1
|
Moghaddam ZS, Dehghan A, Halimi S, Najafi F, Nokhostin A, Naeini AE, Akbarzadeh I, Ren Q. Bacterial Extracellular Vesicles: Bridging Pathogen Biology and Therapeutic Innovation. Acta Biomater 2025:S1742-7061(25)00352-6. [PMID: 40349898 DOI: 10.1016/j.actbio.2025.05.028] [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: 12/16/2024] [Revised: 04/14/2025] [Accepted: 05/07/2025] [Indexed: 05/14/2025]
Abstract
The main role of bacterial extracellular vesicles (BEVs) has been associated with various processes such as intercellular communication and host-pathogen interactions. This comprehensive review explores the multifaceted functions of BEVs across different biological domains, emphasizing their dual nature as contributors to disease and potential vehicles for therapeutic intervention. We examine the intricate interactions of BEVs within bacterial communities and between bacteria and hosts, their involvement in disease development through cargo delivery mechanisms, and their beneficial impact on microbial ecology. The review also highlights BEVs' applications in biomedical field, where they are revolutionizing vaccine development, targeted drug delivery, and cancer therapy. By utilizing the inherent properties of BEVs for controlled drug release, targeted antigen delivery, and immune modulation, they offer a promising frontier in precision medicine. In addition, the diagnostic potential of BEVs is explored through their utility as biomarkers, providing valuable insights into disease states and treatment efficacy. Looking forward, this review underscores the challenges and opportunities in translating BEV research to clinical practice, promoting the need of standardized methods in BEV characterization and scaling up production. The diverse abilities of BEVs, ranging from contributing to pathogen virulence to driving therapeutic innovation, highlight their potential as a cornerstone in the future of biomedical advancements. STATEMENT OF SIGNIFICANCE: Bacterial extracellular vesicles (BEVs) are emerging as pivotal players in both pathogenesis and therapeutic innovation. This review explores their dual nature as agents of disease and as promising biomaterials for biomedical applications, and provides a comprehensive survey on their involvement in disease mechanisms and microbial ecology, and their potential in biomedical applications such as vaccine development, targeted drug delivery, cancer therapy, and diagnosis. It highlights the complex interactions of BEVs within bacterial communities and between bacteria and hosts. This review also addresses current advancements, challenges, and opportunities in translating BEV research into clinical practice. The insights presented here position BEVs as a cornerstone in the future of biomedical advancements, advocating for standardized methods in BEV characterization and scalable production techniques.
Collapse
Affiliation(s)
| | - Ashkan Dehghan
- W Booth School of Engineering Practice and Technology Faculty of Engineering, McMaster University Hamilton, ON, Canada, L8S 0A3
| | - Saba Halimi
- Department of Microbial Biotechnology, School of Biology, College of Science, University of Tehran, 14155-6455 Tehran, Iran
| | - Fatemeh Najafi
- Department of Chemical Engineering, Pennsylvania State University, University Park, Pennsylvania 16802-1503, United States
| | - Ali Nokhostin
- Medical Sciences & Technologies Faculty, Science and Research Branch, Islamic Azad University, Tehran 1477893855, Iran
| | | | - Iman Akbarzadeh
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW, Australia.
| | - Qun Ren
- Laboratory for Biointerfaces, Empa Swiss Federal Laboratories for Materials Science and Technology, 9014 St. Gallen, Switzerland.
| |
Collapse
|
2
|
Tang CT, Wu Y, Tao Q, Zeng CY, Chen YX. Thalidomide mitigates Crohn's disease colitis by modulating gut microbiota, metabolites, and regulatory T cell immunity. J Pharm Anal 2025; 15:101121. [PMID: 40309194 PMCID: PMC12041782 DOI: 10.1016/j.jpha.2024.101121] [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: 06/26/2024] [Revised: 09/30/2024] [Accepted: 10/15/2024] [Indexed: 05/02/2025] Open
Abstract
Thalidomide (THA) is renowned for its potent anti-inflammatory properties. This study aimed to elucidate its underlying mechanisms in the context of Crohn's disease (CD) development. Mouse colitis models were established by dextran sulfate sodium (DSS) treatment. Fecal microbiota and metabolites were analyzed by metagenomic sequencing and mass spectrometry, respectively. Antibiotic-treated mice served as models for microbiota depletion and transplantation. The expression of forkhead box P3+ (FOXP3+) regulatory T cells (Tregs) was measured by flow cytometry and immunohistochemical assay in colitis model and patient cohort. THA inhibited colitis in DSS-treated mice by altering the gut microbiota profile, with an increased abundance of probiotics Bacteroides fragilis, while pathogenic bacteria were depleted. In addition, THA increased beneficial metabolites bile acids and significantly restored gut barrier function. Transcriptomic profiling revealed that THA inhibited interleukin-17 (IL-17), IL-1β and cell cycle signaling. Fecal microbiota transplantation from THA-treated mice to microbiota-depleted mice partly recapitulated the effects of THA. Specifically, increased level of gut commensal B. fragilis was observed, correlated with elevated levels of the microbial metabolite 3alpha-hydroxy-7-oxo-5beta-cholanic acid (7-ketolithocholic acid, 7-KA) following THA treatment. This microbial metabolite may stable FOXP3 expression by targeting the receptor FMR1 autosomal homolog 1 (FXR1) to inhibit autophagy. An interaction between FOXP3 and FXR1 was identified, with binding regions localized to the FOXP3 domain (aa238-335) and the FXR1 domain (aa82-222), respectively. Conclusively, THA modulates the gut microbiota and metabolite profiles towards a more beneficial composition, enhances gut barrier function, promotes the differentiation of FOXP3+ Tregs and curbs pro-inflammatory pathways.
Collapse
Affiliation(s)
- Chao-Tao Tang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Postdoctoral Innovation Practice Base, The First Affiliated Hospital of Nanchang University, Nanchang, 330006, China
| | - Yonghui Wu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Qing Tao
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Chun-Yan Zeng
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- Department of Gastroenterology, Jiangxi Province Hospital of Integrated Chinese and Western Medicine, Nanchang, 330003, China
| | - You-Xiang Chen
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| |
Collapse
|
3
|
El Leithy AA, Youssef ASED, Nassar A, Aziz RK, Khaled NM, Mahrous MT, Farahat GN, Mohamed AH, Bakr YM. Long-read 16S rRNA amplicon sequencing reveals microbial characteristics in patients with colorectal adenomas and carcinoma lesions in Egypt. Gut Pathog 2025; 17:8. [PMID: 39894814 PMCID: PMC11789410 DOI: 10.1186/s13099-025-00681-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 01/23/2025] [Indexed: 02/04/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) is among the five leading causes of cancer incidence and mortality. During the past decade, the role of the gut microbiota and its dysbiosis in colorectal tumorigenesis has been emphasized. Metagenomics and amplicon-based microbiome profiling provided insights into the potential role of microbial dysbiosis in the development of CRC. AIM To address the scarcity of information on differential microbiome composition of tumor tissue in comparison to adenomas and the lack of such data from Egyptian patients with CRC. MATERIALS AND METHODS Long-read nanopore sequencing of 16S rRNA amplicons was used to profile the colonic microbiota from fresh colonoscopic biopsy samples of Egyptian patients with CRC and patients with colonic polyps. RESULTS Species richness of CRC lesions was significantly higher than that in colonic polyps (p-value = 0.0078), while evenness of the CRC group was significantly lower than the colonic polyps group (p-value = 0.0055). Both species richness and Shannon diversity index of the late onset CRC samples were significantly higher than those of the early onset ones. The Firmicutes-to-Bacteroidetes (F/B) ratio was significantly higher in the CRC group than in the colonic polyps group (p-value = 0.0054), and significantly higher in samples from early-onset CRC. The Enterococcus spp. were significantly overabundant in patients with rectal cancer and early-onset CRC, while Staphylococcus spp. were significantly higher in patients with sigmoid cancer and late-onset CRC. In addition, the relative abundance of Fusobacterium nucleatum was significantly higher in CRC patients. CONCLUSION Differentiating trends were identified at phylum, genus, and species levels, despite the inter-individual differences. In summary, this study addressed the microbial dysbiosis associated with CRC and colonic polyps groups, paving the way for a better understanding of the pathogenesis of early and late-onset CRC in Egyptian patients.
Collapse
Affiliation(s)
- Asmaa A El Leithy
- College of Biotechnology, Misr University for Science and Technology, Giza, Egypt.
| | - Amira Salah El-Din Youssef
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Kasr Al-Aini st., Fom El-Khaleeg, Cairo, 11976, Egypt.
| | - Auhood Nassar
- Virology and Immunology Unit, Cancer Biology Department, National Cancer Institute, Cairo University, Kasr Al-Aini st., Fom El-Khaleeg, Cairo, 11976, Egypt
| | - Ramy K Aziz
- Department of Microbiology and Immunology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Center for Genome and Microbiome Research, Cairo University, Cairo, Egypt
| | - Nadin M Khaled
- College of Biotechnology, Misr University for Science and Technology, Giza, Egypt
| | - Mina T Mahrous
- College of Biotechnology, Misr University for Science and Technology, Giza, Egypt
| | - Ghobrial N Farahat
- College of Biotechnology, Misr University for Science and Technology, Giza, Egypt
| | - Aya H Mohamed
- College of Biotechnology, Misr University for Science and Technology, Giza, Egypt
| | - Yasser Mabrouk Bakr
- Cancer Biology Department, National Cancer Institute, Cairo University, Cairo, Egypt
| |
Collapse
|
4
|
Tasoujlu M, Sharifi Y, Ghahremani M, Alizadeh K, Babaie F, Hosseiniazar MM. Evaluation of variations in predominant gut microbiota members in inflammatory bowel disease using real-time PCR. Mol Biol Rep 2025; 52:143. [PMID: 39836282 DOI: 10.1007/s11033-025-10254-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Inflammatory Bowel Disease (IBD) is a persistent ailment that impacts many individuals worldwide. The interaction between the immune system and gut microbiome is thought to influence IBD development. This study aimed to assess some microbiota in IBD patients compared to healthy individuals. The investigation involved a selected group of twenty patients suffering from IBD and an equal number of healthy participants. Stool specimens were obtained and analyzed for Lactobacillus, Bifidobacterium, Bacteroides, Clostridium leptum, Akkermansia muciniphila, Fusobacterium and Enterobacteriaceae using real-time PCR. The findings indicated significantly higher levels of Bifidobacterium in IBD patients (Pv = 0.009) and lower levels of A. muciniphila (Pv = 0.003) healthy individuals. Other bacteria tested did not show significant differences. The study suggests that the progression of IBD patients could be influenced by the rising of Bifidobacterium and the declining of A. muciniphila. Targeting these bacteria could lead to improved treatments and quality of life for those with IBD.
Collapse
Affiliation(s)
- Mina Tasoujlu
- Cellular and Molecular Research Center, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Department of Microbiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Yaeghob Sharifi
- Cellular and Molecular Research Center, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
- Department of Microbiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran.
| | - Maryam Ghahremani
- Department of Microbiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Kasra Alizadeh
- Department of Microbiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Farhad Babaie
- Department of Immunology and Genetics, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | | |
Collapse
|
5
|
Lakemeyer M, Latorre R, Blazkova K, Jensen D, Wood HM, Shakil N, Thomas SC, Saxena D, Mulpuri Y, Poolman D, de Haro PD, Keller LJ, Reed DE, Schmidt BL, Lomax AE, Bunnett NW, Bogyo M. Identification of a secreted protease from Bacteroides fragilis that induces intestinal pain and inflammation by cleavage of PAR 2. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633241. [PMID: 39868234 PMCID: PMC11761754 DOI: 10.1101/2025.01.15.633241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Protease-activated receptor 2 (PAR2) is a central regulator of intestinal barrier function, inflammation and pain. Upregulated intestinal proteolysis and PAR2-signaling are implicated in inflammatory bowel diseases (IBDs) and irritable bowel syndrome (IBS). To identify potential bacterial regulators of PAR2 activity, we developed a functional assay for PAR2 processing and used it to screen conditioned media from a library of diverse gut commensal microbes. We found that multiple bacteria secrete proteases that cleave host PAR2. Using chemoproteomic profiling with a covalent irreversible inhibitor, we identified a previously uncharacterized Bacteroides fragilis serine protease Bfp1, and showed that it cleaves and activates PAR2 in multicellular and murine models. PAR2 cleavage by Bfp1 disrupts the intestinal barrier, sensitizes nociceptors, and triggers colonic inflammation and abdominal pain. Collectively, our findings uncover Bfp1-mediated PAR2-processing as a new axis of host-commensal-interaction in the gut that has the potential to be targeted for therapeutic intervention in IBD or IBS.
Collapse
Affiliation(s)
- Markus Lakemeyer
- Institute for Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller-University Jena; Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena; Jena, Germany
- Department of Pathology, Stanford University School of Medicine; Stanford, CA, USA
| | - Rocco Latorre
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
| | - Kristyna Blazkova
- Department of Pathology, Stanford University School of Medicine; Stanford, CA, USA
| | - Dane Jensen
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
- Translational Research Center, College of Dentistry, New York University; New York, USA
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, Bluestone Center for Clinical Research; New York, USA
| | - Hannah M Wood
- Gastrointestinal Diseases Research Unit, Queen's University; Kingston, Ontario, Canada
| | - Nayab Shakil
- Institute for Organic Chemistry and Macromolecular Chemistry, Friedrich-Schiller-University Jena; Jena, Germany
| | - Scott C Thomas
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
| | - Deepak Saxena
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
| | - Yatendra Mulpuri
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
| | - David Poolman
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
| | - Paz Duran de Haro
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
- Translational Research Center, College of Dentistry, New York University; New York, USA
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, Bluestone Center for Clinical Research; New York, USA
| | - Laura J Keller
- Department of Pathology, Stanford University School of Medicine; Stanford, CA, USA
| | - David E Reed
- Gastrointestinal Diseases Research Unit, Queen's University; Kingston, Ontario, Canada
| | - Brian L Schmidt
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
- Translational Research Center, College of Dentistry, New York University; New York, USA
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, Bluestone Center for Clinical Research; New York, USA
| | - Alan E Lomax
- Gastrointestinal Diseases Research Unit, Queen's University; Kingston, Ontario, Canada
| | - Nigel W Bunnett
- Department of Molecular Pathobiology, College of Dentistry, New York University; New York, USA
- Pain Research Center, New York University; New York, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University School of Medicine; Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine; Stanford, CA, USA
| |
Collapse
|
6
|
Wang T, Huang Y, Jiang P, Yuan X, Long Q, Yan X, Huang Y, Wang Z, Li C. Research progress on anti-inflammatory drugs for preventing colitis-associated colorectal cancer. Int Immunopharmacol 2025; 144:113583. [PMID: 39580861 DOI: 10.1016/j.intimp.2024.113583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 11/04/2024] [Accepted: 11/05/2024] [Indexed: 11/26/2024]
Abstract
Colorectal cancer (CRC) is the third most prevalent malignancy worldwide. Inflammatory bowel diseases (IBD) encompass a group of chronic intestinal inflammatory disorders, including ulcerative colitis (UC) and Crohn's disease (CD). As a chronic inflammatory bowel disease, UC may persist and elevate the risk of malignancy, thereby contributing to the development of colorectal cancer, known as colitis-associated colorectal cancer (CAC). Chronic intestinal inflammation is a significant risk factor for colorectal cancer, and the incidence of colitis-associated colorectal cancer continues to rise. Current studies indicate that therapeutic agents targeting inflammation and key molecules or signaling pathways involved in the inflammatory process may effectively prevent and treat CAC. Mechanistically, drugs with anti-inflammatory or modulatory effects on inflammation-related pathways may exert preventive or therapeutic roles in CAC through multiple molecules or signaling pathways implicated in tumor development. Moreover, the development or discovery of novel drugs with anti-inflammatory properties to prevent or delay CAC progression is becoming an emerging field in fighting against CRC. Therefore, this review aims to summarize drugs that prevent or delay CAC through modulating anti-inflammatory pathways. First, we categorize the published studies exploring the role of anti-inflammatory in CAC prevention. Second, we highlight the specific molecular mechanisms underlying the anti-inflammatory effect of the above-mentioned drugs. Finally, we discuss the potential and challenges associated with clinical application of these drugs. It is hoped that this review offers new insights for further drug development and mechanism exploration.
