1
|
Siang W, Li Jin J, Yinming J, Wenji L, Yan F. Effects of Dietary Fiber and Acetate on Alcoholic Heart Disease and Intestinal Microbes in Mice. Mol Nutr Food Res 2025:e70040. [PMID: 40249113 DOI: 10.1002/mnfr.70040] [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: 12/17/2024] [Revised: 02/16/2025] [Accepted: 03/10/2025] [Indexed: 04/19/2025]
Abstract
Alcoholic heart disease (AHD) is a severe cardiovascular condition linked to chronic alcohol consumption. This study investigates the effects of a high-fiber diet and acetate on gut microbiota and cardiac function in AHD mouse models. Sixty male C57BL/6 mice were divided into six groups, receiving either a control diet, high-fiber diet, or acetate supplementation alongside alcohol treatment. Results revealed that cardiac fibrosis and heart failure were notably improved in the AHD mice receiving high-fiber or acetate diets. Transcriptomic analyses indicated that dietary interventions modulated the expression of genes involved in lipid metabolism and the TGF-β signaling pathway. Additionally, 16S rRNA sequencing showed that the high-fiber diet and acetate altered gut microbiota composition, enhancing the abundance of beneficial bacteria such as Akkermansia muciniphila, Lactobacillus intestinalis, and Bacteroides acidifaciens. These microbes exhibited positive correlations with genes related to fat metabolism and TGF-β signaling, suggesting a potential mechanism for gut microbiota's role in AHD pathology. ROC analysis identified these bacteria as promising biomarkers for AHD detection. Overall, our findings underscore the therapeutic potential of dietary fiber and acetate in modulating gut microbiota and improving cardiac function in AHD, highlighting the intricate relationship between gut health and cardiovascular disease management.
Collapse
Affiliation(s)
- Wei Siang
- College of Life Science, Shanxi Agricultural University, Jinzhong, People's Republic of China
- Beijing Medi Intelligent Pharmaceutical Technology Co., Ltd, Beijing, People's Republic of China
- Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, People's Republic of China
| | - Jiang Li Jin
- College of Life Science, Shanxi Agricultural University, Jinzhong, People's Republic of China
| | - Jiao Yinming
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, People's Republic of China
| | - Lin Wenji
- Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, People's Republic of China
| | - Feng Yan
- College of Life Science, Shanxi Agricultural University, Jinzhong, People's Republic of China
| |
Collapse
|
2
|
Liu QL, Zhou H, Wang Z, Chen Y. Exploring the role of gut microbiota in colorectal liver metastasis through the gut-liver axis. Front Cell Dev Biol 2025; 13:1563184. [PMID: 40181829 PMCID: PMC11965903 DOI: 10.3389/fcell.2025.1563184] [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: 01/19/2025] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Colorectal liver metastasis (CRLM) represents a major therapeutic challenge in colorectal cancer (CRC), with complex interactions between the gut microbiota and the liver tumor microenvironment (TME) playing a crucial role in disease progression via the gut-liver axis. The gut barrier serves as a gatekeeper, regulating microbial translocation, which influences liver colonization and metastasis. Through the gut-liver axis, the microbiota actively shapes the TME, where specific microbial species and their metabolites exert dual roles in immune modulation. The immunologically "cold" nature of the liver, combined with the influence of the gut microbiota on liver immunity, complicates effective immunotherapy. However, microbiota-targeted interventions present promising strategies to enhance immunotherapy outcomes by modulating the gut-liver axis. Overall, this review highlights the emerging evidence on the role of the gut microbiota in CRLM and provides insights into the molecular mechanisms driving the dynamic interactions within the gut-liver axis.
Collapse
Affiliation(s)
- Qiu-Luo Liu
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
- Institute of Digestive Surgery, Institute of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Huijie Zhou
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Health Management Center, General Practice Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ziqiang Wang
- Colorectal Cancer Center, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yan Chen
- Department of Gastrointestinal Surgery, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
3
|
Wu S, Bu X, Chen D, Wu X, Wu H, Caiyin Q, Qiao J. Molecules-mediated bidirectional interactions between microbes and human cells. NPJ Biofilms Microbiomes 2025; 11:38. [PMID: 40038292 PMCID: PMC11880406 DOI: 10.1038/s41522-025-00657-2] [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: 10/06/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Complex molecules-mediated interactions, which are based on the bidirectional information exchange between microbes and human cells, enable the defense against diseases and health maintenance. Recently, diverse single-direction interactions based on active metabolites, immunity factors, and quorum sensing signals have largely been summarized separately. In this review, according to a simplified timeline, we proposed the framework of Molecules-mediated Bidirectional Interactions (MBI) between microbe and humans to decipher and understand their intricate interactions systematically. About the microbe-derived interactions, we summarized various molecules, such as short-chain fatty acids, bile acids, tryptophan catabolites, and quorum sensing molecules, and their corresponding human receptors. Concerning the human-derived interactions, we reviewed the effect of human molecules, including hormones, cytokines, and other circulatory metabolites on microbial characteristics and phenotypes. Finally, we discussed the challenges and trends for developing and deciphering molecule-mediated bidirectional interactions and their potential applications in the guard of human health.
Collapse
Affiliation(s)
- Shengbo Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China
| | - Xueying Bu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Danlei Chen
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China
| | - Xueyan Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China
| | - Hao Wu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
| | - Qinggele Caiyin
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
- Key Laboratory of Systems Bioengineering, Ministry of Education (Tianjin University), Tianjin, 300072, China.
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300072, China.
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, 300350, China.
- Zhejiang Institute of Tianjin University, Shaoxing, Shaoxing, 312300, Zhejiang, China.
- Key Laboratory of Systems Bioengineering, Ministry of Education (Tianjin University), Tianjin, 300072, China.
- Frontiers Science Center for Synthetic Biology (Ministry of Education), Tianjin University, Tianjin, 300072, China.
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, 300072, China.
| |
Collapse
|
4
|
Nai S, Song J, Su W, Liu X. Bidirectional Interplay Among Non-Coding RNAs, the Microbiome, and the Host During Development and Diseases. Genes (Basel) 2025; 16:208. [PMID: 40004537 PMCID: PMC11855195 DOI: 10.3390/genes16020208] [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: 12/30/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
It is widely known that the dysregulation of non-coding RNAs (ncRNAs) and dysbiosis of the gut microbiome play significant roles in host development and the progression of various diseases. Emerging evidence has highlighted the bidirectional interplay between ncRNAs and the gut microbiome. This article aims to review the current understanding of the molecular mechanisms underlying the crosstalk between ncRNAs, especially microRNA (miRNA), and the gut microbiome in the context of development and diseases, such as colorectal cancer, inflammatory bowel diseases, neurological disorders, obesity, and cardiovascular disease. Ultimately, this review seeks to provide a foundation for exploring the potential roles of ncRNAs and gut microbiome interactions as biomarkers and therapeutic targets for clinical diagnosis and treatment, such as ncRNA mimics, antisense oligonucleotides, and small-molecule compounds, as well as probiotics, prebiotics, and diets.
Collapse
Affiliation(s)
| | | | | | - Xiaoqian Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (S.N.); (J.S.); (W.S.)
| |
Collapse
|
5
|
Wang L, Xi M, Cao W, Qin H, Qin D, Chen S, Zhou S, Hou Y, Chen Y, Xiao X, Zheng Q, Li D, Li Y. Electroacupuncture alleviates functional constipation by upregulating host-derived miR-205-5p to modulate gut microbiota and tryptophan metabolism. Front Microbiol 2025; 16:1517018. [PMID: 39973939 PMCID: PMC11835812 DOI: 10.3389/fmicb.2025.1517018] [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: 10/28/2024] [Accepted: 01/07/2025] [Indexed: 02/21/2025] Open
Abstract
Electroacupuncture (EA) has shown promise as a treatment for Functional constipation (FC), with growing evidence suggesting it may enhance gut motility. MicroRNAs (miRNAs) serve as key regulatory molecules mediating host-microbiota interactions. However, the specific fecal miRNAs regulating microbiota composition and metabolism in EA-treated constipated mice, along with their key targets, remain unidentified. We examined fecal microbiome composition, metabolism, and colonic miRNA expression in loperamide-induced constipated mice and EA-treated mice to identify differentially expressed miRNAs and assess their relationships with microbial abundance, metabolism, and gut motility. An antibiotic cocktail and adeno-associated virus were employed to interfere with the gut microbiota and target miRNA in vivo, thereby validating the proposed mechanism. Our results indicate that miR-205-5p, significantly upregulated in fecal and colonic tissues of EA-treated constipated mice, promotes intestinal motility in a microbiome-dependent manner. Specifically, EA promoted the growth of Lactobacillus reuteri, enriched in the feces of constipation-recovered mice, through host-derived miR-205-5p regulation. Furthermore, Lactobacillus reuteri and its tryptophan metabolites (indole-3-acetamide, indole-3-acetic acid, and indole-3-carboxaldehyde) alleviated loperamide-induced constipation. These findings underscore the pivotal role of host-derived miR-205-5p in modulating microbial composition and tryptophan metabolites to enhance intestinal motility through EA.
