51
|
Leo S, Lazarevic V, von Dach E, Kaiser L, Prendki V, Schrenzel J, Huttner BD, Huttner A. Effects of antibiotic duration on the intestinal microbiota and resistome: The PIRATE RESISTANCE project, a cohort study nested within a randomized trial. EBioMedicine 2021; 71:103566. [PMID: 34492446 PMCID: PMC8426194 DOI: 10.1016/j.ebiom.2021.103566] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/27/2021] [Accepted: 08/17/2021] [Indexed: 12/04/2022] Open
Abstract
Background Shortening antibiotic-treatment durations is a key recommendation of antibiotic-stewardship programmes, yet it is based on weak evidence. We investigated whether halving antibiotic courses would reduce antibiotic-resistance genes (ARG) in the intestinal microbiomes of patients treated for gram-negative bacteraemia. Methods This nested prospective cohort study included adult patients hospitalized at Geneva University Hospitals (Switzerland) participating in the PIRATE randomized trial assessing non-inferiority of shorter antibiotic courses (7 versus 14 days) for gram-negative bacteraemia (‘cases’) and, simultaneously, hospitalized patients with similar demography and comorbidity yet no antibiotic therapy (‘controls’). Stool was collected from case and control patients on days 7, 14, 30 and 90 after antibiotic initiation (day 1) and days 7 and 14 after admission, respectively, and analysed by whole-metagenome shotgun sequencing. The primary outcome was ARG abundance at day 30; secondary outcomes included microbiota-species composition and clustering over time. Findings Forty-five patients and 11 controls were included and evaluable; ARG analyses were conducted on the 29 per-protocol patients receiving 7 (±2) days or 14 (±3) days of antibiotic therapy. At day 30, ARGs were not detected at similar abundance in patients receiving 7 and 14 days (median counts/million [mCPM]: 96 versus [vs] 71; p=.38). By day 30, total ARG content between both groups was not significantly different from that of controls at D7 (362 and 370 mCPM vs 314 mCPM, p=.24 and 0.19). There were no significant differences amongst antibiotic-treated patients at any timepoint in bacterial diversity or clustering, but Shannon species diversity was significantly reduced compared to controls through day 14 (median 3.12 and 3.24 in the 7-day and 14-day groups vs 3.61 [controls]; p=.04 and 0.012). Patients treated for 14 days had reduced faecal phage content during and after therapy compared to other patient groups. Interpretation Reducing antibiotic durations by half did not result in decreased abundance of ARGs in patients treated for gram-negative bacteraemia, nor did it improve microbiota species diversity. Funding The study was funded by the University of Geneva's Louis-Jeantet Foundation (grant no. S04_12) and the Swiss National Science Foundation (NRP Smarter Healthcare, grant no. 407,440_167359).
Collapse
Affiliation(s)
- Stefano Leo
- Genomic Research Laboratory, Division of Infectious Diseases, University Hospitals and University of Geneva, Geneva, Switzerland.
| | - Vladimir Lazarevic
- Genomic Research Laboratory, Division of Infectious Diseases, University Hospitals and University of Geneva, Geneva, Switzerland
| | - Elodie von Dach
- Clinical Research Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland
| | - Laurent Kaiser
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Virginie Prendki
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland; Division of Internal Medicine for the Aged, Hôpital des Trois-Chêne, Thônex-Genève, Geneva, Switzerland
| | - Jacques Schrenzel
- Genomic Research Laboratory, Division of Infectious Diseases, University Hospitals and University of Geneva, Geneva, Switzerland; Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Benedikt D Huttner
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Angela Huttner
- Clinical Research Center, Geneva University Hospitals and University of Geneva, Geneva, Switzerland; Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland; Faculty of Medicine, University of Geneva, Geneva, Switzerland
| |
Collapse
|
52
|
Vallianou N, Dalamaga M, Stratigou T, Karampela I, Tsigalou C. Do Antibiotics Cause Obesity Through Long-term Alterations in the Gut Microbiome? A Review of Current Evidence. Curr Obes Rep 2021; 10:244-262. [PMID: 33945146 PMCID: PMC8093917 DOI: 10.1007/s13679-021-00438-w] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/21/2021] [Indexed: 02/08/2023]
Abstract
PURPOSE OF REVIEW In this review, we summarize current evidence on the association between antibiotics and the subsequent development of obesity through modulation of the gut microbiome. Particular emphasis is given on (i) animal and human studies and their limitations; (ii) the reservoir of antibiotics in animal feed, emerging antibiotic resistance, gut dysbiosis, and obesity; (iii) the role of infections, specifically viral infections, as a cause of obesity; and (iv) the potential therapeutic approaches other than antibiotics to modulate gut microbiome. RECENT FINDINGS Overall, the majority of animal studies and meta-analyses of human studies on the association between antibiotics and subsequent development of obesity are suggestive of a link between exposure to antibiotics, particularly early exposure in life, and the development of subsequent obesity as a result of alterations in the diversity of gut microbiota. The evidence is strong in animal models whereas evidence in humans is inconclusive requiring well-designed, long-term longitudinal studies to examine this association. Based on recent meta-analyses and epidemiologic studies in healthy children, factors, such as the administration of antibiotics during the first 6 months of life, repeated exposure to antibiotics for ≥ 3 courses, treatment with broad-spectrum antibiotics, and male gender have been associated with increased odds of overweight/obesity. Early antibiotic exposure in animal models has shown that reductions in the population size of specific microbiota, such as Lactobacillus, Allobaculum, Rikenellaceae, and Candidatus Arthromitus, are related to subsequent adiposity. These data suggest that the loss of diversity of the gut microbiome, especially early in life, may have potential long-term detrimental effects on the adult host gut microbiome and metabolic health. Genetic, environmental, and age-related factors influence the gut microbiome throughout the lifetime. More large-scale, longer-term, longitudinal studies are needed to determine whether changes that occur in the microbiome after exposure to antibiotics, particularly early exposure, are causal of subsequent weight gain or consequent of weight gain in humans. Further well-designed, large-scale RCTs in humans are required to evaluate the effects of administration of antibiotics, particularly early administration, and the subsequent development of overweight/obesity. Therapeutic interventions, such as bacteriophage treatment or the use of probiotics, especially genetically engineered ones, need to be evaluated in terms of prevention and management of obesity.
Collapse
Affiliation(s)
- Natalia Vallianou
- grid.414655.70000 0004 4670 4329Department of Internal Medicine and Endocrinology, ‘Evangelismos’ General Hospital of Athens, 45-47 Ypsilantou Street, 10676 Athens, Greece
| | - Maria Dalamaga
- grid.5216.00000 0001 2155 0800Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527 Athens, Greece
| | - Theodora Stratigou
- grid.414655.70000 0004 4670 4329Department of Internal Medicine and Endocrinology, ‘Evangelismos’ General Hospital of Athens, 45-47 Ypsilantou Street, 10676 Athens, Greece
| | - Irene Karampela
- grid.5216.00000 0001 2155 0800Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Mikras Asias 75, Goudi, 11527 Athens, Greece
- grid.5216.00000 0001 2155 0800Second Department of Critical Care, Attikon General University Hospital, Medical School, National and Kapodistrian University of Athens, 1 Rimini St, Haidari, 12462 Athens, Greece
| | - Christina Tsigalou
- grid.12284.3d0000 0001 2170 8022Laboratory of Microbiology, Medical School, Democritus University of Thrace, 6th Km Alexandroupolis-Makri, Alexandroupolis, Greece
| |
Collapse
|
53
|
Ciprofloxacin for treatment of drug-resistant epilepsy. Epilepsy Res 2021; 176:106742. [PMID: 34419770 DOI: 10.1016/j.eplepsyres.2021.106742] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/06/2021] [Accepted: 08/13/2021] [Indexed: 12/18/2022]
Abstract
PURPOSE To investigate the efficacy of short-term treatment with ciprofloxacin in alteration of gut microbiota pattern and reduction of seizure frequency in adult patients with drug-resistant epilepsy. METHODS In a prospective study, we investigated the effect of a 5-day course of treatment with ciprofloxacin on gut microbiota pattern and seizure frequency of 23 adults with drug-resistant epilepsy. Fecal samples were collected before and after treatment and were analyzed for microbial load and species. Changes in seizure frequency were registered for 12 weeks. Responders were defined as patients who experienced ≥50 % seizure reduction in comparison to baseline. Outcome measures were specified as alteration in fecal microbial burden in days 5-7 and responder rate in 4th and 12th weeks. RESULTS The mean baseline frequency of seizures was5.6 ±7.7 per week. All patients were on polytherapy with a mean of 3 ± 1.2 anti-seizure medications. Microbial analysis showed a considerable increase in Bacteroidetes/Firmicutes ratio after treatment. Seizure frequency significantly decreased at the end of first week and the therapeutic effect continued to week 12 (P < 0.001). The responder rate at 4th and 12th weeks were 69.6 % and 73.9 % respectively with a more prominent response in patients with symptomatic generalized epilepsy (P:0.06). CONCLUSION Alteration of abnormal gut microbiota pattern by methods such as short-course antibiotic therapy, prescription of probiotics and fecal microbiota transplant might be effective in treatment of drug-resistant epilepsy.
Collapse
|
54
|
Yang L, Bajinka O, Jarju PO, Tan Y, Taal AM, Ozdemir G. The varying effects of antibiotics on gut microbiota. AMB Express 2021; 11:116. [PMID: 34398323 PMCID: PMC8368853 DOI: 10.1186/s13568-021-01274-w] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 08/03/2021] [Indexed: 12/21/2022] Open
Abstract
Antibiotics are lifesaving therapeutic drugs that have been used by human for decades. They are used both in the fight against bacterial pathogens for both human and for animal feeding. However, of recent, their effects on the gut microbial compositions and diversities have attracted much attention. Existing literature have established the dysbiosis (reduced diversity) in the gut microbiota in association with antibiotic and antibiotic drug doses. In the light of spelling out the varying effects of antibiotic use on gut microbiota, this review aimed at given an account on the degree of gut microbial alteration caused by common antibiotics. While some common antibiotics are found to destroy the common phyla, other debilitating effects were observed. The effects can be attributed to the mode of mechanism, the class of antibiotic, the degree of resistance of the antibiotic used, the dosage used during the treatment, the route of administration, the pharmacokinetic and pharmacodynamics properties and the spectrum of the antibiotic agent. Health status, stress or the type of diet an individual feeds on could be a great proportion as confounding factors. While it is understood that only the bacterial communities are explored in the quest to establishing the role of gut in health, other gut microbial species are somehow contributing to the dysbiosis status of the gut microbiota. Until now, long term natural fluctuations like diseases outbreaks and mutations of the strain might as well rendered alteration to the gut independent of antibiotic treatments.
Collapse
|
55
|
Ghanem S, Kim CJ, Dutta D, Salifu M, Lim SH. Antimicrobial therapy during cancer treatment: Beyond antibacterial effects. J Intern Med 2021; 290:40-56. [PMID: 33372309 DOI: 10.1111/joim.13238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Revised: 11/22/2020] [Accepted: 12/01/2020] [Indexed: 02/07/2023]
Abstract
Cancer treatment options have evolved to include immunotherapy and targeted therapy, in addition to traditional chemoradiation. Chemoradiation places the patient at a higher risk of infection through a myelosuppressive effect. High clinical suspicion and early use of antimicrobials play a major role in decreasing any associated morbidity and mortality. This has led to a widespread use of antimicrobials in cancer patients. Antimicrobial use, however, does not come without its perils. Dysbiosis caused by antimicrobial use affects responses to chemotherapeutic agents and is prognostic in the development and severity of certain cancer treatment-related complications such as graft-versus-host disease and Clostridioides difficile infections. Studies have also demonstrated that an intact gut microbiota is essential in the anticancer immune response. Antimicrobial use can therefore modulate responses and outcomes with immunotherapy targeting immune checkpoints. In this review, we highlight the perils associated with antimicrobial use during cancer therapy and the importance of a more judicious approach. We discuss the nature of the pathologic changes in the gut microbiota resulting from antimicrobial use. We explore the effect these changes have on responses and outcomes to different cancer treatment modalities including chemotherapy and immunotherapy, as well as potential adverse clinical consequences in the setting of stem cell transplant.
Collapse
Affiliation(s)
- S Ghanem
- From the, Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - C J Kim
- From the, Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - D Dutta
- From the, Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - M Salifu
- From the, Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - S H Lim
- From the, Division of Hematology and Oncology, Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| |
Collapse
|
56
|
Stewart AG, Satlin MJ, Schlebusch S, Isler B, Forde BM, Paterson DL, Harris PNA. Completing the Picture-Capturing the Resistome in Antibiotic Clinical Trials. Clin Infect Dis 2021; 72:e1122-e1129. [PMID: 33354717 DOI: 10.1093/cid/ciaa1877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Indexed: 11/12/2022] Open
Abstract
Despite the accepted dogma that antibiotic use is the largest contributor to antimicrobial resistance (AMR) and human microbiome disruption, our knowledge of specific antibiotic-microbiome effects remains basic. Detection of associations between new or old antimicrobials and specific AMR burden is patchy and heterogeneous. Various microbiome analysis tools are available to determine antibiotic effects on microbial communities in vivo. Microbiome analysis of treatment groups in antibiotic clinical trials, powered to measure clinically meaningful endpoints would greatly assist the antibiotic development pipeline and clinician antibiotic decision making.
