1
|
Zhang K, Hu M, Yang W, Hu Z, Rong Y, Luo B, Wang M, Cheng Y, Zhang R, Lv N, Zhou Q, Zhang X. Clinical significance of the genetically variable landscape of the gut microbiome in patients with gestational diabetes mellitus patients. Heliyon 2024; 10:e37986. [PMID: 39347390 PMCID: PMC11438002 DOI: 10.1016/j.heliyon.2024.e37986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 10/01/2024] Open
Abstract
Background The composition of the gut microbiome has been recorted to be strongly associated with gestational diabetes mellitus (GDM), but mutational characterization of the microbiome in patients with GDM has been overlooked. Here, we revealed the genetic variation landscape of the gut microbiome and assessed its clinical significance in a cohort of patients with GDM. Methods We employed a macrogenomic dataset made up of a discovery cohort of 54 cases and a validation cohort of 220 cases to screen for high-abundance microbial flora and identified single nucleotide variants (SNVs) and insertions/deletions (indels). Subsequently, we analyzed the mutation spectra of genomes of the intestinal flora by using the previously identified SNVs and identified mutation signatures. Additionally, we utilized the Random Forest algorithm to identify key differential SNVs and elucidated their biological functions and associations with the clinicopathological parameters of GDM. Results We screened 15 key microbial flora and found that the GDM group had more SNVs and indels in the intestinal flora than the control group, with a significant increase in C > T and T > C base mutations and were more susceptible to sequence mutations. Compared to the control group, the GDM group underwent a more significant evolution, as evidenced by the presence of a unique mutational spectrum and mutational characteristics. Random Forest algorithm analysis showed that the combined characterization of five gut microbial species and 21 SNV-related markers was effective in distinguishing between GDM and control subjects in both discovery (area under the curve (AUC) = 0.86) and validation (AUC = 0.73) sets. These markers also revealed that GDM is strongly associated with sphingolipids, galactose, and proteins containing the DUF structural domain. Conclusions The GDM intestinal flora has unique mutational features that correlate significantly with clinicopathological involvement and may be involved in the development of the disease.
Collapse
Affiliation(s)
- Kunna Zhang
- Department of Obstetrics, the First Hospital of Yongnian District, Handan, Hebei Province, China
| | - Menglu Hu
- School of Medicine, Southeast University, Nanjing Province, China
| | - Wentao Yang
- School of Medicine, Southeast University, Nanjing Province, China
| | - Zhexia Hu
- Department of Obstetrics and Gynecology, the Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Yun Rong
- Department of Obstetrics and Gynecology, the Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Biyun Luo
- Department of Obstetrics and Gynecology, the Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Mengjia Wang
- Department of Obstetrics and Gynecology, the Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| | - Yajuan Cheng
- Department of Obstetrics, the First Hospital of Yongnian District, Handan, Hebei Province, China
| | - Rui Zhang
- Department of Obstetrics, the First Hospital of Yongnian District, Handan, Hebei Province, China
| | - Ning Lv
- Department of Obstetrics & Gynecology Peking Union Medical College Hospital Chinese Academy of Medical Sciences Peking Union Medical College National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Qian Zhou
- Department of Obstetrics & Gynecology Peking Union Medical College Hospital Chinese Academy of Medical Sciences Peking Union Medical College National Clinical Research Center for Obstetric & Gynecologic Diseases, Beijing, China
| | - Xueling Zhang
- Department of Obstetrics and Gynecology, the Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei Province, China
| |
Collapse
|
2
|
Jiang S, Ma W, Ma C, Zhang Z, Zhang W, Zhang J. An emerging strategy: probiotics enhance the effectiveness of tumor immunotherapy via mediating the gut microbiome. Gut Microbes 2024; 16:2341717. [PMID: 38717360 PMCID: PMC11085971 DOI: 10.1080/19490976.2024.2341717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 04/08/2024] [Indexed: 05/12/2024] Open
Abstract
The occurrence and progression of tumors are often accompanied by disruptions in the gut microbiota. Inversely, the impact of the gut microbiota on the initiation and progression of cancer is becoming increasingly evident, influencing the tumor microenvironment (TME) for both local and distant tumors. Moreover, it is even suggested to play a significant role in the process of tumor immunotherapy, contributing to high specificity in therapeutic outcomes and long-term effectiveness across various cancer types. Probiotics, with their generally positive influence on the gut microbiota, may serve as effective agents in synergizing cancer immunotherapy. They play a crucial role in activating the immune system to inhibit tumor growth. In summary, this comprehensive review aims to provide valuable insights into the dynamic interactions between probiotics, gut microbiota, and cancer. Furthermore, we highlight recent advances and mechanisms in using probiotics to improve the effectiveness of cancer immunotherapy. By understanding these complex relationships, we may unlock innovative approaches for cancer diagnosis and treatment while optimizing the effects of immunotherapy.
