1
|
Kennedy MS, Freiburger A, Cooper M, Beilsmith K, St George ML, Kalski M, Cham C, Guzzetta A, Ng SC, Chan FK, DeLeon O, Rubin D, Henry CS, Bergelson J, Chang EB. Diet outperforms microbial transplant to drive microbiome recovery in mice. Nature 2025:10.1038/s41586-025-08937-9. [PMID: 40307551 DOI: 10.1038/s41586-025-08937-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025]
Abstract
A high-fat, low-fibre Western-style diet (WD) induces microbiome dysbiosis characterized by reduced taxonomic diversity and metabolic breadth1,2, which in turn increases risk for a wide array of metabolic3-5, immune6 and systemic pathologies. Recent work has established that WD can impair microbiome resilience to acute perturbations such as antibiotic treatment7,8, although little is known about the mechanism of impairment and the specific consequences for the host of prolonged post-antibiotic dysbiosis. Here we characterize the trajectory by which the gut microbiome recovers its taxonomic and functional profile after antibiotic treatment in mice on regular chow (RC) or WD, and find that only mice on RC undergo a rapid successional process of recovery. Metabolic modelling indicates that a RC diet promotes the development of syntrophic cross-feeding interactions, whereas in mice on WD, a dominant taxon monopolizes readily available resources without releasing syntrophic byproducts. Intervention experiments reveal that an appropriate dietary resource environment is both necessary and sufficient for rapid and robust microbiome recovery, whereas microbial transplant is neither. Furthermore, prolonged post-antibiotic dysbiosis in mice on WD renders them susceptible to infection by the intestinal pathogen Salmonella enterica serovar Typhimurium. Our data challenge widespread enthusiasm for faecal microbiota transplant (FMT) as a strategy to address dysbiosis, and demonstrate that specific dietary interventions are, at a minimum, an essential prerequisite for effective FMT, and may afford a safer, more natural and less invasive alternative.
Collapse
Affiliation(s)
- M S Kennedy
- Medical Scientist Training Program, Pritzker School of Medicine, The University of Chicago, Chicago, IL, USA
- Department of Ecology & Evolution, The University of Chicago, Chicago, IL, USA
| | - A Freiburger
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
- Department of Chemical Engineering, Northwestern University, Evanston, IL, USA
| | - M Cooper
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - K Beilsmith
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - M L St George
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Medical Scientist Training Program, University of Illinois Chicago, Chicago, IL, USA
| | - M Kalski
- Department of Medicine, The University of Chicago, Chicago, IL, USA
- Stritch School of Medicine, Loyola University Chicago, Chicago, IL, USA
| | - C Cham
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - A Guzzetta
- Department of Pathology, The University of Chicago, Chicago, IL, USA
| | - S C Ng
- Microbiota I-Center (MagIC), Department of Medicine and Therapeutics, LKS Institute of Health Science, Institute of Digestive Disease, Center for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
- New Cornerstone Science Laboratory, The Chinese University of Hong Kong, Hong Kong, China
| | - F K Chan
- Microbiota I-Center (MagIC), Department of Medicine and Therapeutics, LKS Institute of Health Science, Institute of Digestive Disease, Center for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - O DeLeon
- Department of Medicine, The University of Chicago, Chicago, IL, USA
| | - D Rubin
- Inflammatory Bowel Disease Center, University of Chicago Medicine, Chicago, IL, USA
| | - C S Henry
- Division of Data Science and Learning, Argonne National Laboratory, Lemont, IL, USA
| | - J Bergelson
- Department of Biology, Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - E B Chang
- Department of Medicine, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
2
|
Clavel T, Faber F, Groussin M, Haller D, Overmann J, Pauvert C, Poyet M, Selkrig J, Stecher B, Typas A, Vehreschild MJGT, Westermann AJ, Wylensek D, Maier L. Enabling next-generation anaerobic cultivation through biotechnology to advance functional microbiome research. Nat Biotechnol 2025:10.1038/s41587-025-02660-6. [PMID: 40301656 DOI: 10.1038/s41587-025-02660-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/31/2025] [Indexed: 05/01/2025]
Abstract
Microbiomes are complex communities of microorganisms that are essential for biochemical processes on Earth and for the health of humans, animals and plants. Many environmental and host-associated microbiomes are dominated by anaerobic microbes, some of which cannot tolerate oxygen. Anaerobic microbial communities have been extensively studied over the last 20 years using molecular techniques, especially next-generation sequencing. However, there is a renewed interest in microbial cultivation because isolates provide the basis for understanding the taxonomic and functional units of biodiversity, elucidating novel biochemical pathways and the mechanisms underlying microbe-microbe and microbe-host interactions and opening new avenues for biotechnological and clinical applications. In this Perspective, we present areas of research and applications that will benefit from advancement in anaerobic microbial cultivation. We highlight key technical and infrastructural hurdles associated with the development and deployment of sophisticated cultivation workflows. Improving the performance of cultivation techniques will set new trends in functional microbiome research in the coming years.
Collapse
Affiliation(s)
- Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany.
| | - Franziska Faber
- Institute for Hygiene and Microbiology, Faculty of Medicine, University of Würzburg, Würzburg, Germany
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Mathieu Groussin
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Jörg Overmann
- Leibniz-Institut DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- Microbiology, Technical University of Braunschweig, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Charlie Pauvert
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Mathilde Poyet
- Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Joel Selkrig
- Host-Microbe Interactomics Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Bärbel Stecher
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, Germany
- Chair of Intestinal Microbiome, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Athanasios Typas
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
| | - Maria J G T Vehreschild
- Goethe University Frankfurt, University Hospital Frankfurt, Department II of Internal Medicine, Infectious Diseases, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Alexander J Westermann
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Department of Microbiology, Biocentre, University of Würzburg, Würzburg, Germany
| | - David Wylensek
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Lisa Maier
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, Tübingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, University of Tübingen, Tübingen, Germany
| |
Collapse
|
3
|
Liu M, Yang W, Zhu W, Yu D. Innovative applications and research advances of bacterial biosensors in medicine. Front Microbiol 2025; 16:1507491. [PMID: 40336836 PMCID: PMC12055861 DOI: 10.3389/fmicb.2025.1507491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/24/2025] [Indexed: 05/09/2025] Open
Abstract
The demand for early disease detection, treatment monitoring, and personalized medicine is increasing, making it more imperative than ever to create effective, accurate, portable, intelligent, multifunctional diagnostic equipment. Bacteria possess a remarkable perception of their surroundings and have the capacity to adapt by altering the expression of specific genes. Bacteria interact with target substances and produce detectable signals in response to their presence or concentration. This unique property has been harnessed in the development of bacterial biosensors. Due to groundbreaking advancements in synthetic biology, genetic engineering now enables the creation of bacteria tailored with exceptional detecting traits. In addition to meeting a wide range of application needs, this allows quick and precise detection in intricate settings and offers a strong technological basis for early disease diagnosis and treatment monitoring. This article reviews the applications and recent advancements of bacterial biosensors in the medical field and discusses the challenges and obstacles that remain in their research and application.
Collapse
Affiliation(s)
- Mengting Liu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University (Hangzhou First People’s Hospital), Hangzhou, China
- Affiliated Hangzhou First People’s Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Wenjie Yang
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University (Hangzhou First People’s Hospital), Hangzhou, China
- Affiliated Hangzhou First People’s Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Wenqi Zhu
- Affiliated Hangzhou First People’s Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Daojun Yu
- The Fourth School of Clinical Medicine, Zhejiang Chinese Medical University (Hangzhou First People’s Hospital), Hangzhou, China
- Affiliated Hangzhou First People’s Hospital, Westlake University School of Medicine, Hangzhou, China
| |
Collapse
|
4
|
Chatrizeh M, Tian J, Rogers M, Feturi F, Wu G, Firek B, Nikonov R, Cass L, Sheppeck A, Ramos-Jiménez RG, Ohja L, Caroll A, Henkel M, Azar J, Aneja RK, Campfield B, Simon D, Morowitz MJ. Plant based enteral nutrition outperforms artificial nutrition in mitigating consequences of antibiotic-induced dysbiosis in mice and humans. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.03.19.25323813. [PMID: 40166543 PMCID: PMC11957089 DOI: 10.1101/2025.03.19.25323813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Malnutrition, gut inflammation, and antibiotic induced dysbiosis (AID) are omnipresent risk factors for poor clinical outcomes among critically ill patients. We previously showed that commercially available plant-based enteral nutrition (PBEN) preserves a commensal microbiome when compared to commonly used forms of commercially available artificial enteral nutrition (AEN). This study reveals that PBEN is superior to artificial enteral nutrition (AEN) in recovering from antibiotic-induced dysbiosis (AID) in mice and humans. PBEN effectively mitigates anemia, leukopenia, restores naïve lymphocyte populations, and reduces bone marrow myeloid cell expansion. Animals randomized to PBEN also fared better in response to infectious challenges after antibiotics. A pilot clinical study validated these findings, showing increased gut commensals, reduced pathogens, and improved leukocyte balance in critically ill patients receiving PBEN compared to AEN. These results suggest PBEN offers a practical dietary approach to mitigate antibiotic-associated complications and improve clinical outcomes among hospitalized patients requiring supplemental nutrition.
Collapse
Affiliation(s)
- Mona Chatrizeh
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Jianmin Tian
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Matthew Rogers
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Firuz Feturi
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Guojun Wu
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Brian Firek
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Roman Nikonov
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lauren Cass
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Alexandra Sheppeck
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | | | - Lavnish Ohja
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ali Caroll
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Mathew Henkel
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Justin Azar
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rajesh K Aneja
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Brian Campfield
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Dennis Simon
- Department of Critical Care Medicine, University of Pittsburgh, Pittsburgh, PA, USA
- Division of Pediatric Critical Care Medicine, Safar Center for Resuscitation Research, UPMC Children's Hospital of Pittsburgh, University of Pittsburgh, Pittsburgh, PA, USA
- UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Michael J Morowitz
- Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- UPMC Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania, USA
- Lead contact
| |
Collapse
|
5
|
Jans M, Vereecke L. A guide to germ-free and gnotobiotic mouse technology to study health and disease. FEBS J 2025; 292:1228-1251. [PMID: 38523409 DOI: 10.1111/febs.17124] [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: 10/30/2023] [Revised: 01/17/2024] [Accepted: 03/11/2024] [Indexed: 03/26/2024]
Abstract
The intestinal microbiota has major influence on human physiology and modulates health and disease. Complex host-microbe interactions regulate various homeostatic processes, including metabolism and immune function, while disturbances in microbiota composition (dysbiosis) are associated with a plethora of human diseases and are believed to modulate disease initiation, progression and therapy response. The vast complexity of the human microbiota and its metabolic output represents a great challenge in unraveling the molecular basis of host-microbe interactions in specific physiological contexts. To increase our understanding of these interactions, functional microbiota research using animal models in a reductionistic setting are essential. In the dynamic landscape of gut microbiota research, the use of germ-free and gnotobiotic mouse technology, in which causal disease-driving mechanisms can be dissected, represents a pivotal investigative tool for functional microbiota research in health and disease, in which causal disease-driving mechanisms can be dissected. A better understanding of the health-modulating functions of the microbiota opens perspectives for improved therapies in many diseases. In this review, we discuss practical considerations for the design and execution of germ-free and gnotobiotic experiments, including considerations around germ-free rederivation and housing conditions, route and timing of microbial administration, and dosing protocols. This comprehensive overview aims to provide researchers with valuable insights for improved experimental design in the field of functional microbiota research.
Collapse
Affiliation(s)
- Maude Jans
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, Belgium
| | - Lars Vereecke
- VIB Center for Inflammation Research, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Ghent University, Belgium
| |
Collapse
|
6
|
Ahmad M, Aduru SV, Smith RP, Zhao Z, Lopatkin AJ. The role of bacterial metabolism in antimicrobial resistance. Nat Rev Microbiol 2025:10.1038/s41579-025-01155-0. [PMID: 39979446 DOI: 10.1038/s41579-025-01155-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 02/22/2025]
Abstract
The relationship between bacterial metabolism and antibiotic treatment is complex. On the one hand, antibiotics leverage cell metabolism to function. On the other hand, increasing research has highlighted that the metabolic state of the cell also impacts all aspects of antibiotic biology, from drug efficacy to the evolution of antimicrobial resistance (AMR). Given that AMR is a growing threat to the current global antibiotic arsenal and ability to treat infectious diseases, understanding these relationships is key to improving both public and human health. However, quantifying the contribution of metabolism to antibiotic activity and subsequent bacterial evolution has often proven challenging. In this Review, we discuss the complex and often bidirectional relationships between metabolism and the various facets of antibiotic treatment and response. We first summarize how antibiotics leverage metabolism for their function. We then focus on the converse of this relationship by specifically delineating the unique contribution of metabolism to three distinct but related arms of antibiotic biology: antibiotic efficacy, AMR evolution and AMR mechanisms. Finally, we note the relevance of metabolism in clinical contexts and explore the future of metabolic-based strategies for personalized antimicrobial therapies. A deeper understanding of these connections is crucial for the broader scientific community to address the growing crisis of AMR and develop future effective therapeutics.
Collapse
Affiliation(s)
- Mehrose Ahmad
- Weill Cornell Medical College, Cornell University, New York, NY, USA
| | - Sai Varun Aduru
- Department of Chemical Engineering, University of Rochester, Rochester, NY, USA
| | - Robert P Smith
- Cell Therapy Institute, Kiran Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
- Department of Medical Education, Kiran Patel College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | - Zirui Zhao
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA
| | - Allison J Lopatkin
- Department of Chemical Engineering, University of Rochester, Rochester, NY, USA.
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY, USA.
- Department of Biomedical Engineering, University of Rochester Medical Center, Rochester, NY, USA.
| |
Collapse
|
7
|
Chen S, Shen C, Zeng X, Sun L, Luo F, Wan R, Zhang Y, Chen X, Hou Y, Wang W, Zheng Q, Li Y. Energy metabolism and the intestinal barrier: implications for understanding and managing intestinal diseases. Front Microbiol 2025; 16:1515364. [PMID: 39959156 PMCID: PMC11826063 DOI: 10.3389/fmicb.2025.1515364] [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: 10/30/2024] [Accepted: 01/13/2025] [Indexed: 02/18/2025] Open
Abstract
The interplay between energy metabolism and the gut barrier is crucial for maintaining intestinal physiological homeostasis. Energy metabolism and the intestinal barrier perform distinct yet complementary roles that uphold intestinal ecological equilibrium. Disruptions in energy metabolism can compromise the integrity of the intestinal barrier; for example, inactivation of the AMPK pathway may lead to reduced expression of proteins associated with tight junctions. Conversely, impairment of the intestinal barrier can result in metabolic dysregulation, such as alterations in the gut microbiota that impede the production of short-chain fatty acids (SCFAs), which are essential substrates for energy metabolism. This disruption can affect energy production and modify the gut's hypoxic environment. Imbalances in these systems have been associated with the onset of various intestinal diseases. Research indicates that dietary interventions, such as a low FODMAP diet, can enhance the colonization of probiotics and improve the fermentation metabolism of SCFAs. Pharmacological strategies to elevate SCFA levels can activate the AMPK pathway and rectify abnormalities in energy metabolism. This review provides a comprehensive summary of recent advancements in elucidating the interactions between energy metabolism and the intestinal barrier.