Collapse
Affiliation(s)
- Adam G Stewart
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia.,Department of Infectious Diseases, Royal Brisbane and Women's Hospital, Brisbane, Australia
| | - Michael J Satlin
- Department of Medicine, Division of Infectious Diseases, Weill Cornell Medicine, New York, New York, USA
| | - Sanmarié Schlebusch
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia.,Department of Microbiology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Forensic and Scientific Services, Health Support Queensland, Queensland Health, Brisbane, Australia
| | - Burcu Isler
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia
| | - Brian M Forde
- School of Chemistry and Molecular Biosciences, The University of Queensland, Queensland, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Queensland, Australia
| | - David L Paterson
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia.,Department of Infectious Diseases, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Queensland, Australia
| | - Patrick N A Harris
- Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Royal Brisbane and Women's Hospital Campus, Brisbane, Australia.,Department of Microbiology, Pathology Queensland, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Australian Infectious Diseases Research Centre, The University of Queensland, Queensland, Australia
| |
Collapse
|
57
|
Shah T, Baloch Z, Shah Z, Cui X, Xia X. The Intestinal Microbiota: Impacts of Antibiotics Therapy, Colonization Resistance, and Diseases. Int J Mol Sci 2021; 22:6597. [PMID: 34202945 PMCID: PMC8235228 DOI: 10.3390/ijms22126597] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/08/2021] [Accepted: 06/14/2021] [Indexed: 12/11/2022] Open
Abstract
Trillions of microbes exist in the human body, particularly the gastrointestinal tract, coevolved with the host in a mutually beneficial relationship. The main role of the intestinal microbiome is the fermentation of non-digestible substrates and increased growth of beneficial microbes that produce key antimicrobial metabolites such as short-chain fatty acids, etc., to inhibit the growth of pathogenic microbes besides other functions. Intestinal microbiota can prevent pathogen colonization through the mechanism of colonization resistance. A wide range of resistomes are present in both beneficial and pathogenic microbes. Giving antibiotic exposure to the intestinal microbiome (both beneficial and hostile) can trigger a resistome response, affecting colonization resistance. The following review provides a mechanistic overview of the intestinal microbiome and the impacts of antibiotic therapy on pathogen colonization and diseases. Further, we also discuss the epidemiology of immunocompromised patients who are at high risk for nosocomial infections, colonization and decolonization of multi-drug resistant organisms in the intestine, and the direct and indirect mechanisms that govern colonization resistance to the pathogens.
Collapse
Affiliation(s)
- Taif Shah
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China;
- Yunnan Key Laboratory of Sustainable Utilization of Panax Notoginseng, Kunming 650500, China
| | - Zulqarnain Baloch
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China;
| | - Zahir Shah
- Faculty of Animal Husbandry and Veterinary Sciences, College of Veterinary Sciences, The University of Agriculture Peshawar, Peshawar 25120, Pakistan;
| | - Xiuming Cui
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China;
- Yunnan Key Laboratory of Sustainable Utilization of Panax Notoginseng, Kunming 650500, China
| | - Xueshan Xia
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming 650500, China;
| |
Collapse
|
58
|
Alterations in the Fecal Microbiome and Metabolome of Horses with Antimicrobial-Associated Diarrhea Compared to Antibiotic-Treated and Non-Treated Healthy Case Controls. Animals (Basel) 2021; 11:ani11061807. [PMID: 34204371 PMCID: PMC8235368 DOI: 10.3390/ani11061807] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Accepted: 05/30/2021] [Indexed: 12/16/2022] Open
Abstract
Diarrhea is an adverse effect of antimicrobial therapy in horses. This matched, case-controlled study compared the fecal microbiome and metabolome of horses on antibiotics that developed diarrhea (AAD, n = 17) to those that did not develop diarrhea (ABX, n = 15) and to a control population not exposed to antibiotics (CON, n = 31). Fecal samples were collected from horses that were matched for diet and antimicrobial agent (including dose, route, and duration of therapy). Illumina sequencing of 16S rRNA genes was performed, and QIIME 2.0 was used to generate alpha and beta diversity metrics. Untargeted metabolomics using GC-MS platforms was performed and analyzed using Metaboanalyst 5.0. Microbiome composition was significantly different in AAD compared to CON (ANOSIM, R = 0.568, p = 0.001) but not to ABX (ANOSIM, R = 0.121, p = 0.0012). AAD and ABX horses had significantly decreased richness and evenness compared to CON horses (p < 0.05). Horses on antimicrobials (AAD and ABX) had significant changes in 14 phyla compared to CON horses. Only Verrucomicrobia distinguished AAD from ABX and CON horses (q = 0.0005). Metabolite profiles of horses with AAD clustered separately from ABX and CON horses. Seven metabolites were found to be significantly different between groups (p < 0.05): L-tyrosine, kynurenic acid, xanthurenic acid, 5-hydroxyindole-3-acetic acid, docosahexaenoic acid ethyl ester, daidzein, and N-acetyltyramine. Metabolite profiles of horses on antimicrobials, especially those with AAD, are altered compared to CON horses.
Collapse
|
59
|
Pancu DF, Scurtu A, Macasoi IG, Marti D, Mioc M, Soica C, Coricovac D, Horhat D, Poenaru M, Dehelean C. Antibiotics: Conventional Therapy and Natural Compounds with Antibacterial Activity-A Pharmaco-Toxicological Screening. Antibiotics (Basel) 2021; 10:401. [PMID: 33917092 PMCID: PMC8067816 DOI: 10.3390/antibiotics10040401] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 03/30/2021] [Accepted: 03/31/2021] [Indexed: 12/11/2022] Open
Abstract
Antibiotics are considered as a cornerstone of modern medicine and their discovery offers the resolution to the infectious diseases problem. However, the excessive use of antibiotics worldwide has generated a critical public health issue and the bacterial resistance correlated with antibiotics inefficiency is still unsolved. Finding novel therapeutic approaches to overcome bacterial resistance is imperative, and natural compounds with antibacterial effects could be considered a promising option. The role played by antibiotics in tumorigenesis and their interrelation with the microbiota are still debatable and are far from being elucidated. Thus, the present manuscript offers a global perspective on antibiotics in terms of evolution from a historical perspective with an emphasis on the main classes of antibiotics and their adverse effects. It also highlights the connection between antibiotics and microbiota, focusing on the dual role played by antibiotics in tumorigenesis. In addition, using the natural compounds with antibacterial properties as potential alternatives for the classical antibiotic therapy is discussed.
Collapse
Affiliation(s)
- Daniel Florin Pancu
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 1, 300041 Timisoara, Romania; (D.F.P.); (D.H.); (M.P.)
| | - Alexandra Scurtu
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.S.); (M.M.); (C.S.); (D.C.); (C.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Ioana Gabriela Macasoi
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.S.); (M.M.); (C.S.); (D.C.); (C.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Daniela Marti
- Faculty of Medicine, Western University Vasile Goldis Arad, 94 Revolutiei Blvd., 310025 Arad, Romania
| | - Marius Mioc
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.S.); (M.M.); (C.S.); (D.C.); (C.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Codruta Soica
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.S.); (M.M.); (C.S.); (D.C.); (C.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Dorina Coricovac
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.S.); (M.M.); (C.S.); (D.C.); (C.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| | - Delia Horhat
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 1, 300041 Timisoara, Romania; (D.F.P.); (D.H.); (M.P.)
| | - Marioara Poenaru
- Faculty of Medicine, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 1, 300041 Timisoara, Romania; (D.F.P.); (D.H.); (M.P.)
| | - Cristina Dehelean
- Faculty of Pharmacy, “Victor Babeș” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania; (A.S.); (M.M.); (C.S.); (D.C.); (C.D.)
- Research Center for Pharmaco-Toxicological Evaluations, Faculty of Pharmacy, “Victor Babes” University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square No. 2, 300041 Timisoara, Romania
| |
Collapse
|
60
|
Lu PH, Tsai TC, Chang JWC, Deng ST, Cheng CY. Association of prior fluoroquinolone treatment with survival outcomes of immune checkpoint inhibitors in Asia. J Clin Pharm Ther 2020; 46:408-414. [PMID: 33332621 DOI: 10.1111/jcpt.13298] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 08/28/2020] [Accepted: 09/30/2020] [Indexed: 01/22/2023]
Abstract
WHAT IS KNOWN AND OBJECTIVE Gut microbiota plays an important role in shaping immune responses. Several studies have reported that antibiotics may alter gut microbiota diversity and compromise the therapeutic response to immune checkpoint inhibitors (ICIs). Nevertheless, the impact of a specific class of antibiotics on ICIs therapy is still not known. The aim of this study was to analyse the influence of antibiotics on the clinical outcomes of non-small cell lung cancer (NSCLC) patients treated with ICIs and to compare the effects of fluoroquinolones vs. other broad-spectrum antibiotics. METHODS This retrospective cohort study (n = 340) analysed data from Chang Gung Research Database, which comprises work from seven medical institutions in Taiwan. Patients with NSCLC who received ICIs between January 2016 and March 2019 were evaluated. The data of patients who received antibiotics (ie fluoroquinolone) within 30 days prior to ICIs therapy were analysed. Overall survival (OS) was the goal of our study and was calculated from the time the ICIs therapy start. Survival analysis was estimated using the Kaplan-Meier and Cox statistics. RESULTS A total of 340 patients were identified for analysis. Of the 340 patients, only over one third (38%) of patients received antibiotics 30 days prior to ICI therapy. These patients exhibited a shorter OS compared with those not receiving antibiotics (median OS, 266 days vs. 455 days; hazard ratio (HR), 2.9; 95% confidence interval (CI), 1.1-8.1, p = 0.003). In this study, 127 out of 128 patients who were exposed to antibiotics had received at least one broad-spectrum antibiotic. We observed patients who had received fluoroquinolone had a shorter OS compared with those receiving other broad-spectrum antibiotics (median OS, 121 days vs. 370 days; HR, 1.582; 95% CI 1.007-2.841; p = 0.047). WHAT IS NEW AND CONCLUSION Antibiotic treatment, especially fluoroquinolone, prior to ICIs therapy was associated with poorer clinical efficacy in NSCLC patients. Antibiotics should not be withheld when there is a clear need for them despite the possibility of interfering with the microbiome, which may, in turn, adversely affect the ICI's effectiveness. However, one should consider avoiding the use of fluoroquinolones antibiotics.
Collapse
Affiliation(s)
- Po-Hsien Lu
- Department of Pharmacy, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Tzu-Cheng Tsai
- Department of Pharmacy, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - John Wen-Cheng Chang
- Division of Hematology-Oncology, Department of Internal Medicine, Chang Gung Memorial Hospital at Linkou, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Shin-Tarng Deng
- Department of Pharmacy, Chang Gung Memorial Hospital, Linkou, Taiwan
| | - Chi-Yuan Cheng
- Department of Pharmacy, Chang Gung Memorial Hospital, Linkou, Taiwan
| |
Collapse
|
61
|
Steed DB, Wang T, Raheja D, Waldman AD, Babiker A, Dhere T, Kraft CS, Woodworth MH. Gram-Negative Taxa and Antimicrobial Susceptibility after Fecal Microbiota Transplantation for Recurrent Clostridioides difficile Infection. mSphere 2020; 5:e00853-20. [PMID: 33055258 PMCID: PMC7565895 DOI: 10.1128/msphere.00853-20] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Accepted: 09/20/2020] [Indexed: 12/15/2022] Open
Abstract
Fecal microbiota transplantation (FMT) has promising applications in reducing multidrug-resistant organism (MDRO) colonization and antibiotic resistance (AR) gene abundance. However, data on clinical microbiology results after FMT are limited. We examined the changes in antimicrobial susceptibility profiles in patients with Gram-negative infections in the year before and the year after treatment with FMT for recurrent Clostridioides difficile infection (RCDI). We also examined whether a history of FMT changed health care provider behavior with respect to culture ordering and antibiotic prescription. Medical records for RCDI patients who underwent FMT at Emory University between July 2012 and March 2017 were reviewed retrospectively. FMT-treated patients with Gram-negative culture data in the 1-year period preceding and the 1-year period following FMT were included. Demographic and clinical data were abstracted, including CDI history, frequency of Gram-negative cultures, microbiological results, and antibiotic prescription in response to positive cultures in the period following FMT. Twelve patients were included in this case series. We pooled data from infections at all body sites and found a decrease in the number of total and Gram-negative cultures post-FMT. We compared susceptibility profiles across taxa given the potential for horizontal transmission of AR elements and observed increased susceptibility to nitrofurantoin, trimethoprim-sulfamethoxazole, and the aminoglycosides. FMT did not drastically influence health care provider ordering of bacterial cultures or antibiotic prescribing practices. We observed a reduction in Gram-negative cultures and a trend toward increased antimicrobial susceptibility. This study supports further investigation of FMT as a means of improving antimicrobial susceptibility.IMPORTANCE Fecal microbiota transplantation (FMT), which is highly efficacious in treating recurrent C. difficile infection (RCDI), has a promising application in decolonization of multidrug-resistant organisms, reduction of antibiotic resistance gene abundance, and restoration of healthy intestinal microbiota. However, data representing clinical microbiology results after FMT are limited. We sought to characterize the differences in culture positivity and antimicrobial susceptibility profiles in patients with Gram-negative infections in the year before and the year after FMT for RCDI. Drawing on prior studies that had demonstrated the success of FMT in eradicating extraintestinal infections and the occurrence of patient-level interspecies transfer of resistance elements, we employed an agnostic analytic approach of reviewing the data irrespective of body site or species. In a small RCDI population, we observed an improvement in the antimicrobial susceptibility profile of Gram-negative bacteria following FMT, which supports further study of FMT as a strategy to combat antibiotic resistance.
