1
|
Daghrery A, Araújo IJDS, Marques JF, Alipour M, Ünsal RBK, Chathoth BM, Sivaramakrishnan G, Delgadillo-Barrera S, Chaurasia A. Role of exosomes in dental and craniofacial regeneration - A review. Tissue Cell 2025; 93:102684. [PMID: 39740273 DOI: 10.1016/j.tice.2024.102684] [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: 08/16/2024] [Revised: 12/05/2024] [Accepted: 12/11/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND The treatment of congenital deformities, traumatic injuries, infectious diseases, and tumors in the craniomaxillofacial (CMF) region is complex due to the intricate nature of the tissues involved. Conventional treatments such as bone grafts and cell transplantation face limitations, including the need for multiple surgeries, complications, and safety concerns. OBJECTIVE This paper aims to provide a comprehensive analysis of the role of exosomes (EXOs) in CMF and dental tissue regeneration and to explore their potential applications in regenerative dental medicine. METHODS An extensive review of advancements in tissue engineering, materials sciences, and nanotechnology was conducted to evaluate the development of delivery systems for EXOs-based therapies. The analysis included how EXOs, as nanovesicles released by cells, can be modified to target specific cells or loaded with functional molecules for drug or gene delivery. RESULTS EXOs have emerged as a promising alternative to cell transplant therapy, offering a safer method for cell communication and epigenetic control. EXOs transport important proteins and genetic materials, facilitating intercellular communication and delivering therapeutics effectively. The potential of EXOs in personalized medicine, particularly in diagnosing, customizing treatment, and predicting patient responses, is highlighted. CONCLUSION EXO-mediated therapy holds significant potential for advancing tissue regeneration, offering targeted, personalized treatment options with reduced side effects. However, challenges in purification, production, and standardized protocols need to be addressed before its clinical application can be fully realized.
Collapse
Affiliation(s)
- Arwa Daghrery
- Department of Restorative Dental Sciences, School of Dentistry, Jazan University, Jazan, Kingdom of Saudi Arabia.
| | | | - Joana Faria Marques
- Faculdade de Medicina Dentária, Universidade de Lisboa, Cidade Universitária, Lisboa 1600-277, Portugal.
| | - Mahdieh Alipour
- Dental and Periodontal Research Center, Faculty of Dentistry, Tabriz University of Medical Sciences, Iran; Departments of Oral and Craniofacial Sciences, School of Dental Medicine, University of Pittsburgh, USA.
| | - Revan Birke Koca Ünsal
- Department of Periodontology, University of Kyrenia, Faculty of Dentistry, Kyrenia, Cyprus.
| | | | | | - Sara Delgadillo-Barrera
- Grupo de Investigacion Básica y Aplicada en Odontología - IBAPO, Facultad de Odontologia, Universidad Nacional de Colombia, Bogotá, Colombia.
| | - Akhilanand Chaurasia
- Department of Oral Medicine and Radiology, Faculty of Dental Sciences. King George's Medical University, Lucknow, India.
| |
Collapse
|
2
|
Grenda A, Iwan E, Kuźnar-Kamińska B, Bomba A, Bielińska K, Krawczyk P, Chmielewska I, Frąk M, Szczyrek M, Rolska-Kopińska A, Jankowski T, Kieszko R, Milanowski J. Gut microbial predictors of first-line immunotherapy efficacy in advanced NSCLC patients. Sci Rep 2025; 15:6139. [PMID: 39979394 PMCID: PMC11842579 DOI: 10.1038/s41598-025-89406-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 02/05/2025] [Indexed: 02/22/2025] Open
Abstract
The composition of the gut microbiome of patients with advanced non-small cell lung cancer is currently considered a factor influencing the effectiveness of treatment with immune checkpoint inhibitors. We aimed to evaluate the baseline gut microbiome composition in patients before receiving first-line immunotherapy alone or combined with chemoimmunotherapy. We performed 16S rRNA sequencing based on hypervariable regions. Stool samples were collected from 52 patients with advanced NSCLC treated with immunotherapy or chemoimmunotherapy before treatment. We found that the Ruminococcaceae family, species Alistipes sp. genus Eubacterium ventriosum group and genus Marvinbryantia may be intestinal, microbiological predictors of response to treatment. Genus Akkermansia and species from the [Clostridum] leptum group predicted the length of PFS (progression-free survival). Longer OS (overall survival) is associated with bacteria from the Ruminococcaceae family genera [Eubacterium] ventriosum group, Marvinbryantia, Colidextribacter and species [Clostridum] leptum. Bacteria that have an adverse effect (shortening of PFS or OS) on the response to treatment using immune checkpoint inhibitors are Rothia genus, Streptococus salivarius, Streptococus, Family XIII AD3011 group and Family XIII AD3011 group, s. uncultured bacterium. The composition of intestinal flora can be a predictive factor for immunotherapy in NSCLC patients. Specific bacteria can be positively or negatively associated with response to treatment, progression-free survival, and overall survival. They can be potentially used as predictive markers in NSCLC patients treated with immunotherapy.
Collapse
Affiliation(s)
- Anna Grenda
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, ul. Jaczewskiego 8, Lublin, 20-954, Poland.
| | - Ewelina Iwan
- Department of Omics Analyses, National Veterinary Research Institute, 57 Partyzantow, Pulawy, 24-100, Poland
| | - Barbara Kuźnar-Kamińska
- Department of Pulmonology, Allergology and Pulmonary Oncology, Poznan University of Medical Sciences, Poznań, Poland
| | - Arkadiusz Bomba
- Department of Omics Analyses, National Veterinary Research Institute, 57 Partyzantow, Pulawy, 24-100, Poland
| | - Katarzyna Bielińska
- Department of Omics Analyses, National Veterinary Research Institute, 57 Partyzantow, Pulawy, 24-100, Poland
| | - Paweł Krawczyk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, ul. Jaczewskiego 8, Lublin, 20-954, Poland
| | - Izabela Chmielewska
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, ul. Jaczewskiego 8, Lublin, 20-954, Poland
| | - Małgorzata Frąk
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, ul. Jaczewskiego 8, Lublin, 20-954, Poland
| | - Michał Szczyrek
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, ul. Jaczewskiego 8, Lublin, 20-954, Poland
| | - Anna Rolska-Kopińska
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, ul. Jaczewskiego 8, Lublin, 20-954, Poland
| | - Tomasz Jankowski
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, ul. Jaczewskiego 8, Lublin, 20-954, Poland
| | - Robert Kieszko
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, ul. Jaczewskiego 8, Lublin, 20-954, Poland
| | - Janusz Milanowski
- Department of Pneumonology, Oncology and Allergology, Medical University of Lublin, ul. Jaczewskiego 8, Lublin, 20-954, Poland
| |
Collapse
|
3
|
Northrop-Albrecht EJ, Kim Y, Taylor WR, Majumder S, Kisiel JB, Lucien F. The proteomic landscape of stool-derived extracellular vesicles in patients with pre-cancerous lesions and colorectal cancer. Commun Biol 2025; 8:228. [PMID: 39948151 PMCID: PMC11825688 DOI: 10.1038/s42003-025-07652-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Colorectal cancer (CRC) is the 2nd most fatal cancer in the United States, but when detected early it is highly curable. Stool-derived extracellular vesicles (EVs) are a novel biomarker source that could augment the sensitivity for detection of CRC precursors. However, standardization of isolation methods for stool-derived EVs remains underexplored. We previously reported that size-exclusion chromatography (SEC) followed by ultrafiltration (UF-100kDa) was suitable for human stool supernatant EV isolation. In this study, we first assess alternative EV concentration methods (ultrafiltration [UF]; 10 kDa, 30 kDa, 50 kDa, 100 kDa and speed vacuum [SV]). Second, we investigate the host/bacterial EV proteomes by mass spectrometry. We report no difference in recovery, RNA and soluble protein contamination among concentration methods. Proteomic analysis reveals a diverse bacterial proteome, while human-derived proteins are more abundant. Specifically, pancreatic enzymes are among the most abundant proteins, further exploration revealed that zymogen granules are likely co-isolated in stool EV preparations. To enable discovery of EV-based molecular signatures of CRC precursors with high sensitivity, immunocapture strategies will likely be needed. Notably, we identified 10 surface proteins that may serve as candidates for the purification of colon-derived EVs. This work serves as framework for the future discovery and validation of EV-based biomarkers for CRC.
Collapse
Affiliation(s)
| | - Yohan Kim
- Department of Urology, Mayo Clinic, Rochester, MN, USA
| | - William R Taylor
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Shounak Majumder
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - John B Kisiel
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, MN, USA.
- Department of Immunology, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
4
|
Mishra S, Tejesvi MV, Hekkala J, Turunen J, Kandikanti N, Kaisanlahti A, Suokas M, Leppä S, Vihinen P, Kuitunen H, Sunela K, Koivunen J, Jukkola A, Kalashnikov I, Auvinen P, Kääriäinen OS, Peñate Medina T, Peñate Medina O, Saarnio J, Meriläinen S, Rautio T, Aro R, Häivälä R, Suojanen J, Laine M, Erawijattari PP, Lahti L, Karihtala P, Ruuska TS, Reunanen J. Gut microbiome-derived bacterial extracellular vesicles in patients with solid tumours. J Adv Res 2025; 68:375-386. [PMID: 38458256 PMCID: PMC11785572 DOI: 10.1016/j.jare.2024.03.003] [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: 12/21/2023] [Revised: 02/17/2024] [Accepted: 03/06/2024] [Indexed: 03/10/2024] Open
Abstract
INTRODUCTION Gut microbiome-derived nanoparticles, known as bacterial extracellular vesicles (bEVs), have garnered interest as promising tools for studying the link between the gut microbiome and human health. The diverse composition of bEVs, including their proteins, mRNAs, metabolites, and lipids, makes them useful for investigating diseases such as cancer. However, conventional approaches for studying gut microbiome composition alone may not be accurate in deciphering host-gut microbiome communication. In clinical microbiome research, there is a gap in the knowledge on the role of bEVs in solid tumor patients. OBJECTIVES Analyzing the functionality of bEVs using (meta)genomics and proteomics could highlight the unique aspects of host-gut microbiome interactions in solid tumor patients. Therefore, we performed a comparative analysis of the proteome and microbiota composition of gut microbiome-derived bEVs isolated from patients with solid tumors and healthy controls. METHODS After isolating bEVs from the feces of solid tumor patients and healthy controls, we performed spectrometry analysis of their proteomes and next-generation sequencing (NGS) of the 16S gene. We also investigated the gut microbiomes of feces from patients and controls using 16S sequencing and used machine learning to classify the samples into patients and controls based on their bEVs and fecal microbiomes. RESULTS Solid tumor patients showed decreased microbiota richness and diversity in both the bEVs and feces. However, the bEV proteomes were more diverse in patients than in the controls and were enriched with proteins associated with the metabolism of amino acids and carbohydrates, nucleotide binding, and oxidoreductase activity. Metadata classification of samples was more accurate using fecal bEVs (100%) compared with fecal samples (93%). CONCLUSION Our findings suggest that bEVs are unique functional entities. There is a need to explore bEVs together with conventional gut microbiome analysis in functional cancer research to decipher the potential of bEVs as cancer diagnostic or therapeutic biomarkers.
Collapse
Affiliation(s)
- Surbhi Mishra
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland.
| | - Mysore Vishakantegowda Tejesvi
- Biocenter Oulu, University of Oulu, Oulu, Finland; Ecology and Genetics, Faculty of Science, University of Oulu, Oulu, Finland
| | - Jenni Hekkala
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Jenni Turunen
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland; Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland
| | - Niyati Kandikanti
- Faculty of Medicine and Health Technology, University of Tampere, Tampere, Finland
| | - Anna Kaisanlahti
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Marko Suokas
- Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Sirpa Leppä
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, Helsinki, Finland
| | - Pia Vihinen
- FICAN West Cancer Centre and Department of Oncology, Turku University Hospital and University of Turku, 20521 Turku, Finland
| | - Hanne Kuitunen
- Department of Oncology, Oulu University Hospital, Oulu, Finland
| | | | - Jussi Koivunen
- Department of Medical Oncology and Radiotherapy and Medical Research Center, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Arja Jukkola
- Tampere Cancer Center, Faculty of Medicine and Health Technology, Tampere University, Tampere, Finland
| | - Ilja Kalashnikov
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, Helsinki, Finland; Research Program Unit, Applied Tumor Genomics, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Päivi Auvinen
- Cancer Center, Kuopio University Hospital, Northern Savonia Healthcare Municipality, Kuopio, Finland
| | - Okko-Sakari Kääriäinen
- Cancer Center, Kuopio University Hospital, Northern Savonia Healthcare Municipality, Kuopio, Finland
| | - T Peñate Medina
- Section Biomedical Imaging, Department of Radiology and Neuroradiology and Institute for Experimental Cancer Research, Kiel University, 24105 Kiel, Germany
| | - O Peñate Medina
- Section Biomedical Imaging, Department of Radiology and Neuroradiology and Institute for Experimental Cancer Research, Kiel University, 24105 Kiel, Germany; Lonza Netherlands B.V., 6167 RB Geleen, the Netherlands
| | - Juha Saarnio
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Sanna Meriläinen
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Tero Rautio
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Raila Aro
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Reetta Häivälä
- Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital, and University of Oulu, Oulu, Finland
| | - Juho Suojanen
- Päijät-Häme Joint Authority for Health and Wellbeing, Department of Oral and Maxillofacial Surgery, Lahti Central Hospital, 15850 Lahti, Finland; Cleft Palate and Craniofacial Centre, Department of Plastic Surgery, Helsinki University Hospital, 00029 Helsinki, Finland; Clinicum, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Mikael Laine
- Department of Abdominal Surgery, Porvoo Hospital, Hospital District of Helsinki and Uusimaa, Porvoo, Finland
| | | | - Leo Lahti
- Department of Computing, University of Turku, Turku, Finland
| | - Peeter Karihtala
- Department of Oncology, Helsinki University Hospital Comprehensive Cancer Center, University of Helsinki, Helsinki, Finland; Department of Oncology, Oulu University Hospital, Oulu, Finland
| | - Terhi S Ruuska
- Biocenter Oulu, University of Oulu, Oulu, Finland; Research Unit of Clinical Medicine, University of Oulu, Oulu, Finland; Department of Pediatrics and Adolescent Medicine, Oulu University Hospital, Oulu, Finland
| | - Justus Reunanen
- Research Unit of Translational Medicine, University of Oulu, Oulu, Finland; Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
5
|
Yang J. Insight into the potential of algorithms using AI technology as in vitro diagnostics utilizing microbial extracellular vesicles. Mol Cell Probes 2024; 78:101992. [PMID: 39580006 DOI: 10.1016/j.mcp.2024.101992] [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: 08/08/2024] [Revised: 10/21/2024] [Accepted: 11/20/2024] [Indexed: 11/25/2024]
Abstract
Recently, the microbiome has been gaining significant attention in the healthcare sector as a next-generation factor. However, there remains a substantial gap in our understanding of the fundamental mechanisms of microbes, particularly regarding the effector microbial products exchanged between the microbiota and the host. Consequently, research on microbial extracellular vesicles (MEVs) has increased. MEVs, which are nano-sized, can circulate throughout the body and penetrate the bloodstream, carrying diverse information. Consequently, they are increasingly being utilized in medical applications. Additionally, AI technologies are being utilized in medicine. The combination of MEVs and AI technology is being explored for the development of algorithm-based in vitro diagnostics (IVD). Therefore, this study aims to review the integration of MEVs and AI technology as diagnostic tools for personalized medicine. This paper reviewed the MEV-based algorithms developed by a variety of human samples and AI technology. Additionally, most of MEV-based diagnostic models showed higher clinical performance. Several important factors are crucial for accurate diagnosis. First, optimizing sample types according to specific diseases is essential. Second, AI technology with higher diagnostic power yields more accurate results. Finally, incorporating additional markers can enhance diagnostic power. However, applying this tool in situ faces several limitations, including method standardization, sample size, and analysis techniques. In the future, we anticipate that research on MEVs will advance our understanding of their role in disease and establish the foundation for precision medicine strategies.
