; 13:1298264. [PMID: 38035338 PMCID: PMC10687483 DOI: 10.3389/fcimb.2023.1298264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 10/31/2023] [Indexed: 12/02/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most commonly diagnosed cancers and poses a major burden on the human health worldwide. At the moment, treatment of CRC consists of surgery in combination with (neo)adjuvant chemotherapy and/or radiotherapy. More recently, immune checkpoint blockers (ICBs) have also been approved for CRC treatment. In addition, recent studies have shown that radiotherapy and ICBs act synergistically, with radiotherapy stimulating the immune system that is activated by ICBs. However, both treatments are also associated with severe toxicity and efficacy issues, which can lead to temporary or permanent discontinuation of these treatment programs. There's growing evidence pointing to the gut microbiome playing a role in these issues. Some microorganisms seem to contribute to radiotherapy-associated toxicity and hinder ICB efficacy, while others seem to reduce radiotherapy-associated toxicity or enhance ICB efficacy. Consequently, fecal microbiota transplantation (FMT) has been applied to reduce radio- and immunotherapy-related toxicity and enhance their efficacies. Here, we have reviewed the currently available preclinical and clinical data in CRC treatment, with a focus on how the gut microbiome influences radio- and immunotherapy toxicity and efficacy and if these treatments could benefit from FMT.
Collapse
Affiliation(s)
- Lena Van Dingenen
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Charlotte Segers
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Shari Wouters
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology and Histology, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Mohamed Mysara
- Bioinformatics Group, Center for Informatics Science, School of Information Technology and Computer Science, Nile University, Giza, Egypt
| | - Natalie Leys
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| | - Samir Kumar-Singh
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
- Molecular Pathology Group, Laboratory of Cell Biology and Histology, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Surbhi Malhotra-Kumar
- Laboratory of Medical Microbiology, Vaccine and Infectious Disease Institute, Faculty of Medicine, University of Antwerp, Antwerp, Belgium
| | - Rob Van Houdt
- Nuclear Medical Applications, Belgian Nuclear Research Centre, SCK CEN, Mol, Belgium
| |
Collapse
|
48
|
Cao B, Gao J, Zhang Q, Xu X, Zhao R, Li H, Wei B. Melatonin supplementation protects against traumatic colon injury by regulating SERPINA3N protein expression. IMETA 2023; 2:e141. [PMID: 38868216 PMCID: PMC10989984 DOI: 10.1002/imt2.141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 09/25/2023] [Indexed: 06/14/2024]
Abstract
Traumatic colon injury (TCI) is a typical injury with high mortality. Prolongation of the intervention time window is a potentially useful approach to improving the outcomes of TCI casualties. This study aimed to identify the pathological mechanisms of TCI and to develop effective strategies to extend the survival time. A semicircular incision was made to prepare a TCI model using C57BL/6 mice. An overview of microbiota dysregulation was achieved by metagenome sequencing. Protein expression reprogramming in the intestinal epithelium was investigated using proteomics profiling. The mice that were subjected to TCI died within a short period of time when not treated. Gut symbiosis showed abrupt turbulence, and specific pathogenic bacteria rapidly proliferated. The protein expression in the intestinal epithelium was also reprogrammed. Among the differentially expressed proteins, SERPINA3N was overexpressed after TCI modeling. Deletion of Serpina3n prolonged the posttraumatic survival time of mice with TCI by improving gut homeostasis in vivo. To promote the translational application of this research, the effects of melatonin (MLT), an oral inhibitor of the SERPINA3N protein, were further investigated. MLT effectively downregulated SERPINA3N expression and mitigated TCI-induced death by suppressing the NF-κB signaling pathway. Our findings prove that preventive administration of MLT serves as an effective regimen to prolong the posttraumatic survival time by restoring gut homeostasis perturbed by TCI. It may become a novel strategy for improving the prognosis of patients suffering from TCI.