Collapse
Affiliation(s)
| | - Tiffany Wang
- Emory University School of Medicine, Atlanta, Georgia, USA
| | | | - Alex D Waldman
- Emory University School of Medicine, Atlanta, Georgia, USA
| | - Ahmed Babiker
- Emory University School of Medicine, Department of Pathology and Laboratory Medicine, Atlanta, Georgia, USA
| | - Tanvi Dhere
- Emory University School of Medicine, Department of Medicine, Division of Digestive Diseases, Atlanta, Georgia, USA
| | - Colleen S Kraft
- Emory University School of Medicine, Department of Pathology and Laboratory Medicine, Atlanta, Georgia, USA
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, Georgia, USA
| | - Michael H Woodworth
- Emory University School of Medicine, Department of Medicine, Division of Infectious Diseases, Atlanta, Georgia, USA
| |
Collapse
|
62
|
Elvers KT, Wilson VJ, Hammond A, Duncan L, Huntley AL, Hay AD, van der Werf ET. Antibiotic-induced changes in the human gut microbiota for the most commonly prescribed antibiotics in primary care in the UK: a systematic review. BMJ Open 2020; 10:e035677. [PMID: 32958481 PMCID: PMC7507860 DOI: 10.1136/bmjopen-2019-035677] [Citation(s) in RCA: 140] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 06/12/2020] [Accepted: 07/02/2020] [Indexed: 12/16/2022] Open
Abstract
OBJECTIVE The gut microbiota influences many aspects of human health. We investigated the magnitude and duration of changes in gut microbiota in response to antibiotics commonly prescribed in UK primary care. METHODS We searched MEDLINE, EMBASE and AMED, all years up to May 2020 including all study designs, collecting and analysing data on the effect of antibiotics prescribed for respiratory and urinary tract infections. We followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses and Cochrane standard methods. Risk of bias was evaluated using the Critical Appraisal Skills Programme. Narrative synthesis was used to report the themes emerging from the data. MAIN OUTCOME MEASURES Primary outcomes were antibiotic-induced changes in the composition and/or diversity of the gut microbiota. Secondary outcome was the time for the microbiota to return to baseline. RESULTS Thirty-one articles with low or unclear risk of bias showed that antibiotics impact the gut microbiota by causing rapid and diminished levels of bacterial diversity and changes in relative abundances. After cessation of treatment, gut bacteria recover, in most individuals, to their baseline state within a few weeks. Some studies suggested longer term effects from 2 to 6 months. Considerable heterogeneity in methodology makes the studies prone to biases and other confounding factors. Doxycycline was associated with a marked short-term decrease in Bifidobacterium diversity. Clarithromycin decreased the populations of Enterobacteria, and the anaerobic bacteria Bifidobacterium sp and Lactobacillus sp in numbers and diversity for up to 5 weeks. Phenoxymethylpenicillin, nitrofurantoin and amoxicillin had very little effect on the gut microbiome. CONCLUSIONS Despite substantial heterogeneity of the studies and small sample sizes, there is evidence that antibiotics commonly used in primary care influence the composition of the gastrointestinal microbiota. Larger population-based studies are needed to fully understand how antibiotics modulate the microbiota, and to determine if these are associated with (longer term) health consequences. PROSPERO REGISTRATION NUMBER CRD42017073750.
Collapse
Affiliation(s)
- Karen T Elvers
- Centre for Academic Primare Care & NIHR Health Protection Research Unit in Behavioural Science and Evaluation, Bristol Medical School, University of Bristol, Bristol, UK
| | - Victoria J Wilson
- Centre of Academic Primary Care, Bristol Medical School, University of Bristol, Bristol, UK
| | - Ashley Hammond
- Centre of Academic Primary Care, Bristol Medical School, University of Bristol, Bristol, UK
| | - Lorna Duncan
- Centre of Academic Primary Care, Bristol Medical School, University of Bristol, Bristol, UK
| | - Alyson L Huntley
- Centre of Academic Primary Care, Bristol Medical School, University of Bristol, Bristol, UK
| | - Alastair D Hay
- Centre of Academic Primary Care, Bristol Medical School, University of Bristol, Bristol, UK
| | - Esther T van der Werf
- Centre of Academic Primary Care, Bristol Medical School, University of Bristol, Bristol, UK
- Department of Integrative Medicine, Louis Bolk Institute, Bunnik, The Netherlands
| |
Collapse
|
63
|
Antibiotics versus no therapy in kidney transplant recipients with asymptomatic bacteriuria (BiRT): a pragmatic, multicentre, randomized, controlled trial. Clin Microbiol Infect 2020; 27:398-405. [PMID: 32919076 DOI: 10.1016/j.cmi.2020.09.005] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/30/2020] [Accepted: 09/01/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVES Many transplant physicians screen for and treat asymptomatic bacteriuria (ASB) during post-kidney-transplant surveillance. We investigated whether antibiotics are effective in reducing the occurrence of symptomatic urinary tract infection (UTI) in kidney transplant recipients with ASB. METHODS We performed this multicentre, randomized, open-label trial in kidney transplant recipients who had ASB and were ≥2 months post-transplantation. We randomly assigned participants to receive antibiotics or no therapy. The primary outcome was the incidence of symptomatic UTI over the subsequent 12 months. RESULTS One hundred and ninety-nine kidney transplant recipients with ASB were randomly assigned to antibiotics (100 participants) or no therapy (99 participants). There was no significant difference in the occurrence of symptomatic UTI between the antibiotic and no-therapy groups (27%, 27/100 versus 31%, 31/99; univariate Cox model: hazard ratio 0.83, 95%CI: 0.50-1.40; log-rank test: p 0.49). Over the 1-year study period, antibiotic use was five times higher in the antibiotic group than in the no-therapy group (30 antibiotic days/participant, interquartile range 20-41, versus 6, interquartile range 0-15, p < 0.001). Overall, 155/199 participants (78%) had at least one further episode of bacteriuria during the follow-up. Compared with the participant's baseline episode of ASB, the second episode of bacteriuria was more frequently caused by bacteria resistant to clinically relevant antibiotics (ciprofloxacin, cotrimoxazole, third-generation cephalosporin) in the antibiotic group than in the no-therapy group (18%, 13/72 versus 4%, 3/83, p 0.003). CONCLUSIONS Applying a screen-and-treat strategy for ASB does not reduce the occurrence of symptomatic UTI in kidney transplant recipients who are more than 2 months post-transplantation. Furthermore, this strategy increases antibiotic use and promotes the emergence of resistant organisms.
Collapse
|
64
|
Wuerz TC, Kassim SS, Atkins KE. Acquisition of extended-spectrum beta-lactamase-producing Enterobacteriaceae (ESBL-PE) carriage after exposure to systemic antimicrobials during travel: Systematic review and meta-analysis. Travel Med Infect Dis 2020; 37:101823. [PMID: 32755674 DOI: 10.1016/j.tmaid.2020.101823] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 03/23/2020] [Accepted: 07/09/2020] [Indexed: 02/01/2023]
Abstract
BACKGROUND International travel is an important risk factor for colonization with extended-spectrum beta-lactamase-producing Enterobacteriaceae (ESBL-PE). Antimicrobial use during travel likely amplifies this risk, yet to what extent, and whether it varies by antimicrobial class, has not been established. METHODS We conducted a systematic review that included prospective cohorts reporting both receipt of systemic antimicrobials and acquired ESBL-PE isolated from stool or rectum during international travel. We performed a random effects meta-analysis to estimate odds of acquiring ESBL-PE due to antimicrobials during travel, overall and by antimicrobial class. RESULTS Fifteen studies were included. The study population was mainly female travellers from high income countries recruited primarily from travel clinics. Participants travelled most frequently to Asia and Africa with 10% reporting antimicrobial use during travel. The combined odds ratio (OR) for ESBL-PE acquisition during travel was 2.37 for antimicrobial use overall (95% confidence interval [CI], 1.69 to 3.33), but there was substantial heterogeneity between studies. Fluoroquinolones were the antibiotic class associated with the highest combined OR of ESBL-PE acquisition, compared to no antimicrobial use (OR 4.68, 95% CI, 2.34 to 9.37). CONCLUSIONS The risk of ESBL-PE colonization during travel is increased substantially with exposure to antimicrobials, especially fluoroquinolones. While a small proportion of colonized individuals will develop a resistant infection, there remains the potential for onward spread among returning travellers. Public health efforts to decrease inappropriate antimicrobial usage during travel are warranted.
Collapse
Affiliation(s)
- Terence C Wuerz
- St. Boniface General Hospital, Winnipeg, Manitoba, Canada; Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; Department of Community Health Sciences, University of Manitoba, Winnipeg, Manitoba, Canada.
| | - Sameer S Kassim
- Department of Family Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Katherine E Atkins
- Centre for Mathematical Modelling of Infectious Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom; Centre for Global Health Research, Usher Institute for Population Health Sciences and Informatics, Edinburgh Medical School, The University of Edinburgh, UK
| |
Collapse
|
65
|
Huttner A, Wijma RA, Stewardson AJ, Olearo F, Von Dach E, Harbarth S, Brüggemann RJM, Mouton JW, Muller AE. The pharmacokinetics of nitrofurantoin in healthy female volunteers: a randomized crossover study. J Antimicrob Chemother 2020; 74:1656-1661. [PMID: 30859184 DOI: 10.1093/jac/dkz095] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 02/14/2019] [Accepted: 02/17/2019] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Use of nitrofurantoin has increased significantly since its recent repositioning as a first-line agent for uncomplicated cystitis by multiple guidelines. However, current dosing regimens were developed in an era before robust pharmacokinetic testing and may not be optimal. Furthermore, formulations have been modified over the years. OBJECTIVES To reassess the plasma and urinary pharmacokinetic profile of macrocrystalline nitrofurantoin in two commonly used dosing regimens. METHODS In this open-label, randomized crossover pharmacokinetic trial, 12 healthy adult female volunteers were randomized to receive oral nitrofurantoin 100 mg q8h on days 1 and 2 and, after a washout period, 50 mg q6h on days 30 and 31, or the same dosing schemes in reversed order. Urine and blood were collected at steady state and analysed by UPLC. Pharmacokinetic analysis was performed by WinNonlin. RESULTS Plasma peak concentrations were low (mean 0.33 mg/L, SD 0.08, and 0.69 mg/L, SD 0.35, after 50 and 100 mg, respectively) and dose dependent. The AUC0-24 was higher (6.49 versus 4.43 mg·h/L, P = 0.021) for the 100 mg q8h dosing regimen, but the dose-normalized AUC was similar for the two regimens. In contrast, urinary concentrations were dose independent: increasing the nitrofurantoin dose delayed the time to peak urinary concentration, while steady-state AUC0-24 values remained unchanged (943.49 and 855.95 mg·h/L at 50 mg q6h and 100 mg q8h, respectively). CONCLUSIONS Plasma concentrations were relatively low and dose dependent. The dose-independent urinary concentrations suggest that excretion of nitrofurantoin into the urine is saturable. Pharmacodynamic studies are urgently required to determine the impact of these findings.
Collapse
Affiliation(s)
- Angela Huttner
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Rixt A Wijma
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Andrew J Stewardson
- Department of Infectious Disease, Alfred Health and Central Clinical School, Monash University, Melbourne, Australia
| | - Flaminia Olearo
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Elodie Von Dach
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland.,Clinical Trials Unit, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Stephan Harbarth
- Division of Infectious Diseases, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland.,Infection Control Program, Geneva University Hospitals and Faculty of Medicine, Geneva, Switzerland
| | - Roger J M Brüggemann
- Department of Pharmacy and Center of Expertise in Mycology, Radboud University, Nijmegen, The Netherlands
| | - Johan W Mouton
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre, Rotterdam, The Netherlands
| | - Anouk E Muller
- Department of Medical Microbiology and Infectious Diseases, Erasmus University Medical Centre, Rotterdam, The Netherlands.,Department of Medical Microbiology, Haaglanden Medical Centre, The Hague, The Netherlands
| |
Collapse
|
66
|
You L, Ying C, Liu K, Zhang X, Lin D, Yin D, Zhang J, Xu P. Changes in the fecal microbiome of the Yangtze finless porpoise during a short-term therapeutic treatment. Open Life Sci 2020; 15:296-310. [PMID: 33817218 PMCID: PMC7988435 DOI: 10.1515/biol-2020-0032] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 02/15/2020] [Accepted: 02/18/2020] [Indexed: 01/15/2023] Open
Abstract
The fecal microbiome is an integral part of aquatic mammals, like an inner organ. But we know very little about this inner organ of the threatened aquatic species, Yangtze finless porpoise (YFP). Four YFPs were placed into a purse seine for skin ulceration treatment, and this opportunity was taken to nurse the animals closer. In particular, we collected the feces of the YFPs before and after the paired healing and therapeutic treatment, along with samples of their fish diet and water habitat, to explore the changes in their fecal microbiome. Firmicutes (20.9–96.1%), Proteobacteria (3.8–78.7%), Actinobacteria (0.1–35.0%) and Tenericutes (0.8–17.1%) were the most dominant phyla present in the feces. The proportion of Proteobacteria and Actinobacteria increased after the treatment. Firmicutes showed a significant decrease, and most potential pathogens were absent, which reflected the administration of ciprofloxacin hydrochloride. Moreover, environmental shifts can also contribute to changes in the fecal microbiome. These results indicate that certain microbial interactions can be affected by environmental shifts, dietary changes and health-care treatments, which can also help maintain the internal environment of YFPs. These findings will inform the future enhanced protection and management of endangered YFPs and other vulnerable aquatic animals.