Collapse
Affiliation(s)
- Lu Wang
- Department of Acupuncture, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Menghan Xi
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Department of Acupuncture and Moxibustion, Chengdu Pidu District Hospital of TCM/The Third Clinical Medical College of Chengdu University of TCM, Chengdu, Sichuan, China
| | - Wei Cao
- Center of Preventive Medicine, Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, Sichuan, China
| | - Haiyan Qin
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Di Qin
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Shuai Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Siyuan Zhou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yujun Hou
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ying Chen
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xianjun Xiao
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qianhua Zheng
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Dehua Li
- Department of Acupuncture, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ying Li
- Acupuncture and Tuina School, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
6
|
Shi Y, Chen Z, Fang T, Chen X, Deng Y, Qin H, Lian M, Shen J, Zong Y, Chu H, Hoebinger C, Guo H, Yuan Z, Zheng J, Zhou Y, Pan Y, Mendes BG, Lang S, Hendrikx T, Zeng S, Cao H, Yang L, Chen L, Chen P, Dai L, Wang H, Yin S, Zhu S, Ma X, Schnabl B, Chen H, Duan Y. Gut microbiota in treating inflammatory digestive diseases: Current challenges and therapeutic opportunities. IMETA 2025; 4:e265. [PMID: 40027479 PMCID: PMC11865350 DOI: 10.1002/imt2.265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 12/14/2024] [Accepted: 12/14/2024] [Indexed: 03/05/2025]
Abstract
Accumulating evidence indicates that the gut microbiota is intricately involved in the initiation and progression of human diseases, forming a multidirectional regulatory axis centered on intestinal microbiota. This article illustrates the challenges in exploring the role of the gut microbiota in inflammatory digestive diseases, such as metabolic dysfunction-associated steatotic liver disease (MASLD) and inflammatory bowel disease (IBD), and summarizes the existing microbiome-focused treatment strategies (probiotics, prebiotics, symbiotics, fecal microbiota transplantation, and bacteriophages therapy), emerging technologies (gut microbiome-on-a-chip and artificial intelligence), as well as possible future research directions. Taken together, these therapeutic strategies and technologies present both opportunities and challenges, which require researchers and clinicians to test the rationality and feasibility of various therapeutic modalities in continuous practice.
Collapse
Affiliation(s)
- Yongpeng Shi
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Zeran Chen
- Department of Nutrition and Food Hygiene, School of Public HealthCapital Medical UniversityBeijingChina
| | - Tingyu Fang
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Xingyao Chen
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Youpeng Deng
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Hao Qin
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience, National Center for Nanoscience and TechnologyBeijingChina
| | - Min Lian
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of MedicineShanghai Jiao Tong University, Shanghai Institute of Digestive DiseaseShanghaiChina
| | - Juntao Shen
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Yuru Zong
- Department of Nutrition and Food Hygiene, School of Public HealthCapital Medical UniversityBeijingChina
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | | | - Hao Guo
- State Key Laboratory of Cellular Stress Biology, Xiang'an Hospital, School of Life Sciences, Faculty of Medicine and Life SciencesXiamen UniversityXiamenChina
| | - Zhongshang Yuan
- Department of Biostatistics, School of Public Health, Cheeloo College of Medicine, Institute for Medical DataologyShandong UniversityJinanChina
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- Shanghai National Clinical Research Center for metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the Peoples R China, Shanghai Key Laboratory for Endocrine Tumour, Shanghai Digital Medicine Innovation Center, Ruijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical SchoolUniversity of BristolBristolUnited Kingdom
| | - Yongjian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of MedicineSouth China University of TechnologyGuangzhouChina
| | - Yue Pan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat‐Sen Memorial HospitalSun Yat‐Sen UniversityGuangzhouChina
- Nanchang Research InstituteSun Yat‐Sen UniversityNanchangChina
| | - Beatriz G. Mendes
- Department of Clinical AnalysisFederal University of Santa CatarinaFlorianópolisBrazil
| | - Sonja Lang
- Department of Gastroenterology and Hepatology, Faculty of Medicine and University Hospital CologneUniversity of CologneCologneGermany
| | - Tim Hendrikx
- Department of Laboratory MedicineMedical University ViennaViennaAustria
| | - Suling Zeng
- Institute of Health and Medicine, Hefei Comprehensive National Science CenterHefeiChina
| | - Hailong Cao
- Department of Gastroenterology and Hepatology, General Hospital, Tianjin Medical UniversityTianjin Institute of Digestive Diseases, Tianjin Key Laboratory of Digestive DiseasesTianjinChina
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Lianmin Chen
- Department of CardiologyThe First Affiliated Hospital of Nanjing Medical University, Nanjing Medical UniversityNanjingChina
| | - Peng Chen
- Department of Pathophysiology, Guangdong Provincial Key Laboratory of Proteomics, School of Basic Medical SciencesSouthern Medical UniversityGuangzhouChina
- Microbiome Medicine Center, Zhujiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Lei Dai
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of SciencesShenzhenChina
| | - Hua Wang
- Department of OncologyThe First Affiliated Hospital of Anhui Medical UniversityHefeiChina
- Inflammation and Immune Mediated Diseases Laboratory of Anhui ProvinceAnhui Medical UniversityHefeiChina
| | - Shi Yin
- Department of Geriatrics, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTCUniversity of Science and Technology of ChinaHefeiChina
- Anhui Key Laboratory of Geriatric Immunology and Nutrition TherapyHefeiChina
| | - Shu Zhu
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| | - Xiong Ma
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, State Key Laboratory for Oncogenes and Related Genes, Renji Hospital, School of MedicineShanghai Jiao Tong University, Shanghai Institute of Digestive DiseaseShanghaiChina
| | - Bernd Schnabl
- Department of MedicineUniversity of California San DiegoLa JollaCaliforniaUSA
| | - Hanqing Chen
- Department of Nutrition and Food Hygiene, School of Public HealthCapital Medical UniversityBeijingChina
| | - Yi Duan
- Department of Infectious Diseases, The First Affiliated Hospital of USTC, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
- Key Laboratory of Immune Response and Immunotherapy, Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefeiChina
| |
Collapse
|
7
|
Guo Y, He J, Li S, Zou S, Zhang H, Yang X, Wang J. Warm and humid environment induces gut microbiota dysbiosis and bacterial translocation leading to inflammatory state and promotes proliferation and biofilm formation of certain bacteria, potentially causing sticky stool. BMC Microbiol 2025; 25:24. [PMID: 39819481 PMCID: PMC11737230 DOI: 10.1186/s12866-024-03730-6] [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: 10/16/2024] [Accepted: 12/23/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND AND AIMS OF THE STUDY Fluctuations in environmental temperature and humidity significantly affect human physiology and disease manifestation. In the Lingnan region of China, high summer temperatures and humidity often cause symptoms like diminished appetite, sticky tongue coating, sticky stool, unsatisfactory defecation, lethargy, and joint heaviness. These are referred to as "Dampness Syndrome" in Traditional Chinese Medicine (TCM). Thick and greasy tongue fur and sticky feces are characteristic symptoms of "Dampness Syndrome" and serve as crucial diagnostic indicators in TCM for assessing health conditions. However, the specific mechanisms that lead to these symptoms, such as sticky feces and thick and greasy tongue fur, have not been fully elucidated. Understanding these external symptoms is essential, as they reflect internal health status. Warm, humid environments favor microorganism growth, potentially disrupting gut microbiota and bacterial translocation, which could induce an immune-inflammatory response. The primary objective of this study is to explore the potential significant role of immune response products in influencing the proliferation and biofilm formation of gut microbiota, which may subsequently lead to changes in fecal characteristics. METHODS In this study, mice were exposed to a controlled warm and humid environment (25 ± 3 °C with 95% humidity) for 16 days to simulate conditions associated with "Dampness Syndrome." After this period, Huoxiang Zhengqi Water, a traditional remedy, has been administrated for four days. On the one hand saliva and tongue coating samples were also taken from human subjects with "Dampness Syndrome" for microorganism culturing and to assess biofilm formation, on the other hand the co-culture products of a macrophage cell line RAW264.7 and Candida albicans and the effect of tumor necrosis factor-α (TNF-α) were evaluated for their impact on the proliferation and biofilm-forming abilities of different bacterial strains. RESULTS Compared to a control group, the treatment group exhibited significant changes in gut microbiota, including increased biofilm formation, which was mitigated by Huoxiang Zhengqi Water. In the model group, fungal translocation was observed, potentially triggering an inflammatory response. Intraperitoneal injections of various bacterial strains in mice reproduced the sticky stool characteristics. Both mice and human subjects with "Dampness Syndrome" displayed elevated serum levels of inflammatory cytokines TNF-α and interleukin-17 A (IL-17A). Interestingly, Saliva samples from individuals with "Dampness Syndrome" showed elevated TNF-α levels, accompanied by thick and greasy tongue fur. Culturing samples from the tongue coating of individuals in the "Dampness Syndrome" group revealed an increased biofilm formation capability. C. albicans co-cultured with RAW264.7 cells increased TNF-α secretion, and the supernatant promoted pathogenic bacterial proliferation and biofilm formation. TNF-α specifically enhanced biofilm formation in microorganism like C. albicans and Staphylococcus aureus, with minimal effect on beneficial bacteria like Lacticaseibacillus paracasei and Lactiplantibacillus plantarum in the tested conditions. CONCLUSIONS These findings provided new insights into the biological mechanisms of 'Dampness Syndrome' and support the therapeutic role of Huoxiang Zhengqi Water in treating symptoms associated with microbial dysbiosis and inflammation. Additionally, they indicate that TNF-α seems to have selective effects in promoting the proliferation and biofilm formation of different microbial species.
