1
|
McCoubrey LE, Shen C, Mwasambu S, Favaron A, Sangfuang N, Thomaidou S, Orlu M, Globisch D, Basit AW. Characterising and preventing the gut microbiota's inactivation of trifluridine, a colorectal cancer drug. Eur J Pharm Sci 2024; 203:106922. [PMID: 39368784 DOI: 10.1016/j.ejps.2024.106922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/01/2024] [Accepted: 10/02/2024] [Indexed: 10/07/2024]
Abstract
The gut microbiome can metabolise hundreds of drugs, potentially affecting their bioavailability and pharmacological effect. As most gut bacteria reside in the colon, drugs that reach the colon in significant proportions may be most impacted by microbiome metabolism. In this study the anti-colorectal cancer drug trifluridine was used as a model drug for characterising metabolism by the colonic microbiota, identifying correlations between bacterial species and individuals' rates of microbiome drug inactivation, and developing strategies to prevent drug inactivation following targeted colonic delivery. High performance liquid chromatography and ultra-high performance liquid chromatography coupled with high resolution tandem mass spectrometry demonstrated trifluridine's variable and multi-route metabolism by the faecal microbiota sourced from six healthy humans. Here, four drug metabolites were linked to the microbiome for the first time. Metagenomic sequencing of the human microbiota samples revealed their composition, which facilitated prediction of individual donors' microbial trifluridine inactivation. Notably, the abundance of Clostridium perfringens strongly correlated with the extent of trifluridine inactivation by microbiota samples after 2 hours (R2 = 0.8966). Finally, several strategies were trialled for the prevention of microbial trifluridine metabolism. It was shown that uridine, a safe and well-tolerated molecule, significantly reduced the microbiota's metabolism of trifluridine by acting as a competitive enzyme inhibitor. Further, uridine was found to provide prebiotic effects. The findings in this study greatly expand knowledge on trifluridine's interactions with the gut microbiome and provide valuable insights for investigating the microbiome metabolism of other drugs. The results demonstrate how protection strategies could enhance the colonic stability of microbiome-sensitive drugs.
Collapse
Affiliation(s)
- Laura E McCoubrey
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Chenghao Shen
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Sydney Mwasambu
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University, 75124 Uppsala, Sweden
| | - Alessia Favaron
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Nannapat Sangfuang
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Stavrina Thomaidou
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Mine Orlu
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom
| | - Daniel Globisch
- Department of Chemistry - BMC, Science for Life Laboratory, Uppsala University, 75124 Uppsala, Sweden
| | - Abdul W Basit
- UCL School of Pharmacy, 29-39 Brunswick Square, London, WC1N 1AX, United Kingdom.
| |
Collapse
|
2
|
Dasriya VL, Samtiya M, Ranveer S, Dhillon HS, Devi N, Sharma V, Nikam P, Puniya M, Chaudhary P, Chaudhary V, Behare PV, Dhewa T, Vemuri R, Raposo A, Puniya DV, Khedkar GD, Vishweswaraiah RH, Vij S, Alarifi SN, Han H, Puniya AK. Modulation of gut-microbiota through probiotics and dietary interventions to improve host health. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:6359-6375. [PMID: 38334314 DOI: 10.1002/jsfa.13370] [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: 11/21/2023] [Revised: 02/02/2024] [Accepted: 02/07/2024] [Indexed: 02/10/2024]
Abstract
Dietary patterns play an important role in regards to the modulation and control of the gut microbiome composition and function. The interaction between diet and microbiota plays an important role in order to maintain intestinal homeostasis, which ultimately affect the host's health. Diet directly impacts the microbes that inhabit the gastrointestinal tract (GIT), which then contributes to the production of secondary metabolites, such as short-chain fatty acids, neurotransmitters, and antimicrobial peptides. Dietary consumption with genetically modified probiotics can be the best vaccine delivery vector and protect cells from various illnesses. A holistic approach to disease prevention, treatment, and management takes these intrinsically linked diet-microbes, microbe-microbe interactions, and microbe-host interactions into account. Dietary components, such as fiber can modulate beneficial gut microbiota, and they have resulting ameliorative effects against metabolic disorders. Medical interventions, such as antibiotic drugs can conversely have detrimental effects on gut microbiota by disputing the balance between Bacteroides and firmicute, which contribute to continuing disease states. We summarize the known effects of various dietary components, such as fibers, carbohydrates, fatty acids, vitamins, minerals, proteins, phenolic acids, and antibiotics on the composition of the gut microbiota in this article in addition to the beneficial effect of genetically modified probiotics and consequentially their role in regards to shaping human health. © 2024 The Authors. Journal of The Science of Food and Agriculture published by John Wiley & Sons Ltd on behalf of Society of Chemical Industry.
Collapse
Affiliation(s)
| | - Mrinal Samtiya
- Department of Nutrition Biology, School of Interdisciplinary and Applied Sciences, Central University of Haryana, Mahendergarh, India
| | - Soniya Ranveer
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, India
| | | | - Nishu Devi
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, India
| | - Vikas Sharma
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, India
| | - Pranali Nikam
- College of Dairy Science and Food Technology, Dau Shri Vasudev Chandrakar, Kamdhenu University, Raipur, India
| | - Monica Puniya
- Science and Standards Division, Food Safety and Standards Authority of India, FDA Bhawan, New Delhi, India
| | - Priya Chaudhary
- Microbiology Department, VCSG Government Institute of Medical Science and Research, Srinagar, India
| | - Vishu Chaudhary
- University Institute of Biotechnology, Chandigarh University, Sahibzada Ajit Singh Nagar, India
| | - Pradip V Behare
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, India
| | - Tejpal Dhewa
- Department of Nutrition Biology, School of Interdisciplinary and Applied Sciences, Central University of Haryana, Mahendergarh, India
| | - Ravichandra Vemuri
- Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC, USA
| | - António Raposo
- CBIOS (Research Center for Biosciences and Health Technologies), Universidade Lusófona de Humanidades e Tecnologias, Lisboa, Portugal
| | - Dharun Vijay Puniya
- Center of One Health, College of Veterinary Science, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, India
| | - Gulab D Khedkar
- Paul Hebert Center for DNA Barcoding and Biodiversity Studies, Dr Babasaheb Ambedkar Marathwada University, Aurangabad, India
| | | | - Shilpa Vij
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, India
| | - Sehad N Alarifi
- Department of Food and Nutrition Science, Al-Quwayiyah College of Sciences and Humanities, Shaqra University, Shaqraa, Saudi Arabia
| | - Heesup Han
- College of Hospitality and Tourism Management, Sejong University, Seoul, South Korea
| | - Anil Kumar Puniya
- Dairy Microbiology Division, ICAR-National Dairy Research Institute, Karnal, India
| |
Collapse
|
3
|
Gu L, Ni Y, Wang B, Kong L, Yu S, Tang Y, Zhu P, Shao S, Tao F, Liu K. Antibiotic exposure associated with nighttime sleep duration and daytime sleepiness in newlyweds. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2024; 31:6350-6371. [PMID: 38148462 DOI: 10.1007/s11356-023-31475-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/06/2023] [Indexed: 12/28/2023]
Abstract
Few studies have explored the relationship between antibiotic exposure and sleep in newlyweds. We applied the actor-partner interdependence moderation model to estimate the relationships of antibiotic exposure with nighttime sleep duration (weekday, weekend, and average sleep durations) and daytime sleepiness in newlyweds. We found that 99.0% of the 2698 enrolled individuals were exposed to at least one antibiotic. Among the newlyweds, exposure to florfenicol (β, - 0.077; 95% confidence interval [CI], - 0.143, - 0.011), exposure to chloramphenicols (- 0.086 [- 0.160, - 0.011]), and exposure to veterinary antibiotics (VAs) (- 0.106 [- 0.201, - 0.010]) were negatively associated with weekday sleep duration. Florfenicol, chloramphenicols, and VAs were also inversely related to average sleep duration in the newlyweds. Ciprofloxacin and cyadox exposure was significantly associated with an increase of 0.264 (0.030, 0.497) and (0.375 [0.088, 0.663]) Epworth Sleepiness Scale (ESS) scores in the newlyweds, respectively. Gender moderated the actor-partner effects of erythromycin and tetracyclines on the newlyweds' weekday sleep duration and ESS scores. Overall, exposure to florfenicol, chloramphenicols, and VAs shortened weekday and average sleep durations of newlyweds. Exposure to ciprofloxacin and cyadox promoted daytime sleepiness. Gender moderated the actor-partner effects of specific antibiotics on the weekday sleep duration and ESS scores of the newlyweds.