Collapse
Affiliation(s)
- Lei You
- Wuxi Fisheries College, Nanjing Agricultural University, Wuxi, China
| | - Congping Ying
- Scientific Observing and Experimental Station of Fishery Resources and Environment in the Lower Reaches of the Changjiang River, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, CAFS, Binhu District, Wuxi, Jiangsu, China
| | - Kai Liu
- Scientific Observing and Experimental Station of Fishery Resources and Environment in the Lower Reaches of the Changjiang River, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, CAFS, Binhu District, Wuxi, Jiangsu, China
| | - Xizhao Zhang
- Scientific Observing and Experimental Station of Fishery Resources and Environment in the Lower Reaches of the Changjiang River, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, CAFS, Binhu District, Wuxi, Jiangsu, China
| | - Danqing Lin
- Scientific Observing and Experimental Station of Fishery Resources and Environment in the Lower Reaches of the Changjiang River, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, CAFS, Binhu District, Wuxi, Jiangsu, China
| | - Denghua Yin
- Scientific Observing and Experimental Station of Fishery Resources and Environment in the Lower Reaches of the Changjiang River, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, CAFS, Binhu District, Wuxi, Jiangsu, China
| | - Jialu Zhang
- Scientific Observing and Experimental Station of Fishery Resources and Environment in the Lower Reaches of the Changjiang River, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, CAFS, Binhu District, Wuxi, Jiangsu, China
| | - Pao Xu
- Scientific Observing and Experimental Station of Fishery Resources and Environment in the Lower Reaches of the Changjiang River, Ministry of Agriculture and Rural Affairs, Freshwater Fisheries Research Center, CAFS, Binhu District, Wuxi, Jiangsu, China
| |
Collapse
|
67
|
Papanicolas LE, Warner M, Wesselingh SL, Rogers GB. Protect commensal gut bacteria to improve antimicrobial stewardship. Clin Microbiol Infect 2020; 26:814-815. [PMID: 32234452 DOI: 10.1016/j.cmi.2020.03.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Revised: 03/18/2020] [Accepted: 03/21/2020] [Indexed: 01/12/2023]
Affiliation(s)
- L E Papanicolas
- Microbiome & Host Health Programme, The South Australian Health and Medical Research Institute, Adelaide, SA, Australia; The SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia
| | - M Warner
- Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - S L Wesselingh
- The South Australian Health and Medical Research Institute, Adelaide, SA, Australia
| | - G B Rogers
- Microbiome & Host Health Programme, The South Australian Health and Medical Research Institute, Adelaide, SA, Australia; The SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, SA, Australia.
| |
Collapse
|
68
|
Antibacterial Prodrugs to Overcome Bacterial Resistance. Molecules 2020; 25:molecules25071543. [PMID: 32231026 PMCID: PMC7180472 DOI: 10.3390/molecules25071543] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 12/17/2022] Open
Abstract
Bacterial resistance to present antibiotics is emerging at a high pace that makes the development of new treatments a must. At the same time, the development of novel antibiotics for resistant bacteria is a slow-paced process. Amid the massive need for new drug treatments to combat resistance, time and effort preserving approaches, like the prodrug approach, are most needed. Prodrugs are pharmacologically inactive entities of active drugs that undergo biotransformation before eliciting their pharmacological effects. A prodrug strategy can be used to revive drugs discarded due to a lack of appropriate pharmacokinetic and drug-like properties, or high host toxicity. A special advantage of the use of the prodrug approach in the era of bacterial resistance is targeting resistant bacteria by developing prodrugs that require bacterium-specific enzymes to release the active drug. In this article, we review the up-to-date implementation of prodrugs to develop medications that are active against drug-resistant bacteria.
Collapse
|
69
|
Tedijanto C, Grad YH, Lipsitch M. Potential impact of outpatient stewardship interventions on antibiotic exposures of common bacterial pathogens. eLife 2020; 9:52307. [PMID: 32022685 PMCID: PMC7025820 DOI: 10.7554/elife.52307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2019] [Accepted: 01/28/2020] [Indexed: 01/30/2023] Open
Abstract
The relationship between antibiotic stewardship and population levels of antibiotic resistance remains unclear. In order to better understand shifts in selective pressure due to stewardship, we use publicly available data to estimate the effect of changes in prescribing on exposures to frequently used antibiotics experienced by potentially pathogenic bacteria that are asymptomatically colonizing the microbiome. We quantify this impact under four hypothetical stewardship strategies. In one scenario, we estimate that elimination of all unnecessary outpatient antibiotic use could avert 6% to 48% (IQR: 17% to 31%) of exposures across pairwise combinations of sixteen common antibiotics and nine bacterial pathogens. All scenarios demonstrate that stewardship interventions, facilitated by changes in clinician behavior and improved diagnostics, have the opportunity to broadly reduce antibiotic exposures across a range of potential pathogens. Concurrent approaches, such as vaccines aiming to reduce infection incidence, are needed to further decrease exposures occurring in ‘necessary’ contexts.
Collapse
Affiliation(s)
- Christine Tedijanto
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, United States
| | - Yonatan H Grad
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, United States.,Division of Infectious Diseases, Brigham and Women's Hospital, Harvard Medical School, Boston, United States
| | - Marc Lipsitch
- Center for Communicable Disease Dynamics, Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, United States.,Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, United States
| |
Collapse
|
70
|
Meng T, Zhang S, Wang D, Zhang H, Song Z, Li S, Chen M, Tang C. Association between antibiotic use and the risk of rheumatoid arthritis: A protocol for a systematic review. Medicine (Baltimore) 2020; 99:e19155. [PMID: 32118718 PMCID: PMC7478554 DOI: 10.1097/md.0000000000019155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 01/14/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND The potential association between antibiotic use and the risk of rheumatoid arthritis (RA) has drawn significant attention from clinicians and researchers in recent years due to the wild usage of antibiotic. This study aimed to perform a systematic review and meta-analysis of the literature to determine if antibiotic use is associated with an increased risk of RA, so as to provide an important reference for clinical decision-making. METHODS Case-control and nest case-control studies of assessing whether antibiotic use is associated with the onset of RA will be identified in searches of 4 databases from their inception to August 2019. All data were assessed and extracted by 2 authors independently. The Newcastle-Ottawa scale was used to assess the quality of the selected studies. Manager Software 5.3 from Cochrane Collaboration (London, UK) and Stata 15.1 (Stata Corp, College Station, TX) will be used to conduct meta-analysis, determining pooled odds ratios and evaluating heterogeneity between studies. RESULT The results of this systemic review and meta-analysis will be submitted to a recognized journal for publication. CONCLUSION This systemic review and meta-analysis will determine if antibiotic use is associated with an increased risk of RA. We hope this study can make a definitive conclusion for the association.
Collapse
Affiliation(s)
- Tingting Meng
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | - Shibin Zhang
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | - Dawei Wang
- Shunde Hospital of Guangzhou University of Chinese Medicine, Shunde District Hospital of Chinese Medicine of Foshan City, Foshan, Guangdong
| | - Haijun Zhang
- Department of Orthopaedics, Gansu Second Provincial People's Hospital
| | - Zhongyang Song
- Clinical Medical College of Traditional Chinese Medicine, Gansu University of Chinese medicine, Lanzhou, Gansu
| | - Shengdong Li
- Department of Rheumatology, The Second Hospital of Yinzhou, Ninbo Zhejiang, China
| | - Min Chen
- Faculty of Chinese Medicine, Macau University of Science and Technology, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Avenida Wai Long, Taipa, Macau
| | - Chunzhi Tang
- Clinical Medical College of Acupuncture, Moxibustion and Rehabilitation, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
71
|
Papanicolas LE, Gordon DL, Wesselingh SL, Rogers GB. Improving Risk-Benefit in Faecal Transplantation through Microbiome Screening. Trends Microbiol 2020; 28:331-339. [PMID: 31952909 DOI: 10.1016/j.tim.2019.12.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 11/21/2019] [Accepted: 12/09/2019] [Indexed: 12/13/2022]
Abstract
Faecal microbiota transplantation (FMT) has been shown to be effective in the treatment of a growing number of conditions, and its clinical use continues to rise. However, recent cases of antibiotic-resistant pathogen transmission through FMT, resulting in at least one case of fatal sepsis, highlight the need to reevaluate current donor screening practices. Commensal gut microbes profoundly influence infection risk but are not routinely assessed in donor stool. Extending the assessment of donor material beyond pathogen populations to include the composition and structure of the wider faecal microbiota has the potential to reduce infectious complications in FMT recipients.
Collapse
Affiliation(s)
- Lito E Papanicolas
- Microbiome and Host Health Programme, the South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia; The SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, South Australia, Australia
| | - David L Gordon
- Department of Microbiology and Infectious Diseases, Flinders University, Adelaide, South Australia, Australia
| | | | - Geraint B Rogers
- Microbiome and Host Health Programme, the South Australian Health and Medical Research Institute (SAHMRI), Adelaide, South Australia, Australia; The SAHMRI Microbiome Research Laboratory, School of Medicine, Flinders University, Adelaide, South Australia, Australia.
| |
Collapse
|
72
|
Gajdács M, Spengler G. The Role of Drug Repurposing in the Development of Novel Antimicrobial Drugs: Non-Antibiotic Pharmacological Agents as Quorum Sensing-Inhibitors. Antibiotics (Basel) 2019; 8:E270. [PMID: 31861228 PMCID: PMC6963710 DOI: 10.3390/antibiotics8040270] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 02/06/2023] Open
Abstract
Background: The emergence of multidrug-resistant organisms (MDROs) is a global public health issue, severely hindering clinicians in administering appropriate antimicrobial therapy. Drug repurposing is a drug development strategy, during which new pharmacological applications are identified for already approved drugs. From the viewpoint of the development of virulence inhibitors, inhibition of quorum sensing (QS) is a promising route because various important features in bacterial physiology and virulence are mediated by QS-dependent gene expression. Methods: Forty-five pharmacological agents, encompassing a wide variety of different chemical structures and mechanisms of action, were tested during our experiments. The antibacterial activity of the compounds was tested using the broth microdilution method. Screening and semi-quantitative assessment of QS-inhibition by the compounds was performed using QS-signal molecule-producing and indicator strains. Results: Fourteen pharmaceutical agents showed antibacterial activity in the tested concentration range, while eight drugs (namely 5-fluorouracil, metamizole-sodium, cisplatin, methotrexate, bleomycin, promethazine, chlorpromazine, and thioridazine) showed dose-dependent QS-inhibitory activity in the in vitro model systems applied during the experiments. Conclusions: Virulence inhibitors represent an attractive alternative strategy to combat bacterial pathogens more efficiently. Some of the tested compounds could be considered potential QS-inhibitory agents, warranting further experiments involving additional model systems to establish the extent of their efficacy.
Collapse
Affiliation(s)
- Márió Gajdács
- Department of Medical Microbiology and Immunobiology, Faculty of Medicine, University of Szeged, Dóm tér 10, 6720 Szeged, Hungary
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös utca 6, 6720 Szeged, Hungary;
| | - Gabriella Spengler
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, Eötvös utca 6, 6720 Szeged, Hungary;
| |
Collapse
|
73
|
Dempsey JL, Little M, Cui JY. Gut microbiome: An intermediary to neurotoxicity. Neurotoxicology 2019; 75:41-69. [PMID: 31454513 PMCID: PMC7703666 DOI: 10.1016/j.neuro.2019.08.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Revised: 08/04/2019] [Accepted: 08/16/2019] [Indexed: 12/12/2022]
Abstract
There is growing recognition that the gut microbiome is an important regulator for neurological functions. This review provides a summary on the role of gut microbiota in various neurological disorders including neurotoxicity induced by environmental stressors such as drugs, environmental contaminants, and dietary factors. We propose that the gut microbiome remotely senses and regulates CNS signaling through the following mechanisms: 1) intestinal bacteria-mediated biotransformation of neurotoxicants that alters the neuro-reactivity of the parent compounds; 2) altered production of neuro-reactive microbial metabolites following exposure to certain environmental stressors; 3) bi-directional communication within the gut-brain axis to alter the intestinal barrier integrity; and 4) regulation of mucosal immune function. Distinct microbial metabolites may enter systemic circulation and epigenetically reprogram the expression of host genes in the CNS, regulating neuroinflammation, cell survival, or cell death. We will also review the current tools for the study of the gut-brain axis and provide some suggestions to move this field forward in the future.
Collapse
Affiliation(s)
- Joseph L Dempsey
- Department of Environmental and Occupational Health Sciences, University of Washington, United States
| | - Mallory Little
- Department of Environmental and Occupational Health Sciences, University of Washington, United States
| | - Julia Yue Cui
- Department of Environmental and Occupational Health Sciences, University of Washington, United States.
| |
Collapse
|
74
|
Tepekule B, Abel Zur Wiesch P, Kouyos RD, Bonhoeffer S. Quantifying the impact of treatment history on plasmid-mediated resistance evolution in human gut microbiota. Proc Natl Acad Sci U S A 2019; 116:23106-23116. [PMID: 31666328 PMCID: PMC6859334 DOI: 10.1073/pnas.1912188116] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
To understand how antibiotic use affects the risk of a resistant infection, we present a computational model of the population dynamics of gut microbiota including antibiotic resistance-conferring plasmids. We then describe how this model is parameterized based on published microbiota data. Finally, we investigate how treatment history affects the prevalence of resistance among opportunistic enterobacterial pathogens. We simulate treatment histories and identify which properties of prior antibiotic exposure are most influential in determining the prevalence of resistance. We find that resistance prevalence can be predicted by 3 properties, namely the total days of drug exposure, the duration of the drug-free period after last treatment, and the center of mass of the treatment pattern. Overall this work provides a framework for capturing the role of the microbiome in the selection of antibiotic resistance and highlights the role of treatment history for the prevalence of resistance.