Collapse
Affiliation(s)
- Jinho Yang
- Department of Occupational Health and Safety, Semyung University, Jecheon, Chungcheongbuk-do, Republic of Korea.
| |
Collapse
|
6
|
Chen L, Tan KML, Xu J, Mishra P, Mir SA, Gong M, Narasimhan K, Ng B, Lai JS, Tint MT, Cai S, Sadananthan SA, Michael N, Yaligar J, Velan SS, Leow MKS, Tan KH, Chan J, Meaney MJ, Chan SY, Chong YS, Eriksson JG. Exploring multi-omics and clinical characteristics linked to accelerated biological aging in Asian women of reproductive age: insights from the S-PRESTO study. Genome Med 2024; 16:128. [PMID: 39516835 PMCID: PMC11549770 DOI: 10.1186/s13073-024-01403-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Phenotypic age (PhenoAge), a widely used marker of biological aging, has been shown to be a robust predictor of all-cause mortality and morbidity in different populations. Existing studies on biological aging have primarily focused on individual domains, resulting in a lack of a comprehensive understanding of the multi-systemic dysregulation that occurs in aging. METHODS PhenoAge was evaluated based on a linear combination of chronological age (CA) and 9 clinical biomarkers in 952 multi-ethnic Asian women of reproductive age. Phenotypic age acceleration (PhenoAgeAccel), an aging biomarker, represents PhenoAge after adjusting for CA. This study conducts an in-depth association analysis of PhenoAgeAccel with clinical, nutritional, lipidomic, gut microbiome, and genetic factors. RESULTS Higher adiposity, glycaemia, plasma saturated fatty acids, kynurenine pathway metabolites, GlycA, riboflavin, nicotinamide, and insulin-like growth factor binding proteins were positively associated with PhenoAgeAccel. Conversely, a healthier diet and higher levels of pyridoxal phosphate, all-trans retinol, betaine, tryptophan, glutamine, histidine, apolipoprotein B, and insulin-like growth factors were inversely associated with PhenoAgeAccel. Lipidomic analysis found 132 lipid species linked to PhenoAgeAccel, with PC(O-36:0) showing the strongest positive association and CE(24:5) demonstrating the strongest inverse association. A genome-wide association study identified rs9864994 as the top genetic variant (P = 5.69E-07) from the ZDHHC19 gene. Gut microbiome analysis revealed that Erysipelotrichaceae UCG-003 and Bacteroides vulgatus were inversely associated with PhenoAgeAccel. Integrative network analysis of aging-related factors underscored the intricate links among clinical, nutritional and lipidomic variables, such as positive associations between kynurenine pathway metabolites, amino acids, adiposity, and insulin resistance. Furthermore, potential mediation effects of blood biomarkers related to inflammation, immune response, and nutritional and energy metabolism were observed in the associations of diet, adiposity, genetic variants, and gut microbial species with PhenoAgeAccel. CONCLUSIONS Our findings provide a comprehensive analysis of aging-related factors across multiple platforms, delineating their complex interconnections. This study is the first to report novel signatures in lipidomics, gut microbiome and blood biomarkers specifically associated with PhenoAgeAccel. These insights are invaluable in understanding the molecular and metabolic mechanisms underlying biological aging and shed light on potential interventions to mitigate accelerated biological aging by targeting modifiable factors.
Collapse
Affiliation(s)
- Li Chen
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| | - Karen Mei-Ling Tan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore.
| | - Jia Xu
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Priti Mishra
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sartaj Ahmad Mir
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Min Gong
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kothandaraman Narasimhan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Bryan Ng
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jun Shi Lai
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mya Thway Tint
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Shirong Cai
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suresh Anand Sadananthan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Navin Michael
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jadegoud Yaligar
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sambasivam Sendhil Velan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Medicine and Human Potential Translational Research Programme. Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Melvin Khee Shing Leow
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kok Hian Tan
- Duke-NUS Medical School, Singapore, Singapore
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Jerry Chan
- Duke-NUS Medical School, Singapore, Singapore
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Michael J Meaney
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Sackler Program for Epigenetics & Psychobiology, McGill University, Montréal, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montréal, Canada
| | - Shiao-Yng Chan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yap Seng Chong
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Johan G Eriksson
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Folkhalsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| |
Collapse
|
7
|
Song Y, Shi M, Wang Y. Deciphering the role of host-gut microbiota crosstalk via diverse sources of extracellular vesicles in colorectal cancer. Mol Med 2024; 30:200. [PMID: 39501131 PMCID: PMC11536884 DOI: 10.1186/s10020-024-00976-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 10/26/2024] [Indexed: 11/09/2024] Open
Abstract
Colorectal cancer is the most common type of cancer in the digestive system and poses a major threat to human health. The gut microbiota has been found to be a key factor influencing the development of colorectal cancer. Extracellular vesicles are important mediators of intercellular communication. Not only do they regulate life activities within the same individual, but they have also been found in recent years to be important mediators of communication between different species, such as the gut microbiota and the host. Their preventive, diagnostic, and therapeutic value in colorectal cancer is being explored. The aim of this review is to provide insights into the complex interactions between host and gut microbiota, particularly those mediated through extracellular vesicles, and how these interactions affect colorectal cancer development. In addition, the potential of extracellular vesicles from various body fluids as biomarkers was evaluated. Finally, we discuss the potential, challenges, and future research directions of extracellular vesicles in their application to colorectal cancer. Overall, extracellular vesicles have great potential for application in medical processes related to colorectal cancer, but their isolation and characterization techniques, intercellular communication mechanisms, and the effectiveness of their clinical application require further research and exploration.
Collapse
Affiliation(s)
- Yun Song
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai, 200336, PR China
| | - Min Shi
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai, 200336, PR China.
- Key Laboratory for Translational Research and Innovative Therapeutics of Gastrointestinal Oncology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, PR China.
| | - Yugang Wang
- Department of Gastroenterology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, 1111 Xianxia Road, Shanghai, 200336, PR China.
| |
Collapse
|
8
|
Wu Q, Kan J, Fu C, Liu X, Cui Z, Wang S, Le Y, Li Z, Liu Q, Zhang Y, Du J. Insights into the unique roles of extracellular vesicles for gut health modulation: Mechanisms, challenges, and perspectives. CURRENT RESEARCH IN MICROBIAL SCIENCES 2024; 7:100301. [PMID: 39525958 PMCID: PMC11550031 DOI: 10.1016/j.crmicr.2024.100301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Extracellular vesicles (EVs), which play significant regulatory roles in maintaining homeostasis and influencing immune responses, significantly impact gut microbiota composition and function, affecting overall gut health. Despite considerable progress, there are still knowledge gaps regarding the mechanisms by which EVs, including plant-derived EVs (PDEVs), animal-derived EVs (ADEVs), and microbiota-derived EVs (MDEVs), modulate gut health. This review delves into the roles and mechanisms of EVs from diverse sources in regulating gut health, focusing on their contributions to maintaining epithelial barrier integrity, facilitating tissue healing, eliciting immune responses, controlling pathogens, and shaping microbiota. We emphasize open challenges and future perspectives for harnessing EVs in the modulation of gut health to gain a deeper understanding of their roles and impact. Importantly, a comprehensive research framework is presented to steer future investigations into the roles and implications of EVs on gut health, facilitating a more profound comprehension of this emerging field.
Collapse
Affiliation(s)
- Qiming Wu
- Nutrilite Health Institute, Shanghai 200031, China
| | - Juntao Kan
- Nutrilite Health Institute, Shanghai 200031, China
| | - Caili Fu
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Xin Liu
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Zhengying Cui
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Sixu Wang
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Yi Le
- Department of Food Science and Technology, National University of Singapore Suzhou Research Institute, Suzhou 215123, China
| | - Zhanming Li
- Department of Food Quality and Safety, Jiangsu University of Science and Technology, Zhenjiang 212100, China
| | - Qin Liu
- Centre for Chinese Medicine Drug Development Limited, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China
| | - Yuyu Zhang
- Key Laboratory of Geriatric Nutrition and Health, Beijing Technology and Business University, Beijing 100048, China
| | - Jun Du
- Nutrilite Health Institute, Shanghai 200031, China
| |
Collapse
|
9
|
Yang C, Wusigale, You L, Li X, Kwok LY, Chen Y. Inflammation, Gut Microbiota, and Metabolomic Shifts in Colorectal Cancer: Insights from Human and Mouse Models. Int J Mol Sci 2024; 25:11189. [PMID: 39456970 PMCID: PMC11508446 DOI: 10.3390/ijms252011189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/09/2024] [Accepted: 10/13/2024] [Indexed: 10/28/2024] Open
Abstract
Colorectal cancer (CRC) arises from aberrant mutations in colorectal cells, frequently linked to chronic inflammation. This study integrated human gut metagenome analysis with an azoxymethane and dextran sulfate sodium-induced CRC mouse model to investigate the dynamics of inflammation, gut microbiota, and metabolomic profiles throughout tumorigenesis. The analysis of stool metagenome data from 30 healthy individuals and 40 CRC patients disclosed a significant escalation in both gut microbiota diversity and abundance in CRC patients compared to healthy individuals (p < 0.05). Marked structural disparities were identified between the gut microbiota of healthy individuals and those with CRC (p < 0.05), characterized by elevated levels of clostridia and diminished bifidobacteria in CRC patients (p < 0.05). In the mouse model, CRC mice exhibited distinct gut microbiota structures and metabolite signatures at early and advanced tumor stages, with subtle variations noted during the intermediate phase. Additionally, inflammatory marker levels increased progressively during tumor development in CRC mice, in contrast to their stable levels in healthy counterparts. These findings suggest that persistent inflammation might precipitate gut dysbiosis and altered microbial metabolism. Collectively, this study provides insights into the interplay between inflammation, gut microbiota, and metabolite changes during CRC progression, offering potential biomarkers for diagnosis. While further validation with larger cohorts is warranted, the data obtained support the development of CRC prevention and diagnosis strategies.
Collapse
Affiliation(s)
- Chengcong Yang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; (C.Y.); (W.); (L.Y.); (X.L.); (L.-Y.K.)
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Wusigale
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; (C.Y.); (W.); (L.Y.); (X.L.); (L.-Y.K.)
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lijun You
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; (C.Y.); (W.); (L.Y.); (X.L.); (L.-Y.K.)
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Xiang Li
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; (C.Y.); (W.); (L.Y.); (X.L.); (L.-Y.K.)
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; (C.Y.); (W.); (L.Y.); (X.L.); (L.-Y.K.)
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Yongfu Chen
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot 010018, China; (C.Y.); (W.); (L.Y.); (X.L.); (L.-Y.K.)
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| |
Collapse
|
10
|
Wyatt M, Choudhury A, Von Dohlen G, Heileson JL, Forsse JS, Rajakaruna S, Zec M, Tfaily MM, Greathouse L. Randomized control trial of moderate dose vitamin D alters microbiota stability and metabolite networks in healthy adults. Microbiol Spectr 2024; 12:e0008324. [PMID: 39189761 PMCID: PMC11448053 DOI: 10.1128/spectrum.00083-24] [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: 01/10/2024] [Accepted: 07/10/2024] [Indexed: 08/28/2024] Open
Abstract
Evidence indicates that both vitamin D and the gut microbiome are involved in the process of colon carcinogenesis. However, it is unclear what effects supplemental vitamin D3 has on the gut microbiome and its metabolites in healthy adults. We conducted a double-blind, randomized, placebo-controlled trial to identify the acute and long-term microbiota structural and metabolite changes that occur in response to a moderate dose (4,000 IU) of vitamin D3 for 12 weeks in healthy adults. Our results demonstrated a significant increase in serum 25-hydroxy-vitamin D (25(OH)D) in the treatment group compared to placebo (P < 0.0001). Vitamin D3 significantly increased compositional similarity (P < 0.0001) in the treatment group, and enriched members of the Bifidobacteriaceae family. We also identified a significant inverse relationship between the percent change in serum 25(OH)D and microbial stability in the treatment group (R = -0.52, P < 0.019). Furthermore, vitamin D3 supplementation resulted in notable metabolic shifts, in addition to resulting in a drastic rewiring of key gut microbial-metabolic associations. In conclusion, we show that a moderate dose of vitamin D3 among healthy adults has unique acute and persistent effects on the fecal microbiota, and suggest novel mechanisms by which vitamin D may affect the host-microbiota relationship. IMPORTANCE Preventative measures to reduce the rise in early-onset colorectal cancer are of critical need. Both vitamin D, dietary and serum levels, and the gut microbiome are implicated in the etiology of colorectal cancer. By understanding the intimate relationship between vitamin D, the gut microbiome, and its metabolites, we may be able to identify key mechanisms that can be targeted for intervention, including inflammation and metabolic dysfunction. Furthermore, the similarity of vitamin D to cholesterol, which is metabolized by the gut microbiome, gives precedence to its ability to produce metabolites that can be further studied and leveraged for controlling colorectal cancer incidence and mortality.
Collapse
Affiliation(s)
- Madhur Wyatt
- Human Health Performance and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
| | - Ankan Choudhury
- Human Science and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
| | - Gabriella Von Dohlen
- Human Science and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
| | - Jeffery L. Heileson
- Human Health Performance and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
- Nutrition Services Division, Walter Reed National Military Medical Center, Bethesda, Maryland, USA
| | - Jeffrey S. Forsse
- Human Health Performance and Recreation, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
- Department of Biology, Baylor University, Waco, Texas, USA
| | - Sumudu Rajakaruna
- Department of Environmental Science, University of Arizona, Tucson, Arizona, USA
- BIO5 Institute, The University of Arizona, Tucson, Arizona, USA
| | - Manja Zec
- Department of Environmental Science, University of Arizona, Tucson, Arizona, USA
- Colorado Program for Musculoskeletal Research, Department of Orthopedics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Malak M. Tfaily
- Department of Environmental Science, University of Arizona, Tucson, Arizona, USA
- BIO5 Institute, The University of Arizona, Tucson, Arizona, USA
| | - Leigh Greathouse
- Human Science and Design, Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
- Department of Biology, Baylor University, Waco, Texas, USA
| |
Collapse
|
11
|
Liang A, Korani L, Yeung CLS, Tey SK, Yam JWP. The emerging role of bacterial extracellular vesicles in human cancers. J Extracell Vesicles 2024; 13:e12521. [PMID: 39377479 PMCID: PMC11460218 DOI: 10.1002/jev2.12521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 08/16/2024] [Accepted: 09/16/2024] [Indexed: 10/09/2024] Open
Abstract
Bacterial extracellular vesicles (BEVs) have emerged as pivotal mediators between bacteria and host. In addition to being crucial players in host homeostasis, they have recently been implicated in disease pathologies such as cancer. Hence, the study of BEVs represents an intriguing and rapidly evolving field with substantial translational potential. In this review, we briefly introduce the fundamentals of BEV characteristics, cargo and biogenesis. We emphatically summarize the current relationship between BEVs across various cancer types, illustrating their role in tumorigenesis, treatment responses and patient survival. We further discuss the inherent advantages of BEVs, such as stability, abundance and specific cargo profiles, that make them attractive candidates for non-invasive diagnostic and prognostic approaches. The review also explores the potential of BEVs as a strategy for cancer therapy, considering their ability to deliver therapeutic agents, modulate the tumour microenvironment (TME) and elicit immunomodulatory responses. Understanding the clinical significance of BEVs may lead to the development of better-targeted and personalized treatment strategies. This comprehensive review evaluates the current progress surrounding BEVs and poses questions to encourage further research in this emerging field to harness the benefits of BEVs for their full potential in clinical applications against cancer.