Collapse
Affiliation(s)
- Tong Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan 610052, PR China
| | | | - Peng Jiang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan 610052, PR China
| | - Xin Yuan
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan 610052, PR China
| | - Qian Long
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan 610052, PR China
| | - Xiaochen Yan
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan 610052, PR China
| | - Yuwei Huang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan 610052, PR China
| | - Zongkui Wang
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan 610052, PR China.
| | - Changqing Li
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences & Peking Union Medical College, Chengdu, Sichuan 610052, PR China.
| |
Collapse
|
7
|
Yao M, Qu Y, Zheng Y, Guo H. The effect of exercise on depression and gut microbiota: Possible mechanisms. Brain Res Bull 2025; 220:111130. [PMID: 39557221 DOI: 10.1016/j.brainresbull.2024.111130] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/09/2024] [Accepted: 11/12/2024] [Indexed: 11/20/2024]
Abstract
Exercise can effectively prevent and treat depression and anxiety, with gut microbiota playing a crucial role in this process. Studies have shown that exercise can influence the diversity and composition of gut microbiota, which in turn affects depression through immune, endocrine, and neural pathways in the gut-brain axis. The effectiveness of exercise varies based on its type, intensity, and duration, largely due to the different changes in gut microbiota. This article summarizes the possible mechanisms by which exercise affects gut microbiota and how gut microbiota influences depression. Additionally, we reviewed literature on the effects of exercise on depression at different intensities, types, and durations to provide a reference for future exercise-based therapies for depression.
Collapse
Affiliation(s)
- Mingchen Yao
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yaqi Qu
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Yalin Zheng
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Hao Guo
- School of Sports Medicine and Rehabilitation, Beijing Sport University, Beijing, China.
| |
Collapse
|
8
|
Wang Y, Bai M, Peng Q, Li L, Tian F, Guo Y, Jing C. Angiogenesis, a key point in the association of gut microbiota and its metabolites with disease. Eur J Med Res 2024; 29:614. [PMID: 39710789 DOI: 10.1186/s40001-024-02224-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
The gut microbiota is a complex and dynamic ecosystem that plays a crucial role in human health and disease, including obesity, diabetes, cardiovascular diseases, neurodegenerative diseases, inflammatory bowel disease, and cancer. Chronic inflammation is a common feature of these diseases and is closely related to angiogenesis (the process of forming new blood vessels), which is often dysregulated in pathological conditions. Inflammation potentially acts as a central mediator. This abstract aims to elucidate the connection between the gut microbiota and angiogenesis in various diseases. The gut microbiota influences angiogenesis through various mechanisms, including the production of metabolites that directly or indirectly affect vascularization. For example, short-chain fatty acids (SCFAs) such as butyrate, propionate, and acetate are known to regulate immune responses and inflammation, thereby affecting angiogenesis. In the context of cardiovascular diseases, the gut microbiota promotes atherosclerosis and vascular dysfunction by producing trimethylamine N-oxide (TMAO) and other metabolites that promote inflammation and endothelial dysfunction. Similarly, in neurodegenerative diseases, the gut microbiota may influence neuroinflammation and the integrity of the blood-brain barrier, thereby affecting angiogenesis. In cases of fractures and wound healing, the gut microbiota promotes angiogenesis by activating inflammatory responses and immune effects, facilitating the healing of tissue damage. In cancer, the gut microbiota can either inhibit or promote tumor growth and angiogenesis, depending on the specific bacterial composition and their metabolites. For instance, some bacteria can activate inflammasomes, leading to the production of inflammatory factors that alter the tumor immune microenvironment and activate angiogenesis-related signaling pathways, affecting tumor angiogenesis and metastasis. Some bacteria can directly interact with tumor cells, activating angiogenesis-related signaling pathways. Diet, as a modifiable factor, significantly influences angiogenesis through diet-derived microbial metabolites. Diet can rapidly alter the composition of the microbiota and its metabolic activity, thereby changing the concentration of microbial-derived metabolites and profoundly affecting the host's immune response and angiogenesis. For example, a high animal protein diet promotes the production of pro-atherogenic metabolites like TMAO, activating inflammatory pathways and interfering with platelet function, which is associated with the severity of coronary artery plaques, peripheral artery disease, and cardiovascular diseases. A diet rich in dietary fiber promotes the production of SCFAs, which act as ligands for cell surface or intracellular receptors, regulating various biological processes, including inflammation, tissue homeostasis, and immune responses, thereby influencing angiogenesis. In summary, the role of the gut microbiota in angiogenesis is multifaceted, playing an important role in disease progression by affecting various biological processes such as inflammation, immune responses, and multiple signaling pathways. Diet-derived microbial metabolites play a crucial role in linking the gut microbiota and angiogenesis. Understanding the complex interactions between diet, the gut microbiota, and angiogenesis has the potential to uncover novel therapeutic targets for managing these conditions. Therefore, interventions targeting the gut microbiota and its metabolites, such as through fecal microbiota transplantation (FMT) and the application of probiotics to alter the composition of the gut microbiota and enhance the production of beneficial metabolites, present a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Mingshuai Bai
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Qifan Peng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Feng Tian
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Ying Guo
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Changqing Jing
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
9
|
Wang D, Zhao L, Lin J, Wang Y, Gao H, Liu W, Li Q, Zhang L, Kang X, Guo K. Analysis of Characteristics of Bovine-Derived Non-Enterotoxigenic Bacteroides fragilis and Validation of Potential Probiotic Effects. Microorganisms 2024; 12:2319. [PMID: 39597708 PMCID: PMC11596406 DOI: 10.3390/microorganisms12112319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/04/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024] Open
Abstract
Bacteroides fragilis is a new generation of probiotics, and its probiotic effects on humans and some animals have been verified. However, research on B. fragilis in cattle is still lacking. In this study, 24 stool samples were collected from two large-scale cattle farms in Wuzhong, Ningxia, including 12 diarrheal and 12 normal stools. A non-toxigenic Bacteroides fragilis (NTBF) was isolated and identified by 16S rRNA high-throughput sequencing and named BF-1153; genome composition and genome functional analyses were carried out to reflect the biological characteristics of the BF-1153 strain. A cluster analysis of BF-1153 was performed using Mega X to explore its genetic relationship. In addition, Cell Counting Kit-8 (CCK8) was used to determine the toxic effects of the strain on human ileocecal colorectal adenocarcinoma cell line cells (HCT-8), Madin-Darby bovine kidney cells (MDBK), and intestinal porcine epithelial cells (IPECs). The results showed that BF-1153 conformed to the biological characteristics of B. fragilis. BF-1153 had no toxic effects on HCT-8, MDBK, and IPEC. Animal experiments have shown that BF-1153 has no toxic effects on healthy SPF Kunming mice. Notably, the supernatant of BF-1153 enhanced cell activity and promoted cell growth in all three cell lines. At the same time, a cluster analysis of the isolated strains showed that the BF-1153 strain belonged to the same branch as the B. fragilis strain 23212, and B. fragilis strain 22998. The results of the animal experiments showed that BF-1153 had a certain preventive effect on diarrhea symptoms in SPF Kunming mice caused by a bovine rotavirus (BRV). In summary, the strain BF-1153 isolated in this experiment is NTBF, which has no toxic effect on MDBK, HCT-8, and IPEC, and has obvious cell growth-promoting effects, especially on MDBK. BF-1153 promotes the growth and development of SPF Kunming mice when compared with the control group. At the same time, BF-1153 alleviated the diarrhea symptoms caused by BRV in SPF Kunming mice. Therefore, BF-1153 has the potential to be a probiotic for cattle.
Collapse
Affiliation(s)
- Dong Wang
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (D.W.); (L.Z.); (J.L.); (Y.W.); (H.G.); (W.L.); (Q.L.); (L.Z.)
| | - Long Zhao
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (D.W.); (L.Z.); (J.L.); (Y.W.); (H.G.); (W.L.); (Q.L.); (L.Z.)
| | - Jingyi Lin
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (D.W.); (L.Z.); (J.L.); (Y.W.); (H.G.); (W.L.); (Q.L.); (L.Z.)
| | - Yajing Wang
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (D.W.); (L.Z.); (J.L.); (Y.W.); (H.G.); (W.L.); (Q.L.); (L.Z.)
| | - Haihui Gao
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (D.W.); (L.Z.); (J.L.); (Y.W.); (H.G.); (W.L.); (Q.L.); (L.Z.)
- Institute of Animal Science, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, China
| | - Wenhui Liu
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (D.W.); (L.Z.); (J.L.); (Y.W.); (H.G.); (W.L.); (Q.L.); (L.Z.)
| | - Qirui Li
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (D.W.); (L.Z.); (J.L.); (Y.W.); (H.G.); (W.L.); (Q.L.); (L.Z.)
| | - Liang Zhang
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (D.W.); (L.Z.); (J.L.); (Y.W.); (H.G.); (W.L.); (Q.L.); (L.Z.)
| | - Xiaodong Kang
- Institute of Animal Science, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, China
| | - Kangkang Guo
- College of Veterinary Medicine, Northwest A&F University, Xianyang 712100, China; (D.W.); (L.Z.); (J.L.); (Y.W.); (H.G.); (W.L.); (Q.L.); (L.Z.)
| |
Collapse
|
10
|
Shagaleeva OY, Kashatnikova DA, Kardonsky DA, Efimov BA, Ivanov VA, Smirnova SV, Evsiev SS, Zubkov EA, Abramova OV, Zorkina YA, Morozova AY, Vorobeva EA, Silantiev AS, Kolesnikova IV, Markelova MI, Olekhnovich EI, Morozov MD, Zoruk PY, Boldyreva DI, Kazakova VD, Vanyushkina AA, Chaplin AV, Grigoryeva TV, Zakharzhevskaya NB. Bacteroides vesicles promote functional alterations in the gut microbiota composition. Microbiol Spectr 2024; 12:e0063624. [PMID: 39345205 PMCID: PMC11537023 DOI: 10.1128/spectrum.00636-24] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2024] [Accepted: 08/09/2024] [Indexed: 10/01/2024] Open
Abstract
Inflammatory bowel diseases are characterized by chronic intestinal inflammation and alterations in the gut microbiota composition. Bacteroides fragilis, which secretes outer membrane vesicles (OMVs) with polysaccharide A (PSA), can moderate the inflammatory response and possibly alter the microbiota composition. In this study, we created a murine model of chronic sodium dextran sulfate (DSS)-induced intestinal colitis and treated it with B. fragilis OMVs. We monitored the efficiency of OMV therapy by determining the disease activity index (DAI) and performing histological examination (HE) of the intestine before and after vesicle exposure. We also analyzed the microbiota composition using 16S rRNA gene sequencing. Finally, we evaluated the volatile compound composition in the animals' stools by HS-GC/MS to assess the functional activity of the microbiota. We observed more effective intestinal repair after OMV treatment according to the DAI and HE. A metabolomic study also revealed changes in the functional activity of the microbiota, with a predominance of phenol and pentanoic acid in the control group compared to the group treated with DSS and the group treated with OMVs (DSS OMVs). We also observed a positive correlation of these metabolites with Saccharibacteria and Acetivibrio in the control group, whereas in the DSS group, there was a negative correlation of phenol and pentanoic acid with Lactococcus and Romboutsia. According to the metabolome and sequencing data, the microbiota composition of the DSS-treated OMV group was intermediate between that of the control and DSS groups. OMVs not only have an anti-inflammatory effect but also contribute to the recovery of the microbiota composition.IMPORTANCEBacteroides fragilis vesicles contain superficially localized polysaccharide A (PSA), which has unique immune-modulating properties. Isolated PSA can prevent chemically induced colitis in a murine model. Outer membrane vesicles (OMVs) also contain digestive enzymes and volatile metabolites that can complement the anti-inflammatory properties of PSA. OMVs showed high therapeutic activity against sodium dextran sulfate-induced colitis, as confirmed by histological assays. 16S rRNA sequencing of fecal samples from different inflammatory stages, supplemented with comprehensive metabolome analysis of volatile compounds conducted by HS-GC/MS, revealed structural and functional alterations in the microbiota composition under the influence of OMVs. Correlation analysis of the OMV-treated and untreated experimental animal groups revealed associations of phenol and pentanoic acid with Lactococcus, Romboutsia, Saccharibacteria, and Acetivibrio.
Collapse
Affiliation(s)
- Olga Yu. Shagaleeva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Daria A. Kashatnikova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry A. Kardonsky
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Boris A. Efimov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Department of Microbiology and Virology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Viktor A. Ivanov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Svetlana V. Smirnova
- The Laboratory of Ecological Genetics, Vavilov Institute of General Genetics, Russian Academy of Sciences, Moscow, Russia
| | - Suleiman S. Evsiev
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Eugene A. Zubkov
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Olga V. Abramova
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Yana A. Zorkina
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Anna Y. Morozova
- Department of Basic and Applied Neurobiology, V. P. Serbsky National Medical Research Center for Psychiatry and Narcology, Moscow, Russia
| | - Elizaveta A. Vorobeva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Artemiy S. Silantiev
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Irina V. Kolesnikova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Maria I. Markelova
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Evgenii I. Olekhnovich
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Maxim D. Morozov
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Polina Y. Zoruk
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Daria I. Boldyreva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Victoriia D. Kazakova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| | - Anna A. Vanyushkina
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow, Russia
| | - Andrei V. Chaplin
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
- Department of Microbiology and Virology, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Tatiana V. Grigoryeva
- Institute of Fundamental Medicine and Biology, Kazan (Volga Region) Federal University, Kazan, Russia
| | - Natalya B. Zakharzhevskaya
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine of Federal Medical Biological Agency, Moscow, Russia
| |
Collapse
|
11
|
Wang M, Ma Y, Yu G, Zeng B, Yang W, Huang C, Dong Y, Tang B, Wu Z. Integration of microbiome, metabolomics and transcriptome for in-depth understanding of berberine attenuates AOM/DSS-induced colitis-associated colorectal cancer. Biomed Pharmacother 2024; 179:117292. [PMID: 39151314 DOI: 10.1016/j.biopha.2024.117292] [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: 06/12/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/19/2024] Open
Abstract
A type of colorectal cancer (CRC),Colitis-associated colorectal cancer (CAC), is closely associated with chronic inflammation and gut microbiota dysbiosis. Berberine (BBR) has a long history in the treatment of intestinal diseases, which has been reported to inhibit colitis and CRC. However, the mechanism of its action is still unclear. Here, this study aimed to explore the potential protective effects of BBR on azoxymethane (AOM)/dextransulfate sodium (DSS)-induced colitis and tumor mice, and to elucidate its potential molecular mechanisms by microbiota, genes and metabolic alterations. The results showed that BBR inhibited the gut inflammation and improved the function of mucosal barrier to ameliorate AOM/DSS-induced colitis. And BBR treatment significantly reduced intestinal tumor development and ki-67 expression of intestinal tissue along with promoted apoptosis. Through microbiota analysis based on the 16 S rRNA gene, we found that BBR treatment improved intestinal microbiota imbalance in AOM/DSS-induced colitis and tumor mice, which were characterized by an increase of beneficial bacteria, for instance Akkermanisa, Lactobacillus, Bacteroides uniformis and Bacteroides acidifaciens. In addition, transcriptome analysis showed that BBR regulated colonic epithelial signaling pathway in CAC mice particularly by tryptophan metabolism and Wnt signaling pathway. Notably, BBR treatment resulted in the enrichment of amino acids metabolism and microbiota-derived SCFA metabolites. In summary, our research findings suggest that the gut microbiota-amino acid metabolism-Wnt signaling pathway axis plays critical role in maintaining intestinal homeostasis, which may provide new insights into the inhibitory effects of BBR on colitis and colon cancer.