Collapse
Affiliation(s)
- Shuai Chen
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Caifei Shen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xiaorui Zeng
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Luqiang Sun
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Fangli Luo
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Renhong Wan
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yupeng Zhang
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Xinyun Chen
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Yujun Hou
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Wen Wang
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qianhua Zheng
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Ying Li
- Acupuncture and Tuina College, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
8
|
Bermúdez-Sánchez S, Bahl MI, Hansen EB, Licht TR, Laursen MF. Oral amoxicillin treatment disrupts the gut microbiome and metabolome without interfering with luminal redox potential in the intestine of Wistar Han rats. FEMS Microbiol Ecol 2025; 101:fiaf003. [PMID: 39779288 PMCID: PMC11775830 DOI: 10.1093/femsec/fiaf003] [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: 01/03/2024] [Revised: 11/04/2024] [Accepted: 01/07/2025] [Indexed: 01/11/2025] Open
Abstract
Oral antibiotic treatment is well known to be one of the main factors affecting gut microbiota composition by altering bacterial diversity. It decreases the abundance of butyrate-producing bacteria such as Lachnospiraceae and Ruminococcaceae, while increasing abundance of Enterobacteriaceae. The recovery time of commensal bacteria post-antibiotic treatment varies among individuals, and often, complete recovery is not achieved. Recently, gut microbiota disruption has been associated with increased gut oxygen levels and higher redox potential in faecal samples. Given that redox balance is crucial for microbial metabolism and gut health, influencing fermentation processes and maintaining anaerobic conditions, we investigated the impact of oral amoxicillin treatment on the redox potential in the caecum. We used 24 Wistar Han male rats and measured caecal redox potential in situ with a probe, before and after 7 days of amoxicillin treatment, as well as after 7 days of recovery. Additionally, we analysed caecal weight, pH, antioxidant capacity, caecal microbiota, metabolome, and colonic tissue expression of relevant genes involved in the redox potential state. Our findings show that oral amoxicillin treatment significantly reduced archaeal load, and decreased the bacterial alpha diversity and affected bacterial composition of the caecal microbiome. The caecal metabolome was also significantly affected, exemplified by reduced amounts of short chain fatty acids during amoxicillin treatment. While the caecal metabolome fully recovered 7 days post amoxicillin treatment, the microbiome did not fully recover within this time frame. However, amoxicillin did not lead to an increase in luminal redox potential in the cecum during or post amoxicillin treatment. Limited differences were observed for colonic expression of genes involved in intestinal barrier function and generation of reactive oxygen species, except for the catalase gene, which was significantly upregulated post-amoxicillin treatment. Our results suggest that while oral amoxicillin disrupts the gut microbiome and metabolome, it does not directly interfere with gut luminal redox state.
Collapse
Affiliation(s)
- Sandra Bermúdez-Sánchez
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Martin Iain Bahl
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Egon Bech Hansen
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Tine Rask Licht
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| | - Martin Frederik Laursen
- National Food Institute, Technical University of Denmark, Kemitorvet, 2800 Kongens Lyngby, Denmark
| |
Collapse
|
9
|
Smith KM, Francisco SG, Zhu Y, LeRoith T, Davis ML, Crott JW, Barger K, Greenberg AS, Smith DE, Taylor A, Yeruva L, Rowan S. Dietary prevention of antibiotic-induced dysbiosis and mortality upon aging in mice. FASEB J 2024; 38:e70241. [PMID: 39655692 PMCID: PMC11629448 DOI: 10.1096/fj.202402262r] [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: 09/23/2024] [Revised: 11/08/2024] [Accepted: 11/27/2024] [Indexed: 12/13/2024]
Abstract
Oral antibiotic use is both widespread and frequent in older adults and has been linked to dysbiosis of the gut microbiota, enteric infection, and chronic diseases. Diet and nutrients, particularly prebiotics, may modify the susceptibility of the gut microbiome to antibiotic-induced dysbiosis. We fed 12-month-old mice a high glycemic (HG) or low glycemic (LG) diet with or without antibiotics (ampicillin and neomycin) for an additional 11 months. The glycemic index was modulated by the ratio of rapidly digested amylopectin starch to slowly digested amylose, a type-2-resistant starch. We observed a significant decrease in survival of mice fed a HG diet containing antibiotics (HGAbx) relative to those fed a LG diet containing antibiotics (LGAbx). HGAbx mice died with an enlarged and hemorrhagic cecum, which is associated with colonic hyperplasia and goblet cell depletion. Gut microbiome analysis revealed a pronounced expansion of Proteobacteria and a near-complete loss of Bacteroidota and Firmicutes commensal bacteria in HGAbx, whereas the LGAbx group maintained a population of Bacteroides and more closely resembled the LG microbiome. The predicted functional capacity for bile salt hydrolase activity was lost in HGAbx mice but retained in LGAbx mice. An LG diet containing amylose may therefore be a potential therapeutic to prevent antibiotic-induced dysbiosis and morbidity.
Collapse
Affiliation(s)
- Kelsey M. Smith
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- The Friedman School of Nutrition Science & PolicyTufts UniversityBostonMassachusettsUSA
| | - Sarah G. Francisco
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Ying Zhu
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- The Friedman School of Nutrition Science & PolicyTufts UniversityBostonMassachusettsUSA
| | - Tanya LeRoith
- Department of Biomedical Sciences and PathobiologyVA‐MD College of Veterinary Medicine, Virginia TechBlacksburgVirginiaUSA
| | - Meredith L. Davis
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Jimmy W. Crott
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- Department of Pathology & Laboratory MedicineBoston University School of MedicineBostonMassachusettsUSA
| | - Kathryn Barger
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Andrew S. Greenberg
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- The Friedman School of Nutrition Science & PolicyTufts UniversityBostonMassachusettsUSA
| | - Donald E. Smith
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
| | - Allen Taylor
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- The Friedman School of Nutrition Science & PolicyTufts UniversityBostonMassachusettsUSA
| | - Laxmi Yeruva
- USDA‐ARS, Microbiome and Metabolism Research UnitArkansas Children's Nutrition CenterLittle RockArkansasUSA
| | - Sheldon Rowan
- Jean Mayer USDA Human Nutrition Research Center on AgingTufts UniversityBostonMassachusettsUSA
- The Friedman School of Nutrition Science & PolicyTufts UniversityBostonMassachusettsUSA
| |
Collapse
|
10
|
Zhong K, Du X, Niu Y, Li Z, Tao Y, Wu Y, Zhang R, Guo L, Bi Y, Tang L, Dou T, Wang L. Progress in the mechanism of functional dyspepsia: roles of mitochondrial autophagy in duodenal abnormalities. Front Med (Lausanne) 2024; 11:1491009. [PMID: 39655235 PMCID: PMC11627220 DOI: 10.3389/fmed.2024.1491009] [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: 09/04/2024] [Accepted: 11/12/2024] [Indexed: 12/12/2024] Open
Abstract
Mitochondria are the main source of energy for cellular activity. Their functional damage or deficiency leads to cellular deterioration, which in turn triggers autophagic reactions. Taking mitochondrial autophagy as a starting point, the present review explored the mechanisms of duodenal abnormalities in detail, including mucosal barrier damage, release of inflammatory factors, and disruption of intracellular signal transduction. We summarized the key roles of mitochondrial autophagy in the abnormal development of the duodenum and examined the in-depth physiological and pathological mechanisms involved, providing a comprehensive theoretical basis for understanding the pathogenesis of functional dyspepsia. At present, it has been confirmed that an increase in the eosinophil count and mast cell degranulation in the duodenum can trigger visceral hypersensitive reactions and cause gastrointestinal motility disorders. In the future, it is necessary to continue exploring the molecular mechanisms and signaling pathways of mitochondrial autophagy in duodenal abnormalities. A deeper understanding of mitochondrial autophagy provides important references for developing treatment strategies for functional dyspepsia, thereby improving clinical efficacy and patient quality of life.
Collapse
Affiliation(s)
- Kexin Zhong
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Xiaojuan Du
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yuanyuan Niu
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Zhengju Li
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yongbiao Tao
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yuqian Wu
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Ruiting Zhang
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Linjing Guo
- Clinical College of Traditional Chinese Medicine, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Yurong Bi
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, School of Life Sciences, Lanzhou University, Lanzhou, China
| | - Lijuan Tang
- College of Pharmacy, Gansu University of Traditional Chinese Medicine, Lanzhou, China
| | - Tianyu Dou
- College of Life Sciences, China Jiliang University, Hangzhou, China
| | - Longde Wang
- Department of Gastroenterology, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| |
Collapse
|
11
|
Das S, Preethi B, Kushwaha S, Shrivastava R. Therapeutic strategies to modulate gut microbial health: Approaches for sarcopenia management. Histol Histopathol 2024; 39:1395-1425. [PMID: 38497338 DOI: 10.14670/hh-18-730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Sarcopenia is a progressive and generalized loss of skeletal muscle and functions associated with ageing with currently no definitive treatment. Alterations in gut microbial composition have emerged as a significant contributor to the pathophysiology of multiple diseases. Recently, its association with muscle health has pointed to its potential role in mediating sarcopenia. The current review focuses on the association of gut microbiota and mediators of muscle health, connecting the dots between the influence of gut microbiota and their metabolites on biomarkers of sarcopenia. It further delineates the mechanism by which the gut microbiota affects muscle health with progressing age, aiding the formulation of a multi-modal treatment plan involving nutritional supplements and pharmacological interventions along with lifestyle changes compiled in the review. Nutritional supplements containing proteins, vitamin D, omega-3 fatty acids, creatine, curcumin, kefir, and ursolic acid positively impact the gut microbiome. Dietary fibres foster a conducive environment for the growth of beneficial microbes such as Bifidobacterium, Faecalibacterium, Ruminococcus, and Lactobacillus. Probiotics and prebiotics act by protecting against reactive oxygen species (ROS) and inflammatory cytokines. They also increase the production of gut microbiota metabolites like short-chain fatty acids (SCFAs), which aid in improving muscle health. Foods rich in polyphenols are anti-inflammatory and have an antioxidant effect, contributing to a healthier gut. Pharmacological interventions like faecal microbiota transplantation (FMT), non-steroidal anti-inflammatory drugs (NSAIDs), ghrelin mimetics, angiotensin-converting enzyme inhibitors (ACEIs), and butyrate precursors lead to the production of anti-inflammatory fatty acids and regulate appetite, gut motility, and microbial impact on gut health. Further research is warranted to deepen our understanding of the interaction between gut microbiota and muscle health for developing therapeutic strategies for ameliorating sarcopenic muscle loss.
Collapse
Affiliation(s)
- Shreya Das
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, India
| | - B Preethi
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, India
| | - Sapana Kushwaha
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Lucknow, India.
| | - Richa Shrivastava
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, India.
| |
Collapse
|
12
|
Nonejuie P, Wilantho A, McDonald D, Htoo HH, Chalerm J, Tripathi A, Ngamphiw C, Tongsima S, Knight R, Paiboonsukwong K, Fucharoen S. Differential gut microbiota composition in β-Thalassemia patients and its correlation with iron overload. Sci Rep 2024; 14:23858. [PMID: 39394230 PMCID: PMC11470119 DOI: 10.1038/s41598-024-75456-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Accepted: 10/04/2024] [Indexed: 10/13/2024] Open
Abstract
Recent research highlights the significant impact of the gut microbiota on health and disease. Thalassemia, a hereditary blood disorder, requires regular blood transfusions, leading to an accumulation of iron in the body. Such changes could potentially alter the intestinal microbiota, thereby increasing the susceptibility of thalassemic patients to infection. In this study, we analyzed the fecal microbiota of 70 non-transfusion-dependent (NTDT) β-thalassemia/HbE patients and 30 healthy controls. Our findings indicate that iron chelation intervention had no detectable effect on the microbiome profile of thalassemic patients. However, the cross-sectional analysis revealed that the bacterial diversity and community structure in patients were significantly less diverse and distinct compared to those of healthy subjects. Using reference frames, we were also able to demonstrate that bacterial taxa that are known to produce short chain fatty acids, from the genera Alistipes, Coprococcus, and Oscillospira, and those from the family Ruminococcaceae, were less prevalent in the patients. In contrast, bacterial taxa associated with an unhealthy gut, including the genus Clostridium and those from the families Fusobacteriaceae, Enterobacteriaceae, and Peptostrptococcaceae, were more prevalent in patients and found to be correlated with higher levels of ferritin. Collectively, these changes in the microbiota could be regarded as markers of raised ferritin levels, and therefore, awareness should be exercised as they could interfere, albeit indirectly, with the treatment of the co-morbidities of thalassemia.
Collapse
Affiliation(s)
- Poochit Nonejuie
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Alisa Wilantho
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Biobank of Thailand, Pathum Thani, Thailand
| | - Daniel McDonald
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
| | - Htut Htut Htoo
- Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Jenjira Chalerm
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Anupriya Tripathi
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Chumpol Ngamphiw
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Biobank of Thailand, Pathum Thani, Thailand
| | - Sissades Tongsima
- National Center for Genetic Engineering and Biotechnology (BIOTEC), National Biobank of Thailand, Pathum Thani, Thailand
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Engineering, University of California San Diego, La Jolla, CA, USA
- Department of Computer Science and Engineering, University of California San Diego, La Jolla, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, La Jolla, CA, USA
| | - Kittiphong Paiboonsukwong
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand
| | - Suthat Fucharoen
- Thalassemia Research Center, Institute of Molecular Biosciences, Mahidol University, Nakhon Pathom, Thailand.
| |
Collapse
|
13
|
Uttarwar RG, Mekonnen SA, Van Beeck W, Wang A, Finnegan P, Roberts RF, Merenstein D, Slupsky CM, Marco ML. Effects of Bifidobacterium animalis subsp. lactis BB-12 and yogurt on mice during oral antibiotic administration. Microbiol Res 2024; 286:127794. [PMID: 38852301 DOI: 10.1016/j.micres.2024.127794] [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: 02/16/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/11/2024]
Abstract
Probiotics have the potential to prevent disruptions to normal gastrointestinal function caused by oral antibiotic use. In this study, we examined the capacity of Bifidobacterium animalis subspecies lactis BB-12 (BB-12) and yogurt, separately and combined, to mitigate the effects of the antibiotic amoxicillin-clavulanate (AMC) on the gut microbiota and metabolomes of C57BL/6 J mice. Male and female mice were administered either BB-12, yogurt, BB-12 in yogurt, or saline for 10 days concurrent with the inclusion of AMC in the drinking water. Male mice exposed to AMC exhibited significant reductions (p<0.05) in body weight over the course of the study compared to sham (no AMC) controls whereas no such effects were observed for female mice. AMC administration resulted in rapid alterations to the intestinal microbiota in both sexes irrespective of BB-12 or yogurt treatment, including significant (p<0.05) losses in bacterial cell numbers and changes in microbial alpha-diversity and beta-diversity in the feces and cecal contents. The effects of AMC on the gut microbiota were observed within one day of administration and the bacterial contents continued to change over time, showing a succession marked by rapid reductions in Muribaculaceae and Lachnospiraceae and temporal increases in proportions of Acholeplasmataceae (day 1) and Streptococcaceae and Leuconostocaceae (day 5). By day 10 of AMC intake, high proportions of Gammaproteobacteria assigned as Erwiniaceae or Enterobacteriaceae (average of 63 %), were contained in the stools and were similarly enriched in the cecum. The cecal contents of mice given AMC harbored significantly reduced concentrations of (branched) short-chain fatty acids (SCFA), aspartate, and other compounds, whereas numerous metabolites, including formate, lactate, and several amino acids and amino acid derivatives were significantly enriched. Despite the extensive impact of AMC, starting at day 7 of the study, the body weights of male mice given yogurt or BB-12 (in saline) with AMC were similar to the healthy controls. BB-12 (in saline) and yogurt intake was associated with increased Streptococcaceae and both yogurt and BB-12 resulted in lower proportions of Erwiniaceae in the fecal and cecal contents. The cecal contents of mice fed BB-12 in yogurt contained levels of formate, glycine, and glutamine that were equivalent to the sham controls. These findings highlight the potential of BB-12 and yogurt to mitigate antibiotic-induced gut dysbiosis.
