1
|
Simoni A, Schwartz L, Junquera GY, Ching CB, Spencer JD. Current and emerging strategies to curb antibiotic-resistant urinary tract infections. Nat Rev Urol 2024; 21:707-722. [PMID: 38714857 PMCID: PMC11540872 DOI: 10.1038/s41585-024-00877-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/26/2024] [Indexed: 05/23/2024]
Abstract
Rising rates of antibiotic resistance in uropathogenic bacteria compromise patient outcomes and prolong hospital stays. Consequently, new strategies are needed to prevent and control the spread of antibiotic resistance in uropathogenic bacteria. Over the past two decades, sizeable clinical efforts and research advances have changed urinary tract infection (UTI) treatment and prevention strategies to conserve antibiotic use. The emergence of antimicrobial stewardship, policies from national societies, and the development of new antimicrobials have shaped modern UTI practices. Future UTI management practices could be driven by the evolution of antimicrobial stewardship, improved and readily available diagnostics, and an improved understanding of how the microbiome affects UTI. Forthcoming UTI treatment and prevention strategies could employ novel bactericidal compounds, combinations of new and classic antimicrobials that enhance bacterial killing, medications that prevent bacterial attachment to uroepithelial cells, repurposing drugs, and vaccines to curtail the rising rates of antibiotic resistance in uropathogenic bacteria and improve outcomes in people with UTI.
Collapse
Affiliation(s)
- Aaron Simoni
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
| | - Laura Schwartz
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- Department of Pediatrics, Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Guillermo Yepes Junquera
- Department of Pediatrics, Division of Infectious Diseases, Nationwide Children's, Columbus, OH, USA
| | - Christina B Ching
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- Department of Urology, Nationwide Children's, Columbus, OH, USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- Department of Pediatrics, Division of Nephrology and Hypertension, Nationwide Children's, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
2
|
Zou P, Huang L, Li Y, Liu D, Che J, Zhao T, Li H, Li J, Cui YN, Yang G, Li Z, Li LL, Gao C. Phase-Separated Nano-Antibiotics Enhanced Survival in Multidrug-Resistant Escherichia coli Sepsis by Precise Periplasmic EcDsbA Targeting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407152. [PMID: 39279551 DOI: 10.1002/adma.202407152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/27/2024] [Indexed: 09/18/2024]
Abstract
Disulfide bond (Dsb) proteins, especially DsbA, represent a promising but as-yet-unrealized target in combating multidrug-resistant (MDR) bacteria because their precise subcellular targeting through multibarrier remains a significant challenge. Here, a novel heterogenization-phase-separated nano-antibiotics (NCefoTs) is proposed, through the co-assembly of enzyme-inhibiting lipopeptides (ELp component), membrane-recognizing and disrupting lipopeptides (MLp component), and cefoperazone. The self-sorting components of MLp "concentrated island-liked clusters" on the surface of NCefoTs promote the efficient penetration of NCefoTs through the outer membrane. Triggered by the DsbA, the precisely spatiotemporal engineered NCefoTs transform to nanofibers in situ and further significantly enhance the inhibition of DsbA. The hydrolytic activity of β-lactamase and the motility function of flagella are thereby impeded, confirming the efficacy of NCefoTs in restoring susceptibility to antibiotics and inhibiting infection dissemination. By these synergistic effects of NCefoTs, the minimum inhibitory concentration of antibiotics decreases from over 300 µM to 1.56 µM for clinically isolated E. coli MDR. The survival rate of sepsis-inflicted mice is significantly enhanced from 0% to 92% upon encapsulation of cefoperazone in NCefoTs, which rapidly eliminates invading pathogens and mitigates inflammation. The universally applicable delivery system, based on an "on demands" strategy, presents a promising prospect for undruggable antibiotic targets in the periplasm to combat MDR bacteria.
Collapse
Affiliation(s)
- Pengfei Zou
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- School of Material Science and Engineering, Beijing Institute of Technology, Beijing, 100081, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Lin Huang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Yi Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Dan Liu
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, 050018, China
| | - Junwei Che
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong, 250117, China
| | - Te Zhao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- School of Chemical and Pharmaceutical Engineering, Hebei University of Science and Technology, Shijiazhuang, Hebei, 050018, China
| | - Hui Li
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100083, China
| | - Jiaxin Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
- School of Pharmacy, Henan University, Kaifeng, Henan, 475004, China
| | - Ya-Nan Cui
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Guobao Yang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Zhiping Li
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| | - Li-Li Li
- School of Material Science and Engineering, Beijing Institute of Technology, Beijing, 100081, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing, 100190, China
| | - Chunsheng Gao
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing, 100850, China
| |
Collapse
|
3
|
Cocker D, Fitzgerald R, Brown CS, Holmes A. Protecting healthcare and patient pathways from infection and antimicrobial resistance. BMJ 2024; 387:e077927. [PMID: 39374953 PMCID: PMC11450933 DOI: 10.1136/bmj-2023-077927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Affiliation(s)
- Derek Cocker
- David Price Evans Global Health and Infectious Diseases Research Group, University of Liverpool, Liverpool, UK
| | - Richard Fitzgerald
- NIHR Royal Liverpool and Broadgreen Clinical Research Facility, Liverpool, UK
- Department of Pharmacology and Therapeutics, University of Liverpool, Liverpool, UK
| | - Colin S Brown
- UK Health Security Agency, London, UK
- National Institute of Health Research, Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, Imperial College London, London, UK
| | - Alison Holmes
- David Price Evans Global Health and Infectious Diseases Research Group, University of Liverpool, Liverpool, UK
- National Institute of Health Research, Health Protection Research Unit in Healthcare Associated Infection and Antimicrobial Resistance, Imperial College London, London, UK
| |
Collapse
|
4
|
Lin YC, Yang KY, Peng CK, Chan MC, Sheu CC, Feng JY, Wang SH, Huang WH, Chen CM, Chen DH, Chen CL. Clinical outcomes of carbapenem-resistant gram-negative bacterial bloodstream infection in patients with end-stage renal disease in intensive care units: a multicenter retrospective observational study. Infection 2024:10.1007/s15010-024-02343-5. [PMID: 38995550 DOI: 10.1007/s15010-024-02343-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 07/02/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND Carbapenem-resistant gram-negative bacteria (CRGNB) present a considerable global threat due to their challenging treatment and increased mortality rates, with bloodstream infection (BSI) having the highest mortality rate. Patients with end-stage renal disease (ESRD) undergoing renal replacement therapy (RRT) face an increased risk of BSI. Limited data are available regarding the prognosis and treatment outcomes of CRGNB-BSI in patients with ESRD in intensive care units (ICUs). METHODS This multi-center retrospective observational study included a total of 149 ICU patients with ESRD and CRGNB-BSI in Taiwan from January 2015 to December 2019. Clinical and microbiological outcomes were assessed, and multivariable regression analysis was used to evaluate the independent risk factors for day-28 mortality and the impact of antimicrobial therapy regimen on treatment outcomes. RESULTS Among the 149 patients, a total of 127 patients (85.2%) acquired BSI in the ICU, with catheter-related infections (47.7%) and pneumonia (32.2%) being the most common etiologies. Acinetobacter baumannii (49.0%) and Klebsiella pneumoniae (31.5%) were the most frequently isolated pathogens. The day-28 mortality rate from BSI onset was 52.3%, and in-hospital mortality was 73.2%, with survivors experiencing prolonged hospital stays. A higher Sequential Organ Failure Assessment (SOFA) score (adjusted hazards ratio [aHR], 1.25; 95% confidence interval [CI] 1.17-1.35) and shock status (aHR, 2.12; 95% CI 1.14-3.94) independently predicted day-28 mortality. Colistin-based therapy reduced day-28 mortality in patients with shock, a SOFA score of ≥ 13, and Acinetobacter baumannii-related BSI. CONCLUSIONS CRGNB-BSI led to high mortality in critically ill patients with ESRD. Day-28 mortality was independently predicted by a higher SOFA score and shock status. In patients with higher disease severity and Acinetobacter baumannii-related BSI, colistin-based therapy improved treatment outcomes.
Collapse
Affiliation(s)
- Yu-Chao Lin
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Kuang-Yao Yang
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Emergency and Critical Care Medicine, School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chung-Kan Peng
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Ming-Cheng Chan
- Department of Critical Care Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Chau-Chyun Sheu
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- Department of Internal Medicine, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Jia-Yih Feng
- Department of Chest Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Sheng-Huei Wang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
- Institute of Medical Sciences, National Defense Medical Center, Taipei, Taiwan
| | - Wei-Hsuan Huang
- Division of Infectious Diseases, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Chia-Min Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ding-Han Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.
| | - Chieh-Lung Chen
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan.
| |
Collapse
|
5
|
Zang Y, Zhao Y, Peng R, Xiao G, Liu X, Qu Y, Zhang X, Zhang J, Hong J. Incidence of Cytomegalovirus Infection After Repeat Keratoplasty and Associated Rate of Graft Failure. Ophthalmol Ther 2024; 13:1967-1980. [PMID: 38789667 PMCID: PMC11178760 DOI: 10.1007/s40123-024-00968-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 05/07/2024] [Indexed: 05/26/2024] Open
Abstract
INTRODUCTION The aim of this work was to compare the prognosis and characteristics of patients with Cytomegalovirus (CMV) infection (CMV+) with those of patients without virus infection (Virus-) undergoing repeat keratoplasty. METHODS This prospective propensity score-matched cohort study enrolled patients who underwent repeat keratoplasty for graft failure at the Peking University Third Hospital between January 2016 and May 2022. Patients with prior viral keratitis before the first keratoplasty were excluded. The primary outcome measure was the graft failure rate. The secondary outcome measures included the anterior segment characteristics, intraocular pressure (IOP), and endothelial cell density. RESULTS Ninety-four matched patient pairs were included. The graft failure rate in the CMV+ group (71%) was higher than that in the Virus- group (29%) (P < 0.001). CMV infection in the cornea increased the risk of repeat graft failure and shortened the median survival time (hazard ratio, 3.876; 95% confidence intervals, 2.554-5.884; P < 0.001). The characteristics of graft failure included exacerbation of ocular surface inflammation, neovascularization, and opacification. Epithelial defects, high IOP, and endothelial decompensation were observed at an increased frequency in the CMV+ group (all P < 0.005). Recurrent CMV infection presented as early endothelial infection in the CMV+ group. Recurrence of CMV infection was confined to the graft endothelium without involving the stroma and epithelium post-repeat endothelial keratoplasty. CONCLUSIONS CMV infection post-keratoplasty leads to persistent endothelial damage and graft opacification and significantly increases the risk of repeat graft failure. Localized recurrence of CMV infection in the endothelial grafts underscores the importance of monitoring and treatment. TRIAL REGISTRATION Chictr.org.cn, ChiCTR1800014684.