Collapse
Affiliation(s)
- Mengxia Wang
- Dpartment of Medical Science, Shunde Polytechnic, Foshan, China; Academician Workstation,NingBo College of Health Sciences, NingBo, China
| | - Yan Ma
- Dpartment of Medical Science, Shunde Polytechnic, Foshan, China
| | - Guodong Yu
- Dpartment of Medical Science, Shunde Polytechnic, Foshan, China
| | - Bao Zeng
- Dpartment of Medical Science, Shunde Polytechnic, Foshan, China
| | - Wenhao Yang
- Dpartment of Medical Science, Shunde Polytechnic, Foshan, China
| | - Cuihong Huang
- Dpartment of Medical Science, Shunde Polytechnic, Foshan, China
| | - Yujuan Dong
- GuangDong Second Traditional Chinese Medicine Hospital, Guangzhou, China.
| | - Benqin Tang
- Dpartment of Medical Science, Shunde Polytechnic, Foshan, China.
| | - Zhengzhi Wu
- Academician Workstation,NingBo College of Health Sciences, NingBo, China; The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; Shenzhen Institute of Geriatrics, Shenzhen, China.
| |
Collapse
|
12
|
Sharma S, Gilberto VS, Levens CL, Chatterjee A, Kuhn KA, Nagpal P. Microbiome- and Host Inflammasome-Targeting Inhibitor Nanoligomers Are Therapeutic in the Murine Colitis Model. ACS Pharmacol Transl Sci 2024; 7:2677-2693. [PMID: 39296260 PMCID: PMC11406689 DOI: 10.1021/acsptsci.4c00102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 07/30/2024] [Accepted: 08/20/2024] [Indexed: 09/21/2024]
Abstract
Autoimmune and autoinflammatory diseases account for more than 80 chronic conditions affecting more than 24 million people in the US. Among these autoinflammatory diseases, noninfectious chronic inflammation of the gastrointestinal (GI) tract causes inflammatory bowel diseases (IBDs), primarily Crohn's and ulcerative colitis (UC). IBD is a complex disease, and one hypothesis is that these are either caused or worsened by compounds produced by bacteria in the gut. While traditional approaches have focused on pan immunosuppressive techniques (e.g., steroids), low remission rates, prolonged illnesses, and an increased frequency of surgical procedures have prompted the search for more targeted and precision therapeutic approaches. IBD is a complex disease resulting from both genetic and environmental factors, but several recent studies have highlighted the potential pivotal contribution of gut microbiota dysbiosis. Gut microbiota are known to modulate the immune status of the gut by producing metabolites that are encoded in biosynthetic gene clusters (BGCs) of the bacterial genome. Here, we show a targeted and high-throughput screening of more than 90 biosynthetic genes in 41 gut anaerobes, through downselection using available bioinformatics tools, targeted gene manipulation in these genetically intractable organisms using the Nanoligomer platform, and identification and synthesis of top microbiome targets as a Nanoligomer BGC cocktail (SB_BGC_CK1, abbreviated as CK1) as a feasible precision therapeutic approach. Further, we used a host-directed immune target screening to identify the NF-κB and NLRP3 cocktail SB_NI_112 (or NI112 for short) as a targeted inflammasome inhibitor. We used these top two microbe- and host-targeted Nanoligomer cocktails in acute and chronic dextran sulfate sodium (DSS) mouse colitis and in TNFΔARE/+ transgenic mice that develop spontaneous Crohn's like ileitis. The mouse microbiome was humanized to replicate that in human IBD through antibiotic treatment, followed by mixed fecal gavage from 10 human donors and spiked with IBD-inducing microbial species. Following colonization, colitis was induced in mice using 1 week of 3% DSS (acute) or 6 weeks of 3 rounds of 2.5% DSS induction for a week followed by 1 week of no DSS (chronic colitis model). Both Nanoligomer cocktails (CK1 and NI112) showed a strong reduction in disease severity, significant improvement in disease histopathology, and profound downregulation of disease biomarkers in colon tissue, as assessed by multiplexed ELISA. Further, we used two different formulations of intraperitoneal injections (IP) and Nanoligomer pills in the chronic DSS colitis model. Although both formulations were highly effective, the oral pill formulation demonstrated a greater reduction in biochemical markers compared to IP. A similar therapeutic effect was observed in the TNFΔARE/+ model. Overall, these results point to the potential for further development and testing of this inflammasome-targeting host-directed therapy (NI112) and more personalized microbiome cocktails (CK1) for patients with recalcitrant IBD.
Collapse
Affiliation(s)
- Sadhana Sharma
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 8002, United States
| | - Vincenzo S Gilberto
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 8002, United States
| | - Cassandra L Levens
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Anushree Chatterjee
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 8002, United States
| | - Kristine A Kuhn
- Division of Rheumatology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Prashant Nagpal
- Sachi Bio, Colorado Technology Center, 685 S Arthur Avenue, Louisville, Colorado 8002, United States
| |
Collapse
|
13
|
Huang Y, Cao J, Zhu M, Wang Z, Jin Z, Xiong Z. Nontoxigenic Bacteroides fragilis: A double-edged sword. Microbiol Res 2024; 286:127796. [PMID: 38870618 DOI: 10.1016/j.micres.2024.127796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 04/12/2024] [Accepted: 05/31/2024] [Indexed: 06/15/2024]
Abstract
The contribution of commensal microbes to human health and disease is unknown. Bacteroides fragilis (B. fragilis) is an opportunistic pathogen and a common colonizer of the human gut. Nontoxigenic B. fragilis (NTBF) and enterotoxigenic B. fragilis (ETBF) are two kinds of B. fragilis. NTBF has been shown to affect the host immune system and interact with gut microbes and pathogenic microbes. Previous studies indicated that certain strains of B. fragilis have the potential to serve as probiotics, based on their observed relationship with the immune system. However, several recent studies have shown detrimental effects on the host when beneficial gut bacteria are found in the digestive system or elsewhere. In some pathological conditions, NTBF may have adverse reactions. This paper presents a comprehensive analysis of NTBF ecology from the host-microbe perspective, encompassing molecular disease mechanisms analysis, bacteria-bacteria interaction, bacteria-host interaction, and the intricate ecological context of the gut. Our review provides much-needed insights into the precise application of NTBF.
Collapse
Affiliation(s)
- Yumei Huang
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiali Cao
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Mengpei Zhu
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ziwen Wang
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ze Jin
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Zhifan Xiong
- Department of Gastroenterology, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
14
|
Arnaiz-Villena A, Juarez I, Vaquero-Yuste C, Lledo T, Martin-Villa JM, Suarez-Trujillo F. Complex Interactions between the Human Major Histocompatibility Complex (MHC) and Microbiota: Their Roles in Disease Pathogenesis and Immune System Regulation. Biomedicines 2024; 12:1928. [PMID: 39200390 PMCID: PMC11352054 DOI: 10.3390/biomedicines12081928] [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: 07/04/2024] [Revised: 08/19/2024] [Accepted: 08/19/2024] [Indexed: 09/02/2024] Open
Abstract
The relationship between microbiota and the immune system is complex and characterized by the ways in which microbiota directs immune function interactions, both innate and acquired and also keeps activating the immune system throughout an individual's life. In this respect, the human Major Histocompatibility Complex (MHC, referred to as HLA in humans) plays a crucial role and is also established in self-defense against microbes by presenting microbial-derived peptides to the immune cells. However, this assumption has some unclear aspects that should be investigated. For example, how is the microbiota shaped by microbe species diversity, quantity and functions of the immune system, as well as the role and molecular mechanisms of the HLA complex during this process. There are autoimmune diseases related to both HLA and specific microbiota changes or alterations, many of which are mentioned in the present review. In addition, the HLA peptide presenting function should be put in a framework together with its linkage to diseases and also with HLA compatibility necessary for transplants to be successful. These are still quite an enigmatically statistical and phenomenological approach, but no firm pathogenic mechanisms have been described; thus, HLA's real functioning is still to be fully unveiled. After many years of HLA single-genes studies, firm pathogenesis mechanisms underlying disease linkage have been discovered. Finally, microbiota has been defined as conformed by bacteria, protozoa, archaea, fungi, and viruses; notwithstanding, endogenous viral sequences integrated into the human genome and other viral particles (obelisks) recently found in the digestive mucosa should be taken into account because they may influence both the microbiome and the immune system and their interactions. In this context, we propose to integrate these microbial-genetic particle components into the microbiome concept and designate it as "microgenobiota".
Collapse
Affiliation(s)
- Antonio Arnaiz-Villena
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| | - Ignacio Juarez
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| | - Christian Vaquero-Yuste
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| | - Tomás Lledo
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| | - José Manuel Martin-Villa
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| | - Fabio Suarez-Trujillo
- Department of Immunology, School of Medicine, Complutense University of Madrid, 28040 Madrid, Spain; (I.J.); (C.V.-Y.); (T.L.); (J.M.M.-V.); (F.S.-T.)
- Instituto de Investigacion Sanitaria Gegorio Marañon, 28009 Madrid, Spain
| |
Collapse
|
15
|
Mitchell C, Staley S, Williams MC, Saxena A, Bogdon R, Roark K, Hailey M, Miranda K, Becker W, Dopkins N, Pena MM, Hogan KM, Baird M, Wilson K, Nagarkatti P, Nagarkatti M, Busbee PB. Regulation of Bacteroides acidifaciens by the aryl hydrocarbon receptor in IL-22-producing immune cells has sex-dependent consequential impact on colitis. Front Immunol 2024; 15:1444045. [PMID: 39229279 PMCID: PMC11368719 DOI: 10.3389/fimmu.2024.1444045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024] Open
Abstract
Introduction Colitis is an inflammatory bowel disease (IBD) characterized by immune cell dysregulation and alterations in the gut microbiome. In our previous report, we showed a natural product in cruciferous vegetables and ligand of the aryl hydrocarbon receptor (AhR), indole-3-carbinol (I3C), was able to reduce colitis-induced disease severity and microbial dysbiosis in an interleukin-22 (IL-22) dependent manner. Methods In the current study, we performed single-cell RNA sequencing (scRNAseq) from colonocytes during colitis induction and supplementation with I3C and show how this treatment alters expression of genes involved in IL-22 signaling. To further define the role of IL-22 signaling in I3C-mediated protection during colitis and disease-associated microbial dysbiosis, we generated mice with AhR deficiency in RAR-related orphan receptor c (Rorc)-expressing cells (AhR ΔRorc ) which depletes this receptor in immune cells involved in production of IL-22. Colitis was induced in wildtype (WT), AhR ΔRorc , and littermate (LM) mice with or without I3C treatment. Results Results showed AhR ΔRorc mice lost the efficacy effects of I3C treatment which correlated with a loss of ability to increase IL-22 by innate lymphoid type 3 (ILC3s), not T helper 22 (Th22) cells. 16S rRNA microbiome profiling studies showed AhR ΔRorc mice were unable to regulate disease-associated increases in Bacteroides, which differed between males and females. Lastly, inoculation with a specific disease-associated Bacteroides species, Bacteroides acidifaciens (B. acidifaciens), was shown to exacerbate colitis in females, but not males. Discussion Collectively, this report highlights the cell and sex-specific role of AhR in regulating microbes that can impact colitis disease.
Collapse
Affiliation(s)
- Chandani Mitchell
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Shanieka Staley
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Michal Claire Williams
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Archana Saxena
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Raymond Bogdon
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Kasie Roark
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Michele Hailey
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Kathryn Miranda
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - William Becker
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Nicholas Dopkins
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Maria Marjorette Pena
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC, United States
| | - Kristen M. Hogan
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC, United States
| | - Maredith Baird
- Department of Biological Sciences, College of Arts and Sciences, University of South Carolina, Columbia, SC, United States
| | - Kiesha Wilson
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Prakash Nagarkatti
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Mitzi Nagarkatti
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| | - Philip Brandon Busbee
- Department of Pathology, Microbiology, and Immunology, School of Medicine, University of South Carolina, Columbia, SC, United States
| |
Collapse
|
16
|
Leung HKM, Lo EKK, Zhang F, Felicianna, Ismaiah MJ, Chen C, El-Nezami H. Modulation of Gut Microbial Biomarkers and Metabolites in Cancer Management by Tea Compounds. Int J Mol Sci 2024; 25:6348. [PMID: 38928054 PMCID: PMC11203446 DOI: 10.3390/ijms25126348] [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: 04/29/2024] [Revised: 05/24/2024] [Accepted: 05/27/2024] [Indexed: 06/28/2024] Open
Abstract
Cancers are causing millions of deaths and leaving a huge clinical and economic burden. High costs of cancer drugs are limiting their access to the growing number of cancer cases. The development of more affordable alternative therapy could reach more patients. As gut microbiota plays a significant role in the development and treatment of cancer, microbiome-targeted therapy has gained more attention in recent years. Dietary and natural compounds can modulate gut microbiota composition while providing broader and more accessible access to medicine. Tea compounds have been shown to have anti-cancer properties as well as modulate the gut microbiota and their related metabolites. However, there is no comprehensive review that focuses on the gut modulatory effects of tea compounds and their impact on reshaping the metabolic profiles, particularly in cancer models. In this review, the effects of different tea compounds on gut microbiota in cancer settings are discussed. Furthermore, the relationship between these modulated bacteria and their related metabolites, along with the mechanisms of how these changes led to cancer intervention are summarized.
Collapse
Affiliation(s)
- Hoi Kit Matthew Leung
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Emily Kwun Kwan Lo
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Fangfei Zhang
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Felicianna
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Marsena Jasiel Ismaiah
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Congjia Chen
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
| | - Hani El-Nezami
- School of Biological Sciences, University of Hong Kong, Pokfulam, Hong Kong SAR 999077, China; (H.K.M.L.); (E.K.K.L.); (F.Z.); (F.); (M.J.I.); (C.C.)
- Institute of Public Health and Clinical Nutrition, School of Medicine, University of Eastern Finland, FI-70211 Kuopio, Finland
| |
Collapse
|
17
|
Keener JE, Goh B, Yoo JS, Oh SF, Brodbelt JS. Top-Down Characterization of Bacterial Lipopolysaccharides and Lipooligosaccharides Using Activated-Electron Photodetachment Mass Spectrometry. Anal Chem 2024; 96:9151-9158. [PMID: 38758019 PMCID: PMC11384421 DOI: 10.1021/acs.analchem.4c00952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2024]
Abstract
Lipopolysaccharides (LPS) and lipooligosaccharides (LOS) are located in the outer membrane of Gram-negative bacteria and are comprised of three distinctive parts: lipid A, core oligosaccharide (OS), and O-antigen. The structure of each region influences bacterial stability, toxicity, and pathogenesis. Here, we highlight the use of targeted activated-electron photodetachment (a-EPD) tandem mass spectrometry to characterize LPS and LOS from two crucial players in the human gut microbiota, Escherichia coli Nissle and Bacteroides fragilis. a-EPD is a hybrid activation method that uses ultraviolet photoirradiation to generate charge-reduced radical ions followed by collisional activation to produce informative fragmentation patterns. We benchmark the a-EPD method for top-down characterization of triacyl LOS from E. coli R2, then focus on characterization of LPS from E. coli Nissle and B. fragilis. Notably, a-EPD affords extensive fragmentation throughout the backbone of the core OS and O-antigen regions of LPS from E. coli Nissle. This hybrid approach facilitated the elucidation of structural details for LPS from B. fragilis, revealing a putative hexuronic acid (HexA) conjugated to lipid A.