Collapse
Affiliation(s)
- Yinrui Guo
- Guangdong Engineering Research Center of Early Clinical Trials of Biotechnology Drugs, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Jianlang He
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Shaojie Li
- Zhuhai Hospital of Integrated Traditional Chinese and Western Medicine, Zhuhai, Guangdong, China
| | - Shiqi Zou
- Guangdong Engineering Research Center of Early Clinical Trials of Biotechnology Drugs, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Haiting Zhang
- The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Xin Yang
- Guangdong Engineering Research Center of Early Clinical Trials of Biotechnology Drugs, The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, Guangdong, China.
| | - Jian Wang
- School of Basic Medical Science, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China.
- Shenzhen Hospital (Futian) of Guangzhou University of Chinese Medicine, Shenzhen, Guangdong, China.
| |
Collapse
|
8
|
Li JH, Xu J, Huang C, Hu JX, Xu HM, Guo X, Zhang Y, Xu JK, Peng Y, Zhang Y, Zhu MZ, Zhou YL, Nie YQ. Houttuynia cordata-Derived Exosome-Like Nanoparticles Mitigate Colitis in Mice via Inhibition of the NLRP3 Signaling Pathway and Modulation of the Gut Microbiota. Int J Nanomedicine 2024; 19:13991-14018. [PMID: 39742094 PMCID: PMC11687308 DOI: 10.2147/ijn.s493434] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/19/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND Plant-derived exosome-like nanoparticles (PELNs) have received widespread attention in treating ulcerative colitis (UC). However, the role of Houttuynia cordata-derived exosome-like nanoparticles (HELNs) in UC remains unclear. This study aims to evaluate the efficacy of HELNs in treating colitis in mice and investigate its potential mechanisms. METHODS HELNs were isolated from H. cordata for characterization, and their safety and stability were evaluated. A dextran sulfate sodium (DSS)-induced colitis mouse model was utilized to assess the therapeutic potential of HELNs in UC. In vivo, imaging and flow cytometry were utilized to investigate the targeting effect of HELNs on inflamed colonic sites and their modulation of the immune environment. RNA-seq analysis and molecular docking were performed to identify potential pathways recruited by HELNs. Guided by transcriptomic findings, NLRP3-/- mice were used in conjunction with Western blotting, qPCR, immunofluorescence, and other techniques to verify that HELNs alleviated DSS-induced colitis by inhibiting NLRP3/NOD-like receptor signaling pathways. Lastly, the impact of HELNs on the gut microbiota was investigated through 16S rRNA sequencing. RESULTS HELNs significantly reduced the severity of DSS-induced colitis in mice, alleviating colitis symptoms and histopathological damage. Furthermore, HELNs can specifically target inflamed colon tissue, regulate the immune environment, and decrease inflammation. RNA-seq analysis, coupled with the use of NLRP3-/- mice, demonstrated that HELNs inhibited the NLRP3/NOD-like receptor signaling pathways. Lastly, HELNs balanced the gut microbiota composition in mice with colitis, decreasing the abundance of harmful bacteria and increasing the abundance of beneficial bacteria in the intestinal tract of these mice. CONCLUSION In summary, HELNs exhibit the potential to protect the colon from DSS-induced damage by inhibiting the NLRP3/NOD-like receptor signaling pathway and modulating the gut microbiota, presenting a promising therapeutic option for the management of UC.
Collapse
Affiliation(s)
- Jian-Hong Li
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Jing Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Chen Huang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Jin-Xia Hu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Hao-Ming Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Xue Guo
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yan Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Jing-Kui Xu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yao Peng
- Department of Gastroenterology and Hepatology, Shenzhen General Hospital, Shenzhen, People’s Republic of China
| | - Yong Zhang
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Min-Zheng Zhu
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - You-Lian Zhou
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| | - Yu-Qiang Nie
- Department of Gastroenterology and Hepatology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
- Department of Gastroenterology and Hepatology, Guangzhou First People’s Hospital, South China University of Technology, Guangzhou, Guangdong, People’s Republic of China
| |
Collapse
|
9
|
Pepke ML, Hansen SB, Limborg MT. Unraveling host regulation of gut microbiota through the epigenome-microbiome axis. Trends Microbiol 2024; 32:1229-1240. [PMID: 38839511 DOI: 10.1016/j.tim.2024.05.006] [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: 01/26/2024] [Revised: 05/13/2024] [Accepted: 05/15/2024] [Indexed: 06/07/2024]
Abstract
Recent studies of dynamic interactions between epigenetic modifications of a host organism and the composition or activity of its associated gut microbiota suggest an opportunity for the host to shape its microbiome through epigenetic alterations that lead to changes in gene expression and noncoding RNA activity. We use insights from microbiota-induced epigenetic changes to review the potential of the host to epigenetically regulate its gut microbiome, from which a bidirectional 'epigenome-microbiome axis' emerges. This axis embeds environmentally induced variation, which may influence the adaptive evolution of host-microbe interactions. We furthermore present our perspective on how the epigenome-microbiome axis can be understood and investigated within a holo-omic framework with potential applications in the applied health and food sciences.
