1
|
Sastre-Dominguez J, DelaFuente J, Toribio-Celestino L, Herencias C, Herrador-Gómez P, Costas C, Hernández-García M, Cantón R, Rodríguez-Beltrán J, Santos-Lopez A, San Millan A. Plasmid-encoded insertion sequences promote rapid adaptation in clinical enterobacteria. Nat Ecol Evol 2024; 8:2097-2112. [PMID: 39198572 PMCID: PMC7616626 DOI: 10.1038/s41559-024-02523-4] [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: 03/15/2024] [Accepted: 07/23/2024] [Indexed: 09/01/2024]
Abstract
Plasmids are extrachromosomal genetic elements commonly found in bacteria. They are known to fuel bacterial evolution through horizontal gene transfer, and recent analyses indicate that they can also promote intragenomic adaptations. However, the role of plasmids as catalysts of bacterial evolution beyond horizontal gene transfer is poorly explored. In this study, we investigated the impact of a widespread conjugative plasmid, pOXA-48, on the evolution of several multidrug-resistant clinical enterobacteria. Combining experimental and within-patient evolution analyses, we unveiled that plasmid pOXA-48 promotes bacterial evolution through the transposition of plasmid-encoded insertion sequence 1 (IS1) elements. Specifically, IS1-mediated gene inactivation expedites the adaptation rate of clinical strains in vitro and fosters within-patient adaptation in the gut microbiota. We deciphered the mechanism underlying the plasmid-mediated surge in IS1 transposition, revealing a negative feedback loop regulated by the genomic copy number of IS1. Given the overrepresentation of IS elements in bacterial plasmids, our findings suggest that plasmid-mediated IS1 transposition represents a crucial mechanism for swift bacterial adaptation.
Collapse
Affiliation(s)
| | | | | | - Cristina Herencias
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal-Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- Centro de Investigación Biológica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Coloma Costas
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain
| | - Marta Hernández-García
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal-Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- Centro de Investigación Biológica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Rafael Cantón
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal-Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- Centro de Investigación Biológica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Jerónimo Rodríguez-Beltrán
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal-Instituto Ramón y Cajal de Investigación Sanitaria, Madrid, Spain
- Centro de Investigación Biológica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, Madrid, Spain
| | - Alfonso Santos-Lopez
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
- Centro de Investigación Biológica en Red de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain.
| | - Alvaro San Millan
- Centro Nacional de Biotecnología (CNB-CSIC), Madrid, Spain.
- Centro de Investigación Biológica en Red de Epidemiología y Salud Pública (CIBERESP), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
2
|
Liu W, Wang Z, Huang Y, Liu Y, Li R, Wang M, Zhang H, Meng C, Xiao X. Acetylshikonin reduces the spread of antibiotic resistance via plasmid conjugation. Int J Antimicrob Agents 2024:107370. [PMID: 39481662 DOI: 10.1016/j.ijantimicag.2024.107370] [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: 04/22/2024] [Revised: 09/27/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
The plasmid-mediated conjugative transfer of antibiotic resistance genes (ARGs) stands out as the primary driver behind the dissemination of antimicrobial resistance (AMR). Developing effective inhibitors that target conjugative transfer represents an efficient strategy for addressing the issue of AMR. Here, we studied the effect of acetylshikonin (ASK), a botanical derivative, on plasmid conjugation. The conjugative transfer of RP4-7 plasmid inter and intra species was notably reduced by ASK. The conjugation process of IncI2 and IncX4 plasmids harboring the mobile colistin resistance gene (mcr-1), IncX4 and IncX3 plasmids containing the carbapenem resistance gene (blaNDM-5), and IncFI and IncFII plasmids possessing the tetracycline resistance gene [tet(X4)] were also reduced by ASK. Importantly, the conjugative transfer frequency of mcr-1 positive IncI2 plasmid in mouse peritoneal conjugation model and gut conjugation model was reduced by ASK. The mechanism investigation showed that ASK disrupt the functionality of the bacterial cell membrane. Furthermore, the proton motive force (PMF) was dissipated. In addition, ASK blocked the electron transmission in bacteria's electron transport chain (ETC) through disturbing the quinone interaction, resulting in an insufficient energy supply for conjugation. Collectively, ASK is a potential conjugative transfer inhibitor, providing novel strategies to prevent the spread of AMR.
Collapse
Affiliation(s)
- Wei Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009; Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Yanhu Huang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yuan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Ruichao Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| | - Mianzhi Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China
| | - Haijie Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Chuang Meng
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009
| | - Xia Xiao
- College of Veterinary Medicine, Yangzhou University, Yangzhou, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China; Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009.
| |
Collapse
|
3
|
Muhi S, Marshall JL, O'Brien DP, Johnson PD, Ross G, Ramakrishnan A, Mackay LK, Doerflinger M, McCarthy JS, Jamrozik E, Osowicki J, Stinear TP. A human model of Buruli ulcer: Provisional protocol for a Mycobacterium ulcerans controlled human infection study. Wellcome Open Res 2024; 9:488. [PMID: 39386965 PMCID: PMC11462124 DOI: 10.12688/wellcomeopenres.22719.1] [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] [Accepted: 10/18/2024] [Indexed: 10/12/2024] Open
Abstract
Critical knowledge gaps have impeded progress towards reducing the global burden of disease due to Mycobacterium ulcerans, the cause of the neglected tropical disease Buruli ulcer (BU). Development of a controlled human infection model of BU has been proposed as an experimental platform to explore host-pathogen interactions and evaluate tools for prevention, diagnosis, and treatment. We have previously introduced the use case for a new human model and identified M. ulcerans JKD8049 as a suitable challenge strain. Here, we present a provisional protocol for an initial study, for transparent peer review during the earliest stages of protocol development. Following simultaneous scientific peer review and community/stakeholder consultation of this provisional protocol, we aim to present a refined protocol for institutional review board (IRB) evaluation.
Collapse
Affiliation(s)
- Stephen Muhi
- Department of Microbiology and Immunology, Doherty Institute, University of Melbourne, Melbourne, Victoria, 3000, Australia
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of General Medicine, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Julia L. Marshall
- Department of Infectious Diseases, Doherty Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Daniel P. O'Brien
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Department of Infectious Diseases, Doherty Institute, University of Melbourne, Melbourne, Victoria, Australia
- Department of Infectious Diseases, Barwon Health, Geelong, Victoria, Australia
| | - Paul D.R. Johnson
- Department of Microbiology and Immunology, Doherty Institute, University of Melbourne, Melbourne, Victoria, 3000, Australia
- Austin Health, Heidelberg, Victoria, Australia
| | - Gayle Ross
- Department of Dermatology, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Anand Ramakrishnan
- Department of Plastic and Reconstructive Surgery, The Royal Melbourne Hospital, Parkville, Victoria, Australia
| | - Laura K. Mackay
- Department of Microbiology and Immunology, Doherty Institute, University of Melbourne, Melbourne, Victoria, 3000, Australia
| | - Marcel Doerflinger
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - James S. McCarthy
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, Australia
- Department of Infectious Diseases, Doherty Institute, University of Melbourne, Melbourne, Victoria, Australia
| | - Euzebiusz Jamrozik
- Department of General Medicine, The Royal Melbourne Hospital, Parkville, Victoria, Australia
- WHO Collaborating Centre for Bioethics, Monash University, Clayton, Victoria, Australia
| | - Joshua Osowicki
- Tropical Diseases Research Group, Murdoch Children’s Research Institute, The Royal Children’s Hospital, Parkville, Victoria, Australia
- Infectious Diseases Unit, Department of General Medicine, The Royal Children's Hospital, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
| | - Timothy P. Stinear
- Department of Microbiology and Immunology, Doherty Institute, University of Melbourne, Melbourne, Victoria, 3000, Australia
- WHO Collaborating Centre for Mycobacterium ulcerans, Doherty Institute, Melbourne, Victoria, Australia
| |
Collapse
|
4
|
Han M, Wang X, Su L, Pan S, Liu N, Li D, Liu L, Cui J, Zhao H, Yang F. Intestinal microbiome dysbiosis increases Mycobacteria pulmonary colonization in mice by regulating the Nos2-associated pathways. eLife 2024; 13:RP99282. [PMID: 39412514 PMCID: PMC11483126 DOI: 10.7554/elife.99282] [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] [Indexed: 10/19/2024] Open
Abstract
Increasing researches reveal gut microbiota was associated with the development of tuberculosis (TB). How to prevent or reduce Mycobacterium tuberculosis colonization in the lungs is a key measure to prevent TB. However, the data on gut microbiota preventing Mycobacterium colonization in the lungs were scarce. Here, we established the clindamycin-inducing intestinal microbiome dysbiosis and fecal microbial transplantation models in mice to identify gut microbiota's effect on Mycobacterium's colonization in the mouse lungs and explore its potential mechanisms. The results showed that clindamycin treatment altered the diversity and composition of the intestinal bacterial and fungal microbiome, weakened the trans-kingdom network interactions between bacteria and fungi, and induced gut microbiome dysbiosis in the mice. Gut microbiota dysbiosis increases intestinal permeability and enhances the susceptibility of Mycobacterium colonization in the lungs of mice. The potential mechanisms were gut microbiota dysbiosis altered the lung transcriptome and increased Nos2 expression through the 'gut-lung axis'. Nos2 high expression disrupts the intracellular antimicrobial and anti-inflammatory environment by increasing the concentration of nitric oxide, decreasing the levels of reactive oxygen species and Defb1 in the cells, and promoting Mycobacteria colonization in the lungs of mice. The present study raises a potential strategy for reducing the risks of Mycobacteria infections and transmission by regulating the gut microbiome balance.
Collapse
Affiliation(s)
- MeiQing Han
- Department of Tuberculosis, The First Affiliated Hospital of Xinxiang Medical UniversityWeihuiChina
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Xia Wang
- Department of Tuberculosis, The First Affiliated Hospital of Xinxiang Medical UniversityWeihuiChina
| | - Lin Su
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Shiqi Pan
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Ningning Liu
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Duan Li
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Liang Liu
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - JunWei Cui
- Department of Tuberculosis, The First Affiliated Hospital of Xinxiang Medical UniversityWeihuiChina
| | - Huajie Zhao
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| | - Fan Yang
- Department of Tuberculosis, The First Affiliated Hospital of Xinxiang Medical UniversityWeihuiChina
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical UniversityXinxiangChina
| |
Collapse
|
5
|
He H, Fang C, Liu L, Li M, Liu W. Environmental Driving of Adaptation Mechanism on Rumen Microorganisms of Sheep Based on Metagenomics and Metabolomics Data Analysis. Int J Mol Sci 2024; 25:10957. [PMID: 39456741 PMCID: PMC11508146 DOI: 10.3390/ijms252010957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/04/2024] [Accepted: 10/05/2024] [Indexed: 10/28/2024] Open
Abstract
Natural or artificial selection causes animals to adapt to their environment. The adaptive changes generated by the rumen population and metabolism form the basis of ruminant evolution. In particular, the adaptive drive for environmental adaptation reflects the high-quality traits of sheep that have migrated from other places or have been distant from their origins for a long time. The Hu sheep is the most representative sheep breed in the humid and low-altitude environments (Tai Lake region) in East Asia and has been widely introduced into the arid and high-altitude environments (Tibetan Plateau and Hotan region), resulting in environmental adaptive changes in the Hu sheep. In this study, a joint analysis of the rumen microbial metagenome and metabolome was conducted on Hu sheep from different regions (area of origin and area of introduction) with the objective of investigating the quality traits of Hu sheep and identifying microorganisms that influence the adaptive drive of ruminants. The results demonstrated that the growth performance of Hu sheep was altered due to changes in rumen tissue and metabolism following their introduction to the arid area at relatively high altitude. Metagenomic and metabolomic analyses (five ramsper area) revealed that 3580 different microorganisms and 732 different metabolites were identified in the rumen fluid of arid sheep. Among these, the representative upregulated metabolites were 4,6-isocanedione, methanesulfonic acid and N2-succinyl-L-arginine, while the dominant microorganism was Prevotella ruminicola. The downregulated metabolites were identified as campesterol, teprenone and dihydroclavaminic acid, while the disadvantaged microorganisms were Dialister_succinatiphilus, Prevotella_sp._AGR2160, Prevotella_multisaccharivorax and Selenomonas_bovis. The results of the Pearson analysis indicated that the rumen microbiota and metabolite content of sheep were significantly altered and highly correlated following their relocation from a humid lowland to an arid upland. In particular, the observed changes in rumen microorganisms led to an acceleration of body metabolism, rendering sheep highly adaptable to environmental stress. Prevotella_ruminicola was identified as playing an important role in this process. These findings provide insights into the environmental adaptation mechanisms of sheep.
Collapse
Affiliation(s)
- Haiying He
- Department of Animal Science and Biotechnology, Xinjiang Agricultural University, Urumqi 830052, China; (H.H.); (L.L.); (M.L.)
| | - Chao Fang
- Faculte de Medecine Veterinaire, Universite de Liege, Quartier Vallee 2, Avenue de Cureghem 6 (B43), 4000 Liege, Belgium;
| | - Lingling Liu
- Department of Animal Science and Biotechnology, Xinjiang Agricultural University, Urumqi 830052, China; (H.H.); (L.L.); (M.L.)
| | - Mingming Li
- Department of Animal Science and Biotechnology, Xinjiang Agricultural University, Urumqi 830052, China; (H.H.); (L.L.); (M.L.)
| | - Wujun Liu
- Department of Animal Science and Biotechnology, Xinjiang Agricultural University, Urumqi 830052, China; (H.H.); (L.L.); (M.L.)
| |
Collapse
|
6
|
Zhao M, Cao L, Bai Q, Lu Y, Li B, Wu W, Ye J, Chen X, Wang Z, Liu B, Mao D. Light-Activated Nanocatalyst for Precise In-Situ Antimicrobial Synthesis via Photoredox-Catalytic Click Reaction. Angew Chem Int Ed Engl 2024; 63:e202408918. [PMID: 39013139 DOI: 10.1002/anie.202408918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/14/2024] [Accepted: 07/14/2024] [Indexed: 07/18/2024]
Abstract
The excessive and prolonged use of antibiotics contributes to the emergence of drug-resistant S. aureus strains and potential dysbacteriosis-related diseases, necessitating the exploration of alternative therapeutic approaches. Herein, we present a light-activated nanocatalyst for synthesizing in situ antimicrobials through photoredox-catalytic click reaction, achieving precise, site-directed elimination of S. aureus skin infections. Methylene blue (MB), a commercially available photosensitizer, was encapsulated within the CuII-based metal-organic framework, MOF-199, and further enveloped with Pluronic F-127 to create the light-responsive nanocatalyst MB@PMOF. Upon exposure to red light, MB participates in a photoredox-catalytic cycle, driven by the 1,3,5-benzenetricarboxylic carboxylate salts (BTC-) ligand presented in the structure of MOF-199. This light-activated MB then catalyzes the reduction of CuII to CuI through a single-electron transfer (SET) process, efficiently initiating the click reaction to form active antimicrobial agents under physiological conditions. Both in vitro and in vivo results demonstrated the effectiveness of MB@PMOF-catalyzed drug synthesis in inhibiting S. aureus, including their methicillin-resistant strains, thereby accelerating skin healing in severe bacterial infections. This study introduces a novel design paradigm for controlled, on-site drug synthesis, offering a promising alternative to realize precise treatment of bacterial infections without undesirable side effects.