Collapse
Affiliation(s)
- James E Keener
- Department of Chemistry, University of Texas, Austin, Texas 78712, United States
| | - Byoungsook Goh
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
| | - Ji-Sun Yoo
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
| | - Sungwhan F Oh
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women's Hospital, Boston, Massachusetts 02115, United States
- Program in Immunology, Division of Medical Sciences, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jennifer S Brodbelt
- Department of Chemistry, University of Texas, Austin, Texas 78712, United States
| |
Collapse
|
18
|
Giampazolias E, da Costa MP, Lam KC, Lim KHJ, Cardoso A, Piot C, Chakravarty P, Blasche S, Patel S, Biram A, Castro-Dopico T, Buck MD, Rodrigues RR, Poulsen GJ, Palma-Duran SA, Rogers NC, Koufaki MA, Minutti CM, Wang P, Vdovin A, Frederico B, Childs E, Lee S, Simpson B, Iseppon A, Omenetti S, Kelly G, Goldstone R, Nye E, Suárez-Bonnet A, Priestnall SL, MacRae JI, Zelenay S, Patil KR, Litchfield K, Lee JC, Jess T, Goldszmid RS, Sousa CRE. Vitamin D regulates microbiome-dependent cancer immunity. Science 2024; 384:428-437. [PMID: 38662827 PMCID: PMC7615937 DOI: 10.1126/science.adh7954] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 03/04/2024] [Indexed: 05/03/2024]
Abstract
A role for vitamin D in immune modulation and in cancer has been suggested. In this work, we report that mice with increased availability of vitamin D display greater immune-dependent resistance to transplantable cancers and augmented responses to checkpoint blockade immunotherapies. Similarly, in humans, vitamin D-induced genes correlate with improved responses to immune checkpoint inhibitor treatment as well as with immunity to cancer and increased overall survival. In mice, resistance is attributable to the activity of vitamin D on intestinal epithelial cells, which alters microbiome composition in favor of Bacteroides fragilis, which positively regulates cancer immunity. Our findings indicate a previously unappreciated connection between vitamin D, microbial commensal communities, and immune responses to cancer. Collectively, they highlight vitamin D levels as a potential determinant of cancer immunity and immunotherapy success.
Collapse
Affiliation(s)
- Evangelos Giampazolias
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Cancer Immunosurveillance Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | | | - Khiem C. Lam
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology (LICI), Center for Cancer Research (CCR), National Cancer Institute (NCI), 37 Convent Drive, Bethesda, MD 20892-0001, USA
| | - Kok Haw Jonathan Lim
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Immunology and Inflammation, Imperial College, London, UK
| | - Ana Cardoso
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Cécile Piot
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Probir Chakravarty
- Bioinformatics and Biostatistics STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sonja Blasche
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Swara Patel
- Cancer Immunosurveillance Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Adi Biram
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Tomas Castro-Dopico
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Michael D. Buck
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Richard R. Rodrigues
- Basic Science Program, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
- Microbiome and Genetics Core, LICI, CCR, NCI, 37 Convent Drive, Bethesda, MD 20892-0001, USA
| | - Gry Juul Poulsen
- National Center of Excellence for Molecular Prediction of Inflammatory Bowel Disease, PREDICT, Faculty of Medicine, Aalborg University, Department of Gastroenterology and Hepatology, Aalborg University Hospital, A.C. Meyers Vænge 15, A DK-2450 Copenhagen, Denmark
| | | | - Neil C. Rogers
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Maria A. Koufaki
- Cancer Inflammation and Immunity Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Carlos M. Minutti
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Pengbo Wang
- Cancer Immunosurveillance Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Alexander Vdovin
- Cancer Immunosurveillance Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Bruno Frederico
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Eleanor Childs
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sonia Lee
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Ben Simpson
- Tumor ImmunoGenomics and Immunosurveillance (TIGI) Lab, UCL Cancer Institute, 72 Huntley St, London WC1E 6DD, UK
| | - Andrea Iseppon
- AhRimmunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Sara Omenetti
- AhRimmunity Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Gavin Kelly
- Bioinformatics and Biostatistics STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Robert Goldstone
- Bioinformatics and Biostatistics STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Emma Nye
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Alejandro Suárez-Bonnet
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK
| | - Simon L. Priestnall
- Experimental Histopathology, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Department of Pathobiology and Population Sciences, The Royal Veterinary College, Hawkshead Lane, North Mymms, Hatfield, Hertfordshire AL9 7TA, UK
| | - James I. MacRae
- Metabolomics STP, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Santiago Zelenay
- Cancer Inflammation and Immunity Group, Cancer Research UK Manchester Institute, The University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - Kiran Raosaheb Patil
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Kevin Litchfield
- Tumor ImmunoGenomics and Immunosurveillance (TIGI) Lab, UCL Cancer Institute, 72 Huntley St, London WC1E 6DD, UK
| | - James C. Lee
- Genetic Mechanisms of Disease Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
- Institute of Liver and Digestive Health, Division of Medicine, Royal Free Hospital, University College London, London, NW3 2QG, UK
| | - Tine Jess
- National Center of Excellence for Molecular Prediction of Inflammatory Bowel Disease, PREDICT, Faculty of Medicine, Aalborg University, Department of Gastroenterology and Hepatology, Aalborg University Hospital, A.C. Meyers Vænge 15, A DK-2450 Copenhagen, Denmark
| | - Romina S. Goldszmid
- Inflammatory Cell Dynamics Section, Laboratory of Integrative Cancer Immunology (LICI), Center for Cancer Research (CCR), National Cancer Institute (NCI), 37 Convent Drive, Bethesda, MD 20892-0001, USA
| | - Caetano Reis e Sousa
- Immunobiology Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| |
Collapse
|
19
|
Ali A, Wu L, Ali SS. Gut microbiota and acute kidney injury: immunological crosstalk link. Int Urol Nephrol 2024; 56:1345-1358. [PMID: 37749436 DOI: 10.1007/s11255-023-03760-5] [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: 04/14/2023] [Accepted: 08/14/2023] [Indexed: 09/27/2023]
Abstract
The gut microbiota, often called the "forgotten organ," plays a crucial role in bidirectional communication with the host for optimal physiological function. This communication helps regulate the host's immunity and metabolism positively and negatively. Many factors influence microbiota homeostasis and subsequently lead to an immune system imbalance. The correlation between an unbalanced immune system and acute diseases such as acute kidney injury is not fully understood, and the role of gut microbiota in disease pathogenesis is still yet uncovered. This review summarizes our understanding of gut microbiota, focusing on the interactions between the host's immune system and the microbiome and their impact on acute kidney injury.
Collapse
Affiliation(s)
- Asmaa Ali
- Department of Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, China.
- Department of Pulmonary Medicine, Abbassia Chest Hospital, MOH, Cairo, Egypt.
- Department of Respiratory Allergy, A Al-Rashed Allergy Center, Ministry of Health, Kuwait, Kuwait.
| | - Liang Wu
- Yizheng Hospital, Nanjing Drum Tower Hospital Group, Yizheng, 210008, China.
| | - Sameh Samir Ali
- School of the Environment and Safety Engineering, Biofuels Institute, Jiangsu University, Zhenjiang, 212013, China
- Botany Department, Faculty of Science, Tanta University, Tanta, 31527, Egypt
| |
Collapse
|
20
|
Rajesh KM, Kinra M, Ranadive N, Pawaskar GM, Mudgal J, Raval R. Effect of chronic low-dose treatment with chitooligosaccharides on microbial dysbiosis and inflammation associated chronic ulcerative colitis in Balb/c mice. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:1611-1622. [PMID: 37695333 PMCID: PMC10858833 DOI: 10.1007/s00210-023-02710-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
The study aimed to investigate the potential of low dose chitooligosaccharide (COS) in ameliorating dextran sodium sulfate (DSS) induced chronic colitis by regulating microbial dysbiosis and pro-inflammatory responses. Chronic colitis was induced in BALB/c mice by DSS (4% w/v, 3 cycles of 5 days) administration. The mice were divided into four groups: vehicle, DSS, DSS + mesalamine and DSS+COS. COS and mesalamine were administered orally, daily once, from day 1 to day 30 at a dose of 20 mg/kg and 50 mg/kg respectively. The disease activity index (DAI), colon length, histopathological score, microbial composition, and pro-inflammatory cytokine expression were evaluated. COS (20 mg/kg, COSLow) administration reduced the disease activity index, and colon shortening, caused by DSS significantly. Furthermore, COSLow restored the altered microbiome in the gut and inhibited the elevated pro-inflammatory cytokines (IL-1 and IL-6) in the colon against DSS-induced chronic colitis in mice. Moreover, COSLow treatment improved the probiotic microflora thereby restoring the gut homeostasis. In conclusion, this is the first study where microbial dysbiosis and pro-inflammatory responses were modulated by chronic COSLow treatment against DSS-induced chronic colitis in Balb/c mice. Therefore, COS supplementation at a relatively low dose could be efficacious for chronic inflammatory bowel disease.
Collapse
Affiliation(s)
- K M Rajesh
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Manas Kinra
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Niraja Ranadive
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Goutam Mohan Pawaskar
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Jayesh Mudgal
- Department of Pharmacology, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Ritu Raval
- Department of Biotechnology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| |
Collapse
|
21
|
Fogarty EC, Schechter MS, Lolans K, Sheahan ML, Veseli I, Moore RM, Kiefl E, Moody T, Rice PA, Yu MK, Mimee M, Chang EB, Ruscheweyh HJ, Sunagawa S, Mclellan SL, Willis AD, Comstock LE, Eren AM. A cryptic plasmid is among the most numerous genetic elements in the human gut. Cell 2024; 187:1206-1222.e16. [PMID: 38428395 PMCID: PMC10973873 DOI: 10.1016/j.cell.2024.01.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 10/03/2023] [Accepted: 01/25/2024] [Indexed: 03/03/2024]
Abstract
Plasmids are extrachromosomal genetic elements that often encode fitness-enhancing features. However, many bacteria carry "cryptic" plasmids that do not confer clear beneficial functions. We identified one such cryptic plasmid, pBI143, which is ubiquitous across industrialized gut microbiomes and is 14 times as numerous as crAssphage, currently established as the most abundant extrachromosomal genetic element in the human gut. The majority of mutations in pBI143 accumulate in specific positions across thousands of metagenomes, indicating strong purifying selection. pBI143 is monoclonal in most individuals, likely due to the priority effect of the version first acquired, often from one's mother. pBI143 can transfer between Bacteroidales, and although it does not appear to impact bacterial host fitness in vivo, it can transiently acquire additional genetic content. We identified important practical applications of pBI143, including its use in identifying human fecal contamination and its potential as an alternative approach to track human colonic inflammatory states.
Collapse
Affiliation(s)
- Emily C Fogarty
- Committee on Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA; Department of Medicine, University of Chicago, Chicago, IL 60637, USA.
| | - Matthew S Schechter
- Committee on Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA; Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Karen Lolans
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Madeline L Sheahan
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA; Department of Microbiology, University of Chicago, Chicago, IL 60637, USA
| | - Iva Veseli
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Ryan M Moore
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, USA
| | - Evan Kiefl
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Thomas Moody
- Department of Systems Biology, Columbia University, New York, NY 10032, USA
| | - Phoebe A Rice
- Committee on Microbiology, University of Chicago, Chicago, IL 60637, USA; Department of Biochemistry, University of Chicago, Chicago, IL 60637, USA
| | - Michael K Yu
- Toyota Technological Institute at Chicago, Chicago, IL 60637, USA
| | - Mark Mimee
- Committee on Microbiology, University of Chicago, Chicago, IL 60637, USA; Department of Microbiology, University of Chicago, Chicago, IL 60637, USA; Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Eugene B Chang
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Hans-Joachim Ruscheweyh
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zurich, Zurich 8093, Switzerland
| | - Shinichi Sunagawa
- Department of Biology, Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zurich, Zurich 8093, Switzerland
| | - Sandra L Mclellan
- School of Freshwater Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI 53204, USA
| | - Amy D Willis
- Department of Biostatistics, University of Washington, Seattle, WA 98195, USA
| | - Laurie E Comstock
- Committee on Microbiology, University of Chicago, Chicago, IL 60637, USA; Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA; Department of Microbiology, University of Chicago, Chicago, IL 60637, USA.
| | - A Murat Eren
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA; Marine Biological Laboratory, Woods Hole, MA 02543, USA; Alfred Wegener Institute Helmholtz Centre for Polar and Marine Research, 27570 Bremerhaven, Germany; Institute for Chemistry and Biology of the Marine Environment, University of Oldenburg, 26129 Oldenburg, Germany; Max Planck Institute for Marine Microbiology, 28359 Bremen, Germany; Helmholtz Institute for Functional Marine Biodiversity, 26129 Oldenburg, Germany.
| |
Collapse
|
22
|
Lin CL, Ying TH, Yang SF, Lin CL, Chiou HL, Hsieh YH. Magnolin targeting of the JNK/Sp1/MMP15 signaling axis suppresses cervical cancer microenvironment and metastasis via microbiota modulation. Cancer Lett 2024; 583:216584. [PMID: 38123014 DOI: 10.1016/j.canlet.2023.216584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 12/23/2023]
Abstract
Magnolin (MGL), a compound derived from the magnolia plant, has inhibitory effects on tumor cell invasion and growth. His study aims to explore the antitumor effect and underlying molecular mechanism of MGL against human cervical cancer. We found that MGL inhibited the proliferation, migration, and invasiveness of cervical cancer cells in vitro and in vivo. The underlying mechanism was shown to involve MGL-induced inhibition of JNK/Sp1-mediated MMP15 transcription and translation. Overexpression of JNK/Sp1 resulted in significant restoration of MMP15 expression and the migration and invasion capabilities of MGL-treated cervical cancer cells. MGL modulated the cervical cancer microenvironment by inhibiting cell metastasis via targeting IL-10/IL-10 receptor B (IL-10RB) expression, thereby attenuating JNK/Sp1-mediated MMP15 expression. Analysis of the gut microbiota of mice fed MGL revealed a significant augmentation in Lachnospiraceae bacteria, known for their production of sodium butyrate. In vivo experiments also demonstrated synergistic inhibition of cervical cancer cell metastasis by MGL and sodium butyrate co-administration. Our study provides pioneering evidence of a novel mechanism by which MGL inhibits tumor growth and metastasis through the IL-10/IL-10RB targeting of the JNK/Sp1/MMP15 axis in human cervical cancer cells.