Collapse
Affiliation(s)
- Lvfen Gu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Yachao Ni
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Baolin Wang
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Li Kong
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Shuixin Yu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Ying Tang
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Peng Zhu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Shanshan Shao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Fangbiao Tao
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China
| | - Kaiyong Liu
- School of Public Health, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Key Laboratory of Population Health Across Life Cycle (Anhui Medical University), Ministry of Education of the People's Republic of China, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- NHC Key Laboratory of Study On Abnormal Gametes and Reproductive Tract, No 81 Meishan Road, Hefei, 230032, Anhui, China.
- Anhui Provincial Key Laboratory of Population Health and Aristogenics, Anhui Medical University, No 81 Meishan Road, Hefei, 230032, Anhui, China.
| |
Collapse
|
4
|
Andrew B, Zhou A, Trinh S, Srinivasan A, Vasudevan A. Letter: Antibiotics in gut dysbiosis and development of inflammatory bowel disease-More thought is needed. Aliment Pharmacol Ther 2023; 58:838-839. [PMID: 37768293 DOI: 10.1111/apt.17678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/29/2023]
Abstract
LINKED CONTENTThis article is linked to Oh et al papers. To view these articles, visit https://doi.org/10.1111/apt.17542 and https://doi.org/10.1111/apt.17696.
Collapse
Affiliation(s)
- Bridgette Andrew
- Department of Gastroenterology, Eastern Health, Melbourne, Australia
| | - Annie Zhou
- Department of Gastroenterology, Eastern Health, Melbourne, Australia
| | - Steven Trinh
- Department of Gastroenterology, Eastern Health, Melbourne, Australia
| | - Ashish Srinivasan
- Department of Gastroenterology, Eastern Health, Melbourne, Australia
- Monash University, Melbourne, Australia
| | - Abhinav Vasudevan
- Department of Gastroenterology, Eastern Health, Melbourne, Australia
- Monash University, Melbourne, Australia
| |
Collapse
|
5
|
Vázquez-Cuesta S, Lozano García N, Fernández AI, Olmedo M, Kestler M, Alcalá L, Marín M, Bermejo J, Díaz FFA, Muñoz P, Bouza E, Reigadas E. Microbiome profile and calprotectin levels as markers of risk of recurrent Clostridioides difficile infection. Front Cell Infect Microbiol 2023; 13:1237500. [PMID: 37780848 PMCID: PMC10534046 DOI: 10.3389/fcimb.2023.1237500] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 08/23/2023] [Indexed: 10/03/2023] Open
Abstract
Introduction Clostridioides difficile infection (CDI) is the main cause of nosocomial diarrhoea in developed countries. Recurrent CDI (R-CDI), which affects 20%-30% of patients and significantly increases hospital stay and associated costs, is a key challenge. The main objective of this study was to explore the role of the microbiome and calprotectin levels as predictive biomarkers of R-CDI. Methods We prospectively (2019-2021) included patients with a primary episode of CDI. Clinical data and faecal samples were collected. The microbiome was analysed by sequencing the hypervariable V4 region of the 16S rRNA gene on an Illumina Miseq platform. Results We enrolled 200 patients with primary CDI, of whom 54 developed R-CDI and 146 did not. We analysed 200 primary samples and found that Fusobacterium increased in abundance, while Collinsella, Senegalimassilia, Prevotella and Ruminococcus decreased in patients with recurrent versus non-recurrent disease. Elevated calprotectin levels correlated significantly with R-CDI (p=0.01). We built a risk index for R-CDI, including as prognostic factors age, sex, immunosuppression, toxin B amplification cycle, creatinine levels and faecal calprotectin levels (overall accuracy of 79%). Discussion Calprotectin levels and abundance of microbial genera such as Fusobacterium and Prevotella in primary episodes could be useful as early markers of R-CDI. We propose a readily available model for prediction of R-CDI that can be applied at the initial CDI episode. The use of this tool could help to better tailor treatments according to the risk of R-CDI.
Collapse
Affiliation(s)
- Silvia Vázquez-Cuesta
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Biochemistry and Molecular Biology Department, Faculty of Biology, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Nuria Lozano García
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Ana I. Fernández
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - María Olmedo
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Martha Kestler
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| | - Luis Alcalá
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Mercedes Marín
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Javier Bermejo
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Francisco Fernández-Avilés Díaz
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Cardiology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - Patricia Muñoz
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Emilio Bouza
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
- Centro de Investigación Biomédica en red de Enfermedades Respiratorias (CIBERES CB06/06/0058), Madrid, Spain
| | - Elena Reigadas
- Department of Clinical Microbiology and Infectious Diseases, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Medicine Department, School of Medicine, Universidad Complutense de Madrid (UCM), Madrid, Spain
| |
Collapse
|
6
|
Gnatzy L, Ismailos G, Vertzoni M, Reppas C. Managing the clinical effects of drug-induced intestinal dysbiosis with a focus to antibiotics: Challenges and opportunities. Eur J Pharm Sci 2023; 188:106510. [PMID: 37380062 DOI: 10.1016/j.ejps.2023.106510] [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: 04/03/2023] [Revised: 06/23/2023] [Accepted: 06/26/2023] [Indexed: 06/30/2023]
Abstract
The term "intestinal dysbiosis" is used for indicating change(s) of the intestinal microbiota which have been associated with the development of diseases and the deterioration of disease treatments in humans. In this review, documented clinical effects of drug-induced intestinal dysbiosis are briefly presented, and methodologies which could be considered for the management of drug-induced intestinal dysbiosis based on clinical data are critically reviewed. Until relevant methodologies are optimized and/or their effectiveness to the general population is confirmed, and, since drug-induced intestinal dysbiosis refers predominantly to antibiotic-specific intestinal dysbiosis, a pharmacokinetically-based approach for mitigating the impact of antimicrobial therapy on intestinal dysbiosis is proposed.