Collapse
Affiliation(s)
- Ruchita G Uttarwar
- Department of Food Science & Technology, University of California, Davis, USA
| | - Solomon A Mekonnen
- Department of Food Science & Technology, University of California, Davis, USA
| | - Wannes Van Beeck
- Department of Food Science & Technology, University of California, Davis, USA
| | - Aidong Wang
- Department of Food Science & Technology, University of California, Davis, USA; Department of Nutrition, University of California, Davis, CA, USA
| | - Peter Finnegan
- Department of Food Science & Technology, University of California, Davis, USA
| | | | - Daniel Merenstein
- Department of Family Medicine, Georgetown University Medical Center, Washington, DC, USA
| | - Carolyn M Slupsky
- Department of Food Science & Technology, University of California, Davis, USA; Department of Nutrition, University of California, Davis, CA, USA
| | - Maria L Marco
- Department of Food Science & Technology, University of California, Davis, USA.
| |
Collapse
|
14
|
Lin K, Peng F, He K, Qian Z, Mei X, Su Z, Wujimaiti Y, Xia X, Zhang T. Research progress on intestinal microbiota regulating cognitive function through the gut-brain axis. Neurol Sci 2024; 45:3711-3721. [PMID: 38632176 DOI: 10.1007/s10072-024-07525-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 04/05/2024] [Indexed: 04/19/2024]
Abstract
The intestinal microbiota community is a fundamental component of the human body and plays a significant regulatory role in maintaining overall health and in the management disease states.The intestinal microbiota-gut-brain axis represents a vital connection in the cognitive regulation of the central nervous system by the intestinal microbiota.The impact of intestinal microbiota on cognitive function is hypothesized to manifest through both the nervous system and circulatory system. Imbalances in intestinal microbiota during the perioperative period could potentially contribute to perioperative neurocognitive dysfunction. This article concentrates on a review of existing literature to explore the potential influence of intestinal microbiota on brain and cognitive functions via the nervous and circulatory systems.Additionally, it summarizes recent findings on the impact of perioperative intestinal dysbacteriosis on perioperative neurocognitive dysfunction and suggests novel approaches for prevention and treatment of this condition.
Collapse
Affiliation(s)
- Kaijie Lin
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Feng Peng
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
- The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China
| | - Kunyang He
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Zhengyu Qian
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Xuan Mei
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | - Zhikun Su
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China
| | | | - Xun Xia
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China.
- The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China.
| | - Tianyao Zhang
- School of Clinical Medicine, Chengdu Medical College, Chengdu, Sichuan, China.
- The First Affiliated Hospital Of Chengdu Medical College, Chengdu, Sichuan, China.
| |
Collapse
|
15
|
Zhan ZS, Zheng ZS, Shi J, Chen J, Wu SY, Zhang SY. Unraveling colorectal cancer prevention: The vitamin D - gut flora - immune system nexus. World J Gastrointest Oncol 2024; 16:2382-2391. [DOI: 10.4251/wjgo.v16.i6.2382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/02/2024] [Accepted: 04/11/2024] [Indexed: 06/13/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers diagnosed in the world. Although environmental and genetic factors play a major role in the pathogenesis of CRC, extensive research has suggested that vitamin D may play a pivotal role in the development of CRC. Vitamin D, primarily obtained through sunlight exposure, dietary sources, and supplements, has long been recognized for its essential functions in maintaining health, including immune regulation. This article delves into the intricate relationship between vitamin D, the immune system, gut flora, and the prevention of CRC. It presents a synthesis of epidemiological data, experimental studies, and clinical trials, highlighting the mechanisms by which vitamin D influences immune cell function, cytokine production, and inflammation. By enhancing the immune system’s surveillance and anti-tumor activity, vitamin D may offer a promising avenue for CRC prevention. Furthermore, this comprehensive review delves into the prospective clinical applications of vitamin D supplementation and delineates the forthcoming avenues of research in this dynamic domain. Additionally, the paper tentatively outlines a spectrum of prophylactic impacts of vitamin D on CRC, emphasizing its significant potential in reducing CRC risk through shedding light on its mechanisms, encompassing antineoplastic mechanisms, influences on the immune system, and modulation of the gut microbiome.
Collapse
Affiliation(s)
- Zhi-Song Zhan
- Department of Dentistry, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| | - Zu-Shun Zheng
- Department of Physical Examination, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| | - Jing Shi
- Department of Anesthesiology, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| | - Juan Chen
- Department of Clinical Laboratory, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| | - Si-Yi Wu
- Department of Surgery, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| | - Shi-Yan Zhang
- Department of Clinical Laboratory, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| |
Collapse
|
16
|
Zhan ZS, Zheng ZS, Shi J, Chen J, Wu SY, Zhang SY. Unraveling colorectal cancer prevention: The vitamin D - gut flora - immune system nexus. World J Gastrointest Oncol 2024; 16:2394-2403. [PMID: 38994172 PMCID: PMC11236262 DOI: 10.4251/wjgo.v16.i6.2394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 03/02/2024] [Accepted: 04/11/2024] [Indexed: 06/14/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most common cancers diagnosed in the world. Although environmental and genetic factors play a major role in the pathogenesis of CRC, extensive research has suggested that vitamin D may play a pivotal role in the development of CRC. Vitamin D, primarily obtained through sunlight exposure, dietary sources, and supplements, has long been recognized for its essential functions in maintaining health, including immune regulation. This article delves into the intricate relationship between vitamin D, the immune system, gut flora, and the prevention of CRC. It presents a synthesis of epidemiological data, experimental studies, and clinical trials, highlighting the mechanisms by which vitamin D influences immune cell function, cytokine production, and inflammation. By enhancing the immune system's surveillance and anti-tumor activity, vitamin D may offer a promising avenue for CRC prevention. Furthermore, this comprehensive review delves into the prospective clinical applications of vitamin D supplementation and delineates the forthcoming avenues of research in this dynamic domain. Additionally, the paper tentatively outlines a spectrum of prophylactic impacts of vitamin D on CRC, emphasizing its significant potential in reducing CRC risk through shedding light on its mechanisms, encompassing antineoplastic mechanisms, influences on the immune system, and modulation of the gut microbiome.
Collapse
Affiliation(s)
- Zhi-Song Zhan
- Department of Dentistry, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| | - Zu-Shun Zheng
- Department of Physical Examination, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| | - Jing Shi
- Department of Anesthesiology, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| | - Juan Chen
- Department of Clinical Laboratory, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| | - Si-Yi Wu
- Department of Surgery, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| | - Shi-Yan Zhang
- Department of Clinical Laboratory, Fuding Hospital, Fujian University of Traditional Chinese Medicine, Fuding 355200, Fujian Province, China
| |
Collapse
|
17
|
Hu J, Zou H, Qiao X, Wang Y, Lv M, Zhang K, Wang F. The relationship between oxidative balance scores and chronic diarrhea and constipation: a population-based study. BMC Public Health 2024; 24:1366. [PMID: 38773415 PMCID: PMC11106991 DOI: 10.1186/s12889-024-18683-8] [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: 09/12/2023] [Accepted: 04/22/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Oxidative stress is closely related to gut health. Exposures to oxidative stress in one's diet and lifestyle can be evaluated by the oxidative balance score (OBS). However, the relationship between OBS and intestinal habits is unknown. This study aimed to investigate the relationships between OBS and intestinal habits (chronic diarrhea and chronic constipation) and the underlying mechanisms involved. METHODS Using data from the National Health and Nutrition Examination Survey (NHANES) database from 2005 to 2010, we included a total of 8065 participants. Twenty dietary and lifestyle factors were selected for the OBS calculates. Chronic constipation and chronic diarrhea were defined using the Bristol stool form scale (BSFS) types 1 and 2 and the BSFS 6 and 7, respectively. Multivariate logistic regression, subgroup analysis, and restricted cubic splines (RCS) analysis were used to evaluate the relationship between OBS and defecation habits. Finally, we used mediation analysis to explore the indirect effects of oxidative stress and inflammatory markers on these associations. RESULTS After adjusting for all the covariates, multivariate logistic regression analysis revealed that OBS was negatively correlated with diarrhea (OR = 0.57; 95%CI = 0.39-0.83; P = 0.008)and positively correlated with constipation (OR = 1.75; 95%CI = 1.19-2.25; P = 0.008). The RCS showed a nonlinear relationship between OBS and diarrhea (P for nonlinearity = 0.02) and a linear relationship between OBS and constipation (P for nonlinearity = 0.19). Mediation analysis showed that the C-reactive protein (CRP) concentration and white blood cell (WBC) count mediated the correlation between OBS and diarrhea by 6.28% and 6.53%, respectively (P < 0.05). CONCLUSIONS OBS is closely related to changes in patients' defecation habits. Oxidative stress and inflammation may play a role in the relationship between the two. This result emphasizes the importance of the public adjusting their lifestyle and dietary habits according to their own situation. However, further prospective studies are needed to analyze the relationship between oxidative stress and changes in defecation habits.
Collapse
Affiliation(s)
- Jiayan Hu
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Hede Zou
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Xiyun Qiao
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuxi Wang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | - Mi Lv
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Kunli Zhang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China
| | - Fengyun Wang
- Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, 100091, China.
- Institute of Digestive Diseases, Xiyuan Hospital of China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
18
|
Dey P. Good girl goes bad: Understanding how gut commensals cause disease. Microb Pathog 2024; 190:106617. [PMID: 38492827 DOI: 10.1016/j.micpath.2024.106617] [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: 12/23/2023] [Revised: 03/09/2024] [Accepted: 03/10/2024] [Indexed: 03/18/2024]
Abstract
This review examines the complex connection between commensal microbiota and the development of opportunistic infections. Several underlying conditions, such as metabolic diseases and weakened immune systems, increase the vulnerability of patients to opportunistic infections. The increasing antibiotic resistance adds significant complexity to the management of infectious diseases. Although commensals have long been considered beneficial, recent research contradicts this notion by uncovering chronic illnesses linked to atypical pathogens or commensal bacteria. This review examines conditions in which commensal bacteria, which are usually beneficial, contribute to developing diseases. Commensals' support for opportunistic infections can be categorized based on factors such as colonization fitness, pathoadaptive mutation, and evasion of host immune response. Individuals with weakened immune systems are especially susceptible, highlighting the importance of mucosal host-microbiota interaction in promoting infection when conditions are inappropriate. Dysregulation of gut microbial homeostasis, immunological modulation, and microbial interactions are caused by several factors that contribute to the development of chronic illnesses. Knowledge about these mechanisms is essential for developing preventive measures, particularly for susceptible populations, and emphasizes the importance of maintaining a balanced gut microbiota in reducing the impact of opportunistic infections.
Collapse
Affiliation(s)
- Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, Punjab, India.
| |
Collapse
|
19
|
Letourneau J, Carrion VM, Jiang S, Osborne OW, Holmes ZC, Fox A, Epstein P, Tan CY, Kirtley M, Surana NK, David LA. Interplay between particle size and microbial ecology in the gut microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.26.591376. [PMID: 38712077 PMCID: PMC11071529 DOI: 10.1101/2024.04.26.591376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Physical particles can serve as critical abiotic factors that structure the ecology of microbial communities. For non-human vertebrate gut microbiomes, fecal particle size (FPS) has been known to be shaped by chewing efficiency and diet. However, little is known about what drives FPS in the human gut. Here, we analyzed FPS by laser diffraction across a total of 76 individuals and found FPS to be strongly individualized. Surprisingly, a behavioral intervention with 41 volunteers designed to increase chewing efficiency did not impact FPS. Dietary patterns could also not be associated with FPS. Instead, we found evidence that mammalian and human gut microbiomes shaped FPS. Fecal samples from germ-free and antibiotic-treated mice exhibited increased FPS relative to colonized mice. In humans, markers of longer transit time were correlated with smaller FPS. Gut microbiota diversity and composition were also associated with FPS. Finally, ex vivo culture experiments using human fecal microbiota from distinct donors showed that differences in microbiota community composition can drive variation in particle size. Together, our results support an ecological model in which the human gut microbiome plays a key role in reducing the size of food particles during digestion, and that the microbiomes of individuals vary in this capacity. These new insights also suggest FPS in humans to be governed by processes beyond those found in other mammals and emphasize the importance of gut microbiota in shaping their own abiotic environment.
Collapse
Affiliation(s)
- Jeffrey Letourneau
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710
| | - Verónica M Carrion
- Duke Office of Clinical Research, Duke University School of Medicine, Durham, NC 27710
| | - Sharon Jiang
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710
| | - Olivia W Osborne
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710
| | - Zachary C Holmes
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710
| | - Aiden Fox
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710
| | - Piper Epstein
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710
| | - Chin Yee Tan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710
| | - Michelle Kirtley
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710
| | - Neeraj K Surana
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710
- Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710
| | - Lawrence A David
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710
- Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710
- Program in Computational Biology and Bioinformatics, Duke University School of Medicine, Durham, NC 27710
| |
Collapse
|
20
|
Ortiz de Ora L, Balsamo JM, Uyeda KS, Bess EN. Discovery of a Gut Bacterial Metabolic Pathway that Drives α-Synuclein Aggregation. ACS Chem Biol 2024; 19:1011-1021. [PMID: 38517270 PMCID: PMC11040608 DOI: 10.1021/acschembio.4c00095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 03/06/2024] [Accepted: 03/11/2024] [Indexed: 03/23/2024]
Abstract
Parkinson's disease (PD) etiology is associated with aggregation and accumulation of α-synuclein (α-syn) proteins in midbrain dopaminergic neurons. Emerging evidence suggests that in certain subtypes of PD, α-syn aggregates originate in the gut and subsequently spread to the brain. However, mechanisms that instigate α-syn aggregation in the gut have remained elusive. In the brain, the aggregation of α-syn is induced by oxidized dopamine. Such a mechanism has not been explored in the context of the gastrointestinal tract, a niche harboring 46% of the body's dopamine reservoirs. Here, we report that Enterobacteriaceae, a bacterial family prevalent in human gut microbiotas, induce α-syn aggregation. More specifically, our in vitro data indicate that respiration of nitrate by Escherichia coli K-12, which results in production of nitrite that mediates oxidation of Fe2+ to Fe3+, creates an oxidizing redox potential. These oxidizing conditions enabled the formation of dopamine-derived quinones and α-syn aggregates. Exposing nitrite, but not nitrate, to enteroendocrine STC-1 cells induced aggregation of α-syn that is natively expressed in these cells, which line the intestinal tract. Taken together, our findings indicate that bacterial nitrate reduction may be critical for initiating intestinal α-syn aggregation.