Collapse
Affiliation(s)
- Burcu Tepekule
- Department of Environmental Systems Science, Eidgenössische Technische Hochschule Zurich, 8092 Zurich, Switzerland;
| | - Pia Abel Zur Wiesch
- Department of Pharmacy, Faculty of Health Sciences, UiT-The Arctic University of Norway, 9037 Tromsø, Norway
- Centre for Molecular Medicine Norway, 0318 Oslo, Norway
| | - Roger D Kouyos
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, 8091 Zurich, Switzerland
- Institute of Medical Virology, University of Zurich, 8057 Zurich, Switzerland
| | - Sebastian Bonhoeffer
- Department of Environmental Systems Science, Eidgenössische Technische Hochschule Zurich, 8092 Zurich, Switzerland
| |
Collapse
|
75
|
Kim H, Lee JE, Hong SH, Lee MA, Kang JH, Kim IH. The effect of antibiotics on the clinical outcomes of patients with solid cancers undergoing immune checkpoint inhibitor treatment: a retrospective study. BMC Cancer 2019; 19:1100. [PMID: 31718585 PMCID: PMC6852740 DOI: 10.1186/s12885-019-6267-z] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 10/15/2019] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND This study aimed to assess the effect of antibiotics on the clinical outcomes of patients with solid cancers undergoing treatment with immune checkpoint inhibitors (ICIs). METHODS The medical records of 234 patients treated with ICIs for any type of solid cancer between February 2012 and May 2018 at the Seoul St. Mary's Hospital were retrospectively reviewed. The data of patients who received antibiotics within 60 days before the initiation of ICI treatment were analyzed. The patients' responses to ICI treatment and their survival were evaluated. RESULTS Non-small-cell lung carcinoma was the most common type of cancer. About half of the patients were treated with nivolumab (51.9%), and cephalosporin (35.2%) was the most commonly used class of antibiotics. The total objective response rate was 21%. Antibiotics use was associated with a decreased objective response (odds ratio 0.466, 95% confidence interval [CI] 0.225-0.968, p = 0.040). The antibiotics group exhibited shorter progression-free survival (PFS) and overall survival (OS) than the no antibiotics group (median PFS: 2 months vs. 4 months, p < 0.001; median OS: 5 months vs. 17 months, p < 0.001). In the multivariate analysis, antibiotics use was a significant predictor of patient survival (PFS: hazard ratio [HR] 1.715, 95% CI 1.264-2.326, p = 0.001; OS: HR 1.785, 95% CI 1.265-2.519, p = 0.001). CONCLUSIONS The use of antibiotics may affect the clinical outcomes of patients with solid cancers treated with ICIs. Careful prescription of antibiotics is warranted in candidates who are scheduled for ICI treatment. TRIAL REGISTRATION Not applicable (retrospective study).
Collapse
Affiliation(s)
- Hyunho Kim
- Division of Medical Oncology, Department of Internal Medicine, The Catholic University of Korea, St. Vincent's Hospital, Suwon, Republic of Korea
| | - Ji Eun Lee
- Division of Medical Oncology, Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea
| | - Sook Hee Hong
- Division of Medical Oncology, Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea
| | - Myung Ah Lee
- Division of Medical Oncology, Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea
| | - Jin Hyoung Kang
- Division of Medical Oncology, Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, The Catholic University of Korea, Seoul St. Mary's Hospital, Seoul, Republic of Korea. .,Department of Internal Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea College of Medicine, 222 Banpo-daero, Seocho-gu, Seoul, 137-701, Korea.
| |
Collapse
|
76
|
Jurburg SD, Cornelissen JJBWJ, de Boer P, Smits MA, Rebel JMJ. Successional Dynamics in the Gut Microbiome Determine the Success of Clostridium difficile Infection in Adult Pig Models. Front Cell Infect Microbiol 2019; 9:271. [PMID: 31448240 PMCID: PMC6691177 DOI: 10.3389/fcimb.2019.00271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2019] [Accepted: 07/15/2019] [Indexed: 01/27/2023] Open
Abstract
Clostridium difficile infections (CDI) are a major cause of antibiotic-associated diarrhea. It is hypothesized that CDI develops due to the antibiotic-induced disruption of the intestinal microbial community structure, which allows C. difficile to flourish. Here, we pre-treated weaned pigs with the antibiotics Clindamycin or Ciprofloxacin for 1 day, and subsequently inoculated them with a human and pig enteropathogenic C. difficile strain 078 spores. Body temperature, clinical signs of disease, and the fecal microbiome were monitored daily for 15 days. Clindamycin had a stronger effect on the pigs than Ciprofloxacin, resulting in drastic shifts in the fecal microbiome, decreases in microbial diversity and significant increases in body temperature, even in the absence of C. difficile. Fecal shedding of C. difficile was detectable for 3 and 9 days in Ciprofloxacin and Clindamycin treated pigs inoculated with C. difficile, respectively, and in both cases decreased cell proliferation rates were detected in colon tissue. The timing of C. difficile shedding coincided with the decrease in a large cluster of Firmicutes following Clindamycin treatment, a pattern which was also independent of C. difficile inoculation. The observed community patterns suggest that successional dynamics following antibiotic treatment facilitate C. difficile establishment. The similarities between the microbiome responses observed in our study and those previously reported in CDI-infected humans further support the utility of adult pigs as models for the study of CDI.
Collapse
Affiliation(s)
- Stephanie D Jurburg
- Wageningen Bioveterinary Research, Lelystad, Netherlands.,iDiv - German Centre for Integrative Biodiversity Research, Leipzig, Germany
| | | | | | - Mari A Smits
- Wageningen Bioveterinary Research, Lelystad, Netherlands
| | - Johanna M J Rebel
- Wageningen Bioveterinary Research, Lelystad, Netherlands.,Wageningen Livestock Research, Wageningen, Netherlands
| |
Collapse
|
77
|
Morley VJ, Woods RJ, Read AF. Bystander Selection for Antimicrobial Resistance: Implications for Patient Health. Trends Microbiol 2019; 27:864-877. [PMID: 31288975 PMCID: PMC7079199 DOI: 10.1016/j.tim.2019.06.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/29/2019] [Accepted: 06/13/2019] [Indexed: 12/15/2022]
Abstract
Antimicrobial therapy promotes resistance emergence in target infections and in off-target microbiota. Off-target resistance emergence threatens patient health when off-target populations are a source of future infections, as they are for many important drug-resistant pathogens. However, the health risks of antimicrobial exposure in off-target populations remain largely unquantified, making rational antibiotic stewardship challenging. Here, we discuss the contribution of bystander antimicrobial exposure to the resistance crisis, the implications for antimicrobial stewardship, and some novel opportunities to limit resistance evolution while treating target pathogens.
Collapse
Affiliation(s)
- Valerie J Morley
- Center for Infectious Disease Dynamics, Departments of Biology and Entomology, The Pennsylvania State University, University Park, PA, USA.
| | - Robert J Woods
- Division of Infectious Diseases, Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Andrew F Read
- Center for Infectious Disease Dynamics, Departments of Biology and Entomology, The Pennsylvania State University, University Park, PA, USA; Huck Institutes for the Life Sciences, The Pennsylvania State University, University Park, PA, USA
| |
Collapse
|
78
|
Antibiotic treatment and flares of rheumatoid arthritis: a self-controlled case series study analysis using CPRD GOLD. Sci Rep 2019; 9:8941. [PMID: 31222078 PMCID: PMC6586671 DOI: 10.1038/s41598-019-45435-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 06/06/2019] [Indexed: 12/27/2022] Open
Abstract
There is emerging evidence of the impact of infections on rheumatoid arthritis pathogenesis and flares. We aimed to study the association between antibiotic use (and timing of use), and the occurrence of flares in patients with RA. We nested a self-controlled case series (SCCS) of patients who have RA flares within a newly diagnosed RA cohort (n = 31,992) from the UK Clinical Practice Research Datalink (CPRD) GOLD dataset. We determined associations between exposure to antibiotics (beta-lactam, imidazole, macrolide, nitrofurantoin, quinolone, sulphonamide and trimethoprim, and tetracycline) and the occurrence of RA flares. Conditional fixed-effects Poisson regression models were used to determine incidence rate ratios (IRR), offset by the natural logarithm of risk periods. A total of 1,192 (3.7%) of RA subjects had one or more flare/s during the study period, and were therefore included. Use of sulphonamide and trimethoprim was associated with an increased risk of RA flare at 29–90 days (IRR 1.71, CI 1.12–2.59, p = 0.012); 91–183 days (IRR 1.57, CI 1.06–2.33, p = 0.025); and 184–365 days (IRR 1.44, CI 1.03–2.02, p = 0.033) after commencement of antibiotic treatment. No other antibiotic group/s appear associated with RA flare/s risk. Usage of sulphonamide and trimethoprim antibiotics, is associated with a 70% increased risk of RA flare at 1–3 months, which decreases but remains significant up to 12 months after treatment. We hypothesise that the delayed onset of RA flares after specific antibiotics is mediated through the gut or urinary microbiomes. Further epidemiological and mechanistic research is needed to determine the role of infections in RA.
Collapse
|
79
|
Evans LE, Krishna A, Ma Y, Webb TE, Marshall DC, Tooke CL, Spencer J, Clarke TB, Armstrong A, Edwards AM. Exploitation of Antibiotic Resistance as a Novel Drug Target: Development of a β-Lactamase-Activated Antibacterial Prodrug. J Med Chem 2019; 62:4411-4425. [PMID: 31009558 PMCID: PMC6511942 DOI: 10.1021/acs.jmedchem.8b01923] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Indexed: 02/08/2023]
Abstract
Expression of β-lactamase is the single most prevalent determinant of antibiotic resistance, rendering bacteria resistant to β-lactam antibiotics. In this article, we describe the development of an antibiotic prodrug that combines ciprofloxacin with a β-lactamase-cleavable motif. The prodrug is only bactericidal after activation by β-lactamase. Bactericidal activity comparable to ciprofloxacin is demonstrated against clinically relevant E. coli isolates expressing diverse β-lactamases; bactericidal activity was not observed in strains without β-lactamase. These findings demonstrate that it is possible to exploit antibiotic resistance to selectively target β-lactamase-producing bacteria using our prodrug approach, without adversely affecting bacteria that do not produce β-lactamase. This paves the way for selective targeting of drug-resistant pathogens without disrupting or selecting for resistance within the microbiota, reducing the rate of secondary infections and subsequent antibiotic use.
Collapse
Affiliation(s)
- Lindsay E. Evans
- MRC
Centre for Molecular Bacteriology and Infection, Imperial College London, SW7 2AZ London, United Kingdom
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, W12 0BZ London, United Kingdom
| | - Aishwarya Krishna
- MRC
Centre for Molecular Bacteriology and Infection, Imperial College London, SW7 2AZ London, United Kingdom
| | - Yajing Ma
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, W12 0BZ London, United Kingdom
| | - Thomas E. Webb
- Department
of Medicine, Imperial College London, SW7 2AZ London, United Kingdom
| | - Dominic C. Marshall
- Department
of Medicine, Imperial College London, SW7 2AZ London, United Kingdom
| | - Catherine L. Tooke
- School
of Cellular and Molecular Medicine, University
of Bristol, Biomedical Sciences Building, University Walk, BS8 1TD Bristol, United Kingdom
| | - James Spencer
- School
of Cellular and Molecular Medicine, University
of Bristol, Biomedical Sciences Building, University Walk, BS8 1TD Bristol, United Kingdom
| | - Thomas B. Clarke
- MRC
Centre for Molecular Bacteriology and Infection, Imperial College London, SW7 2AZ London, United Kingdom
| | - Alan Armstrong
- Department
of Chemistry, Molecular Sciences Research Hub, Imperial College London, W12 0BZ London, United Kingdom
| | - Andrew M. Edwards
- MRC
Centre for Molecular Bacteriology and Infection, Imperial College London, SW7 2AZ London, United Kingdom
| |
Collapse
|
80
|
Dynamics of Resistance Plasmids in Extended-Spectrum-β-Lactamase-Producing Enterobacteriaceae during Postinfection Colonization. Antimicrob Agents Chemother 2019; 63:AAC.02201-18. [PMID: 30745391 DOI: 10.1128/aac.02201-18] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 02/05/2019] [Indexed: 01/01/2023] Open
Abstract
Extended-spectrum β-lactamase-producing Enterobacteriaceae (EPE) are a major cause of bloodstream infections, and the colonization rate of EPE in the gut microbiota of individuals lacking prior hospitalization or comorbidities is increasing. In this study, we performed an in-depth investigation of the temporal dynamics of EPE and their plasmids during one year by collecting fecal samples from three patients initially seeking medical care for urinary tract infections. In two of the patients, the same strain that caused the urinary tract infection (UTI) was found at all consecutive samplings from the gut microbiota, and no other EPEs were detected, while in the third patient the UTI strain was only found in the initial UTI sample. Instead, this patient presented a complex situation where a mixed microbiota of different EPE strain types, including three different E. coli ST131 variants, as well as different bacterial species, was identified over the course of the study. Different plasmid dynamics were displayed in each of the patients, including the spread of plasmids between different strain types over time and the transposition of bla CTX-M-15 from the chromosome to a plasmid, followed by subsequent loss through homologous recombination. Small cryptic plasmids were found in all isolates from all patients, and they appear to move frequently between different strains in the microbiota. In conclusion, we could demonstrate an extensive variation of EPE strain types, plasmid composition, rearrangements, and horizontal gene transfer of genetic material illustrating the high dynamics nature and interactive environment of the gut microbiota during post-UTI carriage.
Collapse
|
81
|
McConnell MJ. Where are we with monoclonal antibodies for multidrug-resistant infections? Drug Discov Today 2019; 24:1132-1138. [PMID: 30853568 DOI: 10.1016/j.drudis.2019.03.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/21/2019] [Accepted: 03/01/2019] [Indexed: 01/17/2023]
Abstract
Widespread antibiotic resistance threatens the continued efficacy of antimicrobial therapy based on small-molecule antibiotics. Infections caused by multidrug-resistant Gram-negative bacteria are particularly worrisome owing to the lack of antimicrobials retaining sufficient activity against these microorganisms. Despite the explosion in monoclonal antibody therapies that have been developed for oncologic and rheumatic indications, only three antibacterial monoclonal antibodies have been approved for clinical use. In the present review, the therapeutic potential of this drug class for treating multidrug-resistant infections is discussed, and considerations for the development of antibacterial monoclonal antibodies are presented. Finally, the state of development of monoclonal antibody therapies for some of the most problematic multidrug-resistant Gram-negative infections is summarized.