Collapse
Affiliation(s)
- Minyang Zhao
- Department of Laboratory Medicine, Institute of Precision Medicine The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, 1510080, Guangzhou, China
| | - Lei Cao
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore, Singapore
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, 350207, Fuzhou, China
| | - Qingqing Bai
- Department of Laboratory Medicine, Institute of Precision Medicine The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, 1510080, Guangzhou, China
| | - Yaru Lu
- Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, 300072, Tianjin, China
| | - Bowen Li
- Institute of Transplant Medicine School of Medicine, Nankai University, 300071, Tianjin, China
| | - Wenbo Wu
- Department of Chemistry, Institute of Molecular Aggregation Science, Tianjin University, 300072, Tianjin, China
| | - Jinzhou Ye
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, 518132, Shenzhen, Guangdong, China
| | - Xinhan Chen
- Institute of Infectious Diseases, Shenzhen Bay Laboratory, 518132, Shenzhen, Guangdong, China
| | - Zhihong Wang
- Institute of Transplant Medicine School of Medicine, Nankai University, 300071, Tianjin, China
| | - Bin Liu
- Department of Chemical and Biomolecular Engineering, National University of Singapore, 117585, Singapore, Singapore
- Joint School of National University of Singapore and Tianjin University, International Campus of Tianjin University, 350207, Fuzhou, China
| | - Duo Mao
- Department of Laboratory Medicine, Institute of Precision Medicine The First Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, 1510080, Guangzhou, China
| |
Collapse
|
7
|
Radu AF, Bungau SG, Corb Aron RA, Tarce AG, Bodog R, Bodog TM, Radu A. Deciphering the Intricate Interplay in the Framework of Antibiotic-Drug Interactions: A Narrative Review. Antibiotics (Basel) 2024; 13:938. [PMID: 39452205 PMCID: PMC11505481 DOI: 10.3390/antibiotics13100938] [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: 08/16/2024] [Revised: 09/20/2024] [Accepted: 10/04/2024] [Indexed: 10/26/2024] Open
Abstract
Drug interactions are a significant and integral part of the concept of medication-related adverse events, whether referring to potential interactions or those currently observed in real-world conditions. The high global consumption of antibiotics and their pharmacokinetic and pharmacodynamic mechanisms make antibiotic-drug interactions a key element that requires continuous study due to their clinical relevance. In the present work, the current state of knowledge on antibiotic-drug interactions, which are less studied than other drug-drug interactions despite their frequent use in acute settings, has been consolidated and updated. The focus was on the interactions of the commonly used antibiotics in clinical practice, on the characteristics of the geriatric population susceptible to interactions, and on the impact of online drug interaction checkers. Additionally, strategies for optimizing the management of these interactions, including spacing out administrations, monitoring, or avoiding certain combinations, are suggested. Sustained research and careful monitoring are critical for improving antibiotic safety and efficacy, especially in susceptible populations, to enhance precision in managing antibiotic-drug interactions.
Collapse
Affiliation(s)
- Andrei-Flavius Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (A.-F.R.); (R.B.); (T.M.B.)
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Simona Gabriela Bungau
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (A.-F.R.); (R.B.); (T.M.B.)
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| | - Raluca Anca Corb Aron
- Department of Preclinical Disciplines, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Alexandra Georgiana Tarce
- Medicine Program of Study, Faculty of Medicine and Pharmacy, University of Oradea, 410073 Oradea, Romania;
| | - Ruxandra Bodog
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (A.-F.R.); (R.B.); (T.M.B.)
| | - Teodora Maria Bodog
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (A.-F.R.); (R.B.); (T.M.B.)
| | - Ada Radu
- Doctoral School of Biological and Biomedical Sciences, University of Oradea, 410087 Oradea, Romania; (A.-F.R.); (R.B.); (T.M.B.)
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, 410028 Oradea, Romania
| |
Collapse
|
8
|
Guo C, An Q, Zhang L, Wei X, Xu J, Yu J, Wu G, Ma J. Intratumoral microbiota as cancer therapeutic target. Aging Med (Milton) 2024; 7:636-644. [PMID: 39507228 PMCID: PMC11535161 DOI: 10.1002/agm2.12359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024] Open
Abstract
Intratumoral microbiota, which affects the physiological and pathological processes of the host, has attracted increasing attention from researchers. Microbials have been found in normal as well as tumor tissues that were originally thought to be sterile. Intratumoral microbiota is considered to play a significant role in the development of tumors and the reduction of clinical benefits. In addition, intratumoral microbiota are heterogeneous, which have different distribution in various types of tumors, and can influence tumor development through different mechanisms, including genome mutations, inflammatory responses, activated cancer pathways, and immunosuppressive microenvironments. Therefore, eliminating the intratumoral microbiota is considered one of the most promising ways to slow down the tumor progression and improve therapeutic outcomes. In this review, we systematically categorized the intratumoral microbiota and elucidated its role in the pathogenesis and therapeutic response of cancer. We have also described the novel strategies to mitigate the impact of tumor progression. We hope this review will provide new insights for the anti-tumor treatment, particularly for the elderly population, where such insights could significantly enhance treatment outcomes.
Collapse
Affiliation(s)
- Chang Guo
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
- Medical SchoolUniversity of Chinese Academy of SciencesBeijingPeople's Republic of China
| | - Qi An
- General Surgery Department, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Lu‐yao Zhang
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Xun‐dong Wei
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Jing Xu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Jiang‐yong Yu
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Guo‐ju Wu
- General Surgery Department, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| | - Jie Ma
- Center of Biotherapy, Beijing Hospital, National Center of Gerontology; Institute of Geriatric MedicineChinese Academy of Medical SciencesBeijingPeople's Republic of China
| |
Collapse
|
9
|
Zhong B, Liang W, Zhao Y, Li F, Zhao Z, Gao Y, Yang G, Li S. Combination of Lactiplantibacillus Plantarum ELF051 and Astragalus Polysaccharides Improves Intestinal Barrier Function and Gut Microbiota Profiles in Mice with Antibiotic-Associated Diarrhea. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10368-3. [PMID: 39354215 DOI: 10.1007/s12602-024-10368-3] [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] [Accepted: 09/17/2024] [Indexed: 10/03/2024]
Abstract
The purpose of this study was to investigate the improvement of the intestinal barrier and gut microbiota in mice with antibiotic-associated diarrhea (AAD) using Lactiplantibacillus plantarum ELF051 combined with Astragalus polysaccharides. The amoxicillin, clindamycin, and streptomycin triple-mixed antibiotic-induced AAD models were administered with L. plantarum ELF051 or Astragalus polysaccharides or L. plantarum ELF051 + Astragalus polysaccharides for 14 days. Our findings revealed that the combination of L. plantarum ELF051 and Astragalus polysaccharides elevated the number of goblet cells and enhanced the proportion of mucous within the colon tissue. Furthermore, the expression of sIgA and IgG were upregulated, while the levels of IL-17A, IL-4, DAO, D-LA, LPS, and TGF-β1 were downregulated. L. plantarum ELF051 combined with Astragalus polysaccharides elevated the expression of tight junction (TJ) proteins, facilitating intestinal mucosal repair via Smad signaling nodes. Furthermore, their combination effectively increased the relative abundance of lactic acid bacteria (LAB) and Allobaculum, and decreased the relative abundance of Bacteroides and Blautia. Spearman rank correlation analysis demonstrated that LAB were closely related to permeability factors, immune factors, and indicators of intestinal barrier function. In summary, the effect of combining L. plantarum ELF051 and Astragalus polysaccharides on AAD mice was achieved by enhancing intestinal barrier function and regulating the composition of the gut microbiota.
Collapse
Affiliation(s)
- Bao Zhong
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
- College of Food Science and Nutritional Engineering, Jilin Agriculture Science and Technology University, Jilin, 132101, P.R. China
- Brewing Technology Innovation Center of Jilin Province, Jilin Agriculture Science and Technology University, Jilin, 132101, P.R. China
| | - Wei Liang
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
- Anshan Hospital of Traditional Chinese Medicine, Anshan, 114004, P.R. China
| | - Yujuan Zhao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
| | - Fenglin Li
- College of Food Science and Nutritional Engineering, Jilin Agriculture Science and Technology University, Jilin, 132101, P.R. China
- Brewing Technology Innovation Center of Jilin Province, Jilin Agriculture Science and Technology University, Jilin, 132101, P.R. China
| | - Zijian Zhao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
| | - Yansong Gao
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
| | - Ge Yang
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China
| | - Shengyu Li
- Institute of Agro-Food Technology, Jilin Academy of Agricultural Sciences (Northeast Agricultural Research Center of China), Changchun, 130033, P.R. China.
| |
Collapse
|
10
|
Kouraki A, Zheng AS, Miller S, Kelly A, Ashraf W, Bazzani D, Bonadiman A, Tonidandel G, Bolzan M, Vijay A, Nightingale J, Menni C, Ollivere BJ, Valdes AM. Metagenomic changes in response to antibiotic treatment in severe orthopedic trauma patients. iScience 2024; 27:110783. [PMID: 39286492 PMCID: PMC11403444 DOI: 10.1016/j.isci.2024.110783] [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: 05/13/2024] [Revised: 06/21/2024] [Accepted: 08/19/2024] [Indexed: 09/19/2024] Open
Abstract
We investigated changes in microbiome composition and abundance of antimicrobial resistance (AMR) genes post-antibiotic treatment in severe trauma patients. Shotgun sequencing revealed beta diversity (Bray-Curtis) differences between 16 hospitalized multiple rib fractures patients and 10 age- and sex-matched controls (p = 0.043), and between antibiotic-treated and untreated patients (p = 0.015). Antibiotic-treated patients had lower alpha diversity (Shannon) at discharge (p = 0.003) and 12-week post-discharge (p = 0.007). At 12 weeks, they also exhibited a 5.50-fold (95% confidence interval [CI]: 2.86-8.15) increase in Escherichia coli (p = 0.0004) compared to controls. Differential analysis identified nine AMRs that increased in antibiotic-treated compared to untreated patients between hospital discharge and 6 and 12 weeks follow-up (false discovery rate [FDR] < 0.20). Two aminoglycoside genes and a beta-lactamase gene were directly related to antibiotics administered, while five were unrelated. In trauma patients, lower alpha diversity, higher abundance of pathobionts, and increases in AMRs persisted for 12 weeks post-discharge, suggesting prolonged microbiome disruption. Probiotic or symbiotic therapies may offer future treatment avenues.
Collapse
Affiliation(s)
- Afroditi Kouraki
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Amy S Zheng
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Suzanne Miller
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Anthony Kelly
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Waheed Ashraf
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | | | | | | | | | - Amrita Vijay
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Jessica Nightingale
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Cristina Menni
- Department of Twin Research, King's College London, London SE1 7EH, UK
| | - Benjamin J Ollivere
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| | - Ana M Valdes
- Academic Unit of Injury, Recovery and Inflammation Sciences, School of Medicine, University of Nottingham, Nottingham NG7 2UH, UK
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and the University of Nottingham, Nottingham NG7 2UH, UK
| |
Collapse
|
11
|
Sawaed J, Zelik L, Levin Y, Feeney R, Naama M, Gordon A, Zigdon M, Rubin E, Telpaz S, Modilevsky S, Ben-Simon S, Awad A, Harshuk-Shabso S, Nuriel-Ohayon M, Werbner M, Schroeder BO, Erez A, Bel S. Antibiotics damage the colonic mucus barrier in a microbiota-independent manner. SCIENCE ADVANCES 2024; 10:eadp4119. [PMID: 39259805 PMCID: PMC11389797 DOI: 10.1126/sciadv.adp4119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
Antibiotic use is a risk factor for development of inflammatory bowel diseases (IBDs). IBDs are characterized by a damaged mucus layer, which does not separate the intestinal epithelium from the microbiota. Here, we hypothesized that antibiotics affect the integrity of the mucus barrier, which allows bacterial penetrance and predisposes to intestinal inflammation. We found that antibiotic treatment led to breakdown of the colonic mucus barrier and penetration of bacteria into the mucus layer. Using fecal microbiota transplant, RNA sequencing followed by machine learning, ex vivo mucus secretion measurements, and antibiotic treatment of germ-free mice, we determined that antibiotics induce endoplasmic reticulum stress in the colon that inhibits colonic mucus secretion in a microbiota-independent manner. This antibiotic-induced mucus secretion flaw led to penetration of bacteria into the colonic mucus layer, translocation of microbial antigens into circulation, and exacerbation of ulcerations in a mouse model of IBD. Thus, antibiotic use might predispose to intestinal inflammation by impeding mucus production.
Collapse
Affiliation(s)
- Jasmin Sawaed
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Lilach Zelik
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Yehonatan Levin
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
| | - Rachel Feeney
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå, Sweden
- Umeå Center for Microbial Research (UCMR), Umeå, Sweden
| | - Maria Naama
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Ateret Gordon
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Mor Zigdon
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Elad Rubin
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Shahar Telpaz
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | | | - Shira Ben-Simon
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Aya Awad
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | | | | | - Michal Werbner
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Bjoern O Schroeder
- Department of Molecular Biology, Umeå University, Umeå, Sweden
- Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå, Sweden
- Umeå Center for Microbial Research (UCMR), Umeå, Sweden
| | - Amir Erez
- Racah Institute of Physics, The Hebrew University, Jerusalem, Israel
| | - Shai Bel
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
12
|
Zhao C, Chen G, Huang Y, Zhang Y, Li S, Jiang Z, Peng H, Wang J, Li D, Hou R, Peng C, Wan X, Cai H. Alleviation of fluoride-induced colitis by tea polysaccharides: Insights into the role of Limosilactobacillus vaginalis and butyric acid. JOURNAL OF HAZARDOUS MATERIALS 2024; 476:134858. [PMID: 38905983 DOI: 10.1016/j.jhazmat.2024.134858] [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: 03/13/2024] [Revised: 05/23/2024] [Accepted: 06/06/2024] [Indexed: 06/23/2024]
Abstract
Endemic fluorosis has gained increasing attention as a public health concern, and the escalating risk of colitis resulting from excessive fluoride intake calls for effective mitigation strategies. This study aimed to investigate the potential mechanisms underlying the alleviation of fluoride-induced colitis by Tea polysaccharides (TPS). Under conditions of excessive fluoride intake, significant changes were observed in the gut microbiota of rats, leading to aggravated colitis. However, the intervention of TPS exerted a notable alleviating effect on colitis symptoms. Antibiotic intervention and fecal microbiota transplantation (FMT) experiments provided evidence that TPS-mediated relief of fluoride-induced colitis is mediated through its effects on the gut microbiota. Furthermore, TPS supplementation was found to modulate the structure of gut microbiota, enhance the relative abundance of Limosilactobacillus vaginalis in the gut microbiota, and promote the expression of short-chain fatty acid (SCFAs) receptors in colonic tissue. Notably, L. vaginalis played a significant role in alleviating fluoride-induced colitis and facilitating the absorption of butyric acid in the rat colon. Subsequent butyric acid intervention experiments confirmed its remarkable alleviating effect on fluoride-induced colitis. Overall, these findings provide a potential preventive strategy for fluoride-induced colitis by TPS intervention, which is mediated by L. vaginalis and butyric acid.