Collapse
Affiliation(s)
- Lea Gnatzy
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece
| | - George Ismailos
- Experimental, Research and Training Center ELPEN, ELPEN Pharmaceuticals, Pikermi, Greece; National Antimicrobial Testing Committee, Athens, Greece
| | - Maria Vertzoni
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece
| | - Christos Reppas
- Department of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece.
| |
Collapse
|
7
|
Mokrani D, Chommeloux J, Pineton de Chambrun M, Hékimian G, Luyt CE. Antibiotic stewardship in the ICU: time to shift into overdrive. Ann Intensive Care 2023; 13:39. [PMID: 37148398 PMCID: PMC10163585 DOI: 10.1186/s13613-023-01134-9] [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: 01/05/2023] [Accepted: 04/20/2023] [Indexed: 05/08/2023] Open
Abstract
Antibiotic resistance is a major health problem and will be probably one of the leading causes of deaths in the coming years. One of the most effective ways to fight against resistance is to decrease antibiotic consumption. Intensive care units (ICUs) are places where antibiotics are widely prescribed, and where multidrug-resistant pathogens are frequently encountered. However, ICU physicians may have opportunities to decrease antibiotics consumption and to apply antimicrobial stewardship programs. The main measures that may be implemented include refraining from immediate prescription of antibiotics when infection is suspected (except in patients with shock, where immediate administration of antibiotics is essential); limiting empiric broad-spectrum antibiotics (including anti-MRSA antibiotics) in patients without risk factors for multidrug-resistant pathogens; switching to monotherapy instead of combination therapy and narrowing spectrum when culture and susceptibility tests results are available; limiting the use of carbapenems to extended-spectrum beta-lactamase-producing Enterobacteriaceae, and new beta-lactams to difficult-to-treat pathogen (when these news beta-lactams are the only available option); and shortening the duration of antimicrobial treatment, the use of procalcitonin being one tool to attain this goal. Antimicrobial stewardship programs should combine these measures rather than applying a single one. ICUs and ICU physicians should be at the frontline for developing antimicrobial stewardship programs.
Collapse
Affiliation(s)
- David Mokrani
- Service de Médecine Intensive Réanimation, Institut de Cardiologie, ICAN, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne-Université, Hôpital Pitié-Salpêtrière, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Juliette Chommeloux
- Service de Médecine Intensive Réanimation, Institut de Cardiologie, ICAN, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne-Université, Hôpital Pitié-Salpêtrière, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Marc Pineton de Chambrun
- Service de Médecine Intensive Réanimation, Institut de Cardiologie, ICAN, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne-Université, Hôpital Pitié-Salpêtrière, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Guillaume Hékimian
- Service de Médecine Intensive Réanimation, Institut de Cardiologie, ICAN, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne-Université, Hôpital Pitié-Salpêtrière, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France
| | - Charles-Edouard Luyt
- Service de Médecine Intensive Réanimation, Institut de Cardiologie, ICAN, Groupe Hospitalier Pitié-Salpêtrière, Assistance Publique-Hôpitaux de Paris, Sorbonne-Université, Hôpital Pitié-Salpêtrière, 47-83, Boulevard de l'Hôpital, 75651, Paris Cedex 13, France.
- Sorbonne Université, INSERM, UMRS_1166-ICAN Institute of Cardiometabolism and Nutrition, Paris, France.
| |
Collapse
|
8
|
Li Y, Fu R, Li R, Zeng J, Liu T, Li X, Jiang W. Causality of gut microbiome and hypertension: A bidirectional mendelian randomization study. Front Cardiovasc Med 2023; 10:1167346. [PMID: 37215554 PMCID: PMC10192878 DOI: 10.3389/fcvm.2023.1167346] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 04/18/2023] [Indexed: 05/24/2023] Open
Abstract
Background & Aims The pathogenesis of hypertension involves a diverse range of genetic, environmental, hemodynamic, and more causative factors. Recent evidence points to an association between the gut microbiome and hypertension. Given that the microbiota is in part determined by host genetics, we used the two-sample Mendelian randomization (MR) analysis to address the bidirectional causal link between gut microbiota and hypertension. Methods We selected genetic variants (P < 1 × 10-5) for gut microbiota (n = 18,340) from the MiBioGen study. Genetic association estimates for hypertension were extracted from genome-wide association study (GWAS) summary statistics on 54,358 cases and 408,652 controls. Seven complementary MR methods were implemented, including the inverse-variance weighted (IVW) method, followed by sensitivity analyses to verify the robustness of the results. Reverse-direction MR analyses were further conducted to probe if there was a reverse causative relationship. Bidirectional MR analysis then examines a modulation of gut microbiota composition by hypertension. Results At the genus level, our MR estimates from gut microbiome to hypertension showed that there were 5 protective factors Allisonella, Parabacteroide, Phascolarctobacterium, Senegalimassilia, and unknowngenus (id.1000000073), while 6 genera Clostridiuminnocuum, Eubacteriumcoprostanoligenes, Eubacteriumfissicatena, Anaerostipes, LachnospiraceaeFCS020, and unknowngenus (id.2041) are risk factors. The Alcaligenaceae and ClostridialesvadinBB60 were detrimental and beneficial at the family level, respectively. In contrast, the MR results of hypertension-gut flora showed hypertensive states can lead to an increased abundance of Eubacteriumxylanophilum, Eisenbergiella, and Lachnospiraceae and a lower abundance of Alistipes, Bilophila, Butyricimonas, and Phascolarctobacterium. Conclusion Altered gut microbiota is a causal factor in the development of hypertension, and hypertension causes imbalances in the intestinal flora. Substantial research is still needed to find the key gut flora and explore the specific mechanisms of their effects so that new biomarkers can be found for blood pressure control.
Collapse
|
9
|
Mileva R, Petkova T, Yaneva Z, Milanova A. Investigation of the Effect of pH on the Adsorption-Desorption of Doxycycline in Feed for Small Ruminants. Antibiotics (Basel) 2023; 12:antibiotics12020268. [PMID: 36830179 PMCID: PMC9952683 DOI: 10.3390/antibiotics12020268] [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: 01/12/2023] [Revised: 01/24/2023] [Accepted: 01/26/2023] [Indexed: 02/03/2023] Open
Abstract
Orally administered tetracycline antibiotics interact with feed, which may impact their bioavailability and efficacy. Therefore, the pH-dependent adsorption of doxycycline and its interaction with feed for ruminants was studied in vitro. Adsorption experiments on animal feed (135 and 270 mg) with initial doxycycline concentrations of 35, 75, and 150 µg/mL were performed. Desorption experiments were conducted by agitation of a predetermined mass of doxycycline-loaded animal feed in PBS, at pH = 3.0, 6.0, and 7.4, to simulate changes in the gastrointestinal tract. Antibiotic concentrations were determined by LC-MS/MS analysis. The adsorption/desorption of doxycycline was described by mathematical models. Chemisorption with strong intermolecular interactions between the active functional groups of doxycycline and the organic biomass was found. The experimental release curve comprised three sections: initial prolonged 27-30% release (pH = 6.0), followed by moderate 56-59% release (pH = 3.0), and final 63-74% release (pH = 7.4). The sigmoidal model showed a considerable role of diffusion with an initial prevalence of desorption and a decreased desorption rate thereafter. The Weibull equation revealed an initial release stage followed by a lag time section and sustained release. The study of doxycycline adsorption by the animal feed proved a maximum 80% encapsulation efficiency and revealed initial diffusion followed by chemisorption. The highest release efficiency of 74% suggests high bioavailability of doxycycline after oral administration in ruminants.
Collapse
|
10
|
McCoubrey LE, Favaron A, Awad A, Orlu M, Gaisford S, Basit AW. Colonic drug delivery: Formulating the next generation of colon-targeted therapeutics. J Control Release 2023; 353:1107-1126. [PMID: 36528195 DOI: 10.1016/j.jconrel.2022.12.029] [Citation(s) in RCA: 45] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/08/2022] [Accepted: 12/10/2022] [Indexed: 12/26/2022]
Abstract
Colonic drug delivery can facilitate access to unique therapeutic targets and has the potential to enhance drug bioavailability whilst reducing off-target effects. Delivering drugs to the colon requires considered formulation development, as both oral and rectal dosage forms can encounter challenges if the colon's distinct physiological environment is not appreciated. As the therapeutic opportunities surrounding colonic drug delivery multiply, the success of novel pharmaceuticals lies in their design. This review provides a modern insight into the key parameters determining the effective design and development of colon-targeted medicines. Influential physiological features governing the release, dissolution, stability, and absorption of drugs in the colon are first discussed, followed by an overview of the most reliable colon-targeted formulation strategies. Finally, the most appropriate in vitro, in vivo, and in silico preclinical investigations are presented, with the goal of inspiring strategic development of new colon-targeted therapeutics.