Collapse
Affiliation(s)
- Michael L Pepke
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark.
| | - Søren B Hansen
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark
| | - Morten T Limborg
- Center for Evolutionary Hologenomics, Globe Institute, Faculty of Health and Medical Sciences, University of Copenhagen, Øster Farimagsgade 5, DK-1353 Copenhagen, Denmark.
| |
Collapse
|
10
|
Zhang C, Liu H, Jiang X, Zhang Z, Hou X, Wang Y, Wang D, Li Z, Cao Y, Wu S, Huws SA, Yao J. An integrated microbiome- and metabolome-genome-wide association study reveals the role of heritable ruminal microbial carbohydrate metabolism in lactation performance in Holstein dairy cows. MICROBIOME 2024; 12:232. [PMID: 39529146 PMCID: PMC11555892 DOI: 10.1186/s40168-024-01937-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Despite the growing number of studies investigating the connection between host genetics and the rumen microbiota, there remains a dearth of systematic research exploring the composition, function, and metabolic traits of highly heritable rumen microbiota influenced by host genetics. Furthermore, the impact of these highly heritable subsets on lactation performance in cows remains unknown. To address this gap, we collected and analyzed whole-genome resequencing data, rumen metagenomes, rumen metabolomes and short-chain fatty acids (SCFAs) content, and lactation performance phenotypes from a cohort of 304 dairy cows. RESULTS The results indicated that the proportions of highly heritable subsets (h2 ≥ 0.2) of the rumen microbial composition (55%), function (39% KEGG and 28% CAZy), and metabolites (18%) decreased sequentially. Moreover, the highly heritable microbes can increase energy-corrected milk (ECM) production by reducing the rumen acetate/propionate ratio, according to the structural equation model (SEM) analysis (CFI = 0.898). Furthermore, the highly heritable enzymes involved in the SCFA synthesis metabolic pathway can promote the synthesis of propionate and inhibit the acetate synthesis. Next, the same significant SNP variants were used to integrate information from genome-wide association studies (GWASs), microbiome-GWASs, metabolome-GWASs, and microbiome-wide association studies (mWASs). The identified single nucleotide polymorphisms (SNPs) of rs43470227 and rs43472732 on SLC30A9 (Zn2+ transport) (P < 0.05/nSNPs) can affect the abundance of rumen microbes such as Prevotella_sp., Prevotella_sp._E15-22, Prevotella_sp._E13-27, which have the oligosaccharide-degradation enzymes genes, including the GH10, GH13, GH43, GH95, and GH115 families. The identified SNPs of chr25:11,177 on 5s_rRNA (small ribosomal RNA) (P < 0.05/nSNPs) were linked to ECM, the abundance alteration of Pseudobutyrivibrio_sp. (a genus that was also showed to be linked to the ECM production via the mWASs analysis), GH24 (lysozyme), and 9,10,13-TriHOME (linoleic acid metabolism). Moreover, ECM, and the abundances of Pseudobutyrivibrio sp., GH24, and 9,10,13-TRIHOME were significantly greater in the GG genotype than in the AG genotype at chr25:11,177 (P < 0.05). By further the SEM analysis, GH24 was positively correlated with Pseudobutyrivibrio sp., which was positively correlated with 9,10,13-triHOME and subsequently positively correlated with ECM (CFI = 0.942). CONCLUSION Our comprehensive study revealed the distinct heritability patterns of rumen microbial composition, function, and metabolism. Additionally, we shed light on the influence of host SNP variants on the rumen microbes with carbohydrate metabolism and their subsequent effects on lactation performance. Collectively, these findings offer compelling evidence for the host-microbe interactions, wherein cows actively modulate their rumen microbiota through SNP variants to regulate their own lactation performance. Video Abstract.
Collapse
Affiliation(s)
- Chenguang Zhang
- College of Animal Science and Technology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
| | - Huifeng Liu
- College of Animal Science and Technology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
| | - Xingwei Jiang
- College of Animal Science and Technology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
| | - Zhihong Zhang
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
- JUNLEBAO-Northwest A&F University Cooperation Dairy Research Institute, Leyuan Animal Husbandry, JUNLEBAO Company, Shijiazhuang, Hebei, China
| | - Xinfeng Hou
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
- JUNLEBAO-Northwest A&F University Cooperation Dairy Research Institute, Leyuan Animal Husbandry, JUNLEBAO Company, Shijiazhuang, Hebei, China
| | - Yue Wang
- College of Animal Science and Technology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
| | - Dangdang Wang
- College of Animal Science and Technology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
| | - Zongjun Li
- College of Animal Science and Technology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
| | - Yangchun Cao
- College of Animal Science and Technology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China
| | - Shengru Wu
- College of Animal Science and Technology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China.
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China.
| | - Sharon A Huws
- Institute of Global Food Security, School of Biological Sciences, Queen's University Belfast, 19 Chlorine Gardens, Belfast, Northern Ireland, BT9 5DL, UK.
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China.
- Key Laboratory of Livestock Biology, Northwest A&F University, 22 Nt, Xinong Road, Yangling, Shaanxi, China.
| |
Collapse
|
11
|
Wang W, Wei Z, Li Z, Ren J, Song Y, Xu J, Liu A, Li X, Li M, Fan H, Jin L, Niyazbekova Z, Wang W, Gao Y, Jiang Y, Yao J, Li F, Wu S, Wang Y. Integrating genome- and transcriptome-wide association studies to uncover the host-microbiome interactions in bovine rumen methanogenesis. IMETA 2024; 3:e234. [PMID: 39429883 PMCID: PMC11487568 DOI: 10.1002/imt2.234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 10/22/2024]
Abstract
The ruminal microbiota generates biogenic methane in ruminants. However, the role of host genetics in modifying ruminal microbiota-mediated methane emissions remains mysterious, which has severely hindered the emission control of this notorious greenhouse gas. Here, we uncover the host genetic basis of rumen microorganisms by genome- and transcriptome-wide association studies with matched genome, rumen transcriptome, and microbiome data from a cohort of 574 Holstein cattle. Heritability estimation revealed that approximately 70% of microbial taxa had significant heritability, but only 43 genetic variants with significant association with 22 microbial taxa were identified through a genome-wide association study (GWAS). In contrast, the transcriptome-wide association study (TWAS) of rumen microbiota detected 28,260 significant gene-microbe associations, involving 210 taxa and 4652 unique genes. On average, host genetic factors explained approximately 28% of the microbial abundance variance, while rumen gene expression explained 43%. In addition, we highlighted that TWAS exhibits a strong advantage in detecting gene expression and phenotypic trait associations in direct effector organs. For methanogenic archaea, only one significant signal was detected by GWAS, whereas the TWAS obtained 1703 significant associated host genes. By combining multiple correlation analyses based on these host TWAS genes, rumen microbiota, and volatile fatty acids, we observed that substrate hydrogen metabolism is an essential factor linking host-microbe interactions in methanogenesis. Overall, these findings provide valuable guidelines for mitigating methane emissions through genetic regulation and microbial management strategies in ruminants.
Collapse
Affiliation(s)
- Wei Wang
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Zhenyu Wei
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Zhuohui Li
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Jianrong Ren
- Department of Animal Nutrition and Environmental HealthCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Yanliang Song
- Department of Clinical VeterinaryCollege of Veterinary MedicineNorthwest A&F UniversityYanglingChina
| | - Jingyi Xu
- Department of Animal Nutrition and Environmental HealthCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Anguo Liu
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Xinmei Li
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Manman Li
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Huimei Fan
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Liangliang Jin
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Zhannur Niyazbekova
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Wen Wang
- School of Ecology and EnvironmentFaculty of Life Sciences and MedicineNorthwestern Polytechnical UniversityXi'anChina
| | - Yuanpeng Gao
- Department of Clinical VeterinaryCollege of Veterinary MedicineNorthwest A&F UniversityYanglingChina
- Key Laboratory of Livestock BiologyNorthwest A&F UniversityYanglingChina
| | - Yu Jiang
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Key Laboratory of Livestock BiologyNorthwest A&F UniversityYanglingChina
| | - Junhu Yao
- Department of Animal Nutrition and Environmental HealthCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Key Laboratory of Livestock BiologyNorthwest A&F UniversityYanglingChina
| | - Fuyong Li
- Department of Animal Science and TechnologyCollege of Animal SciencesZhejiang UniversityHangzhouChina
| | - Shengru Wu
- Department of Animal Nutrition and Environmental HealthCollege of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Key Laboratory of Livestock BiologyNorthwest A&F UniversityYanglingChina
| | - Yu Wang
- Department of Animal GeneticsBreeding and Reproduction, College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Key Laboratory of Livestock BiologyNorthwest A&F UniversityYanglingChina
| |
Collapse
|
12
|
Zheng D, Zhang H, Zheng X, Zhao A, Jia W. Novel microbial modifications of bile acids and their functional implications. IMETA 2024; 3:e243. [PMID: 39429880 PMCID: PMC11487544 DOI: 10.1002/imt2.243] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/19/2024] [Accepted: 09/20/2024] [Indexed: 10/22/2024]
Abstract
This review outlines the recent discoveries of bile acids that have undergone novel microbial modifications, highlighting their biological roles and the profound implications for the development of innovative therapeutic strategies. The review aims to provide valuable insights and breakthroughs for future drug candidates in the expanding field of bile acid therapeutics.