Collapse
Affiliation(s)
- Chenjun Zhao
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Guijie Chen
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Ying Huang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Yuxuan Zhang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Sichen Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Zhiliang Jiang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Huihui Peng
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Juan Wang
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Daxiang Li
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Ruyan Hou
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Chuanyi Peng
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China
| | - Xiaochun Wan
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China.
| | - Huimei Cai
- State Key Laboratory of Tea Plant Biology and Utilization, School of Tea & Food Science and Technology, Anhui Agricultural University, Hefei 230036, Anhui, PR China; Joint Research Center for Food Nutrition and Health of IHM, Anhui Agricultural University, Hefei 230036, Anhui, PR China.
| |
Collapse
|
13
|
Shah H, Trivedi M, Gurjar T, Sahoo DK, Jergens AE, Yadav VK, Patel A, Pandya P. Decoding the Gut Microbiome in Companion Animals: Impacts and Innovations. Microorganisms 2024; 12:1831. [PMID: 39338505 PMCID: PMC11433972 DOI: 10.3390/microorganisms12091831] [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: 07/15/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/30/2024] Open
Abstract
The changing notion of "companion animals" and their increasing global status as family members underscores the dynamic interaction between gut microbiota and host health. This review provides a comprehensive understanding of the intricate microbial ecology within companion animals required to maintain overall health and prevent disease. Exploration of specific diseases and syndromes linked to gut microbiome alterations (dysbiosis), such as inflammatory bowel disease, obesity, and neurological conditions like epilepsy, are highlighted. In addition, this review provides an analysis of the various factors that impact the abundance of the gut microbiome like age, breed, habitual diet, and microbe-targeted interventions, such as probiotics. Detection methods including PCR-based algorithms, fluorescence in situ hybridisation, and 16S rRNA gene sequencing are reviewed, along with their limitations and the need for future advancements. Prospects for longitudinal investigations, functional dynamics exploration, and accurate identification of microbial signatures associated with specific health problems offer promising directions for future research. In summary, it is an attempt to provide a deeper insight into the orchestration of multiple microbial species shaping the health of companion animals and possible species-specific differences.
Collapse
Affiliation(s)
- Harsh Shah
- TREE Lab, Department of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara 391410, India (M.T.)
| | - Mithil Trivedi
- TREE Lab, Department of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara 391410, India (M.T.)
| | - Tejas Gurjar
- TREE Lab, Department of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara 391410, India (M.T.)
| | - Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Albert E. Jergens
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Virendra Kumar Yadav
- Marwadi University Research Center, Department of Microbiology, Faculty of Sciences, Marwadi University, Rajkot 360003, India;
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University, Patan 384265, India;
| | - Parth Pandya
- TREE Lab, Department of Biomedical and Life Sciences, School of Science, Navrachana University, Vadodara 391410, India (M.T.)
| |
Collapse
|
14
|
Wang M, Chen Y, Song AX, Weng X, Meng Y, Lin J, Mao YH. The Combination of Exercise and Konjac Glucomannan More Effectively Prevents Antibiotics-Induced Dysbiosis in Mice Compared with Singular Intervention. Nutrients 2024; 16:2942. [PMID: 39275258 PMCID: PMC11397520 DOI: 10.3390/nu16172942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/16/2024] Open
Abstract
Our previous studies have demonstrated that konjac glucomannan (KGM) can prevent dysbiosis induced by antibiotics. While exercise may also impact the gut microbiome, there are limited studies reporting its protective effect on antibiotic-induced dysbiosis. Therefore, this study investigated the preventive and regulatory effects of a combination of 6-week exercise and KGM intervention on antibiotic-induced dysbiosis in C57BL/6J mice compared with a single intervention. The results showed that combined exercise and KGM intervention could restore the changes in the relative abundance of Bacteroides (3.73% with CTL versus 14.23% with ATBX versus 4.46% with EK) and Prevotellaceae_Prevotella (0.33% with CTL versus 0.00% with ATBX versus 0.30% with EK) induced by antibiotics (p < 0.05), and minimized the Bray-Curtis distance induced by antibiotics (0.55 with CTL versus 0.81 with ATBX versus 0.80 with EXC versus 0.83 with KGM versus 0.75 with EK). Compared with the combined intervention, exercise intervention also produced a certain level of recovery effects; the relative abundance of Rikenellaceae (1.96% with CTL versus 0.09% with ATBX versus 0.49% with EXC) was restored, while KGM supplementation showed the best preventive effect. In addition, the combination of exercise and KGM significantly enriched microbial purine metabolic pathways (p < 0.05). These findings indicate that combining exercise with KGM could be a promising approach to reducing the side effects of antibiotics on the gut microbiome.
Collapse
Affiliation(s)
- Minghan Wang
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Yonglin Chen
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Ang-Xin Song
- Key Laboratory of Plant Resource Conservation and Germplasm Innovation in Mountainous Region (Ministry of Education), School of Liquor and Food Engineering, Guizhou University, Guiyang 550025, China
| | - Xiquan Weng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Yan Meng
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Jieru Lin
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
| | - Yu-Heng Mao
- School of Exercise and Health, Guangzhou Sport University, Guangzhou 510500, China
- Guangdong Key Laboratory of Human Sports Performance Science, Guangzhou Sport University, Guangzhou 510500, China
| |
Collapse
|
15
|
Brdová D, Ruml T, Viktorová J. Mechanism of staphylococcal resistance to clinically relevant antibiotics. Drug Resist Updat 2024; 77:101147. [PMID: 39236354 DOI: 10.1016/j.drup.2024.101147] [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: 04/16/2024] [Revised: 08/17/2024] [Accepted: 08/26/2024] [Indexed: 09/07/2024]
Abstract
Staphylococcus aureus, a notorious pathogen with versatile virulence, poses a significant challenge to current antibiotic treatments due to its ability to develop resistance mechanisms against a variety of clinically relevant antibiotics. In this comprehensive review, we carefully dissect the resistance mechanisms employed by S. aureus against various antibiotics commonly used in clinical settings. The article navigates through intricate molecular pathways, elucidating the mechanisms by which S. aureus evades the therapeutic efficacy of antibiotics, such as β-lactams, vancomycin, daptomycin, linezolid, etc. Each antibiotic is scrutinised for its mechanism of action, impact on bacterial physiology, and the corresponding resistance strategies adopted by S. aureus. By synthesising the knowledge surrounding these resistance mechanisms, this review aims to serve as a comprehensive resource that provides a foundation for the development of innovative therapeutic strategies and alternative treatments for S. aureus infections. Understanding the evolving landscape of antibiotic resistance is imperative for devising effective countermeasures in the battle against this formidable pathogen.
Collapse
Affiliation(s)
- Daniela Brdová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| | - Jitka Viktorová
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Technicka 3, Prague 16628, Czech Republic.
| |
Collapse
|
16
|
Nilson R, Penumutchu S, Pagano FS, Belenky P. Metabolic changes associated with polysaccharide utilization reduce susceptibility to some β-lactams in Bacteroides thetaiotaomicron. mSphere 2024; 9:e0010324. [PMID: 39109911 PMCID: PMC11351048 DOI: 10.1128/msphere.00103-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 07/07/2024] [Indexed: 08/29/2024] Open
Abstract
Antibiotic therapy alters bacterial abundance and metabolism in the gut microbiome, leading to dysbiosis and opportunistic infections. Bacteroides thetaiotaomicron (Bth) is both a commensal in the gut and an opportunistic pathogen in other body sites. Past work has shown that Bth responds to β-lactam treatment differently depending on the metabolic environment both in vitro and in vivo. Studies of other bacteria show that an increase in respiratory metabolism independent of growth rate promotes susceptibility to bactericidal antibiotics. We propose that Bth enters a protected state linked to an increase in polysaccharide utilization and a decrease in the use of simple sugars. Here, we apply antibiotic susceptibility testing, transcriptomic analysis, and genetic manipulation to characterize this polysaccharide-mediated tolerance (PM tolerance) phenotype. We found that a variety of mono- and disaccharides increased the susceptibility of Bth to several different β-lactams compared to polysaccharides. Transcriptomics indicated a metabolic shift from reductive to oxidative branches of the tricarboxylic acid cycle on polysaccharides. Accordingly, supplementation with intermediates of central carbon metabolism had varying effects on PM tolerance. Transcriptional analysis also showed a decrease in the expression of the electron transport chain (ETC) protein NQR and an increase in the ETC protein NUO, when given fiber versus glucose. Deletion of NQR increased Bth susceptibility while deletion of NUO and a third ETC protein NDH2 had no effect. This work confirms that carbon source utilization modulates antibiotic susceptibility in Bth and that anaerobic respiratory metabolism and the ETC play an essential role.IMPORTANCEAntibiotics are indispensable medications that revolutionized modern medicine. However, their effectiveness is challenged by a large array of resistance and tolerance mechanisms. Treatment with antibiotics also disrupts the gut microbiome which can adversely affect health. Bacteroides are prevalent in the gut microbiome and yet are frequently involved in anaerobic infections. Thus, understanding how antibiotics affect these bacteria is necessary to implement proper treatment. Recent work has investigated the role of metabolism in antibiotic susceptibility in distantly related bacteria such as Escherichia coli. Using antibiotic susceptibility testing, transcriptomics, and genetic manipulation, we demonstrate that polysaccharides reduce β-lactam susceptibility when compared to monosaccharides. This finding underscores the profound impact of metabolic adaptation on the therapeutic efficacy of antibiotics. In the long term, this work indicates that modulation of metabolism could make Bacteroides more susceptible during infections or protect them in the context of the microbiome.
Collapse
Affiliation(s)
- Rachael Nilson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Swathi Penumutchu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Francesco S. Pagano
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
17
|
Beekman CN, Penumutchu S, Peterson R, Han G, Belenky M, Hasan MH, Belenky A, Beura LK, Belenky P. Spatial analysis of murine microbiota and bile acid metabolism during amoxicillin treatment. Cell Rep 2024; 43:114572. [PMID: 39116202 DOI: 10.1016/j.celrep.2024.114572] [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: 01/30/2024] [Revised: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
Antibiotics cause collateral damage to resident microbes that is associated with various health risks. To date, studies have largely focused on the impacts of antibiotics on large intestinal and fecal microbiota. Here, we employ a gastrointestinal (GI) tract-wide integrated multiomic approach to show that amoxicillin (AMX) treatment reduces bacterial abundance, bile salt hydrolase activity, and unconjugated bile acids in the small intestine (SI). Losses of fatty acids (FAs) and increases in acylcarnitines in the large intestine (LI) correspond with spatially distinct expansions of Proteobacteria. Parasutterella excrementihominis engage in FA biosynthesis in the SI, while multiple Klebsiella species employ FA oxidation during expansion in the LI. We subsequently demonstrate that restoration of unconjugated bile acids can mitigate losses of commensals in the LI while also inhibiting the expansion of Proteobacteria during AMX treatment. These results suggest that the depletion of bile acids and lipids may contribute to AMX-induced dysbiosis in the lower GI tract.
Collapse
Affiliation(s)
- Chapman N Beekman
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA.
| | - Swathi Penumutchu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Rachel Peterson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Geongoo Han
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Marina Belenky
- Felicitex Therapeutics Inc., 27 Strathmore Road, Natick, MA 01760, USA
| | - Mohammad H Hasan
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Alexei Belenky
- Felicitex Therapeutics Inc., 27 Strathmore Road, Natick, MA 01760, USA
| | - Lalit K Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
18
|
Akhmedov M, Espinoza JL. Addressing the surge of infections by multidrug-resistant Enterobacterales in hematopoietic cell transplantation. Blood Rev 2024:101229. [PMID: 39217051 DOI: 10.1016/j.blre.2024.101229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 08/17/2024] [Accepted: 08/19/2024] [Indexed: 09/04/2024]
Abstract
Patients undergoing hematopoietic cell transplantation (HCT) have an increased risk of developing severe infections. In recent years, bloodstream infections caused by Gram-negative bacteria have been increasingly reported among HCT recipients, and many of these infections are caused by bacterial strains of the Enterobacterales order. Among these pathogens, particularly concerning are the multidrug-resistant Enterobacterales (MDRE), such as Extended Spectrum β-lactamase-producing Enterobacterales and Carbapenem-resistant Enterobacterales, since infections caused by these pathogens are difficult to treat due to the limited antimicrobial options and are associated with worse transplant outcomes. We summarized the evidence from studies published in PubMed and Scopus on the burden of MDRE infections in HCT recipients, and strategies for the management and prevention of these infections, including strict adherence to recommended infection control practices and multidisciplinary antimicrobial stewardship, the use of probiotics, and fecal microbiota transplantation, are also discussed.
Collapse
Affiliation(s)
- Mobil Akhmedov
- Department of High-dose Chemotherapy and Bone Marrow Transplantation, P. Hertsen Moscow Oncology Research Institute, Russia; Department of Oncology and Oncosurgery, Russian University of Medicine, Russia
| | | |
Collapse
|
19
|
Lee EB, Lee GY, Hossain MA, Awji EG, Park SC. Gut microbiome perturbation and its correlation with tylosin pharmacokinetics in healthy and infected pigs. Sci Rep 2024; 14:18670. [PMID: 39134586 PMCID: PMC11319344 DOI: 10.1038/s41598-024-69566-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/06/2024] [Indexed: 08/15/2024] Open
Abstract
Tylosin, an antibiotic with a long history in treating respiratory bacterial infections, has unknown effects on the gut microbiota of healthy and infected pigs. The study aimed to investigate the effect of a therapeutic dose of tylosin on swine gut microbiota and explored the relationship between this effect and tylosin pharmacokinetics (PK). We also assessed whether changes in gut microbiota after tylosin administration differ between healthy animals (n = 7) and animals intranasally co-infected (n = 7) with Actinobacillus pleuropneumoniae and Pasteurella multocida. Both groups were intramuscularly administered with tylosin (20 mg/kg). The 16S rRNA gene analyses revealed a significantly lower species richness and diversity, after tylosin treatment, in the infected than the healthy pigs, with infected pigs having lower levels of Bacteroidetes and Firmicutes and higher levels of Proteobacteria. Greater tylosin exposure (greater area under curve (AUC) and maximum plasma concentration (Cmax), and slower elimination (longer terminal half-life, T1/2) were observed in healthy than infected pigs. Relative abundance of Lactobacillus, Oscillibacter, Prevotella, and Sporobacter was positively and significantly correlated with AUC and Cmax, whereas the abundance of Acinetobacter, Alishewanella, and Pseudomonas was positively and significantly correlated with T1/2 and mean residence time (MRT) of tylosin. Our findings, for the first time, demonstrated significant changes in swine gut microbiota after a single therapeutic dose of tylosin was administered, whereas the effect of these changes on tylosin PK was not evident.
Collapse
Affiliation(s)
- Eon-Bee Lee
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, Institute for Veterinary Biomedical Science, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Ga-Yeong Lee
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, Institute for Veterinary Biomedical Science, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Md Akil Hossain
- Institute for Tuberculosis Research, College of Pharmacy, University of Illinois Chicago, 833 S Wood St, Chicago, IL, 60612, USA
| | - Elias Gebru Awji
- Independent Researcher, 263 Congressional Ln, Rockville, MD, 20852, USA
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, Institute for Veterinary Biomedical Science, College of Veterinary Medicine, Kyungpook National University, Daegu, 41566, Republic of Korea.
