1
|
Smibert OC, Vogrin S, Sinclair M, Majumdar A, Nasra M, Pandey D, Jahanabadi H, Trubiano JA, Markey KA, Slavin MA, Testro A, Kwong JC. Antibiotic Exposure and Risk of Allograft Rejection and Survival After Liver Transplant: An Observational Cohort Study From a Tertiary Referral Centre. Transpl Infect Dis 2025:e70026. [PMID: 40153422 DOI: 10.1111/tid.70026] [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: 02/02/2025] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/30/2025]
Abstract
INTRODUCTION Our goal is to understand whether there is an association between Abx exposure-and the inferred downstream damage to the intestinal microbiome-and the key patient outcomes of overall survival and rejection following liver transplant. METHODS We conducted a retrospective cohort study of 462 liver transplant recipients treated at a multistate liver transplant (LTx) service during a 7-year period. The association between antibiotic exposure and outcome was tested across models that addressed antibiotic spectrum, duration, and timing relative to transplant. Cox proportional hazard regression was used to evaluate the relationship between antibiotics with survival and rejection. RESULTS The observed 1-year survival in this cohort was 95% (95% CI: 93%, 97%), and 20.8% of patients (96/462) experienced rejection at 1 year. In multivariable analyses, exposure to anaerobe-targeting antibiotics for longer than 14 days pretransplant (p = 0.055) or posttransplant (p = 0.040) was significantly associated with reduced 1-year survival. In multivariable analyses, exposure to any anaerobe-targeting Abx posttransplant was significantly associated with an increased risk of rejection (p = 0.001). CONCLUSIONS Exposure to anaerobic spectrum antibiotics either before or after LTx was associated with poor outcomes during the first year posttransplant and provides an impetus to further characterize the relationship between antibiotic use, microbiota disruption, and cellular immunity in liver transplantation.
Collapse
Affiliation(s)
- Olivia C Smibert
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Infectious Diseases and Immunology, Austin Health, Melbourne, Victoria, Australia
- Department of Infectious Diseases, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sara Vogrin
- Department of Medicine, St Vincent's Hospital, Victoria Parade, University of Melbourne, Fitzroy, Australia
| | - Marie Sinclair
- Liver Transplant Unit, Austin Health, Melbourne, Victoria, Australia
- School of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Victoria, Australia
| | - Avik Majumdar
- Liver Transplant Unit, Austin Health, Melbourne, Victoria, Australia
- School of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Victoria, Australia
| | - Mohamed Nasra
- Department of Infectious Diseases and Immunology, Austin Health, Melbourne, Victoria, Australia
| | - Dinesh Pandey
- Data Analytics Research and Evaluation (DARE) Centre, The University of Melbourne and Austin Hospital, Melbourne, Victoria, Australia
- Clinical Analytics and Reporting, Performance Reporting and Decision Support, Austin Health, Melbourne, Victoria, Australia
| | - Hossein Jahanabadi
- Data Analytics Research and Evaluation (DARE) Centre, The University of Melbourne and Austin Hospital, Melbourne, Victoria, Australia
- Clinical Analytics and Reporting, Performance Reporting and Decision Support, Austin Health, Melbourne, Victoria, Australia
| | - Jason A Trubiano
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Infectious Diseases and Immunology, Austin Health, Melbourne, Victoria, Australia
- Department of Infectious Diseases, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Kate A Markey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center (FHCC), Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Monica A Slavin
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Infectious Diseases, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Adam Testro
- Liver Transplant Unit, Austin Health, Melbourne, Victoria, Australia
- School of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Victoria, Australia
| | - Jason C Kwong
- Department of Infectious Diseases and Immunology, Austin Health, Melbourne, Victoria, Australia
- Department of Microbiology & Immunology, The Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
2
|
Chen Q, Huang X, Zhang H, Jiang X, Zeng X, Li W, Su H, Chen Y, Lin F, Li M, Gu X, Jin H, Wang R, Diao D, Wang W, Li J, Wei S, Zhang W, Liu W, Huang Z, Deng Y, Luo W, Liu Z, Zhang B. Characterization of tongue coating microbiome from patients with colorectal cancer. J Oral Microbiol 2024; 16:2344278. [PMID: 38686186 PMCID: PMC11057396 DOI: 10.1080/20002297.2024.2344278] [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: 09/29/2023] [Accepted: 04/13/2024] [Indexed: 05/02/2024] Open
Abstract
Background Tongue coating microbiota has aroused particular interest in profiling oral and digestive system cancers. However, little is known on the relationship between tongue coating microbiome and colorectal cancer (CRC). Methods Metagenomic shotgun sequencing was performed on tongue coating samples collected from 30 patients with CRC, 30 patients with colorectal polyps (CP), and 30 healthy controls (HC). We further validated the potential of the tongue coating microbiota to predict the CRC by a random forest model. Results We found a greater species diversity in CRC samples, and the nucleoside and nucleotide biosynthesis pathway was more apparent in the CRC group. Importantly, various species across participants jointly shaped three distinguishable fur types.The tongue coating microbiome profiling data gave an area under the receiver operating characteristic curve (AUC) of 0.915 in discriminating CRC patients from control participants; species such as Atopobium rimae, Streptococcus sanguinis, and Prevotella oris aided differentiation of CRC patients from healthy participants. Conclusion These results elucidate the use of tongue coating microbiome in CRC patients firstly, and the fur-types observed contribute to a better understanding of the microbial community in human. Furthermore, the tongue coating microbiota-based biomarkers provide a valuable reference for CRC prediction and diagnosis.
Collapse
Affiliation(s)
- Qubo Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Xiaoting Huang
- Medical Research Center, Huazhong University of Science and Technology Union Shenzhen, Shenzhen, China
| | - Haiyan Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Xuanting Jiang
- Department of Scientific Research, KMHD, Shenzhen, China
| | - Xuan Zeng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Wanhua Li
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Hairong Su
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ying Chen
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Fengye Lin
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Man Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
| | - Xiangyu Gu
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Huihui Jin
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Ruohan Wang
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Dechang Diao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Colorectal surgery of Guangdong Provincial Hospital of TCM, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Wei Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Gastrointestinal Surgery Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jin Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Gastrointestinal Surgery Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Sufen Wei
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Weizheng Zhang
- Medical Laboratory, Guangzhou Cadre Health Management Center, Guangzhou No.11 People’s Hospital, Guangzhou, China
| | - Wofeng Liu
- Medical Laboratory, Guangzhou Cadre Health Management Center, Guangzhou No.11 People’s Hospital, Guangzhou, China
| | - Zhiping Huang
- Information Department, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yusheng Deng
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Biological Resource Center, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou, China
- Department of Scientific Research, KMHD, Shenzhen, China
| | - Wen Luo
- Department of Scientific Research, KMHD, Shenzhen, China
| | - Zuofeng Liu
- Department of Scientific Research, KMHD, Shenzhen, China
| | - Beiping Zhang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, Department of Gastroenterology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
3
|
Zhang Y, Xue G, Wang F, Zhang J, Xu L, Yu C. The impact of antibiotic exposure on antibiotic resistance gene dynamics in the gut microbiota of inflammatory bowel disease patients. Front Microbiol 2024; 15:1382332. [PMID: 38694799 PMCID: PMC11061493 DOI: 10.3389/fmicb.2024.1382332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/26/2024] [Indexed: 05/04/2024] Open
Abstract
Background While antibiotics are commonly used to treat inflammatory bowel disease (IBD), their widespread application can disturb the gut microbiota and foster the emergence and spread of antibiotic resistance. However, the dynamic changes to the human gut microbiota and direction of resistance gene transmission under antibiotic effects have not been clearly elucidated. Methods Based on the Human Microbiome Project, a total of 90 fecal samples were collected from 30 IBD patients before, during and after antibiotic treatment. Through the analysis workflow of metagenomics, we described the dynamic process of changes in bacterial communities and resistance genes pre-treatment, during and post-treatment. We explored potential consistent relationships between gut microbiota and resistance genes, and established gene transmission networks among species before and after antibiotic use. Results Exposure to antibiotics can induce alterations in the composition of the gut microbiota in IBD patients, particularly a reduction in probiotics, which gradually recovers to a new steady state after cessation of antibiotics. Network analyses revealed intra-phylum transfers of resistance genes, predominantly between taxonomically close organisms. Specific resistance genes showed increased prevalence and inter-species mobility after antibiotic cessation. Conclusion This study demonstrates that antibiotics shape the gut resistome through selective enrichment and promotion of horizontal gene transfer. The findings provide insights into ecological processes governing resistance gene dynamics and dissemination upon antibiotic perturbation of the microbiota. Optimizing antibiotic usage may help limit unintended consequences like increased resistance in gut bacteria during IBD management.
Collapse
Affiliation(s)
- Yufei Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Gaogao Xue
- Beijing Hotgen Biotech Co., Ltd., Beijing, China
| | - Fan Wang
- Beijing YuGen Pharmaceutical Co., Ltd., Beijing, China
| | - Jing Zhang
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| | - Lida Xu
- Beijing Hotgen Biotech Co., Ltd., Beijing, China
- Beijing YuGen Pharmaceutical Co., Ltd., Beijing, China
| | - Changyuan Yu
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, China
| |
Collapse
|
4
|
Baindara P, Mandal SM. Gut-Antimicrobial Peptides: Synergistic Co-Evolution with Antibiotics to Combat Multi-Antibiotic Resistance. Antibiotics (Basel) 2023; 12:1732. [PMID: 38136766 PMCID: PMC10740742 DOI: 10.3390/antibiotics12121732] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 12/11/2023] [Accepted: 12/13/2023] [Indexed: 12/24/2023] Open
Abstract
Due to huge diversity and dynamic competition, the human gut microbiome produces a diverse array of antimicrobial peptides (AMPs) that play an important role in human health. The gut microbiome has an important role in maintaining gut homeostasis by the AMPs and by interacting with other human organs via established connections such as the gut-lung, and gut-brain axis. Additionally, gut AMPs play a synergistic role with other gut microbiota and antimicrobials to maintain gut homeostasis by fighting against multi-antibiotic resistance (MAR) bacteria. Further, conventional antibiotics intake creates a synergistic evolutionary pressure for gut AMPs, where antibiotics and gut AMPs fight synergistically against MAR. Overall, gut AMPs are evolving under a complex and highly synergistic co-evolutionary pressure created by the various interactions between gut microbiota, gut AMPs, and antibiotics; however, the complete mechanism is not well understood. The current review explores the synergistic action of gut AMPs and antibiotics along with possibilities to fight against MAR bacteria.
Collapse
Affiliation(s)
- Piyush Baindara
- Radiation Oncology, NextGen Precision Health, School of Medicine, University of Missouri, Columbia, MO 65211, USA
| | - Santi M. Mandal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India;
| |
Collapse
|
5
|
Erb T, Mihai S, Strauß R, Herbst L, Castellanos I, Diesch K, Cipa F, Bihlmaier K, Lang AK, Ganslmayer M, Willam C, Bremer F, Fürst J, Beyer C, Bogdan C, Rath A, Held J. β-(1→3)-D-glucan- and mannan-guided early termination of antifungal therapy in ICU patients: a randomized controlled study. Antimicrob Agents Chemother 2023; 67:e0072523. [PMID: 37823695 PMCID: PMC10648872 DOI: 10.1128/aac.00725-23] [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: 06/01/2023] [Accepted: 08/20/2023] [Indexed: 10/13/2023] Open
Abstract
Candida spp. are frequently encountered in specimens from ICUs. However, most of these detections represent colonization. Nevertheless, clinical practice shows that a considerable proportion of these patients will receive antifungal therapy (AT). β-(1→3)-D-glucan (BDG) and mannan are fungal biomarkers with high negative predictive values. We aimed to examine whether biomarker-guided discontinuation of AT can reduce the antifungal consumption. Therefore, we conducted a prospective, randomized intervention study between 1 April 2019 and 31 March 2020. All adult ICU patients with a newly started systemic AT but without fungal infection were eligible for inclusion. Enrolled patients were randomized into an intervention and a control group. In both groups, serum BDG and mannan were determined on days 1 and 2 of AT. If all measurements were negative, AT was discontinued in the intervention group. The primary endpoint was antifungal use. The study was terminated after 12 months. Until this time-point, 41 patients had been included. In the intervention group (n = 19), AT was stopped in only two patients because all others showed either positive BDG and/or mannan levels. One of these two patients developed candidemia and AT had to be restarted. There was no significant difference in the primary and secondary endpoints. In summary, the strategy of using two negative BDG and mannan levels to stop AT failed to reduce antifungal consumption in our cohort. Indeed, there will inevitably be patients with invasive candidiasis in whom necessary AT is discontinued. The optimal patient population, biomarker set, and termination criteria are critical to the success of biomarker-based termination strategies.