Collapse
Affiliation(s)
- Shuaiming Jiang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Wenyao Ma
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Chenchen Ma
- Department of Human Cell Biology and Genetics, Southern University of Science and Technology, Shenzhen, PR China
| | - Zeng Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Wanli Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| | - Jiachao Zhang
- School of Food Science and Engineering, Hainan University, Haikou, PR China
| |
Collapse
|
3
|
Ma C, Zhang Y, Jiang S, Teng F, Huang S, Zhang J. Cross-cohort single-nucleotide-variant profiling of gut microbiota suggests a novel gut-health assessment approach. mSystems 2023; 8:e0082823. [PMID: 37905808 PMCID: PMC10734426 DOI: 10.1128/msystems.00828-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/21/2023] [Indexed: 11/02/2023] Open
Abstract
IMPORTANCE Most studies focused much on the change in abundance and often failed to explain the microbiome variation related to disease conditions, Herein, we argue that microbial genetic changes can precede the ecological changes associated with the host physiological changes and, thus, would offer a new information layer from metagenomic data for predictive modeling of diseases. Interestingly, we preliminarily found a few genetic biomarkers on SCFA production can cover most chronic diseases involved in the meta-analysis. In the future, it is of both scientific and clinical significance to further explore the dynamic interactions between adaptive evolution and ecology of gut microbiota associated with host health status.
Collapse
Affiliation(s)
- Chenchen Ma
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Yufeng Zhang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Shuaiming Jiang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
| | - Fei Teng
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, China
| | - Shi Huang
- Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Jiachao Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, School of Food Science and Engineering, Hainan University, Haikou, China
- One Health Institute, Hainan University, Haikou, Hainan, China
| |
Collapse
|
4
|
Tong X, Chen L, He S, Liu S, Yao J, Shao Z, Ye Y, Yao S, Lin Z, Zuo J. Forsythia suspensa (Thunb.) Vahl extract ameliorates ulcerative colitis via inhibiting NLRP3 inflammasome activation through the TLR4/MyD88/NF-κB pathway. Immun Inflamm Dis 2023; 11:e1069. [PMID: 38018571 PMCID: PMC10629261 DOI: 10.1002/iid3.1069] [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: 05/04/2023] [Revised: 10/04/2023] [Accepted: 10/18/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Ulcerative colitis (UC), a chronic inflammatory disease, is caused by abnormal immune system reactions resulting in inflammation and ulcers in the large intestine. Phillygenin (PHI) is a natural compound found in Forsythia suspensa (Thunb.) Vahl, which is known for its antipyretic, anti-inflammatory, antiobesity, and other biological activities. However, the therapeutic role and molecular mechanisms of PHI on UC are still insufficiently researched. METHODS In this study, dextran sulfate sodium (DSS) and 2.5% 2,4,6-trinitro-Benzenesulfonic acid (TNBS)-induced acute UC were used to investigate the therapeutic effects of PHI. We evaluated the effects of PHI on disease activity index (DAI), body weight, mortality, intestinal mucosal barrier, cytokine secretion, and macrophage infiltration into colon tissue using various techniques such as flow cytometry, immunofluorescence, enzyme-linked immunosorbent assay (ELISA), RT-qPCR, and Western blot analysis. RESULTS Our findings revealed that PHI has therapeutic properties in UC treatment. PHI was able to maintain body weight, reduce DAI and mortality, restore the intestinal mucosal barrier, and inhibit cytokine secretion. Flow cytometry assay and immunofluorescence indicated that PHI reduces macrophage infiltration into colon tissue. Mechanistically, PHI may exert anti-inflammatory effects by downregulating the TLR4/MyD88/NF-κB pathway and inhibiting the activation of NLRP3 inflammasome. CONCLUSION In conclusion, PHI possesses significant anti-inflammatory properties and is expected to be a potential drug for UC treatment. Our study delves into the underlying mechanisms of PHI therapy and highlights the potential for further research in developing PHI-based treatments for UC.