- Cardiovascular Research Institute, Kyungpook National University, Daegu, 41566, Republic of Korea.
| |
Collapse
|
20
|
Gu H, Tao E, Fan Y, Long G, Jia X, Yuan T, Chen L, Shu X, Zheng W, Jiang M. Effect of β-lactam antibiotics on the gut microbiota of term neonates. Ann Clin Microbiol Antimicrob 2024; 23:69. [PMID: 39113137 PMCID: PMC11308410 DOI: 10.1186/s12941-024-00730-2] [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: 04/27/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
β-Lactam antibiotics are a class of antibiotics commonly used to treat bacterial infections. However, the effects of β-lactam antibiotics on term neonatal intestinal flora have not been fully elucidated. Hospitalized full-term newborns receiving β-lactam antibiotics formed the antibiotic group (n = 67), while those without antibiotic treatment comprised the non-antibiotic group (n = 47). A healthy group included healthy full-term newborns (n = 16). Stool samples were collected for 16 S rDNA sequencing to analyze gut microbiota variations. Further investigation was carried out within the β-lactam antibiotic group, exploring the effects of antibiotic use on the newborns' gut microbiota in relation to the duration and type of antibiotic administration, delivery method, and feeding practices. The antibiotic group exhibited significant difference of microbial community composition compared to the other groups. Genera like Klebsiella, Enterococcus, Streptococcus, Alistipes, and Aeromonas were enriched, while Escherichia-Shigella, Clostridium sensu stricto 1, Bifidobacterium, and Parabacteroides were reduced. Klebsiella negatively correlated with Escherichia-Shigella, positively with Enterobacter, while Escherichia-Shigella negatively correlated with Enterococcus and Streptococcus. Regardless of neonatal age, β-lactam antibiotics induced an elevated abundance of Klebsiella and Enterococcus. The impact on gut microbiota varied with the duration and type of antibiotic (cefotaxime or ampicillin/sulbactam). Compared to vaginal delivery, cesarean delivery after β-lactam treatment heightened the abundance of Klebsiella, Enterobacteriaceae_Unclassified, Lactobacillales_Unclassified, and Pectobacterium. Feeding patterns minimally influenced β-lactam-induced alterations. In conclusion, β-lactam antibiotic treatment for neonatal pneumonia and sepsis markedly disrupted intestinal microbiota, favoring Klebsiella, Enterococcus, Streptococcus, Alistipes, and Aeromonas. The impact of β-lactam varied by duration, type, and delivery method, emphasizing heightened disruptions post-cesarean delivery.
Collapse
Affiliation(s)
- Hongdan Gu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
- Department of Pediatrics, Wenling Maternal and Child Health Care Hospital, Wenling, Zhejiang, 317500, China
| | - Enfu Tao
- Department of Neonatology and NICU, Wenling Maternal and Child Health Care Hospital, Wenling, Zhejiang, 317500, China
| | - Yijia Fan
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Gao Long
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Xinyi Jia
- Department of Gastroenterology and Pediatric Endoscopy Center, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Road, Hangzhou, Zhejiang, 310052, China
| | - Tianming Yuan
- Department of Neonatology, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310052, China
| | - Lihua Chen
- Department of Neonatology, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310052, China
| | - Xiaoli Shu
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China
| | - Wei Zheng
- Department of Gastroenterology and Pediatric Endoscopy Center, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Road, Hangzhou, Zhejiang, 310052, China
| | - Mizu Jiang
- Pediatric Endoscopy Center and Gastrointestinal Laboratory, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310052, China.
- Department of Gastroenterology and Pediatric Endoscopy Center, National Clinical Research Center for Child Health, National Children's Regional Medical Center, Children's Hospital, Zhejiang University School of Medicine, No. 3333, Binsheng Road, Hangzhou, Zhejiang, 310052, China.
| |
Collapse
|
21
|
Cao Q, Liu C, Chen L, Qin Y, Wang T, Wang C. Synergistic impacts of antibiotics and heavy metals on Hermetia illucens: Unveiling dynamics in larval gut bacterial communities and microbial metabolites. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2024; 365:121632. [PMID: 38950506 DOI: 10.1016/j.jenvman.2024.121632] [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: 03/14/2024] [Revised: 06/16/2024] [Accepted: 06/26/2024] [Indexed: 07/03/2024]
Abstract
Hermetia illucens larvae showcases remarkable bioremediation capabilities for both antibiotics and heavy metal contaminants. However, the distinctions in larval intestinal microbiota arising from the single and combined effects of antibiotics and heavy metals remain poorly elucidated. In this study, we delved into the details of larval intestinal bacterial communities and microbial metabolites when exposed to single and combined contaminants of oxytetracycline (OTC) and hexavalent chromium (Cr(VI)). After conversion, single contaminant-spiked substrate showed 75.5% of OTC degradation and 95.2% of Cr(VI) reductiuon, while combined contaminant-spiked substrate exhibited 71.3% of OTC degradation and 93.4% of Cr(VI) reductiuon. Single and combined effects led to differences in intestinal bacterial communities, mainly reflected in the genera of Enterococcus, Pseudogracilibacillus, Gracilibacillus, Wohlfahrtiimonas, Sporosarcina, Lysinibacillus, and Myroide. Moreover, these effects also induced differences across various categories of microbial metabolites, which categorized into amino acid and its metabolites, benzene and substituted derivatives, carbohydrates and its metabolites, heterocyclic compounds, hormones and hormone-related compounds, nucleotide and its metabolites, and organic acid and its derivatives. In particular, the differences induced OTC was greater than that of Cr(VI), and combined effects increased the complexity of microbial metabolism compared to that of single contaminant. Correlation analysis indicated that the bacterial genera, Preudogracilibacillus, Enterococcus, Sporosarcina, Lysinibacillus, Wohlfahrtiimonas, Ignatzschineria, and Fusobacterium exhibited significant correlation with significant differential metabolites, these might be used as indicators for the resistance and bioremediation of OTC and Cr(VI) contaminants. These findings are conducive to further understanding that the metabolism of intestinal microbiota determines the resistance of Hermetia illucens to antibiotics and heavy metals.
Collapse
Affiliation(s)
- Qingcheng Cao
- Key Laboratory of Green Chemical Process of Ministry of Education, Key Laboratory of Novel Reactor and Green Chemical Technology of Hubei Province, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, China
| | - Cuncheng Liu
- Key Laboratory of Green Chemical Process of Ministry of Education, Key Laboratory of Novel Reactor and Green Chemical Technology of Hubei Province, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, China; Research Center for Environmental Ecology and Engineering, School of Environmental Ecology and Biological Engineering, Wuhan Institute of Technology, Wuhan, 430205, China.
| | - Li Chen
- Key Laboratory of Green Chemical Process of Ministry of Education, Key Laboratory of Novel Reactor and Green Chemical Technology of Hubei Province, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, China
| | - Yuanhang Qin
- Key Laboratory of Green Chemical Process of Ministry of Education, Key Laboratory of Novel Reactor and Green Chemical Technology of Hubei Province, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, China
| | - Tielin Wang
- Key Laboratory of Green Chemical Process of Ministry of Education, Key Laboratory of Novel Reactor and Green Chemical Technology of Hubei Province, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, China
| | - Cunwen Wang
- Key Laboratory of Green Chemical Process of Ministry of Education, Key Laboratory of Novel Reactor and Green Chemical Technology of Hubei Province, School of Chemical Engineering and Pharmacy, Wuhan Institute of Technology, Wuhan, 430205, China.
| |
Collapse
|
22
|
Wu M, Fan Q, Li X, Wang P, Feng C, Wang L, Chang Z, Wu W, Chong R. Surface coordination induced a quasi p-n junction for efficient visible light driven degradation of tetracycline over hydroxyapatite. J Colloid Interface Sci 2024; 667:312-320. [PMID: 38640651 DOI: 10.1016/j.jcis.2024.04.077] [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: 01/31/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/21/2024]
Abstract
The removal of antibiotics from aquatic solutions remains a global environmental challenge. In this work, the photocatalytic removal of a typical antibiotic-tetracycline (TC) using hydroxyapatite (HAp) as a catalyst was investigated. It was impressive that TC could be efficiently degraded by HAp under visible light irradiation, even though both HAp and TC exhibited poor harvesting in visible light region. The experimental and theoretical explorations were undertaken to thoroughly investigate the underlying mechanism of visible light degradation of TC over HAp. The results indicated that the formed TC-HAp complexes via surface coordination played an important role as photosensitizers for the visible light response. Together with the formation of a quasi p-n junction via band alignment, the photogenerated electrons in the highest unoccupied molecular orbital (HOMO) of TC-HAp were excited to the lowest unoccupied molecular orbital (LUMO) and subsequently migrated to the conduction band of HAp to achieve the efficient charge separation. Superoxide radicals and holes were found to be the major active species for TC degradation. The toxicity evaluation showed that TC could be transferred to the lower toxic intermediates, and deep oxidation with prolonged reaction time was necessary to eliminate the toxicity of TC. This work demonstrates the surface coordination with subsequent quasi p-n junction mechanism of TC degradation over HAp under visible light, which will stimulate us to explore new efficient photocatalytic systems for the degradation of various contaminants.
Collapse
Affiliation(s)
- Mingwei Wu
- Henan Provincial Engineering Research Center of Green Anticorrosion Technology for Magnesium Alloys, Henan Engineering Research Center for Control and Remediation of Soil Heavy Pollution, College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, PR China
| | - Qinge Fan
- Henan Provincial Engineering Research Center of Green Anticorrosion Technology for Magnesium Alloys, Henan Engineering Research Center for Control and Remediation of Soil Heavy Pollution, College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, PR China
| | - Xiaohui Li
- Henan Provincial Engineering Research Center of Green Anticorrosion Technology for Magnesium Alloys, Henan Engineering Research Center for Control and Remediation of Soil Heavy Pollution, College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, PR China
| | - Penglong Wang
- Henan Provincial Engineering Research Center of Green Anticorrosion Technology for Magnesium Alloys, Henan Engineering Research Center for Control and Remediation of Soil Heavy Pollution, College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, PR China
| | - Caixia Feng
- Henan Provincial Engineering Research Center of Green Anticorrosion Technology for Magnesium Alloys, Henan Engineering Research Center for Control and Remediation of Soil Heavy Pollution, College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, PR China
| | - Li Wang
- Henan Provincial Engineering Research Center of Green Anticorrosion Technology for Magnesium Alloys, Henan Engineering Research Center for Control and Remediation of Soil Heavy Pollution, College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, PR China
| | - Zhixian Chang
- Henan Provincial Engineering Research Center of Green Anticorrosion Technology for Magnesium Alloys, Henan Engineering Research Center for Control and Remediation of Soil Heavy Pollution, College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, PR China.
| | - Wenpeng Wu
- Henan Provincial Engineering Research Center of Green Anticorrosion Technology for Magnesium Alloys, Henan Engineering Research Center for Control and Remediation of Soil Heavy Pollution, College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, PR China.
| | - Ruifeng Chong
- Henan Provincial Engineering Research Center of Green Anticorrosion Technology for Magnesium Alloys, Henan Engineering Research Center for Control and Remediation of Soil Heavy Pollution, College of Chemistry and Molecular Sciences, Henan University, Kaifeng 475004, PR China.
| |
Collapse
|
23
|
Guevara-Cruz M, Hernández-Gómez KG, Condado-Huerta C, González-Salazar LE, Peña-Flores AK, Pichardo-Ontiveros E, Serralde-Zúñiga AE, Sánchez-Tapia M, Maya O, Medina-Vera I, Noriega LG, López-Barradas A, Rodríguez-Lima O, Mata I, Olin-Sandoval V, Torres N, Tovar AR, Velázquez-Villegas LA. Intermittent fasting, calorie restriction, and a ketogenic diet improve mitochondrial function by reducing lipopolysaccharide signaling in monocytes during obesity: A randomized clinical trial. Clin Nutr 2024; 43:1914-1928. [PMID: 39003957 DOI: 10.1016/j.clnu.2024.06.036] [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: 02/27/2024] [Revised: 06/24/2024] [Accepted: 06/30/2024] [Indexed: 07/16/2024]
Abstract
BACKGROUND Mitochondrial dysfunction occurs in monocytes during obesity and contributes to a low-grade inflammatory state; therefore, maintaining good mitochondrial conditions is a key aspect of maintaining health. Dietary interventions are primary strategies for treating obesity, but little is known about their impact on monocyte bioenergetics. Thus, the aim of this study was to evaluate the effects of calorie restriction (CR), intermittent fasting (IF), a ketogenic diet (KD), and an ad libitum habitual diet (AL) on mitochondrial function in monocytes and its modulation by the gut microbiota. METHODS AND FINDINGS A randomized controlled clinical trial was conducted in which individuals with obesity were assigned to one of the 4 groups for 1 month. Subsequently, the subjects received rifaximin and continued with the assigned diet for another month. The oxygen consumption rate (OCR) was evaluated in isolated monocytes, as was the gut microbiota composition in feces and anthropometric and biochemical parameters. Forty-four subjects completed the study, and those who underwent CR, IF and KD interventions had an increase in the maximal respiration OCR (p = 0.025, n2p = 0.159 [0.05, 0.27] 95% confidence interval) in monocytes compared to that in the AL group. The improvement in mitochondrial function was associated with a decrease in monocyte dependence on glycolysis after the IF and KD interventions. Together, diet and rifaximin increased the gut microbiota diversity in the IF and KD groups (p = 0.0001), enriched the abundance of Phascolarctobacterium faecium (p = 0.019) in the CR group and Ruminococcus bromii (p = 0.020) in the CR and KD groups, and reduced the abundance of lipopolysaccharide (LPS)-producing bacteria after CR, IF and KD interventions compared to the AL group at the end of the study according to ANCOVA with covariate adjustment. Spearman's correlation between the variables measured highlighted LPS as a potential modulator of the observed effects. In line with this findings, serum LPS and intracellular signaling in monocytes decreased with the three interventions (CR, p = 0.002; IF, p = 0.001; and KD, p = 0.001) compared to those in the AL group at the end of the study. CONCLUSIONS We conclude that these dietary interventions positively regulate mitochondrial bioenergetic health and improve the metabolic profile of monocytes in individuals with obesity via modulation of the gut microbiota. Moreover, the evaluation of mitochondrial function in monocytes could be used as an indicator of metabolic and inflammatory status, with potential applications in future clinical trials. TRIAL REGISTRATION This trial was registered with ClinicalTrials.gov (NCT05200468).