Collapse
Affiliation(s)
- Dan Zheng
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Huiheng Zhang
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xiaojiao Zheng
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Aihua Zhao
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wei Jia
- Center for Translational Medicine and Shanghai Key Laboratory of Diabetes MellitusShanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of MedicineShanghaiChina
- Department of Pharmacology and PharmacyThe University of Hong KongHong KongChina
| |
Collapse
|
13
|
Wei T, Lu C, Du H, Yang Q, Qi X, Liu Y, Zhang Y, Chen C, Li Y, Tang Y, Zhang WH, Tao X, Jiang N. DeepPBI-KG: a deep learning method for the prediction of phage-bacteria interactions based on key genes. Brief Bioinform 2024; 25:bbae484. [PMID: 39344712 PMCID: PMC11440089 DOI: 10.1093/bib/bbae484] [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: 05/30/2024] [Revised: 08/18/2024] [Accepted: 09/13/2024] [Indexed: 10/01/2024] Open
Abstract
Phages, the natural predators of bacteria, were discovered more than 100 years ago. However, increasing antimicrobial resistance rates have revitalized phage research. Methods that are more time-consuming and efficient than wet-laboratory experiments are needed to help screen phages quickly for therapeutic use. Traditional computational methods usually ignore the fact that phage-bacteria interactions are achieved by key genes and proteins. Methods for intraspecific prediction are rare since almost all existing methods consider only interactions at the species and genus levels. Moreover, most strains in existing databases contain only partial genome information because whole-genome information for species is difficult to obtain. Here, we propose a new approach for interaction prediction by constructing new features from key genes and proteins via the application of K-means sampling to select high-quality negative samples for prediction. Finally, we develop DeepPBI-KG, a corresponding prediction tool based on feature selection and a deep neural network. The results show that the average area under the curve for prediction reached 0.93 for each strain, and the overall AUC and area under the precision-recall curve reached 0.89 and 0.92, respectively, on the independent test set; these values are greater than those of other existing prediction tools. The forward and reverse validation results indicate that key genes and key proteins regulate and influence the interaction, which supports the reliability of the model. In addition, intraspecific prediction experiments based on Klebsiella pneumoniae data demonstrate the potential applicability of DeepPBI-KG for intraspecific prediction. In summary, the feature engineering and interaction prediction approaches proposed in this study can effectively improve the robustness and stability of interaction prediction, can achieve high generalizability, and may provide new directions and insights for rapid phage screening for therapy.
Collapse
Affiliation(s)
- Tongqing Wei
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Chenqi Lu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Hanxiao Du
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Qianru Yang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Xin Qi
- Shanghai Sci-Tech Inno Center for Infection & Immunity, No. 1688 Guoquan Bei Road, Shanghai, China
| | - Yankun Liu
- Shanghai Sci-Tech Inno Center for Infection & Immunity, No. 1688 Guoquan Bei Road, Shanghai, China
| | - Yi Zhang
- Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan Univerisy, No. 12 Wulumuqi Zhong Road, Shanghai, China
| | - Chen Chen
- Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan Univerisy, No. 12 Wulumuqi Zhong Road, Shanghai, China
| | - Yutong Li
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Yuanhao Tang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
| | - Wen-Hong Zhang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, No. 1688 Guoquan Bei Road, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan Univerisy, No. 12 Wulumuqi Zhong Road, Shanghai, China
| | - Xu Tao
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, No. 1688 Guoquan Bei Road, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan Univerisy, No. 12 Wulumuqi Zhong Road, Shanghai, China
| | - Ning Jiang
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, No. 2005 Songhu Road, Shanghai, 200433, China
- Shanghai Sci-Tech Inno Center for Infection & Immunity, No. 1688 Guoquan Bei Road, Shanghai, China
- Department of Infectious Diseases, Huashan Hospital, Shanghai Medical College, Fudan Univerisy, No. 12 Wulumuqi Zhong Road, Shanghai, China
| |
Collapse
|
14
|
Ma Z, Zuo T, Frey N, Rangrez AY. A systematic framework for understanding the microbiome in human health and disease: from basic principles to clinical translation. Signal Transduct Target Ther 2024; 9:237. [PMID: 39307902 PMCID: PMC11418828 DOI: 10.1038/s41392-024-01946-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024] Open
Abstract
The human microbiome is a complex and dynamic system that plays important roles in human health and disease. However, there remain limitations and theoretical gaps in our current understanding of the intricate relationship between microbes and humans. In this narrative review, we integrate the knowledge and insights from various fields, including anatomy, physiology, immunology, histology, genetics, and evolution, to propose a systematic framework. It introduces key concepts such as the 'innate and adaptive genomes', which enhance genetic and evolutionary comprehension of the human genome. The 'germ-free syndrome' challenges the traditional 'microbes as pathogens' view, advocating for the necessity of microbes for health. The 'slave tissue' concept underscores the symbiotic intricacies between human tissues and their microbial counterparts, highlighting the dynamic health implications of microbial interactions. 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and prudent antibiotic use. The 'homeostatic reprogramming hypothesis' integrates the microbiome into the internal environment theory, potentially explaining the change in homeostatic indicators post-industrialization. The 'cell-microbe co-ecology model' elucidates the symbiotic regulation affecting cellular balance, while the 'meta-host model' broadens the host definition to include symbiotic microbes. The 'health-illness conversion model' encapsulates the innate and adaptive genomes' interplay and dysbiosis patterns. The aim here is to provide a more focused and coherent understanding of microbiome and highlight future research avenues that could lead to a more effective and efficient healthcare system.
Collapse
Affiliation(s)
- Ziqi Ma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|
15
|
Chen JL, Liu L, Peng XR, Wang Y, Xiang X, Chen Y, Xu DX, Chen DZ. Role of the GalNAc-galectin pathway in the healing of premature rupture of membranes. Mol Med 2024; 30:138. [PMID: 39232672 PMCID: PMC11375961 DOI: 10.1186/s10020-024-00908-6] [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: 03/05/2023] [Accepted: 08/23/2024] [Indexed: 09/06/2024] Open
Abstract
BACKGROUND Premature rupture of the membranes (PROM) is a key cause of preterm birth and represents a major cause of neonatal mortality and morbidity. Natural products N-acetyl-d-galactosamine (GalNAc), which are basic building blocks of important polysaccharides in biological cells or tissues, such as chitin, glycoproteins, and glycolipids, may improve possible effects of wound healing. METHODS An in vitro inflammation and oxidative stress model was constructed using tumor necrosis-α (TNF-α) and lipopolysaccharide (LPS) action on WISH cells. Human amniotic epithelial cells (hAECs) were primarily cultured by digestion to construct a wound model. The effects of GalNAc on anti-inflammatory and anti-oxidative stress, migration and proliferation, epithelial-mesenchymal transition (EMT), glycosaminoglycan (GAG)/hyaluronic acid (HA) production, and protein kinase B (Akt) pathway in hAECs and WISH cells were analyzed using the DCFH-DA fluorescent probe, ELISA, CCK-8, scratch, transwell migration, and western blot to determine the mechanism by which GalNAc promotes amniotic wound healing. RESULTS GalNAc decreased IL-6 expression in TNF-α-stimulated WISH cells and ROS expression in LPS-stimulated WISH cells (P < 0.05). GalNAc promoted the expression of Gal-1 and Gal-3 with anti-inflammatory and anti-oxidative stress effects. GalNAc promoted the migration of hAECs (50% vs. 80%) and WISH cells through the Akt signaling pathway, EMT reached the point of promoting fetal membrane healing, and GalNAc did not affect the activity of hAECs and WISH cells (P > 0.05). GalNAc upregulated the expression of sGAG in WISH cells (P < 0.05) but did not affect HA levels (P > 0.05). CONCLUSIONS GalNAc might be a potential target for the prevention and treatment of PROM through the galectin pathway, including (i) inflammation; (ii) epithelial-mesenchymal transition; (iii) proliferation and migration; and (iv) regression, remodeling, and healing.
Collapse
Affiliation(s)
- Jia-Le Chen
- The School of Public Health, Anhui Medical University, Hefei, China
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China
- Hospital Infection Management Section, Changzhou Wujin Hospital of Traditional Chinese Medicine, Changzhou, China
| | - Lou Liu
- Department of obstetrics, Longgang District Maternity & Child Healthcare Hospital of Shenzhen City, Shenzhen, China
| | - Xin-Rui Peng
- The School of Public Health, Anhui Medical University, Hefei, China
| | - Yan Wang
- The School of Public Health, Anhui Medical University, Hefei, China
| | - Xiang Xiang
- The School of Public Health, Anhui Medical University, Hefei, China
| | - Yu Chen
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China.
| | - De-Xiang Xu
- The School of Public Health, Anhui Medical University, Hefei, China.
| | - Dao-Zhen Chen
- The School of Public Health, Anhui Medical University, Hefei, China.