Collapse
Affiliation(s)
- Timothy Erb
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene; Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Sidonia Mihai
- Zentrallabor, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Richard Strauß
- Medizinische Klinik 1, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Larissa Herbst
- Medizinische Klinik 4, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Ixchel Castellanos
- Anästhesiologische Klinik, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Katharina Diesch
- Medizinisches Zentrum für Informations- und Kommunikationstechnik, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Franziska Cipa
- Zentrallabor, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Karl Bihlmaier
- Medizinische Klinik 4, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Anne-Katharina Lang
- Anästhesiologische Klinik, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Marion Ganslmayer
- Medizinische Klinik 1, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Carsten Willam
- Medizinische Klinik 4, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Frank Bremer
- Anästhesiologische Klinik, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Fürst
- Medizinische Klinik 1, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Beyer
- Medizinische Klinik 1, Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Christian Bogdan
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene; Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Anca Rath
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene; Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| | - Jürgen Held
- Mikrobiologisches Institut - Klinische Mikrobiologie, Immunologie und Hygiene; Universitätsklinikum Erlangen und Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
6
|
Girma A. The effect of novel antimicrobial agents on the normal functioning of human intestinal microbiota: a systematic review. FRONTIERS IN GASTROENTEROLOGY 2023; 2. [DOI: 10.3389/fgstr.2023.1159352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Abstract
Antimicrobial agents have significant effects on the ecological balance of the human microbiota through incomplete absorption (e.g., orally administered antimicrobial agents) or secretion (e.g., by the salivary glands, in the bile, or from the intestinal mucosa) of the agents. This study aimed to examine the effects of novel antimicrobial agents on the normal functioning of the intestinal microbiota. The articles, written in English, were recovered from PubMed, ScienceDirect, Web of Science, Google Scholar, and DOAJ, as well as from manual searches using a reference list. “Microbiota”, “Intestinal Microbiota”, “Eubiotic Microbiota”, “Ecological Impact”, “Antimicrobial Agents,”, “Antibiotics”, “Dysbiosis”, “Gut Microbiota”, and “Probiotics” were the search terms used to retrieve the articles. The PRISMA 2009 checklist was applied for article search strategy, article selection, data extraction, and result reporting for the review process. A total of eight original research articles were included from a total of 379 articles obtained in different search strategies. The eight new antimicrobial agents demonstrated significant impacts on the ecological balance of the human intestinal microbiota. Therefore, eubiosis is crucial in preventing the establishment of exogenous antimicrobial-resistant strains as well as their gene transfer.Systematic review registration[PRISMA], identifier [2009].
Collapse
|
7
|
Ashraf M, Ahammad SZ, Chakma S. Advancements in the dominion of fate and transport of pharmaceuticals and personal care products in the environment-a bibliometric study. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:64313-64341. [PMID: 37067715 PMCID: PMC10108824 DOI: 10.1007/s11356-023-26796-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Accepted: 03/30/2023] [Indexed: 05/11/2023]
Abstract
The study on the fate and transport of Pharmaceuticals and Personal Care Products, PPCPs (FTP) in the environment, has received particular attention for over two decades. The PPCPs threaten ecology and human health even at low concentrations due to their synergistic effects and long-range transport. The research aims to provide an inclusive map of the scientific background of FTP research over the last 25 years, from 1996 to 2020, to identify the main characteristics, evolution, salient research themes, trends, and research hotspots in the field of interest. Bibliometric networks were synthesized and analyzed for 577 journal articles extracted from the Scopus database. Consequently, seven major themes of FTP research were identified as follows: (i) PPCPs category; (ii) hazardous effects; (iii) occurrence of PPCPs; (iv) PPCPs in organisms; (v) remediation; (vi) FTP-governing processes; and (vii) assessment in the environment. The themes gave an in-depth picture of the sources of PPCPs and their transport and fate processes in the environment, which originated from sewage treatment plants and transported further to sediment/soils/groundwater/oceans that act as the PPCPs' major sink. The article provided a rigorous analysis of the research landscape in the FTP study conducted during the specified years. The prominent research themes, content analysis, and research hotspots identified in the study may serve as the basis of real-time guidance to lead future research areas and a prior review for policymakers and practitioners.
Collapse
Affiliation(s)
- Maliha Ashraf
- School of Interdisciplinary Research, Indian Institute of Technology, Delhi, India.
| | - Shaikh Ziauddin Ahammad
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi, India
| | - Sumedha Chakma
- Department of Civil Engineering, Indian Institute of Technology, Delhi, India
| |
Collapse
|
8
|
Shams M, Hamdy E, Abd-elsadek D. Are multiple courses of antibiotics a potential risk factor for COVID-19 infection and severity? ONE HEALTH BULLETIN 2023; 3:10. [DOI: 10.4103/2773-0344.378589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
|
9
|
Yoon S, Lee G, Yu J, Lee K, Lee K, Si J, You HJ, Ko G. Distinct Changes in Microbiota-Mediated Intestinal Metabolites and Immune Responses Induced by Different Antibiotics. Antibiotics (Basel) 2022; 11:antibiotics11121762. [PMID: 36551419 PMCID: PMC9774394 DOI: 10.3390/antibiotics11121762] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/28/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The cocktails of antibiotics are utilized to study the functions of microbiota. There have been studies on the alteration of not only the microbiota composition but also the host's metabolism or immunity. However, the bacterial species associated with these altered physiologic markers are still unclear. Therefore, we supplied mice with drinking water containing ampicillin (AMP), vancomycin (VAN), neomycin (NEO), or metronidazole (MET) to observe the effect of each antibiotic on helper T cells and inflammation-related gene expression and metabolism, including amino acid metabolism and changes in gut microbiota. We observed major changes in gut microbiota in mice treated with AMP and VAN, respectively, immediately after administration. The abundance of the genera Parabacteroides and Akkermansia increased in the AMP and VAN groups, while Prevotella almost disappeared from both groups. The compositional changes in intestinal metabolites in the AMP and VAN groups were more distinct than those in the NEO and MET groups, which was similar to the microbiome results. In particular, the most distinct changes were observed in amino acid related metabolism in AMP and VAN groups; the amounts of phenylalanine and tyrosine were increased in the AMP group while those were decreased in the VAN group. The changed amounts of intestinal amino acids in each of the AMP and VAN groups were correlated with increases in the abundance of the genera Parabacteroides and Akkermansia in the AMP and VAN groups, respectively. The most distinctive changes in intestinal gene expression were observed in the ileum, especially the expression Th17-related genes such as rorgt, il17a, and il17f, which decreased dramatically in the guts of most of the antibiotic-treated groups. These changes were also associated with a significant decrease in Prevotella in both the AMP and VAN groups. Taken together, these findings indicate that changes in gut microbiota as well as host physiology, including host metabolism and immunity, differ depending on the types of antibiotics, and the antibiotic-induced gut microbiota alteration has a correlation with host physiology such as host metabolic or immunological status. Thus, the immune and metabolic status of the host should be taken into account when administering antibiotics.
Collapse
Affiliation(s)
- Sunghyun Yoon
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Giljae Lee
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- Bio-MAX/N-Bio, Seoul National University, Seoul 08826, Republic of Korea
| | - Junsun Yu
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Kiuk Lee
- KoBioLabs, Inc., Seoul 13488, Republic of Korea
| | - Kyeongju Lee
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
| | - Jiyeon Si
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
- Natural Products Research Center, Korea Institute of Science and Technology (KIST), Gangneung 25451, Republic of Korea
- Center for Human and Environmental Microbiome, Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
| | - Hyun Ju You
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- KoBioLabs, Inc., Seoul 13488, Republic of Korea
- Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
- Center for Human and Environmental Microbiome, Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
- Correspondence: (H.J.Y.); (G.K.)
| | - GwangPyo Ko
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul 08826, Republic of Korea
- Bio-MAX/N-Bio, Seoul National University, Seoul 08826, Republic of Korea
- KoBioLabs, Inc., Seoul 13488, Republic of Korea
- Center for Human and Environmental Microbiome, Institute of Health and Environment, Seoul National University, Seoul 08826, Republic of Korea
- Correspondence: (H.J.Y.); (G.K.)
| |
Collapse
|
10
|
Lima O, Sousa A, Filgueira A, González-Novoa MC, Domínguez-López C, Ávila-Nuñez M, Represa M, Rubiñán P, Martínez-Lamas L, Pérez-Castro S, Rubianes M, Pérez-Rodríguez MT. Gastrointestinal colonization by OXA-48-producing Enterobacterales: risk factors for persistent carriage. Eur J Clin Microbiol Infect Dis 2022; 41:1399-1405. [PMID: 36205803 DOI: 10.1007/s10096-022-04504-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 09/28/2022] [Indexed: 11/25/2022]
Abstract
Carbapenem-resistant Enterobacterales (CRE) infections are a major health problem. Intestinal colonization is a key factor in developing infection. However, factors associated with persistent colonization by CRE are unknown. The aim of the study was to identify factors associated with persistent CRE gut colonization. This is a retrospective, single-centre, observational study of adult patients with CRE gut colonization between January 2015 and January 2020. Epidemiologic characteristics, comorbidities, infectious events, duration of hospitalization and antimicrobial treatment received in the follow-up period were collected. Colonization was defined as isolation in at least 2 rectal swab culture samples of CRE. Decolonization was defined as 3 negative rectal swab cultures or 2 negative cultures and a negative molecular test. A cohort of 86 patients with CRE gut colonization was selected: 44 patients with spontaneous decolonization (DC) and 42 patients with persistent colonization (PC). The mean follow-up period was 24 months (IQR 14-33) in the DC group vs. 25 months (IQR 16-36) in the PC group (p = 0.478). Patient characteristics were similar between both groups. Colonization by other MDR microorganisms was high (44 patients, 51%) and slightly more common in the PC group (PC 60% vs. DC 43%, p = 0.139). The use of ceftazidime-avibactam was more common among the PC group (PC 33% vs. DC 14%, p = 0.041). We observed a higher percentage of antimicrobial therapy in the previous 30 days (PC 68% vs. DC 57%, p = 0.371) and 90 days (PC 81% vs. DC 82%, p = 0.353) in the PC group. Multivariable analysis showed that patients that have received ceftazidime-avibactam therapy (OR 4.9 95% CI [1.45-16.39], p = 0.010), and those colonized by other MDR microorganisms (OR 2.5, 95% CI [0.96-6.25], p = 0.060) presented a higher risk of PC. Ceftazidime-avibactam use and colonization by other MDR microorganisms might be associated with CRE persistent gut colonization.
Collapse
Affiliation(s)
- O Lima
- Infectious Diseases Unit, Internal Medicine, Complexo Hospitalario Universitario de Vigo, Estrada Clara Campoamor, 341 Pontevedra, 36213, Vigo, Spain.
- Biomedical Research Institute Galicia Sur, Vigo, Spain.
| | - A Sousa
- Infectious Diseases Unit, Internal Medicine, Complexo Hospitalario Universitario de Vigo, Estrada Clara Campoamor, 341 Pontevedra, 36213, Vigo, Spain
- Biomedical Research Institute Galicia Sur, Vigo, Spain
| | - A Filgueira
- Vascular Surgery Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain
| | | | | | - M Ávila-Nuñez
- Infectious Diseases Unit, Internal Medicine, Complexo Hospitalario Universitario de Vigo, Estrada Clara Campoamor, 341 Pontevedra, 36213, Vigo, Spain
| | - M Represa
- Infectious Diseases Unit, Internal Medicine, Complexo Hospitalario Universitario de Vigo, Estrada Clara Campoamor, 341 Pontevedra, 36213, Vigo, Spain
| | - P Rubiñán
- Infectious Diseases Unit, Internal Medicine, Complexo Hospitalario Universitario de Vigo, Estrada Clara Campoamor, 341 Pontevedra, 36213, Vigo, Spain
| | - L Martínez-Lamas
- Microbiology Department, Complexo Hospitalario Universitario de Vigo, Vigo, Spain
| | - Sonia Pérez-Castro
- Service of Preventive Medicine, University Hospital Complex of Vigo, Vigo, Spain
| | - M Rubianes
- Infectious Diseases Unit, Internal Medicine, Complexo Hospitalario Universitario de Vigo, Estrada Clara Campoamor, 341 Pontevedra, 36213, Vigo, Spain
| | - M T Pérez-Rodríguez
- Infectious Diseases Unit, Internal Medicine, Complexo Hospitalario Universitario de Vigo, Estrada Clara Campoamor, 341 Pontevedra, 36213, Vigo, Spain
- Biomedical Research Institute Galicia Sur, Vigo, Spain
| |
Collapse
|
11
|
Hobson CA, Vigue L, Naimi S, Chassaing B, Magnan M, Bonacorsi S, Gachet B, El Meouche I, Birgy A, Tenaillon O. MiniBioReactor Array (MBRA) in vitro gut model: a reliable system to study microbiota-dependent response to antibiotic treatment. JAC Antimicrob Resist 2022; 4:dlac077. [PMID: 35795241 PMCID: PMC9252984 DOI: 10.1093/jacamr/dlac077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 06/14/2022] [Indexed: 11/18/2022] Open
Abstract
Background Antimicrobial drugs are mostly studied for their impact on emergence of bacterial antibiotic resistance, but their impact on the gut microbiota is also of tremendous interest. In vitro gut models are important tools to study such complex drug–microbiota interactions in humans. Methods The MiniBioReactor Array (MBRA) in vitro microbiota system; a single-stage continuous flow culture model, hosted in an anaerobic chamber; was used to evaluate the impact of three concentrations of a third-generation cephalosporin (ceftriaxone) on faecal microbiota from two healthy donors (treatment versus control: three replicates per condition). We conducted 16S microbiome profiling and analysed microbial richness, diversity and taxonomic changes. β-Lactamase activities were evaluated and correlated with the effects observed in the MBRA in vitro system. Results The MBRA preserved each donor’s specificities, and differences between the donors were maintained through time. Before treatment, all faecal cultures belonging to the same donor were comparable in composition, richness, and diversity. Treatment with ceftriaxone was associated with a decrease in α-diversity, and an increase in β-diversity index, in a concentration-dependent manner. The maximum effect on diversity was observed after 72 h of treatment. Importantly, one donor had a stronger microbiota β-lactamase activity that was associated with a reduced impact of ceftriaxone on microbiota composition. Conclusions MBRA can reliably mimic the intestinal microbiota and its modifications under antibiotic selective pressure. The impact of the treatment was donor- and concentration-dependent. We hypothesize these results could be explained, at least in part, by the differences in β-lactamase activity of the microbiota itself. Our results support the relevance and promise of the MBRA system to study drug–microbiota interactions.