Collapse
Affiliation(s)
- Martha Guevara-Cruz
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Karla G Hernández-Gómez
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Citlally Condado-Huerta
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Luis E González-Salazar
- Servicio de Nutriología Clínica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Ana Karen Peña-Flores
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Edgar Pichardo-Ontiveros
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Aurora E Serralde-Zúñiga
- Servicio de Nutriología Clínica, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Mónica Sánchez-Tapia
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Otoniel Maya
- Chalmers e-Commons. Chalmers University of Technology, Gotemburg, Vastra Gotaland, Sweden
| | - Isabel Medina-Vera
- Departamento de Metodología de la Investigación, Instituto Nacional de Pediatría, Ciudad de México, Mexico
| | - Lilia G Noriega
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Adriana López-Barradas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Oscar Rodríguez-Lima
- Departamento de Microbiología y Parasitología, Facultad de Medicina, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | - Irma Mata
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Viridiana Olin-Sandoval
- Laboratorio 43. Departamento de Biotecnología y Bioingeniería, Cinvestav-Zacatenco, Ciudad de México, Mexico
| | - Nimbe Torres
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Armando R Tovar
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | - Laura A Velázquez-Villegas
- Departamento de Fisiología de la Nutrición, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico.
| |
Collapse
|
24
|
Eberspacher C, Mascagni D, Pontone S, Arcieri FL, Arcieri S. Topical metronidazole after haemorrhoidectomy to reduce postoperative pain: a systematic review. Updates Surg 2024; 76:1161-1167. [PMID: 39117876 PMCID: PMC11341760 DOI: 10.1007/s13304-024-01930-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/26/2024] [Indexed: 08/10/2024]
Abstract
Excisional haemorrhoidectomy is the gold standard for operating haemorrhoids, but it is accompanied by a significant problem: postoperative pain. Several strategies have been adopted to minimize this condition. Oral metronidazole has been proven to reduce postoperative pain but with some complications. This systematic review was conducted to investigate the effects and general efficacy of topical metronidazole administration and to evaluate its potential superiority over the oral formula. A systematic review of the literature was carried out. Randomized controlled trials published until September 2023 on PubMed, Central, and Web of Science were considered. The primary outcome considered was postoperative pain, which was evaluated using visual analogue scores. The secondary outcomes were analgesic use, return to work, and complications. Six randomized controlled trials were included, with a total of 536 patients. Topical metronidazole was compared with placebo in two studies, with oral formula in three studies, and with placebo and oral administration in one study. Topical metronidazole was found to be effective for treating postoperative pain when compared to a placebo but had no significant advantage over the oral formula. No complications were reported in the studies. Topical and oral metronidazole are effective solutions for postoperative pain after excisional haemorrhoidectomy. No superiority was demonstrated based on the route of administration, and complications were marginal for both formulas. Further studies are required to determine the best metronidazole solution.
Collapse
Affiliation(s)
- Chiara Eberspacher
- Department of Surgery, University of Rome "Sapienza", Viale Regina Elena 324, 00161, Rome, Italy.
| | - Domenico Mascagni
- Department of Surgery, University of Rome "Sapienza", Viale Regina Elena 324, 00161, Rome, Italy
| | - Stefano Pontone
- Department of Surgery, University of Rome "Sapienza", Viale Regina Elena 324, 00161, Rome, Italy
| | - Francesco Leone Arcieri
- Department of Surgery, University of Rome "Sapienza", Viale Regina Elena 324, 00161, Rome, Italy
| | - Stefano Arcieri
- Department of Surgery, University of Rome "Sapienza", Viale Regina Elena 324, 00161, Rome, Italy
| |
Collapse
|
25
|
Liu Y, Wang Y, Wang H. Effects of ciprofloxacin and levofloxacin on initial colonization of intestinal microbiota in Bufo gargarizans at embryonic stages. CHEMOSPHERE 2024; 361:142587. [PMID: 38871193 DOI: 10.1016/j.chemosphere.2024.142587] [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: 04/03/2024] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024]
Abstract
Ciprofloxacin (CIP) and levofloxacin (LEV) are broad-spectrum antibiotics with potent antibacterial activity. Although many studies have shown that antibiotics can lead to gut microbiota disruption, the effects of CIP and LEV on gut microbial colonization at the embryonic stage remain poorly characterized. Here, we evaluated the response of Bufo gargarizans embryos in terms of gut microbiota colonization, growth and developmental stages to CIP and LEV exposure. Embryos treated with 100 μg/L CIP and LEV exhibited significantly reduced diversity and richness of the gut microbiota, as well as altered community structure. Both CIP and LEV treatments resulted in an increase in the pathogenic bacteria Bosea and Aeromonas, and they appeared to be more resistant to CIP than LEV. Additionally, CIP exposure caused reduced total length and delayed the development in B. gargarizans embryos, while LEV increased the total length and promoted embryonic development. The present study revealed the adverse effects of CIP and LEV exposure on host gut microbiota, growth and development during the embryonic stage, and contributed new perspectives to the evaluation of early aquatic ecological risk under CIP and LEV exposure.
Collapse
Affiliation(s)
- Ying Liu
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| | - Yaxi Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China
| | - Hongyuan Wang
- College of Life Sciences, Shaanxi Normal University, Xi'an, Shaanxi, 710119, China.
| |
Collapse
|
26
|
Jia Y, Zhang T, He M, Yang B, Wang Z, Liu Y. Melatonin Protects Against Colistin-Induced Intestinal Inflammation and Microbiota Dysbiosis. J Pineal Res 2024; 76:e12989. [PMID: 38978438 DOI: 10.1111/jpi.12989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/17/2024] [Accepted: 06/25/2024] [Indexed: 07/10/2024]
Abstract
Colistin is renowned as a last-resort antibiotic due to the emergence of multidrug-resistant pathogens. However, its potential toxicity significantly hampers its clinical utilization. Melatonin, chemically known as N-acetyl-5-hydroxytryptamine, is an endogenous hormone produced by the pineal gland and possesses diverse biological functions. However, the protective role of melatonin in alleviating antibiotic-induced intestinal inflammation remains unknown. Herein, we reveal that colistin stimulation markedly elevates intestinal inflammatory levels and compromises the gut barrier. In contrast, pretreatment with melatonin safeguards mice against intestinal inflammation and mucosal damage. Microbial diversity analysis indicates that melatonin supplementation prevents a reduction in the abundance of Erysipelotrichales and Bifidobacteriales, as well as an increase in Desulfovibrionales abundance, following colistin exposure. Remarkably, short-chain fatty acids (SCFAs) analysis shows that propanoic acid contributes to the protective effect of melatonin on colistin-induced intestinal inflammation. Furthermore, the protection effects of melatonin and propanoic acid on LPS-induced cellular inflammation in RAW 264.7 cells are confirmed. Mechanistic investigations suggest that intervention with melatonin and propanoic acid can repress the activation of the TLR4 signal and its downstream NF-κB and MAPK signaling pathways, thereby mitigating the toxic effects of colistin. Our work highlights the unappreciated role of melatonin in preventing the potential detrimental effects of colistin on intestinal health and suggests a combined therapeutic strategy to effectively manage intestinal infectious diseases.
Collapse
Affiliation(s)
- Yuqian Jia
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Tingting Zhang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Mengping He
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Bingqing Yang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Zhiqiang Wang
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
| | - Yuan Liu
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou, China
- Institute of Comparative Medicine, Yangzhou University, Yangzhou, China
| |
Collapse
|
27
|
Wang Y, Fu X, Zhu Y, Lin M, Cai R, Zhu Y, Wu T. An intratumor bacteria-targeted DNA nanocarrier for multifaceted tumor microenvironment intervention. Mater Today Bio 2024; 27:101144. [PMID: 39070095 PMCID: PMC11279327 DOI: 10.1016/j.mtbio.2024.101144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/30/2024] Open
Abstract
Intratumor bacteria, which are involved with complex tumor development mechanisms, can compromise the therapeutic efficiencies of cancer chemotherapeutics. Therefore, the development of anti-tumor agents targeting intratumor bacteria is crucial in overcoming the drug inactivation induced by bacteria colonization. In this study, a double-bundle DNA tetrahedron-based nanocarrier is developed for intratumor bacteria-targeted berberine (Ber) delivery. The combination of aptamer modification and high drug loading efficacy endow the DNA nanocarrier TA@B with enhanced delivery performance in anti-tumor therapy without obvious systemic toxicity. The loaded natural isoquinoline alkaloid Ber exhibits enhanced antimicrobial, anticancer, and immune microenvironment regulation effects, ultimately leading to efficient inhibition of tumor proliferation. This intratumor bacteria-targeted DNA nanoplatform provides a promising strategy in intervening the bacteria-related microenvironment and facilitating tumor therapy.
Collapse
Affiliation(s)
- Yibiao Wang
- Department of Neurosurgery/Department of Pediatrics/Department of Neonatal, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570311, China
| | - Xiaomei Fu
- Department of Neurosurgery/Department of Pediatrics/Department of Neonatal, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570311, China
| | - Yang Zhu
- Department of Neurosurgery/Department of Pediatrics/Department of Neonatal, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570311, China
| | - Mingjing Lin
- Department of Neurosurgery/Department of Pediatrics/Department of Neonatal, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570311, China
| | - Renduan Cai
- Department of Neurosurgery/Department of Pediatrics/Department of Neonatal, Hainan General Hospital/Hainan Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, 570311, China
| | - Yang Zhu
- Fujian Provincial Institutes of Brain Disorders and Brain Sciences/Department of Neurosurgery, Neurosurgery Research Institute, The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, Fujian, China
| | - Tiantian Wu
- School of Pharmacy, Hainan Medical University, Haikou, China
| |
Collapse
|
28
|
Lear L, Hesse E, Buckling A. Disturbances can facilitate prior invasions more than subsequent invasions in microbial communities. Ecol Lett 2024; 27:e14493. [PMID: 39140430 DOI: 10.1111/ele.14493] [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: 01/16/2024] [Revised: 07/02/2024] [Accepted: 07/26/2024] [Indexed: 08/15/2024]
Abstract
Invasions are commonly found to benefit from disturbance events. However, the importance of the relative timing of the invasion and disturbance for invader success and impact on community composition remains uncertain. Here, we experimentally test this by invading a five-species bacterial community on eight separate occasions-four before a disturbance and four after. Invader success and impact on community composition was greatest when the invasion immediately followed the disturbance. However, the subsequent invasions had negligible success or impact. Pre-disturbance, invader success and impact was greatest when the invader was added just before the disturbance. Importantly, however, the first three pre-disturbance invasion events had significantly greater success than the last three post-disturbance invasions. Moreover, these findings were consistent across a range of propagule pressures. Overall, we demonstrate that timing is highly important for both the success and impact on community composition of an invader, with both being lower as time since disturbance progresses.
Collapse
Affiliation(s)
- Luke Lear
- Environment and Sustainability Institute, University of Exeter, Penryn, Cornwall, UK
| | - Elze Hesse
- Environment and Sustainability Institute, University of Exeter, Penryn, Cornwall, UK
| | - Angus Buckling
- Environment and Sustainability Institute, University of Exeter, Penryn, Cornwall, UK
| |
Collapse
|
29
|
Abouelkhair AA, Seleem MN. Exploring novel microbial metabolites and drugs for inhibiting Clostridioides difficile. mSphere 2024; 9:e0027324. [PMID: 38940508 PMCID: PMC11288027 DOI: 10.1128/msphere.00273-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 06/07/2024] [Indexed: 06/29/2024] Open
Abstract
Clostridioides difficile is an enteric pathogen that can cause a range of illnesses from mild diarrhea to pseudomembranous colitis and even death. This pathogen often takes advantage of microbial dysbiosis provoked by antibiotic use. With the increasing incidence and severity of infections, coupled with high recurrence rates, there is an urgent need to identify innovative therapies that can preserve the healthy state of the gut microbiota. In this study, we screened a microbial metabolite library against C. difficile. From a collection of 527 metabolites, we identified 18 compounds with no previously identified antimicrobial activity and metabolites that exhibited potent activity against C. difficile growth. Of these 18 hits, five drugs and three metabolites displayed the most potent anti-C. difficile activity and were subsequently assessed against 20 clinical isolates of C. difficile. These potent agents included ecteinascidin 770 (minimum inhibitory concentration against 50% of isolates [MIC50] ≤0.06 µg/mL); 8-hydroxyquinoline derivatives, such as broxyquinoline and choloroquinaldol (MIC50 = 0.125 µg/mL); ionomycin calcium salt, carbadox, and robenidine hydrochloride (MIC50 = 1 µg/mL); and dronedarone and milbemycin oxime (MIC50 = 4 µg/mL). Unlike vancomycin and fidaxomicin, which are the standard-of-care anti-C. difficile antibiotics, most of these metabolites showed robust bactericidal activity within 2-8 h with minimal impact on the growth of representative members of the normal gut microbiota. These results suggest that the drugs and microbial metabolite scaffolds may offer alternative avenues to address unmet needs in C. difficile disease prevention and treatment. IMPORTANCE The most frequent infection associated with hospital settings is Clostridioides difficile, which can cause fatal diarrhea and severe colitis, toxic megacolon, sepsis, and leaky gut. Those who have taken antibiotics for other illnesses that affect the gut's healthy microbiota are more susceptible to C. difficile infection (CDI). Recently, some reports showed higher recurrence rates and resistance to anti-C. difficile, which may compromise the efficacy of CDI treatment. Our study is significant because it is anticipated to discover novel microbial metabolites and drugs with microbial origins that are safe for the intestinal flora, effective against C. difficile, and reduce the risk of recurrence associated with CDI.
Collapse
Affiliation(s)
- Ahmed A. Abouelkhair
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
- Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
- Department of Bacteriology, Mycology, and Immunology, Faculty of Veterinary Medicine, University of Sadat City, Sadat City, Menoufia, Egypt
| | - Mohamed N. Seleem
- Department of Biomedical Sciences and Pathobiology, Virginia-Maryland College of Veterinary Medicine, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
- Center for One Health Research, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| |
Collapse
|
30
|
Elkrief A, Méndez-Salazar EO, Maillou J, Vanderbilt CM, Gogia P, Desilets A, Messaoudene M, Kelly D, Ladanyi M, Hellmann MD, Zitvogel L, Rudin CM, Routy B, Derosa L, Schoenfeld AJ. Antibiotics are associated with worse outcomes in lung cancer patients treated with chemotherapy and immunotherapy. NPJ Precis Oncol 2024; 8:143. [PMID: 39014160 PMCID: PMC11252311 DOI: 10.1038/s41698-024-00630-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 06/13/2024] [Indexed: 07/18/2024] Open
Abstract
Anti-PD(L)-1 inhibition combined with platinum doublet chemotherapy (Chemo-IO) has become the most frequently used standard of care regimen in patients with non-small cell lung cancer (NSCLC). The negative impact of antibiotics on clinical outcomes prior to anti-PD(L)-1 inhibition monotherapy (IO) has been demonstrated in multiple studies, but the impact of antibiotic exposure prior to initiation of Chemo-IO is controversial. We assessed antibiotic exposures at two time windows: within 60 days prior to therapy (-60 d window) and within 60 days prior to therapy and 42 days after therapy (-60 + 42d window) in 2028 patients with advanced NSCLC treated with Chemo-IO and IO monotherapy focusing on objective response rate (ORR: rate of partial response and complete response), progression-free survival (PFS), and overall survival (OS). We also assessed impact of antibiotic exposure in an independent cohort of 53 patients. Univariable and multivariable analyses were conducted along with a meta-analysis from similar studies. For the -60 d window, in the Chemo-IO group (N = 769), 183 (24%) patients received antibiotics. Antibiotic exposure was associated with worse ORR (27% vs 40%, p = 0.001), shorter PFS (3.9 months vs. 5.9 months, hazard ratio [HR] 1.35, 95%CI 1.1,1.6, p = 0.0012), as well as shorter OS (10 months vs. 15 months, HR 1.50, 95%CI 1.2,1.8, p = 0.00014). After adjusting for known prognostic factors in NSCLC, antibiotic exposure was independently associated with worse PFS (HR 1.39, 95%CI 1.35,1.7, p = 0.002) and OS (HR 1.61, 95%CI 1.28,2.03, p < 0.001). Similar results were obtained in the -60 + 42d window, and also in an independent cohort. In a meta-analysis of patients with NSCLC treated with Chemo-IO (N = 4) or IO monotherapy (N = 13 studies) antibiotic exposure before treatment was associated with worse OS among all patients (n = 11,351) (HR 1.93, 95% CI 1.52, 2.45) and Chemo-IO-treated patients (n = 1201) (HR 1.54, 95% CI 1.28, 1.84). Thus, antibiotics exposure prior to Chemo-IO is common and associated with worse outcomes, even after adjusting for other factors. These results highlight the need to implement antibiotic stewardship in routine oncology practice.