Collapse
Affiliation(s)
- Chia-Liang Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Biochemistry, School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Tsung-Ho Ying
- Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chu-Liang Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan; Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
23
|
Yang J, Wang X, Hu T, Huang H, Chen G, Jin B, Zeng G, Liu J. Entero-toxigenic Bacteroides fragilis contributes to intestinal barrier injury and colorectal cancer progression by mediating the BFT/STAT3/ZEB2 pathway. Cell Cycle 2024; 23:70-82. [PMID: 38273425 PMCID: PMC11005799 DOI: 10.1080/15384101.2024.2309005] [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: 06/27/2023] [Accepted: 12/21/2023] [Indexed: 01/27/2024] Open
Abstract
Our previous findings confirmed the high enrichment of Bacteroides fragilis (BF) in fecal samples from patients with colorectal cancer (CRC). The intestinal mucosal barrier is the first defense of the organism against commensal flora and intestinal pathogens and is closely associated with the occurrence and development of CRC. Therefore, this study aimed to investigate the molecular mechanisms through which BF mediates intestinal barrier injury and CRC progression. SW480 cells and a Caco2 intestinal barrier model were treated with entero-toxigenic BF (ETBF), its enterotoxin (B. fragilis toxin, BFT), and non-toxigenic BF (NTBF). Cell counting kit-8, flow cytometry, wound healing and transwell assays were performed to analyze the proliferation, apoptosis, migration, and invasion of SW480 cells. Transmission electron microscopy, FITC-dextran, and transepithelial electrical resistance (TEER) were used to analyze damage in the Caco2 intestinal barrier model. The Azoxymethane/Dextran Sulfate Sodium (AOM/DSS) animal model was established to evaluate the effect of ETBF on intestinal barrier injury and CRC progression in vivo. ETBF and BFT enhanced the viability, wound healing ratio, invasion, and EMT of SW480 cells. In addition, ETBF and BFT disrupted the tight junctions and villus structure in the intestinal barrier model, resulting in increased permeability and reduced TEER. Similarly, the expression of intestinal barrier-related proteins (MUC2, Occludin and Zo-1) was restricted by ETBF and BFT. Interestingly, the STAT3/ZEB2 axis was activated by ETBF and BFT, and treatment with Brevilin A (a STAT3 inhibitor) or knockdown of ZEB2 limited the promotional effect of ETBF and BFT on the SW480 malignant phenotype. In vivo experiments also confirmed that ETBF colonization accelerated tumor load, carcinogenesis, and intestinal mucosal barrier damage in the colorectum of the AOM/DSS animal model, and that treatment with Brevilin A alleviated these processes. ETBF-secreted BFT accelerated intestinal barrier damage and CRC by activating the STAT3/ZEB2 axis. Our findings provide new insights and perspectives for the application of ETBF in CRC treatment.
Collapse
Affiliation(s)
- Jian Yang
- Department of General Surgery, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
- The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Chengdu Fifth People’s Hospital, Cancer Prevention and Treatment Institute of Chengdu, Chengdu, Sichuan, China
| | - Xue Wang
- Department of General Surgery, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
- The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Chengdu Fifth People’s Hospital, Cancer Prevention and Treatment Institute of Chengdu, Chengdu, Sichuan, China
| | - Tao Hu
- Department of General Surgery, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
- The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Chengdu Fifth People’s Hospital, Cancer Prevention and Treatment Institute of Chengdu, Chengdu, Sichuan, China
| | - He Huang
- Department of General Surgery, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
- The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Chengdu Fifth People’s Hospital, Cancer Prevention and Treatment Institute of Chengdu, Chengdu, Sichuan, China
| | - Gang Chen
- Department of General Surgery, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
- The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Chengdu Fifth People’s Hospital, Cancer Prevention and Treatment Institute of Chengdu, Chengdu, Sichuan, China
| | - Bo Jin
- Department of General Surgery, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
- The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Chengdu Fifth People’s Hospital, Cancer Prevention and Treatment Institute of Chengdu, Chengdu, Sichuan, China
| | - Guilin Zeng
- The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Chengdu Fifth People’s Hospital, Cancer Prevention and Treatment Institute of Chengdu, Chengdu, Sichuan, China
- Department of Medical Oncology, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
| | - Jian Liu
- Department of General Surgery, Chengdu Fifth People’s Hospital, Chengdu, Sichuan, China
- The Second Clinical Medical College, Affiliated Fifth People’s Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- Chengdu Fifth People’s Hospital, Cancer Prevention and Treatment Institute of Chengdu, Chengdu, Sichuan, China
| |
Collapse
|
24
|
Wu L, Park SH, Kim H. Direct and Indirect Evidence of Effects of Bacteroides spp. on Obesity and Inflammation. Int J Mol Sci 2023; 25:438. [PMID: 38203609 PMCID: PMC10778732 DOI: 10.3390/ijms25010438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 12/22/2023] [Accepted: 12/23/2023] [Indexed: 01/12/2024] Open
Abstract
Metabolic disorders present a significant public health challenge globally. The intricate relationship between the gut microbiome, particularly Bacteroides spp. (BAC), and obesity, including their specific metabolic functions, remains partly unresolved. This review consolidates current research on BAC's role in obesity and lipid metabolism, with three objectives: (1) To summarize the gut microbiota's impact on obesity; (2) To assess BAC's efficacy in obesity intervention; (3) To explore BAC's mechanisms in obesity and lipid metabolism management. This review critically examines the role of BAC in obesity, integrating findings from clinical and preclinical studies. We highlight the changes in BAC diversity and concentration following successful obesity treatment and discuss the notable differences in BAC characteristics among individuals with varying obesity levels. Furthermore, we review recent preclinical studies demonstrating the potential of BAC in ameliorating obesity and related inflammatory conditions, providing detailed insights into the methodologies of these in vivo experiments. Additionally, certain BAC-derived metabolites have been shown to be involved in the regulation of host lipid metabolism-related pathways. The enhanced TNF production by dendritic cells following BAC administration, in response to LPS, also positions BAC as a potential adjunctive therapy in obesity management.
Collapse
Affiliation(s)
- Liangliang Wu
- Department of Rehabilitation Medicine of Korean Medicine, Ilsan Hospital of Dongguk University, Goyang 10326, Republic of Korea;
| | - Seo-Hyun Park
- Department of Rehabilitation Medicine of Korean Medicine, Bundang Hospital of Dongguk University, Seongnam 13601, Republic of Korea;
| | - Hojun Kim
- Department of Rehabilitation Medicine of Korean Medicine, Bundang Hospital of Dongguk University, Seongnam 13601, Republic of Korea;
| |
Collapse
|
25
|
Jo M, Hwang S, Lee CG, Hong JE, Kang DH, Yoo SH, Kim WS, Yoo JY, Rhee KJ. Promotion of Colitis in B Cell-Deficient C57BL/6 Mice Infected with Enterotoxigenic Bacteroides fragilis. Int J Mol Sci 2023; 25:364. [PMID: 38203534 PMCID: PMC10778593 DOI: 10.3390/ijms25010364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 12/22/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Enterotoxigenic Bacteroides fragilis (ETBF) causes colitis and is implicated in inflammatory bowel diseases and colorectal cancer. The ETBF-secreted B. fragilis toxin (BFT) causes cleavage of the adherence junction, the E-cadherin, resulting in the large intestine showing IL-17A inflammation in wild-type (WT) mice. However, intestinal pathology by ETBF infection is not fully understood in B-cell-deficient mice. In this study, ETBF-mediated inflammation was characterized in B-cell-deficient mice (muMT). WT or muMT C57BL/6J mice were orally inoculated with ETBF and examined for intestinal inflammation. The indirect indicators for colitis (loss of body weight and cecum weight, as well as mortality) were increased in muMT mice compared to WT mice. Histopathology and inflammatory genes (Nos2, Il-1β, Tnf-α, and Cxcl1) were elevated and persisted in the large intestine of muMT mice compared with WT mice during chronic ETBF infection. However, intestinal IL-17A expression was comparable between WT and muMT mice during infection. Consistently, flow cytometry analysis applied to the mesenteric lymph nodes showed a similar Th17 immune response in both WT and muMT mice. Despite elevated ETBF colonization, the ETBF-infected muMT mice showed no histopathology or inflammation in the small intestine. In conclusion, B cells play a protective role in ETBF-induced colitis, and IL-17A inflammation is not attributed to prompted colitis in B-cell-deficient mice. Our data support the fact that B cells are required to ameliorate ETBF infection-induced colitis in the host.
Collapse
Affiliation(s)
- Minjeong Jo
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
- Department of Molecular Cell Biology, Sungkyunkwan University School of Medicine, Suwon 16419, Republic of Korea
| | - Soonjae Hwang
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
- Department of Biochemistry, Lee Gil Ya Cancer and Diabetes Institute, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Chang-Gun Lee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Ju-Eun Hong
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Da-Hye Kang
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Sang-Hyeon Yoo
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Woo-Seung Kim
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Jung-Yoon Yoo
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| | - Ki-Jong Rhee
- Department of Biomedical Laboratory Science, College of Software and Digital Healthcare Convergence, Yonsei University Mirae Campus, Wonju 26493, Republic of Korea; (M.J.); (S.H.); (C.-G.L.); (J.-E.H.); (D.-H.K.); (S.-H.Y.); (W.-S.K.); (J.-Y.Y.)
| |
Collapse
|
26
|
Yang S, Li X, Cang W, Mu D, Ji S, An Y, Wu R, Wu J. Biofilm tolerance, resistance and infections increasing threat of public health. MICROBIAL CELL (GRAZ, AUSTRIA) 2023; 10:233-247. [PMID: 37933277 PMCID: PMC10625689 DOI: 10.15698/mic2023.11.807] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/28/2023] [Accepted: 09/14/2023] [Indexed: 11/08/2023]
Abstract
Microbial biofilms can cause chronic infection. In the clinical setting, the biofilm-related infections usually persist and reoccur; the main reason is the increased antibiotic resistance of biofilms. Traditional antibiotic therapy is not effective and might increase the threat of antibiotic resistance to public health. Therefore, it is urgent to study the tolerance and resistance mechanism of biofilms to antibiotics and find effective therapies for biofilm-related infections. The tolerance mechanism and host reaction of biofilm to antibiotics are reviewed, and bacterial biofilm related diseases formed by human pathogens are discussed thoroughly. The review also explored the role of biofilms in the development of bacterial resistance mechanisms and proposed therapeutic intervention strategies for biofilm related diseases.
Collapse
Affiliation(s)
- Shanshan Yang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Xinfei Li
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
| | - Weihe Cang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
| | - Delun Mu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Shuaiqi Ji
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Yuejia An
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| | - Junrui Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang 110866, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang 110866, P.R. China
| |
Collapse
|
27
|
Parizadeh M, Arrieta MC. The global human gut microbiome: genes, lifestyles, and diet. Trends Mol Med 2023; 29:789-801. [PMID: 37516570 DOI: 10.1016/j.molmed.2023.07.002] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 07/03/2023] [Accepted: 07/07/2023] [Indexed: 07/31/2023]
Abstract
A growing number of human gut microbiome studies consistently describe differences between human populations. Here, we review how factors related to host genetics, ethnicity, lifestyle, and geographic location help explain this variation. Studies from contrasting environmental scenarios point to diet and lifestyle as the most influential. The effect of human migration and displacement demonstrates how the microbiome adapts to newly adopted lifestyles and contributes to the profound biological and health consequences attributed to migration. This information strongly suggests against a universal scale for healthy or dysbiotic gut microbiomes, and prompts for additional microbiome population surveys, particularly from less industrialized nations. Considering these important differences will be critical for designing strategies to diagnose and restore dysbiosis in various human populations.
Collapse
Affiliation(s)
- Mona Parizadeh
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada; Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada; International Microbiome Center, University of Calgary, Calgary, Alberta, Canada
| | - Marie-Claire Arrieta
- Department of Physiology and Pharmacology, University of Calgary, Calgary, Alberta, Canada; Department of Pediatrics, University of Calgary, Calgary, Alberta, Canada; International Microbiome Center, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
28
|
Pant A, Das B, Arimbasseri GA. Host microbiome in tuberculosis: disease, treatment, and immunity perspectives. Front Microbiol 2023; 14:1236348. [PMID: 37808315 PMCID: PMC10559974 DOI: 10.3389/fmicb.2023.1236348] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/29/2023] [Indexed: 10/10/2023] Open
Abstract
Tuberculosis (TB), an airborne pulmonary disease caused by Mycobacterium tuberculosis (M. tb), poses an unprecedented health and economic burden to most of the developing countries. Treatment of TB requires prolonged use of a cocktail of antibiotics, which often manifest several side effects, including stomach upset, nausea, and loss of appetite spurring on treatment non-compliance and the emergence of antibiotic resistant M. tb. The anti-TB treatment regimen causes imbalances in the composition of autochthonous microbiota associated with the human body, which also contributes to major side effects. The microbiota residing in the gastrointestinal tract play an important role in various physiological processes, including resistance against colonization by pathogens, boosting host immunity, and providing key metabolic functions. In TB patients, due to prolonged exposure to anti-tuberculosis drugs, the gut microbiota significantly loses its diversity and several keystone bacterial taxa. This loss may result in a significant reduction in the functional potency of the microbiota, which is a probable reason for poor treatment outcomes. In this review, we discuss the structural and functional changes of the gut microbiota during TB and its treatment. A major focus of the review is oriented to the gut microbial association with micronutrient profiles and immune cell dynamics during TB infection. Furthermore, we summarize the acquisition of anti-microbial resistance in M. tb along with the microbiome-based therapeutics to cure the infections. Understanding the relationship between these components and host susceptibility to TB disease is important to finding potential targets that may be used in TB prevention, progression, and cure.
Collapse
Affiliation(s)
- Archana Pant
- Molecular Genetics Lab, National Institute of Immunology, New Delhi, India
| | - Bhabatosh Das
- Functional Genomics Laboratory, Infection and Immunology Division, Translational Health Science and Technology Institute, Faridabad, India
| | | |
Collapse
|
29
|
He Q, Niu M, Bi J, Du N, Liu S, Yang K, Li H, Yao J, Du Y, Duan Y. Protective effects of a new generation of probiotic Bacteroides fragilis against colitis in vivo and in vitro. Sci Rep 2023; 13:15842. [PMID: 37740010 PMCID: PMC10517118 DOI: 10.1038/s41598-023-42481-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 09/11/2023] [Indexed: 09/24/2023] Open
Abstract
Bacteroides fragilis, one of the potential next-generation probiotics, but its protective mechanism is not yet known. We aimed to characterize the anti-inflammatory effect of B. fragilisATCC25285 and to elucidate its mechanism through in vivo and in vitro experiments. An in vitro model of inflammation by induction of colonic cells with TNF-a, and co-cultured with B. fragilis to detect cell viability, apoptosis and invasive capacity. Furthermore, critical proteins of the TLR/NF-κB pathway and the inflammatory cytokines were measured. For animal trials, C57BL/6 J male mice were orally administered B. fragilis or PBS once daily for 21 days. Colitis was induced by drinking 2.5% DSS from days 0 to 7. The mice were weighed daily and rectal bleeding, stool condition and blood in the stool were recorded. We found that B. fragilis treatment alone was harmless and had no effect on cell viability or apoptosis. While predictably TNF-α decreased cell viability and increased apoptosis, B. fragilis attenuated this deterioration. The NF-κB pathway and inflammatory cytokines IL-6 and IL-1β activated by TNF-α were also blocked by B. fragilis. Notably, the metabolic supernatant of B. fragilis also has an anti-inflammatory effect. Animal studies showed that live B. fragilis rather than dead strain ameliorated DSS-induced colitis, as evidenced by weight loss, shortened colon length and enhanced barrier function. The colonic tissue levels of inflammatory cytokines (TNF-α, IL-1β, IL-6) were decreased and IL-10 was increased as a result of B. fragilis administration. In conclusion, B. fragilis ATCC25285 exhibited anti-inflammatory effects whether in vivo or in vitro, and it may be a potential probiotic agent for improving colitis.