Collapse
Affiliation(s)
- C A Hobson
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| | - L Vigue
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| | - S Naimi
- INSERM U1016, Team ‘Mucosal Microbiota in Chronic Inflammatory diseases’, CNRS UMR 8104, Université de Paris , Paris , France
| | - B Chassaing
- INSERM U1016, Team ‘Mucosal Microbiota in Chronic Inflammatory diseases’, CNRS UMR 8104, Université de Paris , Paris , France
| | - M Magnan
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| | - S Bonacorsi
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
- Laboratoire de Microbiologie, Hôpital Robert Debré, AP-HP , 75019 Paris , France
| | - B Gachet
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| | - I El Meouche
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| | - A Birgy
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
- Laboratoire de Microbiologie, Hôpital Robert Debré, AP-HP , 75019 Paris , France
| | - O Tenaillon
- IAME, UMR 1137, INSERM, Université de Paris, AP-HP , Paris , France
| |
Collapse
|
12
|
Holkem AT, Silva MPD, Favaro-Trindade CS. Probiotics and plant extracts: a promising synergy and delivery systems. Crit Rev Food Sci Nutr 2022; 63:9561-9579. [PMID: 35445611 DOI: 10.1080/10408398.2022.2066623] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
There is a current interest in healthy diets and supplements, indicating the relevance of novel delivery systems for plant extracts rich in bioactive compounds and probiotics. This simultaneous delivery system can be prospective for health. In this sense, investigating foods rich in bioactive compounds or supplemented by them for incorporating probiotics and some approaches to improve probiotic survivability, such as the choice of resistant probiotic strains or microencapsulation, is valuable. This review addresses a brief discussion about the role of phenolic compounds, chlorophyll and carotenoids from plants and probiotics in gut health, indicating the benefits of this association. Also, an overview of delivery systems used in recent studies is shown, considering their advantages for incorporation in food matrices. Delivery systems containing compounds recovered from plants can reduce probiotic oxidative stress, improving survivability. However, investigating the beneficial concentration of some bioactive compounds from plant extracts is relevant due to their antimicrobial potential. In addition, further clinical trials and toxicological studies of plant extracts are pertinent to ensure safety. Thus, the recovery of extracts from plants emerges as an alternative to providing multiple compounds with antioxidant potential, increasing the preservation of probiotics and allowing the fortification or enrichment of food matrices.
Collapse
Affiliation(s)
- Augusto Tasch Holkem
- Department of Biomedical Sciences, Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, QC, Canada
| | - Marluci Palazzolli da Silva
- Department of Food Engineering, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| | - Carmen Silvia Favaro-Trindade
- Department of Food Engineering, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| |
Collapse
|
13
|
Bassetti M, Vena A, Giacobbe DR, Trucchi C, Ansaldi F, Antonelli M, Adamkova V, Alicino C, Almyroudi MP, Atchade E, Azzini AM, Brugnaro P, Carannante N, Peghin M, Berruti M, Carnelutti A, Castaldo N, Corcione S, Cortegiani A, Dimopoulos G, Dubler S, García-Garmendia JL, Girardis M, Cornely OA, Ianniruberto S, Kullberg BJ, Lagrou K, Lebihan C, Luzzati R, Malbrain M, Merelli M, Marques AJ, Martin-Loeches I, Mesini A, Paiva JA, Raineri SM, Rautemaa-Richardson R, Schouten J, Spapen H, Tasioudis P, Timsit JF, Tisa V, Tumbarello M, Van den Berg CHSB, Veber B, Venditti M, Voiriot G, Wauters J, Zappella N, Montravers P. Risk Factors for Intra-Abdominal Candidiasis in Intensive Care Units: Results from EUCANDICU Study. Infect Dis Ther 2022; 11:827-840. [PMID: 35182353 PMCID: PMC8960530 DOI: 10.1007/s40121-021-00585-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 12/14/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Intra-abdominal infections represent the second most frequently acquired infection in the intensive care unit (ICU), with mortality rates ranging from 20% to 50%. Candida spp. may be responsible for up to 10-30% of cases. This study assesses risk factors for development of intra-abdominal candidiasis (IAC) among patients admitted to ICU. METHODS We performed a case-control study in 26 European ICUs during the period January 2015-December 2016. Patients at least 18 years old who developed an episode of microbiologically documented IAC during their stay in the ICU (at least 48 h after admission) served as the case cohort. The control group consisted of adult patients who did not develop episodes of IAC during ICU admission. Matching was performed at a ratio of 1:1 according to time at risk (i.e. controls had to have at least the same length of ICU stay as their matched cases prior to IAC onset), ICU ward and period of study. RESULTS During the study period, 101 case patients with a diagnosis of IAC were included in the study. On univariate analysis, severe hepatic failure, prior receipt of antibiotics, prior receipt of parenteral nutrition, abdominal drain, prior bacterial infection, anastomotic leakage, recurrent gastrointestinal perforation, prior receipt of antifungal drugs and higher median number of abdominal surgical interventions were associated with IAC development. On multivariate analysis, recurrent gastrointestinal perforation (OR 13.90; 95% CI 2.65-72.82, p = 0.002), anastomotic leakage (OR 6.61; 95% CI 1.98-21.99, p = 0.002), abdominal drain (OR 6.58; 95% CI 1.73-25.06, p = 0.006), prior receipt of antifungal drugs (OR 4.26; 95% CI 1.04-17.46, p = 0.04) or antibiotics (OR 3.78; 95% CI 1.32-10.52, p = 0.01) were independently associated with IAC. CONCLUSIONS Gastrointestinal perforation, anastomotic leakage, abdominal drain and prior receipt of antifungals or antibiotics may help to identify critically ill patients with higher probability of developing IAC. Prospective studies are needed to identify which patients will benefit from early antifungal treatment.
Collapse
Affiliation(s)
- Matteo Bassetti
- Clinica Malattie Infettive. Ospedale Policlinico San Martino - IRCCS, L.go R. Benzi 10, 16132, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Antonio Vena
- Clinica Malattie Infettive. Ospedale Policlinico San Martino - IRCCS, L.go R. Benzi 10, 16132, Genoa, Italy.
- Department of Health Sciences, University of Genoa, Genoa, Italy.
| | - Daniele R Giacobbe
- Clinica Malattie Infettive. Ospedale Policlinico San Martino - IRCCS, L.go R. Benzi 10, 16132, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Cecilia Trucchi
- A.Li.Sa. Liguria Health Authority, Genoa, Italy
- Healthcare Planning Unit, Ospedale Policlinico San Martino - IRCCS, Genoa, Italy
| | - Filippo Ansaldi
- A.Li.Sa. Liguria Health Authority, Genoa, Italy
- Healthcare Planning Unit, Ospedale Policlinico San Martino - IRCCS, Genoa, Italy
| | - Massimo Antonelli
- Department of Intensive Care Anesthesiology and Emercency Medicine, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
- Istituto di Anestesiologia e Rianimazione, Università Cattolica del Sacro Cuore, Milan, Italy
| | - Vaclava Adamkova
- Clinical Microbiology and ATB Centre, Institute of Medical Biochemistry and Laboratory Diagnostics, General University Hospital, Prague, Czech Republic
- Department of Medical Microbiology, Medical Faculty of Palackeho University, Olomouc, Czech Republic
| | - Cristiano Alicino
- Medical Direction, Santa Corona Hospital, ASL 2 Regional Health System of Liguria, Pietra Ligure, Italy
| | | | - Enora Atchade
- Département d'Anesthésie-Réanimation, CHU Bichat-Claude Bernard, HUPNVS, APHP, Paris, France
| | - Anna M Azzini
- Department of Diagnostics and Public Health, Infectious Disease Unit, University of Verona, Verona, Italy
| | | | - Novella Carannante
- First Division of Infectious Diseases, Cotugno Hospital, AORN dei Colli, Naples, Italy
| | - Maddalena Peghin
- Infectious Diseases Clinic, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Marco Berruti
- Clinica Malattie Infettive. Ospedale Policlinico San Martino - IRCCS, L.go R. Benzi 10, 16132, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Alessia Carnelutti
- Infectious Diseases Clinic, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Nadia Castaldo
- Infectious Diseases Clinic, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Silvia Corcione
- Department of Medical Sciences, Infectious Diseases, University of Turin, Turin, Italy
| | - Andrea Cortegiani
- Department of Surgical, Oncological and Oral Science (Di.Chir.On.S.), University of Palermo, Palermo, Italy
- Department of Anesthesia, Intensive Care and Emergency, Policlinico Paolo Giaccone, Palermo, Italy
| | - George Dimopoulos
- Department of Critical Care, University Hospital Attikon, Attikon Medical School, Νational and Kapodistrian University of Athens, Athens, Greece
| | - Simon Dubler
- Department of Anesthesiology and Intensive Care Medicine, Heidelberg University Hospital, Heidelberg, Germany
| | - José L García-Garmendia
- Servicio de Cuidados Críticos y Urgencias, Hospital San Juan de Dios del Aljarafe, Bormujos, Seville, Spain
| | - Massimo Girardis
- Department of Anesthesia and Intensive Care, University Hospital of Modena, Modena, Italy
| | - Oliver A Cornely
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department I of Internal Medicine, Excellence Center for Medical Mycology (ECMM), Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Chair Translational Research, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), Cologne, Germany
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Clinical Trials Centre Cologne (ZKS Köln), Cologne, Germany
- German Centre for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Stefano Ianniruberto
- Infectious Diseases Unit, Department of Medical and Surgical Science, S. Orsola-Malpighi Hospital, University of Bologna, Bologna, Italy
| | - Bart Jan Kullberg
- Radboud Umc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Katrien Lagrou
- Department of Laboratory Medicine and National Reference Centre for Mycosis, University Hospitals of Leuven, Leuven, Belgium
- Department of Microbiology and Immunology and Transplantation, KU Leuven, Leuven, Belgium
| | - Clement Lebihan
- APHP; Medical and Infectious Diseases ICU (MI2), Bichat Hospital, 75018, Paris, France
| | - Roberto Luzzati
- Infectious Diseases Department, Azienda Sanitaria Universitaria Integrata Di Trieste, Trieste, Italy
| | - Manu Malbrain
- Department of Intensive Care Medicine, University Hospital Brussels (UZB), 1090, Jette, Belgium
- Faculty of Medicine and Pharmacy, Vrije Unversiteit Brussel (VUB), 1090, Brussels, Belgium
| | - Maria Merelli
- Infectious Diseases Clinic, Santa Maria della Misericordia University Hospital of Udine, ASUFC, Udine, Italy
| | - Ana J Marques
- C.H. Vila Nova de Gaia/Espinho, Vila Nova de Gaia, Portugal
| | - Ignacio Martin-Loeches
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St. James's Hospital, Dublin, Ireland
- Centro de Investigación Biomedica En Red-Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
- Pneumology Department, Respiratory Institute, Hospital Clinic of Barcelona - Institut d'Investigacions Biomèdiques August Pi I Sunyer - University of Barcelona, Barcelona, Spain
| | - Alessio Mesini
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - José-Artur Paiva
- Department of Emergency and Intensive Care Medicine, Centro Hospitalar Universitário São João, Faculdade de Medicina da Universidade Do Porto E Grupo de Infecção E Sépsis, Porto, Portugal
| | - Santi Maurizio Raineri
- Department of Surgical, Oncological and Oral Science (Di.Chir.On.S.), University of Palermo, Palermo, Italy
- Institute for Biomedical Research and Innovation (IRIB) - National Research Council (CNR), Palermo, Italy
| | - Riina Rautemaa-Richardson
- Faculty of Biology, Medicine and Health, Division of Evolution, Infection and Genomics, University of Manchester, Manchester, UK
- Department of Infectious Diseases, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Manchester, UK
| | - Jeroen Schouten
- Radboud Umc Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Herbert Spapen
- Universitair Ziekenhuis Brussel, VUB University, Brussels, Belgium
| | | | - Jean-François Timsit
- Université Paris Diderot/Hopital Bichat-Réanimation Medicale et Des Maladies Infectieuses, Paris, France
- UMR 1137-IAME Team 5-DeSCID: Decision Sciences in Infectious Diseases, Control and Care, Inserm/Univ Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Valentino Tisa
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Mario Tumbarello
- Institute of Infectious Diseases, Fondazione Policlinico Universitario A. Gemelli IRCCS - Università Cattolica del Sacro Cuore, Rome, Italy
| | | | - Benoit Veber
- Pole Anesthésie-Réanimation-SAMU, Rouen University Hospital, Rouen, France
| | - Mario Venditti
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Guillaume Voiriot
- Service de Réanimation Et USC Médico-Chirurgicale, AP-HP, Hôpitaux Universitaires de L'Est Parisien, Pôle TVAR, Hôpital Tenon, Paris, France
| | - Joost Wauters
- Department of General Internal Medicine, Medical Intensive Care Unit, University Hospitals Leuven, Leuven, UK
| | | | | |
Collapse
|
14
|
Okeahialam NA, Thakar R, Sultan AH. Healing of disrupted perineal wounds after vaginal delivery: a poorly understood condition. BRITISH JOURNAL OF NURSING (MARK ALLEN PUBLISHING) 2021; 30:S8-S16. [PMID: 34781764 DOI: 10.12968/bjon.2021.30.sup20.s8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Perineal injury following childbirth can result in complications such as wound infection and dehiscence. The reported incidence of these complications in the literature range between 0.1-23.6% and 0.2-24.6%, respectively. However, the healing of disrupted perineal wounds is poorly understood. In addition, it is a neglected area in maternity services. In this review, the authors explore the process of wound healing in the context of infected perineal wounds following childbirth. In addition, the authors describe the management of complications including hypergranulation, perineal pain and dyspareunia.