Collapse
Affiliation(s)
- Lizett Ortiz de Ora
- Department
of Chemistry, University of California, Irvine, California 92617, United States
| | - Julia M. Balsamo
- Department
of Chemistry, University of California, Irvine, California 92617, United States
| | - Kylie S. Uyeda
- Department
of Chemistry, University of California, Irvine, California 92617, United States
| | - Elizabeth N. Bess
- Department
of Chemistry, University of California, Irvine, California 92617, United States
- Department
of Molecular Biology and Biochemistry, University
of California, Irvine, California 92617, United States
| |
Collapse
|
21
|
Letourneau J, Walker L, Han SH, David LA, Younge N. A pilot study of fecal pH and redox as functional markers in the premature infant gut microbiome. PLoS One 2024; 19:e0290598. [PMID: 38261587 PMCID: PMC10805279 DOI: 10.1371/journal.pone.0290598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/28/2023] [Indexed: 01/25/2024] Open
Abstract
The infant gut microbiome is a crucial factor in health and development. In preterm infants, altered gut microbiome composition and function have been linked to serious neonatal complications such as necrotizing enterocolitis and sepsis, which can lead to long-term disability. Although many studies have described links between microbiome composition and disease risk, there is a need for biomarkers to identify infants at risk of these complications in practice. In this pilot study, we obtained stool samples from preterm infant participants longitudinally during the first postnatal months, and measured pH and redox, as well as SCFA content and microbiome composition by 16S rRNA gene amplicon sequencing. These outcomes were compared to clinical data to better understand the role of pH and redox in infant gut microbiome development and overall health, and to assess the potential utility of pH and redox as biomarkers. We found that infants born earlier or exposed to antibiotics exhibited increased fecal pH, and that redox potential increased with postnatal age. These differences may be linked to changes in SCFA content, which was correlated with pH and increased with age. Microbiome composition was also related to birth weight, age, pH, and redox. Our findings suggest that pH and redox may serve as biomarkers of metabolic state in the preterm infant gut.
Collapse
Affiliation(s)
- Jeffrey Letourneau
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States of America
| | - LaShawndra Walker
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States of America
| | - Se Hyang Han
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States of America
| | - Lawrence A. David
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, United States of America
- Program in Computational Biology and Bioinformatics, Duke University School of Medicine, Durham, NC, United States of America
| | - Noelle Younge
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States of America
| |
Collapse
|
22
|
Bhattarai SK, Du M, Zeamer AL, Morzfeld BM, Kellogg TD, Firat K, Benjamin A, Bean JM, Zimmerman M, Mardi G, Vilbrun SC, Walsh KF, Fitzgerald DW, Glickman MS, Bucci V. Commensal antimicrobial resistance mediates microbiome resilience to antibiotic disruption. Sci Transl Med 2024; 16:eadi9711. [PMID: 38232140 PMCID: PMC11017772 DOI: 10.1126/scitranslmed.adi9711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 12/15/2023] [Indexed: 01/19/2024]
Abstract
Despite their therapeutic benefits, antibiotics exert collateral damage on the microbiome and promote antimicrobial resistance. However, the mechanisms governing microbiome recovery from antibiotics are poorly understood. Treatment of Mycobacterium tuberculosis, the world's most common infection, represents the longest antimicrobial exposure in humans. Here, we investigate gut microbiome dynamics over 20 months of multidrug-resistant tuberculosis (TB) and 6 months of drug-sensitive TB treatment in humans. We find that gut microbiome dynamics and TB clearance are shared predictive cofactors of the resolution of TB-driven inflammation. The initial severe taxonomic and functional microbiome disruption, pathobiont domination, and enhancement of antibiotic resistance that initially accompanied long-term antibiotics were countered by later recovery of commensals. This resilience was driven by the competing evolution of antimicrobial resistance mutations in pathobionts and commensals, with commensal strains with resistance mutations reestablishing dominance. Fecal-microbiota transplantation of the antibiotic-resistant commensal microbiome in mice recapitulated resistance to further antibiotic disruption. These findings demonstrate that antimicrobial resistance mutations in commensals can have paradoxically beneficial effects by promoting microbiome resilience to antimicrobials and identify microbiome dynamics as a predictor of disease resolution in antibiotic therapy of a chronic infection.
Collapse
Affiliation(s)
- Shakti K Bhattarai
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Muxue Du
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School, New York, NY 10065, USA
| | - Abigail L Zeamer
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Benedikt M Morzfeld
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Tasia D Kellogg
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Kaya Firat
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Anna Benjamin
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - James M Bean
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ 07110, USA
| | - Gertrude Mardi
- Haitian Study Group for Kaposi’s Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - Stalz Charles Vilbrun
- Haitian Study Group for Kaposi’s Sarcoma and Opportunistic Infections (GHESKIO), Port-au-Prince, Haiti
| | - Kathleen F Walsh
- Center for Global Health, Weill Cornell Medicine, New York, NY 10065, USA
- Division of General Internal Medicine, Weill Cornell Medicine, New York, NY 10065, USA
| | | | - Michael S Glickman
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Immunology and Microbial Pathogenesis Graduate Program, Weill Cornell Graduate School, New York, NY 10065, USA
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA 01605, USA
- Immunology and Microbiology Program, UMass Chan Medical School, Worcester, MA 01605, USA
| |
Collapse
|
23
|
Feng N, Xu R, Wang D, Li L, Su Y, Feng X. The Fecal Redox Potential in Healthy and Diarrheal Pigs and Their Correlation with Microbiota. Antioxidants (Basel) 2024; 13:96. [PMID: 38247520 PMCID: PMC10812559 DOI: 10.3390/antiox13010096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 01/10/2024] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
The redox potential plays a critical role in sustaining the stability of gut microbiota. This study measured the fecal redox potential in healthy and diarrheal pigs using direct and dilution methods and investigated their correlation with microbiota. The results showed that the fluctuations in the redox potential of healthy pig feces were consistent using two different methods and the two methods are equivalent based on an equivalence test. The redox potential was positively correlated with the number of fungi and negatively related to the total bacteria. The relative or absolute abundances of many bacteria at the phyla and genus levels were associated with redox potential. In diarrheal pigs, the potentiometric trends of the two methods demonstrated an opposing pattern and the correlation with total bacteria was reversed. Precipitously elevated redox potential was detected post-diarrhea using dilution methods. The absolute abundance of Escherichia-Shigella and Fuurnierella was positively correlated with redox potential, while both relative and absolute abundances of Limosilactobacillus were positively correlated. These results suggest that both methods are suitable for detecting gut redox potential in healthy pigs, while the dilution method is more suitable for diarrheal pigs. The findings on the correlation of Limosilactobacillus, Prevotella, and Escherichia-Shigella with redox potential offer novel insights for targeted modulation of intestinal health.
Collapse
Affiliation(s)
- Ni Feng
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (N.F.); (R.X.); (D.W.); (L.L.)
| | - Rongying Xu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (N.F.); (R.X.); (D.W.); (L.L.)
| | - Dongfang Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (N.F.); (R.X.); (D.W.); (L.L.)
| | - Lian Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (N.F.); (R.X.); (D.W.); (L.L.)
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China; (N.F.); (R.X.); (D.W.); (L.L.)
| | - Xiaobo Feng
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210002, China
| |
Collapse
|
24
|
Ruiz-Valdepeñas Montiel V, Vargas E, Ben Hassine A, Simon I, Duvvuri A, Chang AY, Nandhakumar P, Bulbarello A, Düsterloh A, Mak T, Wang J. Decentralized ORP Measurements for Gut Redox Status Monitoring: Toward Personalized Gut Microbiota Balance. Anal Chem 2024; 96:480-487. [PMID: 38150379 DOI: 10.1021/acs.analchem.3c04570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2023]
Abstract
Gut microbiome targeting has emerged as a new generation of personalized medicine and a potential wellness and disease driver. Specifically, the gut redox balance plays a key role in shaping the gut microbiota and its link with the host, immune system, and disease evolution. In this sense, precise and personalized nutrition has proven synergy and capability to modulate the gut microbiome environment through the formulation of dietary interventions, such as vitamin support. Accordingly, there are urgent demands for simple and effective analytical platforms for understanding the relationship between the tailored vitamin administration and the gut microbiota balance by rapid noninvasive on-the-spot oxidation/reduction potential monitoring for frequent and close surveillance of the gut redox status and targeting by personalized nutrition interventions. Herein, we present a disposable potentiometric sensor chip and a homemade multiwell potentiometric array to address the interplay of vitamin levels with the oxidation/reduction potential in human feces and saliva. The potentiometric ORP sensing platforms have been successfully validated and scaled up for the setup of a multiapplication prototype for cross-talk-free simple screening of many specimens. The interpersonal variability of the gut microbiota environment illustrates the potential of feces and saliva samples for noninvasive, frequent, and decentralized monitoring of the gut redox status to support timely human microbiota surveillance and guide precise dietary intervention toward restoring and promoting personalized gut redox balance.
Collapse
Affiliation(s)
- Víctor Ruiz-Valdepeñas Montiel
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
- Department of Analytical Chemistry, Chemistry Faculty, University Complutense of Madrid, E-28040 Madrid, Spain
| | - Eva Vargas
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Amira Ben Hassine
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Ignasi Simon
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Andres Duvvuri
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - An-Yi Chang
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | - Ponnusamy Nandhakumar
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| | | | | | - Tim Mak
- DSM-Firmenich AG, Kaiseraugst 4303, Switzerland
| | - Joseph Wang
- Department of Nanoengineering, University of California San Diego, La Jolla, California 92093, United States
| |
Collapse
|
25
|
Xu R, Feng N, Li Q, Wang H, Li L, Feng X, Su Y, Zhu W. Pectin supplementation accelerates post-antibiotic gut microbiome reconstitution orchestrated with reduced gut redox potential. THE ISME JOURNAL 2024; 18:wrae101. [PMID: 38857378 PMCID: PMC11203915 DOI: 10.1093/ismejo/wrae101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 05/31/2024] [Accepted: 06/09/2024] [Indexed: 06/12/2024]
Abstract
Antibiotic-induced gut dysbiosis (AID) presents a big challenge to host health, and the recovery from this dysbiosis is often slow and incomplete. AID is typically characterized by elevation in redox potential, Enterobacteriaceae load, and aerobic metabolism. In our previous study, a pectin-enriched diet was demonstrated to decrease fecal redox potential and modulate the gut microbiome. Therefore, we propose that pectin supplementation may modulate gut redox potential and favor post-antibiotic gut microbiome reconstitution from dysbiosis. In the present study, rats with AIDwere used to investigate the effects of pectin supplementation on post-antibiotic gut microbiome reconstitution from dysbiosis. The results showed that pectin supplementation accelerated post-antibiotic reconstitution of gut microbiome composition and function and led to enhancement of anabolic reductive metabolism and weakening of catabolic oxidative pathways. These results were corroborated by the measurement of redox potential, findings suggesting that pectin favors post-antibiotic recovery from dysbiosis. Pectin-modulated fecal microbiota transplantation accelerated the decrease in antibiotics-elevated redox potential and Enterobacteriaceae load similarly to pectin supplementation. Moreover, both pectin supplementation and Pectin-modulated fecal microbiota transplantation enriched anaerobic members, primarily from Lachnospiraceae orchestration with enhancement of microbial reductive metabolism in post-antibiotic rats. These findings suggested that pectin supplementation accelerated post-antibiotic gut microbiome reconstitution orchestrated with reduced gut redox potential and that the effect of pectin on redox potential was mediated by remodeling of the intestinal microbiota.
Collapse
Affiliation(s)
- Rongying Xu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Ni Feng
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Qiuke Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Hongyu Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Lian Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Xiaobo Feng
- Research Institute of General Surgery, Jinling Hospital, Nanjing University School of Medicine, Nanjing 210095, China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
26
|
Letourneau J, Carrion VM, Zeng J, Jiang S, Osborne OW, Holmes ZC, Fox A, Epstein P, Tan CY, Kirtley M, Surana NK, David LA. Interplay between particle size and microbial ecology in the gut microbiome. THE ISME JOURNAL 2024; 18:wrae168. [PMID: 39214074 PMCID: PMC11406467 DOI: 10.1093/ismejo/wrae168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 07/30/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Physical particles can serve as critical abiotic factors that structure the ecology of microbial communities. For non-human vertebrate gut microbiomes, fecal particle size (FPS) has been known to be shaped by chewing efficiency and diet. However, little is known about what drives FPS in the human gut. Here, we analyzed FPS by laser diffraction across a total of 76 individuals and found FPS to be strongly individualized. Contrary to our initial hypothesis, a behavioral intervention with 41 volunteers designed to increase chewing efficiency did not impact FPS. Dietary patterns could also not be associated with FPS. Instead, we found evidence that human and mouse gut microbiomes shaped FPS. Fecal samples from germ-free and antibiotic-treated mice exhibited increased FPS relative to colonized mice. In humans, markers of longer transit time were correlated with smaller FPS. Gut microbiota diversity and composition were also associated with FPS. Finally, ex vivo culture experiments using human fecal microbiota from distinct donors showed that differences in microbiota community composition can drive variation in particle size. Together, our results support an ecological model in which the human gut microbiome plays a key role in reducing the size of food particles during digestion. This finding has important implications for our understanding of energy extraction and subsequent uptake in gastrointestinal tract. FPS may therefore be viewed as an informative functional readout, providing new insights into the metabolic state of the gut microbiome.
Collapse
Affiliation(s)
- Jeffrey Letourneau
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Verónica M Carrion
- Duke Office of Clinical Research, Duke University School of Medicine, Durham, NC 27710, United States
| | - Jun Zeng
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Sharon Jiang
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Olivia W Osborne
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Zachary C Holmes
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Aiden Fox
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Piper Epstein
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Chin Yee Tan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, United States
| | - Michelle Kirtley
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
| | - Neeraj K Surana
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27710, United States
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC 27710, United States
- Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, United States
| | - Lawrence A David
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27710, United States
- Duke Microbiome Center, Duke University School of Medicine, Durham, NC 27710, United States
- Program in Computational Biology and Bioinformatics, Duke University School of Medicine, Durham, NC 27710, United States
| |
Collapse
|
27
|
Rhoades NS, Cinco IR, Hendrickson SM, Prongay K, Haertel AJ, Flores GE, Slifka MK, Messaoudi I. Infant diarrheal disease in rhesus macaques impedes microbiome maturation and is linked to uncultured Campylobacter species. Commun Biol 2024; 7:37. [PMID: 38182754 PMCID: PMC10770169 DOI: 10.1038/s42003-023-05695-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Accepted: 12/12/2023] [Indexed: 01/07/2024] Open
Abstract
Diarrheal diseases remain one of the leading causes of death for children under 5 globally, disproportionately impacting those living in low- and middle-income countries (LMIC). Campylobacter spp., a zoonotic pathogen, is one of the leading causes of food-borne infection in humans. Yet to be cultured Campylobacter spp. contribute to the total burden in diarrheal disease in children living in LMIC thus hampering interventions. We performed microbiome profiling and metagenomic genome assembly on samples collected from over 100 infant rhesus macaques longitudinally and during cases of clinical diarrhea within the first year of life. Acute diarrhea was associated with long-lasting taxonomic and functional shifts of the infant gut microbiome indicative of microbiome immaturity. We constructed 36 Campylobacter metagenomic assembled genomes (MAGs), many of which fell within 4 yet to be cultured species. Finally, we compared the uncultured Campylobacter MAGs assembled from infant macaques with publicly available human metagenomes to show that these uncultured species are also found in human fecal samples from LMIC. These data highlight the importance of unculturable Campylobacter spp. as an important target for reducing disease burden in LMIC children.