Collapse
Affiliation(s)
- Qiuyue He
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
- Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, 650032, China
| | - Min Niu
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
- Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, 650032, China
| | - Jiandie Bi
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Department of Blood Transfusion, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, 650032, China
| | - Na Du
- Department of Clinical Laboratory, The No. 1 Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, 650032, China
| | - Shumin Liu
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
- Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, 650032, China
| | - Kai Yang
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China
- Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, 650032, China
| | - Huanqin Li
- Department of Clinical Laboratory, The No. 1 Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, 650032, China
| | - Jing Yao
- Department of Clinical Laboratory, The No. 1 Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, 650032, China
| | - Yan Du
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China.
- Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, 650032, China.
| | - Yong Duan
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, 650032, China.
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, 650032, China.
- Yunnan Province Clinical Research Center for Laboratory Medicine, Kunming, 650032, China.
| |
Collapse
|
30
|
Aly E, Ros-Berruezo G, López Nicolás R, Ali Darwish A, Sánchez-Moya T, Planes-Muñoz D, Frontela-Saseta C. In vitro prebiotic activity of rhLf and galactooligosaccharides on infant intestinal microbiota. NUTR HOSP 2023; 40:701-710. [PMID: 37073751 DOI: 10.20960/nh.04366] [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] [Indexed: 04/20/2023] Open
Abstract
Introduction Objective: human lactoferrin (Lf) and human milk oligosaccharides possess a wide range of functions. So, the present study focusses on the role of Lf and/or galactooligosaccharides (GOS) in the modulation of gut microbiota composition. Methods: recombinant human lactoferrin (rhLf) was added to the first infant formula (0.10, 0.15, 0.20 %) alone or in combination with GOS (1 %) in vessels of a small-scale batch culture fermentation model. Short-chain fatty acids (SCFAs), microbial population groups, and pH were monitored through fermentation for 24 hours. Results: insignificant changes were observed in pH values and acetic acid accumulated during fermentation. Propionic acid content has been insignificantly increased while butyric acid has been insignificantly decreased. Moreover, increments in all bacterial groups except for Bacteroides were observed through the fermentation process. Lactobacillus and Bifidobacterium showed an increase in relation to initial time over the fermentation process, demonstrating the prebiotic effect of lactoferrin and GOS. After 24 hours of fermentation, all tested ingredients showed significant similarities in Enterococcus for controls except for 0.20 % rhLf + 1 % GOS, which provoked a diminution of Enterococci growth. Conclusion: despite the importance of the batch culture fermentation technique in uncovering the prebiotic activity of food ingredients, it is not useful for detecting the prebiotic nature of Lf due to its nature as a protein. Thus, Lf maybe shows its prebiotic activity on the gut microbiota through other mechanisms.
Collapse
Affiliation(s)
- Esmat Aly
- Food Science and Nutrition Department. Faculty of Veterinary Sciences. Regional Campus of International Excellence "Campus Mare Nostrum". Universidad de Murcia. Food Technology Research Institute (FTRI). Agricultural Research Center (ARC)
| | - Gaspar Ros-Berruezo
- Departamento de Tecnología de Alimentos, Nutrición y Bromatología. Área de Conocimiento de Nutrición y Bromatología. Campus Universitario de Espinardo. Universidad de Murcia
| | - Rubén López Nicolás
- Departamento de Tecnología de los Alimentos, Nutrición y Bromatología. Facultad de Veterinaria. Campus de Excelencia Internacional "Campus Mare Nostrum". Universidad de Murcia
| | - Aliaa Ali Darwish
- Dairy Technology Research Department. Food Technology Research Institute (FTRI). Agricultural Research Center (ARC)
| | - Teresa Sánchez-Moya
- Departamento de Tecnología de los Alimentos, Nutrición y Bromatología. Facultad de Veterinaria. Campus de Excelencia Internacional "Campus Mare Nostrum". Universidad de Murcia
| | - David Planes-Muñoz
- Departamento de Tecnología de los Alimentos, Nutrición y Bromatología. Facultad de Veterinaria. Campus de Excelencia Internacional "Campus Mare Nostrum". Universidad de Murcia
| | - Carmen Frontela-Saseta
- Departamento de Tecnología de los Alimentos, Nutrición y Bromatología. Facultad de Veterinaria. Campus de Excelencia Internacional "Campus Mare Nostrum". Universidad de Murcia
| |
Collapse
|
31
|
Yan Y, Lei Y, Qu Y, Fan Z, Zhang T, Xu Y, Du Q, Brugger D, Chen Y, Zhang K, Zhang E. Bacteroides uniformis-induced perturbations in colonic microbiota and bile acid levels inhibit TH17 differentiation and ameliorate colitis developments. NPJ Biofilms Microbiomes 2023; 9:56. [PMID: 37580334 PMCID: PMC10425470 DOI: 10.1038/s41522-023-00420-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 07/14/2023] [Indexed: 08/16/2023] Open
Abstract
Inflammatory bowel disease (IBD) is associated with gut dysbiosis and can lead to colitis-associated malignancies. Bacteroides uniformis (Bu) regulates animal intestinal homeostasis; however, the mechanism by which it alleviates colitis in mice remains unknown. We investigated the effects of B. uniformis JCM5828 and its metabolites on female C57BL/6J mice with dextran sulfate sodium salt (DSS) induced colitis. Treatment with Bu considerably alleviated colitis progression and restored the mechanical and immune barrier protein expression. Additionally, Bu increased the abundance of the symbiotic bacteria Bifidobacterium and Lactobacillus vaginalis while decreasing that of pathogenic Escherichia-Shigella, and modulated intestinal bile acid metabolism. Bu largely regulated the expression of key regulatory proteins of the NF-κB and mitogen-activated protein kinase (MAPK) signaling pathways in colonic tissues and the differentiation of TH17 cells. However, Bu could not directly inhibit TH17 cell differentiation in vitro; it modulated the process in the lamina propria by participating in bile acid metabolism and regulating key metabolites (alpha-muricholic, hyodeoxycholic, and isolithocholic acid), thereby modulating the intestinal immune response. Our findings suggest that Bu or bile acid supplements are potential therapies for colitis and other diseases associated with intestinal barrier dysfunction.
Collapse
Affiliation(s)
- YiTing Yan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yu Lei
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Ying Qu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Zhen Fan
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Ting Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Yangbin Xu
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Qian Du
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, China
| | - Daniel Brugger
- Institute of Animal Nutrition and Dietetics, Vetsuisse-Faculty, University of Zurich, 8057, Zurich, Switzerland
| | - Yulin Chen
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China
| | - Ke Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| | - Enping Zhang
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling, 712100, China.
| |
Collapse
|
32
|
Gong D, Adomako-Bonsu AG, Wang M, Li J. Three specific gut bacteria in the occurrence and development of colorectal cancer: a concerted effort. PeerJ 2023; 11:e15777. [PMID: 37554340 PMCID: PMC10405800 DOI: 10.7717/peerj.15777] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/29/2023] [Indexed: 08/10/2023] Open
Abstract
Colorectal cancer (CRC), which develops from the gradual evolution of tubular adenomas and serrated polyps in the colon and rectum, has a poor prognosis and a high mortality rate. In addition to genetics, lifestyle, and chronic diseases, intestinal integrity and microbiota (which facilitate digestion, metabolism, and immune regulation) could promote CRC development. For example, enterotoxigenic Bacteroides fragilis, genotoxic Escherichia coli (pks+ E. coli), and Fusobacterium nucleatum, members of the intestinal microbiota, are highly correlated in CRC. This review describes the roles and mechanisms of these three bacteria in CRC development. Their interaction during CRC initiation and progression has also been proposed. Our view is that in the precancerous stage of colorectal cancer, ETBF causes inflammation, leading to potential changes in intestinal ecology that may provide the basic conditions for pks+ E. coli colonization and induction of oncogenic mutations, when cancerous intestinal epithelial cells can further recruit F. nucleatum to colonise the lesion site and F. nucleatum may contribute to CRC advancement by primarily the development of cancer cells, stemization, and proliferation, which could create new and tailored preventive, screening and therapeutic interventions. However, there is the most dominant microbiota in each stage of CRC development, not neglecting the possibility that two or even all three bacteria could be engaged at any stage of the disease. The relationship between the associated gut microbiota and CRC development may provide important information for therapeutic strategies to assess the potential use of the associated gut microbiota in CRC studies, antibiotic therapy, and prevention strategies.
Collapse
Affiliation(s)
- Dengmei Gong
- Institute of Zoonosis, College of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| | - Amma G Adomako-Bonsu
- Institute of Toxicology and Pharmacology, University Medical School Schleswig-Holstein, Kiel, Germany
| | - Maijian Wang
- Gastrointestinal Surgery, Affiliate Hospital of Zunyi Medical University, Zunyi, Guizhou, China
| | - Jida Li
- Institute of Zoonosis, College of Public Health, Zunyi Medical University, Zunyi, Guizhou, China
| |
Collapse
|
33
|
Cameron G, Nguyen T, Ciula M, Williams SJ, Godfrey DI. Glycolipids from the gut symbiont Bacteroides fragilis are agonists for natural killer T cells and induce their regulatory differentiation. Chem Sci 2023; 14:7887-7896. [PMID: 37502334 PMCID: PMC10370605 DOI: 10.1039/d3sc02124f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 07/29/2023] Open
Abstract
Natural Killer T (NKT) cells are a lipid-antigen reactive T cell subset that is restricted to the antigen presenting molecule CD1d. They possess diverse functional properties that contribute to inflammatory and regulatory immune responses. The most studied lipid antigen target for these T cells is α-galactosylceramide (αGC). The commensal organism Bacteroides fragilis (B. fragilis) produces several forms of αGC, but conflicting information exists about the influence of these lipids on NKT cells. Herein, we report the total synthesis of a major form of αGC from B. fragilis (Bf αGC), and several analogues thereof. We confirm the T cell receptor (TCR)-mediated recognition of these glycolipids by mouse and human NKT cells. Despite the natural structure of Bf αGC containing lipid branching that limits potency, we demonstrate that Bf αGC drives mouse NKT cells to proliferate and differentiate into producers of the immunoregulatory cytokine, interleukin-10 (IL-10). These Bf αGC-experienced NKT cells display regulatory function by inhibiting the expansion of naïve NKT cells upon subsequent exposure to this antigen. Moreover, this regulatory activity impacts more than just NKT cells, as demonstrated by the NKT cell-mediated inhibition of antigen-stimulated mucosal-associated invariant T (MAIT) cells (a T cell subset restricted to a different antigen presenting molecule, MR1). These findings reveal that B. fragilis-derived NKT cell agonists may have broad immunoregulatory activity, providing insight into the mechanisms influencing immune tolerance to commensal bacteria and highlighting a potential means to manipulate NKT cell function for therapeutic benefit.
Collapse
Affiliation(s)
- Garth Cameron
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne Melbourne VIC 3000 Australia
| | - Tram Nguyen
- School of Chemistry, University of Melbourne Parkville VIC 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne Parkville VIC 3010 Australia
| | - Marcin Ciula
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne Melbourne VIC 3000 Australia
| | - Spencer J Williams
- School of Chemistry, University of Melbourne Parkville VIC 3010 Australia
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne Parkville VIC 3010 Australia
| | - Dale I Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne Melbourne VIC 3000 Australia
| |
Collapse
|
34
|
Khatiebi S, Kiprotich K, Onyando Z, Wekesa C, Chi CN, Mulambalah C, Okoth P. Shotgun Metagenomic Analyses of Microbial Assemblages in the Aquatic Ecosystem of Winam Gulf of Lake Victoria, Kenya Reveals Multiclass Pollution. BIOMED RESEARCH INTERNATIONAL 2023; 2023:3724531. [PMID: 37521121 PMCID: PMC10382247 DOI: 10.1155/2023/3724531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 08/01/2023]
Abstract
Lake Victoria, the second-largest freshwater lake in the world, provides an important source of food and income, particularly fish for both domestic consumption and for export market. In recent years, Lake Victoria has suffered massive pollution from both industrial and wastewater discharge. Microplastic biomes, pharmaceutical residues, drugs of abuse, heavy metals, agrochemicals, and personal care products are ubiquitous in the aquatic ecosystem of Winam Gulf. These pollutants are known to alter microbial assemblages in aquatic ecosystems with far-reaching ramification including a calamitous consequence to human health. Indeed, some of these pollutants have been associated with human cancers and antimicrobial resistance. There is a paucity of data on the microbial profiles of this important but heavily polluted aquatic ecosystem. The current study sought to investigate the metagenomic profiles of microbial assemblages in the Winam Gulf ecosystem. Water and sediment samples were collected from several locations within the study sites. Total genomic DNA pooled from all sampling sites was extracted and analyzed by whole-genome shotgun sequencing. Analyses revealed three major kingdoms: bacteria, archaea and eukaryotes belonging to 3 phyla, 13 classes, 14 families, 9 orders, 14 genera, and 10 species. Proteobacteria, Betaproteobacteria, Comamonadaceae, Burkholdariales, and Arcobacter were the dominated phyla, class, family, order, genera, and species, respectively. The Kyoto Encyclopedia of Genes and Genomes indicated the highest number of genes involved in metabolism. The presence of carbohydrate metabolism genes and enzymes was used to infer organic pollutions from sewage and agricultural runoffs. Similarly, the presence of xylene and nutrotoluene degradation genes and enzyme was used to infer industrial pollution into the lake. Drug metabolism genes lend credence to the possibility of pharmaceutical pollutants in water. Taken together, there is a clear indication of massive pollution. In addition, carbohydrate-active enzymes were the most abundant and included genes in glycoside hydrolases. Shotgun metagenomic analyses conveyed an understanding of the microbial communities of the massively polluted aquatic ecosystem of Winam Gulf, Lake Vicoria, Kenya. The current study documents the presence of multiclass pollutants in Lake Victoria and reveals information that might be useful for a potential bioremediation strategy using the native microbial communities.
Collapse
Affiliation(s)
- Sandra Khatiebi
- Department of Biological Sciences, School of Natural Science, Masinde Muliro University of Science and Technology, P.O. Box 190, 50100 Kakamega, Kenya
| | - Kelvin Kiprotich
- Department of Biological Sciences, School of Natural Science, Masinde Muliro University of Science and Technology, P.O. Box 190, 50100 Kakamega, Kenya
| | - Zedekiah Onyando
- Department of Biological Sciences, School of Natural Science, Masinde Muliro University of Science and Technology, P.O. Box 190, 50100 Kakamega, Kenya
| | - Clabe Wekesa
- Department of Biological Sciences, School of Natural Science, Masinde Muliro University of Science and Technology, P.O. Box 190, 50100 Kakamega, Kenya
| | - Celestine N. Chi
- Department of Medical Biochemistry and Microbiology, University of Uppsala, P.O. Box 582, 75123 Uppsala, Sweden
| | - Chrispinus Mulambalah
- Department of Medical Microbiology & Parasitology, School of Medicine, Moi University, P.O. Box 4606, 30100 Eldoret, Kenya
| | - Patrick Okoth
- Department of Biological Sciences, School of Natural Science, Masinde Muliro University of Science and Technology, P.O. Box 190, 50100 Kakamega, Kenya
| |
Collapse
|
35
|
Hoffman K, Brownell Z, Doyle WJ, Ochoa-Repáraz J. The immunomodulatory roles of the gut microbiome in autoimmune diseases of the central nervous system: Multiple sclerosis as a model. J Autoimmun 2023; 137:102957. [PMID: 36435700 PMCID: PMC10203067 DOI: 10.1016/j.jaut.2022.102957] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
Abstract
The gut-associated lymphoid tissue is a primary activation site for immune responses to infection and immunomodulation. Experimental evidence using animal disease models suggests that specific gut microbes significantly regulate inflammation and immunoregulatory pathways. Furthermore, recent clinical findings indicate that gut microbes' composition, collectively named gut microbiota, is altered under disease state. This review focuses on the functional mechanisms by which gut microbes promote immunomodulatory responses that could be relevant in balancing inflammation associated with autoimmunity in the central nervous system. We also propose therapeutic interventions that target the composition of the gut microbiota as immunomodulatory mechanisms to control neuroinflammation.