Collapse
Affiliation(s)
| | - Ranee Thakar
- Consultant Obstetrician and Urogynaecologist, Croydon University Hospital, London, and Honorary Senior Lecturer, St George's University of London
| | - Abdul H Sultan
- Consultant Obstetrician and Urogynaecologist, Croydon University Hospital, London, and Honorary Reader, St George's University of London
| |
Collapse
|
15
|
Huang R, Cai GQ, Li J, Li XS, Liu HT, Shang XL, Zhou JD, Nie XM, Gui R. Platelet membrane-camouflaged silver metal-organic framework drug system against infections caused by methicillin-resistant Staphylococcus aureus. J Nanobiotechnology 2021; 19:229. [PMID: 34348721 PMCID: PMC8336064 DOI: 10.1186/s12951-021-00978-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Accepted: 07/27/2021] [Indexed: 12/30/2022] Open
Abstract
Background Due to the intelligent survival strategy and self-preservation of methicillin-resistant Staphylococcus aureus (MRSA), many antibiotics are ineffective in treating MRSA infections. Nano-drug delivery systems have emerged as a new method to overcome this barrier. The aim of this study was to construct a novel nano-drug delivery system for the treatment of MRSA infection, and to evaluate the therapeutic effect and biotoxicity of this system. We prepared a nano silver metal-organic framework using 2-methylimidazole as ligand and silver nitrate as ion provider. Vancomycin (Vanc) was loaded with Ag-MOF, and nano-sized platelet vesicles were prepared to encapsulate Ag-MOF-Vanc, thus forming the novel platelet membrane-camouflaged nanoparticles PLT@Ag-MOF-Vanc. Results The synthesized Ag-MOF particles had uniform size and shape of radiating corona. The mean nanoparticle size and zeta potential of PLT@Ag-MOF-Vanc were 148 nm and − 25.6 mV, respectively. The encapsulation efficiency (EE) and loading efficiency (LE) of vancomycin were 81.0 and 64.7 %, respectively. PLT@Ag-MOF-Vanc was shown to be a pH-responsive nano-drug delivery system with good biocompatibility. Ag-MOF had a good inhibitory effect on the growth of three common clinical strains (Escherichia coli, Pseudomonas aeruginosa, and S. aureus). PLT@Ag-MOF-Vanc showed better antibacterial activity against common clinical strains in vitro than free vancomycin. PLT@Ag-MOF-Vanc killed MRSA through multiple approaches, including interfering with the metabolism of bacteria, catalyzing reactive oxygen species production, destroying the integrity of cell membrane, and inhibiting biofilm formation. Due to the encapsulation of the platelet membrane, PLT@Ag-MOF-Vanc can bind to the surface of the MRSA bacteria and the sites of MRSA infection. PLT@Ag-MOF-Vanc had a good anti-infective effect in mouse MRSA pneumonia model, which was significantly superior to free vancomycin, and has no obvious toxicity. Conclusions PLT@Ag-MOF-Vanc is a novel effective targeted drug delivery system, which is expected to be used safely in anti-infective therapy of MRSA. Graphic abstract ![]()
Collapse
Affiliation(s)
- Rong Huang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Hunan, Changsha, China.,Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Hunan, Changsha, China
| | - Guang-Qing Cai
- Department of Orthopedics, Changsha Hospital of Traditional Chinese Medicine, Changsha Eighth Hospital, Hunan, Changsha, China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Hunan, Changsha, China
| | - Xi-Sheng Li
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Hunan, Changsha, China
| | - Hai-Ting Liu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Hunan, Changsha, China
| | - Xue-Ling Shang
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Hunan, Changsha, China
| | - Jian-Dang Zhou
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Hunan, Changsha, China
| | - Xin-Min Nie
- Department of Laboratory Medicine, The Third Xiangya Hospital, Central South University, Hunan, Changsha, China.
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Hunan, Changsha, China.
| |
Collapse
|
16
|
Living fabrication of functional semi-interpenetrating polymeric materials. Nat Commun 2021; 12:3422. [PMID: 34103521 PMCID: PMC8187375 DOI: 10.1038/s41467-021-23812-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 05/11/2021] [Indexed: 01/02/2023] Open
Abstract
Cell-mediated living fabrication has great promise for generating materials with versatile, programmable functions. Here, we demonstrate the engineering of living materials consisting of semi-interpenetrating polymer networks (sIPN). The fabrication process is driven by the engineered bacteria encapsulated in a polymeric microcapsule, which serves as the initial scaffold. The bacteria grow and undergo programmed lysis in a density-dependent manner, releasing protein monomers decorated with reactive tags. Those protein monomers polymerize with each other to form the second polymeric component that is interlaced with the initial crosslinked polymeric scaffold. The formation of sIPN serves the dual purposes of enhancing the mechanical property of the living materials and anchoring effector proteins for diverse applications. The material is resilient to perturbations because of the continual assembly of the protein mesh from the monomers released by the engineered bacteria. We demonstrate the adoption of the platform to protect gut microbiota in animals from antibiotic-mediated perturbations. Our work lays the foundation for programming functional living materials for diverse applications.
Collapse
|
17
|
Guo M, Zhang C, Zhang C, Zhang X, Wu Y. Lacticaseibacillus rhamnosus Reduces the Pathogenicity of Escherichia coli in Chickens. Front Microbiol 2021; 12:664604. [PMID: 34140939 PMCID: PMC8203825 DOI: 10.3389/fmicb.2021.664604] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/23/2021] [Indexed: 11/13/2022] Open
Abstract
Lacticaseibacillus rhamnosus is a recognized probiotic that is widely used in scientific research and clinical applications. This study found that the Lacticaseibacillus rhamnosus GG (LGG) strain can reduce the adhesion of Escherichia coli (E. coli) to primary chicken intestinal epithelial cells by 75.7% and inhibit 41.7% of the E. coli that adhere to intestinal epithelial cells. Additionally, LGG showed strong inhibitory ability on the growth of E. coli, Staphylococcus aureus, Salmonella Paratyphi B, and Salmonella Enteritidis in vitro. Furthermore, the influence of LGG on the growth performance, intestinal flora, immunity, and disease resistance of chickens was explored. Chickens fed with LGG exhibited increased average daily weight gain and concentrations of sIgA, IgG, and IgM than did controls. After 21 days of feeding, a diet with LGG increased the diversity of intestinal microbiota and maintained intestinal health. Moreover, LGG promoted immunologic barriers by upregulating cytokines and chemokines via the Toll-like receptor. The major pro-inflammatory factors, including Myd88, NF-κB, Il6, and Il8, were upregulated compared to controls. After being challenged with E. coli, the survival rate of chickens fed with LGG was significantly higher than those in the control group, and decreased numbers of E. coli were detected in the heart and lungs of the LGG group. In summary, oral administration of LGG to chickens could improve growth performance, maintain intestinal homeostasis, and enhance innate immune response and disease resistance.
Collapse
Affiliation(s)
- Mengjiao Guo
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Congyue Zhang
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Chengcheng Zhang
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Xiaorong Zhang
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China
| | - Yantao Wu
- Jiangsu Co-Innovation Center for Prevention of Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou, China.,Joint International Research Laboratory of Agriculture and Agri-Product Safety, Yangzhou University (JIRLAAPS), Yangzhou, China
| |
Collapse
|
18
|
Shvetsov YB, Ogino MH, Glibetic N, Asato CB, Wilkens LR, Le Marchand L, Matter ML. Association of Sepsis Mortality with Specific Cancer Sites and Treatment Type: The Multiethnic Cohort Study. J Pers Med 2021; 11:jpm11020146. [PMID: 33669565 PMCID: PMC7922684 DOI: 10.3390/jpm11020146] [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: 12/30/2020] [Revised: 02/12/2021] [Accepted: 02/16/2021] [Indexed: 12/05/2022] Open
Abstract
Sepsis is a severe dysregulated immune response to infection. Sepsis deaths represent 9% of cancer deaths in the U.S. Evidence of the effect of specific cancer sites on sepsis mortality risk remains limited, and no research has evaluated the effect of cancer treatment on the risk of sepsis death. We examined whether cancer sites and treatments differentially affect the risk of sepsis death compared to other-cause mortality, among the 94,784 Hawaii participants in the Multiethnic Cohort, including 29,255 cancer cases, using competing risk Cox proportional hazards regression. Cancer diagnosis at any site was associated with similar increases in sepsis and non-sepsis mortality risk (HR: 3.39 and 3.51, resp.). Colorectal cancer differentially affected the risk of sepsis and non-sepsis mortality with a 40% higher effect on the risk of sepsis death compared with non-sepsis mortality (RRR: 1.40; 95% CI: 1.14–1.72). Lung cancer was associated with a significantly lower increase in sepsis compared to non-sepsis mortality (HR: 1.22 and 3.0, resp.; RRR: 0.39). Radiation therapy had no effect on sepsis mortality but was associated with higher risk of non-sepsis mortality (HR: 0.90 and 1.16, resp.; RRR: 0.76), whereas chemotherapy was associated with higher risk of both sepsis and non-sepsis mortality (HR: 1.31 and 1.21, resp.). We conclude that the risk of sepsis-related mortality is differentially affected by cancer sites and treatments. These associations were consistent across sexes and ethnic groups.
Collapse
|
19
|
Tranberg A, Samuelsson C, Klarin B. Disturbance in the oropharyngeal microbiota in relation to antibiotic and proton pump inhibitor medication and length of hospital stay. APMIS 2021; 129:14-22. [PMID: 32981186 PMCID: PMC7756849 DOI: 10.1111/apm.13087] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 09/21/2020] [Indexed: 12/30/2022]
Abstract
The aim of this study was to investigate the appearance of a disturbed oropharyngeal microbiota during hospitalization and explore the patient characteristics that maybe associated with such a disturbance. Oropharyngeal swabs were collected from 134 patients at hospital admission and every 3-4 days thereafter. The samples were cultivated to determine the presence of a disturbed microbiota, which, in turn, was subcategorized into respiratory tract pathogens, gut microbiota and yeast species. Demographics, medical history data and hospitalization events were compared. The percentage of disturbed oropharyngeal microbiota increased significantly with length of stay (LOS). Receiving antibiotic treatment during the hospitalization tended to be associated with a disturbed microbiota (OR 2.75 [0.99-7.60]). Proton pump inhibitor (PPI) medication and receiving antibiotics before hospitalization were associated with the development of a disturbed oropharyngeal microbiota with colonization of gut pathogens (OR 3.49 [1.19-10.2] and OR 4.52 [1.13-18.1], respectively), while acute hospital admission was associated with a lower risk of colonization of gut pathogens (OR: 0.23 [0.074-0.72]). The risk of developing a disturbed oropharyngeal microbiota increased with LOS in hospitalized patients. PPI medication and receiving antibiotics before hospitalization were independent risk factors for developing oropharyngeal colonization of gut pathogens.
Collapse
Affiliation(s)
- Anna Tranberg
- Division of Intensive and Perioperative CareSkane University HospitalLundSweden
| | - Carolina Samuelsson
- Division of Intensive and Perioperative CareSkane University HospitalLundSweden
| | - Bengt Klarin
- Division of Intensive and Perioperative CareSkane University HospitalLundSweden
| |
Collapse
|
20
|
Vanamala K, Tatiparti K, Bhise K, Sau S, Scheetz MH, Rybak MJ, Andes D, Iyer AK. Novel approaches for the treatment of methicillin-resistant Staphylococcus aureus: Using nanoparticles to overcome multidrug resistance. Drug Discov Today 2021; 26:31-43. [PMID: 33091564 PMCID: PMC7855522 DOI: 10.1016/j.drudis.2020.10.011] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 09/15/2020] [Accepted: 10/14/2020] [Indexed: 02/07/2023]
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) causes serious infections in both community and hospital settings, with high mortality rates. Treatment of MRSA infections is challenging because of the rapidly evolving resistance mechanisms combined with the protective biofilms of S. aureus. Together, these characteristic resistance mechanisms continue to render conventional treatment modalities ineffective. The use of nanoformulations with unique modes of transport across bacterial membranes could be a useful strategy for disease-specific delivery. In this review, we summarize treatment approaches for MRSA, including novel techniques in nanoparticulate designing for better therapeutic outcomes; and facilitate an understanding that nanoparticulate delivery systems could be a robust approach in the successful treatment of MRSA.