Collapse
Affiliation(s)
- Yunxiao Zang
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, No. 49 Garden North Road, Haidian, Beijing, 100191, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Yaning Zhao
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, No. 49 Garden North Road, Haidian, Beijing, 100191, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Rongmei Peng
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, No. 49 Garden North Road, Haidian, Beijing, 100191, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Gege Xiao
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, No. 49 Garden North Road, Haidian, Beijing, 100191, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Xiaozhen Liu
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, No. 49 Garden North Road, Haidian, Beijing, 100191, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Yi Qu
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, No. 49 Garden North Road, Haidian, Beijing, 100191, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Xuanjun Zhang
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, No. 49 Garden North Road, Haidian, Beijing, 100191, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Jiaxin Zhang
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, No. 49 Garden North Road, Haidian, Beijing, 100191, China
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China
| | - Jing Hong
- Department of Ophthalmology, Peking University Eye Center, Peking University Third Hospital, No. 49 Garden North Road, Haidian, Beijing, 100191, China.
- Key Laboratory of Vision Loss and Restoration, Ministry of Education, Beijing, China.
| |
Collapse
|
6
|
Cusack R, Little E, Martin-Loeches I. Practical Lessons on Antimicrobial Therapy for Critically Ill Patients. Antibiotics (Basel) 2024; 13:162. [PMID: 38391547 PMCID: PMC10886263 DOI: 10.3390/antibiotics13020162] [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: 12/19/2023] [Revised: 01/30/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
Sepsis stands as a formidable global health challenge, with persistently elevated mortality rates in recent decades. Each year, sepsis not only contributes to heightened morbidity but also imposes substantial healthcare costs on survivors. This narrative review aims to highlight the targeted measures that can be instituted to alleviate the incidence and impact of sepsis in intensive care. Here we discuss measures to reduce nosocomial infections and the prevention of equipment and patient colonisation by resilient pathogens. The overarching global crisis of bacterial resistance to newly developed antimicrobial agents intensifies the imperative for antimicrobial stewardship and de-escalation. This urgency has been accentuated in recent years, notably during the COVID-19 pandemic, as high-dose steroids and opportunistic infections presented escalating challenges. Ongoing research into airway colonisation's role in influencing disease outcomes among critically ill patients underscores the importance of tailoring treatments to disease endotypes within heterogeneous populations, which are important lessons for intensivists in training. Looking ahead, the significance of novel antimicrobial delivery systems and drug monitoring is poised to increase. This narrative review delves into the multifaceted barriers and facilitators inherent in effectively treating critically ill patients vulnerable to nosocomial infections. The future trajectory of intensive care medicine hinges on the meticulous implementation of vigilant stewardship programs, robust infection control measures, and the continued exploration of innovative and efficient technological solutions within this demanding healthcare landscape.
Collapse
Affiliation(s)
- Rachael Cusack
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St James' Hospital, D08 NHY1 Dublin, Ireland
| | - Elizabeth Little
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St James' Hospital, D08 NHY1 Dublin, Ireland
| | - Ignacio Martin-Loeches
- Department of Intensive Care Medicine, Multidisciplinary Intensive Care Research Organization (MICRO), St James' Hospital, D08 NHY1 Dublin, Ireland
- Hospital Clinic, Universitat de Barcelona, IDIBAPS, CIBERES, 08180 Barcelona, Spain
| |
Collapse
|
7
|
Chagas RA, Silva CDS, de Cássia Teixeira Birro J, Dos Santos KV. Effects of Vancomycin Subtherapeutic Concentration on Staphylococcus aureus Isolated from Hemodialysis Patients with Low Serum Trough Concentrations. Curr Microbiol 2024; 81:65. [PMID: 38231442 DOI: 10.1007/s00284-023-03588-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/12/2023] [Indexed: 01/18/2024]
Abstract
Blood bactericidal activity and antimicrobial therapy are crucial against catheter-related infection in patients undergoing hemodialysis (HD). It is well-known that catheters colonized by biofilm-producing bacteria are a risk factor for bacteremia in HD-patients. Methicillin-resistant S. aureus bacteremia in HD-patients justify the use of vancomycin as empiric therapy. The recommended vancomycin target for therapeutic efficacy is a minimum serum concentration of 10 µg mL-1 to avoid resistance. However, subtherapeutic concentrations of vancomycin have frequently occurred in HD-patients. Thus, we aim to investigate the effect of subtherapeutic vancomycin concentration on S. aureus growth, susceptibility to antimicrobials, resistance to whole blood activity, and biofilm formation. Seventeen S. aureus strains isolated from bacteremia in HD-patients and two reference strains were exposed to a vancomycin-gradient (0-10 µg mL-1) for five consecutive days to mimic the dosing interval of vancomycin in HD-patients. After that, we observed the following: no effect on growth curve; decreased susceptibility to vancomycin and daptomycin; increased S. aureus survival to whole blood bactericidal action; and a strain-dependent biofilm production after drug exposure. In conclusion, our findings suggest that the subtherapeutic concentration of vancomycin decrease S. aureus susceptibility to vancomycin and daptomycin and increases its survival to whole blood bactericidal action.
Collapse
Affiliation(s)
- Rodrigo Altoé Chagas
- Department of Pathology, Health Sciences Center, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | - Cristiana de Souza Silva
- Department of Pathology, Health Sciences Center, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil
| | | | - Kênia Valéria Dos Santos
- Department of Pathology, Health Sciences Center, Federal University of Espírito Santo (UFES), Vitória, Espírito Santo, Brazil.
| |
Collapse
|
8
|
Weikert B, Kramer TS, Schwab F, Graf-Allgeier C, Clausmeyer JO, Wolke SI, Gastmeier P, Geffers C. Implementation of a new surveillance system for dialysis-associated infection events in outpatient dialysis facilities in Germany. J Hosp Infect 2023; 142:67-73. [PMID: 37734681 DOI: 10.1016/j.jhin.2023.08.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/29/2023] [Accepted: 08/30/2023] [Indexed: 09/23/2023]
Abstract
INTRODUCTION Patients receiving maintenance haemodialysis are at risk of catheter-related infections. Up to now, there has been no standardized surveillance tool in Germany to evaluate infection events in haemodialysis outpatients. As such, this study aimed to implement an online-based surveillance tool in outpatient dialysis facilities, and to report the first national surveillance data for haemodialysis patients in Germany from October 2019 until September 2021. METHODS Outpatient dialysis facilities reported three types of dialysis-associated infection event (DAIE): bloodstream infections, intravenous antimicrobial starts, and local access site infections. Denominator data were provided by the number of haemodialysis treatments at each facility per month. DAIE rates stratified by vascular access type were calculated. RESULTS In total, 43 outpatient dialysis facilities reported 723 DAIEs, including 63 bloodstream infections, 439 intravenous antimicrobial starts, and 221 local access site infections. The overall incidence of DAIEs was 0.51 per 1000 dialysis treatments (723/1,413,457) during the surveillance period. The overall incidence of DAIEs was 0.13 per 1000 dialysis treatments among patients with arteriovenous fistulas (AVFs; 126/990,392), 0.41 per 1000 dialysis treatments among patients with arteriovenous grafts (41/99,499), and 1.68 per 1000 dialysis treatments among patients with central venous catheters (CVCs; 535/318,757). The rate ratio of DAIEs between CVC and AVF rates was 13.2 (95% confidence interval 10.9-16.0; P<0.001). DISCUSSION These 2-year infection data represent the first standardized data of outpatient dialysis facilities in Germany. Rates of infection were highest among patients with CVCs compared with other vascular access types. This online-based surveillance tool may be helpful to identify effective targets for infection prevention measures in haemodialysis patients.
Collapse
Affiliation(s)
- B Weikert
- Institute of Hygiene and Environmental Medicine, Charité - University Hospital Berlin, Berlin, Germany; National Reference Centre for the Surveillance of Nosocomial Infections, Charité - University Hospital Berlin, Berlin, Germany.
| | - T S Kramer
- Institute of Hygiene and Environmental Medicine, Charité - University Hospital Berlin, Berlin, Germany; National Reference Centre for the Surveillance of Nosocomial Infections, Charité - University Hospital Berlin, Berlin, Germany
| | - F Schwab
- Institute of Hygiene and Environmental Medicine, Charité - University Hospital Berlin, Berlin, Germany; National Reference Centre for the Surveillance of Nosocomial Infections, Charité - University Hospital Berlin, Berlin, Germany
| | - C Graf-Allgeier
- Patienten Heimversorgung Gemeinnützige Stiftung, Bad Homburg, Germany
| | - J-O Clausmeyer
- Institute of Hygiene and Environmental Medicine, Charité - University Hospital Berlin, Berlin, Germany; National Reference Centre for the Surveillance of Nosocomial Infections, Charité - University Hospital Berlin, Berlin, Germany
| | - S I Wolke
- Institute of Hygiene and Environmental Medicine, Charité - University Hospital Berlin, Berlin, Germany; National Reference Centre for the Surveillance of Nosocomial Infections, Charité - University Hospital Berlin, Berlin, Germany
| | - P Gastmeier
- Institute of Hygiene and Environmental Medicine, Charité - University Hospital Berlin, Berlin, Germany; National Reference Centre for the Surveillance of Nosocomial Infections, Charité - University Hospital Berlin, Berlin, Germany
| | - C Geffers
- Institute of Hygiene and Environmental Medicine, Charité - University Hospital Berlin, Berlin, Germany; National Reference Centre for the Surveillance of Nosocomial Infections, Charité - University Hospital Berlin, Berlin, Germany
| |
Collapse
|
9
|
Schwartz L, de Dios Ruiz-Rosado J, Stonebrook E, Becknell B, Spencer JD. Uropathogen and host responses in pyelonephritis. Nat Rev Nephrol 2023; 19:658-671. [PMID: 37479904 PMCID: PMC10913074 DOI: 10.1038/s41581-023-00737-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/21/2023] [Indexed: 07/23/2023]
Abstract
Urinary tract infections (UTIs) are among the most common bacterial infections seen in clinical practice. The ascent of UTI-causing pathogens to the kidneys results in pyelonephritis, which can trigger kidney injury, scarring and ultimately impair kidney function. Despite sizable efforts to understand how infections develop or are cleared in the bladder, our appreciation of the mechanisms by which infections develop, progress or are eradicated in the kidney is limited. The identification of virulence factors that are produced by uropathogenic Escherichia coli to promote pyelonephritis have begun to fill this knowledge gap, as have insights into the mechanisms by which kidney tubular epithelial cells oppose uropathogenic E. coli infection to prevent or eradicate UTIs. Emerging data also illustrate how specific cellular immune responses eradicate infection whereas other immune cell populations promote kidney injury. Insights into the mechanisms by which uropathogenic E. coli circumvent host immune defences or antibiotic therapy to cause pyelonephritis is paramount to the development of new prevention and treatment strategies to mitigate pyelonephritis and its associated complications.