Collapse
Affiliation(s)
- Xiao Tong
- University of Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Drug ResearchChinese Academy of SciencesShanghaiChina
| | - Li Chen
- State Key Laboratory of Drug ResearchChinese Academy of SciencesShanghaiChina
| | - Shijun He
- Innovation Research Institute of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghaiChina
| | | | - Jiaying Yao
- College of PharmacyJiangxi University of Traditional Chinese MedicineNanchangChina
| | - Zhenguang Shao
- University of Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Drug Research & Natural Products Research CenterChinese Academy of SciencesShanghaiChina
| | - Yang Ye
- University of Chinese Academy of SciencesBeijingChina
- College of PharmacyJiangxi University of Traditional Chinese MedicineNanchangChina
- School of Life Science and TechnologyShanghaiTech UniversityShanghaiChina
| | - Sheng Yao
- University of Chinese Academy of SciencesBeijingChina
- College of PharmacyJiangxi University of Traditional Chinese MedicineNanchangChina
- Zhongshan Institute for Drug DiscoveryChinese Academy of SciencesZhongshanChina
| | - Zemin Lin
- State Key Laboratory of Drug ResearchChinese Academy of SciencesShanghaiChina
| | - Jianping Zuo
- University of Chinese Academy of SciencesBeijingChina
- State Key Laboratory of Drug ResearchChinese Academy of SciencesShanghaiChina
- Laboratory of Immunology and VirologyShanghai University of Traditional Chinese MedicineShanghaiChina
| |
Collapse
|
5
|
Yin R, Wang T, Dai H, Han J, Sun J, Liu N, Dong W, Zhong J, Liu H. Immunogenic molecules associated with gut bacterial cell walls: chemical structures, immune-modulating functions, and mechanisms. Protein Cell 2023; 14:776-785. [PMID: 37013853 PMCID: PMC10599643 DOI: 10.1093/procel/pwad016] [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: 02/05/2023] [Accepted: 03/25/2023] [Indexed: 04/05/2023] Open
Abstract
Interactions between gut microbiome and host immune system are fundamental to maintaining the intestinal mucosal barrier and homeostasis. At the host-gut microbiome interface, cell wall-derived molecules from gut commensal bacteria have been reported to play a pivotal role in training and remodeling host immune responses. In this article, we review gut bacterial cell wall-derived molecules with characterized chemical structures, including peptidoglycan and lipid-related molecules that impact host health and disease processes via regulating innate and adaptive immunity. Also, we aim to discuss the structures, immune responses, and underlying mechanisms of these immunogenic molecules. Based on current advances, we propose cell wall-derived components as important sources of medicinal agents for the treatment of infection and immune diseases.