Collapse
Affiliation(s)
- Michael J McConnell
- Antimicrobial Resistance and Hospital Acquired Infections Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain.
| |
Collapse
|
82
|
Lasota S, Stephan I, Horn MA, Otto W, Noll M. Copper in Wood Preservatives Delayed Wood Decomposition and Shifted Soil Fungal but Not Bacterial Community Composition. Appl Environ Microbiol 2019; 85:e02391-18. [PMID: 30530712 PMCID: PMC6365821 DOI: 10.1128/aem.02391-18] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/01/2018] [Indexed: 12/17/2022] Open
Abstract
Copper-based fungicides are routinely used for wood and plant protection, which can lead to an enrichment of copper-tolerant microbial communities in soil. To investigate the effect of such wood preservatives on the soil fungal and bacterial community compositions, five different vineyard and fruit-growing soil environments were evaluated using incubation studies over time. Pine sapwood specimens were impregnated with either water or different biocide treatment solutions containing a mixture of copper, triazoles, and quaternary ammonium compounds (CuTriQAC), a mixture of triazoles and quaternary ammonium compounds (TriQAC), or copper alone (Cu). Specimens were incubated in soil from each sample site for 8, 16, 24, and 32 weeks. The effects of preservative treatment on the modulus of elasticity (MOE) of the wood specimens and on the soil fungal as well as bacterial community composition at the soil-wood interface were assessed by quantitative PCR and amplicon sequencing of the fungal internal transcribed spacer (ITS) region and bacterial 16S rRNA gene. Specimens impregnated with CuTriQAC and Cu showed decreased MOE and reduced fungal and bacterial copy numbers over time compared to those impregnated with water and TriQAC. Fungal but not bacterial community composition was significantly affected by wood preservative treatment. The relative abundance of members of the family Trichocomaceae compared to other genera increased in the presence of the Cu and CuTriQAC treatments at three sites, suggesting these to be Cu-tolerant fungi. In conclusion, the copper-containing treatments resulted in marginally increased MOE, lowered microbial gene copy numbers compared to those in the TriQAC and water treatments, and thus enhanced wood protection against soil microbial wood degradation.IMPORTANCE Copper-containing rather than TRIQAC formulations are efficient wood preservatives irrespective of the origin and composition of the soil microbial communities. However, some fungi appear to be naturally insensitive to copper and should be the focus of future wood preservative formulations to facilitate the life span of wooden construction in contact with soil while also minimizing the overall environmental impact.
Collapse
Affiliation(s)
- Sandra Lasota
- Institute for Bioanalysis, Department of Applied Sciences, Coburg University of Applied Sciences and Arts, Coburg, Germany
| | - Ina Stephan
- Bundesanstalt für Materialforschung und -prüfung, Division 4.1, Biodeterioration and Reference Organisms, Berlin, Germany
| | - Marcus A Horn
- Institute of Microbiology, Leibniz University of Hannover, Hannover, Germany
| | - Wolfgang Otto
- Institute of Informatics, University of Leipzig, Leipzig, Germany
| | - Matthias Noll
- Institute for Bioanalysis, Department of Applied Sciences, Coburg University of Applied Sciences and Arts, Coburg, Germany
| |
Collapse
|
83
|
Gardiner BJ, Stewardson AJ, Abbott IJ, Peleg AY. Nitrofurantoin and fosfomycin for resistant urinary tract infections: old drugs for emerging problems. Aust Prescr 2019; 42:14-19. [PMID: 30765904 DOI: 10.18773/austprescr.2019.002] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Uncomplicated urinary tract infection is one of the most common indications for antibiotic use in the community However the Gram-negative organisms that can cause the infection are becoming more resistant to antibiotics Many multidrug resistant organisms retain susceptibility to two old antibiotics nitrofurantoin and fosfomycin Advantages over newer drugs include their high urinary concentrations and minimal toxicity Fosfomycin is a potential treatment option for patients with uncomplicated urinary tract infection due to resistant organisms Nitrofurantoin may be more effective and can be used for urinary infections in pregnant women
Collapse
Affiliation(s)
- Bradley J Gardiner
- Department of Infectious Disease, Alfred Health and Central Clinical School, Monash University, Melbourne.,Department of Medical Microbiology and Infectious Diseases, Research and Development Unit, Erasmus Medical Centre, Rotterdam, The Netherlands.,Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne
| | - Andrew J Stewardson
- Department of Infectious Disease, Alfred Health and Central Clinical School, Monash University, Melbourne.,Department of Medical Microbiology and Infectious Diseases, Research and Development Unit, Erasmus Medical Centre, Rotterdam, The Netherlands.,Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne
| | - Iain J Abbott
- Department of Infectious Disease, Alfred Health and Central Clinical School, Monash University, Melbourne.,Department of Medical Microbiology and Infectious Diseases, Research and Development Unit, Erasmus Medical Centre, Rotterdam, The Netherlands.,Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne
| | - Anton Y Peleg
- Department of Infectious Disease, Alfred Health and Central Clinical School, Monash University, Melbourne.,Department of Medical Microbiology and Infectious Diseases, Research and Development Unit, Erasmus Medical Centre, Rotterdam, The Netherlands.,Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne
| |
Collapse
|
84
|
Peeters T, Penders J, Smeekens SP, Galazzo G, Houben B, Netea MG, Savelkoul PH, Gyssens IC. The fecal and mucosal microbiome in acute appendicitis patients: an observational study. Future Microbiol 2019; 14:111-127. [PMID: 30663346 DOI: 10.2217/fmb-2018-0203] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Accepted: 11/27/2018] [Indexed: 01/08/2023] Open
Abstract
AIM We aimed to study the mucosal microbiota of the appendix in a prospective appendicitis cohort and to compare the fecal microbiota of patients and controls. We hypothesized that the microbiota may be associated with susceptibility to appendicitis. PATIENTS & METHODS The fecal microbiota of 99 patients and 106 controls were characterized using 16S-23S intergenic spacer profiling. Richness, diversity and community structure were compared. The appendiceal microbiota from 90 patients was analyzed according to the severity of appendicitis. RESULTS Overall fecal microbial richness and diversity were similar in patients and controls, yet richness and diversity within the group of Firmicutes, Actinobacteria, Fusobacteria and Verrucomicrobia phyla were lower in patients. Discriminant analyses could correctly classify patients and controls with fair accuracy. No differences were found according to severity in appendiceal or fecal microbiota. CONCLUSION This study demonstrates differences in the composition of intestinal microbiota of appendicitis patients and healthy individuals.
Collapse
Affiliation(s)
- Toon Peeters
- Department of Infectious Diseases & Immunity, Jessa Hospital, Hasselt, Belgium
- Faculty of Medicine & Life Sciences, Hasselt University, Hasselt, Belgium
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - John Penders
- School of Nutrition & Translational Research in Metabolism (NUTRIM) & School for Public Health & Primary Care (CAPHRI), Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Sanne P Smeekens
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gianluca Galazzo
- School of Nutrition & Translational Research in Metabolism (NUTRIM) & School for Public Health & Primary Care (CAPHRI), Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Bert Houben
- Department of Abdominal & Oncological Surgery, Jessa Hospital, Hasselt, Belgium
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| | - Paul Hm Savelkoul
- School of Nutrition & Translational Research in Metabolism (NUTRIM) & School for Public Health & Primary Care (CAPHRI), Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands
- Department of Medical Microbiology & Infection Control, VU University Medical Center, Amsterdam, The Netherlands
| | - Inge C Gyssens
- Department of Infectious Diseases & Immunity, Jessa Hospital, Hasselt, Belgium
- Faculty of Medicine & Life Sciences, Hasselt University, Hasselt, Belgium
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Radboudumc Center for Infectious Diseases (RCI), Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
85
|
Ferguson KM, Jacob ME, Theriot CM, Callahan BJ, Prange T, Papich MG, Foster DM. Dosing Regimen of Enrofloxacin Impacts Intestinal Pharmacokinetics and the Fecal Microbiota in Steers. Front Microbiol 2018; 9:2190. [PMID: 30283418 PMCID: PMC6156522 DOI: 10.3389/fmicb.2018.02190] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Accepted: 08/27/2018] [Indexed: 12/25/2022] Open
Abstract
Objective: The intestinal concentrations of antimicrobial drugs that select for resistance in fecal bacteria of cattle are poorly understood. Our objective was to associate active drug concentrations in the intestine of steers with changes in the resistance profile and composition of the fecal microbiome. Methods: Steers were administered either a single dose (12.5 mg/kg) or 3 multiple doses (5 mg/kg) of enrofloxacin subcutaneously every 24 h. Enrofloxacin and ciprofloxacin concentrations in intestinal fluid were measured over 96 h, and the abundance and MIC of E. coli in culture and the composition of the fecal microbiota by 16S rRNA gene sequencing were assessed over 192 h after initial treatment. Results: Active drug concentrations in the ileum and colon exceeded plasma and interstitial fluid concentrations, but were largely eliminated by 48 h after the last dose. The concentration of E. coli in the feces significantly decreased during peak drug concentrations, but returned to baseline by 96 h in both groups. The median MIC of E. coli isolates increased for 24 h in the single dose group, and for 48 h in the multiple dose group. The median MIC was higher in the multiple dose group when compared to the single dose group starting 12 h after the initial dose. The diversity of the fecal microbiota did not change in either treatment group, and taxa-specific changes were primarily seen in phyla commonly associated with the rumen. Conclusions: Both dosing regimens of enrofloxacin achieve high concentrations in the intestinal lumen, and the rapid elimination mitigates long-term impacts on fecal E. coli resistance and the microbiota.
Collapse
Affiliation(s)
- Kaitlyn M Ferguson
- Department of Population Health and Pathobiology, College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| | - Megan E Jacob
- Department of Population Health and Pathobiology, College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| | - Casey M Theriot
- Department of Population Health and Pathobiology, College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| | - Benjamin J Callahan
- Department of Population Health and Pathobiology, College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| | - Timo Prange
- Department of Clinical Sciences, College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| | - Mark G Papich
- Department of Molecular and Biomedical Sciences, College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| | - Derek M Foster
- Department of Population Health and Pathobiology, College of Veterinary Medicine, NC State University, Raleigh, NC, United States
| |
Collapse
|
86
|
Akgül T, Karakan T. The role of probiotics in women with recurrent urinary tract infections. Turk J Urol 2018; 44:377-383. [PMID: 30487041 DOI: 10.5152/tud.2018.48742] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/18/2018] [Indexed: 12/21/2022]
Abstract
Urinary tract infections (UTIs) still represent a significant bother for women and result in high costs to the health system. Increasing antimicrobial resistance has stimulated interest in non-antibiotic prophylaxis of recurrent UTIs. Evidence shows that the microorganisms inhabit many sites of the body, including the urinary tract which has long been assumed to be sterile in healthy individuals, might have a role in maintaining urinary health. Studies of the urinary microbiota (UM) have identified remarkable differences between healthy populations and those with urologic diseases. The depletion of these organisms in women susceptible to UTIs raised the question of whether artificial supplementation of these microorganisms as probiotics could lower infection rates. In the literature, probiotic interventions were shown to have some efficacy in the treatment and prevention of urogenital infections. Despite previous controversy regarding the use of probiotics, as treatment for UTIs, there are increasing signs that it may be possible to use them as a first step in regulating the UM so as to reduce the risk of or as a treatment for certain urinary diseases. However, further future clinical trials, involving large numbers of patients, will be mandatory to achieve definite evidence of the preventive and curative role of probiotics in UTIs. Details about correct formulations in terms of amount of bacteria, viability and associated growth factors, will be required in order to standardize the administration schedule and achieve homogeneous and comparable results on selected patients.
Collapse
Affiliation(s)
- Turgay Akgül
- Department of Urology, University of Health Sciences Ankara Training and Research Hospital, Ankara, Turkey
| | - Tolga Karakan
- Department of Urology, University of Health Sciences Ankara Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
87
|
Cižman M, Plankar Srovin T. Antibiotic consumption and resistance of gram-negative pathogens (collateral damage). GMS INFECTIOUS DISEASES 2018; 6:Doc05. [PMID: 30671336 PMCID: PMC6301726 DOI: 10.3205/id000040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Antibiotics are commonly prescribed in community and hospital care. Overuse and misuse favors emergence and spread of resistant bacteria. The ATC/DDD methodology is commonly used for presenting the drug utilization data. In primary care, the consumption is usually expressed in DDD per 1,000 inhabitants per day, in hospital, preferably in DDD per 100 bed days and DDD per 100 admissions. The alternative metric is days of therapy (DOT), which needs IT support. Antibiotics have ecological adverse effects at individual and population level. Antibiotics select resistant bacteria among pathogens and normal flora. Broad-spectrum antibiotics, low dosage and prolonged antibiotic therapy favor the development of resistance. Although total use of antibiotics in hospital is much less than in the community, the intensity of use magnified by cross infection ensures a multitude of resistant bacteria in today's hospitals. Reversal of resistance is complex and might persist for many years despite the introduction of antimicrobial containment and stewardship programs.
Collapse
Affiliation(s)
- Milan Cižman
- University Medical Center, Department of Infectious Diseases, Ljubljana, Slovenia
| | - Tina Plankar Srovin
- University Medical Center, Department of Infectious Diseases, Ljubljana, Slovenia
| |
Collapse
|
88
|
Rea D, Coppola G, Palma G, Barbieri A, Luciano A, Del Prete P, Rossetti S, Berretta M, Facchini G, Perdonà S, Turco MC, Arra C. Microbiota effects on cancer: from risks to therapies. Oncotarget 2018; 9:17915-17927. [PMID: 29707157 PMCID: PMC5915165 DOI: 10.18632/oncotarget.24681] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 02/27/2018] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota, a group of 1014 bacteria, eukaryotes and virus living in gastrointestinal tract, is crucial for many physiological processes in particular plays an important role in inflammatory and immune reactions. Several internal and external factors can influence this population, and shifts in their composition, have been demonstrated to contribute and affect different diseases. During dysbiosis several bacteria related to inflammation, one of the most necessary factors in carcinogenesis; it has been shown that some bacterial strains through deregulation of different signals/pathways may affect tumor development through the production of many factors. Gut microbiota might be considered as a holistic hub point for cancer development: direct and indirect involvements have been studying in several neoplasms such as colon rectal cancer, hepatocellular carcinoma and breast cancer. This review discuss over the evidence of crosstalk between gut microbiota and cancer, its ability to modulate chemotherapy, radiotherapy and immunotherapy, and the possibility that the intestinal microbial is a new target for therapeutic approaches to improve the prognosis and quality of life of cancer patients.