- Wuxi Maternity and Child Health Care Hospital, Wuxi, China.
- Department of Laboratory, Haidong No.2 People's Hospital, Haidong, China.
| |
Collapse
|
16
|
Hu M, Du Y, Li W, Zong X, Du W, Sun H, Liu H, Zhao K, Li J, Farooq MZ, Wu J, Xu Q. Interplay of Food-Derived Bioactive Peptides with Gut Microbiota: Implications for Health and Disease Management. Mol Nutr Food Res 2024; 68:e2400251. [PMID: 39097954 DOI: 10.1002/mnfr.202400251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 06/19/2024] [Indexed: 08/06/2024]
Abstract
Bioactive peptides (BPs) are protein fragments with beneficial effects on metabolism, physiology, and diseases. This review focuses on proteolytic BPs, which are produced by the action of gut microbiota on proteins in food and have demonstrated to influence the composition of gut microbes. And gut microbiota are candidate targets of BPs to alleviate oxidative stress, enhance immunity, and control diseases, including diabetes, hypertension, obesity, cancer, and immune and neurodegenerative diseases. Despite promising results, further research is needed to understand the mechanisms underlying the interactions between BPs and gut microbes, and to identify and screen more BPs for industrial applications. Overall, BPs offer potential as therapeutic agents for various diseases through their interactions with gut microbes, highlighting the importance of continued research in this area.
Collapse
Affiliation(s)
- Mingyang Hu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yufeng Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenyue Li
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xiaomei Zong
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Wenjuan Du
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Huizeng Sun
- Institute of Dairy Science, MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Hongyun Liu
- Institute of Dairy Science, MoE Key Laboratory of Molecular Animal Nutrition, College of Animal Sciences, Zhejiang University, Hangzhou, 310058, China
| | - Ke Zhao
- Institute of Food Science, Zhejiang Academy of Agricultural Sciences, Hangzhou, 310058, China
| | - Jianxiong Li
- Wuhan Jason Biotech Co., Ltd., Wuhan, 430070, China
| | - Muhammad Zahid Farooq
- Department of Animal Science, University of Veterinary and Animal Science, Lahore, 54000, Pakistan
| | - Jianping Wu
- Department of Agricultural, Food and Nutritional Science, University of Alberta, 4-10 Ag/For Building, Edmonton, Alberta, T6G 2P5, Canada
| | - Qingbiao Xu
- College of Animal Sciences and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| |
Collapse
|
17
|
Xue M, Xie Y, Zang X, Zhong Y, Ma X, Sun H, Liu J. Deciphering functional groups of rumen microbiome and their underlying potentially causal relationships in shaping host traits. IMETA 2024; 3:e225. [PMID: 39135684 PMCID: PMC11316931 DOI: 10.1002/imt2.225] [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: 05/31/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 08/15/2024]
Abstract
Over the years, microbiome research has achieved tremendous advancements driven by culture-independent meta-omics approaches. Despite extensive research, our understanding of the functional roles and causal effects of the microbiome on phenotypes remains limited. In this study, we focused on the rumen metaproteome, combining it with metatranscriptome and metabolome data to accurately identify the active functional distributions of rumen microorganisms and specific functional groups that influence feed efficiency. By integrating host genetics data, we established the potentially causal relationships between microbes-proteins/metabolites-phenotype, and identified specific patterns in which functional groups of rumen microorganisms influence host feed efficiency. We found a causal link between Selenomonas bovis and rumen carbohydrate metabolism, potentially mediated by bacterial chemotaxis and a two-component regulatory system, impacting feed utilization efficiency of dairy cows. Our study on the nutrient utilization functional groups in the rumen of high-feed-efficiency dairy cows, along with the identification of key microbiota functional proteins and their potentially causal relationships, will help move from correlation to causation in rumen microbiome research. This will ultimately enable precise regulation of the rumen microbiota for optimized ruminant production.
Collapse
Affiliation(s)
- Ming‐Yuan Xue
- Institute of Dairy Science, College of Animal SciencesZhejiang UniversityHangzhouChina
- Xianghu LaboratoryHangzhouChina
| | - Yun‐Yi Xie
- Institute of Dairy Science, College of Animal SciencesZhejiang UniversityHangzhouChina
| | - Xin‐Wei Zang
- Institute of Dairy Science, College of Animal SciencesZhejiang UniversityHangzhouChina
| | - Yi‐Fan Zhong
- Institute of Dairy Science, College of Animal SciencesZhejiang UniversityHangzhouChina
| | - Xiao‐Jiao Ma
- Institute of Dairy Science, College of Animal SciencesZhejiang UniversityHangzhouChina
| | - Hui‐Zeng Sun
- Institute of Dairy Science, College of Animal SciencesZhejiang UniversityHangzhouChina
- Ministry of Education Key Laboratory of Molecular Animal NutritionZhejiang UniversityHangzhouChina
| | - Jian‐Xin Liu
- Institute of Dairy Science, College of Animal SciencesZhejiang UniversityHangzhouChina
- Ministry of Education Key Laboratory of Molecular Animal NutritionZhejiang UniversityHangzhouChina
| |
Collapse
|
18
|
Wang M, Zhang L, Jiang X, Song Y, Wang D, Liu H, Wu S, Yao J. Multiomics analysis revealed that the metabolite profile of raw milk is associated with lactation stage of dairy cows and could be affected by variations in the ruminal microbiota. J Dairy Sci 2024:S0022-0302(24)00919-6. [PMID: 38876221 DOI: 10.3168/jds.2024-24753] [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: 02/02/2024] [Accepted: 05/12/2024] [Indexed: 06/16/2024]
Abstract
The nutritional components and quality of milk are influenced by the rumen microbiota and its metabolites at different lactation stages. Hence, rumen fluid and milk samples from 6 dairy cows fed the same diet were collected during peak, early mid- and later mid-lactation. Untargeted metabolomics and 16S rRNA sequencing were applied for analyzing milk and rumen metabolites, as well as rumen microbial composition, respectively. The levels of lipid-related metabolites, L-glutamate, glucose-1-phosphate and acetylphosphate in milk exhibited lactation-dependent attenuation. Maltol, N-acetyl-D-glucosamine, and choline, which are associated with milk flavor or coagulation properties, as well as L-valine, lansioside-A, clitocine and ginsenoside-La increased significantly in early mid- and later mid-lactation, especially in later mid-lactation. The obvious increase in rumen microbial diversities (Ace and Shannon indices) were observed in early mid-lactation compared with peak lactation. Twenty-one differential bacterial genera of the rumen were identified, with Succinivibrionaceae_UCG-001, Candidatus Saccharimonas, Fibrobacter, and SP3-e08 being significantly enriched in peak lactation. Rikenellaceae_RC9_gut_group, Eubacterium_ruminantium_group, Lachnospira, Butyrivibrio, Eubacterium_hallii_group, and Schwartzia were most significantly enriched in early mid-lactation. In comparison, only 2 bacteria (unclassified_f__Prevotellaceae and Prevotellaceae_UCG-001) were enriched in later mid-lactation. For rumen metabolites, LPE(16:0), L-glutamate and L-tyrosine had higher levels in peak lactation, whereas PE(17:0/0:0), PE(16:0/0:0), PS(18:1(9Z)/0:0), L-phenylalanine, dulcitol, 2-(methoxymethyl)furan and 3-phenylpropyl acetate showed higher levels in early mid- and later mid-lactation. Multiomics integrated analysis revealed that a greater abundance of Fibrobacter contributed to phospholipid content in milk by increasing ruminal acetate, L-glutamate and LysoPE(16:0). Prevotellaceae_UCG-001 and unclassified_f_Prevotellaceae provide substrates for milk metabolites of the same category by increasing ruminal L-phenylalanine and dulcitol contents. These results demonstrated that milk metabolomic fingerprints and critical functional metabolites during lactation, and the key bacteria in rumen related to them. These findings provide new insights into the development of functional dairy products.