Collapse
Affiliation(s)
- Arielle Elkrief
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- University of Montreal Research Center (CR-CHUM), Montreal, QC, Canada.
- Department of Hematology-Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada.
| | | | - Jade Maillou
- University of Montreal Research Center (CR-CHUM), Montreal, QC, Canada
| | - Chad M Vanderbilt
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Pooja Gogia
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Antoine Desilets
- Department of Hematology-Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | | | - Daniel Kelly
- Informatics Systems, Memorial Sloan Kettering Cancer, New York, NY, USA
| | - Marc Ladanyi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew D Hellmann
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Laurence Zitvogel
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Charles M Rudin
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Bertrand Routy
- University of Montreal Research Center (CR-CHUM), Montreal, QC, Canada
- Department of Hematology-Oncology, Centre Hospitalier de l'Université de Montréal (CHUM), Montreal, QC, Canada
| | - Lisa Derosa
- INSERM U1015, Gustave Roussy Cancer Campus, Université Paris-Saclay, Villejuif, France
| | - Adam J Schoenfeld
- Department of Medicine, Thoracic Oncology Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
- Department of Medicine, Weill Cornell Medical College, New York, NY, USA.
| |
Collapse
|
31
|
Huang J, Lin Y, Ding X, Lin S, Li X, Yan W, Chen M. Alteration of the gut microbiome in patients with heart failure: A systematic review and meta-analysis. Microb Pathog 2024; 192:106647. [PMID: 38788811 DOI: 10.1016/j.micpath.2024.106647] [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: 12/18/2023] [Revised: 04/02/2024] [Accepted: 04/13/2024] [Indexed: 05/26/2024]
Abstract
Recent research has revealed that alterations of the gut microbiome (GM) play a comprehensive role in the pathophysiology of HF. However, findings in this field remain controversial. In this study, we focus on differences in GM diversity and abundance between HF patients and non-HF people, based on previous 16 S ribosomal RNA (16rRNA) gene sequencing. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines, we conducted a comprehensive search of PubMed, Web of Science, Embase, Cochrane Library, and Ovid databases using the keyword "Heart failure" and "Gastrointestinal Microbiome". A significant decrease in alpha diversity was observed in the HF patients (Chao1, I2 = 87.5 %, p < 0.001; Shannon index, I2 = 62.8 %, p = 0.021). At the phylum level, the HF group exhibited higher abundances of Proteobacteria (I2 = 92.0 %, p = 0.004) and Actinobacteria (I2 = 82.5 %, p = 0.010), while Bacteroidetes (I2 = 45.1 %, p = 0.017) and F/B ratio (I2 = 0.0 %, p<0.001) were lower. The Firmicutes showed a decreasing trend but did not reach statistical significance (I2 = 82.3 %, p = 0.127). At the genus level, the relative abundances of Streptococcus, Bacteroides, Alistipes, Bifidobacterium, Escherichia-Shigella, Enterococcus and Klebsiella were increased in the HF group, whereas Ruminococcus, Faecalibacterium, Dorea and Megamona exhibited decreased relative abundances. Dialister, Blautia and Prevotella showed decreasing trends but without statistical significance. This observational meta-analysis suggests that GM changes are associated with HF, manifesting as alterations in GM abundance, disruptions in the production of short-chain fatty acids (SCFAs) bacteria, and an increase in trimethylamine N-oxide (TMAO) producing bacteria.
Collapse
Affiliation(s)
- Jiayi Huang
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China
| | - Yongping Lin
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China
| | - Xiangwei Ding
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China
| | - Song Lin
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China
| | - Xin Li
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China
| | - Wei Yan
- Department of Cardiology, Nanjing Pukou People's Hospital, Nanjing, 211800, China
| | - Minglong Chen
- Department of Cardiology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical Medicine, Nanjing Medical University, Taizhou, 225300, China; Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
32
|
Reveles KR, Gonzales-Luna AJ, Golan Y, Alonso CD, Guthmueller B, Tan X, Bidell MR, Pokhilko V, Crawford CV, Skinner AM. Efficacy of Fecal Microbiota, Live-jslm (REBYOTA®), Among Patients Exposed to Non- Clostridioides difficile Infection Antibiotics: Post Hoc Subgroup Analysis of a Phase 2 Open-Label Study. Open Forum Infect Dis 2024; 11:ofae341. [PMID: 39006315 PMCID: PMC11244191 DOI: 10.1093/ofid/ofae341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/13/2024] [Indexed: 07/16/2024] Open
Abstract
Background Antibiotic use is a major risk factor for recurrent Clostridioides difficile infection (CDI) due to the associated disruption in gut microbiota. Fecal microbiota, live-jslm (REBYOTA®; RBL, previously RBX2660), is the first microbiota-based live biotherapeutic approved by the US Food and Drug Administration to prevent recurrent CDI in adults following standard-of-care antibiotic treatment. To investigate the impact of non-CDI antibiotics on the durability of RBL, a subgroup analysis was conducted on PUNCH™ Open-Label study participants who received non-CDI antibiotics during the period between RBL administration and up to 2 years after. Methods Participants in PUNCH™ Open-Label who received non-CDI antibiotics after RBL administration were included in this subgroup analysis. Treatment response was defined as the absence of CDI diarrhea needing retreatment at the last evaluable time point (8 weeks, 6 months, 1 year, or 2 years) after RBL administration. Results Among participants from PUNCH™ Open-Label, 43 received non-CDI antibiotics after RBL administration but before CDI recurrence as evaluated over a 2-year period. Across all evaluable time points, 86% (37/43) of participants had a treatment response regardless of when non-CDI antibiotic exposure occurred. Treatment response was sustained for a median 470 days (IQR, 212-648) from the first day of non-CDI antibiotic use. Most participants (5/6) with CDI recurrences received a high-risk antibiotic. Conclusions RBL remained efficacious in participants with a history of recurrent CDI after subsequent non-CDI antibiotic exposure. Clinical Trials Registration NCT02589847 (https://clinicaltrials.gov/study/NCT02589847).
Collapse
Affiliation(s)
- Kelly R Reveles
- College of Pharmacy, The University of Texas at Austin, Austin, Texas, USA
| | - Anne J Gonzales-Luna
- Department of Pharmacy Practice and Translational Research, College of Pharmacy, University of Houston, Houston, Texas, USA
| | - Yoav Golan
- Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Carolyn D Alonso
- Division of Infectious Diseases, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | | | - Xing Tan
- Ferring Pharmaceuticals, Parsippany, New Jersey, USA
| | | | | | - Carl V Crawford
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, New York, USA
| | - Andrew M Skinner
- Department of Internal Medicine, School of Medicine, University of Utah, Salt Lake City, Utah, USA
- Infectious Diseases Section, George E. Wahlen Veterans Affairs Medical Center, Salt Lake City, Utah, USA
| |
Collapse
|
33
|
Puerarin alleviates atherosclerosis via the inhibition of Prevotella copri and its trimethylamine production. Gut 2024:gutjnl-2024-331880. [PMID: 38777572 DOI: 10.1136/gutjnl-2024-331880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 04/30/2024] [Indexed: 05/25/2024]
Abstract
OBJECTIVE Puerarin (PU) is a natural compound that exhibits limited oral bioavailability but has shown promise in the treatment of atherosclerosis (AS). However, the precise mechanisms underlying its therapeutic effects remain incompletely understood. This study aimed to investigate the effects of PU and its mechanisms in mitigating AS in both mice and humans. DESIGN The impact of PU on AS was examined in ApoE -/- mice fed a high-fat diet (HFD) and in human patients with carotid artery plaque. To explore the causal link between PU-associated gut microbiota and AS, faecal microbiota transplantation (FMT) and mono-colonisation of mice with Prevotella copri (P. copri) were employed. RESULTS PU alleviated AS by modulating the gut microbiota, as evidenced by alterations in gut microbiota composition and the amelioration of AS following FMT from PU-treated mice into ApoE-/- mice fed HFD. Specifically, PU reduced the abundance of P. copri, which exacerbated AS by producing trimethylamine (TMA). Prolonged mono-colonisation of P. copri undermines the beneficial effects of PU on AS. In clinical, the plaque scores of AS patients were positively correlated with the abundance of P. copri and plasma trimethylamine-N-oxide (TMAO) levels. A 1-week oral intervention with PU effectively decreased P. copri levels and reduced TMAO concentrations in patients with carotid artery plaque. CONCLUSION PU may provide therapeutic benefits in combating AS by targeting P. copri and its production of TMA. TRIAL REGISTRATION NUMBER ChiCTR1900022488.
Collapse
|
34
|
Zebian G, Kreitmann L, Houard M, Piantoni A, Piga G, Ruffier des Aimes S, Holik B, Wallet F, Labreuche J, Nseir S. Immunosuppression at ICU admission is not associated with a higher incidence of ICU-acquired bacterial bloodstream infections: the COCONUT study. Ann Intensive Care 2024; 14:83. [PMID: 38837065 PMCID: PMC11153408 DOI: 10.1186/s13613-024-01314-1] [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: 10/31/2023] [Accepted: 05/16/2024] [Indexed: 06/06/2024] Open
Abstract
BACKGROUND Immunosuppression at intensive care unit (ICU) admission has been associated with a higher incidence of ICU-acquired infections, some of them related to opportunistic pathogens. However, the association of immunosuppression with the incidence, microbiology and outcomes of ICU-acquired bacterial bloodstream infections (BSI) has not been thoroughly investigated. METHODS Retrospective single-centered cohort study in France. All adult patients hospitalized in the ICU of Lille University-affiliated hospital for > 48 h between January 1st and December 31st, 2020, were included, regardless of their immune status. Immunosuppression was defined as active cancer or hematologic malignancy, neutropenia, hematopoietic stem cell and solid organ transplants, use of steroids or immunosuppressive drugs, human immunodeficiency virus infection and genetic immune deficiency. The primary objective was to compare the 28-day cumulative incidence of ICU-acquired bacterial BSI between immunocompromised and non-immunocompromised patients. Secondary objectives were to assess the microbiology and outcomes of ICU-acquired bacterial BSI in the two groups. RESULTS A total of 1313 patients (66.9% males, median age 62 years) were included. Among them, 271 (20.6%) were immunocompromised at ICU admission. Severity scores at admission, the use of invasive devices and antibiotic exposure during ICU stay were comparable between groups. Both prior to and after adjustment for pre-specified baseline confounders, the 28-day cumulative incidence of ICU-acquired bacterial BSI was not statistically different between immunocompromised and non-immunocompromised patients. The distribution of bacteria was comparable between groups, with a majority of Gram-negative bacilli (~ 64.1%). The proportion of multidrug-resistant bacteria was also similar between groups. Occurrence of ICU-acquired bacterial BSI was associated with a longer ICU length-of-stay and a longer duration of invasive mechanical ventilation, with no significant association with mortality. Immune status did not modify the association between occurrence of ICU-acquired bacterial BSI and these outcomes. CONCLUSION The 28-day cumulative incidence of ICU-acquired bacterial BSI was not statistically different between patients with and without immunosuppression at ICU admission.
Collapse
Affiliation(s)
- Ghadi Zebian
- Médecine Intensive-Réanimation, Hôpital R. Salengro, CHU de Lille, Rue E. Laine, 59037, Lille Cedex, France
| | - Louis Kreitmann
- Department of Intensive Care Medicine, Imperial College Healthcare NHS Trust, London, UK
- Centre for Antimicrobial Optimisation, Department of Infectious Disease, Faculty of Medicine, Imperial College London, London, W12 0HS, UK
| | - Marion Houard
- Médecine Intensive-Réanimation, Hôpital R. Salengro, CHU de Lille, Rue E. Laine, 59037, Lille Cedex, France
| | - Antoine Piantoni
- Médecine Intensive-Réanimation, Hôpital R. Salengro, CHU de Lille, Rue E. Laine, 59037, Lille Cedex, France
| | - Gaetan Piga
- Médecine Intensive-Réanimation, Hôpital R. Salengro, CHU de Lille, Rue E. Laine, 59037, Lille Cedex, France
| | - Sarah Ruffier des Aimes
- Médecine Intensive-Réanimation, Hôpital R. Salengro, CHU de Lille, Rue E. Laine, 59037, Lille Cedex, France
| | - Bérénice Holik
- Médecine Intensive-Réanimation, Hôpital R. Salengro, CHU de Lille, Rue E. Laine, 59037, Lille Cedex, France
| | - Frédéric Wallet
- Laboratoire de Bactériologie-Hygiène, CHU de Lille, Centre de Biologie Pathologie, 59000, Lille, France
| | | | - Saad Nseir
- Médecine Intensive-Réanimation, Hôpital R. Salengro, CHU de Lille, Rue E. Laine, 59037, Lille Cedex, France.
- Inserm U1285, Université de Lille, CNRS, UMR 8576-UGSF, 59000, Lille, France.
| |
Collapse
|
35
|
Alves J, Prendki V, Chedid M, Yahav D, Bosetti D, Rello J. Challenges of antimicrobial stewardship among older adults. Eur J Intern Med 2024; 124:5-13. [PMID: 38360513 DOI: 10.1016/j.ejim.2024.01.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/06/2024] [Accepted: 01/15/2024] [Indexed: 02/17/2024]
Abstract
Older adults hospitalized in internal medicine wards or long-term care facilities (LTCF) are progressively increasing. Older adults with multimorbidity are more susceptible to infections, as well as to more vulnerable to adverse effects (and interactions) of antibiotics, resulting in a need for effective and safer strategies for antimicrobial stewardship (ASM), both in hospitalization wards and long-term care facilities. Studies on antimicrobial stewardship in older patients are scarce and guidelines are required. Given the peculiarities of the optimization of antimicrobial prescription in individual older adults for common infections, tactics to overcome barriers need an update. The use of rapid diagnosis tests, biomarkers, de-escalation and switching from intravenous to oral/subcutaneous therapy strategies are examples of successful AMS interventions. AMS interventions are associated with reduced side effects, lower mortality, shorter hospital stays, and reduced costs. The proposed AMS framework in LTCF should focus on five domains: strategic vision, team, interventions, patient-centred care and awareness. Internists can partner with geriatrists, pharmacists and infectious disease specialists to address barriers and to improve patient care.