Collapse
Affiliation(s)
- Domenica Rea
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Giovanni Coppola
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Giuseppe Palma
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Antonio Barbieri
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Antonio Luciano
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Paola Del Prete
- Direzione Scientifica, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Sabrina Rossetti
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Massimiliano Berretta
- Department of Medical Oncology, CRO- Aviano, National Cancer Institute, Aviano, Italy
| | - Gaetano Facchini
- Division of Medical Oncology, Department of Uro-Gynaecological Oncology, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Sisto Perdonà
- Department of Urology, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| | - Maria Caterina Turco
- Department of Medicine and Surgery, University of Salerno, Baronissi, Salerno, Italy
| | - Claudio Arra
- S.S.D Sperimentazione Animale, Istituto Nazionale Tumori, IRCCS, “Fondazione G. Pascale”, Naples, Italy
| |
Collapse
|
89
|
Iizumi T, Battaglia T, Ruiz V, Perez Perez GI. Gut Microbiome and Antibiotics. Arch Med Res 2017; 48:727-734. [PMID: 29221800 DOI: 10.1016/j.arcmed.2017.11.004] [Citation(s) in RCA: 118] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 11/15/2017] [Indexed: 12/12/2022]
Abstract
Despite that the human gastrointestinal tract is the most populated ecological niche by bacteria in the human body, much is still unknown about its characteristics. This site is highly susceptible to the effects of many external factors that may affect in the quality and the quantity of the microbiome. Specific factors such as diet, personal hygiene, pharmacological drugs and the use of antibiotics can produce a significant impact on the gut microbiota. The effect of these factors is more relevant early in life, when the gut microbiota has not yet fully established. In this review, we discussed the effect of type and doses of the antibiotics on the gut microbiota and what the major consequences in the use and abuse of these antimicrobial agents.
Collapse
Affiliation(s)
- Tadasu Iizumi
- Department of Medicine, New York University School of Medicine NYU Langone Health, New York, NY, USA.
| | - Thomas Battaglia
- Department of Medicine, New York University School of Medicine NYU Langone Health, New York, NY, USA
| | - Victoria Ruiz
- Department of Medicine, New York University School of Medicine NYU Langone Health, New York, NY, USA
| | - Guillermo I Perez Perez
- Department of Medicine, New York University School of Medicine NYU Langone Health, New York, NY, USA
| |
Collapse
|
90
|
de Lastours V, Maugy E, Mathy V, Chau F, Rossi B, Guérin F, Cattoir V, Fantin B. Ecological impact of ciprofloxacin on commensal enterococci in healthy volunteers. J Antimicrob Chemother 2017; 72:1574-1580. [PMID: 28333351 DOI: 10.1093/jac/dkx043] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/20/2017] [Indexed: 12/15/2022] Open
Abstract
Background The ecological impact of ciprofloxacin on commensal enterococci is unknown. Methods Forty-eight healthy volunteers received ciprofloxacin from day (D) 0 to D14; stools were collected on D7, D14 and D42. Fluoroquinolone-susceptible and -resistant enterococci (FQ-SE and FQ-RE) were detected and quantified by culture, and identified by MALDI-TOF MS. The relative abundance of FQ-RE over FQ-SE was determined. The genetic basis of fluoroquinolone resistance was deciphered by partial sequencing of gyrA and parC genes. Clonal relatedness was determined by random amplification of polymorphic DNA PCR. Clinical trial no.: NCT00190151. Results Enterococci were carried by 47/48 (98%) subjects. Total counts were reduced during ciprofloxacin therapy (4.0 and 3.9 log cfu/g on D7 and D14 versus 5.9 log cfu/g before and 6.9 log cfu/g after treatment; P < 0.05). Twenty-one out of 48 (44%) carried FQ-RE; among them, 21/21 carried Enterococcus faecium , 19 carried Enterococcus faecalis and 11 carried other species. Five out of 48 (10%) harboured FQ-RE (ciprofloxacin MIC >4 mg/L) before treatment (all E. faecium ), 6 on D7 (3 E. faecium and 3 E. faecalis ), 8 on D14 (4 E. faecium and 4 E. faecalis ) and 10 (21%) on D42 (9 E. faecium and 1 E. faecalis ). The relative abundance of FQ-RE increased from 44% on D0 to 73% and 75% on D7 and D14, respectively. No acquisition of fluoroquinolone resistance among endogenous D0 strains was evidenced. All (14/14) distinct Fluoroquinolone-resistant E. faecalis clones were gyrA / parC double mutants with high-level resistance (ciprofloxacin MIC >64 mg/L). In contrast, 34/35 E. faecium exhibited low-level resistance (ciprofloxacin MIC 4-32 mg/L) with no gyrA / parC mutation, but overexpressed the chromosomal Efm qnr gene. As compared with Fluoroquinolone-susceptible strains, Fluoroquinolone-resistant E. faecium were more frequently ampicillin resistant and Fluoroquinolone-resistant E. faecalis were more highly resistant to gentamicin. Conclusions Although intrinsically poorly susceptible to fluoroquinolones, gut populations of enterococci are highly impacted both quantitatively and qualitatively by ciprofloxacin.
Collapse
Affiliation(s)
- Victoire de Lastours
- IAME, UMR-1137, Inserm and Université Paris Diderot, Paris, France.,Service de Médecine Interne, Hôpital Beaujon, APHP, Paris, France
| | - Elena Maugy
- Université de Caen Normandie, EA4655 (équipe 'Antibio-Résistance'), Caen, France
| | - Vincent Mathy
- Université de Caen Normandie, EA4655 (équipe 'Antibio-Résistance'), Caen, France
| | - Françoise Chau
- IAME, UMR-1137, Inserm and Université Paris Diderot, Paris, France
| | - Benjamin Rossi
- IAME, UMR-1137, Inserm and Université Paris Diderot, Paris, France.,Service de Médecine Interne, Hôpital Beaujon, APHP, Paris, France
| | - François Guérin
- Université de Caen Normandie, EA4655 (équipe 'Antibio-Résistance'), Caen, France.,CNR de la Résistance aux Antibiotiques (Laboratoire Associé 'Entérocoques'), Caen, France
| | - Vincent Cattoir
- Université de Caen Normandie, EA4655 (équipe 'Antibio-Résistance'), Caen, France.,CNR de la Résistance aux Antibiotiques (Laboratoire Associé 'Entérocoques'), Caen, France
| | - Bruno Fantin
- IAME, UMR-1137, Inserm and Université Paris Diderot, Paris, France.,Service de Médecine Interne, Hôpital Beaujon, APHP, Paris, France
| | | |
Collapse
|
91
|
Ruppé E, Burdet C, Grall N, de Lastours V, Lescure FX, Andremont A, Armand-Lefèvre L. Impact of antibiotics on the intestinal microbiota needs to be re-defined to optimize antibiotic usage. Clin Microbiol Infect 2017; 24:3-5. [PMID: 28970162 DOI: 10.1016/j.cmi.2017.09.017] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2017] [Revised: 09/19/2017] [Accepted: 09/21/2017] [Indexed: 12/22/2022]
Affiliation(s)
- E Ruppé
- INSERM, IAME, UMR 1137, Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Bactériologie, Paris, France.
| | - C Burdet
- INSERM, IAME, UMR 1137, Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, Paris, France; AP-HP, Hôpital Bichat, Département of d'Epidémiologie, Biostatistiques et Recherche Clinique, Paris, France
| | - N Grall
- INSERM, IAME, UMR 1137, Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Bactériologie, Paris, France
| | - V de Lastours
- INSERM, IAME, UMR 1137, Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, Paris, France; AP-HP, Hôpital Beaujon, Equipe Mobile d'Antibiothérapie, Clichy, France
| | - F-X Lescure
- INSERM, IAME, UMR 1137, Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, Paris, France; AP-HP, Hôpital Bichat, Service des Maladies Infectieuses, Paris, France
| | - A Andremont
- INSERM, IAME, UMR 1137, Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Bactériologie, Paris, France
| | - L Armand-Lefèvre
- INSERM, IAME, UMR 1137, Paris, France; Université Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Bactériologie, Paris, France
| |
Collapse
|
92
|
Tamanai-Shacoori Z, Smida I, Bousarghin L, Loreal O, Meuric V, Fong SB, Bonnaure-Mallet M, Jolivet-Gougeon A. Roseburia spp.: a marker of health? Future Microbiol 2017; 12:157-170. [PMID: 28139139 DOI: 10.2217/fmb-2016-0130] [Citation(s) in RCA: 476] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The genus Roseburia consists of obligate Gram-positive anaerobic bacteria that are slightly curved, rod-shaped and motile by means of multiple subterminal flagella. It includes five species: Roseburia intestinalis, R. hominis, R. inulinivorans, R. faecis and R. cecicola. Gut Roseburia spp. metabolize dietary components that stimulate their proliferation and metabolic activities. They are part of commensal bacteria producing short-chain fatty acids, especially butyrate, affecting colonic motility, immunity maintenance and anti-inflammatory properties. Modification in Roseburia spp. representation may affect various metabolic pathways and is associated with several diseases (including irritable bowel syndrome, obesity, Type 2 diabetes, nervous system conditions and allergies). Roseburia spp. could also serve as biomarkers for symptomatic pathologies (e.g., gallstone formation) or as probiotics for restoration of beneficial flora.
Collapse
Affiliation(s)
- Zohreh Tamanai-Shacoori
- INSERM 1241/NUtrition MEtabolism CANcer/CIMIAD, 2 avenue du Professeur Léon Bernard, Rennes, France
| | - Imen Smida
- INSERM 1241/NUtrition MEtabolism CANcer/CIMIAD, 2 avenue du Professeur Léon Bernard, Rennes, France
| | - Latifa Bousarghin
- INSERM 1241/NUtrition MEtabolism CANcer/CIMIAD, 2 avenue du Professeur Léon Bernard, Rennes, France
| | - Olivier Loreal
- INSERM 1241/NUtrition MEtabolism CANcer/CIMIAD, 2 avenue du Professeur Léon Bernard, Rennes, France
| | - Vincent Meuric
- INSERM 1241/NUtrition MEtabolism CANcer/CIMIAD, 2 avenue du Professeur Léon Bernard, Rennes, France.,CHU Rennes, 2 rue Henri Le Guilloux, 35000 Rennes, France.,Université de Rennes 1, 2 avenue du Professeur Léon Bernard, 35043 Rennes, France
| | - Shao Bing Fong
- Université de Rennes 1, 2 avenue du Professeur Léon Bernard, 35043 Rennes, France
| | - Martine Bonnaure-Mallet
- INSERM 1241/NUtrition MEtabolism CANcer/CIMIAD, 2 avenue du Professeur Léon Bernard, Rennes, France.,CHU Rennes, 2 rue Henri Le Guilloux, 35000 Rennes, France.,Université de Rennes 1, 2 avenue du Professeur Léon Bernard, 35043 Rennes, France
| | - Anne Jolivet-Gougeon
- INSERM 1241/NUtrition MEtabolism CANcer/CIMIAD, 2 avenue du Professeur Léon Bernard, Rennes, France.,CHU Rennes, 2 rue Henri Le Guilloux, 35000 Rennes, France.,Université de Rennes 1, 2 avenue du Professeur Léon Bernard, 35043 Rennes, France
| |
Collapse
|
93
|
Continuous long-term electricity-driven bioproduction of carboxylates and isopropanol from CO 2 with a mixed microbial community. J CO2 UTIL 2017. [DOI: 10.1016/j.jcou.2017.04.014] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
|
94
|
Grall N, Lazarevic V, Gaïa N, Couffignal C, Laouénan C, Ilic-Habensus E, Wieder I, Plesiat P, Angebault C, Bougnoux ME, Armand-Lefevre L, Andremont A, Duval X, Schrenzel J. Unexpected persistence of extended-spectrum β-lactamase-producing Enterobacteriaceae in the faecal microbiota of hospitalised patients treated with imipenem. Int J Antimicrob Agents 2017; 50:81-87. [PMID: 28499958 DOI: 10.1016/j.ijantimicag.2017.02.018] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Revised: 02/10/2017] [Accepted: 02/22/2017] [Indexed: 11/29/2022]
Abstract
Imipenem is active against extended-spectrum β-lactamase-producing Enterobacteriaceae (ESBL-E) but favours the intestinal emergence of resistance. The effects of imipenem on intestinal microbiota have been studied using culture-based techniques. In this study, the effects were investigated in patients using culture and metagenomic techniques. Seventeen hospitalised adults receiving imipenem were included in a multicentre study (NCT01703299, http://www.clinicaltrials.gov). Most patients had a history of antibiotic use and/or hospitalisation. Stools were collected before, during and after imipenem treatment. Bacterial and fungal colonisation was assessed by culture, and microbiota changes were assessed using metagenomics. Unexpectedly, high colonisation rates by imipenem-susceptible ESBL-E before treatment (70.6%) remained stable over time, suggesting that imipenem intestinal concentrations were very low. Carriage rates of carbapenem-resistant Gram-negative bacilli (0-25.0%) were also stable over time, whereas those of yeasts (64.7% before treatment) peaked at 76.5% during treatment and decreased thereafter. However, these trends were not statistically significant. Yeasts included highly diverse colonising Candida spp. Metagenomics showed no global effect of imipenem on the bacterial taxonomic profiles at the sequencing depth used but demonstrated specific changes in the microbiota not detected with culture, attributed to factors other than imipenem, including sampling site or treatment with other antibiotics. In conclusion, culture and metagenomics were highly complementary in characterising the faecal microbiota of patients. The changes observed during imipenem treatment were unexpectedly limited, possibly because the microbiota was already disturbed by previous antibiotic exposure or hospitalisation.