Collapse
Affiliation(s)
- Mengya Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China; Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Lei Zhang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China; Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Xingwei Jiang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China; Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Yuxuan Song
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China; Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Dangdang Wang
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China; Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Huifeng Liu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China; Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, China
| | - Shengru Wu
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China; Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| | - Junhu Yao
- College of Animal Science and Technology, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China; Key Laboratory of Livestock Biology, Northwest A&F University, Yangling 712100, Shaanxi, China.
| |
Collapse
|
19
|
Wang L, Villafuerte Gálvez JA, Lee C, Wu S, Kelly CP, Chen X, Cao Y. Understanding host immune responses in Clostridioides difficile infection: Implications for pathogenesis and immunotherapy. IMETA 2024; 3:e200. [PMID: 38898983 PMCID: PMC11183162 DOI: 10.1002/imt2.200] [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: 04/02/2024] [Revised: 04/23/2024] [Accepted: 04/23/2024] [Indexed: 06/21/2024]
Abstract
Clostridioides difficile (C. difficile) is the predominant causative agent of nosocomial diarrhea worldwide. Infection with C. difficile occurs due to the secretion of large glycosylating toxin proteins, which can lead to toxic megacolon or mortality in susceptible hosts. A critical aspect of C. difficile's biology is its ability to persist asymptomatically within the human host. Individuals harboring asymptomatic colonization or experiencing a single episode of C. difficile infection (CDI) without recurrence exhibit heightened immune responses compared to symptomatic counterparts. The significance of these immune responses cannot be overstated, as they play critical roles in the development, progression, prognosis, and outcomes of CDI. Nonetheless, our current comprehension of the immune responses implicated in CDI remains limited. Therefore, further investigation is imperative to elucidate their underlying mechanisms. This review explores recent advancements in comprehending CDI pathogenesis and how the host immune system response influences disease progression and severity, aiming to enhance our capacity to develop immunotherapy-based treatments for CDI.
Collapse
Affiliation(s)
- Lamei Wang
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Javier A. Villafuerte Gálvez
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Christina Lee
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Shengru Wu
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
| | - Ciaran P. Kelly
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Xinhua Chen
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| | - Yangchun Cao
- College of Animal Science and TechnologyNorthwest A&F UniversityYanglingChina
- Division of Gastroenterology, Department of Medicine, Beth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusettsUSA
| |
Collapse
|
20
|
Tian Z, Zhang X, Yao G, Jin J, Zhang T, Sun C, Wang Z, Zhang Q. Intestinal flora and pregnancy complications: Current insights and future prospects. IMETA 2024; 3:e167. [PMID: 38882493 PMCID: PMC11170975 DOI: 10.1002/imt2.167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/27/2023] [Accepted: 12/22/2023] [Indexed: 06/18/2024]
Abstract
Numerous studies have demonstrated the pivotal roles of intestinal microbiota in many physiopathological processes through complex interactions with the host. As a unique period in a woman's lifespan, pregnancy is characterized by changes in hormones, immunity, and metabolism. The gut microbiota also changes during this period and plays a crucial role in maintaining a healthy pregnancy. Consequently, anomalies in the composition and function of the gut microbiota, namely, gut microbiota dysbiosis, can predispose individuals to various pregnancy complications, posing substantial risks to both maternal and neonatal health. However, there are still many controversies in this field, such as "sterile womb" versus "in utero colonization." Therefore, a thorough understanding of the roles and mechanisms of gut microbiota in pregnancy and its complications is essential to safeguard the health of both mother and child. This review provides a comprehensive overview of the changes in gut microbiota during pregnancy, its abnormalities in common pregnancy complications, and potential etiological implications. It also explores the potential of gut microbiota in diagnosing and treating pregnancy complications and examines the possibility of gut-derived bacteria residing in the uterus/placenta. Our aim is to expand knowledge in maternal and infant health from the gut microbiota perspective, aiding in developing new preventive and therapeutic strategies for pregnancy complications based on intestinal microecology.
Collapse
Affiliation(s)
- Zhenyu Tian
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology Qilu Hospital of Shandong University Jinan China
| | - Xinjie Zhang
- Department of Biology University College London London UK
| | - Guixiang Yao
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology Qilu Hospital of Shandong University Jinan China
| | - Jiajia Jin
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology Qilu Hospital of Shandong University Jinan China
| | - Tongxue Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology Qilu Hospital of Shandong University Jinan China
| | - Chunhua Sun
- Department of Health Management Center, Qilu Hospital, Cheeloo College of Medicine Shandong University Jinan China
| | - Zhe Wang
- Department of Geriatrics Shandong Provincial Hospital Affiliated to Shandong First Medical University Jinan China
| | - Qunye Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences; Department of Cardiology Qilu Hospital of Shandong University Jinan China
- Cardiovascular Disease Research Center of Shandong First Medical University Central Hospital Affiliated to Shandong First Medical University Jinan China
| |
Collapse
|
21
|
Ma L, Tao S, Song T, Lyu W, Li Y, Wang W, Shen Q, Ni Y, Zhu J, Zhao J, Yang H, Xiao Y. Clostridium butyricum and carbohydrate active enzymes contribute to the reduced fat deposition in pigs. IMETA 2024; 3:e160. [PMID: 38868506 PMCID: PMC10989082 DOI: 10.1002/imt2.160] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 10/06/2023] [Indexed: 06/14/2024]
Abstract
Pig gastrointestinal tracts harbor a heterogeneous and dynamic ecosystem populated with trillions of microbes, enhancing the ability of the host to harvest energy from dietary carbohydrates and contributing to host adipogenesis and fatness. However, the microbial community structure and related mechanisms responsible for the differences between the fatty phenotypes and the lean phenotypes of the pigs remained to be comprehensively elucidated. Herein, we first found significant differences in microbial composition and potential functional capacity among different gut locations in Jinhua pigs with distinct fatness phenotypes. Second, we identified that Jinhua pigs with lower fatness exhibited higher levels of short-chain fatty acids in the colon, highlighting their enhanced carbohydrate fermentation capacity. Third, we explored the differences in expressed carbohydrate-active enzyme (CAZyme) in pigs, indicating their involvement in modulating fat storage. Notably, Clostridium butyricum might be a representative bacterial species from Jinhua pigs with lower fatness, and a significantly higher percentage of its genome was dedicated to CAZyme glycoside hydrolase family 13 (GH13). Finally, a subsequent mouse intervention study substantiated the beneficial effects of C. butyricum isolated from experimental pigs, suggesting that it may possess characteristics that promote the utilization of carbohydrates and hinder fat accumulation. Remarkably, when Jinhua pigs were administered C. butyricum, similar alterations in the gut microbiome and host fatness traits were observed, further supporting the potential role of C. butyricum in modulating fatness. Taken together, our findings reveal previously overlooked links between C. butyricum and CAZyme function, providing insight into the basic mechanisms that connect gut microbiome functions to host fatness.