Collapse
Affiliation(s)
- Ruopeng Yin
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Tao Wang
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Huanqin Dai
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Junjie Han
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jingzu Sun
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Ningning Liu
- CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
| | - Wang Dong
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Jin Zhong
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, 100101 Beijing, China
| | - Hongwei Liu
- State Key Laboratory of Mycology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, China
- Savaid Medical School, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
6
|
Shen S, Wang J, Ma C, Chen Y, Ding H, Zhang J. Understanding the "individual drug reaction" from the perspective of the interaction between probiotics and lovastatin in vitro and in vivo. MICROBIOME 2023; 11:209. [PMID: 37749663 PMCID: PMC10518969 DOI: 10.1186/s40168-023-01658-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 08/25/2023] [Indexed: 09/27/2023]
Abstract
BACKGROUND The existence of the gut microbiota produces an "individual drug reaction." As members of the intestinal microbiota, probiotics, although they have prebiotic functions, may accelerate the degradation of drugs, thereby affecting drug efficacy. Lovastatin is one of the well-recognized lipid-lowering drugs. Its main action site is the liver. Therefore, if it is degraded in advance by gastrointestinal probiotics, its efficacy may be reduced. RESULTS Here, we designed a two-stage experiment in vitro and in vivo to explore the degradation of lovastatin by probiotics. In vitro, the degradation of lovastatin by 83 strains of Lactiplantibacillus plantarum and the "star strain" Lacticaseibacillus paracasei strain Shirota was investigated by high-performance liquid chromatography (HPLC). The results showed that probiotics could degrade lovastatin to varying degrees. Subsequently, we selected Lactiplantibacillus plantarum A5 (16.87%) with the strongest ability to degrade lovastatin, Lactiplantibacillus plantarum C3 (4.61%) with the weakest ability to degrade lovastatin and Lacticaseibacillus paracasei strain Shirota (17.6%) as representative probiotics for in vivo experiments. In vivo, the therapeutic effect of lovastatin combined with probiotics on golden hamsters with mixed hyperlipidemia was evaluated by measuring blood indicators, intestinal microbiota metagenomic sequencing, and the liver transcriptome. The results showed that the intake of probiotics did not affect the efficacy of lovastatin and could slow the inflammatory reaction of the liver. CONCLUSIONS The supplementation of probiotics produced beneficial metabolites in the intestine by promoting beneficial microbes. Intestinal metabolites affected the expression of the liver genes through the gut-liver axis, increased the relative content of the essential amino acids, and finally improved the liver inflammatory response of the host. This study aims to reveal the impact of probiotics on the human body from a unique perspective, suggesting the impact of taking probiotics while taking drugs. Video Abstract.
Collapse
Affiliation(s)
- Siyuan Shen
- School of Food Science and Engineering, Hainan University, Haikou, China
- One Health Institute, Hainan University, Haikou, 570228, Hainan, China
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, 570228, China
| | - Jun Wang
- School of Food Science and Engineering, Hainan University, Haikou, China
- One Health Institute, Hainan University, Haikou, 570228, Hainan, China
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, 570228, China
| | - Chenchen Ma
- School of Food Science and Engineering, Hainan University, Haikou, China
- One Health Institute, Hainan University, Haikou, 570228, Hainan, China
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, 570228, China
| | - Yanni Chen
- School of Food Science and Engineering, Hainan University, Haikou, China
- One Health Institute, Hainan University, Haikou, 570228, Hainan, China
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, 570228, China
| | - Hao Ding
- School of Food Science and Engineering, Hainan University, Haikou, China
- One Health Institute, Hainan University, Haikou, 570228, Hainan, China
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, 570228, China
| | - Jiachao Zhang
- School of Food Science and Engineering, Hainan University, Haikou, China.
- One Health Institute, Hainan University, Haikou, 570228, Hainan, China.
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Haikou, 570228, China.
| |
Collapse
|
7
|
Chen A, Zhang J, Zhang Y. Gut microbiota in heart failure and related interventions. IMETA 2023; 2:e125. [PMID: 38867928 PMCID: PMC10989798 DOI: 10.1002/imt2.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 05/28/2023] [Accepted: 06/04/2023] [Indexed: 06/14/2024]
Abstract
Heart failure (HF) is a sophisticated syndrome with structural or functional impairment of ventricular filling or ejection of blood, either causing symptoms and signs or being asymptomatic. HF is a major global health issue affecting about 64.3 million people worldwide. The gut microbiota refers to the complex ecosystem of microorganisms, mainly bacteria, in the gut. Studies have revealed that the gut microbiota is associated with many diseases ranging from neurodegenerative diseases to inflammatory bowel disease and cardiovascular diseases. The gut hypothesis of HF suggests that low cardiac output and systemic circulation congestion would cause insufficient intestinal perfusion, leading to ischemia and intestinal barrier dysfunction. The resulting bacterial translocation would contribute to inflammation. Recent studies have refined the hypothesis that changes of metabolites in the gut microbiota have a close relationship with HF. Thus, the gut microbiota has emerged as a potential therapeutic target for HF due to both its critical role in regulating host physiology and metabolism and its pivotal role in the development of HF. This review article aims to provide an overview of the current understanding of the gut microbiota's involvement in HF, including the introduction of the gut hypothesis of HF, its association with HF progression, the potential mechanisms involved mediated by the gut microbiota metabolites, and the impact of various interventions on the gut microbiota, including dietary interventions, probiotic therapy, fecal microbiota transplantation, antibiotics, and so on. While the gut hypothesis of HF is refined with up-to-date knowledge and the gut microbiota presents a promising target for HF therapy, further research is still needed to further understand the underlying mechanisms between gut microbiota and HF, the efficacy of these interventions, and contribute to the health of HF patients.