Collapse
Affiliation(s)
- Nicholas S Rhoades
- Department of Molecular biology and Biochemistry, University of California Irvine, Irvine, CA, USA
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Isaac R Cinco
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA
| | - Sara M Hendrickson
- Division of Neuroscience, Oregon National Primate Research Center, Portland, OR, USA
| | - Kamm Prongay
- Division of Animal Resources and Research Support, Oregon National Primate Research Center, Oregon Health and Science University West Campus, Portland, OR, USA
| | - Andrew J Haertel
- Division of Animal Resources and Research Support, Oregon National Primate Research Center, Oregon Health and Science University West Campus, Portland, OR, USA
| | - Gilberto E Flores
- Department of Biology, California State University, Northridge, Northridge, CA, USA
| | - Mark K Slifka
- Division of Neuroscience, Oregon National Primate Research Center, Portland, OR, USA
| | - Ilhem Messaoudi
- Department of Microbiology, Immunology and Molecular Genetics, College of Medicine, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
28
|
Sarkar A, McInroy CJA, Harty S, Raulo A, Ibata NGO, Valles-Colomer M, Johnson KVA, Brito IL, Henrich J, Archie EA, Barreiro LB, Gazzaniga FS, Finlay BB, Koonin EV, Carmody RN, Moeller AH. Microbial transmission in the social microbiome and host health and disease. Cell 2024; 187:17-43. [PMID: 38181740 PMCID: PMC10958648 DOI: 10.1016/j.cell.2023.12.014] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 01/07/2024]
Abstract
Although social interactions are known to drive pathogen transmission, the contributions of socially transmissible host-associated mutualists and commensals to host health and disease remain poorly explored. We use the concept of the social microbiome-the microbial metacommunity of a social network of hosts-to analyze the implications of social microbial transmission for host health and disease. We investigate the contributions of socially transmissible microbes to both eco-evolutionary microbiome community processes (colonization resistance, the evolution of virulence, and reactions to ecological disturbance) and microbial transmission-based processes (transmission of microbes with metabolic and immune effects, inter-specific transmission, transmission of antibiotic-resistant microbes, and transmission of viruses). We consider the implications of social microbial transmission for communicable and non-communicable diseases and evaluate the importance of a socially transmissible component underlying canonically non-communicable diseases. The social transmission of mutualists and commensals may play a significant, under-appreciated role in the social determinants of health and may act as a hidden force in social evolution.
Collapse
Affiliation(s)
- Amar Sarkar
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
| | - Cameron J A McInroy
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Siobhán Harty
- Independent, Tandy Court, Spitalfields, Dublin, Ireland
| | - Aura Raulo
- Department of Biology, University of Oxford, Oxford, UK; Department of Computing, University of Turku, Turku, Finland
| | - Neil G O Ibata
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Mireia Valles-Colomer
- Department of Medicine and Life Sciences, Pompeu Fabra University, Barcelona, Spain; Department of Cellular, Computational and Integrative Biology, University of Trento, Trento, Italy
| | - Katerina V-A Johnson
- Institute of Psychology, Leiden University, Leiden, the Netherlands; Department of Psychiatry, University of Oxford, Oxford, UK
| | - Ilana L Brito
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY, USA
| | - Joseph Henrich
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Elizabeth A Archie
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN, USA
| | - Luis B Barreiro
- Committee on Immunology, University of Chicago, Chicago, IL, USA; Department of Medicine, University of Chicago, Chicago, IL, USA; Committee on Genetics, Genomics and Systems Biology, University of Chicago, Chicago, IL, USA
| | - Francesca S Gazzaniga
- Molecular Pathology Unit, Cancer Center, Massachusetts General Hospital Research Institute, Charlestown, MA, USA; Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - B Brett Finlay
- Department of Microbiology and Immunology, University of British Columbia, Vancouver, BC, Canada; Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada; Department of Biochemistry, University of British Columbia, Vancouver, BC, Canada
| | - Eugene V Koonin
- National Center for Biotechnology Information, National Library of Medicine, Bethesda, MD, USA
| | - Rachel N Carmody
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA
| | - Andrew H Moeller
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
29
|
Zhang M, Mo R, Wang H, Liu T, Zhang G, Wu Y. Grape seed proanthocyanidin improves intestinal inflammation in canine through regulating gut microbiota and bile acid compositions. FASEB J 2023; 37:e23285. [PMID: 37933950 DOI: 10.1096/fj.202300819rr] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 10/04/2023] [Accepted: 10/18/2023] [Indexed: 11/08/2023]
Abstract
Although certain progress has been made in treating canine inflammatory bowel disease (IBD), a large proportion of dogs have a poor prognosis and may develop resistance and side effects. Therefore, it is of great significance to prevent or alleviate canine IBD through nutritional intervention. Plant polyphenol can interact with intestinal bacteria and has important prospects in the intestinal health improvement. This study evaluated the effect of grape seed proanthocyanidin (GSP), a plant-derived natural polyphenol, on Labrador Retrievers with mild IBD. In Experiment 1 of this study, GSP alleviated persistent intestinal inflammation in canines by improving inflammatory indexes and reducing intestinal permeability. Moreover, GSP treatment increased the abundance of bacteria with potential anti-inflammatory properties and engaging bile acid metabolism, including Ruminococcaceae, Faecalibacterium, Ruminococcus_torques_group, and Lachnospiraceae_NK4A136_group. Notably, targeted metabolomic analysis identified elevated productions of fecal chenodeoxycholic acid and its microbial transformation product lithocholic acid, which might contribute to relieving canine intestinal inflammation. Further, in Experiment 2, fecal microbiota transplantation was used to determine whether gut microbiota is a potential mechanism for GSP efficacy. Dogs with mild IBD received the fecal microbiota from the group administered GSP and mirrored the improvement effects of GSP, which results verified that gut microbial alteration could be an underlying mechanism for GSP efficiency on canine IBD. Our findings highlight that the mechanism of the GSP function on canine IBD is mediated by altering gut microbial composition and improving bile acid metabolism. This study proposes a natural polyphenol-based dietary strategy for improving canine intestinal health.
Collapse
Affiliation(s)
- Mingrui Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, People's Republic of China
| | - Ruixia Mo
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, People's Republic of China
| | - Haotian Wang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, People's Republic of China
| | - Tianyi Liu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, People's Republic of China
| | - Gang Zhang
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, People's Republic of China
| | - Yi Wu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing, People's Republic of China
| |
Collapse
|
30
|
Luo D, Shi CY, Wei NS, Yang BY, Qin K, Liu G, Dong BQ, Qin YX, Qin XL, Chen SY, Guo XJ, Gan L, Xu RL, Li H, Li J. The potential mechanism of the progression from latent to active tuberculosis based on the intestinal microbiota alterations. Tuberculosis (Edinb) 2023; 143:102413. [PMID: 37832478 DOI: 10.1016/j.tube.2023.102413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 09/10/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023]
Abstract
INTRODUCTION Tuberculosis (TB) poses a serious challenge to global health systems. The altered intestinal microbiota is associated with the pathogenesis of TB, but the exact links remain unclear. METHODS 16 S rDNA sequencing was performed to comprehensively detect the changes in the intestinal microbiota of feces from active TB (ATB), latent TB infection (LTBI) and healthy controls (HC). RESULTS The rarefaction curves demonstrated the sequencing results' validity. The alpha diversity was lowest in ATB, while highest in HC. Boxplot of beta diversity showed significant differences in every two groups. LDA Effect Size (LEfSe) Analysis revealed differences in probiotic bacteria like Romboutsia, Bifidobacterium and Lactobacillus in LTBI, and pro-inflammatory bacteria like R. gnavus, Streptococcus and Erysipelatoclostridium in ATB, corresponding to the cluster analysis. PICRUST2 analysis revealed the pentose phosphate pathway was active in ATB and LTBI (more active in ATB). The differences between the groups are statistically significant at the P<0.05 level. CONCLUSION Our study indicated that from LTBI to ATB, some intestinal microbiota inhibit the synthesis of interferon (INF)-γ and interleukin (IL)-17, promoting the survival and spread of Mycobacterium tuberculosis (M. tb). In addition, the metabolites secreted by intestinal microbiota and dysbiosis in intestine also have an effect on the development of LTBI to ATB.
Collapse
Affiliation(s)
- Dan Luo
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China; Guangxi Key Laboratory of Translational Medicine for Treating High-Incidence Infectious Diseases with Integrative Medicine, Nanning, China
| | - Chong-Yu Shi
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Nian-Sa Wei
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Bo-Yi Yang
- The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kai Qin
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Gang Liu
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Bai-Qing Dong
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Yi-Xiang Qin
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiao-Ling Qin
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Shi-Yi Chen
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Xiao-Jing Guo
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Li Gan
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Ruo-Lan Xu
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China
| | - Hai Li
- Department of Biostatistics, School of Public Health and Management, Guangxi University of Chinese Medicine, Nanning, China.
| | - Jing Li
- Department of Physiology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, China.
| |
Collapse
|
31
|
Homolak J, Babic Perhoc A, Knezovic A, Osmanovic Barilar J, Virag D, Salkovic-Petrisic M. Exploratory Study of Gastrointestinal Redox Biomarkers in the Presymptomatic and Symptomatic Tg2576 Mouse Model of Familial Alzheimer's Disease: Phenotypic Correlates and Effects of Chronic Oral d-Galactose. ACS Chem Neurosci 2023; 14:4013-4025. [PMID: 37932005 PMCID: PMC10655039 DOI: 10.1021/acschemneuro.3c00495] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 10/26/2023] [Accepted: 10/26/2023] [Indexed: 11/08/2023] Open
Abstract
The gut might play an important role in the etiopathogenesis of Alzheimer's disease (AD) as gastrointestinal alterations often precede the development of neuropathological changes in the brain and correlate with disease progression in animal models. The gut has an immense capacity to generate free radicals whose role in the etiopathogenesis of AD is well-known; however, it remains to be clarified whether gastrointestinal redox homeostasis is associated with the development of AD. The aim was to (i) examine gastrointestinal redox homeostasis in the presymptomatic and symptomatic Tg2576 mouse model of AD; (ii) investigate the effects of oral d-galactose previously shown to alleviate cognitive deficits and metabolic changes in animal models of AD and reduce gastrointestinal oxidative stress; and (iii) investigate the association between gastrointestinal redox biomarkers and behavioral alterations in Tg2576 mice. In the presymptomatic stage, Tg2576 mice displayed an increased gastrointestinal electrophilic tone, characterized by higher lipid peroxidation and elevated Mn/Fe-SOD activity. In the symptomatic stage, these alterations are rectified, but the total antioxidant capacity is decreased. Chronic oral d-galactose increased the antioxidant capacity and reduced lipid peroxidation in the Tg2576 but had the opposite effects in the wild-type animals. The total antioxidant capacity of the gastrointestinal tract was associated with greater spatial memory. Gut redox homeostasis might be involved in the development and progression of AD pathophysiology and should be further explored in this context.
Collapse
Affiliation(s)
- Jan Homolak
- Department
of Pharmacology, University of Zagreb School
of Medicine, Zagreb 10000, Croatia
- Croatian
Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia
- Interfaculty
Institute of Microbiology and Infection Medicine, University of Tübingen, Tübingen 72076, Germany
- Cluster
of Excellence “Controlling Microbes to Fight Infections”, University of Tübingen, Tübingen 72076, Germany
| | - Ana Babic Perhoc
- Department
of Pharmacology, University of Zagreb School
of Medicine, Zagreb 10000, Croatia
- Croatian
Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia
| | - Ana Knezovic
- Department
of Pharmacology, University of Zagreb School
of Medicine, Zagreb 10000, Croatia
- Croatian
Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia
| | - Jelena Osmanovic Barilar
- Department
of Pharmacology, University of Zagreb School
of Medicine, Zagreb 10000, Croatia
- Croatian
Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia
| | - Davor Virag
- Department
of Pharmacology, University of Zagreb School
of Medicine, Zagreb 10000, Croatia
- Croatian
Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia
| | - Melita Salkovic-Petrisic
- Department
of Pharmacology, University of Zagreb School
of Medicine, Zagreb 10000, Croatia
- Croatian
Institute for Brain Research, University of Zagreb School of Medicine, Zagreb 10000, Croatia
| |
Collapse
|
32
|
Zhang Y, Aldamarany WAS, Song G, Liu J, Liu S, Chen Y, Jiang W, Zhong G. Influence of konjac glucomannan and its derivatives on the oral pharmacokinetics of antimicrobial agent in antibiotics cocktails: Keep vigilant on dietary fiber supplement. Int J Biol Macromol 2023; 251:126306. [PMID: 37573922 DOI: 10.1016/j.ijbiomac.2023.126306] [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: 04/11/2023] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 08/15/2023]
Abstract
In this study, konjac glucomannan (KGM) and its derivatives were gavaged as dietary fiber supplements, followed by a single dose of antibiotic cocktail (Abx) containing amoxicillin, neomycin, metronidazole and vancomycin in mice. The effects of dietary fiber on the pharmacokinetics and tissue distribution of each antibiotic were investigated. The results showed that the specific effects of KGM and its derivatives on the absorption, distribution, and elimination of certain antibiotics varied and depended on the nature of the fibers and the characteristics of the antibiotics. Explicitly, the ingestion of KGM and its derivatives enhanced the absorption of metronidazole by 1.7 times and hindered that of amoxicillin by nearly 36 % without affecting the absorption of neomycin sulfate and vancomycin. KGM and its derivatives had no effect on the distribution of amoxicillin and metronidazole, but DKGM and KGM hindered the distributions of neomycin sulfate (from 1.25 h to 1.62 h) and vancomycin (from 0.95 h to 1.14 h), respectively. KGM and its derivatives promoted the elimination of amoxicillin by nearly 38 % while prolonging that of metronidazole by >50 %. KOGM boosted the elimination of neomycin sulfate and vancomycin, but KGM differed from DKGM in acting on the elimination of both.