Collapse
Affiliation(s)
- Aijun Liang
- Department of Hepatobiliary Surgery IIZhujiang Hospital, Southern Medical UniversityGuangzhouGuangdongChina
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Lavisha Korani
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Cherlie Lot Sum Yeung
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Sze Keong Tey
- Department of Surgery, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| | - Judy Wai Ping Yam
- Department of Hepatobiliary Surgery IIZhujiang Hospital, Southern Medical UniversityGuangzhouGuangdongChina
- Department of Pathology, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong KongHong KongHong Kong
| |
Collapse
|
12
|
Fu J, Li G, Li X, Song S, Cheng L, Rui B, Jiang L. Gut commensal Alistipes as a potential pathogenic factor in colorectal cancer. Discov Oncol 2024; 15:473. [PMID: 39331213 PMCID: PMC11436608 DOI: 10.1007/s12672-024-01393-3] [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: 08/14/2024] [Accepted: 09/24/2024] [Indexed: 09/28/2024] Open
Abstract
Although previous research has shown that inflammation is associated with development of colorectal cancer (CRC), questions remain about whether inflammatory factor-secreting bacteria play a crucial role in CRC development. The potential role of gut microbiota in secreting inflammatory factors involved in the carcinogenesis of CRC among Chinese patients was explored in this study. 16S rRNA sequencing was utilized to evaluate the distinct microbial characteristics between patients with CRC and colorectal adenoma. The serum levels of TNF-α, IL-6 and IL-10 were measured using Enzyme-linked immunosorbent assay (ELISA), while the expression of LRG1 and TGF-β1 in tissues was evaluated by immunohistochemistry. The correlation between gut microbiota and inflammatory factor signaling was analyzed. Compared with the adenoma group, CRC patients exhibit distinct pathologies. Moreover, elevated levels of CEA, erythrocytes and haemoglobin in the blood of CRC patients were found. In addition, CRC patients have significantly higher levels of TNF-α, IL-6, IL-10, LRG1 and TGF-β1. Spearman correlation analysis revealed that LRG1 was positively related to IL-6 and TNF-α, respectively. The correlation analysis results of TGF-β1 were consistent with the above. The abundance of Blautia and Streptococcus was lower in CRC patients, while the relative abundance of Alistipes, Peptostreptococcus and Porphyromonas was significantly elevated. Moreover, positive correlations between Alistipes and inflammatory factor signaling were also found. Our results suggest that gut commensal Alistipes is a key bacterium with pro-inflammatory properties in the CRC carcinogenesis. TNF-α and IL-6 associated with Alistipes might activate LRG1/TGF-β1 signaling which contributed to the carcinogenesis of CRC.
Collapse
Affiliation(s)
- Jingjing Fu
- Department of Pharmacy, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
- Anhui No.2 Provincial People's Hospital Clinical College, Anhui Medical University, Hefei, 230041, Anhui, China
| | - Guangyao Li
- Department of Gastrointestinal Surgery, The Second People's Hospital of Wuhu, Wuhu, 241000, Anhui, China
| | - Xiaoping Li
- Department of Gastroenterology Department 1, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
| | - Shasha Song
- Department of Gastroenterology, the Second Hospital of Anhui Medical University, Hefei, 230601, Anhui, China
| | - Lijuan Cheng
- Department of Pharmacy, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
- Anhui No.2 Provincial People's Hospital Clinical College, Anhui Medical University, Hefei, 230041, Anhui, China
| | - Beibei Rui
- Department of Pharmacy, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China
- Anhui No.2 Provincial People's Hospital Clinical College, Anhui Medical University, Hefei, 230041, Anhui, China
| | - Lei Jiang
- Department of Pharmacy, Anhui No.2 Provincial People's Hospital, Hefei, 230041, Anhui, China.
- Anhui No.2 Provincial People's Hospital Clinical College, Anhui Medical University, Hefei, 230041, Anhui, China.
| |
Collapse
|
13
|
Kalluri R. The biology and function of extracellular vesicles in immune response and immunity. Immunity 2024; 57:1752-1768. [PMID: 39142276 PMCID: PMC11401063 DOI: 10.1016/j.immuni.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 01/02/2024] [Accepted: 07/15/2024] [Indexed: 08/16/2024]
Abstract
Extracellular vesicles (EVs), such as ectosomes and exosomes, contain DNA, RNA, proteins and are encased in a phospholipid bilayer. EVs provide intralumenal cargo for delivery into the cytoplasm of recipient cells with an impact on the function of immune cells, in part because their biogenesis can also intersect with antigen processing and presentation. Motile EVs from activated immune cells may increase the frequency of immune synapses on recipient cells in a proximity-independent manner for local and long-distance modulation of systemic immunity in inflammation, autoimmunity, organ fibrosis, cancer, and infections. Natural and engineered EVs exhibit the ability to impact innate and adaptive immunity and are entering clinical trials. EVs are likely a component of an optimally functioning immune system, with the potential to serve as immunotherapeutics. Considering the evolving evidence, it is possible that EVs could be the original primordial organic units that preceded the creation of the first cell.
Collapse
Affiliation(s)
- Raghu Kalluri
- Department of Cancer Biology, Metastasis Research Center, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Bioengineering, Rice University, Houston, TX, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
14
|
Khalili L, Park G, Nagpal R, Salazar G. The Role of Akkermansia muciniphila on Improving Gut and Metabolic Health Modulation: A Meta-Analysis of Preclinical Mouse Model Studies. Microorganisms 2024; 12:1627. [PMID: 39203469 PMCID: PMC11356609 DOI: 10.3390/microorganisms12081627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Akkermansia muciniphila (A. muciniphila) and its derivatives, including extracellular vesicles (EVs) and outer membrane proteins, are recognized for enhancing intestinal balance and metabolic health. However, the mechanisms of Akkermansia muciniphila's action and its effects on the microbiome are not well understood. In this study, we examined the influence of A. muciniphila and its derivatives on gastrointestinal (GI) and metabolic disorders through a meta-analysis of studies conducted on mouse models. A total of 39 eligible studies were identified through targeted searches on PubMed, Web of Science, Science Direct, and Embase until May 2024. A. muciniphila (alive or heat-killed) and its derivatives positively affected systemic and gut inflammation, liver enzyme level, glycemic response, and lipid profiles. The intervention increased the expression of tight-junction proteins in the gut, improving gut permeability in mouse models of GI and metabolic disorders. Regarding body weight, A. muciniphila and its derivatives prevented weight loss in animals with GI disorders while reducing body weight in mice with metabolic disorders. Sub-group analysis indicated that live bacteria had a more substantial effect on most analyzed biomarkers. Gut microbiome analysis using live A. muciniphila identified a co-occurrence cluster, including Desulfovibrio, Family XIII AD3011 group, and Candidatus Saccharimonas. Thus, enhancing the intestinal abundance of A. muciniphila and its gut microbial clusters may provide more robust health benefits for cardiometabolic, and age-related diseases compared with A. muciniphila alone. The mechanistic insight elucidated here will pave the way for further exploration and potential translational applications in human health.
Collapse
Affiliation(s)
- Leila Khalili
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Gwoncheol Park
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Ravinder Nagpal
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
| | - Gloria Salazar
- Department of Health, Nutrition and Food Sciences, Florida State University, Tallahassee, FL 32306, USA; (L.K.); (G.P.); (R.N.)
- Center for Advancing Exercise and Nutrition Research on Aging (CAENRA), Florida State University, Tallahassee, FL 32306, USA
| |
Collapse
|
15
|
Lv H, Guo M, Guo C, He K. The Interrelationships between Cytokines and Schizophrenia: A Systematic Review. Int J Mol Sci 2024; 25:8477. [PMID: 39126046 PMCID: PMC11313682 DOI: 10.3390/ijms25158477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/26/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024] Open
Abstract
Schizophrenia (SCZ) imposes a significant burden on patients and their families because of its high prevalence rate and disabling nature. Given the lack of definitive conclusions regarding its pathogenesis, physicians heavily rely on patients' subjective symptom descriptions for diagnosis because reliable diagnostic biomarkers are currently unavailable. The role of the inflammatory response in the pathogenesis of SCZ has been supported by some studies. The findings of these studies showed abnormal changes in the levels of inflammatory factors, such as cytokines (CKs), in both peripheral blood and cerebrospinal fluid (CSF) among individuals affected by SCZ. The findings imply that inflammatory factors could potentially function as risk indicators for the onset of SCZ. Consequently, researchers have directed their attention towards investigating the potential utility of CKs as viable biomarkers for diagnosing SCZ. Extracellular vesicles (EVs) containing disease-specific components exhibit remarkable stability and abundance, making them promising candidates for biomarker discovery across various diseases. CKs encapsulated within EVs secreted by immune cells offer valuable insights into disease progression. This review presents a comprehensive analysis summarizing the relationship between CKs and SCZ and emphasizes the vital role of CKs encapsulated within EVs in the pathogenesis and development of SCZ.
Collapse
Affiliation(s)
- Haibing Lv
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao 028000, China; (H.L.); (C.G.)
| | - Meng Guo
- Finance Office, Inner Mongolia Minzu University, Tongliao 028000, China;
| | - Chuang Guo
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao 028000, China; (H.L.); (C.G.)
| | - Kuanjun He
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao 028000, China; (H.L.); (C.G.)
| |
Collapse
|
16
|
Zhang H, Lin Y, Li S, Bi J, Zeng J, Mo C, Xu S, Jia B, Lu Y, Liu C, Liu Z. Effects of bacterial extracellular vesicles derived from oral and gastrointestinal pathogens on systemic diseases. Microbiol Res 2024; 285:127788. [PMID: 38833831 DOI: 10.1016/j.micres.2024.127788] [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: 03/04/2024] [Revised: 04/18/2024] [Accepted: 05/26/2024] [Indexed: 06/06/2024]
Abstract
Oral microbiota and gastrointestinal microbiota, the two largest microbiomes in the human body, are closely correlated and frequently interact through the oral-gut axis. Recent research has focused on the roles of these microbiomes in human health and diseases. Under normal conditions, probiotics and commensal bacteria can positively impact health. However, altered physiological states may induce dysbiosis, increasing the risk of pathogen colonization. Studies suggest that oral and gastrointestinal pathogens contribute not only to localized diseases at their respective colonized sites but also to the progression of systemic diseases. However, the mechanisms by which bacteria at these local sites are involved in systemic diseases remain elusive. In response to this gap, the focus has shifted to bacterial extracellular vesicles (BEVs), which act as mediators of communication between the microbiota and the host. Numerous studies have reported the targeted delivery of bacterial pathogenic substances from the oral cavity and the gastrointestinal tract to distant organs via BEVs. These pathogenic components subsequently elicit specific cellular responses in target organs, thereby mediating the progression of systemic diseases. This review aims to elucidate the extensive microbial communication via the oral-gut axis, summarize the types and biogenesis mechanisms of BEVs, and highlight the translocation pathways of oral and gastrointestinal BEVs in vivo, as well as the impacts of pathogens-derived BEVs on systemic diseases.
Collapse
Affiliation(s)
- Han Zhang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yunhe Lin
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Siwei Li
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiaming Bi
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Jiawei Zeng
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chuzi Mo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Bo Jia
- Department of Oral and Maxillofacial Surgery, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Yu Lu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Chengxia Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China
| | - Zhongjun Liu
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong 510515, China.
| |
Collapse
|
17
|
Di Paola FJ, Alquati C, Conti G, Calafato G, Turroni S, D'Amico F, Ceccarelli C, Buttitta F, Bernardi A, Cuicchi D, Poggioli G, Turchetti D, Ferrari S, Cannizzaro R, Realdon S, Brigidi P, Ricciardiello L. Interplay between WNT/PI3K-mTOR axis and the microbiota in APC-driven colorectal carcinogenesis: data from a pilot study and possible implications for CRC prevention. J Transl Med 2024; 22:631. [PMID: 38970018 PMCID: PMC11227240 DOI: 10.1186/s12967-024-05305-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 05/16/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND Wnt/β-catenin signalling impairment accounts for 85% of colorectal cancers (CRCs), including sporadic and familial adenomatous polyposis (FAP) settings. An altered PI3K/mTOR pathway and gut microbiota also contribute to CRC carcinogenesis. We studied the interplay between the two pathways and the microbiota composition within each step of CRC carcinogenesis. METHODS Proteins and target genes of both pathways were analysed by RT-qPCR and IHC in tissues from healthy faecal immunochemical test positive (FIT+, n = 17), FAP (n = 17) and CRC (n = 15) subjects. CRC-related mutations were analysed through NGS and Sanger. Oral, faecal and mucosal microbiota was profiled by 16 S rRNA-sequencing. RESULTS We found simultaneous hyperactivation of Wnt/β-catenin and PI3K/mTOR pathways in FAP-lesions compared to CRCs. Wnt/β-catenin molecular markers positively correlated with Clostridium_sensu_stricto_1 and negatively with Bacteroides in FAP faecal microbiota. Alistipes, Lachnospiraceae, and Ruminococcaceae were enriched in FAP stools and adenomas, the latter also showing an overabundance of Lachnoclostridium, which positively correlated with cMYC. In impaired-mTOR-mutated CRC tissues, p-S6R correlated with Fusobacterium and Dialister, the latter also confirmed in the faecal-ecosystem. CONCLUSIONS Our study reveals an interplay between Wnt/β-catenin and PI3K/mTOR, whose derangement correlates with specific microbiota signatures in FAP and CRC patients, and identifies new potential biomarkers and targets to improve CRC prevention, early adenoma detection and treatment.
Collapse
Affiliation(s)
| | - Chiara Alquati
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
- Centre for Applied Biomedical Research (CRBA), University of Bologna, Bologna, Italy
| | - Gabriele Conti
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Giulia Calafato
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Silvia Turroni
- Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Federica D'Amico
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Claudio Ceccarelli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | - Alice Bernardi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Dajana Cuicchi
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Gilberto Poggioli
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Daniela Turchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Simona Ferrari
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Renato Cannizzaro
- Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Stefano Realdon
- Oncological Gastroenterology, Centro di Riferimento Oncologico di Aviano (CRO) IRCCS, Aviano, Italy
| | - Patrizia Brigidi
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Luigi Ricciardiello
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy.