Collapse
Affiliation(s)
- Kristina Hoffman
- Department of Biological Sciences, Boise State University, Boise, ID, 83725, USA
| | - Zackariah Brownell
- Department of Biological Sciences, Arizona State University, Tempe, AZ, 85281, USA
| | - William J Doyle
- Department of Biological Sciences, Boise State University, Boise, ID, 83725, USA
| | - Javier Ochoa-Repáraz
- Department of Biological Sciences, Boise State University, Boise, ID, 83725, USA.
| |
Collapse
|
36
|
Fiyouzi T, Pelaez-Prestel HF, Reyes-Manzanas R, Lafuente EM, Reche PA. Enhancing Regulatory T Cells to Treat Inflammatory and Autoimmune Diseases. Int J Mol Sci 2023; 24:ijms24097797. [PMID: 37175505 PMCID: PMC10177847 DOI: 10.3390/ijms24097797] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 04/19/2023] [Accepted: 04/21/2023] [Indexed: 05/15/2023] Open
Abstract
Regulatory T cells (Tregs) control immune responses and are essential to maintain immune homeostasis and self-tolerance. Hence, it is no coincidence that autoimmune and chronic inflammatory disorders are associated with defects in Tregs. These diseases have currently no cure and are treated with palliative drugs such as immunosuppressant and immunomodulatory agents. Thereby, there is a great interest in developing medical interventions against these diseases based on enhancing Treg cell function and numbers. Here, we give an overview of Treg cell ontogeny and function, paying particular attention to mucosal Tregs. We review some notable approaches to enhance immunomodulation by Tregs with therapeutic purposes including adoptive Treg cell transfer therapy and discuss relevant clinical trials for inflammatory bowel disease. We next introduce ways to expand mucosal Tregs in vivo using microbiota and dietary products that have been the focus of clinical trials in various autoimmune and chronic-inflammatory diseases.
Collapse
Affiliation(s)
- Tara Fiyouzi
- Laboratory of Immunomedicine, Faculty of Medicine, University Complutense of Madrid, Ave Complutense S/N, 28040 Madrid, Spain
| | - Hector F Pelaez-Prestel
- Laboratory of Immunomedicine, Faculty of Medicine, University Complutense of Madrid, Ave Complutense S/N, 28040 Madrid, Spain
| | - Raquel Reyes-Manzanas
- Laboratory of Immunomedicine, Faculty of Medicine, University Complutense of Madrid, Ave Complutense S/N, 28040 Madrid, Spain
| | - Esther M Lafuente
- Laboratory of Immunomedicine, Faculty of Medicine, University Complutense of Madrid, Ave Complutense S/N, 28040 Madrid, Spain
| | - Pedro A Reche
- Laboratory of Immunomedicine, Faculty of Medicine, University Complutense of Madrid, Ave Complutense S/N, 28040 Madrid, Spain
| |
Collapse
|
37
|
Fogarty EC, Schechter MS, Lolans K, Sheahan ML, Veseli I, Moore R, Kiefl E, Moody T, Rice PA, Yu MK, Mimee M, Chang EB, Mclellan SL, Willis AD, Comstock LE, Eren AM. A highly conserved and globally prevalent cryptic plasmid is among the most numerous mobile genetic elements in the human gut. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.25.534219. [PMID: 36993556 PMCID: PMC10055365 DOI: 10.1101/2023.03.25.534219] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/13/2023]
Abstract
Plasmids are extrachromosomal genetic elements that often encode fitness enhancing features. However, many bacteria carry 'cryptic' plasmids that do not confer clear beneficial functions. We identified one such cryptic plasmid, pBI143, which is ubiquitous across industrialized gut microbiomes, and is 14 times as numerous as crAssphage, currently established as the most abundant genetic element in the human gut. The majority of mutations in pBI143 accumulate in specific positions across thousands of metagenomes, indicating strong purifying selection. pBI143 is monoclonal in most individuals, likely due to the priority effect of the version first acquired, often from one's mother. pBI143 can transfer between Bacteroidales and although it does not appear to impact bacterial host fitness in vivo, can transiently acquire additional genetic content. We identified important practical applications of pBI143, including its use in identifying human fecal contamination and its potential as an inexpensive alternative for detecting human colonic inflammatory states.
Collapse
Affiliation(s)
- Emily C Fogarty
- Committee on Microbiology, University of Chicago, Chicago, IL 60637, USA
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Matthew S Schechter
- Committee on Microbiology, University of Chicago, Chicago, IL 60637, USA
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Karen Lolans
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Madeline L. Sheahan
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
- Department of Microbiology, University of Chicago, Chicago, IL, 60637, USA
| | - Iva Veseli
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Ryan Moore
- Center for Bioinformatics and Computational Biology, University of Delaware, Newark, DE, USA
| | - Evan Kiefl
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
- Graduate Program in Biophysical Sciences, University of Chicago, Chicago, IL 60637, USA
| | - Thomas Moody
- Department of Systems Biology, Columbia University, New York, NY, 10032 USA
| | - Phoebe A Rice
- Committee on Microbiology, University of Chicago, Chicago, IL 60637, USA
- Department of Biochemistry, University of Chicago, Chicago, IL, 60637, USA
| | | | - Mark Mimee
- Committee on Microbiology, University of Chicago, Chicago, IL 60637, USA
- Department of Microbiology, University of Chicago, Chicago, IL, 60637, USA
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL 60637, USA
| | - Eugene B Chang
- Department of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Sandra L Mclellan
- School of Freshwater Sciences, University of Wisconsin-Milwaukee, Milwaukee, WI, 53204, USA
| | - Amy D Willis
- Department of Biostatistics, University of Washington, Seattle, WA, 98195, USA
| | - Laurie E Comstock
- Duchossois Family Institute, University of Chicago, Chicago, IL 60637, USA
- Department of Microbiology, University of Chicago, Chicago, IL, 60637, USA
| | - A Murat Eren
- Marine Biological Laboratory, Woods Hole, MA, 02543, USA
- Alfred Wegener Institute, Helmholtz Centre for Polar and Marine Research, 27570 Bremerhaven, Germany
- Institute for Chemistry and Biology of the Marine Environment, University of Oldenburg, 26129 Oldenburg, Germany
- Helmholtz Institute for Functional Marine Biodiversity, 26129 Oldenburg, Germany
| |
Collapse
|
38
|
Liu Y, Lei S, Hou R, Li D, Wan X, Cai H, Chen G. Tea polysaccharides from Taiping Houkui may serve as a potential candidate for regulation of lipid metabolism: Roles of gut microbiota and metabolite in vitro. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/23/2023] Open
|
39
|
Eribo OA, Naidoo CC, Theron G, Walzl G, du Plessis N, Chegou NN. An Archetypical Model for Engrafting Bacteroides fragilis into Conventional Mice Following Reproducible Antibiotic Conditioning of the Gut Microbiota. Microorganisms 2023; 11:microorganisms11020451. [PMID: 36838416 PMCID: PMC9966493 DOI: 10.3390/microorganisms11020451] [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: 12/16/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023] Open
Abstract
Bacteroides fragilis is a commonly investigated commensal bacterium for its protective role in host diseases. Here, we aimed to develop a reproducible antibiotic-based model for conditioning the gut microbiota and engrafting B. fragilis into a conventional murine host. Initially, we selected different combinations of antibiotics, including metronidazole, imipenem, and clindamycin, and investigated their efficacy in depleting the mouse Bacteroides population. We performed 16S rRNA sequencing of DNA isolated from fecal samples at different time points. The α-diversity was similar in mice treated with metronidazole (MET) and differed only at weeks 1 (p = 0.001) and 3 (p = 0.009) during metronidazole/imipenem (MI) treatment. Bacteroides compositions, during the MET and MI exposures, were similar to the pre-antibiotic exposure states. Clindamycin supplementation added to MET or MI regimens eliminated the Bacteroides population. We next repeated metronidazole/clindamycin (MC) treatment in two additional independent experiments, followed by a B. fragilis transplant. MC consistently and reproducibly eliminated the Bacteroides population. The depleted Bacteroides did not recover in a convalescence period of six weeks post-MC treatment. Finally, B. fragilis was enriched for ten days following engraftment into Bacteroides-depleted mice. Our model has potential use in gut microbiota studies that selectively investigate Bacteroides' role in diseases of interest.
Collapse
Affiliation(s)
- Osagie A. Eribo
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa
| | - Charissa C. Naidoo
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa
- African Microbiome Institute, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa
| | - Grant Theron
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa
- African Microbiome Institute, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa
| | - Gerhard Walzl
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa
| | - Nelita du Plessis
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa
| | - Novel N. Chegou
- DSI-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town 8000, South Africa
- Correspondence:
| |
Collapse
|
40
|
Pither MD, Silipo A, Molinaro A, Di Lorenzo F. Extraction, Purification, and Chemical Degradation of LPS from Gut Microbiota Strains. Methods Mol Biol 2023; 2613:153-179. [PMID: 36587078 DOI: 10.1007/978-1-0716-2910-9_13] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
It is estimated that more than 500 different bacterial species colonize the human gut, and they are collectively known as the gut microbiota. Such a massive bacterial presence is now considered an additional organ of the human body, thus becoming the object of an intense and daily growing research activity. Gram-negative bacteria represent a large percentage of the gut microbiota strains. The main constituent of the outer membrane of Gram-negatives is the lipopolysaccharide (LPS). Since its first discovery, LPS has been extensively studied for its structure-dependent capability to elicit a potent immune inflammatory reaction when perceived by specific immune receptors present in our body. Therefore, traditionally, LPS, due to its peculiar chemistry, has been associated with pathogenic bacteria, and it has been extensively studied for its dangerous effects on human health. However, LPS is also expressed on the cell surface of harmless and beneficial bacteria that colonize our intestines. This necessarily implies that the LPS from harmless gut microbes is "chemically different" from that owned by pathogenic ones, hence enabling successful colonization of the intestinal tract without creating a threat to the host immune system. Deciphering the structural features of LPS from these gut bacteria is essential to improve our still scarce knowledge of how the human host lives in a harmonious relationship with its own microbiota. To this end, LPS extraction and purification are essential steps in this field of research. Yet working with gut bacteria is extremely complex for a number of reasons, one being related to the fact that they produce an array of other glycans and glycoconjugates, such as capsular polysaccharides and/or exopolysaccharides, which render the isolation and characterization of the sole LPS not at all trivial. Here, we provide a protocol that might help when dealing with LPS from gut microbial species. We describe the preliminary manipulations and checks, extraction, and purification approaches, as well as the necessary chemical manipulations that should be performed to enable the characterization of the structure of an LPS by means of techniques like nuclear magnetic resonance spectroscopy and mass spectrometry.
Collapse
Affiliation(s)
- Molly Dorothy Pither
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy.
| | - Alba Silipo
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Antonio Molinaro
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Flaviana Di Lorenzo
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy.
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy.
- Department of Agricultural Sciences, University of Naples Federico II, Portici, Naples, Italy.
| |
Collapse
|
41
|
Wang J, Ding Y, Li D, Zhu N, Nishiyama A, Yuan Y. (Pro)renin receptor promotes colorectal cancer progression through inhibiting the NEDD4L-mediated Wnt3 ubiquitination and modulating gut microbiota. Cell Commun Signal 2023; 21:2. [PMID: 36597142 PMCID: PMC9809055 DOI: 10.1186/s12964-022-01015-x] [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: 06/05/2022] [Accepted: 12/11/2022] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND We previously found that (pro)renin receptor ((P)RR) augments Wnt3 protein without affecting Wnt3 gene transcription in colorectal cancer (CRC) cells, thus contributes to CRC initiation. The present study aims to investigate whether (P)RR further promotes CRC progression following oncogenesis and the related mechanisms. Notably, we deeply elaborate how (P)RR affects Wnt3 protein level and the key enzyme that mediates this process. METHODS Immunohistochemistry, western blotting and immunofluorescence were performed to detect protein expression status. A kind of gastrointestinal epithelium-specific ATP6AP2 ((P)RR encoding gene) knock-in mice were generated using Crispr/Cas9 system. RESULTS We found that increased (P)RR expression in primary CRC lesions is positively associated with higher Wnt3 protein level and disease progression. Progressive CRC presents less colocalization of Wnt3 and an E3 ubiquitin ligase NEDD4L in primary lesions than non-progressive CRC. In colon cancer cells, (P)RR dramatically inhibits the NEDD4L-mediated Wnt3 protein ubiquitination. ATP6AP2 knock-in mice show more diminished Wnt3-NEDD4L colocalization in their gut epithelium in comparison to wildtype mice. They also have abnormal gut bacterial flora distribution. Especially, Lachnospiraceae_NK4A136 and Bacteroides genus, which are generally protective against CRC, are suppressed in guts of ATP6AP2 knock-in mice. CONCLUSIONS Collectively, (P)RR promotes CRC progression through inhibiting the NEDD4L-mediated Wnt3 ubiquitination and modulating gut microbiota. Video Abstract.
Collapse
Affiliation(s)
- Juan Wang
- grid.412465.0Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences of Zhejiang Province), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China ,Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center of Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Yuwei Ding
- grid.412465.0Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences of Zhejiang Province), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China ,Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center of Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Dan Li
- grid.412465.0Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences of Zhejiang Province), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China ,Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center of Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Ning Zhu
- grid.412465.0Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences of Zhejiang Province), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China ,Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center of Zhejiang University, Hangzhou, 310058 Zhejiang China
| | - Akira Nishiyama
- grid.258331.e0000 0000 8662 309XDepartment of Pharmacology, Faculty of Medicine, Kagawa University, Takamatsu, Kagawa 761-0793 Japan
| | - Ying Yuan
- grid.412465.0Department of Medical Oncology (Key Laboratory of Cancer Prevention and Intervention, China National Ministry of Education, Key Laboratory of Molecular Biology in Medical Sciences of Zhejiang Province), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009 Zhejiang China ,Zhejiang Provincial Clinical Research Center for Cancer, Hangzhou, 310009 Zhejiang China ,grid.13402.340000 0004 1759 700XCancer Center of Zhejiang University, Hangzhou, 310058 Zhejiang China
| |
Collapse
|
42
|
Xia C, Cai Y, Ren S, Xia C. Role of microbes in colorectal cancer therapy: Cross-talk between the microbiome and tumor microenvironment. Front Pharmacol 2022; 13:1051330. [PMID: 36438840 PMCID: PMC9682563 DOI: 10.3389/fphar.2022.1051330] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 10/24/2022] [Indexed: 08/10/2023] Open
Abstract
The human gut microbiota is associated with the development and progression of colorectal cancer, and manipulation of the gut microbiota is a novel strategy for the prevention and treatment of colorectal cancer. Some bacteria have antitumor activity against colorectal cancer, where specific bacteria can improve the tumor microenvironment, activate immune cells including dendritic cells, helper T cells, natural killer cells, and cytotoxic T cells, and upregulate the secretion of pro-tumor immune cytokines such as interleukin-2 and interferon. In this paper, we summarize some bacteria with potential benefits in colorectal cancer and describe their roles in the tumor microenvironment, demonstrate the application of gut microbes in combination with immunosuppressive agents, and provide suggestions for further experimental studies and clinical practice applications.