Collapse
Affiliation(s)
- Yuan Zhang
- College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing 400715, China
| | - Waleed A S Aldamarany
- College of Food Science, Southwest University, Chongqing 400715, China; Food Science and Technology Department, Faculty of Agriculture, Al-Azhar University (Assiut Branch), Assiut 71524, Egypt
| | - Guangming Song
- College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing 400715, China
| | - Jie Liu
- College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing 400715, China
| | - Sha Liu
- College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing 400715, China
| | - Yuanyuan Chen
- College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing 400715, China
| | - Wenjing Jiang
- College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing 400715, China
| | - Geng Zhong
- College of Food Science, Southwest University, Chongqing 400715, China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing 400715, China.
| |
Collapse
|
33
|
Zhang Y, Aldamarany WAS, Deng L, Zhong G. Carbohydrate supplementation retains intestinal barrier and ameliorates bacterial translocation in an antibiotic-induced mouse model. Food Funct 2023; 14:8186-8200. [PMID: 37599609 DOI: 10.1039/d3fo01343j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2023]
Abstract
Bacterial translocation (BT), with antibiotic use as an inducer, is associated with increased risk of developing multiple inflammatory disorders, and is closely associated with intestinal barrier integrity. Deacetylated konjac glucomannan (DKGM) and konjac oligo-glucomannan (KOGM) are two of the most widely used derivatives in the food industry. They are structurally and physiologically distinct from konjac glucomannan (KGM), and previous studies have confirmed their prebiotic effects. But whether they play a role in antibiotic-induced BT is unknown. Here, we applied an antibiotic cocktail (Abx) to a mouse model and investigated whether and how KGM and its derivatives function in BT and inflammation response amelioration during and after antibiotics, and which intervention plan is more effective. The results showed that KGM and its derivatives all inhibited BT. The colon tissue lesions caused by BT were largely alleviated, and short-chain fatty acid (SCFA) production was highly improved with the supplementation of carbohydrates. The prolonged intervention plan using KGM and its derivatives was more efficient than intervention only during the Abx administration period. Among the three dietary fibers, KGM behaved best, while DKGM and KOGM behaved equivalently. Additionally, KGM and its derivatives all reduced the inflammatory response accompanying BT, but DKGM may have a direct inhibitory efficacy in inflammation other than that through IL-10, unlike KGM or KOGM.
Collapse
Affiliation(s)
- Yuan Zhang
- College of Food Science, Southwest University, Chongqing, 400715, China.
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing, 400715, China
| | - Waleed A S Aldamarany
- College of Food Science, Southwest University, Chongqing, 400715, China.
- Food Science and Technology Department, Faculty of Agriculture, Al-Azhar University (Assiut Branch), Assiut 71524, Egypt
| | - Liling Deng
- Chongqing Key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing 401331, China
| | - Geng Zhong
- College of Food Science, Southwest University, Chongqing, 400715, China.
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Southwest University, Chongqing, 400715, China
| |
Collapse
|
34
|
Wang X, Li X, Zhang L, An L, Guo L, Huang L, Gao W. Recent progress in plant-derived polysaccharides with prebiotic potential for intestinal health by targeting gut microbiota: a review. Crit Rev Food Sci Nutr 2023; 64:12242-12271. [PMID: 37651130 DOI: 10.1080/10408398.2023.2248631] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Natural products of plant origin are of high interest and widely used, especially in the food industry, due to their low toxicity and wide range of bioactive properties. Compared to other plant components, the safety of polysaccharides has been generally recognized. As dietary fibers, plant-derived polysaccharides are mostly degraded in the intestine by polysaccharide-degrading enzymes secreted by gut microbiota, and have potential prebiotic activity in both non-disease and disease states, which should not be overlooked, especially in terms of their involvement in the treatment of intestinal diseases and the promotion of intestinal health. This review elucidates the regulatory effects of plant-derived polysaccharides on gut microbiota and summarizes the mechanisms involved in targeting gut microbiota for the treatment of intestinal diseases. Further, the structure-activity relationships between different structural types of plant-derived polysaccharides and the occurrence of their prebiotic activity are further explored. Finally, the practical applications of plant-derived polysaccharides in food production and food packaging are summarized and discussed, providing important references for expanding the application of plant-derived polysaccharides in the food industry or developing functional dietary supplements.
Collapse
Affiliation(s)
- Xiaozhen Wang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Xia Li
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Luyao Zhang
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Lingzhuo An
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Lanping Guo
- National Resource Center for Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, China
| | - Luqi Huang
- National Resource Center for Chinese Materia Medica, Academy of Chinese Medical Sciences, Beijing, China
| | - Wenyuan Gao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
35
|
Penumutchu S, Korry BJ, Hewlett K, Belenky P. Fiber supplementation protects from antibiotic-induced gut microbiome dysbiosis by modulating gut redox potential. Nat Commun 2023; 14:5161. [PMID: 37620319 PMCID: PMC10449846 DOI: 10.1038/s41467-023-40553-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 07/31/2023] [Indexed: 08/26/2023] Open
Abstract
Antibiotic-induced gut dysbiosis (AID) is a frequent and serious side effect of antibiotic use and mitigating this dysbiosis is a critical therapeutic target. We propose that the host diet can modulate the chemical environment of the gut resulting in changes to the structure and function of the microbiome during antibiotic treatment. Gut dysbiosis is typically characterized by increases in aerobic respiratory bacterial metabolism, redox potential, and abundance of Proteobacteria. In this study, we explore dietary fiber supplements as potential modulators of the chemical environment in the gut to reduce this pattern of dysbiosis. Using defined-diets and whole-genome sequencing of female murine microbiomes during diet modulation and antibiotic treatment, we find that fiber prebiotics significantly reduced the impact of antibiotic treatment on microbiome composition and function. We observe reduced abundance of aerobic bacteria as well as metabolic pathways associated with oxidative metabolism. These metatranscriptomic results are corroborated by chemical measurements of eH and pH suggesting that fiber dampens the dysbiotic effects of antibiotics. This work indicates that fiber may act as a potential therapeutic for AID by modulating bacterial metabolism in the gut to prevent an increase in redox potential and protect commensal microbes during antibiotic treatment.
Collapse
Affiliation(s)
- Swathi Penumutchu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Benjamin J Korry
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA
| | - Katharine Hewlett
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
36
|
Jiang Y, Cui W, Zhang Y, Wang T, Zheng X, Li H, Shang J. FG-4592 relieves diabetic kidney disease severity by influencing metabolic profiles via gut microbiota reconstruction in both human and mouse models. Front Physiol 2023; 14:1195441. [PMID: 37654676 PMCID: PMC10465800 DOI: 10.3389/fphys.2023.1195441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 07/25/2023] [Indexed: 09/02/2023] Open
Abstract
Objective: Diabetic kidney disease (DKD) is one of the most prevalent complications of diabetes mellitus (DM) and is highly associated with devastating outcomes. Hypoxia-inducible factor (HIF), the main transcription factor that regulates cellular responses to hypoxia, plays an important role in regulating erythropoietin (EPO) synthesis. FG-4592 is the HIF stabilizer that is widely used in patients with renal anemia. We investigated the effect of FG-4592 on DKD phenotypes and the pharmacologic mechanism from the perspective of gut microbiota and systemic metabolism. Design: We collected the clinical data of 73 participants, including 40 DKD patients with combined renal anemia treated with FG-4592, and 33 clinical index-matched DKD patients without FG-4592 treatment from The First Affiliated Hospital of Zhengzhou University at the beginning and after a 3-6-month follow-up period. We established DKD mouse models treated by FG-4592 and performed fecal microbiota transplantation from FG-4592-treated DKD mice to investigate the effects of FG-4592 on DKD and to understand this mechanism from a microbial perspective. Untargeted metabolome-microbiome combined analysis was implemented to globally delineate the mechanism of FG-4592 from both microbial and metabolomic aspects. Result: DKD phenotypes significantly improved after 3-6 months of FG-4592 treatment in DKD patients combined with renal anemia, including a decreased level of systolic blood pressure, serum creatinine, and increased estimated glomerular infiltration rate. Such effects were also achieved in the DKD mouse model treated with FG-4592 and can be also induced by FG-4592-influenced gut microbiota. Untargeted plasma metabolomics-gut microbiota analysis showed that FG-4592 dramatically altered both the microbial and metabolic profiles of DKD mice and relieved DKD phenotypes via upregulating beneficial gut microbiota-associated metabolites. Conclusion: FG-4592 can globally relieve the symptoms of DKD patients combined with renal anemia. In the animal experiment, FG-4592 can reconstruct the intestinal microbial profiles of DKD to further upregulate the production of gut-associated beneficial metabolites, subsequently improving DKD phenotypes.
Collapse
Affiliation(s)
- Yumin Jiang
- Department of Emergency Intensive Care Unit, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wen Cui
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou University, Zhengzhou, Henan, China
| | - Yiding Zhang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou University, Zhengzhou, Henan, China
| | - Ting Wang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou University, Zhengzhou, Henan, China
| | - Xuejun Zheng
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou University, Zhengzhou, Henan, China
| | - Huangmin Li
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou University, Zhengzhou, Henan, China
| | - Jin Shang
- Department of Nephrology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
37
|
Letourneau J, Walker L, Han SH, David LA, Younge N. Fecal pH and redox as functional markers in the premature infant gut microbiome. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.14.553216. [PMID: 37645803 PMCID: PMC10462032 DOI: 10.1101/2023.08.14.553216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
The infant gut microbiome is a crucial factor in health and development. In preterm infants, altered gut microbiome composition and function have been linked to serious neonatal complications such as necrotizing enterocolitis and sepsis, which can lead to long-term disability. Although many studies have described links between microbiome composition and disease risk, there is a need for biomarkers to identify infants at risk of these complications in practice. In this study, we obtained stool samples from preterm infant participants longitudinally during the first postnatal months, and measured pH and redox, as well as SCFA content and microbiome composition by 16S rRNA gene amplicon sequencing. These outcomes were compared to clinical data to better understand the role of pH and redox in infant gut microbiome development and overall health, and to assess the potential utility of pH and redox as biomarkers. We found that infants born earlier or exposed to antibiotics exhibited increased fecal pH, and that redox potential increased with postnatal age. These differences may be linked to changes in SCFA content, which was correlated with pH and increased with age. Microbiome composition was also related to birth weight, age, pH, and redox. Our findings suggest that pH and redox may serve as biomarkers of metabolic state in the preterm infant gut.
Collapse
Affiliation(s)
- Jeffrey Letourneau
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27708
| | - LaShawndra Walker
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27708
| | - Se Hyang Han
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27708
| | - Lawrence A David
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC 27708
- Program in Computational Biology and Bioinformatics, Duke University School of Medicine, Durham, NC 27708
| | - Noelle Younge
- Department of Pediatrics, Duke University School of Medicine, Durham, NC 27708
| |
Collapse
|
38
|
Horrocks V, King OG, Yip AYG, Marques IM, McDonald JAK. Role of the gut microbiota in nutrient competition and protection against intestinal pathogen colonization. MICROBIOLOGY (READING, ENGLAND) 2023; 169:001377. [PMID: 37540126 PMCID: PMC10482380 DOI: 10.1099/mic.0.001377] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/25/2023] [Indexed: 08/05/2023]
Abstract
The human gut microbiota can restrict the growth of pathogens to prevent them from colonizing the intestine ('colonization resistance'). However, antibiotic treatment can kill members of the gut microbiota ('gut commensals') and reduce competition for nutrients, making these nutrients available to support the growth of pathogens. This disturbance can lead to the growth and expansion of pathogens within the intestine (including antibiotic-resistant pathogens), where these pathogens can exploit the absence of competitors and the nutrient-enriched gut environment. In this review, we discuss nutrient competition between the gut microbiota and pathogens. We also provide an overview of how nutrient competition can be harnessed to support the design of next-generation microbiome therapeutics to restrict the growth of pathogens and prevent the development of invasive infections.
Collapse
Affiliation(s)
- Victoria Horrocks
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Olivia G. King
- Centre for Bacterial Resistance Biology, Department of Infectious Disease, Imperial College London, London SW7 2AZ, UK
| | - Alexander Y. G. Yip
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Inês Melo Marques
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| | - Julie A. K. McDonald
- Centre for Bacterial Resistance Biology, Department of Life Sciences, Imperial College London, London SW7 2AZ, UK
| |
Collapse
|
39
|
Inda-Webb ME, Jimenez M, Liu Q, Phan NV, Ahn J, Steiger C, Wentworth A, Riaz A, Zirtiloglu T, Wong K, Ishida K, Fabian N, Jenkins J, Kuosmanen J, Madani W, McNally R, Lai Y, Hayward A, Mimee M, Nadeau P, Chandrakasan AP, Traverso G, Yazicigil RT, Lu TK. Sub-1.4 cm 3 capsule for detecting labile inflammatory biomarkers in situ. Nature 2023; 620:386-392. [PMID: 37495692 DOI: 10.1038/s41586-023-06369-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 06/26/2023] [Indexed: 07/28/2023]
Abstract
Transient molecules in the gastrointestinal tract such as nitric oxide and hydrogen sulfide are key signals and mediators of inflammation. Owing to their highly reactive nature and extremely short lifetime in the body, these molecules are difficult to detect. Here we develop a miniaturized device that integrates genetically engineered probiotic biosensors with a custom-designed photodetector and readout chip to track these molecules in the gastrointestinal tract. Leveraging the molecular specificity of living sensors1, we genetically encoded bacteria to respond to inflammation-associated molecules by producing luminescence. Low-power electronic readout circuits2 integrated into the device convert the light emitted by the encapsulated bacteria to a wireless signal. We demonstrate in vivo biosensor monitoring in the gastrointestinal tract of small and large animal models and the integration of all components into a sub-1.4 cm3 form factor that is compatible with ingestion and capable of supporting wireless communication. With this device, diseases such as inflammatory bowel disease could be diagnosed earlier than is currently possible, and disease progression could be more accurately tracked. The wireless detection of short-lived, disease-associated molecules with our device could also support timely communication between patients and caregivers, as well as remote personalized care.