Collapse
Affiliation(s)
- N Grall
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris-Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Microbiologie, F-75018 Paris, France.
| | - V Lazarevic
- Genomic Research Laboratory, Geneva University Hospitals, Geneva, Switzerland
| | - N Gaïa
- Genomic Research Laboratory, Geneva University Hospitals, Geneva, Switzerland
| | - C Couffignal
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris-Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, Service de Biostatistique, F-75018 Paris, France
| | - C Laouénan
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris-Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, Service de Biostatistique, F-75018 Paris, France
| | - E Ilic-Habensus
- AP-HP, Hôpital Bichat, INSERM CIC 1425, F-75018 Paris, France
| | - I Wieder
- AP-HP, Hôpital Bichat, Laboratoire de Microbiologie, F-75018 Paris, France
| | - P Plesiat
- Laboratoire de Bactériologie EA4266, Faculté de Médecine-Pharmacie, Université de Franche-Comté, Besançon, France
| | - C Angebault
- AP-HP, Hôpital Necker-Enfants Malades, Unité de Parasitologie-Mycologie, Service de Microbiologie, F-75015 Paris, France; Université Paris Descartes, Sorbonne Paris-Cité, F-75015 Paris, France
| | - M E Bougnoux
- AP-HP, Hôpital Necker-Enfants Malades, Unité de Parasitologie-Mycologie, Service de Microbiologie, F-75015 Paris, France; Université Paris Descartes, Sorbonne Paris-Cité, F-75015 Paris, France
| | - L Armand-Lefevre
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris-Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Microbiologie, F-75018 Paris, France
| | - A Andremont
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris-Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, Laboratoire de Microbiologie, F-75018 Paris, France
| | - X Duval
- INSERM, IAME, UMR 1137, F-75018 Paris, France; Université Paris-Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France; AP-HP, Hôpital Bichat, INSERM CIC 1425, F-75018 Paris, France
| | - J Schrenzel
- Genomic Research Laboratory, Geneva University Hospitals, Geneva, Switzerland; Laboratory of Bacteriology, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
95
|
Antimicrobial Resistance Patterns in Women with Positive Urine Culture: Does Menopausal Status Make a Significant Difference? BIOMED RESEARCH INTERNATIONAL 2017; 2017:4192908. [PMID: 28497048 PMCID: PMC5406742 DOI: 10.1155/2017/4192908] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 03/03/2017] [Accepted: 03/22/2017] [Indexed: 11/19/2022]
Abstract
Aim. Urinary tract infection (UTI) is considered one of the most common bacterial infections in women. The aim of this study was to investigate the types of uropathogens present, as well as the degree of antimicrobial drug resistance seen among premenopausal (n = 2748) and postmenopausal (n = 1705) women with uncomplicated UTI. Methods. Urinary samples (n = 4453) collected from women with UTI were analyzed in terms of uropathogens present. These were considered as positive if bacterial growth was ≥105 colony forming units (CFUs)/mL. Susceptibility and resistance testing for commonly used antibiotics was subsequently assessed. Results. The most common uropathogens cultured from urine samples were Escherichia coli (65.5%), followed by Enterococcus faecalis (12.2%), Klebsiella pneumoniae (4.7%), and Proteus mirabilis (4.2%). The resistance to ampicillin exceeded 40%, independently of menopausal status. Of note, resistance to ciprofloxacin exceeded 25% among postmenopausal patients. Moreover, resistance of all uropathogens to commonly used antimicrobials was significantly higher in postmenopausal women. Conclusion. Due to the high resistance rate, ampicillin, ciprofloxacin, and the trimethoprim/sulfamethoxazole combination should be avoided in treating postmenopausal women affected by UTI without being indicated by initial urine culture report. Finally, cephalexin and cefuroxime are promising alternatives as initial treatment in postmenopausal women.
Collapse
|
96
|
Søgaard M, Heide-Jørgensen U, Vandenbroucke JP, Schønheyder HC, Vandenbroucke-Grauls CMJE. Risk factors for extended-spectrum β-lactamase-producing Escherichia coli urinary tract infection in the community in Denmark: a case-control study. Clin Microbiol Infect 2017; 23:952-960. [PMID: 28377310 DOI: 10.1016/j.cmi.2017.03.026] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 01/06/2023]
Abstract
OBJECTIVE To verify the role of proton pump inhibitors (PPI) and nitrofurantoin, which have appeared as novel risk factors for carriage of extended-spectrum β-lactamase (ESBL) -producing Escherichia coli, as risk factors for ESBL E. coli urinary tract infection (UTI). We included known risk factors to ascertain whether our findings are comparable with those of previous studies. METHODS Population-based case-control study including 339 cases with community-onset ESBL E. coli UTI in 2007-2012, 3390 non-ESBL E. coli UTI controls and 3390 population controls. We investigated potential risk factors by estimating ORs and 95% CIs adjusting for sex, age and co-morbidity. RESULTS Comparing cases with non-ESBL E. coli UTI, PPI use yielded an OR of 1.6 (95% CI 1.2-2.0) and antibiotic exposure gave an OR of 1.4 (95% CI 1.1-1.8); these were driven by nitrofurantoin (OR 1.8; 95% CI 1.3-2.6) and macrolides (OR 1.7; 95% CI 1.2-2.3). Other risk factors included previous hospitalization with one or two and more than two hospitalizations versus none yielding ORs of 1.9 (95% CI 1.4-2.5) and 4.6 (95% CI 3.2-6.8), recent surgery (OR 2.0; 95% CI 1.5-2.8), renal disease (OR 2.2; 95% CI 1.4-3.4), chronic pulmonary disease (OR 1.4; 95% CI 1.0-2.0) and cancer (OR 1.5; 95% CI 1.1-2.1). Comparing cases with population controls, we found that most risk factors were also risk factors for non-ESBL UTI. CONCLUSIONS ESBL E. coli UTI were associated with previous hospitalization and surgery. Nitrofurantoin and macrolides augmented the risk. PPIs had a moderate effect but may be important facilitators of ESBL carriage due to their widespread use.
Collapse
Affiliation(s)
- M Søgaard
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark.
| | - U Heide-Jørgensen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark
| | - J P Vandenbroucke
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus, Denmark; Department of Clinical Epidemiology, Leiden University, Utrecht, The Netherlands
| | - H C Schønheyder
- Department of Clinical Microbiology, Aalborg University Hospital, Aalborg, Denmark; Department of Clinical Medicine, Aalborg University, Aalborg, Denmark
| | - C M J E Vandenbroucke-Grauls
- Department of Medical Microbiology & Infection Control, VU University Medical Centre, Amsterdam, The Netherlands
| |
Collapse
|
97
|
Deng YF, Wang YJ, Zou Y, Azarfar A, Wei XL, Ji SK, Zhang J, Wu ZH, Wang SX, Dong SZ, Xu Y, Shao DF, Xiao JX, Yang KL, Cao ZJ, Li SL. Influence of dairy by-product waste milk on the microbiomes of different gastrointestinal tract components in pre-weaned dairy calves. Sci Rep 2017; 7:42689. [PMID: 28281639 PMCID: PMC5345013 DOI: 10.1038/srep42689] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2016] [Accepted: 01/13/2017] [Indexed: 12/25/2022] Open
Abstract
The community structure of colonised bacteria in the gastrointestinal tracts (GITs) of pre-weaned calves is affected by extrinsic factors, such as the genetics and diet of the calves; however, the dietary impact is not fully understood and warrants further research. Our study revealed that a total of 6, 5, 2 and 10 bacterial genera showed biologically significant differences in the GITs of pre-weaned calves fed four waste-milk diets: acidified waste milk, pasteurised waste milk, untreated bulk milk, and untreated waste milk, respectively. Specifically, generic biomarkers were observed in the rumen (e.g., Bifidobacterium, Parabacteroides, Fibrobacter, Clostridium, etc.), caecum (e.g., Faecalibacterium, Oxalobacter, Odoribacter, etc.) and colon (e.g., Megamonas, Comamonas, Stenotrophomonas, etc.) but not in the faeces. In addition, the predicted metabolic pathways showed that the expression of genes related to metabolic diseases was increased in the calves fed untreated waste milk, which indicated that untreated waste milk is not a suitable liquid diet for pre-weaned calves. This is the first study to demonstrate how different types of waste milk fed to pre-weaned calves affect the community structure of colonised bacteria, and the results may provide insights for the intentional adjustment of diets and gastrointestinal bacterial communities.
Collapse
Affiliation(s)
- Y F Deng
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Y J Wang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Y Zou
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - A Azarfar
- Department of Animal Science, Faculty of Agriculture, Lorestan University, PO Box 465, Khorramabad, Iran
| | - X L Wei
- Sichuan Animal Science Academy, Animal Breeding and Genetics key Laboratory of Sichuan Province, Chengdu 610066, P. R. China
| | - S K Ji
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - J Zhang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Z H Wu
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - S X Wang
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - S Z Dong
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - Y Xu
- Beijing Computing Center, Beijing 100094, P. R. China
| | - D F Shao
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - J X Xiao
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - K L Yang
- College of Animal Science, Xinjiang Agricultural University, Wulumuqi 830052, P. R. China
| | - Z J Cao
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| | - S L Li
- State Key Laboratory of Animal Nutrition, Beijing Engineering Technology Research Center of Raw Milk Quality and Safety Control, College of Animal Science and Technology, China Agricultural University, Beijing 100193, P. R. China
| |
Collapse
|
98
|
|
99
|
Staley C, Vaughn BP, Graiziger CT, Sadowsky MJ, Khoruts A. Gut-sparing treatment of urinary tract infection in patients at high risk of Clostridium difficile infection. J Antimicrob Chemother 2017; 72:522-528. [PMID: 27999027 PMCID: PMC6075516 DOI: 10.1093/jac/dkw499] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2016] [Revised: 10/17/2016] [Accepted: 10/20/2016] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Recipients of faecal microbiota transplantation (FMT) in treatment of recurrent Clostridium difficile infection (RCDI) remain at markedly increased risk of re-infection with C. difficile with new antibiotic provocations. Urinary tract infections (UTIs) are common indications for antibiotics in these patients, often resulting in C. difficile re-infection. METHODS We present a case series of 19 patients treated with parenteral aminoglycosides for UTI following FMT for RCDI. A 3 day outpatient regimen of once-daily intramuscular administration of gentamicin was used to treat 18 consecutive FMT recipients with uncomplicated UTI. One other patient was treated for a complicated UTI with intravenous amikacin. Profiling of 16S rRNA genes was used to track changes in faecal microbial community structure during this regimen in three patients. RESULTS The protocol was highly effective in treating UTI symptoms. None of the patients suffered a re-infection with C. difficile The faecal microbial communities remained undisturbed by treatment with intramuscular administration of gentamicin. CONCLUSIONS Despite falling out of favour in recent years, aminoglycoside antibiotics given parenterally have the advantage of minimal penetration into the gut lumen. A brief (3 day) course of parenteral gentamicin was safe and effective in curing UTI in patients at high risk of C. difficile infection without perturbing their gut microbiota.
Collapse
Affiliation(s)
- Christopher Staley
- BioTechnology Institute, University of Minnesota, 140 Gortner Labs, 1479 Gortner Avenue, St Paul, MN 55108, USA
| | - Byron P Vaughn
- Department of Medicine Division of Gastroenterology, University of Minnesota, 401 East River Parkway, Variety Club Research Center, 1st Floor - Suite 131, Minneapolis, MN, 55455, USA
| | - Carolyn T Graiziger
- Department of Medicine Division of Gastroenterology, University of Minnesota, 401 East River Parkway, Variety Club Research Center, 1st Floor - Suite 131, Minneapolis, MN, 55455, USA
| | - Michael J Sadowsky
- BioTechnology Institute, University of Minnesota, 140 Gortner Labs, 1479 Gortner Avenue, St Paul, MN 55108, USA
| | - Alexander Khoruts
- BioTechnology Institute, University of Minnesota, 140 Gortner Labs, 1479 Gortner Avenue, St Paul, MN 55108, USA
- Wallin Biomedical Sciences Building, Center for Immunology, University of Minnesota, 2101 6th St SE, Minneapolis, MN 55414, USA
| |
Collapse
|
100
|
Wong ACN, Vanhove AS, Watnick PI. The interplay between intestinal bacteria and host metabolism in health and disease: lessons from Drosophila melanogaster. Dis Model Mech 2016; 9:271-81. [PMID: 26935105 PMCID: PMC4833331 DOI: 10.1242/dmm.023408] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
All higher organisms negotiate a truce with their commensal microbes and battle pathogenic microbes on a daily basis. Much attention has been given to the role of the innate immune system in controlling intestinal microbes and to the strategies used by intestinal microbes to overcome the host immune response. However, it is becoming increasingly clear that the metabolisms of intestinal microbes and their hosts are linked and that this interaction is equally important for host health and well-being. For instance, an individual's array of commensal microbes can influence their predisposition to chronic metabolic diseases such as diabetes and obesity. A better understanding of host-microbe metabolic interactions is important in defining the molecular bases of these disorders and could potentially lead to new therapeutic avenues. Key advances in this area have been made using Drosophila melanogaster. Here, we review studies that have explored the impact of both commensal and pathogenic intestinal microbes on Drosophila carbohydrate and lipid metabolism. These studies have helped to elucidate the metabolites produced by intestinal microbes, the intestinal receptors that sense these metabolites, and the signaling pathways through which these metabolites manipulate host metabolism. Furthermore, they suggest that targeting microbial metabolism could represent an effective therapeutic strategy for human metabolic diseases and intestinal infection.
Collapse
Affiliation(s)
- Adam C N Wong
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Audrey S Vanhove
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Paula I Watnick
- Division of Infectious Diseases, Boston Children's Hospital, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| |
Collapse
|