Collapse
Affiliation(s)
- Laura Schwartz
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| | - Juan de Dios Ruiz-Rosado
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Emily Stonebrook
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - Brian Becknell
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA
- The Ohio State University College of Medicine, Columbus, OH, USA
| | - John David Spencer
- The Kidney and Urinary Tract Center, Nationwide Children's Abigail Wexner Research Institute, Columbus, OH, USA.
- The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
10
|
Dey TK, Lindahl JF, Lundkvist Å, Grace D, Deka RP, Shome R, Bandyopadhyay S, Goyal NK, Sharma G, Shome BR. Analyses of Extended-Spectrum-β-Lactamase, Metallo-β-Lactamase, and AmpC-β-Lactamase Producing Enterobacteriaceae from the Dairy Value Chain in India. Antibiotics (Basel) 2023; 12:1449. [PMID: 37760745 PMCID: PMC10650101 DOI: 10.3390/antibiotics12091449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 09/03/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
The consumption of milk contaminated with antibiotic-resistant bacteria poses a significant health threat to humans. This study aimed to investigate the prevalence of Enterobacteriaceae producing β-lactamases (ESBL, MBL, and AmpC) in cow and buffalo milk samples from two Indian states, Haryana and Assam. A total of 401 milk samples were collected from dairy farmers and vendors in the specified districts. Microbiological assays, antibiotic susceptibility testing, and PCR-based genotyping were employed to analyze 421 Gram-negative bacterial isolates. The overall prevalence of β-lactamase genes was 10% (confidence interval (CI) (7-13)), with higher rates in Haryana (13%, CI (9-19)) compared to Assam (7%, CI (4-11)). The identified β-lactamase genes in isolates were blaCMY, blaMOX, blaFOX, blaEBC, and blaDHA, associated with AmpC production. Additionally, blaCTX-M1, blaSHV, and blaTEM were detected as ESBL producers, while blaVIM, blaIMP, blaSPM, blaSIM, and blaGIM were identified as MBL producers. Notably, Shigella spp. were the dominant β-lactamase producers among identified Enterobacteriaceae. This study highlights the presence of various prevalent β-lactamase genes in milk isolates, indicating the potential risk of antimicrobial-resistant bacteria in dairy products. The presence of β-lactam resistance raises concern as this could restrict antibiotic options for treatment. The discordance between genotypic and phenotypic methods emphasizes the necessity for comprehensive approaches that integrate both techniques to accurately assess antibiotic resistance. Urgent collaborative action incorporating rational and regulated use of antibiotics across the dairy value chain is required to address the global challenge of β-lactam resistance.
Collapse
Affiliation(s)
- Tushar Kumar Dey
- Department of Biosciences, International Livestock Research Institute, Nairobi 00100, Kenya
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, 75123 Uppsala, Sweden
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru 560064, India
| | - Johanna Frida Lindahl
- Department of Biosciences, International Livestock Research Institute, Nairobi 00100, Kenya
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, 75123 Uppsala, Sweden
- Department of Clinical Sciences, Swedish University of Agricultural Sciences, 75007 Uppsala, Sweden
| | - Åke Lundkvist
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, 75123 Uppsala, Sweden
| | - Delia Grace
- Department of Biosciences, International Livestock Research Institute, Nairobi 00100, Kenya
- Food and Markets Department, Natural Resources Institute, Chatham Maritime ME4 4TB, UK
| | - Ram Pratim Deka
- International Livestock Research Institute, Regional Office for South Asia, New Delhi 110012, India
| | - Rajeswari Shome
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru 560064, India
| | - Samiran Bandyopadhyay
- Eastern Regional Station, ICAR-Indian Veterinary Research Institute, Kolkata 700037, India
| | - Naresh Kumar Goyal
- Dairy Microbiology Division, National Dairy Research Institute, Karnal 132001, India
| | - Garima Sharma
- Department of Biosciences, International Livestock Research Institute, Nairobi 00100, Kenya
- Zoonosis Science Center, Department of Medical Biochemistry and Microbiology, Uppsala University, 75123 Uppsala, Sweden
| | - Bibek Ranjan Shome
- ICAR-National Institute of Veterinary Epidemiology and Disease Informatics, Bengaluru 560064, India
| |
Collapse
|
11
|
Alasmari F, Alasmari MS, Muwainea HM, Alomar HA, Alasmari AF, Alsanea S, Alshamsan A, Rasool MF, Alqahtani F. Physiologically-based pharmacokinetic modeling for single and multiple dosing regimens of ceftriaxone in healthy and chronic kidney disease populations: a tool for model-informed precision dosing. Front Pharmacol 2023; 14:1200828. [PMID: 37547336 PMCID: PMC10398570 DOI: 10.3389/fphar.2023.1200828] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/04/2023] [Indexed: 08/08/2023] Open
Abstract
Introduction: Ceftriaxone is one of commonly prescribed beta-lactam antibiotics with several label and off-label clinical indications. A high fraction of administered dose of ceftriaxone is excreted renally in an unchanged form, and it may accumulate significantly in patients with impaired renal functions, which may lead to toxicity. Methods: In this study, we employed a physiologically-based pharmacokinetic (PBPK) modeling, as a tool for precision dosing, to predict the biological exposure of ceftriaxone in a virtually-constructed healthy and chronic kidney disease patient populations, with subsequent dosing optimizations. We started developing the model by integrating the physicochemical properties of the drug with biological system information in a PBPK software platform. A PBPK model in an adult healthy population was developed and evaluated visually and numerically with respect to experimental pharmacokinetic data. The model performance was evaluated based on the fold error criteria of the predicted and reported values for different pharmacokinetic parameters. Then, the model was applied to predict drug exposure in CKD patient populations with various degrees of severity. Results: The developed PBPK model was able to precisely describe the pharmacokinetic behavior of ceftriaxone in adult healthy population and in mild, moderate, and severe CKD patient populations. Decreasing the dose by approximately 25% in mild and 50% in moderate to severe renal disease provided a comparable exposure to the healthy population. Based on the simulation of multiple dosing regimens in severe CKD population, it has been found that accumulation of 2 g every 24 h is lower than the accumulation of 1 g every 12 h dosing regimen. Discussion: In this study, the observed concentration time profiles and pharmacokinetic parameters for ceftriaxone were successfully reproduced by the developed PBPK model and it has been shown that PBPK modeling can be used as a tool for precision dosing to suggest treatment regimens in population with renal impairment.
Collapse
Affiliation(s)
- Fawaz Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed S. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hussa Mubarak Muwainea
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Hatun A. Alomar
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah F. Alasmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Sary Alsanea
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Aws Alshamsan
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| | - Muhammad F. Rasool
- Department of Pharmacy Practice, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, Pakistan
| | - Faleh Alqahtani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
12
|
Soraci L, Cherubini A, Paoletti L, Filippelli G, Luciani F, Laganà P, Gambuzza ME, Filicetti E, Corsonello A, Lattanzio F. Safety and Tolerability of Antimicrobial Agents in the Older Patient. Drugs Aging 2023; 40:499-526. [PMID: 36976501 PMCID: PMC10043546 DOI: 10.1007/s40266-023-01019-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/27/2023] [Indexed: 03/29/2023]
Abstract
Older patients are at high risk of infections, which often present atypically and are associated with high morbidity and mortality. Antimicrobial treatment in older individuals with infectious diseases represents a clinical challenge, causing an increasing burden on worldwide healthcare systems; immunosenescence and the coexistence of multiple comorbidities determine complex polypharmacy regimens with an increase in drug-drug interactions and spread of multidrug-resistance infections. Aging-induced pharmacokinetic and pharmacodynamic changes can additionally increase the risk of inappropriate drug dosing, with underexposure that is associated with antimicrobial resistance and overexposure that may lead to adverse effects and poor adherence because of low tolerability. These issues need to be considered when starting antimicrobial prescriptions. National and international efforts have been made towards the implementation of antimicrobial stewardship (AMS) interventions to help clinicians improve the appropriateness and safety of antimicrobial prescriptions in both acute and long-term care settings. AMS programs were shown to decrease consumption of antimicrobials and to improve safety in hospitalized patients and older nursing home residents. With the abundance of antimicrobial prescriptions and the recent emergence of multidrug resistant pathogens, an in-depth review of antimicrobial prescriptions in geriatric clinical practice is needed. This review will discuss the special considerations for older individuals needing antimicrobials, including risk factors that shape risk profiles in geriatric populations as well as an evidence-based description of antimicrobial-induced adverse events in this patient population. It will highlight agents of concern for this age group and discuss interventions to mitigate the effects of inappropriate antimicrobial prescribing.