Collapse
Affiliation(s)
- Joana Alves
- Infectious Diseases Specialist, Head of Local Unit of the Program for Prevention and Control of Infection and Antimicrobial Resistance, Hospital de Braga, Portugal.
| | - Virginie Prendki
- Department of Internal Medicine for the Aged, Department of Rehabilitation and Geriatrics, Geneva University Hospitals, Geneva, Switzerland; Department of Infectious Disease, Geneva University Hospital, Switzerland
| | - Marie Chedid
- Department of Infectious Disease, Geneva University Hospital, Switzerland
| | - Dafna Yahav
- Infectious Diseases Unit, Sheba Medical Centre, Ramat Gan, Israel
| | - Davide Bosetti
- Department of Infectious Disease, Geneva University Hospital, Switzerland; Infection Control Programme and WHO Collaborating Centre for Infection Prevention and Control and Antimicrobial Resistance, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Jordi Rello
- Medicine Department, Universitat Internacional de Catalunya, Spain; Clinical Research Pneumonia and Sepsis (CRIPS) Research Group-Vall d'Hebrón Institute Research (VHIR), Barcelona, Spain; Formation, Recherche, Evaluation (FOREVA), CHU Nîmes, Nîmes, France
| |
Collapse
|
36
|
Pimenta AI, Bernardino RM, Pereira IAC. Role of sulfidogenic members of the gut microbiota in human disease. Adv Microb Physiol 2024; 85:145-200. [PMID: 39059820 DOI: 10.1016/bs.ampbs.2024.04.003] [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: 07/28/2024]
Abstract
The human gut flora comprises a dynamic network of bacterial species that coexist in a finely tuned equilibrium. The interaction with intestinal bacteria profoundly influences the host's development, metabolism, immunity, and overall health. Furthermore, dysbiosis, a disruption of the gut microbiota, can induce a variety of diseases, not exclusively associated with the intestinal tract. The increased consumption of animal protein, high-fat and high-sugar diets in Western countries has been implicated in the rise of chronic and inflammatory illnesses associated with dysbiosis. In particular, this diet leads to the overgrowth of sulfide-producing bacteria, known as sulfidogenic bacteria, which has been linked to inflammatory bowel diseases and colorectal cancer, among other disorders. Sulfidogenic bacteria include sulfate-reducing bacteria (Desulfovibrio spp.) and Bilophila wadsworthia among others, which convert organic and inorganic sulfur compounds to sulfide through the dissimilatory sulfite reduction pathway. At high concentrations, sulfide is cytotoxic and disrupts the integrity of the intestinal epithelium and mucus barrier, triggering inflammation. Besides producing sulfide, B. wadsworthia has revealed significant pathogenic potential, demonstrated in the ability to cause infection, adhere to intestinal cells, promote inflammation, and compromise the integrity of the colonic mucus layer. This review delves into the mechanisms by which taurine and sulfide-driven gut dysbiosis contribute to the pathogenesis of sulfidogenic bacteria, and discusses the role of these gut microbes, particularly B. wadsworthia, in human diseases.
Collapse
Affiliation(s)
- Andreia I Pimenta
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Raquel M Bernardino
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Inês A C Pereira
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal.
| |
Collapse
|
37
|
An R, Zhou X, He P, Lyu C, Wang D. Inulin mitigated antibiotic-induced intestinal microbiota dysbiosis - a comparison of different supplementation stages. Food Funct 2024; 15:5429-5438. [PMID: 38644728 DOI: 10.1039/d3fo05186b] [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: 04/23/2024]
Abstract
Antibiotics are unavoidable to be prescribed to subjects due to different reasons, and they decrease the relative abundance of beneficial microbes. Inulin, a fructan type of polysaccharide carbohydrate, on the contrary, could promote the growth of beneficial microbes. In this study, we investigated the effect of inulin on antibiotic-induced intestinal microbiota dysbiosis and compared their overall impact at different supplementation stages, i.e., post-antibiotic, at the time of antibiotic administration or prior to antibiotic treatment, in the C57BL/6 mice model. Although supplementation of inulin after antibiotic treatment could aid in the reconstruction of the intestinal microbial community its overall impact was limited and no remarkable differences were identified as compared to the spontaneous restoration. On the contrary, the effect of simultaneous and pre-supplementation was more remarkable. Simultaneous inulin supplementation significantly mitigated the antibiotic-induced dysbiosis based on alterations as evaluated using weighted and unweighted UniFrac distance between baseline and after treatment. Moreover, comparing the effect of simultaneous supplementation, pre-supplemented inulin further mitigated the antibiotic-induced dysbiosis, especially on the relative abundance of dominant microbes. Collectively, the current study found that the use of inulin could alleviate antibiotic-induced microbiota dysbiosis, and the best supplementation stage (overall effect as evaluated by beta diversity distance changes) was before the antibiotic treatment, then simultaneous supplementation and supplementation after the antibiotic treatment.
Collapse
Affiliation(s)
- Ran An
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.
| | - Xilong Zhou
- State Key Laboratory of Dairy Biotechnology, Dairy Research Institute, Bright Dairy and Food Co., Ltd, Shanghai, China
| | - Penglin He
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.
| | - Chenang Lyu
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.
| | - Dapeng Wang
- Department of Food Science and Technology, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
38
|
Sandes V, Figueras A, Lima EC. Pharmacovigilance Strategies to Address Resistance to Antibiotics and Inappropriate Use-A Narrative Review. Antibiotics (Basel) 2024; 13:457. [PMID: 38786184 PMCID: PMC11117530 DOI: 10.3390/antibiotics13050457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/11/2024] [Accepted: 05/15/2024] [Indexed: 05/25/2024] Open
Abstract
The spread of antimicrobial resistance (AMR) is a global challenge. Close and continuous surveillance for quick detection of AMR can be difficult, especially in remote places. This narrative review focuses on the contributions of pharmacovigilance (PV) as an auxiliary tool for identifying and monitoring the ineffectiveness, resistance, and inappropriate use of antibiotics (ABs). The terms "drug ineffective", "therapeutic failure", "drug resistance", "pathogen resistance", and "multidrug resistance" were found in PV databases and dictionaries, denoting ineffectiveness. These terms cover a range of problems that should be better investigated because they are useful in warning about possible causes of AMR. "Medication errors", especially those related to dose and indication, and "Off-label use" are highlighted in the literature, suggesting inappropriate use of ABs. Hence, the included studies show that the terms of interest related to AMR and use are not only present but frequent in PV surveillance programs. This review illustrates the feasibility of using PV as a complementary tool for antimicrobial stewardship activities, especially in scenarios where other resources are scarce.
Collapse
Affiliation(s)
- Valcieny Sandes
- Postgraduate Program in Pharmaceutical Sciences, School of Pharmacy, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho-373, Rio de Janeiro 21941-170, RJ, Brazil;
- National Cancer Institute, Pr. da Cruz Vermelha-23, Rio de Janeiro 20230-130, RJ, Brazil
| | | | - Elisangela Costa Lima
- Postgraduate Program in Pharmaceutical Sciences, School of Pharmacy, Federal University of Rio de Janeiro, Av. Carlos Chagas Filho-373, Rio de Janeiro 21941-170, RJ, Brazil;
| |
Collapse
|
39
|
Tao Y, Zheng D, Zou W, Guo T, Liao G, Zhou W. Targeting the cysteine biosynthesis pathway in microorganisms: Mechanism, structure, and drug discovery. Eur J Med Chem 2024; 271:116461. [PMID: 38691891 DOI: 10.1016/j.ejmech.2024.116461] [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: 02/01/2024] [Revised: 04/16/2024] [Accepted: 04/27/2024] [Indexed: 05/03/2024]
Abstract
Owing to the global health crisis of resistant pathogenic infections, researchers are emphasizing the importance of novel prevention and control strategies. Existing antimicrobial drugs predominantly target a few pathways, and their widespread use has pervasively increased drug resistance. Therefore, it is imperative to develop new antimicrobial drugs with novel targets and chemical structures. The de novo cysteine biosynthesis pathway, one of the microbial metabolic pathways, plays a crucial role in pathogenicity and drug resistance. This pathway notably differs from that in humans, thereby representing an unexplored target for developing antimicrobial drugs. Herein, we have presented an overview of cysteine biosynthesis pathways and their roles in the pathogenicity of various microorganisms. Additionally, we have investigated the structure and function of enzymes involved in these pathways as well as have discussed drug design strategies and structure-activity relationships of the enzyme inhibitors. This review provides valuable insights for developing novel antimicrobials and offers new avenues to combat drug resistance.
Collapse
Affiliation(s)
- Ying Tao
- State Key Laboratory of Resource Insects, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Dandan Zheng
- State Key Laboratory of Resource Insects, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Wei Zou
- State Key Laboratory of Resource Insects, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Ting Guo
- State Key Laboratory of Resource Insects, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China
| | - Guojian Liao
- State Key Laboratory of Resource Insects, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| | - Wei Zhou
- State Key Laboratory of Resource Insects, College of Pharmaceutical Sciences, Southwest University, Chongqing, 400715, China.
| |
Collapse
|
40
|
Whiley L, Lawler NG, Zeng AX, Lee A, Chin ST, Bizkarguenaga M, Bruzzone C, Embade N, Wist J, Holmes E, Millet O, Nicholson JK, Gray N. Cross-Validation of Metabolic Phenotypes in SARS-CoV-2 Infected Subpopulations Using Targeted Liquid Chromatography-Mass Spectrometry (LC-MS). J Proteome Res 2024; 23:1313-1327. [PMID: 38484742 PMCID: PMC11002931 DOI: 10.1021/acs.jproteome.3c00797] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 04/06/2024]
Abstract
To ensure biological validity in metabolic phenotyping, findings must be replicated in independent sample sets. Targeted workflows have long been heralded as ideal platforms for such validation due to their robust quantitative capability. We evaluated the capability of liquid chromatography-mass spectrometry (LC-MS) assays targeting organic acids and bile acids to validate metabolic phenotypes of SARS-CoV-2 infection. Two independent sample sets were collected: (1) Australia: plasma, SARS-CoV-2 positive (n = 20), noninfected healthy controls (n = 22) and COVID-19 disease-like symptoms but negative for SARS-CoV-2 infection (n = 22). (2) Spain: serum, SARS-CoV-2 positive (n = 33) and noninfected healthy controls (n = 39). Multivariate modeling using orthogonal projections to latent structures discriminant analyses (OPLS-DA) classified healthy controls from SARS-CoV-2 positive (Australia; R2 = 0.17, ROC-AUC = 1; Spain R2 = 0.20, ROC-AUC = 1). Univariate analyses revealed 23 significantly different (p < 0.05) metabolites between healthy controls and SARS-CoV-2 positive individuals across both cohorts. Significant metabolites revealed consistent perturbations in cellular energy metabolism (pyruvic acid, and 2-oxoglutaric acid), oxidative stress (lactic acid, 2-hydroxybutyric acid), hypoxia (2-hydroxyglutaric acid, 5-aminolevulinic acid), liver activity (primary bile acids), and host-gut microbial cometabolism (hippuric acid, phenylpropionic acid, indole-3-propionic acid). These data support targeted LC-MS metabolic phenotyping workflows for biological validation in independent sample sets.
Collapse
Affiliation(s)
- Luke Whiley
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Nathan G. Lawler
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Annie Xu Zeng
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Alex Lee
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Sung-Tong Chin
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| | - Maider Bizkarguenaga
- Centro
de Investigación Cooperativa en Biociencias—CIC bioGUNE,
Precision Medicine and Metabolism Laboratory, Basque Research and
Technology Alliance, Bizkaia Science and
Technology Park, Building
800, 48160 Derio, Spain
| | - Chiara Bruzzone
- Centro
de Investigación Cooperativa en Biociencias—CIC bioGUNE,
Precision Medicine and Metabolism Laboratory, Basque Research and
Technology Alliance, Bizkaia Science and
Technology Park, Building
800, 48160 Derio, Spain
| | - Nieves Embade
- Centro
de Investigación Cooperativa en Biociencias—CIC bioGUNE,
Precision Medicine and Metabolism Laboratory, Basque Research and
Technology Alliance, Bizkaia Science and
Technology Park, Building
800, 48160 Derio, Spain
| | - Julien Wist
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Chemistry
Department, Universidad del Valle, Cali 76001, Colombia
| | - Elaine Holmes
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Department
of Metabolism Digestion and Reproduction, Faculty of Medicine, Imperial
College London, Sir Alexander Fleming Building, South Kensington, London SW7 2AZ, U.K.
| | - Oscar Millet
- Centro
de Investigación Cooperativa en Biociencias—CIC bioGUNE,
Precision Medicine and Metabolism Laboratory, Basque Research and
Technology Alliance, Bizkaia Science and
Technology Park, Building
800, 48160 Derio, Spain
| | - Jeremy K. Nicholson
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Institute
of Global Health Innovation, Faculty Building South Kensington Campus, Imperial College London, London SW7 2AZ, U.K.
| | - Nicola Gray
- Australian
National Phenome Centre, Health Futures Institute Harry Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
- Centre
for Computational and Systems Medicine, Health Futures Institute Harry
Perkins Institute, Murdoch University, 5 Robin Warren Drive, Perth, WA 6150, Australia
| |
Collapse
|
41
|
Xiao H, Fang LT, Tang AZ, Chen HL, Xu ML, Wei XS, Pang GD, Li CQ. Mycobacterium vaccae alleviates allergic airway inflammation and airway hyper-responsiveness in asthmatic mice by altering intestinal microbiota. Immunology 2024; 171:595-608. [PMID: 38205925 DOI: 10.1111/imm.13750] [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: 07/01/2023] [Accepted: 12/29/2023] [Indexed: 01/12/2024] Open
Abstract
Host immunity can influence the composition of the gut microbiota and consequently affect disease progression. Previously, we reported that a Mycobacterium vaccae vaccine could ameliorate allergic inflammation in asthmatic mice by regulating inflammatory immune processes. Here, we investigated the anti-inflammatory effects of M. vaccae on allergic asthma via gut microbiota modulation. An ovalbumin (OVA)-induced asthmatic murine model was established and treated with M. vaccae. Gut microbiota profiles were determined in 18 BALB/c mice using 16S rDNA gene sequencing and metabolomic profiling was performed using liquid chromatography quadrupole time-of-flight mass spectrometry. Mycobacterium vaccae alleviated airway hyper-reactivity and inflammatory infiltration in mice with OVA-induced allergic asthma. The microbiota of asthmatic mice is disrupted and that this can be reversed with M. vaccae. Additionally, a total of 24 differential metabolites were screened, and the abundance of PI(14:1(9Z)/18:0), a glycerophospholipid, was found to be correlated with macrophage numbers (r = 0.52, p = 0.039). These metabolites may affect chemokine (such as macrophage chemoattractant protein-1) concentrations in the serum, and ultimately affect pulmonary macrophage recruitment. Our data demonstrated that M. vaccae might alleviate airway inflammation and hyper-responsiveness in asthmatic mice by reversing imbalances in gut microbiota. These novel mechanistic insights are expected to pave the way for novel asthma therapeutic strategies.
Collapse
Affiliation(s)
- Huan Xiao
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Li-Ting Fang
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - An-Zhou Tang
- Department of Otorhinolaryngology Head and Neck Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Hong-Liu Chen
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Mei-Li Xu
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiao-Shua Wei
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Guo-Dong Pang
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Chao-Qian Li
- Department of Emergency, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
42
|
Jimonet P, Druart C, Blanquet-Diot S, Boucinha L, Kourula S, Le Vacon F, Maubant S, Rabot S, Van de Wiele T, Schuren F, Thomas V, Walther B, Zimmermann M. Gut Microbiome Integration in Drug Discovery and Development of Small Molecules. Drug Metab Dispos 2024; 52:274-287. [PMID: 38307852 DOI: 10.1124/dmd.123.001605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/04/2024] Open
Abstract
Human microbiomes, particularly in the gut, could have a major impact on the efficacy and toxicity of drugs. However, gut microbial metabolism is often neglected in the drug discovery and development process. Medicen, a Paris-based human health innovation cluster, has gathered more than 30 international leading experts from pharma, academia, biotech, clinical research organizations, and regulatory science to develop proposals to facilitate the integration of microbiome science into drug discovery and development. Seven subteams were formed to cover the complementary expertise areas of 1) pharma experience and case studies, 2) in silico microbiome-drug interaction, 3) in vitro microbial stability screening, 4) gut fermentation models, 5) animal models, 6) microbiome integration in clinical and regulatory aspects, and 7) microbiome ecosystems and models. Each expert team produced a state-of-the-art report of their respective field highlighting existing microbiome-related tools at every stage of drug discovery and development. The most critical limitations are the growing, but still limited, drug-microbiome interaction data to produce predictive models and the lack of agreed-upon standards despite recent progress. In this paper we will report on and share proposals covering 1) how microbiome tools can support moving a compound from drug discovery to clinical proof-of-concept studies and alert early on potential undesired properties stemming from microbiome-induced drug metabolism and 2) how microbiome data can be generated and integrated in pharmacokinetic models that are predictive of the human situation. Examples of drugs metabolized by the microbiome will be discussed in detail to support recommendations from the working group. SIGNIFICANCE STATEMENT: Gut microbial metabolism is often neglected in the drug discovery and development process despite growing evidence of drugs' efficacy and safety impacted by their interaction with the microbiome. This paper will detail existing microbiome-related tools covering every stage of drug discovery and development, current progress, and limitations, as well as recommendations to integrate them into the drug discovery and development process.