Collapse
Affiliation(s)
- Laura E McCoubrey
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Alessia Favaron
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Atheer Awad
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Mine Orlu
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Simon Gaisford
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Abdul W Basit
- 29 - 39 Brunswick Square, UCL School of Pharmacy, University College London, London, WC1N 1AX, UK.
| |
Collapse
|
11
|
Ren M, Lotfipour S. Antibiotic Knockdown of Gut Bacteria Sex-Dependently Enhances Intravenous Fentanyl Self-Administration in Adult Sprague Dawley Rats. Int J Mol Sci 2022; 24:409. [PMID: 36613853 PMCID: PMC9820294 DOI: 10.3390/ijms24010409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 12/28/2022] Open
Abstract
Communication between the brain and gut bacteria impacts drug- and addiction-related behaviors. To investigate the role of gut microbiota on fentanyl reinforcement and reward, we depleted gut bacteria in adult Sprague Dawley male and female rats using an oral, nonabsorbable antibiotic cocktail and allowed rats to intravenously self-administer fentanyl on an escalating schedule of reinforcement. We found that antibiotic treatment enhanced fentanyl self-administration in males, but not females, at the lowest schedule of reinforcement (i.e., fixed ratio 1). Both males and females treated with antibiotics self-administered greater amounts of fentanyl at higher schedules of reinforcement. We then replete microbial metabolites via short-chain fatty acid administration to evaluate a potential mechanism in gut-brain communication and found that restoring metabolites decreases fentanyl self-administration back to controls at higher fixed ratio schedules of reinforcement. Our findings highlight an important relationship between the knockdown and rescue of gut bacterial metabolites and fentanyl self-administration in adult rats, which provides support for a significant relationship between the gut microbiome and opioid use. Further work in this field may lead to effective, targeted treatment interventions in opioid-related disorders.
Collapse
Affiliation(s)
- Michelle Ren
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
| | - Shahrdad Lotfipour
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of California Irvine, Irvine, CA 92697, USA
- Department of Emergency Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
12
|
Sturov NV, Popov SV, Zhukov VA, Lyapunova TV, Rusanova EI, Kobylyanu GN, Kobylyanu GN. Intestinal Microbiota Correction in the Treatment and Prevention of Urinary Tract Infection. Turk J Urol 2022; 48:406-414. [PMID: 36416330 PMCID: PMC9797784 DOI: 10.5152/tud.2022.22119] [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] [Indexed: 11/24/2022]
Abstract
Intestinal microbiota is a topical subject of modern research. The maintenance of a healthy intestinal micro biota is an important component of homeostasis, and violations of its composition and functions, called dysbiosis, are associated with a number of diseases, including urinary tract infections. Antimicrobial therapy leads to significant changes in the intestinal microbiota and causes the possibility of urinary tract infection recurrence. In this regard, it is important to study methods of microbiota correction in order to restore its structural and functional integrity.
Collapse
Affiliation(s)
- Nikolay V. Sturov
- General Medical Practice Department, RUDN University (Peoples’ Friendship University of Russia), Moscow, Russian Federation
| | - Sergey V. Popov
- General Medical Practice Department, RUDN University (Peoples’ Friendship University of Russia), Moscow, Russian Federation
| | - Vladimir A. Zhukov
- General Medical Practice Department, RUDN University (Peoples’ Friendship University of Russia), Moscow, Russian Federation,Corresponding author:Vladimir A. ZhukovE-mail:
| | - Tatiana V. Lyapunova
- Medical Informatics and Telemedicine Department, RUDN University (Peoples’ Friendship University of Russia), Moscow, Russian Federation
| | - Ekaterina I. Rusanova
- General Medical Practice Department, RUDN University (Peoples’ Friendship University of Russia), Moscow, Russian Federation
| | - Georgy N. Kobylyanu
- General Medical Practice Department, RUDN University (Peoples’ Friendship University of Russia), Moscow, Russian Federation
| | | | | | | | | | | | | |
Collapse
|
13
|
Influence of occupational exposure to pigs or chickens on human gut microbiota composition in Thailand. One Health 2022; 15:100463. [DOI: 10.1016/j.onehlt.2022.100463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 11/17/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022] Open
|
14
|
Li P, Li M, Song Y, Huang X, Wu T, Xu ZZ, Lu H. Green Banana Flour Contributes to Gut Microbiota Recovery and Improves Colonic Barrier Integrity in Mice Following Antibiotic Perturbation. Front Nutr 2022; 9:832848. [PMID: 35369097 PMCID: PMC8964434 DOI: 10.3389/fnut.2022.832848] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Green banana flour (GBF) is rich in resistant starch that has been used as a prebiotic to exert beneficial effects on gut microbiota. In this study, GBF was evaluated for its capacity to restore gut microbiota and intestinal barrier integrity from antibiotics (Abx) perturbation by comparing it to natural recovery (NR) treatment. C57B/L 6 J mice were exposed to 3 mg ciprofloxacin and 3.5 mg metronidazole once a day for 2 weeks to induce gut microbiota dysbiosis model. Then, GBF intervention at the dose of 400 mg/kg body weight was conducted for 2 weeks. The results showed that mice treated with Abx displayed increased gut permeability and intestinal barrier disruption, which were restored more quickly with GBF than NR treatment by increasing the secretion of mucin. Moreover, GBF treatment enriched beneficial Bacteroidales S24-7, Lachnospiraceae, Bacteroidaceae, and Porphyromonadaceae that accelerated the imbalanced gut microbiota restoration to its original state. This study puts forward novel insights into the application of GBF as a functional food ingredient to repair gut microbiota from Abx perturbation.