Collapse
Affiliation(s)
- Luca Soraci
- Unit of Geriatric Medicine, IRCCS INRCA, 87100, Cosenza, Italy
| | - Antonio Cherubini
- Geriatria, Accettazione geriatrica e Centro di ricerca per l'invecchiamento, IRCCS INRCA, Ancona, Italy
| | - Luca Paoletti
- Geriatria, Accettazione geriatrica e Centro di ricerca per l'invecchiamento, IRCCS INRCA, Ancona, Italy
| | | | - Filippo Luciani
- Infectious Diseases Unit of Annunziata Hospital, Cosenza, Italy
| | - Pasqualina Laganà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | | | | | | | | |
Collapse
|
13
|
Perdigão Neto LV, Machado AS, da Silva RG, de Souza RBC, Coutinho SM, Comello F, Porto APM, Lima DS, di Gioia TSR, Castro Lima VAC, Farias LABG, Macedo MRF, Noguera SLV, Dos Anjos SN, Tonheiro CMMP, Cocentino BCB, Costa SF, Oliveira MSD. Case Report: Successful Treatment of Recurrent Urinary Tract Infection Due to Extensively Drug-Resistant Klebsiella Pneumoniae in a Kidney Transplant Recipient Using Chloramphenicol. Transplant Proc 2023; 55:654-659. [PMID: 36934054 DOI: 10.1016/j.transproceed.2023.02.019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Accepted: 02/19/2023] [Indexed: 03/17/2023]
Abstract
Effective therapies for multidrug-resistant (MDR) microorganisms, especially Gram-negative bacteria, are becoming rare. Also, solid-organ transplant recipients are at high risk of MDR Gram-negative bacilli infection. Urinary tract infections are the most frequent bacterial infections in kidney transplant recipients and are an important cause of mortality after renal transplantation. We describe a case of complicated urinary tract infection in a kidney transplant patient due to extensively drug-resistant (XDR) K. pneumoniae treated successfully with a regimen comprising a combination of chloramphenicol and ertapenem. We do not recommend chloramphenicol as a first-line choice for treating complicated urinary tract infections. Still, we believe it is an alternative for infections caused by MDR and/or XDR pathogens in renal transplant patients, as other options are nephrotoxic.
Collapse
Affiliation(s)
- Lauro Vieira Perdigão Neto
- Hospital Paulistano, São Paulo, Brazil; Department of Infectious Diseases of Hospital das Clínicas and Laboratório de Investigação Médica - LIM 49, University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | | | | | | - Luis Arthur Brasil Gadelha Farias
- Department of Infectious Diseases of Hospital das Clínicas and Laboratório de Investigação Médica - LIM 49, University of São Paulo, São Paulo, Brazil.
| | - Mariana Rolim Fernandes Macedo
- Department of Infectious Diseases of Hospital das Clínicas and Laboratório de Investigação Médica - LIM 49, University of São Paulo, São Paulo, Brazil
| | - Saidy Liceth Vasconez Noguera
- Department of Infectious Diseases of Hospital das Clínicas and Laboratório de Investigação Médica - LIM 49, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Silvia Figueiredo Costa
- Department of Infectious Diseases of Hospital das Clínicas and Laboratório de Investigação Médica - LIM 49, University of São Paulo, São Paulo, Brazil
| | - Maura Salaroli de Oliveira
- Hospital Paulistano, São Paulo, Brazil; Department of Infectious Diseases of Hospital das Clínicas and Laboratório de Investigação Médica - LIM 49, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
14
|
Bonardi S, Cabassi CS, Fiaccadori E, Cavirani S, Parisi A, Bacci C, Lamperti L, Rega M, Conter M, Marra F, Crippa C, Gambi L, Spadini C, Iannarelli M, Paladini C, Filippin N, Pasquali F. Detection of carbapenemase- and ESBL-producing Klebsiella pneumoniae from bovine bulk milk and comparison with clinical human isolates in Italy. Int J Food Microbiol 2023; 387:110049. [PMID: 36521239 DOI: 10.1016/j.ijfoodmicro.2022.110049] [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: 09/16/2022] [Revised: 11/30/2022] [Accepted: 12/01/2022] [Indexed: 12/12/2022]
Abstract
Klebsiella pneumoniae is the most common Klebsiella species infecting animals and is one of the causing agents of mastitis in cows. The rise of antimicrobial resistance in K. pneumoniae, particularly in strains producing extended-spectrum β-lactamases (ESBLs) and/or carbapenemases, is of concern worldwide. Recently (Regulation UE No 2022/1255), carbapenems and cephalosporins in combination with β-lactamase inhibitors have been reserved only to human treatments in the European Union. The aim of this study was to investigate the role of cattle as carrier of human pathogenic carbapenem-resistant (CR) and ESBL-producing K. pneumoniae. On this purpose, a study involving 150 dairy farms in Parma province (Northern Italy) and 14 non replicate K. pneumoniae isolates from patients admitted at Parma University-Hospital was planned. Four multidrug resistant (MDR) K. pneumoniae strains were detected from 258 milk filters collected between 2019 and 2021. One carbapenemase KPC-3-positive K. pneumoniae ST307 (0.4 %; 95 % CI - 0.07 - 2.2) was detected in milk filters. The isolate also harboured OXA-9, CTX-M-15 and SHV-106 determinants, together with genes conferring resistance to aminoglycosides (aac(3')-IIa, aph (3″)-Ib, aph (6)-Id), fluoroquinolones (oqxA, oqxB, qnrB1), phosphonic acids (fosA6), sulphonamides (sul2), tetracyclines (tet(A)6) and trimethoprim (dfrA14). One KPC-3-producing K. pneumoniae ST307 was identified also among the human isolates, thus suggesting a possible circulation of pathogens out of the clinical settings. The remaining three bovine isolates were MDR ESBL-producing K. pneumoniae characterized by different genomic profiles: CTX-M-15, TEM-1B and SHV-187 genes (ST513); CTX-M-15 and SHV-145 (ST307); SHV-187 and DHA-1 (ST307). Occurrence of ESBL-producing K. pneumoniae in milk filters was 1.2 % (95 % CI 0.4-3.4). All the isolates showed resistance to aminoglycosides, 3rd-generation cephalosporins, and fluoroquinolones. Among the human isolates, two multidrug resistant ESBL-producing K. pneumoniae ST307 were found, thus confirming the circulation of this high-risk lineage between humans and cattle. Our findings suggest that food-producing animals can carry human pathogenic microorganisms harboring resistance genes against carbapenems and 3rd-generation cephalosporins, even if not treated with such antimicrobials. Moreover, on the MDR K. pneumoniae farms, the antimicrobial use was much higher than the Italian median value, thus highlighting the importance of a more prudent use of antibiotics in animal productions.
Collapse
Affiliation(s)
- S Bonardi
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy.
| | - C S Cabassi
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - E Fiaccadori
- Nephrology Unit, Parma University-Hospital, Department of Medicine and Surgery, Parma University, Via Gramsci 24, 43126 Parma, Italy
| | - S Cavirani
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - A Parisi
- Istituto Zooprofilattico Sperimentale della Puglia e della Basilicata, Via Chiancolla, 1, 70017 Putignano, BA, Italy
| | - C Bacci
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - L Lamperti
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - M Rega
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - M Conter
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - F Marra
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - C Crippa
- Food Safety Unit, Department of Agricultural and Food Sciences Alma Mater Studiorum, University of Bologna, Via del Florio, 2, 40064 Ozzano dell'Emilia, BO, Italy
| | - L Gambi
- Food Safety Unit, Department of Agricultural and Food Sciences Alma Mater Studiorum, University of Bologna, Via del Florio, 2, 40064 Ozzano dell'Emilia, BO, Italy
| | - C Spadini
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - M Iannarelli
- Department of Veterinary Science, University of Parma, Strada del Taglio 10, 43126 Parma, Italy
| | - C Paladini
- National Veterinary Service, Via Vasari 13/A, 43126 Parma, Italy
| | - N Filippin
- National Veterinary Service, Via Vasari 13/A, 43126 Parma, Italy
| | - F Pasquali
- Food Safety Unit, Department of Agricultural and Food Sciences Alma Mater Studiorum, University of Bologna, Via del Florio, 2, 40064 Ozzano dell'Emilia, BO, Italy
| |
Collapse
|
15
|
Noor MN, Rahman-Shepherd A, Siddiqui AR, Aftab W, Shakoor S, Hasan R, Khan M. Socioecological factors linked with pharmaceutical incentive-driven prescribing in Pakistan. BMJ Glob Health 2023; 6:e010853. [PMID: 36731921 PMCID: PMC10175940 DOI: 10.1136/bmjgh-2022-010853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 12/02/2022] [Indexed: 02/04/2023] Open
Abstract
Pharmaceutical marketing through financial incentivisation to general practitioners (GPs) is a poorly studied health system problem in Pakistan. Pharmaceutical incentivisation is seen to be distorting GPs prescribing behaviour that can compromise the health and well-being of patients. We draw on a conceptual framework outlined in the ecological system theory to identify multiple factors linked with pharmaceutical incentivisation to GPs in Pakistan. We conducted qualitative interviews with 28 policy actors to seek their views on the health system dynamics, how they sustain pharmaceutical incentivisation and their effect on the quality of care. Our analysis revealed four interlinked factors operating at different levels and how they collectively contribute to pharmaceutical incentivisation. In addition to influences such as the increasing family needs and peers' financial success, sometimes GPs may naturally be inclined to maximise incomes by engaging in pharmaceutical incentivisation. On other hand, the pharmaceutical market dynamics that involve that competition underpinned by a profit-maximisation mindset enable pharmaceutical companies to meet GPs' desires/needs in return for prescribing their products. Inadequate monitoring and health regulations may further permit the pharmaceutical industry and GPs to sustain the incentive-driven relationship. Our findings have important implications for potential health reforms such as introducing regulatory controls, and appropriate monitoring and regulation of the private health sector, required to address pharmaceutical incentivisation to GPs.