Collapse
Affiliation(s)
- Kushal Vanamala
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Katyayani Tatiparti
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Ketki Bhise
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Samaresh Sau
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA
| | - Marc H Scheetz
- Departments of Pharmacy Practice and Pharmacology, Midwestern University Chicago College of Pharmacy and Graduate Studies, Pharmacometric Center of Excellence, Chicago, IL, USA
| | - Michael J Rybak
- Anti-Infective Research Laboratory, Department of Pharmacy Practice, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA; Division of Infectious Diseases, Department of Medicine, School of Medicine, Wayne State University, Detroit, MI, USA
| | - David Andes
- Division of Infectious Disease, Department of Medicine, School of Medicine and Public Health, University of Wisconsin, Madison, WI, USA
| | - Arun K Iyer
- Use-Inspired Biomaterials and Integrated Nano Delivery Systems Laboratory, Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI, USA; Molecular Imaging Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
21
|
Chen Z, Guo J, Jiang Y, Shao Y. High concentration and high dose of disinfectants and antibiotics used during the COVID-19 pandemic threaten human health. ENVIRONMENTAL SCIENCES EUROPE 2021; 33:11. [PMID: 33532166 PMCID: PMC7844811 DOI: 10.1186/s12302-021-00456-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 01/21/2021] [Indexed: 05/04/2023]
Abstract
The issue of coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has created enormous threat to global health. In an effort to contain the spread of COVID-19, a huge amount of disinfectants and antibiotics have been utilized on public health. Accordingly, the concentration of disinfectants and antibiotics is increasing rapidly in various environments, including wastewater, surface waters, soils and sediments. The aims of this study were to analyze the potential ecological environment impacts of disinfectants and antibiotics by summarizing their utilization, environmental occurrence, distribution and toxicity. The paper highlights the promoting effects of disinfectants and antibiotics on antibiotic resistance genes (ARGs) and even antibiotic resistant bacteria (ARB). The scientific evidences indicate that the high concentration and high dose of disinfectants and antibiotics promote the evolution toward antimicrobial resistance through horizontal gene transformation and vertical gene transformation, which threaten human health. Further concerns should be focused more on the enrichment, bioaccumulation and biomagnification of disinfectants, antibiotics, antibiotic resistance genes (ARGs) and even antibiotic resistant bacteria (ARB) in human bodies.
Collapse
Affiliation(s)
- Zhongli Chen
- Key Laboratory of the Three Gorges Reservoir Eco-Environment, Chongqing University, Chongqing, 400045 People’s Republic of China
| | - Jinsong Guo
- Key Laboratory of the Three Gorges Reservoir Eco-Environment, Chongqing University, Chongqing, 400045 People’s Republic of China
| | - Yanxue Jiang
- Key Laboratory of the Three Gorges Reservoir Eco-Environment, Chongqing University, Chongqing, 400045 People’s Republic of China
| | - Ying Shao
- Key Laboratory of the Three Gorges Reservoir Eco-Environment, Chongqing University, Chongqing, 400045 People’s Republic of China
| |
Collapse
|
22
|
Alagawany M, Elnesr SS, Farag MR, Tiwari R, Yatoo MI, Karthik K, Michalak I, Dhama K. Nutritional significance of amino acids, vitamins and minerals as nutraceuticals in poultry production and health - a comprehensive review. Vet Q 2020; 41:1-29. [PMID: 33250002 PMCID: PMC7755404 DOI: 10.1080/01652176.2020.1857887] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 11/23/2020] [Accepted: 11/26/2020] [Indexed: 02/08/2023] Open
Abstract
Nutraceuticals have gained immense importance in poultry science recently considering the nutritional and beneficial health effects of their constituents. Besides providing nutritional requirements to birds, nutraceuticals have beneficial pharmacological effects, for example, they help in establishing normal physiological health status, prevent diseases and thereby improve production performance. Nutraceuticals include amino acids, vitamins, minerals, enzymes, etc. which are important for preventing oxidative stress, regulating the immune response and maintaining normal physiological, biochemical and homeostatic mechanisms. Nutraceuticals help in supplying nutrients in balanced amounts for supporting the optimal growth performance in modern poultry flocks, and as a dietary supplement can reduce the use of antibiotics. The application of antibiotic growth enhancers in poultry leads to the propagation of antibiotic-resistant microbes and drug residues; therefore, they have been restricted in many countries. Thus, there is a demand for natural feed additives that lead to the same growth enhancement without affecting the health. Nutraceuticals substances have an essential role in the development of the animals' normal physiological functions and in protecting them against infectious diseases. In this review, the uses of amino acids, vitamins and minerals as well as their mode of action in growth promotion and elevation of immune system are discussed.
Collapse
Affiliation(s)
- Mahmoud Alagawany
- Faculty of Agriculture, Department of Poultry, Zagazig University, Zagazig, Egypt
| | - Shaaban S. Elnesr
- Faculty of Agriculture, Department of Poultry Production, Fayoum University, Fayoum, Egypt
| | - Mayada R. Farag
- Faculty of Veterinary Medicine, Forensic Medicine and Toxicology Department, Zagazig University, Zagazig, Egypt
| | - Ruchi Tiwari
- Department of Veterinary Microbiology and Immunology, College of Veterinary Sciences, Deen Dayal Upadhayay Pashu Chikitsa Vigyan Vishwavidyalay Evum Go-Anusandhan Sansthan (DUVASU), Mathura, India
| | - Mohd. Iqbal Yatoo
- Sher-E-Kashmir University of Agricultural Sciences and Technology of Kashmir, Srinagar, India
| | - Kumaragurubaran Karthik
- Central University Laboratory, Tamil Nadu Veterinary and Animal Sciences University, Chennai, India
| | - Izabela Michalak
- Faculty of Chemistry, Department of Advanced Material Technologies, Wrocław University of Science and Technology, Wrocław, Poland
| | - Kuldeep Dhama
- Division of Pathology, ICAR-Indian Veterinary Research Institute, Bareilly, India
| |
Collapse
|
23
|
Bioactive Healing Abutment as a Potential Tool for the Treatment of Peri-Implant Disease—In Vitro Study. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10155376] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The common use of dental implants for dental reconstruction poses new treatment challenges for physicians, peri-implantitis being a particularly difficult one. Micro-organisms, including drug-resistant Staphylococcus spp. strains, play a crucial role in the etiology of peri-implantitis. In this paper, the authors assess the efficacy of a bioactive healing abutment (BHA) of their own design for the local release of antibiotics as a potential tool for the treatment of peri-implant disease. BHA filled with a collagen material, combined with the antibiotics clindamycin and tetracycline, was tested in vitro by disk diffusion assay. Antibacterial activity was observed for the chosen Staphylococcus aureus and Staphylococcus epidermidis bacterial strains. In addition, the impact of titanium discs (which were used to make the BHA) was monitored. The results show that the zone diameter breakpoints for BHA are higher than those of standard absorbent paper discs in both tested strains for both tested antibiotics. This work demonstrates that the proposed BHA can serve as an effective and precise drug carrier. The release of antibiotics from the described implant device is easy to control and allows for an effective local antibacterial in vitro treatment. The procedure is inexpensive, easy to perform, and repeatable.
Collapse
|
24
|
Bhalodi AA, van Engelen TSR, Virk HS, Wiersinga WJ. Impact of antimicrobial therapy on the gut microbiome. J Antimicrob Chemother 2020; 74:i6-i15. [PMID: 30690540 PMCID: PMC6382031 DOI: 10.1093/jac/dky530] [Citation(s) in RCA: 137] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The gut microbiome is now considered an organ unto itself and plays an important role in health maintenance and recovery from critical illness. The commensal organisms responsible for the framework of the gut microbiome are valuable in protection against disease and various physiological tasks. Critical illness and the associated interventions have a detrimental impact on the microbiome. While antimicrobials are one of the fundamental and often life-saving modalities in septic patients, they can also pave the way for subsequent harm because of the resulting damage to the gut microbiome. Contributing to many of the non-specific signs and symptoms of sepsis, the balance between the overuse of antimicrobials and the clinical need in these situations is often difficult to delineate. Given the potency of antimicrobials utilized to treat septic patients, the effects on the gut microbiome are often rapid and long-lasting, in which case full recovery may never be observed. The overgrowth of opportunistic pathogens is of significant concern as they can lead to infections that become increasingly difficult to treat. Continued research to understand the disturbances within the gut microbiome of critically ill patients and their outcomes is essential to help develop future therapies to circumvent damage to, or restore, the microbiome. In this review, we discuss the impact of the antimicrobials often used for the treatment of sepsis on the gut microbiota.
Collapse
Affiliation(s)
- Amira A Bhalodi
- Accelerate Diagnostics, Inc., Scientific Affairs, Tucson, AZ, USA
| | - Tjitske S R van Engelen
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands
| | - Harjeet S Virk
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands
| | - W Joost Wiersinga
- Amsterdam UMC, University of Amsterdam, Center for Experimental and Molecular Medicine, Amsterdam, The Netherlands.,Amsterdam UMC, University of Amsterdam, Department of Medicine, Division of Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
25
|
Ma Z, Kang M, Meng S, Tong Z, Yoon SD, Jang Y, Jeong KC. Selective Killing of Shiga Toxin-Producing Escherichia coli with Antibody-Conjugated Chitosan Nanoparticles in the Gastrointestinal Tract. ACS APPLIED MATERIALS & INTERFACES 2020; 12:18332-18341. [PMID: 32239905 DOI: 10.1021/acsami.0c02177] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Shiga toxin-producing Escherichia coli (STEC) are critical foodborne pathogens, which cause serious human health issues, including hemolytic uremic syndrome. Illnesses caused by STEC lack effective treatments that target the elimination of these bacteria from the gastrointestinal tract without causing an adverse effect. Reducing this pathogen from a reservoir of STEC is an effective strategy, but the challenges remain due to the lack of efficient, selective antimicrobial agents. We developed specific antibody-conjugated chitosan nanoparticles (CNs) to selectively target and treat STEC in the gastrointestinal tract. Given the great broad-spectrum antimicrobial activity of CN, we conjugated antibodies to CN. Antibodies were raised and purified from egg yolks after immunization of hens with seven different O-side-chain antigens isolated from STEC (O26, O45, O103, O111, O121, O145, and O157). We prepared CN-immunoglobulin Y (IgY) conjugates by forming amide bonds at different ratios of CN:IgY (10:1, 10:2, and 10:4). The CN-IgY conjugated at a 10:2 ratio demonstrated significantly enhanced antimicrobial activity against E. coli O157:H7. Conjugates of CN and anti-STEC IgY antibodies killed corresponding STEC serotypes specifically and selectively, while showing no significant impact on nontargeted bacteria, including Salmonella enterica and Lactobacillus plantarum. The enhanced antimicrobial activity of CN-IgY against STEC was also confirmed in synthetic intestinal fluid, as well as an in vivo animal model of Caenorhabditis elegans. These results suggest that the CN-IgY conjugates have strong and specific antimicrobial activity and that they are also great candidates to eliminate pathogens selectively in the gastrointestinal tract without inhibiting beneficial bacteria.
Collapse
Affiliation(s)
- Zhengxin Ma
- Emerging Pathogens Institute, University of Florida, 2055 Mowry Road, Gainesville, Florida 32611, United States
- Department of Animal Sciences, University of Florida, Gainesville, Florida 32611, United States
| | - Minyoung Kang
- Emerging Pathogens Institute, University of Florida, 2055 Mowry Road, Gainesville, Florida 32611, United States
- Department of Animal Sciences, University of Florida, Gainesville, Florida 32611, United States
| | - Shanyu Meng
- Department of Agricultural and Biological Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Zhaohui Tong
- Department of Agricultural and Biological Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Soon-Do Yoon
- Department of Chemical and Biomolecular Engineering, Chonnam National University, Yeosu, Jeonnam 59626, Republic of Korea
| | - Yeongseon Jang
- Department of Chemical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - K Casey Jeong
- Emerging Pathogens Institute, University of Florida, 2055 Mowry Road, Gainesville, Florida 32611, United States
- Department of Animal Sciences, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
26
|
Liu Z, Mahale P, Engels EA. Sepsis and Risk of Cancer Among Elderly Adults in the United States. Clin Infect Dis 2020; 68:717-724. [PMID: 29982318 DOI: 10.1093/cid/ciy530] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 06/27/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Sepsis is an important cause of mortality among older adults in the United States. The association between sepsis and subsequent risk of cancer is poorly understood. METHODS Using the Surveillance, Epidemiology, and End Results (SEER)-Medicare linked database, we conducted a case-control study in US adults. We included 1801156 cases with a first cancer diagnosis in SEER during 1992-2013 (ages 66-115 years) and 200000 cancer-free controls from a 5% random sample of Medicare beneficiaries. Sepsis was identified using inpatient Medicare claims. Associations with sepsis were estimated using logistic regression. RESULTS After correction for multiple comparisons, sepsis was significantly associated with increased risk for cancers of the colon (adjusted odds ratio [aOR] = 1.12), rectum (1.13), liver (1.47), lung (1.17), and cervix (1.52), as well as acute myeloid leukemia (AML, 1.19), chronic myeloid leukemia (1.54), and myelodysplastic syndrome (1.30). Inverse associations were observed for cancers of the breast (aOR = 0.86), prostate (0.75), kidney (0.90), and thyroid (0.68) and for melanoma (0.83), diffuse large B-cell lymphoma (0.89), and follicular lymphoma (0.65). Sepsis was significantly associated with the following 9 types of cancer in the period >5 years following sepsis diagnosis: thyroid, prostate, colon, rectum, lung, and liver and follicular lymphoma, melanoma, and AML. CONCLUSIONS Sepsis is associated with increased or decreased risks for a small group of cancers. Factors that may explain these associations include etiologic effects. Other associations may reflect the presence of precursor conditions or patterns in ascertainment of cancer and screening.