Collapse
Affiliation(s)
- M E Inda-Webb
- Synthetic Biology Group, MIT Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - M Jimenez
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Q Liu
- Electrical and Computer Engineering Department, Boston University, Boston, MA, USA
| | - N V Phan
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J Ahn
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - C Steiger
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - A Wentworth
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - A Riaz
- Electrical and Computer Engineering Department, Boston University, Boston, MA, USA
| | - T Zirtiloglu
- Electrical and Computer Engineering Department, Boston University, Boston, MA, USA
| | - K Wong
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - K Ishida
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - N Fabian
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Comparative Medicine, MIT, Cambridge, MA, USA
| | - J Jenkins
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - J Kuosmanen
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - W Madani
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - R McNally
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Y Lai
- Synthetic Biology Group, MIT Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Research Laboratory of Electronics, Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - A Hayward
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Division of Comparative Medicine, MIT, Cambridge, MA, USA
| | - M Mimee
- Department of Microbiology, Biological Sciences Division and Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
| | | | - A P Chandrakasan
- Department of Electrical Engineering and Computer Science, MIT, Cambridge, MA, USA
| | - G Traverso
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- The Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Division of Gastroenterology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - R T Yazicigil
- Electrical and Computer Engineering Department, Boston University, Boston, MA, USA.
| | - T K Lu
- Synthetic Biology Group, MIT Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Research Laboratory of Electronics, Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Senti Biosciences, South San Francisco, CA, USA.
| |
Collapse
|
40
|
Sun P, Wang M, Liu YX, Li L, Chai X, Zheng W, Chen S, Zhu X, Zhao S. High-fat diet-disturbed gut microbiota-colonocyte interactions contribute to dysregulating peripheral tryptophan-kynurenine metabolism. MICROBIOME 2023; 11:154. [PMID: 37468922 PMCID: PMC10355067 DOI: 10.1186/s40168-023-01606-x] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 06/20/2023] [Indexed: 07/21/2023]
Abstract
BACKGROUND Aberrant tryptophan (Trp)-kynurenine (Kyn) metabolism has been implicated in the pathogenesis of human disease. In particular, populations with long-term western-style diets are characterized by an excess of Kyn in the plasma. Host-gut microbiota interactions are dominated by diet and are essential for maintaining host metabolic homeostasis. However, the role of western diet-disturbed gut microbiota-colonocyte interactions in Trp metabolism remains to be elucidated. RESULTS Here, 4-week-old mice were fed with a high-fat diet (HFD), representing a typical western diet, for 4 weeks, and multi-omics approaches were adopted to determine the mechanism by which HFD disrupted gut microbiota-colonocyte interplay causing serum Trp-Kyn metabolism dysfunction. Our results showed that colonocyte-microbiota interactions dominated the peripheral Kyn pathway in HFD mice. Mechanistically, persistent HFD-impaired mitochondrial bioenergetics increased colonic epithelial oxygenation and caused metabolic reprogramming in colonites to support the expansion of Proteobacteria in the colon lumen. Phylum Proteobacteria-derived lipopolysaccharide (LPS) stimulated colonic immune responses to upregulate the indoleamine 2,3-dioxygenase 1 (IDO1)-mediated Kyn pathway, leading to Trp depletion and Kyn accumulation in the circulation, which was further confirmed by transplantation of Escherichia coli (E.coli) indicator strains and colonic IDO1 depletion. Butyrate supplementation promoted mitochondrial functions in colonocytes to remodel the gut microbiota in HFD mice, consequently ameliorating serum Kyn accumulation. CONCLUSIONS Our results highlighted that HFD disrupted the peripheral Kyn pathway in a gut microbiota-dependent manner and that the continuous homeostasis of gut bacteria-colonocytes interplay played a central role in the regulation of host peripheral Trp metabolism. Meanwhile, this study provided new insights into therapies against western diet-related metabolic disorders. Video Abstract.
Collapse
Affiliation(s)
- Penghao Sun
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Mengli Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Yong-Xin Liu
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, 518120, Guangdong, China
| | - Luqi Li
- Life Science Research Core Services, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xuejun Chai
- College of Basic Medicine, Xi'an Medical University, Xi'an, 710000, Shaanxi, China.
| | - Wei Zheng
- College of Resources and Environment Sciences, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Shulin Chen
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China
| | - Xiaoyan Zhu
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| | - Shanting Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, 712100, Shaanxi, China.
| |
Collapse
|
41
|
Molina RA, Villar MD, Miranda MH, Maldonado NC, Vignolo GM, Nader-Macías MEF. A multi-strain probiotic promoted recovery of puppies from gastroenteritis in a randomized, double-blind, placebo-controlled study. THE CANADIAN VETERINARY JOURNAL = LA REVUE VETERINAIRE CANADIENNE 2023; 64:666-673. [PMID: 37397694 PMCID: PMC10286135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 07/04/2023]
Abstract
Objective and animals Acute diarrhea is among the most common causes of veterinary consultations for dogs. A double-blind, placebocontrolled intervention trial was done with 120 puppies with gastroenteritis. These dogs were 1 to 4 mo old, male and female, of various breeds and sizes. Procedure Dogs were randomly allocated into 2 groups: Those in the treated group (TG) received a multi-strain probiotic with Lactobacillus johnsonii CRL1693, Ligilactobacillus murinus CRL1695, Limosilactobacillus mucosae CRL1696, and Ligilactobacillus salivarius CRL1702 (1 × 109 CFU/mL) daily for 7 d, whereas those in the control group (CG) received a placebo. All puppies received intravenous fluids, an antiparasitic, amoxicillin PO, and enrofloxacin SC. Results At the start of the trial, the 2 groups were similar. Probiotic administration for 7 d normalized fecal consistency, with 69, 50, and 80% of small, medium, and large puppies in the TG achieving a fecal score of 1 (separate hard lumps) at 7 d, significantly better than puppies in the CG. After 7 d of treatment, most puppies (70%) in the TG had an excellent recovery, whereas in the CG, recoveries were 35.7% "bad" and 30.4% "fair." Therefore, treatment with probiotics hastened recovery (P < 0.0001). At the end of the trial, there was a significant increase of cultivable lactobacilli in the feces of TG puppies, but no significant differences between the 2 groups in numbers of total mesophylls, enterobacteria, or Gram-positive cocci. Total mortality was 5.8%, including 4 puppies from the CG and 3 from the TG. Conclusion In a randomized, double-blind, placebo-controlled study, puppies with gastroenteritis symptoms receiving a multi-strain probiotic had rapid improvement, implying beneficial effects on the microbiota and its functionality.
Collapse
Affiliation(s)
- Rosa A Molina
- Centro de Referencia para Lactobacilos (CERELA), CONICET, Chacabuco 145, (T4000) Tucumán, Argentina (Molina, Miranda, Maldonado, Vignolo, Nader-Macías); Facultad de Agronomía y Zootecnia (Molina) and Cátedra de Bioestadística, Facultad de Medicina (D'Urso Villar), Universidad Nacional de Tucumán (UNT), (T4000) Tucumán, Argentina
| | - Marcela D'Urso Villar
- Centro de Referencia para Lactobacilos (CERELA), CONICET, Chacabuco 145, (T4000) Tucumán, Argentina (Molina, Miranda, Maldonado, Vignolo, Nader-Macías); Facultad de Agronomía y Zootecnia (Molina) and Cátedra de Bioestadística, Facultad de Medicina (D'Urso Villar), Universidad Nacional de Tucumán (UNT), (T4000) Tucumán, Argentina
| | - María H Miranda
- Centro de Referencia para Lactobacilos (CERELA), CONICET, Chacabuco 145, (T4000) Tucumán, Argentina (Molina, Miranda, Maldonado, Vignolo, Nader-Macías); Facultad de Agronomía y Zootecnia (Molina) and Cátedra de Bioestadística, Facultad de Medicina (D'Urso Villar), Universidad Nacional de Tucumán (UNT), (T4000) Tucumán, Argentina
| | - Natalia C Maldonado
- Centro de Referencia para Lactobacilos (CERELA), CONICET, Chacabuco 145, (T4000) Tucumán, Argentina (Molina, Miranda, Maldonado, Vignolo, Nader-Macías); Facultad de Agronomía y Zootecnia (Molina) and Cátedra de Bioestadística, Facultad de Medicina (D'Urso Villar), Universidad Nacional de Tucumán (UNT), (T4000) Tucumán, Argentina
| | - Graciela M Vignolo
- Centro de Referencia para Lactobacilos (CERELA), CONICET, Chacabuco 145, (T4000) Tucumán, Argentina (Molina, Miranda, Maldonado, Vignolo, Nader-Macías); Facultad de Agronomía y Zootecnia (Molina) and Cátedra de Bioestadística, Facultad de Medicina (D'Urso Villar), Universidad Nacional de Tucumán (UNT), (T4000) Tucumán, Argentina
| | - María E F Nader-Macías
- Centro de Referencia para Lactobacilos (CERELA), CONICET, Chacabuco 145, (T4000) Tucumán, Argentina (Molina, Miranda, Maldonado, Vignolo, Nader-Macías); Facultad de Agronomía y Zootecnia (Molina) and Cátedra de Bioestadística, Facultad de Medicina (D'Urso Villar), Universidad Nacional de Tucumán (UNT), (T4000) Tucumán, Argentina
| |
Collapse
|
42
|
Vitetta L, Bambling M, Strodl E. Probiotics and Commensal Bacteria Metabolites Trigger Epigenetic Changes in the Gut and Influence Beneficial Mood Dispositions. Microorganisms 2023; 11:1334. [PMID: 37317308 DOI: 10.3390/microorganisms11051334] [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: 05/05/2023] [Revised: 05/15/2023] [Accepted: 05/17/2023] [Indexed: 06/16/2023] Open
Abstract
The effect of the intestinal microbiome on the gut-brain axis has received considerable attention, strengthening the evidence that intestinal bacteria influence emotions and behavior. The colonic microbiome is important to health and the pattern of composition and concentration varies extensively in complexity from birth to adulthood. That is, host genetics and environmental factors are complicit in shaping the development of the intestinal microbiome to achieve immunological tolerance and metabolic homeostasis from birth. Given that the intestinal microbiome perseveres to maintain gut homeostasis throughout the life cycle, epigenetic actions may determine the effect on the gut-brain axis and the beneficial outcomes on mood. Probiotics are postulated to exhibit a range of positive health benefits including immunomodulating capabilities. Lactobacillus and Bifidobacterium are genera of bacteria found in the intestines and so far, the benefits afforded by ingesting bacteria such as these as probiotics to people with mood disorders have varied in efficacy. Most likely, the efficacy of probiotic bacteria at improving mood has a multifactorial dependency, relying namely on several factors that include the agents used, the dose, the pattern of dosing, the pharmacotherapy used, the characteristics of the host and the underlying luminal microbial environment (e.g., gut dysbiosis). Clarifying the pathways linking probiotics with improvements in mood may help identify the factors that efficacy is dependent upon. Adjunctive therapies with probiotics for mood disorders could, through DNA methylation molecular mechanisms, augment the intestinal microbial active cohort and endow its mammalian host with important and critical co-evolutionary redox signaling metabolic interactions, that are embedded in bacterial genomes, and that in turn can enhance beneficial mood dispositions.
Collapse
Affiliation(s)
- Luis Vitetta
- Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2005, Australia
| | - Matthew Bambling
- Faculty of Medicine and Health, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Esben Strodl
- Faculty of Health, Queensland University of Technology, Brisbane, QLD 4058, Australia
| |
Collapse
|
43
|
Mazumder MHH, Gandhi J, Majumder N, Wang L, Cumming RI, Stradtman S, Velayutham M, Hathaway QA, Shannahan J, Hu G, Nurkiewicz TR, Tighe RM, Kelley EE, Hussain S. Lung-gut axis of microbiome alterations following co-exposure to ultrafine carbon black and ozone. Part Fibre Toxicol 2023; 20:15. [PMID: 37085867 PMCID: PMC10122302 DOI: 10.1186/s12989-023-00528-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Accepted: 04/17/2023] [Indexed: 04/23/2023] Open
Abstract
BACKGROUND Microbial dysbiosis is a potential mediator of air pollution-induced adverse outcomes. However, a systemic comparison of the lung and gut microbiome alterations and lung-gut axis following air pollution exposure is scant. In this study, we exposed male C57BL/6J mice to inhaled air, CB (10 mg/m3), O3 (2 ppm) or CB + O3 mixture for 3 h/day for either one day or four consecutive days and were euthanized 24 h post last exposure. The lung and gut microbiome were quantified by 16 s sequencing. RESULTS Multiple CB + O3 exposures induced an increase in the lung inflammatory cells (neutrophils, eosinophils and B lymphocytes), reduced absolute bacterial load in the lungs and increased load in the gut. CB + O3 exposure was more potent as it decreased lung microbiome alpha diversity just after a single exposure. CB + O3 co-exposure uniquely increased Clostridiaceae and Prevotellaceae in the lungs. Serum short chain fatty acids (SCFA) (acetate and propionate) were increased significantly only after CB + O3 co-exposure. A significant increase in SCFA producing bacterial families (Ruminococcaceae, Lachnospiraceae, and Eubacterium) were also observed in the gut after multiple exposures. Co-exposure induced significant alterations in the gut derived metabolite receptors/mediator (Gcg, Glp-1r, Cck) mRNA expression. Oxidative stress related mRNA expression in lungs, and oxidant levels in the BALF, serum and gut significantly increased after CB + O3 exposures. CONCLUSION Our study confirms distinct gut and lung microbiome alterations after CB + O3 inhalation co-exposure and indicate a potential homeostatic shift in the gut microbiome to counter deleterious impacts of environmental exposures on metabolic system.
Collapse
Affiliation(s)
- Md Habibul Hasan Mazumder
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Jasleen Gandhi
- Department of Microbiology, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Nairrita Majumder
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Lei Wang
- Department of Microbiology, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Robert Ian Cumming
- Department of Medicine, Duke University Medical Center, Durham, NC, 2927, USA
| | - Sydney Stradtman
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Murugesan Velayutham
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Quincy A Hathaway
- Heart and Vascular Institute, School of Medicine, West Virginia University, Morgantown, WV, USA
| | - Jonathan Shannahan
- School of Health Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Gangqing Hu
- Department of Microbiology, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Timothy R Nurkiewicz
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Robert M Tighe
- Department of Medicine, Duke University Medical Center, Durham, NC, 2927, USA
| | - Eric E Kelley
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Salik Hussain
- Department of Physiology, Pharmacology, and Toxicology, Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA.
- Center for Inhalation Toxicology (iTOX), School of Medicine, West Virginia University, Morgantown, WV, 26506, USA.
- Department of Microbiology, School of Medicine, West Virginia University, Morgantown, WV, 26506, USA.
| |
Collapse
|
44
|
Yin Y, Sichler A, Ecker J, Laschinger M, Liebisch G, Höring M, Basic M, Bleich A, Zhang XJ, Kübelsbeck L, Plagge J, Scherer E, Wohlleber D, Wang J, Wang Y, Steffani M, Stupakov P, Gärtner Y, Lohöfer F, Mogler C, Friess H, Hartmann D, Holzmann B, Hüser N, Janssen KP. Gut microbiota promote liver regeneration through hepatic membrane phospholipid biosynthesis. J Hepatol 2023; 78:820-835. [PMID: 36681162 DOI: 10.1016/j.jhep.2022.12.028] [Citation(s) in RCA: 62] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 12/23/2022] [Accepted: 12/27/2022] [Indexed: 01/19/2023]
Abstract
BACKGROUND & AIMS Hepatocyte growth and proliferation depends on membrane phospholipid biosynthesis. Short-chain fatty acids (SCFAs) generated by bacterial fermentation, delivered through the gut-liver axis, significantly contribute to lipid biosynthesis. We therefore hypothesized that dysbiotic insults like antibiotic treatment not only affect gut microbiota, but also impair hepatic lipid synthesis and liver regeneration. METHODS Stable isotope labeling and 70% partial hepatectomy (PHx) was carried out in C57Bl/6J wild-type mice, in mice treated with broad-spectrum antibiotics, in germ-free mice and mice colonized with minimal microbiota. The microbiome was analyzed by 16S rRNA gene sequencing and microbial culture. Gut content, liver, blood and primary hepatocyte organoids were tested by mass spectrometry-based lipidomics, quantitative reverse-transcription PCR (qRT-PCR), immunoblot and immunohistochemistry for expression of proliferative and lipogenic markers. Matched biopsies from hyperplastic and hypoplastic liver tissue of patients subjected to surgical intervention to induce hyperplasia were analyzed by qRT-PCR for lipogenic enzymes. RESULTS Three days of antibiotic treatment induced persistent dysbiosis with significantly decreased beta-diversity and richness, but a massive increase of Proteobacteria, accompanied by decreased colonic SCFAs. After PHx, antibiotic-treated mice showed delayed liver regeneration, increased mortality, impaired hepatocyte proliferation and decreased hepatic phospholipid synthesis. Expression of the lipogenic enzyme SCD1 was upregulated after PHx but delayed by antibiotic treatment. Germ-free mice essentially recapitulated the phenotype of antibiotic treatment. Phospholipid biosynthesis, hepatocyte proliferation, liver regeneration and survival were rescued in gnotobiotic mice colonized with a minimal SCFA-producing microbial community. SCFAs induced the growth of murine hepatocyte organoids and hepatic SCD1 expression in mice. Further, SCD1 was required for proliferation of human hepatoma cells and was associated with liver regeneration in human patients. CONCLUSION Gut microbiota are pivotal for hepatic membrane phospholipid biosynthesis and liver regeneration. IMPACT AND IMPLICATIONS Gut microbiota affect hepatic lipid metabolism through the gut-liver axis, but the underlying mechanisms are poorly understood. Perturbations of the gut microbiome, e.g. by antibiotics, impair the production of bacterial metabolites, which normally serve as building blocks for membrane lipids in liver cells. As a consequence, liver regeneration and survival after liver surgery is severely impaired. Even though this study is preclinical, its results might allow physicians in the future to improve patient outcomes after liver surgery, by modulation of gut microbiota or their metabolites.