Collapse
Affiliation(s)
- Muhammad Naveed Noor
- Pathology and Laboratory Medicine, Aga Khan University, Karachi, Sindh, Pakistan
- Centre for Social Research in Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Afifah Rahman-Shepherd
- Department of Global Health and Development, London School of Hygiene & Tropical Medicine, London, UK
| | | | - Wafa Aftab
- Community Health Sciences, Aga Khan University, Karachi, Sindh, Pakistan
| | - Sadia Shakoor
- Pathology and Laboratory Medicine, Aga Khan University, Karachi, Sindh, Pakistan
| | - Rumina Hasan
- Pathology and Laboratory Medicine, Aga Khan University, Karachi, Sindh, Pakistan
- Department of Global Health and Development, London School of Hygiene & Tropical Medicine, London, UK
| | - Mishal Khan
- Department of Global Health and Development, London School of Hygiene & Tropical Medicine, London, UK
| |
Collapse
|
16
|
Zou P, Liu J, Li X, Yaseen M, Yao J, Liu L, Luo L, Wang H, Shi X, Li Z, Sun T, Gao Y, Gao C, Li LL. A Membrane Curvature Modulated Lipopeptide to Broadly Combat Multidrug-Resistant Bacterial Pneumonia with Low Resistance Risk. ACS NANO 2022; 16:20545-20558. [PMID: 36375012 DOI: 10.1021/acsnano.2c07251] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The extensive spread of multidrug resistance to Gram-negative bacteria has become a huge threat to human health, where peptide-based antibacterial agents have emerged as a powerful star weapon. Here we report a lipopeptide (LP-20) constructed nanomicelle with a different antibacterial mechanism of membrane curvature modulation, which induced dynamic membrane fission resulting in acceleration and enhancement of antibacterial activity to clinically isolated ESKAPE strains, including multidrug-resistant (MDR) pathogens. The minimum inhibitory concentration was reduced to 2-10 μM, and the minimum duration for killing was shortened to less than an hour by LP-20. This is an improvement over antimicrobial peptides and traditional antibiotics, such as ciprofloxacin and tetracycline, significantly enhancing antibacterial activity for MDR, and we observed no acquisition of resistance for one month. This accelerated germicidal mechanism was attributed to multitargeting with lipopolysaccharides, phosphoethanolamine, phosphatidylglycerol, and cardiolipin, and the synergetic interactions induced a high curvature of the bacterial membrane, which facilitated simultaneously efficient damage to both inner and outer membrane. The LP-20 effectively prolonged the lifetime of myositis mice with Escherichia coli MDR and pneumonia mice with Klebsiella pneumoniae through a hepatic metabolism with ignorable toxicity. This study provides critical information for the fabrication of lipopeptide-based nano-antibiotics for the efficient control of intractable MDR caused by Gram-negative pathogens.
Collapse
Affiliation(s)
- Pengfei Zou
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing100190, China
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing100850, China
- School of Pharmacy, Weifang Medical University, Weifang261053, China
| | - Jiao Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing100190, China
- School of Pharmacy, Weifang Medical University, Weifang261053, China
| | - Xinyu Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing100190, China
| | - Muhammad Yaseen
- Institute of Chemical Sciences, University of Peshawar, Peshawar25120, KP, Pakistan
| | - Jiahui Yao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing100190, China
- Department of Pharmacy, PLA General Hospital, Center of Medicine Clinical Research, Beijing100853, China
| | - Lingling Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing100190, China
- Department of Pharmacy, PLA General Hospital, Center of Medicine Clinical Research, Beijing100853, China
| | - Lujun Luo
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing100190, China
| | - Hui Wang
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology, Beijing100190, China
| | - Xinghua Shi
- Laboratory of Theoretical and Computational Nanoscience, National Center for Nanoscience and Technology, Beijing100190, China
| | - Zhiping Li
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing100850, China
| | - Tongyi Sun
- School of Life Science and Technology, Shandong Key Laboratory of Proteins and Peptides Pharmaceutical Engineering, Shandong Universities Key Laboratory of Biopharmaceuticals, Weifang Medical University, Weifang261053, China
| | - Yuanyuan Gao
- School of Pharmacy, Weifang Medical University, Weifang261053, China
| | - Chunsheng Gao
- State key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing100850, China
| | - Li Li Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology, University of Chinese Academy of Sciences, Beijing100190, China
| |
Collapse
|
17
|
Pan X, Zhou Z, Liu B, Wu Z. A novel therapeutic concern: Antibiotic resistance genes in common chronic diseases. Front Microbiol 2022; 13:1037389. [DOI: 10.3389/fmicb.2022.1037389] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
Infections caused by multidrug-resistant bacteria carrying antibiotic resistance genes pose a severe threat to global public health and human health. In clinical practice, it has been found that human gut microbiota act as a “reservoir” of antibiotic resistance genes (ARGs) since gut microbiota contain a wide variety of ARGs, and that the structure of the gut microbiome is influenced by the profile of the drug resistance genes present. In addition, ARGs can spread within and between species of the gut microbiome in multiple ways. To better understand gut microbiota ARGs and their effects on patients with chronic diseases, this article reviews the generation of ARGs, common vectors that transmit ARGs, the characteristics of gut microbiota ARGs in common chronic diseases, their impact on prognosis, the current state of treatment for ARGs, and what should be addressed in future research.
Collapse
|
18
|
Muacevic A, Adler JR. Antibiotic Antibiogram in Patients With Complicated Urinary Tract Infections in Nephrology Unit of South Waziristan. Cureus 2022; 14:e29803. [PMID: 36337803 PMCID: PMC9619388 DOI: 10.7759/cureus.29803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2022] [Indexed: 11/06/2022] Open
Abstract
Objectives To evaluate the antibiotic antibiogram in patients with complicated urinary tract infections (cUTIs) presenting to a Nephrology unit of South Waziristan. Methods A cross-sectional study was conducted at the Department of Nephrology, Sholam, South Waziristan. The study included all patients who presented with cUTIs and the symptoms included urinary urgency, hematuria, dysuria, suprapubic discomfort, and increased frequency. Those patients with clinical manifestations but are on antibiotics within the past five days were excluded. Results A total of 158 patients were included in the study with 113 (71.5%) females and 45 (28.5%) males. A total of 95 (60%) cases had gram-negative microbes, 47 (30%) had gram-positive cocci, and 16 (10%) had candida infection. In our study, the highly prevalent uropathogenic gram-positive bacteria showed the highest sensitivity to Linezolid, Rifampicin, and Vancomycin. Methicillin-resistant staph aureus was detected in 25% of isolates. All isolates of candida were sensitive to fluconazole. Gram-negative bacteria were highly resistant to ceftazidime, cefepime, ceftriaxone, and ciprofloxacin. Conclusion The development of bacterial resistance against multiple antibiotics is a global crisis that restricts the drug of choice for the treatment of cUTIs. In our study, we showed that overall, E.coli (gram negative) and S. Aureus (gram-positive) showed variable resistance to many antibiotics including ceftazidime, cefepime, piperacillin-tazobactam, ceftriaxone, and clindamycin.
Collapse
|
19
|
Bazaid AS, Punjabi AA, Aldarhami A, Qanash H, Alsaif G, Gattan H, Barnawi H, Alharbi B, Alrashidi A, Alqadi A. Bacterial Infections among Patients with Chronic Diseases at a Tertiary Care Hospital in Saudi Arabia. Microorganisms 2022; 10:microorganisms10101907. [PMID: 36296184 PMCID: PMC9609889 DOI: 10.3390/microorganisms10101907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 09/18/2022] [Accepted: 09/23/2022] [Indexed: 11/30/2022] Open
Abstract
Infections caused by multi-drug-resistant bacteria in patients with chronic diseases have been associated with high mortality and morbidity. While few reports have evaluated bacterial infections in multiple chronic disease patients, the focus of the current study was to investigate the prevalence of bacterial infections and the susceptibility profiles of causative strains among various groups of patients suffering from chronic diseases. Microbiological reports of patients suffering from cancer, diabetes mellitus, cardiovascular diseases, kidney diseases, and skin burns were retrospectively collected from a tertiary hospital in Saudi Arabia. Approximately 54.2% of recruited patients were males, and positive urine was the most prevalent specimen associated with kidney disease patients (25%). Escherichia coli isolates were predominant among cardiovascular, kidney, and cancer patients. Staphylococcus aureus was commonly detected in diabetics and those with burns. Although resistance patterns varied based on the type of specimens and underlying diseases, Escherichia coli showed limited resistance to colistin, carbapenems, and tigecycline, while S. aureus demonstrated susceptibility to ciprofloxacin, gentamicin, and rifampin. These observations are crucial for clinicians and policymakers to ensure effective treatment plans and improve outcomes in these patients with comorbidity.
Collapse
Affiliation(s)
- Abdulrahman S. Bazaid
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Ha’il, Hail 55476, Saudi Arabia
- Correspondence: (A.S.B.); (H.Q.); Tel.: +966-16-5358200 (ext. 1713) (A.S.B.); +966-16-5351752 (H.Q.)
| | - Ahmed A. Punjabi
- Microbiology Unit, Department of Laboratory Medicine and Pathology BB, International Medical Center, Jeddah 21451, Saudi Arabia
| | - Abdu Aldarhami
- Department of Medical Microbiology, Qunfudah Faculty of Medicine, Umm Al-Qura University, Al-Qunfudah 21961, Saudi Arabia
| | - Husam Qanash
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Ha’il, Hail 55476, Saudi Arabia
- Molecular Diagnostics and Personalized Therapeutics Unit, University of Ha’il, Hail 55476, Saudi Arabia
- Correspondence: (A.S.B.); (H.Q.); Tel.: +966-16-5358200 (ext. 1713) (A.S.B.); +966-16-5351752 (H.Q.)
| | - Ghaida Alsaif
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Ha’il, Hail 55476, Saudi Arabia
| | - Hattan Gattan
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
- Special Infectious Agents Unit, King Fahad Medical Research Center, Jeddah 22252, Saudi Arabia
| | - Heba Barnawi
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Ha’il, Hail 55476, Saudi Arabia
| | - Bandar Alharbi
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Ha’il, Hail 55476, Saudi Arabia
| | - Abdulaziz Alrashidi
- Department of Medical Laboratory Science, College of Applied Medical Sciences, University of Ha’il, Hail 55476, Saudi Arabia
| | | |
Collapse
|
20
|
Chronic Kidney Disease-An Underestimated Risk Factor for Antimicrobial Resistance in Patients with Urinary Tract Infections. Biomedicines 2022; 10:biomedicines10102368. [PMID: 36289631 PMCID: PMC9598187 DOI: 10.3390/biomedicines10102368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/18/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
(1) Background: Chronic kidney disease (CKD), as well as antimicrobial resistance (AMR) represent major global health problems, with important social and economic implications. It was reported that CKD is a risk factor for antimicrobial resistance, but evidence is scarce. In addition, CKD is recognized to be a risk factor for complicated urinary tract infections (UTIs). (2) Methods: We conducted an observational study on 564 adult in-hospital patients diagnosed with urinary tract infections. The aim of the study was to identify the risk factors for AMR, as well as multiple drug resistance (MDR) and the implicated resistance patterns. (3) Results: The mean age was 68.63 ± 17.2 years. The most frequently isolated uropathogens were Escherichia coli strains (68.3%) followed by Klebsiella species (spp. (11.2%). In 307 cases (54.4%)), the UTIs were determined by antibiotic-resistant bacteria (ARBs) and 169 cases (30%) were UTIs with MDR strains. Increased age (≥65) OR 2.156 (95% CI: 1.404−3.311), upper urinary tract obstruction OR 1.666 (1.083−2.564), indwelling urinary catheters OR 6.066 (3.919−9.390), chronic kidney disease OR 2.696 (1.832−3.969), chronic hemodialysis OR 4.955 (1.828−13.435) and active malignancies OR 1.962 (1.087−3.540) were independent risk factors for MDR UTIs. In a multivariate logistic regression model, only indwelling urinary catheters (OR 5.388, 95% CI: 3.294−8.814, p < 0.001), CKD (OR 1.779, 95% CI: 1.153−2.745, p = 0.009) and chronic hemodialysis (OR 4.068, 95% 1.413−11.715, p = 0.009) were risk factors for UTIs caused by MDR uropathogens. (4) Conclusions: CKD is an important risk factor for overall antimicrobial resistance, but also for multiple-drug resistance.