Collapse
Affiliation(s)
- Zhiwei Liu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Parag Mahale
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| | - Eric A Engels
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Rockville, Maryland
| |
Collapse
|
27
|
Lakes JE, Richards CI, Flythe MD. Inhibition of Bacteroidetes and Firmicutes by select phytochemicals. Anaerobe 2019; 61:102145. [PMID: 31918362 DOI: 10.1016/j.anaerobe.2019.102145] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/11/2019] [Accepted: 12/23/2019] [Indexed: 01/26/2023]
Abstract
Current research indicates that changes in gut microbiota can impact the host, but it is not always clear how dietary and environmental factors alter gut microbiota. One potential factor is antimicrobial activity of compounds ingested by the host. The goal of this study was to determine the antimicrobial activity of common plant secondary metabolites against pure cultures of paired, structurally and phylogenetically distinct gastrointestinal bacteria of human or bovine origin: Prevotella bryantii B14, Bacteroides fragilis 25285, Acetoanaerobium (Clostridium) sticklandii SR and Clostridioides difficile 9689. When growth media were amended with individual phytochemicals (the alkaloids: berberine, capsaicin, nicotine, piperine and quinine and the phenolic: curcumin), growth of each species was inhibited to varying degrees at the three greatest concentrations tested (0.10-10.00 mg mL-1). The viable cell numbers of all the cultures were reduced, ≥4-logs, by berberine at concentrations ≥1.00 mg mL-1. Quinine performed similarly to berberine for B14, 25285, and SR at the same concentrations. The other phytochemicals were inhibitory, but not as much as quinine or berberine. Nicotine had activity against all four species (≥2-log reduction in viable cell number at 10.00 mg mL-1), but had stronger activity against the Gram-positive bacteria, SR and 9689, (≥4-log reductions at 10.00 mg mL-1). In conclusion, the phytochemicals had varying spectra of antimicrobial activity. These results are consistent with the hypothesis that ingested phytochemicals have the ability to differentially impact gut microbiota through antimicrobial activity.
Collapse
Affiliation(s)
- Jourdan E Lakes
- Department of Chemistry, College of Arts & Sciences, University of Kentucky, Lexington, KY, USA
| | - Christopher I Richards
- Department of Chemistry, College of Arts & Sciences, University of Kentucky, Lexington, KY, USA
| | - Michael D Flythe
- USDA Agricultural Research Service Forage-Animal Production Research Unit, Lexington, KY, USA; Department of Animal and Food Sciences, College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY, USA.
| |
Collapse
|
28
|
Bao C, Wang K, Ding Y, Kong J. Association Between Anti-bacterial Drug Use and Digestive System Neoplasms: A Systematic Review and Meta-analysis. Front Oncol 2019; 9:1298. [PMID: 31828038 PMCID: PMC6890852 DOI: 10.3389/fonc.2019.01298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/08/2019] [Indexed: 12/14/2022] Open
Abstract
Background: Anti-bacterial drugs are thought to be associated with several malignancies. Objective: We conducted a systematic review and meta-analysis to assess the association between antibacterial drug exposure and the risk of digestive system neoplasms. Methods: Relevant publications reporting a relationship between antibiotic use and the risk of cancer were identified in PubMed, EMBASE, and Cochrane Central Register through June 2018. The random-effects model was selected to pool the risk ratios (RRs) and determine 95% confidence intervals (95% CIs). We performed subgroup analyses by tumor organ site, individual antibacterial drug class, and drug dose accumulation. Results: A total of 17 eligible studies (four randomized trials and 13 observational studies) involving 77,284 cancer patients were included in our analyses. Anti-bacterial drug exposure slightly increased the risk of overall digestive system cancer (RR, 1.12; 95% CI, 1.10-1.14), stomach and small intestine (RR, 1.12; 95% CI, 1.07-1.17), anorectocolonic (RR, 1.08; 95% CI, 1.05-1.12), and hepatobiliary and pancreatic cancers (RR, 1.18; 95% CI, 1.14-1.22). For different anti-bacterial drugs classes, nitroimidazoles (RR, 1.17; 95% CI, 1.09-1.26) and quinolones (RR, 1.18; 95% CI, 1.11-1.26) showed a modest association with the risk of cancers incidence. The risks of digestive system cancers increased with the rise of drug dose accumulation: low (RR, 1.08; 95% CI, 1.05-1.11), intermediate (RR, 1.15; 95% CI, 1.12-1.18), and high (RR, 1.22; 95% CI, 1.18-1.26). Conclusions: Anti-bacterial drug exposure was associated with the risks of digestive system cancer occurrence in our analysis.
Collapse
Affiliation(s)
| | - Ke Wang
- Department of Respiratory Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | | | - Jinliang Kong
- Department of Respiratory Disease, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| |
Collapse
|
29
|
Falcone M, Tiseo G, Venditti M, Menichetti F. Updates in the epidemiology and management of candidemia and Clostridioides difficile coinfection. Expert Rev Anti Infect Ther 2019; 17:375-382. [PMID: 30982376 DOI: 10.1080/14787210.2019.1608183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Introduction: In recent years, more and more studies have focused on the association between candidemia and Clostridioides difficile infection (CDI), highlighting the risk of subsequent candidemia in patients with CDI. However, a more recent model focuses on the Candida-Clostridioides difficile coinfection as a clinical entity in which candidemia could occur before or after the CDI episode. Areas covered: In this review we analyzed the physiopathological mechanisms underlying the Candida-Clostridioides difficile coinfection, both in case of candidemia preceding and following the CDI. We highlighted that gut alterations occurring during a CDI play a crucial role in the risk of subsequent candidemia. Moreover, we identified areas of interest about the management of Candida-Clostridioides difficile coinfection and proposed answers to relevant clinical questions. Expert opinion: The evaluation of risk factors for candidemia in patients with CDI and the rational antibiotic use in patients with candidemia remain the most efficacious and cost-free instruments to optimally manage the Candida-Clostridioides difficile coinfection. However, further studies are required to cover some unmet needs, such as the development of rapid diagnostic methods of candidemia and the use of new available drugs with minimal effect on the microbiome biodiversity in patients with CDI at high risk of fungemia.
Collapse
Affiliation(s)
- Marco Falcone
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Giusy Tiseo
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| | - Mario Venditti
- b Department of Public Health and Infectious Diseases , 'Sapienza' University of Rome , Rome , Italy
| | - Francesco Menichetti
- a Department of Clinical and Experimental Medicine , University of Pisa , Pisa , Italy
| |
Collapse
|
30
|
Shi Y, Kellingray L, Zhai Q, Gall GL, Narbad A, Zhao J, Zhang H, Chen W. Structural and Functional Alterations in the Microbial Community and Immunological Consequences in a Mouse Model of Antibiotic-Induced Dysbiosis. Front Microbiol 2018; 9:1948. [PMID: 30186263 PMCID: PMC6110884 DOI: 10.3389/fmicb.2018.01948] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2018] [Accepted: 08/02/2018] [Indexed: 12/11/2022] Open
Abstract
The aim of this study was to establish continuous therapeutic-dose ampicillin (CTDA)-induced dysbiosis in a mouse model, mimicking typical adult exposure, with a view to using this to assess its impact on gut microbiota, intestinal metabolites and host immune responses. Mice were exposed to ampicillin for 14 days and antibiotic-induced dysbiosis was evaluated by alteration of microbiota and gut permeability. The cecal index was increased in the CTDA group, and the gut permeability indicated by fluorescent dextran, endotoxin and D-Lactate in the serum was significantly increased after antibiotic use. The tight-junction proteins ZO-1 and occludin in the colon were reduced to half the control level in CTDA. We found that alpha-diversity was significantly decreased in mice receiving CTDA, and microbial community structure was altered compared with the control. Key taxa were identified as CTDA-specific, and the relative abundance of Enterococcus and Klebsiella was particularly enriched while Lachnospiraceae, Coprobacillus and Dorea were depleted after antibiotic treatment. In particular, a significant increase in succinate and a reduction in butyrate was detected in CTDA mice, and the triggering of NF-κB enhancement reflected that the host immune response was influenced by ampicillin use. The observed perturbation of the microbiota was accompanied by modulation of inflammatory state; this included increase in interferon-γ and RegIIIγ, and a decrease in secretory IgA in the colon mucosa. This study allowed us to identify the key taxa associated with an ampicillin-induced state of dysbiosis in mice and to characterize the microbial communities via molecular profiling. Thus, this work describes the bacterial ecology of antibiotic exposure model in combination with host physiological characteristics at a detailed level of microbial taxa.
Collapse
Affiliation(s)
- Ying Shi
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Wuxi, China.,International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, China.,Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Lee Kellingray
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom.,UK-China Joint Centre on Probiotic Bacteria, Norwich, United Kingdom
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Wuxi, China.,International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi, China
| | - Gwenaelle Le Gall
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom
| | - Arjan Narbad
- Gut Microbes and Health Programme, Quadram Institute Bioscience, Norwich, United Kingdom.,UK-China Joint Centre on Probiotic Bacteria, Norwich, United Kingdom
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Wuxi, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi, China.,National Engineering Research Center for Functional Food, Wuxi, China.,Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing, China
| |
Collapse
|
31
|
Dissection of the antimicrobial and hemolytic activity of Cap18: Generation of Cap18 derivatives with enhanced specificity. PLoS One 2018; 13:e0197742. [PMID: 29852015 PMCID: PMC5978884 DOI: 10.1371/journal.pone.0197742] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 05/08/2018] [Indexed: 01/08/2023] Open
Abstract
Due to the rapid emergence of resistance to classical antibiotics, novel antimicrobial compounds are needed. It is desirable to selectively kill pathogenic bacteria without targeting other beneficial bacteria in order to prevent the negative clinical consequences caused by many broad-spectrum antibiotics as well as reducing the development of antibiotic resistance. Antimicrobial peptides (AMPs) represent an alternative to classical antibiotics and it has been previously demonstrated that Cap18 has high antimicrobial activity against a broad range of bacterial species. In this study we report the design of a positional scanning library consisting of 696 Cap18 derivatives and the subsequent screening for antimicrobial activity against Y. ruckeri, A. salmonicida, S. Typhimurium and L. lactis as well as for hemolytic activity measuring the hemoglobin release of horse erythrocytes. We show that the hydrophobic face of Cap18, in particular I13, L17 and I24, is essential for its antimicrobial activity against S. Typhimurium, Y. ruckeri, A. salmonicida, E. coli, P. aeruginosa, L. lactis, L. monocytogenes and E. faecalis. In particular, Cap18 derivatives harboring a I13D, L17D, L17P, I24D or I24N substitution lost their antimicrobial activity against any of the tested bacterial strains. In addition, we were able to generate species-specific Cap18 derivatives by particular amino acid substitutions either in the hydrophobic face at positions L6, L17, I20, and I27, or in the hydrophilic face at positions K16 and K18. Finally, our data showed the proline residue at position 29 to be essential for the inherent low hemolytic activity of Cap18 and that substitution of the residues K16, K23, or G21 by any hydrophobic residues enhances the hemolytic activity. This study demonstrates the potential of generating species-specific AMPs for the selective elimination of bacterial pathogens.
Collapse
|
32
|
Zeineldin M, Aldridge B, Blair B, Kancer K, Lowe J. Impact of parenteral antimicrobial administration on the structure and diversity of the fecal microbiota of growing pigs. Microb Pathog 2018; 118:220-229. [DOI: 10.1016/j.micpath.2018.03.035] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2018] [Revised: 03/16/2018] [Accepted: 03/21/2018] [Indexed: 12/31/2022]
|
33
|
Falcone M, Venditti M, Sanguinetti M, Posteraro B. Management of candidemia in patients with Clostridium difficile infection. Expert Rev Anti Infect Ther 2017; 14:679-85. [PMID: 27254270 DOI: 10.1080/14787210.2016.1197118] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
INTRODUCTION Patients with C. difficile infection (CDI) experience intestinal microflora changes that can promote the overgrowth and subsequent translocation of gut resident pathogens into the blood. Consistently, CDI due to PCR-ribotype 027 strain, severe or relapsing CDI, and treatment with high-dosage vancomycin are independent risk factors for candidemia. AREAS COVERED We review the role played by the gut microbiota during CDI and its treatment, as well as the clinical profile of CDI patients who are at risk of developing candidemia. Also, we discuss the management of these patients by focusing on pre-emptive strategies aimed at reducing the risk of candidemia, and on innovative anti-C. difficile therapies that may mitigate CDI-related effects such as the altered gut microbiota composition and prolonged intestinal mucosa damage. Expert commentary: A closer clinical and diagnostic monitoring of patients with CDI should help to limit the CDI-associated long-term consequences, including Candida infections, which worsen the outcome of hospitalized patients.