Collapse
Affiliation(s)
- Patrick Jimonet
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Céline Druart
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Stéphanie Blanquet-Diot
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Lilia Boucinha
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Stephanie Kourula
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Françoise Le Vacon
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Sylvie Maubant
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Sylvie Rabot
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Tom Van de Wiele
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Frank Schuren
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Vincent Thomas
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Bernard Walther
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| | - Michael Zimmermann
- Medicen Paris Région, Paris, France (P.J.); Pharmabiotic Research Institute, Narbonne, France (C.D.); UMR 454 MEDIS, Université Clermont Auvergne, Clermont-Ferrand, France (S.B.D.); Global Bioinformatics, Evotec ID, Lyon, France (L.B.); Preclinical Sciences & Translational Safety, JNJ Innovative Medicine, Beerse, Belgium (S.K.); Biofortis, Saint-Herblain, France (F.L.V.); Translational Pharmacology Department, Oncodesign Services, Dijon, France (S.M.); Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, Jouy-en-Josas, France (S.R.); Center of Microbial Ecology and Technology, Faculty of Bioscience Engineering, Ghent University, Gent, Belgium (T.V.W.); TNO, Leiden, The Netherlands (F.S.); Lallemand Health Solutions, Blagnac, France (V.T.); Servier, Saclay, France (B.W.); and Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany (M.Z.)
| |
Collapse
|
43
|
Iqbal S, Begum F, Ullah I, Jalal N, Shaw P. Peeling off the layers from microbial dark matter (MDM): recent advances, future challenges, and opportunities. Crit Rev Microbiol 2024:1-21. [PMID: 38385313 DOI: 10.1080/1040841x.2024.2319669] [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: 07/07/2023] [Accepted: 02/10/2024] [Indexed: 02/23/2024]
Abstract
Microbes represent the most common organisms on Earth; however, less than 2% of microbial species in the environment can undergo cultivation for study under laboratory conditions, and the rest of the enigmatic, microbial world remains mysterious, constituting a kind of "microbial dark matter" (MDM). In the last two decades, remarkable progress has been made in culture-dependent and culture-independent techniques. More recently, studies of MDM have relied on culture-independent techniques to recover genetic material through either unicellular genomics or shotgun metagenomics to construct single-amplified genomes (SAGs) and metagenome-assembled genomes (MAGs), respectively, which provide information about evolution and metabolism. Despite the remarkable progress made in the past decades, the functional diversity of MDM still remains uncharacterized. This review comprehensively summarizes the recently developed culture-dependent and culture-independent techniques for characterizing MDM, discussing major challenges, opportunities, and potential applications. These activities contribute to expanding our knowledge of the microbial world and have implications for various fields including Biotechnology, Bioprospecting, Functional genomics, Medicine, Evolutionary and Planetary biology. Overall, this review aims to peel off the layers from MDM, shed light on recent advancements, identify future challenges, and illuminate the exciting opportunities that lie ahead in unraveling the secrets of this intriguing microbial realm.
Collapse
Affiliation(s)
- Sajid Iqbal
- Oujiang Lab (Zhejiang Laboratory for Regenerative Medicine, Vision, and Brain Health), Wenzhou, China
- School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou, China
| | - Farida Begum
- Department of Biochemistry, Abdul Wali Khan University Mardan, Mardan, Pakistan
| | - Ihsan Ullah
- College of Chemical Engineering, Fuzhou University, Fuzhou, China
| | - Nasir Jalal
- Oujiang Lab (Zhejiang Laboratory for Regenerative Medicine, Vision, and Brain Health), Wenzhou, China
| | - Peter Shaw
- Oujiang Lab (Zhejiang Laboratory for Regenerative Medicine, Vision, and Brain Health), Wenzhou, China
| |
Collapse
|
44
|
Kong J, Yang F, Zong Y, Wang M, Jiang S, Ma Z, Li Z, Li W, Cai Y, Zhang H, Zhao X, Wang J. Early-life antibiotic exposure promotes house dust mite-induced allergic airway inflammation by impacting gut microbiota and lung lipid metabolism. Int Immunopharmacol 2024; 128:111449. [PMID: 38199196 DOI: 10.1016/j.intimp.2023.111449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 12/04/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024]
Abstract
Asthma is a chronic inflammatory respiratory disease. Early-life antibiotic exposure is a unique risk factor for the incidence and severity of asthma later in life. Perturbations in microbial-metabolite-immune interaction caused by antibiotics are closely associated with the pathogenesis of allergy and asthma. We investigated the effect of early intervention with common oral antibiotics on later asthma exacerbations and found that different antibiotic exposures can amplify different types of immune responses induced by HDM. Cefixime (CFX) promoted a biased type 2 inflammation, azithromycin (AZM) enhanced Th17 immune response, and cefuroxime axetil (CFA) induced eosinophils recruitment. Moreover, early-life antibiotic exposure can have short- and long-term effects on the abundance, composition, and diversity of the gut microbiota. In the model of CFX-promoted type 2 airway inflammation, fecal metabolomics indicated abnormal lipid metabolism and T cell response. Lipidomic also suggested allergic airway inflammation amplified by CFX is closely associated with abnormal lipid metabolism in lung tissues. Moreover, abnormalities in lipid metabolism-related genes (LMRGs) were found to have cellular heterogeneity be associated with asthma severity by bioinformatics analysis.
Collapse
Affiliation(s)
- Jingwei Kong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Fan Yang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuhan Zong
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Manting Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Shiyuan Jiang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhaotian Ma
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Zhuqing Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Wenle Li
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yuyang Cai
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Huixian Zhang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China; School of Chinese Medicine, Southern Medical University, Guangzhou, China.
| | - Ji Wang
- School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China; National Institute of TCM Constitution and Preventive Medicine, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
45
|
Shao Y, Qi Z, Sang J, Yu Z, Li M, Wang Z, Tu J, Song X, Qi K. Metagenome-Based Analysis of the Microbial Community Structure and Drug-Resistance Characteristics of Livestock Feces in Anhui Province, China. Vet Sci 2024; 11:87. [PMID: 38393105 PMCID: PMC10892912 DOI: 10.3390/vetsci11020087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/25/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
We analyzed metagenome data of feces from sows at different physiological periods reared on large-scale farms in Anhui Province, China, to provide a better understanding of the microbial diversity of the sow intestinal microbiome and the structure of antibiotic-resistance genes (ARGs) and virulence genes it carries. Species annotation of the metagenome showed that in the porcine intestinal microbiome, bacteria were dominant, representing >97% of the microorganisms at each physiological period. Firmicutes and Proteobacteria dominated the bacterial community. In the porcine gut microbiome, the viral component accounted for an average of 0.65%, and the species annotation results indicated that most viruses were phages. In addition, we analyzed the microbiome for ARGs and virulence genes. Multidrug-like, MLS-like, and tetracycline-like ARGs were most abundant in all samples. Evaluation of the resistance mechanisms indicated that antibiotic inactivation was the main mechanism of action in the samples. It is noteworthy that there was a significant positive correlation between ARGs and the total microbiome. Moreover, comparative analysis with the Virulence Factor Database showed that adhesion virulence factors were most abundant.
Collapse
Affiliation(s)
- Ying Shao
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (J.S.); (Z.Y.); (M.L.); (Z.W.); (J.T.)
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Zhao Qi
- School of Information and Artificial Intelligence, Anhui Agricultural University, Hefei 230036, China;
| | - Jinhui Sang
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (J.S.); (Z.Y.); (M.L.); (Z.W.); (J.T.)
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Zhaorong Yu
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (J.S.); (Z.Y.); (M.L.); (Z.W.); (J.T.)
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Min Li
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (J.S.); (Z.Y.); (M.L.); (Z.W.); (J.T.)
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Zhenyu Wang
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (J.S.); (Z.Y.); (M.L.); (Z.W.); (J.T.)
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Jian Tu
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (J.S.); (Z.Y.); (M.L.); (Z.W.); (J.T.)
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Xiangjun Song
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (J.S.); (Z.Y.); (M.L.); (Z.W.); (J.T.)
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Kezong Qi
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China; (Y.S.); (J.S.); (Z.Y.); (M.L.); (Z.W.); (J.T.)
- Anhui Province Key Laboratory of Veterinary Pathobiology and Disease Control, College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| |
Collapse
|
46
|
Engelsberger V, Gerhard M, Mejías-Luque R. Effects of Helicobacter pylori infection on intestinal microbiota, immunity and colorectal cancer risk. Front Cell Infect Microbiol 2024; 14:1339750. [PMID: 38343887 PMCID: PMC10853882 DOI: 10.3389/fcimb.2024.1339750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 01/10/2024] [Indexed: 02/15/2024] Open
Abstract
Infecting about half of the world´s population, Helicobacter pylori is one of the most prevalent bacterial infections worldwide and the strongest known risk factor for gastric cancer. Although H. pylori colonizes exclusively the gastric epithelium, the infection has also been associated with various extragastric diseases, including colorectal cancer (CRC). Epidemiological studies reported an almost two-fold increased risk for infected individuals to develop CRC, but only recently, direct causal and functional links between the chronic infection and CRC have been revealed. Besides modulating the host intestinal immune response, H. pylori is thought to increase CRC risk by inducing gut microbiota alterations. It is known that H. pylori infection not only impacts the gastric microbiota at the site of infection but also leads to changes in bacterial colonization in the distal large intestine. Considering that the gut microbiome plays a driving role in CRC, H. pylori infection emerges as a key factor responsible for promoting changes in microbiome signatures that could contribute to tumor development. Within this review, we want to focus on the interplay between H. pylori infection, changes in the intestinal microbiota, and intestinal immunity. In addition, the effects of H. pylori antibiotic eradication therapy will be discussed.
Collapse
Affiliation(s)
| | | | - Raquel Mejías-Luque
- Institute for Medical Microbiology, Immunology and Hygiene, TUM School of Medicine and Health, Department Preclinical Medicine, Technical University of Munich, Munich, Germany
| |
Collapse
|
47
|
Liu GY, Yu D, Fan MM, Zhang X, Jin ZY, Tang C, Liu XF. Antimicrobial resistance crisis: could artificial intelligence be the solution? Mil Med Res 2024; 11:7. [PMID: 38254241 PMCID: PMC10804841 DOI: 10.1186/s40779-024-00510-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Antimicrobial resistance is a global public health threat, and the World Health Organization (WHO) has announced a priority list of the most threatening pathogens against which novel antibiotics need to be developed. The discovery and introduction of novel antibiotics are time-consuming and expensive. According to WHO's report of antibacterial agents in clinical development, only 18 novel antibiotics have been approved since 2014. Therefore, novel antibiotics are critically needed. Artificial intelligence (AI) has been rapidly applied to drug development since its recent technical breakthrough and has dramatically improved the efficiency of the discovery of novel antibiotics. Here, we first summarized recently marketed novel antibiotics, and antibiotic candidates in clinical development. In addition, we systematically reviewed the involvement of AI in antibacterial drug development and utilization, including small molecules, antimicrobial peptides, phage therapy, essential oils, as well as resistance mechanism prediction, and antibiotic stewardship.
Collapse
Affiliation(s)
- Guang-Yu Liu
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, Hangzhou, 311121, China
| | - Dan Yu
- National Key Discipline of Pediatrics Key Laboratory of Major Diseases in Children Ministry of Education, Laboratory of Dermatology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Mei-Mei Fan
- Department of Immunology and Pathogen Biology, School of Basic Medical Sciences, Hangzhou Normal University, Key Laboratory of Aging and Cancer Biology of Zhejiang Province, Key Laboratory of Inflammation and Immunoregulation of Hangzhou, Hangzhou Normal University, Hangzhou, 311121, China
| | - Xu Zhang
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ze-Yu Jin
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Christoph Tang
- Sir William Dunn School of Pathology, University of Oxford, Oxford, OX1 3RE, UK.
| | - Xiao-Fen Liu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Key Laboratory of Clinical Pharmacology of Antibiotics, National Health Commission of the People's Republic of China, National Clinical Research Centre for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
48
|
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
|
49
|
Wang S, Ju D, Zeng X. Mechanisms and Clinical Implications of Human Gut Microbiota-Drug Interactions in the Precision Medicine Era. Biomedicines 2024; 12:194. [PMID: 38255298 PMCID: PMC10813426 DOI: 10.3390/biomedicines12010194] [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: 11/05/2023] [Revised: 12/09/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
The human gut microbiota, comprising trillions of microorganisms residing in the gastrointestinal tract, has emerged as a pivotal player in modulating various aspects of human health and disease. Recent research has shed light on the intricate relationship between the gut microbiota and pharmaceuticals, uncovering profound implications for drug metabolism, efficacy, and safety. This review depicted the landscape of molecular mechanisms and clinical implications of dynamic human gut Microbiota-Drug Interactions (MDI), with an emphasis on the impact of MDI on drug responses and individual variations. This review also discussed the therapeutic potential of modulating the gut microbiota or harnessing its metabolic capabilities to optimize clinical treatments and advance personalized medicine, as well as the challenges and future directions in this emerging field.
Collapse
Affiliation(s)
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China;
| | - Xian Zeng
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China;
| |
Collapse
|
50
|
Shen P, Cheng P, Li Y, Zong G, Deng R, Qian C, Zhao Y, Wei Z, Lu Y. Unveiling the covert interaction between gut microbiota and neutrophils to drive colorectal cancer metastasis. Eur J Pharmacol 2024; 962:176217. [PMID: 38036200 DOI: 10.1016/j.ejphar.2023.176217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/26/2023] [Accepted: 11/16/2023] [Indexed: 12/02/2023]
Abstract
The formation of the microenvironment preceding liver metastasis is intricately linked to the intestinal tract. In recent years, mounting evidence has revealed the significant involvement of neutrophil extracellular traps (NETs) in tumor metastasis, particularly in liver metastasis. Disruption of the intestinal barrier can lead to the translocation of bacteria and their metabolites, such as lipopolysaccharide, into the liver. As the primary defense against pathogens, NETs help eliminate gut-derived toxins and shape the liver's inflammatory and immunosuppressive environment. However, this double-edged sword effect can potentially stimulate tumor metastasis by creating a fertile ground for the growth of intestinal tumor cells due to impaired liver tissue and reduced activity of killer immune cells. This comprehensive review systematically describes the influence factors and mechanisms of NETs in colon cancer metastasis and explores their potential as biomarkers and therapeutic targets for liver metastasis.
Collapse
Affiliation(s)
- Peiliang Shen
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Peng Cheng
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yanan Li
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Gangfan Zong
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Rui Deng
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Cheng Qian
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yang Zhao
- School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacolgy and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing, 210023, China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|