Collapse
|
21
|
Commercialized artemisinin derivatives combined with colistin protect against critical Gram-negative bacterial infection. Commun Biol 2022; 5:931. [PMID: 36076060 PMCID: PMC9458715 DOI: 10.1038/s42003-022-03898-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 08/25/2022] [Indexed: 11/25/2022] Open
Abstract
The emergence and spread of the mcr-1 gene and its mutants has immensely compromised the efficient usage of colistin for the treatment of drug-resistant Gram-negative bacterial infection in clinical settings. However, there are currently no clinically available colistin synergis. Here we identify artemisinin derivatives, such as dihydroartemisinin (DHA), that produces a synergistic antibacterial effect with colistin against the majority of Gram-negative bacteria (FIC < 0.5) without induced resistance, particularly those carrying the mcr-1 gene. Mechanism analysis reveals the direct engagement of DHA with the active center of MCR-1 to inhibit the activity of MCR-1. Meanwhile, the results from transcriptome and electron microscope analysis show that DHA could also simultaneously affect the flagellar assembly and the energy metabolism of bacteria. Moreover, in the mouse infection models of Gram-negative bacteria, combination therapy shows remarkable treatment benefits, as shown by an improved survival rate, reduced morbidity, alleviated pathological injury and decreased bacterial loading. Due to the generally safe profile of specialized malaria medication administration in humans, artemisinin derivatives are a promising class of multi-target inhibitors on bacterial resistance and virulence that can be used to extend the usage life of colistin and to tackle the inevitability of serious bacterial infection with colistin. The anti-malaria drugs artemisinin derivatives produce a synergistic antibacterial effect with colistin against Gram-negative bacteria by simultaneously inhibiting MCR-1 function and hindering flagella assembly and energy metabolism.
Collapse
|
22
|
Bankaitis VA, Tripathi A, Chen XR, Igumenova TI. New strategies for combating fungal infections: Inhibiting inositol lipid signaling by targeting Sec14 phosphatidylinositol transfer proteins. Adv Biol Regul 2022; 84:100891. [PMID: 35240534 PMCID: PMC9149032 DOI: 10.1016/j.jbior.2022.100891] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 02/22/2022] [Indexed: 12/13/2022]
Abstract
Virulent fungi represent a particularly difficult problem in the infectious disease arena as these organisms are eukaryotes that share many orthologous activities with their human hosts. The fact that these activities are often catalyzed by conserved proteins places additional demands on development of pharmacological strategies for specifically inhibiting target fungal activities without imposing undesirable secondary effects on the host. While deployment of a limited set of anti-mycotics has to date satisfied the clinical needs for treatment of fungal infections, the recent emergence of multi-drug resistant fungal 'superbugs' now poses a serious global health threat with rapidly diminishing options for treatment. This escalating infectious disease problem emphasizes the urgent need for development of new classes of anti-mycotics. In that regard, Sec14 phosphatidylinositol transfer proteins offer interesting possibilities for interfering with fungal phosphoinositide signaling with exquisite specificity and without targeting the highly conserved lipid kinases responsible for phosphoinositide production. Herein, we review the establishment of proof-of-principle that demonstrates the feasibility of such an approach. We also describe the lead compounds of four chemotypes that directly target fungal Sec14 proteins. The rules that pertain to the mechanism(s) of Sec14 inhibition by validated small molecule inhibitors, and the open questions that remain, are discussed - as are the challenges that face development of next generation Sec14-directed inhibitors.
Collapse
Affiliation(s)
- Vytas A Bankaitis
- Department of Molecular & Cellular Medicine, Texas A&M University, College Station, TX, 77843-0014, USA; Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843-0014, USA.
| | - Ashutosh Tripathi
- Department of Molecular & Cellular Medicine, Texas A&M University, College Station, TX, 77843-0014, USA
| | - Xiao-Ru Chen
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843-0014, USA
| | - Tatyana I Igumenova
- Department of Biochemistry & Biophysics, Texas A&M University, College Station, TX, 77843-0014, USA
| |
Collapse
|
23
|
Li PKT, Chow KM, Cho Y, Fan S, Figueiredo AE, Harris T, Kanjanabuch T, Kim YL, Madero M, Malyszko J, Mehrotra R, Okpechi IG, Perl J, Piraino B, Runnegar N, Teitelbaum I, Wong JKW, Yu X, Johnson DW. ISPD peritonitis guideline recommendations: 2022 update on prevention and treatment. Perit Dial Int 2022; 42:110-153. [PMID: 35264029 DOI: 10.1177/08968608221080586] [Citation(s) in RCA: 240] [Impact Index Per Article: 80.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Peritoneal dialysis (PD)-associated peritonitis is a serious complication of PD and prevention and treatment of such is important in reducing patient morbidity and mortality. The ISPD 2022 updated recommendations have revised and clarified definitions for refractory peritonitis, relapsing peritonitis, peritonitis-associated catheter removal, PD-associated haemodialysis transfer, peritonitis-associated death and peritonitis-associated hospitalisation. New peritonitis categories and outcomes including pre-PD peritonitis, enteric peritonitis, catheter-related peritonitis and medical cure are defined. The new targets recommended for overall peritonitis rate should be no more than 0.40 episodes per year at risk and the percentage of patients free of peritonitis per unit time should be targeted at >80% per year. Revised recommendations regarding management of contamination of PD systems, antibiotic prophylaxis for invasive procedures and PD training and reassessment are included. New recommendations regarding management of modifiable peritonitis risk factors like domestic pets, hypokalaemia and histamine-2 receptor antagonists are highlighted. Updated recommendations regarding empirical antibiotic selection and dosage of antibiotics and also treatment of peritonitis due to specific microorganisms are made with new recommendation regarding adjunctive oral N-acetylcysteine therapy for mitigating aminoglycoside ototoxicity. Areas for future research in prevention and treatment of PD-related peritonitis are suggested.
Collapse
Affiliation(s)
- Philip Kam-Tao Li
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Kai Ming Chow
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Carol and Richard Yu Peritoneal Dialysis Research Centre, Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
| | - Yeoungjee Cho
- Australasian Kidney Trials Network, University of Queensland, Brisbane, Australia
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| | - Stanley Fan
- Translational Medicine and Therapeutic, William Harvey Research Institute, Queen Mary University, London, UK
| | - Ana E Figueiredo
- Nursing School Escola de Ciências da Saúde e da Vida Pontificia Universidade Catolica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Tess Harris
- Polycystic Kidney Disease Charity, London, UK
| | - Talerngsak Kanjanabuch
- Division of Nephrology, Department of Medicine, Chulalongkorn University, Bangkok, Thailand
- Center of Excellence in Kidney Metabolic Disorders, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Yong-Lim Kim
- Department of Internal Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
| | - Magdalena Madero
- Division of Nephrology, Department of Medicine, National Heart Institute, Mexico City, Mexico
| | - Jolanta Malyszko
- Department of Nephrology, Dialysis and Internal Diseases, The Medical University of Warsaw, Poland
| | - Rajnish Mehrotra
- Division of Nephrology, Department of Medicine, Harborview Medical Center, University of Washington, Seattle, Washington, DC, USA
| | - Ikechi G Okpechi
- Department of Medicine, Faculty of Health Sciences, University of Cape Town and Groote Schuur Hospital, South Africa
| | - Jeff Perl
- St Michael's Hospital, University of Toronto, ON, Canada
| | - Beth Piraino
- Department of Medicine, Renal Electrolyte Division, University of Pittsburgh, PA, USA
| | - Naomi Runnegar
- Infectious Management Services, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia
| | - Isaac Teitelbaum
- Division of Nephrology, Department of Medicine, University of Colorado, Aurora, CO, USA
| | | | - Xueqing Yu
- Department of Nephrology, Guangdong Provincial People's Hospital, Guangzhou, China
- Guangdong Academy of Medical Sciences, Guangzhou, China
| | - David W Johnson
- Australasian Kidney Trials Network, University of Queensland, Brisbane, Australia
- Department of Nephrology, Princess Alexandra Hospital, Brisbane, Australia
| |
Collapse
|
24
|
Duan L, Zhou Q, Feng Z, Zhu C, Cai Y, Wang S, Zhu M, Li J, Yuan Y, Liu X, Sun J, Yang Z, Tang L. A Regression Model to Predict Linezolid Induced Thrombocytopenia in Neonatal Sepsis Patients: A Ten-Year Retrospective Cohort Study. Front Pharmacol 2022; 13:710099. [PMID: 35185555 PMCID: PMC8850389 DOI: 10.3389/fphar.2022.710099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 01/05/2022] [Indexed: 02/04/2023] Open
Abstract
Background: Linezolid-induced thrombocytopenia (LIT) is the main factor limiting the clinical application of linezolid (LZD). The incidence and risk factors of LIT in neonatal patients were possibly different from other populations based on pathophysiological characteristics. The purpose of this study was to establish a regression model for predicting LIT in neonatal sepsis patients. Methods: We retrospectively included 518 patients and divided them into the LIT group and the non-LIT group. A logistic regression analysis was used to analyze the factors related to LIT, and a regression model was established. A receiver operating characteristic (ROC) curve was drawn to evaluate the model’s predictive value. We prospectively collected 39 patients’ data to validate the model and evaluate the effect of LZD pharmacokinetics on LIT. Results: Among the 518 patients, 103 patients (19.9%) developed LIT. The Kaplan–Meier plot revealed that the overall median time from the initiation of LZD treatment to the onset of LIT in preterm infants was much shorter when compared with term infants [10 (6, 12) vs. 13 (9.75, 16.5), p = 0.004]. Multiple logistic regression analysis indicated that the independent risk factors of LIT were lower weight at medication, younger gestational ages, late-onset sepsis, necrotizing enterocolitis, mechanical ventilation, longer durations of LZD treatment, and lower baseline of platelet level. We established the above seven-variable prediction regression model and calculated the predictive probability. The ROC curve showed that the predicted probability of combined body weight, gestational age, duration of LZD treatment, and baseline of platelet had better sensitivity (84.4%), specificity (74.2%), and maximum AUC (AUC = 0.873). LIT occurred in 9 out of 39 patients (23.1%), and the accuracies of positive and negative predictions of LIT were 88.9 and 76.7%, respectively. Compared with the non-LIT patients, the LIT patients had higher trough concentration [11.49 (6.86, 15.13) vs. 5.51 (2.80, 11.61) mg/L; p = 0.028] but lower apparent volume of distribution (Vd) [0.778 (0.687, 1.421) vs. 1.322 (1.099, 1.610) L; p = 0.010]. Conclusion: The incidence of LIT was high in neonatal sepsis patients, especially in preterm infants. LIT occurred earlier in preterm infants than in term infants. The regression model of seven variables had a high predictive value for predicting LIT. LIT was correlated with higher trough concentration and lower Vd.