Collapse
Affiliation(s)
- Marco Falcone
- a Department of Public Health and Infectious Diseases , Sapienza University of Rome , Rome , Italy
| | - Mario Venditti
- a Department of Public Health and Infectious Diseases , Sapienza University of Rome , Rome , Italy
| | - Maurizio Sanguinetti
- b Institute of Microbiology , Università Cattolica del Sacro Cuore , Rome , Italy
| | - Brunella Posteraro
- c Institute of Public Health (Section of Hygiene) , Università Cattolica del Sacro Cuore , Rome , Italy
| |
Collapse
|
34
|
Grellmann AP, Sfreddo CS, Maier J, Lenzi TL, Zanatta FB. Systemic antimicrobials adjuvant to periodontal therapy in diabetic subjects: a meta-analysis. J Clin Periodontol 2017; 43:250-60. [PMID: 26790108 DOI: 10.1111/jcpe.12514] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/12/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Adjuvant antibiotics have been suggested to improve periodontal therapy in diabetic subjects. AIM The aim of this study was to systematically review randomized clinical trials assessing systemic antimicrobial use adjuvant to scaling and root planing (SRP) versus SRP alone in diabetic subjects. MATERIAL AND METHODS The PubMed, Cochrane Central Register of Controlled Trials, EMBASE, TRIP, Web of Science and LILACS databases and the grey literature were searched through May 2015. Of 2534 potentially eligible studies, 13 were included in the systematic review. Weighted mean differences (WMDs) in probing depth (PD) reduction and clinical attachment level (CAL) gain (primary outcomes), and plaque index (PI) and bleeding on probing (BOP) reductions, were estimated using a random effect model. RESULTS The WMD in PD reduction [-0.15 mm, n = 11, p = 0.001, 95% confidence interval (CI) -0.24, -0.06] favoured antibiotic use. WMDs in CAL gain, PI and BOP reductions (-0.14 mm, n = 9, p = 0.11, 95% CI -0.32, 0.03; 4.01%, n = 7, p = 0.05, 95% CI -0.04, 8.07; and -1.91%, n = 7, p = 0.39, 95% CI -6.32, 2.51 respectively) did not favour adjunctive antibiotic use. CONCLUSION Adjunctive therapy may improve the efficacy of SRP in reducing PD in diabetic subjects.
Collapse
Affiliation(s)
| | - Camila Silveira Sfreddo
- Post-graduate Program in Dentistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Juliana Maier
- Post-graduate Program in Dentistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | - Tathiane Larissa Lenzi
- Post-graduate Program in Dentistry, Federal University of Santa Maria, Santa Maria, RS, Brazil
| | | |
Collapse
|
35
|
Wardhani, S., Ridho, M. R., Arinafril, Arita, S., Ngudiantoro. Consortium of heterotrophic nitrification bacteria Bacillus sp. and its application on urea fertilizer industrial wastewater treatment. MALAYSIAN JOURNAL OF MICROBIOLOGY 2017. [DOI: 10.1016/s1773-035x(15)72824-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
36
|
Ma H, Stone VN, Wang H, Kellogg GE, Xu P, Zhang Y. Diaminopimelic acid (DAP) analogs bearing isoxazoline moiety as selective inhibitors against meso-diaminopimelate dehydrogenase (m-Ddh) from Porphyromonas gingivalis. Bioorg Med Chem Lett 2017; 27:3840-3844. [PMID: 28668193 PMCID: PMC5546221 DOI: 10.1016/j.bmcl.2017.06.056] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Revised: 06/19/2017] [Accepted: 06/21/2017] [Indexed: 11/18/2022]
Abstract
Two diastereomeric analogs (1 and 2) of diaminopimelic acid (DAP) bearing an isoxazoline moiety were synthesized and evaluated for their inhibitory activities against meso-diaminopimelate dehydrogenase (m-Ddh) from the periodontal pathogen, Porphyromonas gingivalis. Compound 2 showed promising inhibitory activity against m-Ddh with an IC50 value of 14.9µM at pH 7.8. The two compounds were further tested for their antibacterial activities against a panel of periodontal pathogens, and compound 2 was shown to be selectively potent to P. gingivalis strains W83 and ATCC 33277 with minimum inhibitory concentration (MIC) values of 773µM and 1.875mM, respectively. Molecular modeling studies revealed that the inversion of chirality at the C-5 position of these compounds was the primary reason for their different biological profiles. Based on these preliminary results, we believe that compound 2 has properties consistent with it being a lead compound for developing novel pathogen selective antibiotics to treat periodontal diseases.
Collapse
Affiliation(s)
- Hongguang Ma
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, Richmond, VA 23298, USA
| | - Victoria N Stone
- Philips Institute for Oral Health Research, Virginia Commonwealth University, 521 North 11th Street, Richmond, VA 23298, USA; Department of Microbiology and Immunology, Virginia Commonwealth University, 1101 East Marshall Street, Richmond, VA 23298, USA
| | - Huiqun Wang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, Richmond, VA 23298, USA
| | - Glen E Kellogg
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, Richmond, VA 23298, USA; Institute for Structural Biology, Drug Discovery and Development, Virginia Commonwealth University, 800 East Leigh Street, Richmond, VA 23298, USA
| | - Ping Xu
- Philips Institute for Oral Health Research, Virginia Commonwealth University, 521 North 11th Street, Richmond, VA 23298, USA; Department of Microbiology and Immunology, Virginia Commonwealth University, 1101 East Marshall Street, Richmond, VA 23298, USA
| | - Yan Zhang
- Department of Medicinal Chemistry, Virginia Commonwealth University, 800 East Leigh Street, Richmond, VA 23298, USA.
| |
Collapse
|
37
|
Toutain PL, Ferran AA, Bousquet-Melou A, Pelligand L, Lees P. Veterinary Medicine Needs New Green Antimicrobial Drugs. Front Microbiol 2016; 7:1196. [PMID: 27536285 PMCID: PMC4971058 DOI: 10.3389/fmicb.2016.01196] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Accepted: 07/19/2016] [Indexed: 01/08/2023] Open
Abstract
Given that: (1) the worldwide consumption of antimicrobial drugs (AMDs) used in food-producing animals will increase over the coming decades; (2) the prudent use of AMDs will not suffice to stem the rise in human antimicrobial resistance (AMR) of animal origin; (3) alternatives to AMD use are not available or not implementable, there is an urgent need to develop novel AMDs for food-producing animals. This is not for animal health reasons, but to break the link between human and animal resistomes. In this review we establish the feasibility of developing for veterinary medicine new AMDs, termed "green antibiotics," having minimal ecological impact on the animal commensal and environmental microbiomes. We first explain why animal and human commensal microbiota comprise a "turnstile" exchange, between the human and animal resistomes. We then outline the ideal physico-chemical, pharmacokinetic, and pharmacodynamic properties of a veterinary green antibiotic and conclude that they can be developed through a rational screening of currently used AMD classes. The ideal drug will be hydrophilic, of relatively low potency, slow clearance and small volume of distribution. It should be eliminated principally by the kidney as inactive metabolite(s). For oral administration, bioavailability can be enhanced by developing lipophilic pro-drugs. For parenteral administration, slow-release formulations of existing eco-friendly AMDs with a short elimination half-life can be developed. These new eco-friendly veterinary AMDs can be developed from currently used drug classes to provide alternative agents to those currently used in veterinary medicine and mitigate animal contributions to the human AMR problem.
Collapse
Affiliation(s)
- Pierre-Louis Toutain
- Ecole Nationale Vétérinaire de Toulouse, Institut National de la Recherche Agronomique, TOXALIM, Université de ToulouseToulouse, France
| | - Aude A. Ferran
- Ecole Nationale Vétérinaire de Toulouse, Institut National de la Recherche Agronomique, TOXALIM, Université de ToulouseToulouse, France
| | - Alain Bousquet-Melou
- Ecole Nationale Vétérinaire de Toulouse, Institut National de la Recherche Agronomique, TOXALIM, Université de ToulouseToulouse, France
| | - Ludovic Pelligand
- Comparative Biomedical Sciences, The Royal Veterinary CollegeHatfield, UK
| | - Peter Lees
- Comparative Biomedical Sciences, The Royal Veterinary CollegeHatfield, UK
| |
Collapse
|
38
|
Maxson T, Mitchell DA. Targeted Treatment for Bacterial Infections: Prospects for Pathogen-Specific Antibiotics Coupled with Rapid Diagnostics. Tetrahedron 2016; 72:3609-3624. [PMID: 27429480 PMCID: PMC4941824 DOI: 10.1016/j.tet.2015.09.069] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Antibiotics are a cornerstone of modern medicine and have significantly reduced the burden of infectious diseases. However, commonly used broad-spectrum antibiotics can cause major collateral damage to the human microbiome, causing complications ranging from antibiotic-associated colitis to the rapid spread of resistance. Employing narrower spectrum antibiotics targeting specific pathogens may alleviate this predicament as well as provide additional tools to expand an antibiotic repertoire threatened by the inevitability of resistance. Improvements in clinical diagnosis will be required to effectively utilize pathogen-specific antibiotics and new molecular diagnostics are poised to fulfill this need. Here we review recent trends and the future prospects of deploying narrower spectrum antibiotics coupled with rapid diagnostics. Further, we discuss the theoretical advantages and limitations of this emerging approach to controlling bacterial infectious diseases.
Collapse
Affiliation(s)
- Tucker Maxson
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Douglas A. Mitchell
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Department of Microbiology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
- Carle R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
39
|
Verdugo F, Laksmana T, Uribarri A. Systemic antibiotics and the risk of superinfection in peri-implantitis. Arch Oral Biol 2015; 64:39-50. [PMID: 26761363 DOI: 10.1016/j.archoralbio.2015.12.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Revised: 11/29/2015] [Accepted: 12/23/2015] [Indexed: 12/15/2022]
Abstract
Peri-implantitis has emerged in the last few years as a complication difficult to resolve. The etiopathogenesis consensus is mainly attributed to bacteria. Following the preferred reporting items for systematic reviews and meta-analysis (PRISMA) guidelines, a PubMed/Medline literature search was performed using the US National Library of Medicine database up to 2015 to analyze available scientific data on the rationale and risk of superinfection associated to systemic antimicrobials in human peri-implant disease. A hand search was also conducted on relevant medical and microbiology journals. The methodological index for non-randomized studies (MINORS) was independently assessed for quality on the selected papers. Proposed combined therapies use broad-spectrum antibiotics to halt the disease progression. A major associated risk, particularly when prescribed empirically without microbiological follow-up, is the undetected development of superinfections and overgrowth of opportunistic pathogens difficult to eradicate. Peri-implant superinfections with opportunistic bacteria, yeast and viruses, are plausible risks associated to the use of systemic antibiotics in immunocompetent individuals. Lack of microbiological follow-up and antibiotic susceptibility testing may lead to ongoing microbial challenges that exacerbate the disease progression. The increased proliferation of antimicrobial resistance, modern implant surface topography and indiscriminative empiric antibiotic regimens may promote the escalation of peri-implant disease in years to come. A personalized 3-month supportive therapy may help prevent risks by sustaining a normal ecological balance, decreasing specific pathogen proportions and maintaining ideal plaque control.
Collapse
Affiliation(s)
- Fernando Verdugo
- Department of Periodontics, VA Hospital, Greater Los Angeles Healthcare System, USA.
| | - Theresia Laksmana
- Advanced Periodontology, University of Southern California School of Dentistry, Los Angeles, CA, USA
| | - Agurne Uribarri
- Department of Oral Medicine, School of Medicine and Odontology, University of Basque Country, Leioa, Spain
| |
Collapse
|
40
|
Rashid MU, Rosenborg S, Panagiotidis G, Söderberg-Löfdal K, Weintraub A, Nord CE. Ecological Effect of Ceftaroline-Avibactam on the Normal Human Intestinal Microbiota. Antimicrob Agents Chemother 2015; 59:4504-9. [PMID: 25987638 PMCID: PMC4505277 DOI: 10.1128/aac.00530-15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Accepted: 05/09/2015] [Indexed: 11/20/2022] Open
Abstract
Ceftaroline-avibactam is a new combination of the antibiotic ceftaroline with a novel non-β-lactam β-lactamase inhibitor, avibactam. The purpose of the present study was to investigate the effect of ceftaroline-avibactam on the human intestinal microbiota. Fourteen healthy volunteers received ceftaroline-avibactam (600 mg ceftaroline fosamil and 600 mg avibactam) intravenously over 2 h every 8 h on days 1 to 6 and as a single dose on day 7. Fecal samples were collected on day -1 (within 24 h of the first infusion on day 1) and on days 2, 5, 7, 9, 14, and 21. Escherichia coli numbers decreased during the study and normalized on day 21. An increased number of Klebsiella bacteria appeared on day 14 and normalized on day 21. The number of other enterobacteria decreased during the study, and the number of enterococci decreased from days 2 to 7 and normalized on day 9. Candida numbers increased from days 5 to 9 and normalized after day 14. The number of lactobacilli decreased during the study and recovered on day 14. The number of bifidobacteria decreased on day 2 and normalized on day 21. The number of Bacteroides bacteria was unchanged. Clostridium difficile numbers decreased on days 7 and 9 and increased on days 14 and 21. A toxigenic C. difficile strain was detected in one volunteer on day 21 with no reported adverse events. Plasma samples were collected on days -1, 2, 5, and 7. Ceftaroline and avibactam concentrations were 0 to 34.5 mg/liter and 0 to 61.6 mg/liter, respectively, in plasma and 0 to 35.4 mg/kg and 0 to 98.5 mg/kg, respectively, in feces. (This study is registered in the European Clinical Trials Database [https://eudract.ema.europa.eu/] under number EudraCT 2012 004921-25.).
Collapse
Affiliation(s)
- Mamun-Ur Rashid
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Staffan Rosenborg
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Georgios Panagiotidis
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Karin Söderberg-Löfdal
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Andrej Weintraub
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Carl Erik Nord
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
41
|
Dalhoff A, Rashid MU, Kapsner T, Panagiotidis G, Weintraub A, Nord CE. Analysis of effects of MCB3681, the antibacterially active substance of prodrug MCB3837, on human resident microflora as proof of principle. Clin Microbiol Infect 2015; 21:767.e1-4. [PMID: 26047854 DOI: 10.1016/j.cmi.2015.05.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Revised: 05/09/2015] [Accepted: 05/18/2015] [Indexed: 11/26/2022]
Abstract
The water-soluble prodrug MCB3837 is rapidly converted to MCB3681, active against Gram-positive bacterial species, after intravenous infusion. The aim of this study was to prove the principle that MCB3681 is efficacious in vivo by demonstrating its effect on the resident microflora or colonizers of the human skin, nose, oropharynx and intestine. MCB3837 was infused at a daily dose of 6 mg/kg for 5 days. MCB3681 was active against clostridia, bifidobacteria, lactobacilli, enterococci and Staphylococcus aureus, thus proving the principle that MCB3681 is antibacterially efficacious in vivo without affecting the Gram-negative microflora.