Collapse
Affiliation(s)
- Bo Cao
- Department of General Surgery, First Medical CenterChinese PLA General HospitalBeijingChina
| | - Jing‐Wang Gao
- Department of General Surgery, First Medical CenterChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Qing‐Peng Zhang
- Department of General Surgery, First Medical CenterChinese PLA General HospitalBeijingChina
| | - Xing‐Ming Xu
- Department of General Surgery, First Medical CenterChinese PLA General HospitalBeijingChina
| | - Rui‐Yang Zhao
- Department of General Surgery, First Medical CenterChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Hang‐Hang Li
- Department of General Surgery, First Medical CenterChinese PLA General HospitalBeijingChina
- Medical School of Chinese PLABeijingChina
| | - Bo Wei
- Department of General Surgery, First Medical CenterChinese PLA General HospitalBeijingChina
| |
Collapse
|
49
|
Lin X, Xiao HM, Liu HM, Lv WQ, Greenbaum J, Gong R, Zhang Q, Chen YC, Peng C, Xu XJ, Pan DY, Chen Z, Li ZF, Zhou R, Wang XF, Lu JM, Ao ZX, Song YQ, Zhang YH, Su KJ, Meng XH, Ge CL, Lv FY, Luo Z, Shi XM, Zhao Q, Guo BY, Yi NJ, Shen H, Papasian CJ, Shen J, Deng HW. Gut microbiota impacts bone via Bacteroides vulgatus-valeric acid-related pathways. Nat Commun 2023; 14:6853. [PMID: 37891329 PMCID: PMC10611739 DOI: 10.1038/s41467-023-42005-y] [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: 03/26/2020] [Accepted: 09/11/2023] [Indexed: 10/29/2023] Open
Abstract
Although the gut microbiota has been reported to influence osteoporosis risk, the individual species involved, and underlying mechanisms, remain largely unknown. We performed integrative analyses in a Chinese cohort of peri-/post-menopausal women with metagenomics/targeted metabolomics/whole-genome sequencing to identify novel microbiome-related biomarkers for bone health. Bacteroides vulgatus was found to be negatively associated with bone mineral density (BMD), which was validated in US white people. Serum valeric acid (VA), a microbiota derived metabolite, was positively associated with BMD and causally downregulated by B. vulgatus. Ovariectomized mice fed B. vulgatus demonstrated increased bone resorption and poorer bone micro-structure, while those fed VA demonstrated reduced bone resorption and better bone micro-structure. VA suppressed RELA protein production (pro-inflammatory), and enhanced IL10 mRNA expression (anti-inflammatory), leading to suppressed maturation of osteoclast-like cells and enhanced maturation of osteoblasts in vitro. The findings suggest that B. vulgatus and VA may represent promising targets for osteoporosis prevention/treatment.
Collapse
Affiliation(s)
- Xu Lin
- Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), No.1 of Jiazi Road, Lunjiao, Shunde District, Foshan City, 528308, Guangdong Province, China
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Hong-Mei Xiao
- Center of System Biology, Data Information and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, 410008, Hunan Province, China.
| | - Hui-Min Liu
- Center of System Biology, Data Information and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, 410008, Hunan Province, China
| | - Wan-Qiang Lv
- Center of System Biology, Data Information and Reproductive Health, School of Basic Medical Science, Central South University, Changsha, 410008, Hunan Province, China
| | - Jonathan Greenbaum
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Rui Gong
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Qiang Zhang
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Yuan-Cheng Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Cheng Peng
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Xue-Juan Xu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Dao-Yan Pan
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Zhi Chen
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Zhang-Fang Li
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Rou Zhou
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Xia-Fang Wang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Jun-Min Lu
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Zeng-Xin Ao
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Yu-Qian Song
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Yin-Hua Zhang
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China
| | - Kuan-Jui Su
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Xiang-He Meng
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Chang-Li Ge
- LC-Bio Technologies (Hangzhou) CO., LTD., Hangzhou, 310018, Zhejiang Province, China
| | - Feng-Ye Lv
- LC-Bio Technologies (Hangzhou) CO., LTD., Hangzhou, 310018, Zhejiang Province, China
| | - Zhe Luo
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Xing-Ming Shi
- Departments of Neuroscience & Regenerative Medicine and Orthopaedic Surgery, Medical College of Georgia, Augusta University, Augusta, GA, 30914, USA
| | - Qi Zhao
- Department of Preventive Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Bo-Yi Guo
- Department of Biostatistics, University of Alabama at Birmingham, Alabama, 35294, USA
| | - Neng-Jun Yi
- Department of Biostatistics, University of Alabama at Birmingham, Alabama, 35294, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA
| | - Christopher J Papasian
- Department of Biomedical Sciences, School of Medicine, University of Missouri-Kansas City, 2411 Holmes Street, Kansas City, MO, 64108, USA
| | - Jie Shen
- Shunde Hospital of Southern Medical University (The First People's Hospital of Shunde), No.1 of Jiazi Road, Lunjiao, Shunde District, Foshan City, 528308, Guangdong Province, China.