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy.
| |
Collapse
|
18
|
de Wit S, Geerlings L, Shi C, Dronkers J, Schouten EM, Blancke G, Andries V, Yntema T, Meijers WC, Koonen DPY, Vereecke L, Silljé HHW, Aboumsallem JP, de Boer RA. Heart failure-induced microbial dysbiosis contributes to colonic tumour formation in mice. Cardiovasc Res 2024; 120:612-622. [PMID: 38400709 DOI: 10.1093/cvr/cvae038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 12/18/2023] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
AIMS Heart failure (HF) and cancer are the leading causes of death worldwide. Epidemiological studies revealed that HF patients are prone to develop cancer. Preclinical studies provided some insights into this connection, but the exact mechanisms remain elusive. In colorectal cancer (CRC), gut microbial dysbiosis is linked to cancer progression and recent studies have shown that HF patients display microbial dysbiosis. This current study focussed on the effects of HF-induced microbial dysbiosis on colonic tumour formation. METHODS AND RESULTS C57BL/6J mice were subjected to myocardial infarction (MI), with sham surgery as control. After six weeks faeces were collected, processed for 16 s rRNA sequencing, and pooled for faecal microbiota transplantation. CRC tumour growth was provoked in germ-free mice by treating them with Azoxymethane/Dextran sodium sulphate. The CRC mice were transplanted with faeces from MI or sham mice. MI-induced HF resulted in microbial dysbiosis, characterized by a decreased α-diversity and microbial alterations on the genus level, several of which have been associated with CRC. We then performed faecal microbiota transplantation with faeces from HF mice in CRC mice, which resulted in a higher endoscopic disease score and an increase in the number of tumours in CRC mice. CONCLUSION We demonstrated that MI-induced HF contributes to colonic tumour formation by altering the gut microbiota composition, providing a mechanistic explanation for the observed association between HF and increased risk for cancer. Targeting the microbiome may present as a tool to mitigate HF-associated co-morbidities, especially cancer.
Collapse
Affiliation(s)
- Sanne de Wit
- Department of Cardiology, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Lotte Geerlings
- Department of Cardiology, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Canxia Shi
- Department of Cardiology, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
- Thorax Center, Department of Cardiology, Erasmus MC, Cardiovascular Institute, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - Just Dronkers
- Department of Cardiology, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Elisabeth M Schouten
- Department of Cardiology, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Gillian Blancke
- Department of Internal Medicine and Paediatrics, Ghent University, 9000, Ghent, Belgium
- Host-Microbiota Interaction Lab, VIB Center for Inflammation Research, 9052, Ghent, Belgium
| | - Vanessa Andries
- Department of Internal Medicine and Paediatrics, Ghent University, 9000, Ghent, Belgium
- Host-Microbiota Interaction Lab, VIB Center for Inflammation Research, 9052, Ghent, Belgium
| | - Tess Yntema
- Department of Paediatrics, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Wouter C Meijers
- Thorax Center, Department of Cardiology, Erasmus MC, Cardiovascular Institute, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
- Department of Cardiology, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Debby P Y Koonen
- Department of Paediatrics, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Lars Vereecke
- Department of Internal Medicine and Paediatrics, Ghent University, 9000, Ghent, Belgium
- Host-Microbiota Interaction Lab, VIB Center for Inflammation Research, 9052, Ghent, Belgium
| | - Herman H W Silljé
- Department of Cardiology, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Joseph-Pierre Aboumsallem
- Thorax Center, Department of Cardiology, Erasmus MC, Cardiovascular Institute, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
- Department of Cardiology, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| | - Rudolf A de Boer
- Thorax Center, Department of Cardiology, Erasmus MC, Cardiovascular Institute, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
- Department of Cardiology, University Medical Center Groningen, Groningen, 9713 AV, The Netherlands
| |
Collapse
|
19
|
Smith SF, Brewer DS, Hurst R, Cooper CS. Applications of Urinary Extracellular Vesicles in the Diagnosis and Active Surveillance of Prostate Cancer. Cancers (Basel) 2024; 16:1717. [PMID: 38730670 PMCID: PMC11083542 DOI: 10.3390/cancers16091717] [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: 04/14/2024] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/13/2024] Open
Abstract
Prostate cancer is the most common non-cutaneous cancer among men in the UK, causing significant health and economic burdens. Diagnosis and risk prognostication can be challenging due to the genetic and clinical heterogeneity of prostate cancer as well as uncertainties in our knowledge of the underlying biology and natural history of disease development. Urinary extracellular vesicles (EVs) are microscopic, lipid bilayer defined particles released by cells that carry a variety of molecular cargoes including nucleic acids, proteins and other molecules. Urine is a plentiful source of prostate-derived EVs. In this narrative review, we summarise the evidence on the function of urinary EVs and their applications in the evolving field of prostate cancer diagnostics and active surveillance. EVs are implicated in the development of all hallmarks of prostate cancer, and this knowledge has been applied to the development of multiple diagnostic tests, which are largely based on RNA and miRNA. Common gene probes included in multi-probe tests include PCA3 and ERG, and the miRNAs miR-21 and miR-141. The next decade will likely bring further improvements in the diagnostic accuracy of biomarkers as well as insights into molecular biological mechanisms of action that can be translated into opportunities in precision uro-oncology.
Collapse
Affiliation(s)
- Stephanie F. Smith
- Metabolic Health Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK (C.S.C.)
- Department of Urology, Norfolk and Norwich University Hospitals, Norwich NR4 7UY, UK
| | - Daniel S. Brewer
- Metabolic Health Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK (C.S.C.)
| | - Rachel Hurst
- Metabolic Health Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK (C.S.C.)
| | - Colin S. Cooper
- Metabolic Health Research Centre, Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK (C.S.C.)
| |
Collapse
|
20
|
Guo C, Bai Y, Li P, He K. The emerging roles of microbiota-derived extracellular vesicles in psychiatric disorders. Front Microbiol 2024; 15:1383199. [PMID: 38650872 PMCID: PMC11033316 DOI: 10.3389/fmicb.2024.1383199] [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: 02/07/2024] [Accepted: 03/28/2024] [Indexed: 04/25/2024] Open
Abstract
Major depressive disorder, schizophrenia, and bipolar disorder are three major psychiatric disorders that significantly impact the well-being and overall health of patients. Some researches indicate that abnormalities in the gut microbiota can trigger certain psychiatric diseases. Microbiota-derived extracellular vesicles have the ability to transfer bioactive compounds into host cells, altering signaling and biological processes, ultimately influencing the mental health and illness of the host. This review aims to investigate the emerging roles of microbiota-derived extracellular vesicles in these three major psychiatric disorders and discusses their roles as diagnostic biomarkers and therapies for these psychiatric disorders.
Collapse
Affiliation(s)
- Chuang Guo
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, China
| | - Yulong Bai
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, China
| | - Pengfei Li
- Affiliated Hospital of Inner Mongolia Minzu University, Tongliao, China
| | - Kuanjun He
- College of Life Sciences and Food Engineering, Inner Mongolia Minzu University, Tongliao, China
| |
Collapse
|
21
|
Ravi Kumar R, Ndiaye MM, Haddad I, Vinh J, Verdier Y. ChipFilter: Microfluidic-Based Comprehensive Sample Preparation Methodology for Microbial Consortia. J Proteome Res 2024; 23:869-880. [PMID: 38353246 PMCID: PMC10913871 DOI: 10.1021/acs.jproteome.3c00288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 01/04/2024] [Accepted: 01/19/2024] [Indexed: 03/02/2024]
Abstract
The metaproteomic approach is an attractive way to describe a microbiome at the functional level, allowing the identification and quantification of proteins across a broad dynamic range as well as the detection of post-translational modifications. However, it remains relatively underutilized, mainly due to technical challenges that should be addressed, including the complexity of extracting proteins from heterogeneous microbial communities. Here, we show that a ChipFilter microfluidic device coupled to a liquid chromatography tandem mass spectrometry (LC-MS/MS) setup can be successfully used for the identification of microbial proteins. Using cultures of Escherichia coli, Bacillus subtilis, and Saccharomyces cerevisiae, we have shown that it is possible to directly lyse the cells and digest the proteins in the ChipFilter to allow the identification of a higher number of proteins and peptides than that by standard protocols, even at low cell density. The peptides produced are overall longer after ChipFilter digestion but show no change in their degree of hydrophobicity. Analysis of a more complex mixture of 17 species from the gut microbiome showed that the ChipFilter preparation was able to identify and estimate the amounts of 16 of these species. These results show that ChipFilter can be used for the proteomic study of microbiomes, particularly in the case of a low volume or cell density. The mass spectrometry data have been deposited on the ProteomeXchange Consortium via the PRIDE partner repository with the data set identifier PXD039581.
Collapse
Affiliation(s)
- Ranjith
Kumar Ravi Kumar
- Spectrométrie de
Masse Biologique et Protéomique, LPC, UMR ESPCI CNRS 8249, 10 rue Vauquelin, F-75005 Paris, France
| | - Massamba Mbacke Ndiaye
- Spectrométrie de
Masse Biologique et Protéomique, LPC, UMR ESPCI CNRS 8249, 10 rue Vauquelin, F-75005 Paris, France
| | - Iman Haddad
- Spectrométrie de
Masse Biologique et Protéomique, LPC, UMR ESPCI CNRS 8249, 10 rue Vauquelin, F-75005 Paris, France
| | - Joelle Vinh
- Spectrométrie de
Masse Biologique et Protéomique, LPC, UMR ESPCI CNRS 8249, 10 rue Vauquelin, F-75005 Paris, France
| | - Yann Verdier
- Spectrométrie de
Masse Biologique et Protéomique, LPC, UMR ESPCI CNRS 8249, 10 rue Vauquelin, F-75005 Paris, France
| |
Collapse
|
22
|
Kim MG, Kim S, Jeon JY, Moon SJ, Kwak YG, Na JY, Lee S, Park KM, Kim HJ, Lee SM, Choi SY, Shin KH. Profiling of endogenous metabolites and changes in intestinal microbiota distribution after GEN-001 ( Lactococcus lactis) administration. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2024; 28:153-164. [PMID: 38414398 PMCID: PMC10902589 DOI: 10.4196/kjpp.2024.28.2.153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/18/2023] [Accepted: 12/22/2023] [Indexed: 02/29/2024]
Abstract
This study aimed to identify metabolic biomarkers and investigate changes in intestinal microbiota in the feces of healthy participants following administration of Lactococcus lactis GEN-001. GEN-001 is a single-strain L. lactis strain isolated from the gut of a healthy human volunteer. The study was conducted as a parallel, randomized, phase 1, open design trial. Twenty healthy Korean males were divided into five groups according to the GEN-001 dosage and dietary control. Groups A, B, C, and D1 received 1, 3, 6, and 9 GEN-001 capsules (1 × 1011 colony forming units), respectively, without dietary adjustment, whereas group D2 received 9 GEN-001 capsules with dietary adjustment. All groups received a single dose. Fecal samples were collected 2 days before GEN-001 administration to 7 days after for untargeted metabolomics and gut microbial metagenomic analyses; blood samples were collected simultaneously for immunogenicity analysis. Levels of phenylalanine, tyrosine, cholic acid, deoxycholic acid, and tryptophan were significantly increased at 5-6 days after GEN-001 administration when compared with predose levels. Compared with predose, the relative abundance (%) of Parabacteroides and Alistipes significantly decreased, whereas that of Lactobacillus and Lactococcus increased; Lactobacillus and tryptophan levels were negatively correlated. A single administration of GEN-001 shifted the gut microbiota in healthy volunteers to a more balanced state as evidenced by an increased abundance of beneficial bacteria, including Lactobacillus, and higher levels of the metabolites that have immunogenic properties.
Collapse
Affiliation(s)
- Min-Gul Kim
- Center for Clinical Pharmacology, Jeonbuk National University Hospital, Jeonju 54907, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Korea
- Department of Pharmacology, School of Medicine, Jeonbuk National University, Jeonju 54907, Korea
| | - Suin Kim
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Ji-Young Jeon
- Center for Clinical Pharmacology, Jeonbuk National University Hospital, Jeonju 54907, Korea
| | - Seol Ju Moon
- Center for Clinical Pharmacology, Jeonbuk National University Hospital, Jeonju 54907, Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Korea
| | - Yong-Geun Kwak
- Center for Clinical Pharmacology, Jeonbuk National University Hospital, Jeonju 54907, Korea
- Department of Pharmacology, School of Medicine, Jeonbuk National University, Jeonju 54907, Korea
| | - Joo Young Na
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | - SeungHwan Lee
- Department of Clinical Pharmacology and Therapeutics, Seoul National University College of Medicine and Hospital, Seoul 03080, Korea
| | | | | | - Sang-Min Lee
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Seo-Yeon Choi
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| | - Kwang-Hee Shin
- College of Pharmacy, Research Institute of Pharmaceutical Sciences, Kyungpook National University, Daegu 41566, Korea
| |
Collapse
|
23
|
Lee HK, Kim NE, Shin CM, Oh TJ, Yoon H, Park YS, Kim N, Won S, Lee DH. Gut microbiome signature of metabolically healthy obese individuals according to anthropometric, metabolic and inflammatory parameters. Sci Rep 2024; 14:3449. [PMID: 38342934 PMCID: PMC10859373 DOI: 10.1038/s41598-024-53837-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 02/06/2024] [Indexed: 02/13/2024] Open
Abstract
In this study, we investigated the characteristics of gut microbiome in the metabolically healthy obese (MHO) patients, and how they correlate with metabolic and inflammatory profiles. A total of 120 obese people without metabolic comorbidities were recruited, and their clinical phenotypes, metabolic and inflammatory parameters were analysed. The faecal microbial markers originating from bacterial cell and extracellular vesicle (EV) were profiled using 16S rDNA sequencing. The total study population could be classified into two distinct enterotypes (enterotype I: Prevotellaceae-predominant, enterotype II: Akkermansia/Bacteroides-predominant), based on their stool EV-derived microbiome profile. When comparing the metabolic and inflammatory profiles, subjects in enterotype I had higher levels of serum IL-1β [false discovery rate (FDR) q = 0.050] and had a lower level of microbial diversity than enterotype II (Wilcoxon rank-sum test p < 0.01). Subjects in enterotype I had relatively higher abundance of Bacteroidetes, Prevotellaceae and Prevotella-derived EVs, and lower abundance of Actinobacteria, Firmicutes, Proteobacteria, Akkermansia and Bacteroides-derived EVs (FDR q < 0.05). In conclusion, HMO patients can be categorised into two distinct enterotypes by the faecal EV-derived microbiome profile. The enterotyping may be associated with different metabolic and inflammatory profiles. Further studies are warranted to elucidate the long-term prognostic impact of EV-derived microbiome in the obese population.