Collapse
Affiliation(s)
- Ping Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Ming Li
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Ying Song
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Xiaochang Huang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Tao Wu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Zhenjiang Zech Xu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hui Lu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| |
Collapse
|
15
|
Wang Y, Zhao X, Zang J, Li Y, Dong X, Jiang F, Wang N, Jiang L, Jiang Q, Fu C. Estimates of Dietary Exposure to Antibiotics among a Community Population in East China. Antibiotics (Basel) 2022; 11:antibiotics11030407. [PMID: 35326870 PMCID: PMC8944873 DOI: 10.3390/antibiotics11030407] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/08/2022] [Accepted: 03/16/2022] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Antibiotics are widely used in clinics, livestock farms and the aquaculture industry. A variety of antibiotics in foods and drinking water may lead to important and inadvertent dietary exposure However, the profile of dietary exposure to antibiotics in humans is not well-explored. East China is an economically developed area with a high usage of antibiotics and a high rate of antibiotic resistance (ABR). This study aimed to evaluate the total intake level of antibiotics in humans via foods and drinking water based on a community population in East China. METHODS A total of 600 local residents from 194 households were recruited into this study in Deqing County of Zhejiang Province since June 2019. Each subject was asked to fill a food frequency questionnaire to report their daily consumption of foods and drinking water. Tap water samples were collected from ten households and twenty-one antibiotics of five categories were selected to detect in drinking water. Data of antibiotic residues in animal-derived foods were obtained from the notification of unqualified edible agricultural products after special supervision sampling inspection in Deqing County. The human dietary exposure to antibiotics was estimated by combining the data of antibiotic contamination in foods and drinking water, and the information of dietary consumption. RESULTS Of twenty-one antibiotics selected, subjects were exposed to a total of sixteen antibiotics, ranging from 15.12 to 1128 μg/day via two main dietary routes (animal-derived foods and drinking water). The overall dietary exposure level varied greatly in the antibiotics detected and their sources. Compared with other antibiotics, enrofloxacin made the most contributions in terms of dietary exposure, with a median exposure level of 120.19 μg/day (IQR: 8.39-698.78 μg/day), followed by sulfamethazine (median: 32.95 μg/day, IQR: 2.77-162.55 μg/day) and oxytetracycline (median: 28.50 μg/day, IQR: 2.22-146.58 μg/day). The estimated exposure level via drinking water (at the ng/day level, median: 26.74 ng/day, IQR: 16.05-37.44 ng/day) was significantly and substantially lower than those via animal-derived foods (at the μg/day level, median: 216.38 μg/day, IQR: 87.52-323.00 μg/day). The overall dietary exposure level also showed differences in sex and age. Males and youths were more likely to be exposed to antibiotics via dietary routes than others. CONCLUSIONS The community population investigated in East China was extensively exposed to multiple antibiotics via dietary routes. Long-term exposure to low-dose antibiotics in animal-derived foods was the primary dietary exposure route, compared with drinking water. Enrofloxacin contributed to the major body burden of dietary exposure, based on the combination of consumption of aquatic products and considerable enrofloxacin residues in them. Although the human dietary exposure level to antibiotics via drinking water and animal-derived foods ranged from ng/day to μg/L, their chronic toxicity and the accumulation and spread of ABR may be potential hazards to humans. Therefore, long-term monitoring of antibiotic contaminations in foods and drinking water, and human dietary antibiotic exposure is warranted.
Collapse
Affiliation(s)
- Yingying Wang
- NHC Key Laboratory of Health Technology Assessment, School of Public Health, Fudan University, Shanghai 200032, China; (Y.W.); (X.Z.); (J.Z.); (Y.L.); (F.J.); (L.J.); (Q.J.)
| | - Xinping Zhao
- NHC Key Laboratory of Health Technology Assessment, School of Public Health, Fudan University, Shanghai 200032, China; (Y.W.); (X.Z.); (J.Z.); (Y.L.); (F.J.); (L.J.); (Q.J.)
| | - Jinxin Zang
- NHC Key Laboratory of Health Technology Assessment, School of Public Health, Fudan University, Shanghai 200032, China; (Y.W.); (X.Z.); (J.Z.); (Y.L.); (F.J.); (L.J.); (Q.J.)
| | - Yurong Li
- NHC Key Laboratory of Health Technology Assessment, School of Public Health, Fudan University, Shanghai 200032, China; (Y.W.); (X.Z.); (J.Z.); (Y.L.); (F.J.); (L.J.); (Q.J.)
| | - Xiaolian Dong
- Deqing County Center for Disease Prevention and Control, Huzhou 550004, China;
| | - Feng Jiang
- NHC Key Laboratory of Health Technology Assessment, School of Public Health, Fudan University, Shanghai 200032, China; (Y.W.); (X.Z.); (J.Z.); (Y.L.); (F.J.); (L.J.); (Q.J.)
| | - Na Wang
- NHC Key Laboratory of Health Technology Assessment, School of Public Health, Fudan University, Shanghai 200032, China; (Y.W.); (X.Z.); (J.Z.); (Y.L.); (F.J.); (L.J.); (Q.J.)
- Correspondence: (N.W.); (C.F.)
| | - Lufang Jiang
- NHC Key Laboratory of Health Technology Assessment, School of Public Health, Fudan University, Shanghai 200032, China; (Y.W.); (X.Z.); (J.Z.); (Y.L.); (F.J.); (L.J.); (Q.J.)
| | - Qingwu Jiang
- NHC Key Laboratory of Health Technology Assessment, School of Public Health, Fudan University, Shanghai 200032, China; (Y.W.); (X.Z.); (J.Z.); (Y.L.); (F.J.); (L.J.); (Q.J.)
| | - Chaowei Fu
- NHC Key Laboratory of Health Technology Assessment, School of Public Health, Fudan University, Shanghai 200032, China; (Y.W.); (X.Z.); (J.Z.); (Y.L.); (F.J.); (L.J.); (Q.J.)
- Correspondence: (N.W.); (C.F.)
| |
Collapse
|
16
|
Taxonomic Diversity of Fungi and Bacteria in Azoé-NP® Vertical Flow Constructed Wetlands. WATER 2022. [DOI: 10.3390/w14050698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Plants, fungi, bacteria and protozoa are highly interconnected in constructed wetlands. These heterogeneous groups of organisms constitute a single system with complex internal trophic interactions. Thus, the joint activity of micro- and macroorganisms in constructed wetlands provides highly efficient wastewater treatment: both nutrients and complex organic substances can be effectively removed in branched trophic chains. The bacterial community of constructed wetlands has recently received much attention, while the fungal component remains less studied, particularly saprotrophic fungi. This paper reveals a taxonomic analysis of the cultivated saprotrophic fungi combined with the bacterial community in vertical flow constructed wetlands (VSCWs) operated by the Azoé-NP® process. These systems have unique features to affect the microbial community, which results in a high treatment efficiency and nitrogen removal. There are very few studies of saprotrophic fungi in VFCWs, while this work shows their abundance and diversity in VFCWs. We found 62 species of cultivated microscopic fungi and described the taxonomic composition of bacterial and fungal community at all wastewater treatment stages. In the studied VFCWs, we identified the species of micromycetes, which proved effective in the removal of contaminants. The data obtained can provide a deeper insight into the characteristics of Azoé-NP® systems and the treatment processes occurring in constructed wetlands.
Collapse
|
17
|
Antimicrobial Stewardship Program: Reducing Antibiotic's Spectrum of Activity Is not the Solution to Limit the Emergence of Multidrug-Resistant Bacteria. Antibiotics (Basel) 2022; 11:antibiotics11010070. [PMID: 35052947 PMCID: PMC8772858 DOI: 10.3390/antibiotics11010070] [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: 11/07/2021] [Revised: 12/20/2021] [Accepted: 12/30/2021] [Indexed: 12/10/2022] Open
Abstract
Overconsumption of antibiotics in hospitals has led to policy implementation, including the control of antibiotic prescriptions. The impact of these policies on the evolution of antimicrobial resistance remains uncertain. In this work, we review the possible limits of such policies and focus on the need for a more efficient approach. Establishing a causal relationship between the introduction of new antibiotics and the emergence of new resistance mechanisms is difficult. Several studies have demonstrated that many resistance mechanisms existed before the discovery of antibiotics. Overconsumption of antibiotics has worsened the phenomenon of resistance. Antibiotics are responsible for intestinal dysbiosis, which is suspected of being the source of bacterial resistance. The complexity of the intestinal microbiota composition, the impact of the pharmacokinetic properties of antibiotics, and the multiplicity of other factors involved in the acquisition and emergence of multidrug-resistant organisms, lead us to think that de-escalation, in the absence of studies proving its effectiveness, is not the solution to limiting the spread of multidrug-resistant organisms. More studies are needed to clarify the ecological risk caused by different antibiotic classes. In the meantime, we need to concentrate our efforts on limiting antibiotic prescriptions to patients who really need it, and work on reducing the duration of these treatments.