Collapse
Affiliation(s)
- Lingyan Ma
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐Products, Institute of Agro‐product Safety and NutritionZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Shiyu Tao
- Department of Animal Nutrition and Feed Science, College of Animal Sciences and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Tongxing Song
- Department of Animal Nutrition and Feed Science, College of Animal Sciences and TechnologyHuazhong Agricultural UniversityWuhanChina
| | - Wentao Lyu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐Products, Institute of Agro‐product Safety and NutritionZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Ying Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, College of Life Science and EngineeringFoshan UniversityFoshanChina
| | - Wen Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐Products, Institute of Agro‐product Safety and NutritionZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Qicheng Shen
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐Products, Institute of Agro‐product Safety and NutritionZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Yan Ni
- The Children's Hospital, Zhejiang University School of MedicineNational Clinical Research Center for Child HealthHangzhouChina
| | - Jiang Zhu
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐Products, Institute of Agro‐product Safety and NutritionZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Jiangchao Zhao
- Department of Animal Science, Division of AgricultureUniversity of ArkansasFayettevilleArkansasUSA
| | - Hua Yang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐Products, Institute of Agro‐product Safety and NutritionZhejiang Academy of Agricultural SciencesHangzhouChina
| | - Yingping Xiao
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro‐Products, Institute of Agro‐product Safety and NutritionZhejiang Academy of Agricultural SciencesHangzhouChina
| |
Collapse
|
22
|
Liu Y, Duan H, Chen Y, Zhang C, Zhao J, Narbad A, Tian F, Zhai Q, Yu L, Chen W. Intraspecific difference of Latilactobacillus sakei in inflammatory bowel diseases: Insights into potential mechanisms through comparative genomics and metabolomics analyses. IMETA 2023; 2:e136. [PMID: 38868211 PMCID: PMC10989848 DOI: 10.1002/imt2.136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/22/2023] [Accepted: 08/30/2023] [Indexed: 06/14/2024]
Abstract
Inflammatory bowel diseases (IBDs) are chronic inflammatory diseases of the gastrointestinal tract that have become a global health burden. Studies have revealed that Latilactobacillus sakei can effectively alleviate various immune diseases, including colitis, rheumatoid arthritis, and metabolic disorders. Here, we obtained 72 strains of L. sakei from 120 fermentation and fecal samples across China. In total, 16 strains from different sources were initially screened in an in vitro Caco-2 model induced by dextran sulfate sodium. Subsequently, six strains (four exhibiting effectiveness and two exhibiting ineffectiveness) were selected for further validation in an in vivo colitis mouse model. The results demonstrated that L. sakei strains exhibited varying degrees of amelioration of the colitis disease process. Notably, L. sakei CCFM1267, the most effective strain, significantly restored colon length and tight-junction protein expression, and reduced the levels of cytokines and associated inflammatory enzymes. Moreover, L. sakei CCFM1267 upregulated the abundance of Enterorhabdus, Alloprevotella, and Roseburia, leading to increased levels of acetic acid and propionic acid. Conversely, the other four strains (L. sakei QJSSZ1L4, QJSSZ4L10, QGZZYRHMT1L6, and QGZZYRHMT2L6) only exhibited a partial remission effect, while L. sakei QJSNT1L10 displayed minimal impact. Therefore, L. sakei CCFM1267 and QJSNT1L10 were selected for further exploration of the mechanisms underlying their differential mitigating effects. Comparative genomics analysis revealed significant variations between the two strains, particularly in genes associated with carbohydrate-active enzymes, such as the glycoside hydrolase family, which potentially contribute to the diverse profiles of short-chain fatty acids in vivo. Additionally, metabolome analysis demonstrated that acetylcholine and indole-3-acetic acid were the main differentiating metabolites of the two strains. Therefore, the strains of L. sakei exhibited varying degrees of effectiveness in alleviating IBD-related symptoms, and the possible reasons for these variations were attributed to discrepancies in the carbohydrate-active enzymes and metabolites among the strains.
Collapse
Affiliation(s)
- Yaru Liu
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Hui Duan
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Ying Chen
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
| | - Jianxin Zhao
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
- National Engineering Research Center for Functional FoodJiangnan UniversityWuxiChina
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
| | - Arjan Narbad
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
- Gut Health and Microbiome Institute Strategic ProgrammeQuadram Institute BioscienceNorwichUK
| | - Fengwei Tian
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
| | - Qixiao Zhai
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
| | - Leilei Yu
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
| | - Wei Chen
- State Key Laboratory of Food Science and ResourcesJiangnan UniversityWuxiChina
- School of Food Science and TechnologyJiangnan UniversityWuxiChina
- National Engineering Research Center for Functional FoodJiangnan UniversityWuxiChina
- International Joint Research Laboratory for ProbioticsJiangnan UniversityWuxiChina
| |
Collapse
|
23
|
Liu Y, Huang Y, He Q, Dou Z, Zeng M, Wang X, Li S. From heart to gut: Exploring the gut microbiome in congenital heart disease. IMETA 2023; 2:e144. [PMID: 38868221 PMCID: PMC10989834 DOI: 10.1002/imt2.144] [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: 08/16/2023] [Revised: 09/14/2023] [Accepted: 09/26/2023] [Indexed: 06/14/2024]
Abstract
Congenital heart disease (CHD) is a prevalent birth defect and a significant contributor to childhood mortality. The major characteristics of CHD include cardiovascular malformations and hemodynamical disorders. However, the impact of CHD extends beyond the circulatory system. Evidence has identified dysbiosis of the gut microbiome in patients with CHD. Chronic hypoxia and inflammation associated with CHD affect the gut microbiome, leading to alterations in its number, abundance, and composition. The gut microbiome, aside from providing essential nutrients, engages in direct interactions with the host immune system and indirect interactions via metabolites. The abnormal gut microbiome or its products can translocate into the bloodstream through an impaired gut barrier, leading to an inflammatory state. Metabolites of the gut microbiome, such as short-chain fatty acids and trimethylamine N-oxide, also play important roles in the development, treatment, and prognosis of CHD. This review discusses the role of the gut microbiome in immunity, gut barrier, neurodevelopment, and perioperative period in CHD. By fostering a better understanding of the cross-talk between CHD and the gut microbiome, this review aims to contribute to improve clinical management and outcomes for CHD patients.
Collapse
Affiliation(s)
- Yuze Liu
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Yuan Huang
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Qiyu He
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Zheng Dou
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Min Zeng
- Department of Pediatric Intensive Care Unit, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Xu Wang
- Department of Pediatric Intensive Care Unit, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Shoujun Li
- Pediatric Cardiac Surgery Centre, Fuwai Hospital, National Centre for Cardiovascular Diseases, State Key Laboratory of Cardiovascular Disease, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| |
Collapse
|
24
|
Gautam A, Bhowmik D, Basu S, Zeng W, Lahiri A, Huson DH, Paul S. Microbiome Metabolome Integration Platform (MMIP): a web-based platform for microbiome and metabolome data integration and feature identification. Brief Bioinform 2023; 24:bbad325. [PMID: 37771003 DOI: 10.1093/bib/bbad325] [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: 04/17/2023] [Revised: 08/12/2023] [Indexed: 09/30/2023] Open
Abstract
A microbial community maintains its ecological dynamics via metabolite crosstalk. Hence, knowledge of the metabolome, alongside its populace, would help us understand the functionality of a community and also predict how it will change in atypical conditions. Methods that employ low-cost metagenomic sequencing data can predict the metabolic potential of a community, that is, its ability to produce or utilize specific metabolites. These, in turn, can potentially serve as markers of biochemical pathways that are associated with different communities. We developed MMIP (Microbiome Metabolome Integration Platform), a web-based analytical and predictive tool that can be used to compare the taxonomic content, diversity variation and the metabolic potential between two sets of microbial communities from targeted amplicon sequencing data. MMIP is capable of highlighting statistically significant taxonomic, enzymatic and metabolic attributes as well as learning-based features associated with one group in comparison with another. Furthermore, MMIP can predict linkages among species or groups of microbes in the community, specific enzyme profiles, compounds or metabolites associated with such a group of organisms. With MMIP, we aim to provide a user-friendly, online web server for performing key microbiome-associated analyses of targeted amplicon sequencing data, predicting metabolite signature, and using learning-based linkage analysis, without the need for initial metabolomic analysis, and thereby helping in hypothesis generation.
Collapse
Affiliation(s)
- Anupam Gautam
- Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany
- International Max Planck Research School "From Molecules to Organisms", Max Planck Institute for Biology Tübingen, Tübingen, Germany
- Cluster of Excellence: EXC 2124: Controlling Microbes to Fight Infection, Tübingen, Germany
| | - Debaleena Bhowmik
- Cell Biology and Physiology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Sayantani Basu
- Department of Computer Science, University of Illinois at Urbana-Champaign, Urbana, IL 61801, United States
| | - Wenhuan Zeng
- Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany
- Cluster of Excellence: EXC 2064: Machine Learning: New Perspectives for Science, University of Tübingen, Tübingen, Germany
| | - Abhishake Lahiri
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
- Infectious Diseases and Immunology Division, CSIR-Indian Institute of Chemical Biology, Kolkata, India
- Centre for Health Science and Technology, JIS Institute of Advanced Studies and Research Kolkata, JIS University, West Bengal, India
| | - Daniel H Huson
- Algorithms in Bioinformatics, Institute for Bioinformatics and Medical Informatics, University of Tübingen, Tübingen, Germany
- International Max Planck Research School "From Molecules to Organisms", Max Planck Institute for Biology Tübingen, Tübingen, Germany
- Cluster of Excellence: EXC 2124: Controlling Microbes to Fight Infection, Tübingen, Germany
| | - Sandip Paul
- Centre for Health Science and Technology, JIS Institute of Advanced Studies and Research Kolkata, JIS University, West Bengal, India
| |
Collapse
|