Collapse
Affiliation(s)
- Ho-Kyoung Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Nam-Eun Kim
- Institute of Health and Environment, Seoul National University, Seoul, South Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea.
| | - Tae Jung Oh
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea
| | - Sungho Won
- Department of Public Health Sciences, Seoul National University, Seoul, South Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, 82, Gumi-ro 173, Beon-gil, Bundang-gu, Seongnam-si, Gyeonggi-do, 13620, South Korea.
| |
Collapse
|
24
|
Zhang C, Li C, Zhao P, Shao Q, Ma Y, Bai D, Liao C, He L, Huang S, Wang X. Effects of dietary Glycyrrhiza polysaccharide supplementation on growth performance, intestinal antioxidants, immunity and microbiota in weaned piglets. Anim Biotechnol 2023; 34:2273-2284. [PMID: 35714985 DOI: 10.1080/10495398.2022.2086878] [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: 11/01/2022]
Abstract
The aim of this study was to evaluate the effects of dietary Glycyrrhiza polysaccharide (GCP) supplementation on growth performance, intestinal antioxidants, immunity and microbiota in weaned piglets. One hundred and twenty 28-day-old weaned piglets were randomly assigned into five groups (four replicates per group) and fed a basal diet with GCP at 0, 500, 1000, 2000 and 4000 mg/kg for four weeks, respectively. Results showed that 1000 mg/kg GCP improved piglets' ADG and ADFI and reduced FCR (p < .05). Thus, the 0 and 1000 mg/kg GCP dose were selected for subsequent experiments. We found that 1000 mg/GCP increased SOD and T-AOC and decreased MDA in the jejunal mucosa (p < .05). Dietary 1000 mg/kg GCP also resulted in high levels of sIgA, IL-10 and TGF-β, whereas IL-2 dropped dramatically (p < .05). The relative expression levels of ZO-1, CLDN, OCLDN, TLR-4, IL-10, TGF-β, Nrf-2, SOD1 and CAT increased in the jejunal mucosa, whereas INF-γ decreased (p < .05). 1000 mg/kg GCP treatment altered the diversity and community composition of cecal microbiota in pigs, with increasing relative abundance of Bacteroidota and Lactobacillus at phylum and genus levels (p < .05), respectively. The results suggested that dietary 1000 mg/kg GCP could improve growth performance and intestinal health of weaned piglets.
Collapse
Affiliation(s)
- Cai Zhang
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, P. R. China
| | - Chenxu Li
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, P. R. China
| | - Pengli Zhao
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, P. R. China
| | - Qi Shao
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, P. R. China
| | - Yanbo Ma
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, P. R. China
| | - Dongying Bai
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, P. R. China
| | - Chengshui Liao
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, P. R. China
| | - Lei He
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, P. R. China
| | - Shucheng Huang
- College of Animal Science and Veterinary Medicine, Henan Agricultural University, Zhengzhou, P. R. China
| | - Xueying Wang
- Henan International Joint Laboratory of Animal Welfare and Health Breeding, Henan University of Science and Technology, Luoyang, P. R. China
| |
Collapse
|
25
|
Li CC, Hsu WF, Chiang PC, Kuo MC, Wo AM, Tseng YJ. Characterization of markers, functional properties, and microbiome composition in human gut-derived bacterial extracellular vesicles. Gut Microbes 2023; 15:2288200. [PMID: 38038385 PMCID: PMC10730231 DOI: 10.1080/19490976.2023.2288200] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/22/2023] [Indexed: 12/02/2023] Open
Abstract
Past studies have confirmed the etiologies of bacterial extracellular vesicles (BEVs) in various diseases, including inflammatory bowel disease (IBD) and colorectal cancer (CRC). This study aimed to investigate the characteristics of stool-derived bacterial extracellular vesicles (stBEVs) and discuss their association with stool bacteria. First, three culture models - gram-positive (G+)BcBEVs (from B.coagulans), gram-negative (G-)EcBEVs (from E.coli), and eukaryotic cell-derived EVs (EEV, from Colo205 cell line) - were used to benchmark various fractions of stEVs separated from optimized density gradient approach (DG). As such, WB, TEM, NTA, and functional assays, were utilized to analyze properties and distribution of EVs in cultured and stool samples. Stool samples from healthy individuals were interrogated using the approaches developed. Results demonstrated successful separation of most stBEVs (within DG fractions 8&9) from stEEVs (within DG fractions 5&6). Data also suggest the presence of stBEV DNA within vesicles after extraction of BEV DNA and DNase treatment. Metagenomic analysis from full-length (FL) region sequencing results confirmed significant differences between stool bacteria and stBEVs. Significantly, F8&9 and the pooled sample (F5-F9) exhibited a similar microbial composition, indicating that F8&9 were enriched in most stBEV species, primarily dominated by Firmicutes (89.6%). However, F5&6 and F7 still held low-density BEVs with a significantly higher proportion of Proteobacteria (20.5% and 40.7%, respectively) and Bacteroidetes (24% and 13.7%, respectively), considerably exceeding the proportions in stool and F8&9. Importantly, among five healthy individuals, significant variations were observed in the gut microbiota composition of their respective stBEVs, indicating the potential of stBEVs as a target for personalized medicine and research.
Collapse
Affiliation(s)
- Chih-Chi Li
- Graduate Institute of Biomedical Electronics and Bioinformatics, College of Electrical Engineering and Computer Science, National Taiwan University, Taipei, Taiwan
| | - Wei-Fan Hsu
- Institute of Applied Mechanics, National Taiwan University, Taipei, Taiwan
- Department of R&D, Reliance Biosciences Inc, New Taipei City, Taiwan
| | - Po-Chieh Chiang
- Department of R&D, Reliance Biosciences Inc, New Taipei City, Taiwan
| | - Ming-Che Kuo
- Department of Medicine, National Taiwan University Cancer Center, Taipei, Taiwan
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan
| | - Andrew M. Wo
- Institute of Applied Mechanics, National Taiwan University, Taipei, Taiwan
- Department of R&D, Reliance Biosciences Inc, New Taipei City, Taiwan
| | - Yufeng Jane Tseng
- Graduate Institute of Biomedical Electronics and Bioinformatics, College of Electrical Engineering and Computer Science, National Taiwan University, Taipei, Taiwan
- Department of Computer Science and Information Engineering, College of Electrical Engineering and Computer Science, National Taiwan University, Taipei, Taiwan
- Master’s Program in Smart Medicine and Health Informatics, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
26
|
Meng R, Zeng M, Ji Y, Huang X, Xu M. The potential role of gut microbiota outer membrane vesicles in colorectal cancer. Front Microbiol 2023; 14:1270158. [PMID: 38029123 PMCID: PMC10661380 DOI: 10.3389/fmicb.2023.1270158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Colorectal cancer (CRC) is a common malignant digestive tract tumor in colorectal regions. Considerable evidence now shows that the gut microbiota have essential roles in CRC occurrence and development. Most Gram-negative bacteria release outer membrane vesicles (OMVs) via outer membrane blistering, which contain specific cargoes which interact with host cells via intercellular communications, host immune regulation, and gut microbiota homeostasis. Studies have also shown that OMVs selectively cluster near tumor cells, thus cancer treatment strategies based on OMVs have attracted considerable research attention. However, little is known about the possible impact of gut microbiota OMVs in CRC pathophysiology. Therefore, in this review, we summarize the research progress on molecular composition and function of OMV, and review the microbial dysbiosis in CRC. We then focus on the potential role of gut microbiota OMVs in CRC. Finally, we examine the clinical potential of OMVs in CRC treatment, and their main advantages and challenges in tumor therapy.
Collapse
Affiliation(s)
- Ran Meng
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Minmin Zeng
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Ying Ji
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xinxiang Huang
- Department of Biochemistry and Molecular Biology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Institute of Digestive Diseases, Jiangsu University, Zhenjiang, Jiangsu, China
| |
Collapse
|
27
|
Long D, Mao C, Zhang Z, Zou J, Zhu Y. Visual analysis of colorectal cancer and gut microbiota: A bibliometric analysis from 2002 to 2022. Medicine (Baltimore) 2023; 102:e35727. [PMID: 37933041 PMCID: PMC10627710 DOI: 10.1097/md.0000000000035727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Accepted: 09/29/2023] [Indexed: 11/08/2023] Open
Abstract
A growing number of studies have shown that gut microbiota (GM) plays an essential role in the occurrence and development of colorectal cancer (CRC). The current body of research exploring the relationship between CRC and GM is vast. Nevertheless, bibliometric studies in this area have not yet been reported. This study aimed to explore the hotspots and frontiers of research on GM and CRC in the past 20 years, which may provide a reference for researchers in this field. The Web of Science Core Collection database was searched for publications on CRC and GM from 2002 to 2022. The scientometric softwares CiteSpace and VOSviewer were used to visually analyze the countries, institutions, authors, journals, and keywords involved in the literature. Keywords co-occurrence, cluster, and burst analysis were utilized to further explore the current state and development trends of research on GM and CRC. A total of 2158 publications were included in this study, with a noticeably rising annual publication trend. The majority of these papers are from 80 nations, primarily China and the USA. J Yu was the most active author and WS Garrett has the highest citation. Among all institutions, Shanghai Jiao Tong University has the largest number of papers. Most of the publications were published in the International Journal of Molecular Sciences, with Science being the most frequently cited journal. The 4 main clusters mainly involved probiotics, inflammation, molecular mechanisms, and research methods. Current research hotspots included "Fusobacterium nucleatum," "Escherichia coli," etc. Newly emerging research has focused predominantly on immune response, gene expression, and recent strategies for the treatment of CRC with GM. The relationship between GM and CRC will continue to be a hot research area. Changes in the composition of GM in patients with CRC, the potential molecular mechanisms as well as probiotics and natural products used in the treatment of CRC have been the focus of current research and hotspots for future studies.
Collapse
Affiliation(s)
- Dan Long
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Chenhan Mao
- Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zhensheng Zhang
- The First Traditional Chinese Medicine Hospital of Zhanjiang City, Zhanjiang, Guangdong, China
| | - Junjun Zou
- The Second Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Ying Zhu
- The First Hospital of Hunan University of Chinese Medicine, Changsha, Hunan, China
| |
Collapse
|
28
|
Wang Y, Luo X, Xiang X, Hao C, Ma D. Roles of bacterial extracellular vesicles in systemic diseases. Front Microbiol 2023; 14:1258860. [PMID: 37840728 PMCID: PMC10569430 DOI: 10.3389/fmicb.2023.1258860] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Accepted: 09/05/2023] [Indexed: 10/17/2023] Open
Abstract
Accumulating evidence suggests that in various systems, not all bidirectional microbiota-host interactions involve direct cell contact. Bacterial extracellular vesicles (BEVs) may be key participants in this interkingdom crosstalk. BEVs mediate microbiota functions by delivering effector molecules that modulate host signaling pathways, thereby facilitating host-microbe interactions. BEV production during infections by both pathogens and probiotics has been observed in various host tissues. Therefore, these vesicles released by microbiota may have the ability to drive or inhibit disease pathogenesis in different systems within the host. Here, we review the current knowledge of BEVs and particularly emphasize their interactions with the host and the pathogenesis of systemic diseases.
Collapse
Affiliation(s)
- Yanzhen Wang
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xinghong Luo
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| | - Xiaozhen Xiang
- Department of Stomatology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Chunbo Hao
- Hainan General Hospital (Hainan Affiliated Hospital of Hainan Medical University), Haikou, Hainan, China
| | - Dandan Ma
- Department of Endodontics, Stomatological Hospital, School of Stomatology, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
29
|
Li D, Zhu L, Wang Y, Zhou X, Li Y. Bacterial outer membrane vesicles in cancer: Biogenesis, pathogenesis, and clinical application. Biomed Pharmacother 2023; 165:115120. [PMID: 37442066 DOI: 10.1016/j.biopha.2023.115120] [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: 04/30/2023] [Revised: 06/18/2023] [Accepted: 07/01/2023] [Indexed: 07/15/2023] Open
Abstract
Outer membrane vesicles (OMVs) are spherical, nano-sized particles of bilayer lipid structure secreted by Gram-negative bacteria. They contain a series of cargos from bacteria and are important messengers for communication between bacteria and their environment. OMVs play multiple roles in bacterial survival and adaptation and can affect host physiological functions and disease development by acting on host cell membranes and altering host cell signaling pathways. This paper summarizes the mechanisms of OMV genesis and the multiple roles of OMVs in the tumor microenvironment. Also, this paper discusses the prospects of OMVs for a wide range of applications in drug delivery, tumor diagnosis, and therapy.
Collapse
Affiliation(s)
- Deming Li
- Anesthesia Department, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, Liaoning, China
| | - Lisi Zhu
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, Liaoning, China
| | - Yuxiao Wang
- Anesthesia Department, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, Liaoning, China
| | - Xiangyu Zhou
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, Liaoning, China.
| | - Yan Li
- Department of General surgery, The Fourth Affiliated Hospital, China Medical University, Shenyang 110032, Liaoning, China.
| |
Collapse
|
30
|
Liu J, Cai J, Fan P, Dong X, Zhang N, Tai J, Cao Y. Salidroside alleviates dextran sulfate sodium-induced colitis in mice by modulating the gut microbiota. Food Funct 2023; 14:7506-7519. [PMID: 37504971 DOI: 10.1039/d3fo01929b] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
Dysbiosis causes continuous progress of inflammatory bowel disease (IBD). Herein, we aim to explore whether Salidroside (Sal), which is a major glycoside extracted from Rhodiola rosea L., could ameliorate dextran sulfate sodium (DSS)-induced colitis by modulating the microbiota. Results showed that oral treatment with 15 mg kg-1 of Sal inhibited DSS-induced colitis in mice as evidenced by colon length, histological analysis, disease activity index (DAI) score, and the proportion and number of macrophages in the intestine. The gut microbiota of colitic mice was also partly restored by Sal. A fecal microbiota transplantation (FMT) study was designed to verify the causality. Compared with DSS-treated mice, FM from the Sal-treated donor mice significantly mitigated the symptoms of colitic mice, including reducing the DAI score, alleviating tissue damage, boosting the expression of mucin protein (mucin-2) and tight junction (TJ) proteins (occludin and zonula occludens-1 (ZO-1), and decreasing M1 macrophages in the gut. It was found that both Sal and FMT affected the structure and abundance of the gut microbiota as reflected by the decreased relative abundance of Turicibacter, Alistipes, Romboutsia and the increased relative abundance of Lactobacillus at the genus level. Moreover, the anti-inflammatory effect of Sal disappeared when the gut microbiota was depleted by antibiotics, demonstrating that Sal alleviated the intestinal inflammation in a gut microbiota-dependent manner. Thus, Sal could be a remarkable candidate as a functional food for colitis.
Collapse
Affiliation(s)
- Jiuxi Liu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, 130062 Changchun, People's Republic of China.
- CAS Key Laboratory of Regenerative Biology, Guangdong Provincial Key Laboratory of Stem Cell and Regenerative Medicine, Guangzhou Institute of Biomedicine and Health, Chinese Academy of Sciences, 510530 Guangzhou, People's Republic of China
| | - Jiapei Cai
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, 130062 Changchun, People's Republic of China.
| | - Peng Fan
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, 130062 Changchun, People's Republic of China.
| | - Xue Dong
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, 130062 Changchun, People's Republic of China.
| | - Naisheng Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, 130062 Changchun, People's Republic of China.
| | - Jiandong Tai
- Department of Colorectal & Anal Surgery, General Surgery Center, The First Hospital of Jilin University, 130021 Changchun, People's Republic of China.
| | - Yongguo Cao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Jilin University, 130062 Changchun, People's Republic of China.
- Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, 130062 Changchun, People's Republic of China
| |
Collapse
|
31
|
Liu G, Li T, Zhu X, Zhang X, Wang J. An independent evaluation in a CRC patient cohort of microbiome 16S rRNA sequence analysis methods: OTU clustering, DADA2, and Deblur. Front Microbiol 2023; 14:1178744. [PMID: 37560524 PMCID: PMC10408458 DOI: 10.3389/fmicb.2023.1178744] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Accepted: 06/14/2023] [Indexed: 08/11/2023] Open
Abstract
16S rRNA is the universal gene of microbes, and it is often used as a target gene to obtain profiles of microbial communities via next-generation sequencing (NGS) technology. Traditionally, sequences are clustered into operational taxonomic units (OTUs) at a 97% threshold based on the taxonomic standard using 16S rRNA, and methods for the reduction of sequencing errors are bypassed, which may lead to false classification units. Several denoising algorithms have been published to solve this problem, such as DADA2 and Deblur, which can correct sequencing errors at single-nucleotide resolution by generating amplicon sequence variants (ASVs). As high-resolution ASVs are becoming more popular than OTUs and only one analysis method is usually selected in a particular study, there is a need for a thorough comparison of OTU clustering and denoising pipelines. In this study, three of the most widely used 16S rRNA methods (two denoising algorithms, DADA2 and Deblur, along with de novo OTU clustering) were thoroughly compared using 16S rRNA amplification sequencing data generated from 358 clinical stool samples from the Colorectal Cancer (CRC) Screening Cohort. Our findings indicated that all approaches led to similar taxonomic profiles (with P > 0.05 in PERMNAOVA and P <0.001 in the Mantel test), although the number of ASVs/OTUs and the alpha-diversity indices varied considerably. Despite considerable differences in disease-related markers identified, disease-related analysis showed that all methods could result in similar conclusions. Fusobacterium, Streptococcus, Peptostreptococcus, Parvimonas, Gemella, and Haemophilus were identified by all three methods as enriched in the CRC group, while Roseburia, Faecalibacterium, Butyricicoccus, and Blautia were identified by all three methods as enriched in the healthy group. In addition, disease-diagnostic models generated using machine learning algorithms based on the data from these different methods all achieved good diagnostic efficiency (AUC: 0.87-0.89), with the model based on DADA2 producing the highest AUC (0.8944 and 0.8907 in the training set and test set, respectively). However, there was no significant difference in performance between the models (P >0.05). In conclusion, this study demonstrates that DADA2, Deblur, and de novo OTU clustering display similar power levels in taxa assignment and can produce similar conclusions in the case of the CRC cohort.
Collapse
Affiliation(s)
- Guang Liu
- School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
- Guangdong Hongyuan Pukong Medical Technology Co., Ltd., Guangzhou, China
| | - Tong Li
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou, China
| | - Xiaoyan Zhu
- School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Xuanping Zhang
- School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| | - Jiayin Wang
- School of Electronic and Information Engineering, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
32
|
Liu J, Huang X, Chen C, Wang Z, Huang Z, Qin M, He F, Tang B, Long C, Hu H, Pan S, Wu J, Tang W. Identification of colorectal cancer progression-associated intestinal microbiome and predictive signature construction. J Transl Med 2023; 21:373. [PMID: 37291572 PMCID: PMC10249256 DOI: 10.1186/s12967-023-04119-1] [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: 12/20/2022] [Accepted: 04/09/2023] [Indexed: 06/10/2023] Open
Abstract
OBJECTIVE The relationship between intestinal microbiome and colorectal cancer (CRC) progression is unclear. This study aims to identify the intestinal microbiome associated with CRC progression and construct predictive labels to support the accurate assessment and treatment of CRC. METHOD The 192 patients included in the study were divided into stage I-II and stage III-IV CRC patients according to the pathological stages, and preoperative stools were collected from both groups for 16S rDNA sequencing of the intestinal microbiota. Pearson correlation and Spearman correlation coefficient analysis were used to analyze the differential intestinal microbiome and the correlation with tumor microenvironment and to predict the functional pathway. XGBoost model (XGB) and Random Forest model (RF) were used to construct the microbiome-based signature. The total RNA extraction from 17 CRC tumor simples was used for transcriptome sequencing. RESULT The Simpson index of intestinal microbiome in stage III-IV CRC were significantly lower than those in stage I-II CRC. Proteus, Parabacteroides, Alistipes and Ruminococcus etc. are significantly enriched genus in feces of CRC patients with stage III-IV. ko00514: Other types of O - glycan biosynthesis pathway is relevant with CRC progression. Alistipes indistinctus was positively correlated with mast cells, immune activators IL-6 and IL6R, and GOBP_PROTEIN_FOLDING_IN_ENDOPLASMIC_RETICULUM dominantly. The Random Forest (RF) model and eXtreme Gradient Boosting (XGBoost) model constructed with 42 CRC progression-associated differential bacteria were effective in distinguishing CRC patients between stage I-II and stage III-IV. CONCLUSIONS The abundance and diversity of intestinal microbiome may increase gradually with the occurrence and progression of CRC. Elevated fetal abundance of Proteus, Parabacteroides, Alistipes and Ruminococcus may contribute to CRC progression. Enhanced synthesis of O - glycans may result in CRC progression. Alistipes indistinctus may play a facilitated role in mast cell maturation by boosting IL-6 production. Alistipes indistinctus may work in the correct folding of endoplasmic reticulum proteins in CRC, reducing ER stress and prompting the survival and deterioration of CRC, which may owe to the enhanced PERK expression and activation of downstream UPR by Alistipes indistinctus. The CRC progression-associated differential intestinal microbiome identified in our study can be served as potential microbial markers for CRC staging prediction.
Collapse
Affiliation(s)
- Jungang Liu
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People's Republic of China
| | - Xiaoliang Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People's Republic of China
| | - Chuanbin Chen
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People's Republic of China
| | - Zhen Wang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People's Republic of China
| | - Zigui Huang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People's Republic of China
| | - Mingjian Qin
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People's Republic of China
| | - Fuhai He
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People's Republic of China
| | - Binzhe Tang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People's Republic of China
| | - Chenyan Long
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People's Republic of China
| | - Hong Hu
- School of Public Health, Guangxi Medical University, Nanning, The People's Republic of China
| | - Shuibo Pan
- School of Public Health, Guangxi Medical University, Nanning, The People's Republic of China
| | - Junduan Wu
- School of Public Health, Guangxi Medical University, Nanning, The People's Republic of China.
| | - Weizhong Tang
- Division of Colorectal & Anal Surgery, Department of Gastrointestinal Surgery, Guangxi Medical University Cancer Hospital, Nanning, The People's Republic of China.
| |
Collapse
|
33
|
Rodovalho VDR, da Luz BSR, Nicolas A, Jardin J, Briard-Bion V, Folador EL, Santos AR, Jan G, Loir YL, Azevedo VADC, Guédon É. Different culture media and purification methods unveil the core proteome of Propionibacterium freudenreichii-derived extracellular vesicles. MICROLIFE 2023; 4:uqad029. [PMID: 37324655 PMCID: PMC10265600 DOI: 10.1093/femsml/uqad029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 05/12/2023] [Accepted: 05/31/2023] [Indexed: 06/17/2023]
Abstract
Bacterial extracellular vesicles (EVs) are natural lipidic nanoparticles implicated in intercellular communication. Although EV research focused mainly on pathogens, the interest in probiotic-derived EVs is now rising. One example is Propionibacterium freudenreichii, which produces EVs with anti-inflammatory effects on human epithelial cells. Our previous study with P. freudenreichii showed that EVs purified by size exclusion chromatography (SEC) displayed variations in protein content according to bacterial growth conditions. Considering these content variations, we hypothesized that a comparative proteomic analysis of EVs recovered in different conditions would elucidate whether a representative vesicular proteome existed, possibly providing a robust proteome dataset for further analysis. Therefore, P. freudenreichii was grown in two culture media, and EVs were purified by sucrose density gradient ultracentrifugation (UC). Microscopic and size characterization confirmed EV purification, while shotgun proteomics unveiled that they carried a diverse set of proteins. A comparative analysis of the protein content of UC- and SEC-derived EVs, isolated from cultures either in UF (cow milk ultrafiltrate medium) or YEL (laboratory yeast extract lactate medium), showed that EVs from all these conditions shared 308 proteins. This EV core proteome was notably enriched in proteins related to immunomodulation. Moreover, it showed distinctive features, including highly interacting proteins, compositional biases for some specific amino acids, and other biochemical parameters. Overall, this work broadens the toolset for the purification of P. freudenreichii-derived EVs, identifies a representative vesicular proteome, and enumerates conserved features in vesicular proteins. These results hold the potential for providing candidate biomarkers of purification quality, and insights into the mechanisms of EV biogenesis and cargo sorting.
Collapse
Affiliation(s)
- Vinícius de Rezende Rodovalho
- INRAE, Institut Agro, STLO, 35042, Rennes, France
- Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
- Laboratory of Immunoinflammation, Institute of Biology, University of Campinas (UNICAMP), Campinas 13000-000, Brazil
| | - Brenda Silva Rosa da Luz
- INRAE, Institut Agro, STLO, 35042, Rennes, France
- Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | | | | | | | - Edson Luiz Folador
- Center of Biotechnology, Department of Biotechnology, Federal University of Paraíba, João Pessoa 58051-900, Brazil
| | - Anderson Rodrigues Santos
- Faculty of Computer Science, Department of Computer Science, Federal University of Uberlândia, Uberlândia 38400902, Brazil
| | - Gwénaël Jan
- INRAE, Institut Agro, STLO, 35042, Rennes, France
| | - Yves Le Loir
- INRAE, Institut Agro, STLO, 35042, Rennes, France
| | - Vasco Ariston de Carvalho Azevedo
- Laboratory of Cellular and Molecular Genetics, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Brazil
| | - Éric Guédon
- Corresponding author. INRAE, Institut Agro, STLO, 35042, Rennes, France. E-mail:
| |
Collapse
|
34
|
Arunima A, van Schaik EJ, Samuel JE. The emerging roles of long non-coding RNA in host immune response and intracellular bacterial infections. Front Cell Infect Microbiol 2023; 13:1160198. [PMID: 37153158 PMCID: PMC10160451 DOI: 10.3389/fcimb.2023.1160198] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/07/2023] [Indexed: 05/09/2023] Open
Abstract
The long non-coding RNAs (lncRNAs) are evolutionarily conserved classes of non-coding regulatory transcripts of > 200 nucleotides in length. They modulate several transcriptional and post-transcriptional events in the organism. Depending on their cellular localization and interactions, they regulate chromatin function and assembly; and alter the stability and translation of cytoplasmic mRNAs. Although their proposed range of functionality remains controversial, there is increasing research evidence that lncRNAs play a regulatory role in the activation, differentiation and development of immune signaling cascades; microbiome development; and in diseases such as neuronal and cardiovascular disorders; cancer; and pathogenic infections. This review discusses the functional roles of different lncRNAs in regulation of host immune responses, signaling pathways during host-microbe interaction and infection caused by obligate intracellular bacterial pathogens. The study of lncRNAs is assuming significance as it could be exploited for development of alternative therapeutic strategies for the treatment of severe and chronic pathogenic infections caused by Mycobacterium, Chlamydia and Rickettsia infections, as well as commensal colonization. Finally, this review summarizes the translational potential of lncRNA research in development of diagnostic and prognostic tools for human diseases.
Collapse
Affiliation(s)
| | | | - James E. Samuel
- Department of Microbial Pathogenesis and Immunology, School of Medicine, Texas A&M University, Bryan, TX, United States
| |
Collapse
|
35
|
Tajasuwan L, Kettawan A, Rungruang T, Wunjuntuk K, Prombutara P. Role of Dietary Defatted Rice Bran in the Modulation of Gut Microbiota in AOM/DSS-Induced Colitis-Associated Colorectal Cancer Rat Model. Nutrients 2023; 15:nu15061528. [PMID: 36986258 PMCID: PMC10052090 DOI: 10.3390/nu15061528] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
Defatted rice bran (DRB) is a by-product of rice bran derived after the oil extraction. DRB contains several bioactive compounds, including dietary fiber and phytochemicals. The supplementation with DRB manifests chemopreventive effects in terms of anti-chronic inflammation, anti-cell proliferation, and anti-tumorigenesis in the azoxymethane (AOM) and dextran sodium sulfate (DSS)-induced colitis-associated colorectal cancer (CRC) model in rats. However, little is known about its effect on gut microbiota. Herein, we investigated the effect of DRB on gut microbiota and short chain fatty acid (SCFA) production, colonic goblet cell loss, and mucus layer thickness in the AOM/DSS-induced colitis-associated CRC rat model. The results suggested that DRB enhanced the production of beneficial bacteria (Alloprevotella, Prevotellaceae UCG-001, Ruminococcus, Roseburia, Butyricicoccus) and lessened the production of harmful bacteria (Turicibacter, Clostridium sensu stricto 1, Escherichia-Shigella, Citrobacter) present in colonic feces, mucosa, and tumors. In addition, DRB also assisted the cecal SCFAs (acetate, propionate, butyrate) production. Furthermore, DRB restored goblet cell loss and improved the thickness of the mucus layer in colonic tissue. These findings suggested that DRB could be used as a prebiotic supplement to modulate gut microbiota dysbiosis, which decreases the risks of CRC, therefore encouraging further research on the utilization of DRB in various nutritional health products to promote the health-beneficial bacteria in the colon.
Collapse
Affiliation(s)
- Laleewan Tajasuwan
- Graduate Student in Doctor of Philosophy Program in Nutrition, Faculty of Medicine Ramathibodi Hospital and Institute of Nutrition, Mahidol University, Bangkok 10400, Thailand
| | - Aikkarach Kettawan
- Institute of Nutrition, Mahidol University, Nakhon Pathom 73170, Thailand
| | - Thanaporn Rungruang
- Department of Anatomy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kansuda Wunjuntuk
- Department of Home Economics, Faculty of Agriculture, Kasetsart University, Bangkok 10900, Thailand
| | - Pinidphon Prombutara
- OMICS Sciences and Bioinformatics Center, Faculty of Science, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
36
|
Rudar J, Golding GB, Kremer SC, Hajibabaei M. Decision Tree Ensembles Utilizing Multivariate Splits Are Effective at Investigating Beta Diversity in Medically Relevant 16S Amplicon Sequencing Data. Microbiol Spectr 2023; 11:e0206522. [PMID: 36877086 PMCID: PMC10100742 DOI: 10.1128/spectrum.02065-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 02/11/2023] [Indexed: 03/07/2023] Open
Abstract
Developing an understanding of how microbial communities vary across conditions is an important analytical step. We used 16S rRNA data isolated from human stool samples to investigate whether learned dissimilarities, such as those produced using unsupervised decision tree ensembles, can be used to improve the analysis of the composition of bacterial communities in patients suffering from Crohn's disease and adenomas/colorectal cancers. We also introduce a workflow capable of learning dissimilarities, projecting them into a lower dimensional space, and identifying features that impact the location of samples in the projections. For example, when used with the centered log ratio transformation, our new workflow (TreeOrdination) could identify differences in the microbial communities of Crohn's disease patients and healthy controls. Further investigation of our models elucidated the global impact amplicon sequence variants (ASVs) had on the locations of samples in the projected space and how each ASV impacted individual samples in this space. Furthermore, this approach can be used to integrate patient data easily into the model and results in models that generalize well to unseen data. Models employing multivariate splits can improve the analysis of complex high-throughput sequencing data sets because they are better able to learn about the underlying structure of the data set. IMPORTANCE There is an ever-increasing level of interest in accurately modeling and understanding the roles that commensal organisms play in human health and disease. We show that learned representations can be used to create informative ordinations. We also demonstrate that the application of modern model introspection algorithms can be used to investigate and quantify the impacts of taxa in these ordinations, and that the taxa identified by these approaches have been associated with immune-mediated inflammatory diseases and colorectal cancer.