Collapse
|
18
|
Short- and long-term effects of amoxicillin/clavulanic acid or doxycycline on the gastrointestinal microbiome of growing cats. PLoS One 2021; 16:e0253031. [PMID: 34910719 PMCID: PMC8673677 DOI: 10.1371/journal.pone.0253031] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Accepted: 10/18/2021] [Indexed: 02/07/2023] Open
Abstract
Antibiotic treatment in early life influences gastrointestinal (GI) microbial composition and function. In humans, the resultant intestinal dysbiosis is associated with an increased risk for certain diseases later in life. The objective of this study was to determine the temporal effects of antibiotic treatment on the GI microbiome of young cats. Fecal samples were collected from cats randomly allocated to receive either amoxicillin/clavulanic acid (20 mg/kg q12h) for 20 days (AMC group; 15 cats) or doxycycline (10 mg/kg q24h) for 28 days (DOX group;15 cats) as part of the standard treatment of upper respiratory tract infection. In addition, feces were collected from healthy control cats (CON group;15 cats). All cats were approximately two months of age at enrolment. Samples were collected on days 0 (baseline), 20 or 28 (AMC and DOX, respectively; last day of treatment), 60, 120, and 300. DNA was extracted and sequencing of the 16S rRNA gene and qPCR assays were performed. Fecal microbial composition was different on the last day of treatment for AMC cats, and 1 month after the end of antibiotic treatment for DOX cats, compared to CON cats. Species richness was significantly greater in DOX cats compared to CON cats on the last day of treatment. Abundance of Enterobacteriales was increased, and that of Erysipelotrichi was decreased in cats of the AMC group on the last day of treatment compared to CON cats. The abundance of the phylum Proteobacteria was increased in cats of the DOX group on days 60 and 120 compared to cats of the CON group. Only minor differences in abundances between the treatment groups and the control group were present on day 300. Both antibiotics appear to delay the developmental progression of the microbiome, and this effect is more profound during treatment with amoxicillin/clavulanic acid and one month after treatment with doxycycline. Future studies are required to determine if these changes influence microbiome function and whether they have possible effects on disease susceptibility in cats.
Collapse
|
19
|
Moor J, Wüthrich T, Aebi S, Mostacci N, Overesch G, Oppliger A, Hilty M. Influence of pig farming on human Gut Microbiota: role of airborne microbial communities. Gut Microbes 2021; 13:1-13. [PMID: 34060426 PMCID: PMC8172160 DOI: 10.1080/19490976.2021.1927634] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
It has been hypothesized that both genetics and diet influence the composition of the human cecal microbiota. However, it remains unclear whether and how occupational exposure to microbes impacts the microbial communities in human guts. Using a One Health approach, we visited pig farms (n = 26) and collected stool specimens from pig workers (n = 59), pig barn air samples (n = 19), and rectal swabs from pigs at three different growth stages (n = 144). Stool samples from cattle workers were included as a control group (n = 22). Each sample's microbiota was characterized using 16S rRNA gene sequencing and the DADA2 pipeline.We obtained a significantly different clustering of the microbial compositions of pig and cattle workers by permutational multivariate analysis of variance (PERMANOVA; P < .001). Workers primarily exposed to pigs had higher relative abundances of Prevotellaceae and less Bacteroidaceae than workers exposed to cattle. We also found that the microbial compositions of pig workers' stool samples shared extensive fractions with the samples from their pigs. We also identified amplicon sequencing variants (ASVs) in the airborne microbiota which were likely involved in zoonotic transmission events.We hypothesize that ASVs originating from pig feces are aerosolized and, through breathing, get trapped in the pig farm workers' upper respiratory tract from where they can get swallowed. Consequently, some of the animal associated ASVs are transferred into the gastrointestinal tracts (GITs) which leads to changes in the composition of the human gut microbiota. The importance of this finding for human health must be investigated further.
Collapse
Affiliation(s)
- Julia Moor
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland,Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Tsering Wüthrich
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland,Graduate School of Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Suzanne Aebi
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Nadezda Mostacci
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Gudrun Overesch
- Institute of Veterinary Bacteriology, University of Bern, Bern, Switzerland
| | - Anne Oppliger
- Unisante, Department of Occupational and Environmental Health, University of Lausanne, Lausanne, Switzerland
| | - Markus Hilty
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland,Markus Hilty Institute for Infectious Diseases, University of Bern, Friedbühlstrasse 51, 3001Bern, Switzerland, Phone +41 31 632 49 83
| |
Collapse
|
20
|
Problems associated with the use of the term "antibiotics". Naunyn Schmiedebergs Arch Pharmacol 2021; 394:2153-2166. [PMID: 34536087 PMCID: PMC8449524 DOI: 10.1007/s00210-021-02144-9] [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: 05/15/2021] [Accepted: 08/25/2021] [Indexed: 11/24/2022]
Abstract
The term “antibiotics” is a broadly used misnomer to designate antibacterial drugs. In a recent article, we have proposed to replace, e.g., the term “antibiotics” by “antibacterial drugs”, “antibiosis” by “antibacterial therapy”, “antibiogram” by “antibacteriogram”, and “antibiotic stewardship” by “antibacterial stewardship” (Seifert and Schirmer Trends Microbiol, 2021). In the present article, we show that many traditional terms related to antibiotics are used much more widely in the biomedical literature than the respective scientifically precise terms. This practice should be stopped. Moreover, we provide arguments to end the use of other broadly used terms in the biomedical literature such as “narrow-spectrum antibiotics” and “reserve antibiotics”, “chemotherapeutics”, and “tuberculostatics”. Finally, we provide several examples showing that antibacterial drugs are used for non-antibacterial indications and that some non-antibacterial drugs are used for antibacterial indications now. Thus, the increasing importance of drug repurposing renders it important to drop short designations of drug classes such as “antibiotics”. Rather, the term “drug” should be explicitly used, facilitating the inclusion of newly emerging indications such as antipsychotic and anti-inflammatory. This article is part of an effort to implement a new rational nomenclature of drug classes across the entire field of pharmacology.
Collapse
|
21
|
Wu Y, Wu J, Lin Z, Wang Q, Li Y, Wang A, Shan X, Liu J. Administration of a Probiotic Mixture Ameliorates Cisplatin-Induced Mucositis and Pica by Regulating 5-HT in Rats. J Immunol Res 2021; 2021:9321196. [PMID: 34568500 PMCID: PMC8461230 DOI: 10.1155/2021/9321196] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/18/2021] [Accepted: 08/21/2021] [Indexed: 12/24/2022] Open
Abstract
Probiotic-based therapies have been shown to be beneficial for chemotherapy-induced mucositis. Previous research has demonstrated that a probiotic mixture (Bifidobacterium brevis, Lactobacillus acidophilus, Lactobacillus casei, and Streptococcus thermophilus) can ameliorate chemotherapy-induced mucositis and dysbiosis in rats, but the underlying mechanism has not been completely elucidated. We aimed to determine the inhibitory effects of the probiotic mixture on cisplatin-induced mucositis and pica and the underlying mechanism, focusing on the levels of 5-hydroxytryptamine (5-HT, serotonin) regulated by the gut microbiota. A rat model of mucositis and pica was established by daily intraperitoneal injection of cisplatin (6 mg/kg) for 3 days. In the probiotic+cisplatin group, predaily intragastric injection of the probiotic mixture (1 × 109 CFU/kg BW) was administrated for 1 week before cisplatin injection. This was then followed by further daily probiotic injections for 6 days. Histopathology, pro-/anti-inflammatory cytokines, oxidative status, and 5-HT levels were assessed on days 3 and 6. The structure of the gut microbiota was analyzed by 16S rRNA gene sequencing and quantitative PCR. Additionally, 5-HT levels in enterochromaffin (EC) cells (RIN-14B cell line) treated with cisplatin and/or various probiotic bacteria were also determined. The probiotic mixture significantly attenuated kaolin consumption, inflammation, oxidative stress, and the increase in 5-HT concentrations in rats with cisplatin-induced intestinal mucositis and pica. Cisplatin markedly increased the relative abundances of Enterobacteriaceae_other, Blautia, Clostridiaceae_other, and members of Clostridium clusters IV and XIVa. These levels were significantly restored by the probiotic mixture. Importantly, most of the genera increased by cisplatin were significantly positively correlated with colonic 5-HT. Furthermore, in vitro, the probiotic mixture had direct inhibitory effects on the 5-HT secretion by EC cells. The probiotic mixture protects against cisplatin-induced intestine injury, exhibiting both anti-inflammatory and antiemetic properties. These results were closely related to the reestablishment of intestinal microbiota ecology and normalization of the dysbiosis-driven 5-HT overproduction.