Collapse
Affiliation(s)
- Lufen Duan
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Qin Zhou
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Zongtai Feng
- Neonatology Department, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Chenqi Zhu
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Yan Cai
- Neonatology Department, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Sannan Wang
- Neonatology Department, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Meiying Zhu
- Neonatology Department, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Jingjing Li
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Yunlong Yuan
- Medical Laboratory, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Xin Liu
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Jiantong Sun
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
| | - Zuming Yang
- Neonatology Department, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
- *Correspondence: Lian Tang, ; Zuming Yang,
| | - Lian Tang
- Department of Pharmacy, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou, China
- *Correspondence: Lian Tang, ; Zuming Yang,
| |
Collapse
|
25
|
Costa CFFA, Merino-Ribas A, Ferreira C, Campos C, Silva N, Pereira L, Garcia A, Azevedo Á, Mesquita RBR, Rangel AOSS, Manaia CM, Sampaio-Maia B. Characterization of Oral Enterobacteriaceae Prevalence and Resistance Profile in Chronic Kidney Disease Patients Undergoing Peritoneal Dialysis. Front Microbiol 2022; 12:736685. [PMID: 34970231 PMCID: PMC8713742 DOI: 10.3389/fmicb.2021.736685] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 11/10/2021] [Indexed: 11/28/2022] Open
Abstract
Chronic Kidney Disease (CKD) is a growing public-health concern worldwide. Patients exhibit compromised immunity and are more prone to infection than other populations. Therefore, oral colonization by clinically relevant members of the Enterobacteriaceae family, major agents of both nosocomial and dialysis-associated infections with frequent prevalence of antibiotic resistances, may constitute a serious risk. Thus, this study aimed to assess the occurrence of clinically relevant enterobacteria and their antibiotic resistance profiles in the oral cavity of CKD patients undergoing peritoneal dialysis (CKD-PD) and compare it to healthy controls. Saliva samples from all the participants were cultured on MacConkey Agar and evaluated regarding the levels of urea, ammonia, and pH. Bacterial isolates were identified and characterized for antibiotic resistance phenotype and genotype. The results showed that CKD-PD patients exhibited significantly higher salivary pH, urea, and ammonia levels than controls, that was accompanied by higher prevalence and diversity of oral enterobacteria. Out of all the species isolated, only the prevalence of Raoultella ornithinolytica varied significantly between groups, colonizing the oral cavity of approximately 30% of CKD-PD patients while absent from controls. Antibiotic resistance phenotyping revealed mostly putative intrinsic resistance phenotypes (to amoxicillin, ticarcillin, and cephalothin), and resistance to sulfamethoxazole (~43% of isolates) and streptomycin (~17%). However, all isolates were resistant to at least one of the antibiotics tested and multidrug resistance isolates were only found in CKD-PD group (31,6%). Mobile genetic elements and resistance genes were detected in isolates of the species Raoultella ornithinolytica, Klebsiella pneumoniae, Klebsiella oxytoca, Escherichia coli, and Enterobacter asburiae, mostly originated from CKD-PD patients. PD-related infection history revealed that Enterobacteriaceae were responsible for ~8% of peritonitis and ~ 16% of exit-site infections episodes in CKD-PD patients, although no association was found to oral enterobacteria colonization at the time of sampling. The results suggest that the CKD-induced alterations of the oral milieu might promote a dysbiosis of the commensal oral microbiome, namely the proliferation of clinically relevant Enterobacteriaceae potentially harboring acquired antibiotic resistance genes. This study highlights the importance of the oral cavity as a reservoir for pathobionts and antibiotic resistances in CKD patients undergoing peritoneal dialysis.
Collapse
Affiliation(s)
- Carolina F F A Costa
- Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.,Nephrology & Infectious Diseases R&D Group, INEB - Instituto de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Ana Merino-Ribas
- Nephrology & Infectious Diseases R&D Group, INEB - Instituto de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Nephrology Department, Hospital Universitari Doctor Josep Trueta, Girona, Spain
| | - Catarina Ferreira
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Carla Campos
- Instituto Português de Oncologia do Porto Francisco Gentil (IPO), Porto, Portugal
| | - Nádia Silva
- Nephrology Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Luciano Pereira
- Nephrology & Infectious Diseases R&D Group, INEB - Instituto de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Nephrology Department, Centro Hospitalar Universitário de São João, Porto, Portugal
| | - Andreia Garcia
- Nephrology & Infectious Diseases R&D Group, INEB - Instituto de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Álvaro Azevedo
- Faculdade de Medicina Dentária, Universidade do Porto, Porto, Portugal.,Laboratório para a Investigação Integrativa e Translacional em Saúde Populacional (ITR), Porto, Portugal
| | - Raquel B R Mesquita
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - António O S S Rangel
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Célia M Manaia
- Universidade Católica Portuguesa, CBQF - Centro de Biotecnologia e Química Fina - Laboratório Associado, Escola Superior de Biotecnologia, Porto, Portugal
| | - Benedita Sampaio-Maia
- Nephrology & Infectious Diseases R&D Group, INEB - Instituto de Engenharia Biomédica, i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.,Faculdade de Medicina Dentária, Universidade do Porto, Porto, Portugal
| |
Collapse
|
26
|
Nolan VC, Rafols L, Harrison J, Soldevila-Barreda JJ, Crosatti M, Garton NJ, Wegrzyn M, Timms DL, Seaton CC, Sendron H, Azmanova M, Barry NP, Pitto-Barry A, Cox JA. Indole-containing arene-ruthenium complexes with broad spectrum activity against antibiotic-resistant bacteria. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 3:100099. [PMID: 35059676 PMCID: PMC8760505 DOI: 10.1016/j.crmicr.2021.100099] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 12/13/2021] [Accepted: 12/13/2021] [Indexed: 11/10/2022] Open
Abstract
A new family of indole-containing arene ruthenium organometallic compounds are active against several bacterial species and drug resistant strains Bactericidal activity observed against various Gram negative, Gram positive and acid-fast bacteria, demonstrating broad-spectrum inhibitory activity Compound series exhibits low toxicity against human cells Shows considerable promise as next generation antibiotics
Antimicrobial resistant (AMR) bacteria are emerging and spreading globally, threatening our ability to treat common infectious diseases. The development of new classes of antibiotics able to kill or inhibit the growth of such AMR bacteria through novel mechanisms of action is therefore urgently needed. Here, a new family of indole-containing arene ruthenium organometallic compounds are screened against several bacterial species and drug resistant strains. The most active complex [(p-cym)Ru(O-cyclohexyl-1H-indole-2-carbothioate)Cl] (3) shows growth inhibition and bactericidal activity against different organisms (Acinetobacter baumannii, Mycobacterium abscessus, Mycobacterium tuberculosis, Staphylococcus aureus, Salmonella enterica serovar Typhi and Escherichia coli), demonstrating broad-spectrum inhibitory activity. Importantly, this compound series exhibits low toxicity against human cells. Owing to the novelty of the antibiotic family, their moderate cytotoxicity, and their inhibitory activity against Gram positive, Gram negative and acid-fast, antibiotic resistant microorganisms, this series shows significant promise for further development.
Collapse
|
27
|
Bacteriological Profile in Septicaemic Patients with Elevated C-reactive Protein in Intensive Care Units at A Tertiary Care Hospital. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.4.25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Sepsis, the second leading cause of death is due to infections. Intensive care units (ICUs) are having the highest burden of treating the patients with sepsis and nosocomial infections compared to other areas of hospitals. Our objective was to identify the bacteriological profile and their antibiogram of sepsis cases in all ICUs. A sum of 102 blood samples were collected from patients with clinically suspected sepsis with elevated CRP. Processed by an automated method using Bact/Alert & growth were identified by Standard guidelines. Out of 102 samples, 54 (53%) were shown positive by culture. Gram-negative bacilli were the predominant and their number were 33 (61.1% ) and the commonest organisms were from the Enterobacteriaceae family. Escherichia coli was the highest number with 15 (27.7%) followed by Klebsiella pneumoniae 10 (18.51%), & the rest were single isolates of Salmonella typhi, Proteus mirabilis and Citrobacter koseri. Nonfermenter isolated were Acinetobacter baumanii 3 (5.6%), Pseudomonas aeruginosa 2 (3.7%). The Gram-positive cocci were 17 & 32.4% of culture positivity. Coagulase-negative Staphylococcus was the highest isolated accounting for 9 (16.6%) followed by Staphylococcus aureus 6 (11.1%) and Enterococcus faecalis (3.7%). Culture positivity will be more when CRP is also included in the selection of samples for sepsis and Gram-negative bacilli are the leading cause in septicemia and organisms belonging to the Enterobacteriaceae family still dominate in septicemia infections in ICUs and a real challenge for treatment are MDRs which needs to be detected regularly by using screening tests.