Collapse
Affiliation(s)
- An‐Tian Chen
- Department of Cardiology, State Key Laboratory of Cardiovascular Disease, Fuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular DiseasesBeijingChina
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular DiseasesBeijingChina
| | - Jian Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular DiseasesBeijingChina
- Key Laboratory of Clinical Research for Cardiovascular MedicationsNational Health CommitteeBeijingChina
| | - Yuhui Zhang
- State Key Laboratory of Cardiovascular Disease, Heart Failure Center, Fuwai HospitalChinese Academy of Medical Sciences & Peking Union Medical College/National Center for Cardiovascular DiseasesBeijingChina
| |
Collapse
|
8
|
Gao S, Gao X, Zhu R, Wu D, Feng Z, Jiao N, Sun R, Gao W, He Q, Liu Z, Zhu L. Microbial genes outperform species and SNVs as diagnostic markers for Crohn's disease on multicohort fecal metagenomes empowered by artificial intelligence. Gut Microbes 2023; 15:2221428. [PMID: 37278203 PMCID: PMC10246480 DOI: 10.1080/19490976.2023.2221428] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/16/2023] [Indexed: 06/07/2023] Open
Abstract
Dysbiosis of gut microbial community is associated with the pathogenesis of CD and may serve as a promising noninvasive diagnostic tool. We aimed to compare the performances of the microbial markers of different biological levels by conducting a multidimensional analysis on the microbial metagenomes of CD. We collected fecal metagenomic datasets generated from eight cohorts that altogether include 870 CD patients and 548 healthy controls. Microbial alterations in CD patients were assessed at multidimensional levels including species, gene, and SNV level, and then diagnostic models were constructed using artificial intelligence algorithm. A total of 227 species, 1047 microbial genes, and 21,877 microbial SNVs were identified that differed between CD and controls. The species, gene, and SNV models achieved an average AUC of 0.97, 0.95, and 0.77, respectively. Notably, the gene model exhibited superior diagnostic capability, achieving an average AUC of 0.89 and 0.91 for internal and external validations, respectively. Moreover, the gene model was specific for CD against other microbiome-related diseases. Furthermore, we found that phosphotransferase system (PTS) contributed substantially to the diagnostic capability of the gene model. The outstanding performance of PTS was mainly explained by genes celB and manY, which demonstrated high predictabilities for CD with metagenomic datasets and was validated in an independent cohort by qRT-PCR analysis. Our global metagenomic analysis unravels the multidimensional alterations of the microbial communities in CD and identifies microbial genes as robust diagnostic biomarkers across geographically and culturally distinct cohorts.
Collapse
Affiliation(s)
- Sheng Gao
- Department of Gastroenterology, the Shanghai Tenth People’s Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, P. R. China
| | - Xiang Gao
- Department of Gastroenterology, the Shanghai Tenth People’s Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, P. R. China
| | - Ruixin Zhu
- Department of Gastroenterology, the Shanghai Tenth People’s Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, P. R. China
| | - Dingfeng Wu
- National Clinical Research Center for Child Health, the Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Zhongsheng Feng
- Department of Gastroenterology, the Shanghai Tenth People’s Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, P. R. China
| | - Na Jiao
- National Clinical Research Center for Child Health, the Children’s Hospital, Zhejiang University School of Medicine, Hangzhou, P. R. China
| | - Ruicong Sun
- Department of Gastroenterology, the Shanghai Tenth People’s Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, P. R. China
| | - Wenxing Gao
- Department of Gastroenterology, the Shanghai Tenth People’s Hospital, School of Medicine, School of Life Sciences and Technology, Tongji University, Shanghai, P. R. China
| | - Qing He
- Departments of Gastroenterology and Nutrition, the Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, P.R. China
| | - Zhanju Liu
- Center for IBD Research, Department of Gastroenterology, the Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, P. R. China
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases; Biomedical Innovation Center, Sun Yat-Sen University, Guangzhou, P. R. China
| | - Lixin Zhu