Collapse
Affiliation(s)
- Yuanhang Wu
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jianlin Wu
- Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Zhikun Lin
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Qian Wang
- Liaoning CapitalBio Technology Co., Ltd., Dalian, China
| | - Ying Li
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Aman Wang
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Xiu Shan
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Jiwei Liu
- Department of Medical Oncology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| |
Collapse
|
22
|
Lynn MA, Eden G, Ryan FJ, Bensalem J, Wang X, Blake SJ, Choo JM, Chern YT, Sribnaia A, James J, Benson SC, Sandeman L, Xie J, Hassiotis S, Sun EW, Martin AM, Keller MD, Keating DJ, Sargeant TJ, Proud CG, Wesselingh SL, Rogers GB, Lynn DJ. The composition of the gut microbiota following early-life antibiotic exposure affects host health and longevity in later life. Cell Rep 2021; 36:109564. [PMID: 34433065 DOI: 10.1016/j.celrep.2021.109564] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 06/02/2021] [Accepted: 07/29/2021] [Indexed: 12/13/2022] Open
Abstract
Studies investigating whether there is a causative link between the gut microbiota and lifespan have largely been restricted to invertebrates or to mice with a reduced lifespan because of a genetic deficiency. We investigate the effect of early-life antibiotic exposure on otherwise healthy, normal chow-fed, wild-type mice, monitoring these mice for more than 700 days in comparison with untreated control mice. We demonstrate the emergence of two different low-diversity community types, post-antibiotic microbiota (PAM) I and PAM II, following antibiotic exposure. PAM II but not PAM I mice have impaired immunity, increased insulin resistance, and evidence of increased inflammaging in later life as well as a reduced lifespan. Our data suggest that differences in the composition of the gut microbiota following antibiotic exposure differentially affect host health and longevity in later life.
Collapse
Affiliation(s)
- Miriam A Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Georgina Eden
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Feargal J Ryan
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Julien Bensalem
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Xuemin Wang
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Stephen J Blake
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Jocelyn M Choo
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia; Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Yee Tee Chern
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Anastasia Sribnaia
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Jane James
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Saoirse C Benson
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia; Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Lauren Sandeman
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia
| | - Jianling Xie
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia
| | - Sofia Hassiotis
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Emily W Sun
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Alyce M Martin
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Marianne D Keller
- Preclinical, Imaging & Research Laboratories (PIRL), South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia
| | - Damien J Keating
- Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Timothy J Sargeant
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia
| | - Christopher G Proud
- Lifelong Health Theme, South Australian Health & Medical Research Institute, Adelaide, SA 5000, Australia; School of Biological Sciences, University of Adelaide, Adelaide, SA 5005, Australia
| | - Steve L Wesselingh
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia; Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - Geraint B Rogers
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia; Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia
| | - David J Lynn
- Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide, SA 5001, Australia; Flinders Health and Medical Research Institute, Flinders University, Bedford Park, SA 5042, Australia.
| |
Collapse
|
23
|
McCoubrey LE, Gaisford S, Orlu M, Basit AW. Predicting drug-microbiome interactions with machine learning. Biotechnol Adv 2021; 54:107797. [PMID: 34260950 DOI: 10.1016/j.biotechadv.2021.107797] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 02/07/2023]
Abstract
Pivotal work in recent years has cast light on the importance of the human microbiome in maintenance of health and physiological response to drugs. It is now clear that gastrointestinal microbiota have the metabolic power to promote, inactivate, or even toxify the efficacy of a drug to a level of clinically relevant significance. At the same time, it appears that drug intake has the propensity to alter gut microbiome composition, potentially affecting health and response to other drugs. Since the precise composition of an individual's microbiome is unique, one's drug-microbiome relationship is similarly unique. Thus, in the age of evermore personalised medicine, the ability to predict individuals' drug-microbiome interactions is highly sought. Machine learning (ML) offers a powerful toolkit capable of characterising and predicting drug-microbiota interactions at the individual patient level. ML techniques have the potential to learn the mechanisms operating drug-microbiome activities and measure patients' risk of such occurrences. This review will outline current knowledge at the drug-microbiota interface, and present ML as a technique for examining and forecasting personalised drug-microbiome interactions. When harnessed effectively, ML could alter how the pharmaceutical industry and healthcare professionals consider the drug-microbiome axis in patient care.
Collapse
Affiliation(s)
| | | | - Mine Orlu
- University College London, London, United Kingdom
| | | |
Collapse
|
24
|
Li Y, Liu H, Qi H, Tang W, Zhang C, Liu Z, Liu Y, Wei X, Kong Z, Jia S, Du B, Yuan J, Wang C, Li M. Probiotic fermentation of Ganoderma lucidum fruiting body extracts promoted its immunostimulatory activity in mice with dexamethasone-induced immunosuppression. Biomed Pharmacother 2021; 141:111909. [PMID: 34328088 DOI: 10.1016/j.biopha.2021.111909] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 02/06/2023] Open
Abstract
Ganoderma lucidum is a legendary traditional Chinese medicine with various bioactivities. This study was conducted (a) to explore the in vitro fermentation of the water extracts of G. lucidum fruiting body with Lactobacillus acidophilus and Bifidobacterium breve and (b) to investigate the effect of fermentation broth (GLFB) on dexamethasone (DEX)-induced immunosuppressed mice. Our results demonstrated that probiotic fermentation of G. lucidum fruiting body extracts underwent structural changing of major ganoderic acid components, such as ganoderic acid A (GA) into GC2, and this fermentation process involves changing of several metabolic pathways in the probiotic strains. GLFB could significantly improve the immunity, intestinal integrity, and gut microbiota dysbiosis in DEX-treated mice, and the immunostimulatory activity of GLFB was found closely related to its direct regulation on the expansion of CD4+ T cells in Peyer's patches of mice. These data implied that probiotic fermentation of G. lucidum fruiting body extracts promoted its immunostimulatory activity via biotransformation of components such as GA. This research provides a theoretical support for the development and application of G. lucidum fermentation by probiotics.
Collapse
Affiliation(s)
- Yuyuan Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - He Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Huawen Qi
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China
| | - Wei Tang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Caihua Zhang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Zhaiyi Liu
- School of Bioengineering, Dalian University of Technology, Dalian, China
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiaoqing Wei
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Zhen Kong
- Hefei Kangchuntang Pharmaceutical Co.,Ltd, Hefei, China
| | - Shangyi Jia
- Hefei Kangchuntang Pharmaceutical Co.,Ltd, Hefei, China
| | - Borong Du
- People's Hospital of Jiuquan City, Gansu, China
| | - Jieli Yuan
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Chaoran Wang
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, China.