Collapse
Affiliation(s)
- Yuhan Yin
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Anna Sichler
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Josef Ecker
- ZIEL - Inst. for Food & Health, TUM, Freising, Germany
| | - Melanie Laschinger
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Gerhard Liebisch
- Inst. of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Marcus Höring
- Inst. of Clinical Chemistry and Laboratory Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science, Hannover Medical School, Germany
| | - André Bleich
- Institute for Laboratory Animal Science, Hannover Medical School, Germany
| | - Xue-Jun Zhang
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Ludwig Kübelsbeck
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | | | - Emely Scherer
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Dirk Wohlleber
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Jianye Wang
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Yang Wang
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Marcella Steffani
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Pavel Stupakov
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Yasmin Gärtner
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Fabian Lohöfer
- Department of Diagnostic and Interventional Radiology, School of Medicine, Klinikum rechts der Isar, TUM, Munich, Germany
| | - Carolin Mogler
- Institute of Pathology, School of Medicine, TUM, Munich, Germany
| | - Helmut Friess
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Daniel Hartmann
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany
| | - Bernhard Holzmann
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany.
| | - Norbert Hüser
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany.
| | - Klaus-Peter Janssen
- Dept. of Surgery, School of Medicine, Klinikum rechts der Isar, Technical University of Munich (TUM), Munich, Germany.
| |
Collapse
|
45
|
Zhang Y, Qi X, Zhang Z, Jin Z, Wang G, Ling F. Effects of dietary Cetobacterium somerae on the intestinal health, immune parameters and resistance against Nocardia seriolae of largemouth bass, Micropterus salmoides. FISH & SHELLFISH IMMUNOLOGY 2023; 135:108693. [PMID: 36940785 DOI: 10.1016/j.fsi.2023.108693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 03/12/2023] [Accepted: 03/14/2023] [Indexed: 06/18/2023]
Abstract
Largemouth bass (Micropterus salmoides), one of the most important freshwater commercial fish species has been widely cultivated in China. In recent years, the nocardiosis caused by Nocardia seriolae has greatly damaged the M. salmoides industry and there is no effective treatment at present. Currently, Cetobacterium somerae, the predominant bacteria in the gut of many freshwater fishes has been reported to be associated with fish health. However, whether the native C. somerae could protect the host from N. seriolae is unclear. In this study, M. salmoides were fed with three different diets, including control diet (CD), low C. somerae diet (106 CFU/g as LD) and high C. somerae diet (108 CFU/g as HD). After 8-week feeding, growth performance, gut health index, serum enzyme activities and the expression of inflammation-related genes were tested. Results showed that the LD and HD diets had no adverse effects on the growth performance. Moreover, dietary HD enhanced gut barrier and reduced intestinal ROS and ORP, as well as increased serum enzyme activities including ACP, AKP, SOD and LZM compared to the CD group. In addition, the HD diet significantly up-regulated the expression of TNF-α, IL8, IL-1β and IL15, while down-regulating the expression of TGF-β1 and IL10 in kidney. Moreover, the expression of antibacterial genes was significantly increased in HD group after being challenged by N. seriolae. And the fish fed HD diet exhibited higher survival rate (57.5%) than that in CD (37.5%) and LD groups (42.5%). To summarize, our study demonstrates that dietary HD can enhance gut health, improve immune response and strengthen pathogen resistance, suggesting that C. somerae is a potential probiotic for defending against N. seriolae infection in M. salmoides.
Collapse
Affiliation(s)
- Yong Zhang
- Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China
| | - Xiaozhou Qi
- Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China
| | - Zhongyu Zhang
- Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China
| | - Zhanlin Jin
- Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China
| | - Gaoxue Wang
- Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China; Key Laboratory of Livestock Biology, Northwest A&F University, China.
| | - Fei Ling
- Northwest A&F University, Xinong Road 22nd, Yangling, Shaanxi, 712100, China; Key Laboratory of Livestock Biology, Northwest A&F University, China.
| |
Collapse
|
46
|
Xu R, Li Q, Wang H, Su Y, Zhu W. Reduction of Redox Potential Exerts a Key Role in Modulating Gut Microbial Taxa and Function by Dietary Supplementation of Pectin in a Pig Model. Microbiol Spectr 2023; 11:e0328322. [PMID: 36475916 PMCID: PMC9927287 DOI: 10.1128/spectrum.03283-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Accepted: 11/22/2022] [Indexed: 12/13/2022] Open
Abstract
Pectin exists in a vast range of plants and has a long history of acting as a functional food additive with potential prebiotic effects on intestinal health. However, knowledge of how pectin regulates gut microbial communities is still insufficient and limited. Here, metatranscriptome sequencing revealed that a pectin-enriched diet (PEC) decreased the abundances of fungal keystone taxa (e.g., amino acid-producing Kazachstania spp.) and their genes involved in oxidative phosphorylation, while it increased the abundance of sulfate-reducing Desulfovibrio spp., and methane-producing Methanobrevibacter spp. in colon microbiomes. Furthermore, we first confirmed that PEC decreased fecal redox potential in a fistula pig model, which could be supported by the enrichment of antioxidants (e.g., inosine) in feces. Fecal metagenome analysis disclosed that certain microbial taxa promoted inosine biosynthesis from pectin degradation, including Prevotella, which plays an essential role in pectin biodegradation. Overall, these results demonstrate that pectin decreases the redox potential in pig hindgut to modulate microbial composition and functions, and specific microorganisms generate reducing agents in the course of pectin degradation to decrease redox potential of microbial ecosystem. IMPORTANCE Collective studies indicate that pectin degradation promotes extensive microorganisms that can be involved in pectin degradation directly or indirectly, or benefit from the altered physiological conditions caused by pectin ingestions. Our study focuses on effects of pectin on gut microbial taxa and functions, as well as its interactions with altered environmental features. Our results demonstrate pectin-induced proreducing shifts on colon microbial taxa and functions, and first confirm that pectin decreases hindgut redox potential, which is an important environmental feature that can modulate microbial communities. These results infer that there is bidirectional regulation between microbiota and redox potential during pectin degradation. In general, this investigation proposes new insights into the pectin-modulating gut microbial ecosystem and also provides new perspectives for targeting modulation of gut microbiota.
Collapse
Affiliation(s)
- Rongying Xu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Qiuke Li
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Hongyu Wang
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, Jiangsu Key Laboratory of Gastrointestinal Nutrition and Animal Health, College of Animal Science and Technology, Nanjing Agricultural University, Nanjing, China
- National Center for International Research on Animal Gut Nutrition, Nanjing Agricultural University, Nanjing, China
| |
Collapse
|
47
|
Role of Mitophagy in Regulating Intestinal Oxidative Damage. Antioxidants (Basel) 2023; 12:antiox12020480. [PMID: 36830038 PMCID: PMC9952109 DOI: 10.3390/antiox12020480] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 02/09/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023] Open
Abstract
The mitochondrion is also a major site for maintaining redox homeostasis between reactive oxygen species (ROS) generation and scavenging. The quantity, quality, and functional integrity of mitochondria are crucial for regulating intracellular homeostasis and maintaining the normal physiological function of cells. The role of oxidative stress in human disease is well established, particularly in inflammatory bowel disease and gastrointestinal mucosal diseases. Oxidative stress could result from an imbalance between ROS and the antioxidative system. Mitochondria are both the main sites of production and the main target of ROS. It is a vicious cycle in which initial ROS-induced mitochondrial damage enhanced ROS production that, in turn, leads to further mitochondrial damage and eventually massive intestinal cell death. Oxidative damage can be significantly mitigated by mitophagy, which clears damaged mitochondria. In this review, we aimed to review the molecular mechanisms involved in the regulation of mitophagy and oxidative stress and their relationship in some intestinal diseases. We believe the reviews can provide new ideas and a scientific basis for researching antioxidants and preventing diseases related to oxidative damage.
Collapse
|
48
|
Baltsavias S, Van Treuren W, Sawaby A, Baker SW, Sonnenburg JL, Arbabian A. Gut Microbiome Redox Sensors With Ultrasonic Wake-Up and Galvanic Coupling Wireless Links. IEEE Trans Biomed Eng 2023; 70:76-87. [PMID: 35727787 PMCID: PMC9911315 DOI: 10.1109/tbme.2022.3184972] [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] [Indexed: 11/07/2022]
Abstract
Tools to measure in vivo redox activity of the gut microbiome and its influence on host health are lacking. In this paper, we present the design of new in vivo gut oxidation-reduction potential (ORP) sensors for rodents, to study host-microbe and microbe-environment interactions throughout the gut. These are the first in vivo sensors to combine ultrasonic wake-up and galvanic coupling telemetry, allowing for sensor miniaturization, experiment flexibility, and robust wireless measurements in live rodents. A novel study of in situ ORP along the intestine reveals biogeographical redox features that the ORP sensors can uniquely access in future gut microbiome studies.
Collapse
|
49
|
Wang Y, Zhang Z, Li B, He B, Li L, Nice EC, Zhang W, Xu J. New Insights into the Gut Microbiota in Neurodegenerative Diseases from the Perspective of Redox Homeostasis. Antioxidants (Basel) 2022; 11:2287. [PMID: 36421473 PMCID: PMC9687622 DOI: 10.3390/antiox11112287] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 08/27/2023] Open
Abstract
An imbalance between oxidants and antioxidants in the body can lead to oxidative stress, which is one of the major causes of neurodegenerative diseases. The gut microbiota contains trillions of beneficial bacteria that play an important role in maintaining redox homeostasis. In the last decade, the microbiota-gut-brain axis has emerged as a new field that has revolutionized the study of the pathology, diagnosis, and treatment of neurodegenerative diseases. Indeed, a growing number of studies have found that communication between the brain and the gut microbiota can be accomplished through the endocrine, immune, and nervous systems. Importantly, dysregulation of the gut microbiota has been strongly associated with the development of oxidative stress-mediated neurodegenerative diseases. Therefore, a deeper understanding of the relationship between the gut microbiota and redox homeostasis will help explain the pathogenesis of neurodegenerative diseases from a new perspective and provide a theoretical basis for proposing new therapeutic strategies for neurodegenerative diseases. In this review, we will describe the role of oxidative stress and the gut microbiota in neurodegenerative diseases and the underlying mechanisms by which the gut microbiota affects redox homeostasis in the brain, leading to neurodegenerative diseases. In addition, we will discuss the potential applications of maintaining redox homeostasis by modulating the gut microbiota to treat neurodegenerative diseases, which could open the door for new therapeutic approaches to combat neurodegenerative diseases.
Collapse
Affiliation(s)
- Yu Wang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Zhe Zhang
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bowen Li
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Bo He
- West China School of Basic Medical Sciences & Forensic Medicine, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, China
| | - Lei Li
- School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Edouard C. Nice
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | - Wei Zhang
- West China Biomedical Big Data Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Mental Health Center and Psychiatric Laboratory, the State Key Laboratory of Biotherapy, West China Hospital of Sichuan University, Chengdu 610000, China
| | - Jia Xu
- School of Medicine, Ningbo University, Ningbo 315211, China
| |
Collapse
|
50
|
Shi Z, Zhang J, Jiang Y, Wen Y, Gao Z, Deng W, Yin Y, Zhu F. Two low-toxic Klebsiella pneumoniae strains from gut of black soldier fly Hermetia illucens are multi-resistance to sulfonamides and cadmium. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2022; 312:120062. [PMID: 36049579 DOI: 10.1016/j.envpol.2022.120062] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 06/15/2023]
Abstract
In recent years, pollution of antibiotics and heavy metal has often been reported in organic wastes. Saprophytic insects have been recorded as biological control agents in organic waste management. During organic waste conversion, the intestinal bacteria of the saprophytic insects play an important role in digestion, physiology, immunity and prevention of pathogen colonization. Black soldier fly (BSF) Hermetia illucens has been widely used as saprophytic insects and showed tolerance to sulfonamides (SAs) and cadmium (Cd). Diversity and changes in gut microbiota of black soldier fly larvae (BSFL) were evaluated through 16S rRNA high-throughput sequencing, and a decrease in diversity of gut microbiota along with an increase in SAs stress was recorded. Major members identified were Actinomycetaceae, Enterobacteriaceae, and Enterococcaceae. And fourteen multi-resistance Klebsiella pneumoniae strains were isolated. Two strains BSFL7-B-5 (from middle midgut of 7-day BSFL) and BSFL11-C-1 (from posterior midgut of 11-day BSFL) were found to be low-toxic and multi-resistance. The adsorption rate of SAs in 5 mg/kg solutions by these two strains reached 65.2% and 61.6%, respectively. Adsorption rate of Cd in 20 mg/L solutions was 77.2% for BSFL7-B-5. The strain BSFL11-C-1 showed higher than 70% adsorption rates of Cd in 20, 30 and 40 mg/L solutions. This study revealed that the presence of multi-resistance bacterial strains in the gut of BSFL helped the larvae against SAs or Cd stress. After determining how and where they are used, selected BSFL gut bacterial strains might be utilized in managing SAs or Cd contamination at suitable concentrations in the future.
Collapse
Affiliation(s)
- Zhihui Shi
- Hubei International Scientific and Technological Cooperation Base of Waste Conversion by Insects, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Jie Zhang
- Hubei International Scientific and Technological Cooperation Base of Waste Conversion by Insects, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Yijie Jiang
- Hubei International Scientific and Technological Cooperation Base of Waste Conversion by Insects, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Yiting Wen
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Zhenghui Gao
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Wenhui Deng
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Yumeng Yin
- Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Fen Zhu
- Hubei International Scientific and Technological Cooperation Base of Waste Conversion by Insects, Huazhong Agricultural University, Wuhan, 430070, China; Hubei Insect Resources Utilization and Sustainable Pest Management Key Laboratory, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|