Collapse
Affiliation(s)
- A Dalhoff
- Institute for Infection Medicine, University Medical Center Schleswig-Holstein, Kiel, Germany.
| | - M-U Rashid
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - T Kapsner
- Morphochem Aktiengesellschaft für kombinatorische Chemie, Munich, Germany
| | - G Panagiotidis
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - A Weintraub
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - C E Nord
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
42
|
Determining the Long-term Effect of Antibiotic Administration on the Human Normal Intestinal Microbiota Using Culture and Pyrosequencing Methods. Clin Infect Dis 2015; 60 Suppl 2:S77-84. [DOI: 10.1093/cid/civ137] [Citation(s) in RCA: 105] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
|
43
|
Rashid MU, Rosenborg S, Panagiotidis G, Löfdal KS, Weintraub A, Nord CE. Ecological effect of ceftazidime/avibactam on the normal human intestinal microbiota. Int J Antimicrob Agents 2015; 46:60-5. [PMID: 25979639 DOI: 10.1016/j.ijantimicag.2015.02.027] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 02/24/2015] [Accepted: 02/25/2015] [Indexed: 11/25/2022]
Abstract
Ceftazidime/avibactam is a new combination of the antibiotic ceftazidime with the novel, non-β-lactam β-lactamase inhibitor avibactam. The purpose of the present study was to investigate the effect of ceftazidime/avibactam on the human intestinal microbiota following intravenous (i.v.) administration. Twelve healthy volunteers received ceftazidime/avibactam by i.v. infusion (2000mg ceftazidime and 500mg avibactam) given over 2h every 8h on Days 1-6 (inclusive) and a single dose on Day 7. Faecal samples were collected on Day-1 (pre-dose), during administration on Days 2, 5 and 7 and post-dose on Days 9, 14 and 21. Samples were cultured on non-selective and selective media. The number of Escherichia coli and other enterobacteria decreased significantly during administration of ceftazidime/avibactam, whereas the number of enterococci increased. Lactobacilli, bifidobacteria, clostridia and Bacteroides decreased significantly during ceftazidime/avibactam administration. The effects on lactobacilli, bifidobacteria and Bacteroides were similar in the 12 volunteers, whilst clostridia showed different ecological patterns among the volunteers. Toxigenic Clostridium difficile strains were detected in five volunteers during the study. In four of the volunteers, loose stools were reported as adverse events. Plasma samples were collected on Days -1, 2, 5 and 7. Ceftazidime and avibactam concentrations in plasma (ceftazidime 0-224.2mg/L of plasma and avibactam 0-70.5mg/L of plasma) and faeces (ceftazidime 0-468.2mg/kg of faeces and avibactam 0-146.0mg/kg of faeces) were found by bioassay. New colonising resistant clostridia were found in five volunteers and lactobacilli were found in three volunteers.
Collapse
Affiliation(s)
- Mamun-Ur Rashid
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Staffan Rosenborg
- Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden; Clinical Pharmacology Trial Unit, Karolinska University Hospital, Huddinge, Sweden
| | - Georgios Panagiotidis
- Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden; Clinical Pharmacology Trial Unit, Karolinska University Hospital, Huddinge, Sweden
| | - Karin Söderberg Löfdal
- Department of Laboratory Medicine, Division of Clinical Pharmacology, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden; Clinical Pharmacology Trial Unit, Karolinska University Hospital, Huddinge, Sweden
| | - Andrej Weintraub
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Carl Erik Nord
- Department of Laboratory Medicine, Division of Clinical Microbiology, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
| |
Collapse
|
44
|
History of antimicrobial use and the risk of Dientamoeba fragilis infection. Eur J Clin Microbiol Infect Dis 2015; 34:1145-51. [PMID: 25663130 DOI: 10.1007/s10096-015-2334-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Accepted: 01/21/2015] [Indexed: 12/23/2022]
Abstract
Associations between antimicrobial use and risk of enteric infection with intestinal protozoa are scarcely studied. The aim of this study was to quantify the risk of Dientamoeba fragilis infection conferred by exposure to antimicrobials. We conducted a registry-based retrospective cohort study of 9,945 Danish patients investigated for D. fragilis infection between 2008 and 2011, using data from the Danish Register of Medicinal Product Statistics, and calculating relative risks (RR) for D. fragilis infection by stratified binary regression. Furthermore, we conducted a population based case-control study using controls sampled from the Danish Civil Registration System, calculating hazard ratios (HR) for D. fragilis infection by conditional logistic regression. Exposure to metronidazole was found to confer decreased risk of D. fragilis infection; however, similar associations were found for antimicrobials not commonly used to treat D. fragilis, such as broad-spectrum penicillin, fluoroquinolones, and macrolides. In contrast, mebendazole exposure was associated with increased risk. The intake of antimicrobials influences the risk of D. fragilis.
Collapse
|
45
|
Rashid MU, Weintraub A, Nord CE. Development of antimicrobial resistance in the normal anaerobic microbiota during one year after administration of clindamycin or ciprofloxacin. Anaerobe 2015; 31:72-7. [DOI: 10.1016/j.anaerobe.2014.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/02/2014] [Accepted: 10/05/2014] [Indexed: 10/24/2022]
|
46
|
Jang MO, An JH, Jung SI, Park KH. Refractory Clostridium difficile Infection Cured With Fecal Microbiota Transplantation in Vancomycin-Resistant Enterococcus Colonized Patient. Intest Res 2015; 13:80-4. [PMID: 25691847 PMCID: PMC4316227 DOI: 10.5217/ir.2015.13.1.80] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2013] [Revised: 04/03/2014] [Accepted: 04/04/2014] [Indexed: 01/18/2023] Open
Abstract
The rates and severity of Clostridium difficile infections, including pseudomembranous colitis, have increased markedly. However, there are few effective treatments for refractory or recurrent C. difficile infections and the outcomes are poor. Fecal microbiota transplantation is becoming increasingly accepted as an effective and safe intervention in patients with recurrent disease, likely due to the restoration of a disrupted microbiome. Cure rates of >90% are being consistently reported from multiple centers. We cured a case of severe refractory C. difficile infection with fecal microbiota transplantation in a patient colonized by vancomycin-resistant enterococcus.
Collapse
Affiliation(s)
- Mi-Ok Jang
- Department of Internal Medicine, Chonnam National University Medical School, Gwang-ju, Korea
| | - Jun Hwan An
- Department of Internal Medicine, Chonnam National University Medical School, Gwang-ju, Korea
| | - Sook-In Jung
- Department of Internal Medicine, Chonnam National University Medical School, Gwang-ju, Korea
| | - Kyung-Hwa Park
- Department of Internal Medicine, Chonnam National University Medical School, Gwang-ju, Korea
| |
Collapse
|
47
|
Allen HK, Trachsel J, Looft T, Casey TA. Finding alternatives to antibiotics. Ann N Y Acad Sci 2014; 1323:91-100. [DOI: 10.1111/nyas.12468] [Citation(s) in RCA: 149] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Heather K. Allen
- United States Department of Agriculture Food Safety and Enteric Pathogens Research Unit National Animal Disease Center Agricultural Research Service Ames Iowa
| | - Julian Trachsel
- United States Department of Agriculture Food Safety and Enteric Pathogens Research Unit National Animal Disease Center Agricultural Research Service Ames Iowa
| | - Torey Looft
- United States Department of Agriculture Food Safety and Enteric Pathogens Research Unit National Animal Disease Center Agricultural Research Service Ames Iowa
| | - Thomas A. Casey
- United States Department of Agriculture Food Safety and Enteric Pathogens Research Unit National Animal Disease Center Agricultural Research Service Ames Iowa
| |
Collapse
|
48
|
Alternative strategies for proof-of-principle studies of antibacterial agents. Antimicrob Agents Chemother 2014; 58:4257-63. [PMID: 24867989 DOI: 10.1128/aac.02473-14] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The proof that a new antibacterial agent is not only active in vitro but also effective in vivo under clinically relevant conditions is currently provided (i) by using appropriate nonclinical models of infection and pharmacokinetic-pharmacodynamic (PK-PD) analysis providing evidence of the likelihood of clinical efficacy and (ii) by examining the study drug in exploratory clinical trials, as well as dose and schedule finding during phase II of clinical development. This approach is both time-consuming and costly. Furthermore, PK-PD targets for any novel antibacterial agent cannot be derived from studies with experimental animals. Therefore, alternative strategies have to be identified to prove the principle that a novel antibacterial agent is active under clinically relevant conditions. This review summarizes evidence that the quantitative analysis of shifts in the viable counts of pathogens in infected patients or the evaluation of the PD effect of an investigational agent on indicator organisms of the human resident microflora or colonizers of healthy volunteers, if paralleled with PK monitoring of serum and the target site, provides an alternative to a classical proof-of-principle study in the course of a phase II study program.
Collapse
|
49
|
Rashid MU, Dalhoff A, Bäckström T, Björkhem-Bergman L, Panagiotidis G, Weintraub A, Nord CE. Ecological impact of MCB3837 on the normal human microbiota. Int J Antimicrob Agents 2014; 44:125-30. [PMID: 24931053 DOI: 10.1016/j.ijantimicag.2014.03.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2014] [Revised: 03/26/2014] [Accepted: 03/30/2014] [Indexed: 11/30/2022]
Abstract
MCB3837 is a novel, water-soluble, injectable prodrug that is rapidly converted to the active substance MCB3681 in vivo following intravenous (i.v.) administration. Both MCB3837 and MCB3681 are oxazolidinone-quinolone hybrid molecules. The purpose of the present study was to investigate the effect of MCB3681 on the human skin, nose, oropharyngeal and intestinal microbiota following administration of MCB3837. Twelve healthy male subjects received i.v. MCB3837 (6 mg/kg body weight) once daily for 5 days. Skin, nose, saliva and faecal samples were collected on Day -1 (pre dose), during administration on Days 2 and 5, and post dose on Days 8, 12 and 19. Micro-organisms were identified to genus level. No measurable concentrations of MCB3681 were found in any saliva samples or in the faecal samples on Day -1. On Day 2, 10 volunteers had faecal MCB3681 concentrations between 16.5 mg/kg faeces and 275.1mg/kg faeces; no MCB3681 in faeces could be detected in two of the volunteers. On Day 5, all volunteers had faecal concentrations of MCB3681 ranging from 98.9 to 226.3 mg/kg. MCB3681 caused no ecological changes in the skin, nasal and oropharyngeal microbiota. The numbers of enterococci, bifidobacteria, lactobacilli and clostridia decreased in the intestinal microbiota during administration of the drug. Numbers of Escherichia coli, other enterobacteria and Candida were not affected during the study. There was no impact on the number of Bacteroides. The faecal microbiota was normalised on Day 19. No new colonising aerobic or anaerobic Gram-positive bacteria with MCB3681 minimum inhibitory concentrations of ≥4 mg/L were found.
Collapse
Affiliation(s)
- Mamun-Ur Rashid
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Axel Dalhoff
- University Medical Center Schleswig-Holstein, Institute for Infection Medicine, Brunswiker Str. 4, D-24105 Kiel, Germany
| | - Tobias Bäckström
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Linda Björkhem-Bergman
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Georgios Panagiotidis
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Andrej Weintraub
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden
| | - Carl Erik Nord
- Department of Laboratory Medicine, Karolinska University Hospital, Karolinska Institutet, SE-141 86 Stockholm, Sweden.
| |
Collapse
|
50
|
Khan N, Abbasi AM, Dastagir G, Nazir A, Shah GM, Shah MM, Shah MH. Ethnobotanical and antimicrobial study of some selected medicinal plants used in Khyber Pakhtunkhwa (KPK) as a potential source to cure infectious diseases. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2014; 14:122. [PMID: 24708514 PMCID: PMC3977958 DOI: 10.1186/1472-6882-14-122] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 03/31/2014] [Indexed: 11/20/2022]
Abstract
Background Present investigation deals with antimicrobial screening of ten medicinally important plants used by the inhabitants of district Haripur, Khyber Pakhtunkhwa (KPK) for different infectious diseases. Methods Aqueous, n-hexane and ethanolic extracts of each plant were tested for their antimicrobial activity against both Gram positive and Gram negative strains of bacteria, as well as strain of yeast. Agar well diffusion and broth dilution methods were used to determine the antimicrobial activity of different plant extracts. Results The results indicated that all plants exhibited antimicrobial activity against one or more test pathogens. Interestingly, extracts of three plants showed strong and broad spectrum activity as compared to rest of the extracts which demonstrated the moderate activity. On the whole ethanolic extracts exhibited maximum antimicrobial effect than their corresponding aqueous and n-hexane extracts, when compared with standard antibiotics i.e., Streptomycin and Tetracycline. Among various extracts, only ethanloic extract of Azadirachta indica and aqueous and ethanolic extracts of Eucalyptus globulus and Bergenia ciliata and ethanolic extract of Punica granatum were found to have potentially promising activity against test microorganisms. Conclusion Different plant extracts show promising antimicrobial activity justifying their usage in traditional medicines. This study will be continued to identify more plants with potential antimicrobial components.
Collapse
|