- Guangdong Institute of Gastroenterology; Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases; Biomedical Innovation Center, Sun Yat-Sen University, Guangzhou, P. R. China
- Department of General Surgery, The Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, P. R. China
| |
Collapse
|
9
|
Wu Y, Jha R, Li A, Liu H, Zhang Z, Zhang C, Zhai Q, Zhang J. Probiotics (Lactobacillus plantarum HNU082) Supplementation Relieves Ulcerative Colitis by Affecting Intestinal Barrier Functions, Immunity-Related Gene Expression, Gut Microbiota, and Metabolic Pathways in Mice. Microbiol Spectr 2022; 10:e0165122. [PMID: 36321893 PMCID: PMC9769980 DOI: 10.1128/spectrum.01651-22] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 10/14/2022] [Indexed: 11/07/2022] Open
Abstract
Probiotics can effectively improve ulcerative colitis (UC), but the mechanism is still unclear. Here, shotgun metagenome and transcriptome analyses were performed to explore the therapeutic effect and the mechanism of the probiotic Lactobacillus plantarum HNU082 (Lp082) on UC. The results showed that Lp082 treatment significantly ameliorated dextran sulfate sodium (DSS)-induced UC in mice, which was manifested as increases in body weight, water intake, food intake, and colon length and decreases in disease activity index (DAI), immune organ index, inflammatory factors, and histopathological scores after Lp082 intake. An in-depth study discovered that Lp082 could improve the intestinal mucosal barrier and relieve inflammation by cooptimizing the biological barrier, chemical barrier, mechanical barrier, and immune barrier. Specifically, Lp082 rebuilt the biological barrier by regulating the intestinal microbiome and increasing the production of short-chain fatty acids (SCFAs). Lp082 improved the chemical barrier by reducing intercellular cell adhesion molecule-1 (ICAM-1) and vascular cell adhesion molecule (VCAM) and increasing goblet cells and mucin2. Lp082 ameliorated the mechanical barrier by increasing zonula occludens-1 (ZO-1), zonula occludens-2 (ZO-2), and occludin while decreasing claudin-1 and claudin-2. Lp082 optimized the immune barrier by reducing the content of interleukin-1β (IL-1β), IL-6, tumor necrosis factor-α (TNF-α), myeloperoxidase (MPO), and interferon-γ (IFN-γ) and increasing IL-10, transforming growth factor-β1 (TGF-β1), and TGF-β2, inhibiting the NF-κB signaling pathway. Taken together, probiotic Lp082 can play a protective role in a DSS-induced colitis mouse model by protecting the intestinal mucosal barrier, attenuating the inflammatory response, and regulating microbial imbalance. This study provides support for the development of probiotic-based microbial products as an alternative treatment strategy for UC. IMPORTANCE Many studies have focused on the therapeutic effect of probiotics on ulcerative colitis (UC), but few studies have paid attention to the mechanism of probiotics, especially the therapeutic effect. This study suggests that Lp082 has a therapeutic effect on colitis in mice. Its mechanisms of action include protecting the mucosal barrier and actively modulating the gut microbiome, modulating inflammatory pathways, and reducing neutrophil infiltration. Our study enriches the mechanism and provides a new prospect for probiotics in the treatment of colitis, helps to deepen the understanding of the intestinal mucosal barrier, and provides guidance for the future probiotic treatment of human colitis.
Collapse
Affiliation(s)
- Yuqing Wu
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, College of Food Science and Engineering, Hainan University, Haikou, China
| | - Rajesh Jha
- Department of Human Nutrition, Food and Animal Sciences, College of Tropical Agriculture and Human Resources, University of Hawaii at Manoa, Honolulu, Hawaii, USA
| | - Ao Li
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, College of Food Science and Engineering, Hainan University, Haikou, China
| | - Huanwei Liu
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, College of Food Science and Engineering, Hainan University, Haikou, China
| | - Zeng Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, College of Food Science and Engineering, Hainan University, Haikou, China
| | - Chengcheng Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Qixiao Zhai
- School of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Jiachao Zhang
- Key Laboratory of Food Nutrition and Functional Food of Hainan Province, College of Food Science and Engineering, Hainan University, Haikou, China
- One Health Institute, Hainan University, Haikou, China
| |
Collapse
|