- Department of Endocrinology and Metabolism, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510630, Guangdong Province, China.
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, 70112, USA.
| |
Collapse
|
50
|
Cui W, Hull L, Zizzo A, Wang L, Lin B, Zhai M, Xiao M. The gut microbiome changes in wild type and IL-18 knockout mice after 9.0 Gy total body irradiation. Anim Microbiome 2023; 5:42. [PMID: 37679818 PMCID: PMC10485964 DOI: 10.1186/s42523-023-00262-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 08/24/2023] [Indexed: 09/09/2023] Open
Abstract
BACKGROUND Recent studies have shown that gut microbiome plays important roles in response to radiation exposure. IL-18, an inflammatory cytokine, is highly elevated in mice, mini-pigs and nonhuman primates after radiation exposure. Blocking IL-18 using its endogenous binding protein (IL-18BP) increases mice survival after radiation exposure by decreasing bone marrow interferon-gamma levels. METHODS To further characterize the roles of IL-18 in response to radiation, both wild type and IL-18 knockout (IL-18 KO) mice were exposed to 9.0 Gy total body irradiation (TBI). The 30-day survival result demonstrated that IL-18 KO mice were significantly more resistant to radiation compared to the wild type mice (p < 0.0001). Mouse faecal samples were collected at pre-radiation (d0), d1, d3, d7, d14, d21 and d29 after radiation exposure. Microbiome profiling was performed on the faecal samples using 16S and ITS sequencing technology. RESULTS Data analysis showed that there was significant difference in the bacterial microbiome between wild type and IL-18 KO mice. Cohousing of wild type and IL-18 KO mice decreased the bacterial microbiome difference between the two genotypes. Much fewer bacterial genera were significantly changed in wild type mice than the IL-18 KO mice after radiation exposure. The different composition of the IL-18 KO mice and wild type mice persisted even after radiation exposure. Bacterial genera that significantly correlated with other genera were identified in the IL-18 KO and wild type mice. The metabolic pathways that differentially expressed in both genotypes were identified. The animal bacterial microbiome data could be used to predict the animal's radiation status. The fungal microbiome had no significant difference regarding genotype or time after radiation exposure. CONCLUSION The current study helps understand the gut microbiome in different genetic backgrounds and its temporal changes after radiation exposure. Our data provide insight into the mechanisms underlying radiation-induced toxicity and help identify bacteria important in response to radiation.
Collapse
Affiliation(s)
- Wanchang Cui
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA.
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, 20817, USA.
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA.
| | - Lisa Hull
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, 20817, USA
| | - Alex Zizzo
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA
| | - Li Wang
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, 20817, USA
- Department of Pathology, Uniformed Services University of the Health Sciences, Bethesda, MD, 20814, USA
| | - Bin Lin
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, 20817, USA
| | - Min Zhai
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA
- The Henry M. Jackson Foundation for the Advancement of Military Medicine, Inc., Bethesda, MD, 20817, USA
| | - Mang Xiao
- Armed Forces Radiobiology Research Institute, Uniformed Services University of the Health Sciences, 4555 South Palmer Road, Bethesda, MD, 20889-5648, USA.
| |
Collapse
|