Collapse
Affiliation(s)
- Cong Xia
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yantao Cai
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, China
| | - Shuangyi Ren
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Chenglai Xia
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan, China
- School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
Romero-Garmendia I, Garcia-Etxebarria K. Host Genetics and Microbiota Interactions in Colorectal Cancer: Shared or Independent Risk? Microorganisms 2022; 10:2129. [PMID: 36363721 PMCID: PMC9697093 DOI: 10.3390/microorganisms10112129] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 07/30/2023] Open
Abstract
The role of microbiota in colorectal cancer has been studied since alterations in its composition were observed. In addition, there are more and more pieces of evidence that microbiota could be implicated in colorectal cancer progression. Thus, the components of the microbiota could be biomarkers for the diagnosis and prognosis of colorectal cancer. In addition, it is important to address how the microbiota interacts with the host and how the host shapes the microbiota, in order to understand the biological pathways and mechanisms involved in their relationship and the consequences of their interactions in colorectal cancer. Thereby, it could be possible to find feasible measures and treatments to prevent or better diagnose colorectal cancer. In this review, we will try to summarize the role of the microbiota in colorectal cancer and its interactions with the host and the host genetics, coming to some conclusions that could be useful to find the gaps in our knowledge and propose future steps in this field.
Collapse
Affiliation(s)
- Irati Romero-Garmendia
- Department of Genetics, Physical Anthropology and Animal Physiology, University of the Basque Country (Universidad del País Vasco/Euskal Herriko Unibertsitatea), 48940 Leioa, Spain
| | - Koldo Garcia-Etxebarria
- Gastrointestinal Genetics Group, Biodonostia, 20014 San Sebastián, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 08036 Barcelona, Spain
| |
Collapse
|
44
|
Štofilová J, Kvaková M, Kamlárová A, Hijová E, Bertková I, Guľašová Z. Probiotic-Based Intervention in the Treatment of Ulcerative Colitis: Conventional and New Approaches. Biomedicines 2022; 10:2236. [PMID: 36140337 PMCID: PMC9496552 DOI: 10.3390/biomedicines10092236] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 08/31/2022] [Accepted: 09/02/2022] [Indexed: 12/02/2022] Open
Abstract
Although there are number of available therapies for ulcerative colitis (UC), many patients are unresponsive to these treatments or experience secondary failure during treatment. Thus, the development of new therapies or alternative strategies with minimal side effects is inevitable. Strategies targeting dysbiosis of gut microbiota have been tested in the management of UC due to the unquestionable role of gut microbiota in the etiology of UC. Advanced molecular analyses of gut microbiomes revealed evident dysbiosis in UC patients, characterized by a reduced biodiversity of commensal microbiota. Administration of conventional probiotic strains is a commonly applied approach in the management of the disease to modify the gut microbiome, improve intestinal barrier integrity and function, and maintain a balanced immune response. However, conventional probiotics do not always provide the expected health benefits to a patient. Their benefits vary significantly, depending on the type and stage of the disease and the strain and dose of the probiotics administered. Their mechanism of action is also strain-dependent. Recently, new candidates for potential next-generation probiotics have been discovered. This could bring to light new approaches in the restoration of microbiome homeostasis and in UC treatment in a targeted manner. The aim of this paper is to provide an updated review on the current options of probiotic-based therapies, highlight the effective conventional probiotic strains, and outline the future possibilities of next-generation probiotic and postbiotic supplementation and fecal microbiota transplantation in the management of UC.
Collapse
Affiliation(s)
- Jana Štofilová
- Center of Clinical and Preclinical Research MEDIPARK, Faculty of Medicine, Pavol Jozef Safarik University in Kosice, Trieda SNP 1, 040 11 Kosice, Slovakia
| | | | | | | | | | | |
Collapse
|
45
|
Cheng J, Hu J, Geng F, Nie S. Bacteroides utilization for dietary polysaccharides and their beneficial effects on gut health. FOOD SCIENCE AND HUMAN WELLNESS 2022. [DOI: 10.1016/j.fshw.2022.04.002] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
46
|
Kapsetaki SE, Marquez Alcaraz G, Maley CC, Whisner CM, Aktipis A. Diet, Microbes, and Cancer Across the Tree of Life: a Systematic Review. Curr Nutr Rep 2022; 11:508-525. [PMID: 35704266 PMCID: PMC9197725 DOI: 10.1007/s13668-022-00420-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/21/2022] [Indexed: 11/09/2022]
Abstract
PURPOSE OF REVIEW Cancers are a leading cause of death in humans and for many other species. Diet has often been associated with cancers, and the microbiome is an essential mediator between diet and cancers. Here, we review the work on cancer and the microbiome across species to search for broad patterns of susceptibility associated with different microbial species. RECENT FINDINGS Some microbes, such as Helicobacter bacteria, papillomaviruses, and the carnivore-associated Fusobacteria, consistently induce tumorigenesis in humans and other species. Other microbes, such as the milk-associated Lactobacillus, consistently inhibit tumorigenesis in humans and other species. We systematically reviewed over a thousand published articles and identified links between diet, microbes, and cancers in several species of mammals, birds, and flies. Future work should examine a larger variety of host species to discover new model organisms for human preclinical trials, to better understand the observed variance in cancer prevalence across species, and to discover which microbes and diets are associated with cancers across species. Ultimately, this could help identify microbial and dietary interventions to diagnose, prevent, and treat cancers in humans as well as other animals.
Collapse
Affiliation(s)
- Stefania E Kapsetaki
- Arizona Cancer Evolution Center, Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA.
- Biodesign Center for Biocomputing, Security and Society, Arizona State University, Tempe, USA.
| | - Gissel Marquez Alcaraz
- Arizona Cancer Evolution Center, Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Biocomputing, Security and Society, Arizona State University, Tempe, USA
| | - Carlo C Maley
- Arizona Cancer Evolution Center, Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Biodesign Center for Biocomputing, Security and Society, Arizona State University, Tempe, USA
| | - Corrie M Whisner
- College of Health Solutions, Arizona State University, Phoenix, AZ, USA
- Biodesign Center for Health Through Microbiomes, Arizona State University, Tempe, AZ, USA
| | - Athena Aktipis
- Arizona Cancer Evolution Center, Biodesign Institute and School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Department of Psychology, Arizona State University, Tempe, AZ, USA
| |
Collapse
|
47
|
Yinhang W, Wei W, Jing Z, Qing Z, Yani Z, Yangyanqiu W, Shuwen H. Biological roles of toll-like receptors and gut microbiota in colorectal cancer. Future Microbiol 2022; 17:1071-1089. [PMID: 35916158 DOI: 10.2217/fmb-2021-0072] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Colorectal cancer (CRC) is one of the most considerably common malignancies of the alimentary system, with high mortality and incidence rates. The present study suggested that the occurrence of CRC is closely related to bacteria, as the large intestine is a gathering place for human micro-organisms. However, the nosogenesis of bacteria leading to tumorigenesis is still obscure. Recently, many studies have reported that toll-like receptors and their related molecular pathways are involved in the process of gut micro-organisms generating CRC. Gut micro-organisms can promote or inhibit the development of CRC via binding to special toll-like receptors. In this paper, the authors review the relationship among toll-like receptors, gut micro-organisms and CRC in order to provide a reference for future tumor immunotherapy and targeted therapy.
Collapse
Affiliation(s)
- Wu Yinhang
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,The Second School of Clinical Medicine, Zhejiang Chinese Medical University, 548 Binwen Road, Binjiang District, Hangzhou, Zhejiang Province, 310053, China.,Key Laboratory of Multiomics Research & Clinical Transformation of Digestive Cancer of Huzhou,1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Wu Wei
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Key Laboratory of Multiomics Research & Clinical Transformation of Digestive Cancer of Huzhou,1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Zhuang Jing
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Key Laboratory of Multiomics Research & Clinical Transformation of Digestive Cancer of Huzhou,1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Zhou Qing
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Key Laboratory of Multiomics Research & Clinical Transformation of Digestive Cancer of Huzhou,1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| | - Zhou Yani
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Graduate School of Medicine Faculty, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou, Zhejiang Province, 310058, China
| | - Wang Yangyanqiu
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Graduate School of Medicine Faculty, Zhejiang University, 866 Yuhangtang Road, Xihu District, Hangzhou, Zhejiang Province, 310058, China
| | - Han Shuwen
- Huzhou Central Hospital, Affiliated Central Hospital, Huzhou University, 1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China.,Key Laboratory of Multiomics Research & Clinical Transformation of Digestive Cancer of Huzhou,1558 Sanhuan North Road, Wuxing District, Huzhou, Zhejiang Province, 313000, China
| |
Collapse
|
48
|
Cao B, Wang S, Li R, Wang Z, Li T, Zhang Y, Dong B, Li Y, Lin M, Li X, Xiao X, Li C, Li G. Xihuang Pill enhances anticancer effect of anlotinib by regulating gut microbiota composition and tumor angiogenesis pathway. Biomed Pharmacother 2022; 151:113081. [PMID: 35605293 DOI: 10.1016/j.biopha.2022.113081] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/22/2022] [Accepted: 05/04/2022] [Indexed: 11/24/2022] Open
Abstract
Lung cancer poses a serious threat to human health. Although targeted therapies have led to breakthroughs in the treatment of lung cancer, drug resistance and side effects limit their clinical applications. Xihuang pill (XHW), a classical anti-cancer traditional Chinese medicine formula, has been clinically proven to be an effective complementary therapy in the treatment of various of cancers. However, the underlying mechanism for its use in combination with anti-cancer drugs remains unclear. Here, we explored the anti-lung cancer effect of XHW combined with anlotinib in mice bearing Lewis lung cancer (LLC). We used gut microbiota and transcriptomics to elucidate the regulatory properties of XHW in improving anti-lung cancer effect of anlotinib. The results showed that combination treatment of XHW with Anlotinib significantly inhibited tumor growth in LLC-bearing mice. We found that XHW played a key role in the regulation of gut microbiota using 16 s rRNA sequencing analysis. Specifically, XHW increased the proportion of the beneficial bacteria Bacteroides and g_norank_f_Muribaculaceae. Based on transcriptomic analysis of tumor tissues, differentially expressed genes in the combination therapy group were related to biological processes concerning angiogenesis, such as regulation of blood vessel diameter, regulation of tube diameter, and regulation of tube size. Our data suggest that XWH enhances the anticancer effect of anlotinib by regulating gut microbiota composition and tumor angiogenesis pathway. Combination therapy with anlotinib and XHW may be a novel therapeutic strategy for lung cancer patients.
Collapse
Affiliation(s)
- Bo Cao
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Shiyuan Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Ruisheng Li
- Research Center for Clinical and Translational Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhihong Wang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Taifeng Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Yuanyuan Zhang
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Bin Dong
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Yingying Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Mengmeng Lin
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China
| | - Xingjie Li
- Research Center for Clinical and Translational Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Xiaohe Xiao
- China Military Institute of Chinese Medicine, Fifth Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Chunyu Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| | - Guohui Li
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, China.
| |
Collapse
|
49
|
Ustjanzew A, Sencio V, Trottein F, Faber J, Sandhoff R, Paret C. Interaction between Bacteria and the Immune System for Cancer Immunotherapy: The α-GalCer Alliance. Int J Mol Sci 2022; 23:ijms23115896. [PMID: 35682578 PMCID: PMC9180740 DOI: 10.3390/ijms23115896] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Revised: 05/19/2022] [Accepted: 05/22/2022] [Indexed: 11/25/2022] Open
Abstract
Non-conventional T cells, such as γδ T and invariant natural killer T (iNKT) cells, are emerging players in fighting cancer. Alpha-galactosylceramide (α-GalCer) is used as an exogenous ligand to activate iNKT cells. Human cells don’t have a direct pathway producing α-GalCer, which, however, can be produced by bacteria. We searched the literature for bacteria strains that are able to produce α-GalCer and used available sequencing data to analyze their presence in human tumor tissues and their association with survival. The modulatory effect of antibiotics on the concentration of α-GalCer was analyzed in mice. The human gut bacteria Bacteroides fragilis, Bacteroides vulgatus, and Prevotella copri produce α-GalCer structures that are able to activate iNKT cells. In mice, α-GalCer was depleted upon treatment with vancomycin. The three species were detected in colon adenocarcinoma (COAD) and rectum adenocarcinoma tissues, and Prevotella copri was also detected in bone tumors and glioblastoma tissues. Bacteroides vulgatus in COAD tissues correlated with better survival. In conclusion, α-GalCer-producing bacteria are part of the human gut microbiome and can infiltrate tumor tissues. These results suggest a new mechanism of interaction between bacteria and immune cells: α-GalCer produced by bacteria may activate non-conventional T cells in tumor tissues, where they can exert a direct or indirect anti-tumor activity.
Collapse
Affiliation(s)
- Arsenij Ustjanzew
- Institute of Medical Biostatistics, Epidemiology and Informatics (IMBEI), University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Valentin Sencio
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 8204, University of Lille, CHU Lille, Institut Pasteur de Lille, 59000 Lille, France; (V.S.); (F.T.)
| | - François Trottein
- Centre d’Infection et d’Immunité de Lille, Inserm U1019, CNRS UMR 8204, University of Lille, CHU Lille, Institut Pasteur de Lille, 59000 Lille, France; (V.S.); (F.T.)
| | - Jörg Faber
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
| | - Roger Sandhoff
- Lipid Pathobiochemistry, German Cancer Research Center, 69120 Heidelberg, Germany;
| | - Claudia Paret
- Department of Pediatric Hematology/Oncology, Center for Pediatric and Adolescent Medicine, University Medical Center of the Johannes Gutenberg University Mainz, 55131 Mainz, Germany;
- Correspondence:
| |
Collapse
|
50
|
Chemically Induced Colitis-Associated Cancer Models in Rodents for Pharmacological Modulation: A Systematic Review. J Clin Med 2022; 11:jcm11102739. [PMID: 35628865 PMCID: PMC9146029 DOI: 10.3390/jcm11102739] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 04/29/2022] [Accepted: 05/10/2022] [Indexed: 02/01/2023] Open
Abstract
Animal models for colitis-associated colorectal cancer (CACC) represent an important tool to explore the mechanistic basis of cancer-related inflammation, providing important evidence that several inflammatory mediators play specific roles in the initiation and perpetuation of colitis and CACC. Although several original articles have been published describing the CACC model in rodents, there is no consensus about the induction method. This review aims to identify, summarize, compare, and discuss the chemical methods for the induction of CACC through the PRISMA methodology. METHODS We searched MEDLINE via the Pubmed platform for studies published through March 2021, using a highly sensitive search expression. The inclusion criteria were only original articles, articles where a chemically-induced animal model of CACC is described, preclinical studies in vivo with rodents, and articles published in English. RESULTS Chemically inducible models typically begin with the administration of a carcinogenic compound (as azoxymethane (AOM) or 1,2-dimethylhydrazine (DMH)), and inflammation is caused by repeated cycles of colitis-inducing agents (such as 2,4,6-trinitrobenzenesulfonic acid (TNBS) or dextran sulfate sodium (DSS)). The strains mostly used are C57BL/6 and Balb/c with 5-6 weeks. To characterize the preclinical model, the parameters more used include body weight, stool consistency and morbidity, inflammatory biomarkers such as tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β, angiogenesis markers such as proliferating cell nuclear antigen (PCNA), marker of proliferation Ki-67, and caspase 3, the presence of ulcers, thickness or hyperemia in the colon, and histological evaluation of inflammation. CONCLUSION The AOM administration seems to be important to the CACC induction method, since the carcinogenic effect is achieved with just one administration. DSS has been the more used inflammatory agent; however, the TNBS contribution should be more studied, since it allows a reliable, robust, and a highly reproducible animal model of intestinal inflammation.
Collapse
|