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China.
| |
Collapse
|
25
|
McCoubrey LE, Elbadawi M, Orlu M, Gaisford S, Basit AW. Machine Learning Uncovers Adverse Drug Effects on Intestinal Bacteria. Pharmaceutics 2021; 13:1026. [PMID: 34371718 PMCID: PMC8308984 DOI: 10.3390/pharmaceutics13071026] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 06/24/2021] [Accepted: 06/30/2021] [Indexed: 02/07/2023] Open
Abstract
The human gut microbiome, composed of trillions of microorganisms, plays an essential role in human health. Many factors shape gut microbiome composition over the life span, including changes to diet, lifestyle, and medication use. Though not routinely tested during drug development, drugs can exert profound effects on the gut microbiome, potentially altering its functions and promoting disease. This study develops a machine learning (ML) model to predict whether drugs will impair the growth of 40 gut bacterial strains. Trained on over 18,600 drug-bacteria interactions, 13 distinct ML models are built and compared, including tree-based, ensemble, and artificial neural network techniques. Following hyperparameter tuning and multi-metric evaluation, a lead ML model is selected: a tuned extra trees algorithm with performances of AUROC: 0.857 (±0.014), recall: 0.587 (±0.063), precision: 0.800 (±0.053), and f1: 0.666 (±0.042). This model can be used by the pharmaceutical industry during drug development and could even be adapted for use in clinical settings.
Collapse
Affiliation(s)
| | | | | | | | - Abdul W. Basit
- UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; (L.E.M.); (M.E.); (M.O.); (S.G.)
| |
Collapse
|
26
|
Luo M, Zhou DD, Shang A, Gan RY, Li HB. Influences of food contaminants and additives on gut microbiota as well as protective effects of dietary bioactive compounds. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.05.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
27
|
Mulder M, Arp PP, Kiefte-de Jong JC, Uitterlinden AG, Klaassen CHW, Kraaij R, Goessens WHF, Verbon A, Stricker BH. Prevalence of and risk factors for extended-spectrum beta-lactamase genes carriership in a population-based cohort of middle-aged and elderly. Int J Antimicrob Agents 2021; 58:106388. [PMID: 34161788 DOI: 10.1016/j.ijantimicag.2021.106388] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 05/17/2021] [Accepted: 06/13/2021] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Increasing resistance to beta-lactam antibiotics is an alarming development worldwide. Fecal carriership of TEM, SHV, CTX-M and CMY was studied in a community-dwelling population of middle-aged and elderly individuals. PATIENTS AND METHODS Feces was obtained from individuals of the Rotterdam Study. Carriership of the TEM, SHV, CTX-M and CMY genes was determined using real-time polymerase chain reaction (qPCR). Possible associations were investigated between carriership of these genes and several risk factors, such as the use of antimicrobial drugs, diabetes mellitus, protein pump inhibitor (PPI) use, travelling, the composition of the gut microbiota, and intake of certain foods. RESULTS The most prevalent gene was TEM (53.0%), followed by SHV (18.4%), CTX-M (5.4%) and CMY (3.6%). Use of penicillins with extended spectrum was associated with TEM carriership, whereas use of macrolides and lincosamides was associated with TEM and SHV carriership. Interestingly, use of PPIs was associated with a higher prevalence of carriership of TEM, SHV and CMY (TEM: odds ratio [OR] 1.34; 95% confidence interval [CI] 1.05-1.77; SHV: OR 2.17; 95%CI 1.55-2.87; CMY: OR 2.26; 95%CI 1.23-4.11). Furthermore, associations were found between the richness and composition of the gut microbiota and TEM and SHV carriership. CONCLUSIONS The prevalence of carriership of TEM was substantial, but the prevalence of carriership of the extended-spectrum β-lactamase gene, CTX-M and the AmpC β-lactamase gene, CMY was relatively low in this community-dwelling, population-based cohort. The composition of the microbiota might play a role in the retention of resistance genes, but future studies are necessary to further elucidate this relationship.
Collapse
Affiliation(s)
- M Mulder
- Department of Epidemiology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands; Department of Medical Microbiology, Maastricht University Medical Center, PO Box 5800, 6202 AZ Maastricht, The Netherlands
| | - P P Arp
- Department of Internal Medicine, Erasmus MC University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - J C Kiefte-de Jong
- Department of Epidemiology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands; Department of Public Health and Primary Care/LUMC Campus, The Hague, Leiden University Medical Center, The Netherlands
| | - A G Uitterlinden
- Department of Internal Medicine, Erasmus MC University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - C H W Klaassen
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - R Kraaij
- Department of Internal Medicine, Erasmus MC University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - W H F Goessens
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - A Verbon
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands
| | - B H Stricker
- Department of Epidemiology, Erasmus MC University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands; Department of Internal Medicine, Erasmus MC University Medical Center, PO Box 2040, 3000 CA Rotterdam, The Netherlands; Inspectorate of Health Care, PO Box 2518, 6401 DA Heerlen, The Netherlands.
| |
Collapse
|
28
|
Li Y, Liu M, Liu H, Sui X, Liu Y, Wei X, Liu C, Cheng Y, Ye W, Gao B, Wang X, Lu Q, Cheng H, Zhang L, Yuan J, Li M. The Anti-Inflammatory Effect and Mucosal Barrier Protection of Clostridium butyricum RH2 in Ceftriaxone-Induced Intestinal Dysbacteriosis. Front Cell Infect Microbiol 2021; 11:647048. [PMID: 33842393 PMCID: PMC8027357 DOI: 10.3389/fcimb.2021.647048] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 02/26/2021] [Indexed: 12/12/2022] Open
Abstract
This study aimed at determining the beneficial effect of Clostridium butyricum (CB) RH2 on ceftriaxone-induced dysbacteriosis. To this purpose, BALB/c mice were exposed to ceftriaxone (400 mg/ml) or not (control) for 7 days, and administered a daily oral gavage of low-, and high-dose CB RH2 (108 and 1010 CFU/ml, respectively) for 2 weeks. CB RH2 altered the diversity of gut microbiota, changed the composition of gut microbiota in phylum and genus level, decreased the F/B ratio, and decreased the pro-inflammatory bacteria (Deferribacteres, Oscillibacter, Desulfovibrio, Mucispirillum and Parabacteroides) in ceftriaxone-treated mice. Additionally, CB RH2 improved colonic architecture and intestinal integrity by improving the mucous layer and the tight junction barrier. Furthermore, CB RH2 also mitigated intestinal inflammation through decreasing proinflammatory factors (TNF-α and COX-2) and increasing anti-inflammatory factors (IL-10). CB RH2 had direct effects on the expansion of CD4+ T cells in Peyer’s patches (PPs) in vitro, which in turn affected their immune response upon challenge with ceftriaxone. All these data suggested that CB RH2 possessed the ability to modulate the intestinal mucosal and systemic immune system in limiting intestinal alterations to relieve ceftriaxone-induced dysbacteriosis.
Collapse
Affiliation(s)
- Yuyuan Li
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Man Liu
- College of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - He Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xue Sui
- Advanced Institute for Medical Sciences, Dalian Medical University, Dalian, China
| | - Yinhui Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiaoqing Wei
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Chunzheng Liu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Yiqin Cheng
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Weikang Ye
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Binbin Gao
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xin Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Qiao Lu
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Hao Cheng
- Marketing Department, Hangzhou Grand Biologic Pharmaceutical Inc., Hangzhou, China
| | - Lu Zhang
- Marketing Department, Hangzhou Grand Biologic Pharmaceutical Inc., Hangzhou, China
| | - Jieli Yuan
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| |
Collapse
|