Collapse
Affiliation(s)
- Josip Rudar
- Department of Integrative Biology & Centre for Biodiversity Genomics, University of Guelph, Guelph, Ontario, Canada
| | - G. Brian Golding
- Department of Biology, McMaster University, Hamilton, Ontario, Canada
| | - Stefan C. Kremer
- School of Computer Science, University of Guelph, Guelph, Ontario, Canada
| | - Mehrdad Hajibabaei
- Department of Integrative Biology & Centre for Biodiversity Genomics, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
37
|
Rodríguez-Díaz C, Martín-Reyes F, Taminiau B, Ho-Plágaro A, Camargo R, Fernandez-Garcia F, Pinazo-Bandera J, Toro-Ortiz JP, Gonzalo M, López-Gómez C, Rodríguez-Pacheco F, Rodríguez de los Ríos D, Daube G, Alcain-Martinez G, García-Fuentes E. The Metagenomic Composition and Effects of Fecal-Microbe-Derived Extracellular Vesicles on Intestinal Permeability Depend on the Patient's Disease. Int J Mol Sci 2023; 24:ijms24054971. [PMID: 36902401 PMCID: PMC10002483 DOI: 10.3390/ijms24054971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 02/14/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
The composition and impact of fecal-microbe-derived extracellular vesicles (EVs) present in different diseases has not been analyzed. We determined the metagenomic profiling of feces and fecal-microbe-derived EVs from healthy subjects and patients with different diseases (diarrhea, morbid obesity and Crohn's disease (CD)) and the effect of these fecal EVs on the cellular permeability of Caco-2 cells. The control group presented higher proportions of Pseudomonas and Rikenellaceae_RC9_gut_group and lower proportions of Phascolarctobacterium, Veillonella and Veillonellaceae_ge in EVs when compared with the feces from which these EVs were isolated. In contrast, there were significant differences in 20 genera between the feces and EV compositions in the disease groups. Bacteroidales and Pseudomonas were increased, and Faecalibacterium, Ruminococcus, Clostridium and Subdoligranum were decreased in EVs from control patients compared with the other three groups of patients. Tyzzerella, Verrucomicrobiaceae, Candidatus_Paracaedibacter and Akkermansia were increased in EVs from the CD group compared with the morbid obesity and diarrhea groups. Fecal EVs from the morbid obesity, CD and, mainly, diarrhea induced a significant increase in the permeability of Caco-2 cells. In conclusion, the metagenomic composition of fecal-microbe-derived EVs changes depending on the disease of the patients. The modification of the permeability of Caco-2 cells produced by fecal EVs depends on the disease of the patients.
Collapse
Affiliation(s)
- Cristina Rodríguez-Díaz
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Flores Martín-Reyes
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- Facultad de Medicina, Universidad de Málaga, 29010 Malaga, Spain
| | - Bernard Taminiau
- Fundamental and Applied Research for Animals & Health (FARAH), Department of Food Microbiology, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Ailec Ho-Plágaro
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Raquel Camargo
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Felix Fernandez-Garcia
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - José Pinazo-Bandera
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Juan Pedro Toro-Ortiz
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Montserrat Gonzalo
- UCG de Endocrinología y Nutrición, Hospital Regional Universitario, 29009 Malaga, Spain
| | - Carlos López-Gómez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Francisca Rodríguez-Pacheco
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Dámaris Rodríguez de los Ríos
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
| | - Georges Daube
- Fundamental and Applied Research for Animals & Health (FARAH), Department of Food Microbiology, Faculty of Veterinary Medicine, University of Liège, 4000 Liège, Belgium
| | - Guillermo Alcain-Martinez
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- Correspondence: (G.A.-M.); (E.G.-F.)
| | - Eduardo García-Fuentes
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina—IBIMA Plataforma BIONAND, 29590 Malaga, Spain
- UGC de Aparato Digestivo, Hospital Universitario Virgen de la Victoria, 29010 Malaga, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), 29010 Malaga, Spain
- Correspondence: (G.A.-M.); (E.G.-F.)
| |
Collapse
|
38
|
The Gut Microbiota Metabolite Urolithin B Prevents Colorectal Carcinogenesis by Remodeling Microbiota and PD-L1/HLA-B. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6480848. [PMID: 36778211 PMCID: PMC9908333 DOI: 10.1155/2023/6480848] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Revised: 12/19/2022] [Accepted: 12/20/2022] [Indexed: 02/05/2023]
Abstract
Colorectal cancer has risen to the third occurring cancer in the world. Fluorouracil (5-Fu), oxaliplatin, and cisplatin are the most effective chemotherapeutic agents for clinical chemotherapy. Nevertheless, due to chemotherapeutic drug resistance, the survival rate of patients with CRC remains very low. In this study, we used the inflammation-induced or mutation-family-inherited murine CRC models to study the anticancer and immunotherapy effects of urolithin B (UB), the final metabolite of polyphenols in the gastrointestinal tract. The label-free proteomics analysis and the gene ontology (GO) classifications were used to test and analyze the proteins affected by UB. And 16S rDNA sequencing and flow cytometry were utilized to uncover gut microbiome composition and immune defense improved by UB administration. The results indicated that urolithin B prevents colorectal carcinogenesis by remodeling gut microbial and tumor immune microenvironments, such as HLA-B, NK cells, regulatory T cells, and γδ TCR cells, and decreasing the PD-L1. The combination of urolithin B with first-line therapeutic drugs improved the colorectal intestinal hematochezia by shaping gut microbiota, providing a strategy for the treatment of immunotherapy treatment for CRC treatments. UB combined with anti-PD-1 antibody could inhibit the growth of colon cancer. Urolithin B may thus contribute to anticancer treatments and provide a high immune response microenvironment for CRC patients' further immunotherapy.
Collapse
|
39
|
Heo M, Park YS, Yoon H, Kim NE, Kim K, Shin CM, Kim N, Lee DH. Potential of Gut Microbe-Derived Extracellular Vesicles to Differentiate Inflammatory Bowel Disease Patients from Healthy Controls. Gut Liver 2023; 17:108-118. [PMID: 36424722 PMCID: PMC9840915 DOI: 10.5009/gnl220081] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 05/09/2022] [Accepted: 06/03/2022] [Indexed: 11/26/2022] Open
Abstract
Background/Aims This study aimed to evaluate the potential of the stool microbiome and gut microbe-derived extracellular vesicles (EVs) to differentiate between patients with inflammatory bowel disease (IBD) and healthy controls, and to predict relapse in patients with IBD. Methods Metagenomic profiling of the microbiome and bacterial EVs in stool samples of controls (n=110) and patients with IBD (n=110) was performed using 16S rRNA sequencing and then compared. Patients with IBD were divided into two enterotypes based on their microbiome, and the cumulative risk of relapse was evaluated. Results There was a significant difference in the composition of the stool microbiome and gut microbe-derived EVs between patients with IBD and controls. The alpha diversity of the microbiome in patients with IBD was significantly lower than that in controls, while the beta diversity also differed significantly between the two groups. These findings were more prominent in gut microbe-derived EVs than in the stool microbiome. The survival curve tended to be different for enterotypes based on the gut microbe-derived EVs; however, this difference was not statistically significant (log-rank test, p=0.166). In the multivariable analysis, elevated fecal calprotectin (>250 mg/kg) was the only significant risk factor associated with relapse (adjusted hazard ratio, 3.147; 95% confidence interval, 1.545 to 6.408; p=0.002). Conclusions Analysis of gut microbe-derived EVs is better at differentiating patients with IBD from healthy controls than stool microbiome analysis.
Collapse
Affiliation(s)
- Min Heo
- Interdisciplinary Program of Bioinformatics, College of Natural Sciences, Seoul National University, Seoul, Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea
| | - Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea,Corresponding AuthorHyuk Yoon, ORCIDhttps://orcid.org/0000-0002-2657-0349, E-mail
| | - Nam-Eun Kim
- Department of Public Health Science, Graduate School of Public Health, Seoul, Korea
| | - Kangjin Kim
- Institute of Health and Environment, Seoul National University, Seoul, Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, Korea,Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
40
|
Wu W, Ouyang Y, Zheng P, Xu X, He C, Xie C, Hong J, Lu N, Zhu Y, Li N. Research trends on the relationship between gut microbiota and colorectal cancer: A bibliometric analysis. Front Cell Infect Microbiol 2023; 12:1027448. [PMID: 36699721 PMCID: PMC9868464 DOI: 10.3389/fcimb.2022.1027448] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 12/19/2022] [Indexed: 01/10/2023] Open
Abstract
Background Colorectal cancer (CRC)is the third most common cancer in the world and the second leading cause of cancer-related deaths, and over the past two decades, many of these researchers have provided a substantial amount of important information on the role of gut microbes in the development and progression of CRC. A causal relationship between the presence of specific microorganisms and CRC development has also been validated. Although a large number of papers related to this area have been published, no bibliometric study has been conducted to review the current state of research in this area and to highlight the research trends and hotspots in this area. This study aims to analyze the current status and future research trends of gut microbiota and CRC through bibliometric analysis. Methods Publications from 2001 to 2022 were retrieved from the Web of Science Core Collection database and screened according to inclusion criteria. VOSviewer and CiteSpace software were used to visualize the research trends in this field, including the analysis of title, country, institution, author, number of publications, year of publication, number of citations, journal, and H-index. Results A total of 863 studies were eventually identified, and the articles retrieved were cited an average of 44.85 times each. The number of publications on this topic has been increased steadily since 2011. China and the USA have made the largest contribution in the field. FRONTIERS IN MICROBIOLOGY is the top productive journal with 26 papers, and Gut journal has the highest average citation (167.23). Shanghai Jiao Tong University is the most contributive institution. Professor Yu J, Sung, Joseph J. Y and Fang JY are the most productive authors in this field. Keyword co-occurrence analysis showed that the terms of "Gut Microbiota", "Colorectal Cancer", "Inflammation", "Probiotic" and "Fusobacterium Nucleatum" were the most frequent, which revealed the research hotpots and trends in this field. Conclusions There has been a growing number of publications over the past two decades according to the global trends. China and the USA still maintained the leading position in this field. However, collaboration between institutions needs to be strengthened. It's commended to pay attention to the latest hotspots, such as "F. nucleatum" and "probiotics". This bibliometric analysis evaluates the scope and trends of gut microbiota and CRC, providing a useful perspective on current research and future directions for studying the link between the gut microbiota and CRC.
Collapse
Affiliation(s)
- Weigen Wu
- Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yaobin Ouyang
- Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Pan Zheng
- Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xinbo Xu
- Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Cong He
- Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chuan Xie
- Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Junbo Hong
- Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Nonghua Lu
- Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yin Zhu
- Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Nianshuang Li
- Digestive Disease Hospital, The First Affiliated Hospital of Nanchang University, Nanchang, China
- Department of Gastroenterology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
41
|
Yoon H, Kim NE, Park J, Shin CM, Kim N, Lee DH, Park JY, Choi CH, Kim JG, Park YS. Analysis of the gut microbiome using extracellular vesicles in the urine of patients with colorectal cancer. Korean J Intern Med 2023; 38:27-38. [PMID: 36353788 PMCID: PMC9816683 DOI: 10.3904/kjim.2022.112] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/15/2022] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND/AIMS We evaluated the gut microbiome using extracellular vesicles (EVs) in the urine of patients with colorectal cancer (CRC) to determine whether gut-microbe-derived EVs could be a potential biomarker for the diagnosis of CRC. METHODS EVs were isolated from the urine of patients with CRC and healthy controls. DNA was extracted from the EVs, and the bacterial composition was analyzed using next-generation sequencing of the 16S rRNA. RESULTS A total of 91 patients with CRC and 116 healthy controls were enrolled. We found some specific microbiomes that were more or less abundant in the CRC group than in the control group. The alpha-diversity of the gut microbiome was significantly lower in the CRC group than in the control group. A significant difference was observed in the beta-diversity between the groups. The alpha-diversity indices between patients with early- and late-stage CRC showed conflicting results; however, there was no significant difference in the beta-diversity according to the stage of CRC. There was no difference in the alpha- and beta-diversity of the gut microbiome corresponding to the location of CRC (proximal vs. distal). CONCLUSION A distinct gut microbiome is reflected in the urine EVs of patients with CRC compared with that in the healthy controls. Microbial signatures from EVs in urine could serve as potential biomarkers for the diagnosis of CRC.
Collapse
Affiliation(s)
- Hyuk Yoon
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam,
Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul,
Korea
| | - Nam-Eun Kim
- Department of Public Health Sciences, Graduate School of Public Health, Seoul National University, Seoul,
Korea
| | - Jihye Park
- Department of Internal Medicine, Yonsei University College of Medicine, Seoul,
Korea
| | - Cheol Min Shin
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam,
Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul,
Korea
| | - Nayoung Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam,
Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul,
Korea
| | - Dong Ho Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam,
Korea
- Department of Internal Medicine and Liver Research Institute, Seoul National University College of Medicine, Seoul,
Korea
| | - Jae Yong Park
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul,
Korea
| | - Chang Hwan Choi
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul,
Korea
| | - Jae Gyu Kim
- Department of Internal Medicine, Chung-Ang University College of Medicine, Seoul,
Korea
| | - Young Soo Park
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam,
Korea
| |
Collapse
|
42
|
Pérez Martínez G, Giner-Pérez L, Castillo-Romero KF. Bacterial extracellular vesicles and associated functional proteins in fermented dairy products with Lacticaseibacillus paracasei. Front Microbiol 2023; 14:1165202. [PMID: 37152726 PMCID: PMC10157241 DOI: 10.3389/fmicb.2023.1165202] [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: 02/13/2023] [Accepted: 03/20/2023] [Indexed: 05/09/2023] Open
Abstract
Cells of all kingdoms produce extracellular vesicles (EVs); hence, they are present in most environments and body fluids. Lacticaseibacillus paracasei produces EVs that have attached biologically active proteins (P40 and P75). In this study, EV and functional proteins were found in five different commercial dairy-fermented products carrying L. paracasei. Strains present in those products were isolated, and with one exception, all produced small EVs (24-47 d.nm) carrying P40 and P75. In order to winnow bacterial EV from milk EV, products were subjected to centrifugal fractionation at 15,000 × g (15 K), 33,000 × g (33 K), and 100,000 × g (100 K). P75 was present in all supernatants and pellets, but P40 was only found in two products bound to the 15 and 33 K pellets, and 16S rDNA of L. paracasei could be amplified from all 100 K EVs, indicating the presence of L. paracasei EV. To investigate the interactions of bacterial EV and proteins with milk EV, L. paracasei BL23 EV was added to three commercial UHT milk products. Small-size vesicles (50-60 d.nm) similar to L. paracasei BL23 EV were found in samples from 100 K centrifugations, but intriguingly, P40 and P75 were bound to EV in 15 and 33 K pellets, containing bovine milk EV of larger size (200-300 d.nm). Sequencing 16S rDNA bands amplified from EV evidenced the presence of bacterial EVs of diverse origins in milk and fermented products. Furthermore, L. paracasei 16S rDNA could be amplified with species-specific primers from all samples, showing the presence of L. paracasei EV in all EV fractions (15, 33, and 100 K), suggesting that these bacterial EVs possibly aggregate and are co-isolated with EV from milk. P40 and P75 proteins would be interacting with specific populations of milk EV (15 and 33 K) because they were detected bound to them in fermented products and milk, and this possibly forced the sedimentation of part of L. paracasei EV at lower centrifugal forces. This study has solved technically complex problems and essential questions which will facilitate new research focusing on the molecular behavior of probiotics during fermentation and the mechanisms of action mediating the health benefits of fermented products.
Collapse
Affiliation(s)
- Gaspar Pérez Martínez
- Laboratory of Lactic Acid Bacteria and Probiotics, Department of Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
- *Correspondence: Gaspar Pérez Martínez
| | - Lola Giner-Pérez
- Laboratory of Lactic Acid Bacteria and Probiotics, Department of Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
- Laboratory of Neurobiology, Centro de Investigación Príncipe Felipe, Valencia, Spain
| | - Keshia F. Castillo-Romero
- Laboratory of Lactic Acid Bacteria and Probiotics, Department of Biotechnology, Instituto de Agroquímica y Tecnología de Alimentos (C.S.I.C.), Valencia, Spain
- School of Engineering and Sciences, Tecnológico de Monterrey, Monterrey, Mexico
| |
Collapse
|
43
|
Oral Microbiota as Novel Biomarkers for Colorectal Cancer Screening. Cancers (Basel) 2022; 15:cancers15010192. [PMID: 36612188 PMCID: PMC9818409 DOI: 10.3390/cancers15010192] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 12/25/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
Alterations of the gut microbiome in cases of colorectal cancer (CRC) hint at the involvement of host-microbe interactions in the onset and progression of CRC and also, possibly, provide novel ways to detect and prevent CRC early. The aim of the present study was to evaluate whether the oral and fecal microbiomes of an individual can be suitable for CRC screening. Oral and fecal samples (n = 80) were gathered in Taleghani hospital, affiliated with Shahid Beheshti University of Medical Sciences, Tehran-Iran, from CRC stage 0 and I patients and healthy controls (HCs), who were screened for the first time. Microbial metagenomics assays were performed for studying microbiota profiles in all oral and fecal samples gathered. An abundance of top bacterial genera from both types of specimens (fecal and saliva samples) revealed a distinction between CRC patients and HCs. In saliva samples, the α diversity index was different between the microbiome of HCs and CRC patients, while β diversity showed a densely clustered microbiome in the HCs but a more dispersed pattern in CRC cases. The α and β diversity of fecal microbiota between HCs and CRC patients showed no statistically significant differences. Bifidobacterium was identified as a potential bacterial biomarker in CRC saliva samples, while Fusobacterium, Dialister, Catonella, Tennerella, Eubacterium-brachy-group, and Fretibacterium were ideal to distinguish HCs from CRC patients. One of the reasons for the heterogeneity of CRC may be the gastrointestinal (GI) tract microbiota, which can also cause systematic resistance to CRC. Moreover, an evaluation of saliva microbiota might offer a suitable screening test for the early detection of this malignancy, providing more accurate results than its fecal counterpart.
Collapse
|
44
|
Su KY, Koh Kok JY, Chua YW, Ong SD, Ser HL, Pusparajah P, San Saw P, Goh BH, Lee WL. Bacterial extracellular vesicles in biofluids as potential diagnostic biomarkers. Expert Rev Mol Diagn 2022; 22:1057-1062. [PMID: 36629056 DOI: 10.1080/14737159.2022.2166403] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Accepted: 01/05/2023] [Indexed: 01/12/2023]
Abstract
INTRODUCTION Extracellular vesicles (EVs) are spherical membrane-derived lipid bilayers released by cells. The human microbiota consists of trillions of microorganisms, with bacteria being the largest group secreting microbial EVs. The discovery of bacterial EVs (BEVs) has garnered interest among researchers as potential diagnostic markers, given that the microbiota is known to be associated with various diseases and EVs carry important macromolecular cargo for intercellular interaction. AREAS COVERED The differential bacterial composition identified from BEVs isolated from biofluids between patients and healthy controls may be valuable for detecting diseases. Therefore, BEVs may serve as novel diagnostic markers. Literature search on PubMed and Google Scholar databases was conducted. In this special report, we outline the commonly used approach for investigating BEVs in biofluids, the 16S ribosomal RNA gene sequencing of V3-V4 hypervariable regions, and the recent studies exploring the potential of BEVs as biomarkers for various diseases. EXPERT OPINION The emerging field of BEVs offers new possibilities for the diagnosis of various types of diseases, although there remain issues that need to be resolved in this research area to implement BEVs in clinical applications. Hence, it is important for future studies to take these challenges into consideration when investigating the diagnostic value of BEVs.
Collapse
Affiliation(s)
- Kar-Yan Su
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | - Jie-Yi Koh Kok
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | - Yie-Wei Chua
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | - Shearn-Dior Ong
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| | - Hooi Leng Ser
- Department of Biological Sciences, School of Medical and Life Sciences, Sunway University, Kuala Lumpur, Malaysia
| | - Priyia Pusparajah
- Medical Health and Translational Research Group (MHTR), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Subang Jaya, Malaysia
| | - Pui San Saw
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
| | - Bey Hing Goh
- Biofunctional Molecule Exploratory (BMEX) Research Group, School of Pharmacy, Monash University Malaysia, Subang Jaya, Malaysia
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wai-Leng Lee
- School of Science, Monash University Malaysia, Subang Jaya, Malaysia
| |
Collapse
|
45
|
Sun L, Zhu Z, Jia X, Ying X, Wang B, Wang P, Zhang S, Yu J. The difference of human gut microbiome in colorectal cancer with and without metastases. Front Oncol 2022; 12:982744. [PMID: 36387258 PMCID: PMC9665410 DOI: 10.3389/fonc.2022.982744] [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: 06/30/2022] [Accepted: 09/30/2022] [Indexed: 01/24/2023] Open
Abstract
Metastasis of colorectal cancer is deemed to be closely related to the changes in the human gut microbiome. The purpose of our study is to distinguish the differences in gut microbiota between colorectal cancer with and without metastases. Firstly, this study recruited colorectal cancer patients who met the established inclusion and exclusion criteria in the Oncology Department of Zhejiang Hospital of Traditional Chinese Medicine from February 2019 to June 2019. Fresh stool samples from healthy volunteers, non-metastatic patients, and metastatic patients were collected for 16S rRNA gene sequencing, to analyze the diversity and abundance of intestinal microorganisms in each group. The results showed that the microbial composition of the control group was more aplenty than the experimental group, while the difference also happened in the Tumor and the metastases group. At the phylum level, the abundance of Bacteroidetes significantly declined in the Tumor and the metastases group, compared with the control group. At the class level, Bacilli increased in experimental groups, while its abundance in the Tumor group was significantly higher than that in the metastases group. At the order level, the Tumor group had the highest abundance of Lactobacillales, followed by the metastases group and the control group had the lowest abundance. Overall, our study showed that the composition of the flora changed with the occurrence of metastasis in colorectal cancer. Therefore, the analysis of gut microbiota can serve as a supplement biological basis for the diagnosis and treatment of metastatic colorectal cancer which may offer the potential to develop non-invasive diagnostic tests.
Collapse
Affiliation(s)
- Leitao Sun
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Zhenzheng Zhu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xinru Jia
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xiangchang Ying
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Binbin Wang
- Department of Medical Oncology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, China
| | - Peipei Wang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China,*Correspondence: Jieru Yu, ; Shuo Zhang, ; Peipei Wang,
| | - Shuo Zhang
- The Second Affiliated Hospital of Zhejiang Chinese Medical University (Xinhua Hospital of Zhejiang Province), Hangzhou, Zhejiang, China,*Correspondence: Jieru Yu, ; Shuo Zhang, ; Peipei Wang,
| | - Jieru Yu
- School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China,*Correspondence: Jieru Yu, ; Shuo Zhang, ; Peipei Wang,
| |
Collapse
|
46
|
Glass SE, Coffey RJ. Recent Advances in the Study of Extracellular Vesicles in Colorectal Cancer. Gastroenterology 2022; 163:1188-1197. [PMID: 35724732 PMCID: PMC9613516 DOI: 10.1053/j.gastro.2022.06.039] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 06/08/2022] [Accepted: 06/11/2022] [Indexed: 12/15/2022]
Abstract
There has been significant progress in the study of extracellular vesicles (EVs) since the 2017 American Gastroenterological Association-sponsored Freston Conference "Extracellular Vesicles: Biology, Translation and Clinical Application in GI Disorders." The burgeoning interest in this field stems from the increasing recognition that EVs represent an understudied form of cell-to-cell communication and contain cargo replete with biomarkers and therapeutic targets. This short review will highlight recent advances in the field, with an emphasis on colorectal cancer. After a brief introduction to secreted particles, we will describe how our laboratory became interested in EVs, which led to refined methods of isolation and identification of 2 secreted nanoparticles. We will then summarize the cargo found in small EVs released from colorectal cancer cells and other cells in the tumor microenvironment, as well as those found in the circulation of patients with colorectal cancer. Finally, we will consider the continuing challenges and future opportunities in this rapidly evolving field.
Collapse
Affiliation(s)
- Sarah E Glass
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Robert J Coffey
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
47
|
Liang X, Dai N, Sheng K, Lu H, Wang J, Chen L, Wang Y. Gut bacterial extracellular vesicles: important players in regulating intestinal microenvironment. Gut Microbes 2022; 14:2134689. [PMID: 36242585 PMCID: PMC9578468 DOI: 10.1080/19490976.2022.2134689] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Intestinal microenvironment dysbiosis is one of the major causes of diseases, such as obesity, diabetes, inflammatory bowel disease, and colon cancer. Microbiota-based strategies have excellent clinical potential in the treatment of repetitive and refractory diseases; however, the underlying regulatory mechanisms remain elusive. Identification of the internal regulatory mechanism of the gut microbiome and the interaction mechanisms involving bacteria-host is essential to achieve precise control of the gut microbiome and obtain effective clinical data. Gut bacteria-derived extracellular vesicles (GBEVs) are lipid bilayer nanoparticles secreted by the gut microbiota and are considered key players in bacteria-bacteria and bacteria-host communication. This review focusses on the role of GBEVs in gut microbiota interactions and bacteria-host communication, and the potential clinical applications of GBEVs.
Collapse
Affiliation(s)
- Xiao Liang
- School of Life Sciences, Anhui University, Hefei, China,Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China,Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| | - Nini Dai
- School of Life Sciences, Anhui University, Hefei, China,Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China,Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| | - Kangliang Sheng
- School of Life Sciences, Anhui University, Hefei, China,Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China,Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| | - Hengqian Lu
- School of Life Sciences, Anhui University, Hefei, China,Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China,Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| | - Jingmin Wang
- School of Life Sciences, Anhui University, Hefei, China,Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China,Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| | - Liping Chen
- School of Life Sciences, Anhui University, Hefei, China,Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China,Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China
| | - Yongzhong Wang
- School of Life Sciences, Anhui University, Hefei, China,Key Laboratory of Human Microenvironment and Precision Medicine of Anhui Higher Education Institutes, Anhui University, Hefei, China,Anhui Key Laboratory of Modern Biomanufacturing, Anhui University, Hefei, China,Institute of Physical Science and Information Technology, Anhui University, Hefei, China,CONTACT Yongzhong Wang School of Life Sciences, Anhui University, Hefei, China
| |
Collapse
|
48
|
Moreno-Jiménez M, Rocha-Guzmán N, Larrosa M, Bressa C, Segura-Sánchez C, Macías-Salas A, Díaz-Rivas J, Flores-Rodríguez P. Constituents of Quercus eduardii leaf infusion: Their interaction with gut microbiota communities and therapeutic role in colorectal cancer. J Funct Foods 2022. [DOI: 10.1016/j.jff.2022.105258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022] Open
|
49
|
Characteristics of gastric cancer gut microbiome according to tumor stage and age segmentation. Appl Microbiol Biotechnol 2022; 106:6671-6687. [PMID: 36083304 DOI: 10.1007/s00253-022-12156-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 08/22/2022] [Accepted: 08/24/2022] [Indexed: 11/02/2022]
Abstract
With the development of 16S rRNA technology, gut microbiome evaluation has been performed in many diseases, including gastrointestinal tumors. Among these cancers, gastric cancer (GC) exhibits high morbidity and mortality and has been extensively studied in its pathogenesis and diagnosis techniques. The current researches have proved that the gut microbiome may have the potential to distinguish GC patients from healthy patients. However, the change of the gut microbiome according to tumor node metastasis classification (TNM) has not been clarified. Besides, the characteristics of gut microbiome in GC patients and their ages of onset are also ambiguous. To address the above shortcomings, we investigated 226 fecal samples and divided them according to their tumor stage and onset age. The findings revealed that surgery and tumor stage can change the characteristic of GC patients' gut microbiota. In specific, the effect of surgery on early gastric cancer (EGC) was greater than that on advanced gastric cancer (AGC), and the comparison of postoperative microflora with healthy people indicated that EGC has more differential bacteria than AGC. Besides, we found that Collinsella, Blautia, Anaerostipes, Dorea, and Lachnospiraceae_ND3007_group expressed differently between EGC and AGC. More importantly, it is the first time revealed that the composition of gut microbiota in GC is different between different onset ages. KEY POINTS: •Gut microbiota of gastric cancer (GC) patients are either highly associated with TNM stage and surgery or not. It shows surgery has more significant changes in early gastric cancer (EGC) than advanced gastric cancer (AGC). •There existed specific gut microbiota between EGC and AGC which may have potential to distinguish the early or advanced GC. •Onset age of GC may influence the gut microbiota: the composition of gut microbiota of early-onset gastric cancer (EOGC) and late-onset gastric cancer (LOGC) is significantly different.
Collapse
|
50
|
Vidal-Veuthey B, González D, Cárdenas JP. Role of microbial secreted proteins in gut microbiota-host interactions. Front Cell Infect Microbiol 2022; 12:964710. [PMID: 35967863 PMCID: PMC9373040 DOI: 10.3389/fcimb.2022.964710] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 07/06/2022] [Indexed: 11/30/2022] Open
Abstract
The mammalian gut microbiota comprises a variety of commensals including potential probiotics and pathobionts, influencing the host itself. Members of the microbiota can intervene with host physiology by several mechanisms, including the secretion of a relatively well-reported set of metabolic products. Another microbiota influence mechanism is the use of secreted proteins (i.e., the secretome), impacting both the host and other community members. While widely reported and studied in pathogens, this mechanism remains understood to a lesser extent in commensals, and this knowledge is increasing in recent years. In the following minireview, we assess the current literature covering different studies, concerning the functions of secretable proteins from members of the gut microbiota (including commensals, pathobionts, and probiotics). Their effect on host physiology and health, and how these effects can be harnessed by postbiotic products, are also discussed.
Collapse
Affiliation(s)
- Boris Vidal-Veuthey
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Huechuraba, Chile
| | - Dámariz González
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Huechuraba, Chile
| | - Juan P. Cárdenas
- Centro de Genómica y Bioinformática, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Huechuraba, Chile
- Escuela de Biotecnología, Facultad de Ciencias, Ingeniería y Tecnología, Universidad Mayor, Santiago, Chile
- *Correspondence: Juan P. Cárdenas,
| |
Collapse
|