Collapse
|
28
|
Hocking L, Ali GC, d’Angelo C, Deshpande A, Stevenson C, Virdee M, Guthrie S. A rapid evidence assessment exploring whether antimicrobial resistance complicates non-infectious health conditions and healthcare services, 2010-20. JAC Antimicrob Resist 2021; 3:dlab171. [PMID: 34806009 PMCID: PMC8599069 DOI: 10.1093/jacamr/dlab171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Antimicrobial resistance (AMR) is one of the greatest public health threats at this time. While there is a good understanding of the impacts of AMR on infectious diseases, an area of less focus is the effects AMR may be having on non-communicable health conditions (such as cancer) and healthcare services (such as surgery). Therefore, this study aimed to explore what impact AMR is currently having on non-communicable health conditions, or areas of health services, where AMR could be a complicating factor impacting on the ability to treat the condition and/or health outcomes. To do this, a rapid evidence assessment of the literature was conducted, involving a systematic approach to searching and reviewing the evidence. In total, 101 studies were reviewed covering surgery, organ transplants, cancer, ICUs, diabetes, paediatric patients, immunodeficiency conditions, liver and kidney disease, and physical trauma. The results showed limited research in this area and studies often use a selective population, making the results difficult to generalize. However, the evidence showed that for all health conditions and healthcare service areas reviewed, at least one study demonstrated a higher risk of death for patients with resistant infections, compared with no or drug-susceptible infections. Poor health outcomes were also associated with resistant infections in some instances, such as severe sepsis and failure of treatments, as well as a greater need for invasive medical support. While there are gaps in the evidence base requiring further research, efforts are also needed within policy and practice to better understand and overcome these challenges.
Collapse
Affiliation(s)
- Lucy Hocking
- RAND Europe, Westbrook Centre, Milton Road, Cambridge, UK
- Corresponding author. E-mail:
| | | | | | | | | | - Mann Virdee
- RAND Europe, Westbrook Centre, Milton Road, Cambridge, UK
| | - Susan Guthrie
- RAND Europe, Westbrook Centre, Milton Road, Cambridge, UK
| |
Collapse
|
29
|
Jayalath S, Magana-Arachchi D. Dysbiosis of the Human Urinary Microbiome and its Association to Diseases Affecting the Urinary System. Indian J Microbiol 2021; 62:153-166. [DOI: 10.1007/s12088-021-00991-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Accepted: 11/02/2021] [Indexed: 12/15/2022] Open
|
30
|
Abd El-Hamid El-Kady R, Waggas D, AkL A. Microbial Repercussion on Hemodialysis Catheter-Related Bloodstream Infection Outcome: A 2-Year Retrospective Study. Infect Drug Resist 2021; 14:4067-4075. [PMID: 34621127 PMCID: PMC8491864 DOI: 10.2147/idr.s333438] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/21/2021] [Indexed: 12/14/2022] Open
Abstract
Background Albeit growing technical advances in the design of hemodialysis catheters, intravascular catheter-related bloodstream infection (CRBSI) still represents an utmost clinical challenge to the health-care workers (HCWs). Data regarding the influence of the culprit organism on the scenario of CRBSI in the literature are extremely lacking. Thereby, this research was carried out. Methods We undertook a retrospective cohort study over an interval of 2 years, involving patients who underwent regular hemodialysis via catheters in the Renal Dialysis Unit (RDU) of Dr. Soliman Fakeeh Hospital (DSFH), Jeddah, Kingdom of Saudi Arabia (KSA). The study enrolled 139 patients (56.8% females and 43.2% males), with mean age of 60.79 ± 11.45 years. Results The aggregate rate of CRBSI was 5.1/1000 catheter days. Amongst the 139 study candidates confirmed of having CRBSI, while 69.8% of CRBSIs were ascribed to Gram-positive cocci, about one-third of the infectious episodes were secondary to Gram-negative bacilli. Interestingly, fever was the most common presentation of S. aureus CRBSI compared to CoNS and Gram-negative bacilli CRBSIs (20.9% versus 12.9% versus 6.5%, p= 0.0001), whereas CRBSIs due to CoNS were presented mainly with rigors (19.4%). Of note, CRBSIs caused by Gram-negative bacilli had a tendency to manifest with unusual symptoms such as vomiting or hypotension. Besides, they were more prone to involve hospitalization or ICU admission. In this study, no mortality was attributed to CRBSIs. Conclusion Our study disclosed that the illicit organism has a repercussion on the clinical presentation as well as the fate of CRBSI among hemodialysis patients. This highlights the worth of identifying the infected cases in a periodic manner, to avoid the occurrence of devastating complications. A large body of work from various hemodialysis centers should take place in the near future so as to provide more insight in this perspective.
Collapse
Affiliation(s)
- Rania Abd El-Hamid El-Kady
- Department of Medical Microbiology and Immunology, Faculty of Medicine, Mansoura University, Mansoura, Egypt.,Department of Pathological Sciences, Fakeeh College for Medical Sciences, Jeddah, Kingdom of Saudi Arabia
| | - Dania Waggas
- Department of Pathological Sciences, Fakeeh College for Medical Sciences, Jeddah, Kingdom of Saudi Arabia
| | - Ahmed AkL
- Department of Nephrology, Urology and Nephrology Center, Mansoura University, Mansoura, Egypt.,Department of Internal Medicine/Adult Nephrology, Dr. Soliman Fakeeh Hospital, Jeddah, Kingdom of Saudi Arabia
| |
Collapse
|
31
|
Souza GHDAD, Rossato L, Brito GT, Bet GMDS, Simionatto S. Carbapenem-resistant Pseudomonas aeruginosa strains: a worrying health problem in intensive care units. Rev Inst Med Trop Sao Paulo 2021; 63:e71. [PMID: 34586305 PMCID: PMC8494492 DOI: 10.1590/s1678-9946202163071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/12/2021] [Indexed: 11/22/2022] Open
Abstract
Pseudomonas aeruginosa is one of the most common bacterium with a broad spectrum of human-associated infections. It is intrinsically resistant to many antimicrobial drugs, making carbapenems crucial in clinical management. The emergence and dissemination of carbapenemases among P. aeruginosa clinical isolates is a serious public health concern as it limits the options for the treatment of bacterial infections. Here, we described the molecular and epidemiological characteristics of 28 carbapenem-resistant P. aeruginosa strains isolated from patients hospitalized in an intensive care unit (ICU). The antimicrobial susceptibility of carbapenem-resistant P. aeruginosa strains was determined by broth microdilution. The presence of resistance genes was evaluated by PCR and DNA sequencing. Additionally, alterations in genes encoding P. aeruginosa outer membrane proteins were analyzed by PCR as well as SDS-PAGE. Clinical characteristics of the patients and the economic impact of hospitalization on the public health system were evaluated. PCR amplification showed that the blaKPC-2 and blaTEM genes were identified in three isolates (11%) and blaSHV gene in two isolates (7%). Outer membrane profiles obtained by SDS-PAGE indicated that the OprD porin was either absent or was produced at very low levels. A PCR assay using oprD-specific primers failed to show the presence of mutations in this gene. P. aeruginosa strains were isolated from 28 patients, among whom 43% (12/28) had sepsis, 31% (9/28) had respiratory failure, and 31% (9/28) had systemic arterial hypertension. A high mortality rate (39%) was observed in these patients, with an average duration of hospitalization of 34.6 days and a median cost of 3.275 dollars per patient. The production of carbapenemase was not the main mechanism of resistance in these strains. All carbapenem-resistant P. aeruginosa were isolated from patients hospitalized in the ICU. Besides the high mortality rate, many patients remained hospitalized for several days, resulting in a high cost of hospitalization for the public health system. Therefore, the evolution of this resistance and its dissemination should be actively monitored among critically ill patients to improve their health conditions.
Collapse
Affiliation(s)
| | - Luana Rossato
- Universidade Federal da Grande Dourados, Laboratório de Pesquisa em Ciências da Saúde, Dourados, Mato Grosso do Sul, Brazil
| | - Gabriel Teixeira Brito
- Universidade Federal da Grande Dourados, Laboratório de Pesquisa em Ciências da Saúde, Dourados, Mato Grosso do Sul, Brazil
| | - Graciela Mendonça Dos Santos Bet
- Universidade Federal da Grande Dourados, Laboratório de Pesquisa em Ciências da Saúde, Dourados, Mato Grosso do Sul, Brazil.,Universidade Federal da Grande Dourados, Hospital Universitário, Comissão de Controle das Infecções Hospitalares, Dourados, Mato Grosso do Sul, Brazil
| | - Simone Simionatto
- Universidade Federal da Grande Dourados, Laboratório de Pesquisa em Ciências da Saúde, Dourados, Mato Grosso do Sul, Brazil
| |
Collapse
|
32
|
Abbott IJ, Dekker J, van Gorp E, Wijma RA, Raaphorst MN, Klaassen CHW, Meletiadis J, Mouton JW, Peleg AY. Impact of bacterial species and baseline resistance on fosfomycin efficacy in urinary tract infections. J Antimicrob Chemother 2021; 75:988-996. [PMID: 31873748 DOI: 10.1093/jac/dkz519] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 11/17/2019] [Accepted: 11/22/2019] [Indexed: 12/15/2022] Open
Abstract
OBJECTIVES To assess the antibacterial effects of a single 3 g oral fosfomycin dose on Escherichia coli and Klebsiella pneumoniae clinical isolates within a dynamic bladder infection model. METHODS An in vitro model simulating dynamic urinary fosfomycin concentrations was used. Target fosfomycin exposure (Cmax = 1984 mg/L and Tmax = 7.5 h) was validated by LC-MS/MS. Pharmacodynamic responses of 24 E. coli and 20 K. pneumoniae clinical isolates were examined (fosfomycin MIC ≤0.25-128 mg/L). Mutant prevention concentration (MPC), fosfomycin heteroresistance, fosfomycin resistance genes and fosA expression were examined. Pathogen kill and emergence of high-level resistance (HLR; MIC >1024 mg/L) were quantified. RESULTS Following fosfomycin exposure, 20 of 24 E. coli exhibited reductions in bacterial counts below the lower limit of quantification without regrowth, despite baseline fosfomycin MICs up to 128 mg/L. Four E. coli regrew (MIC = 4-32 mg/L) with HLR population replacement. At baseline, these isolates had detectable HLR subpopulations and MPC >1024 mg/L. All E. coli isolates were fosA negative. In contrast, 17 of 20 K. pneumoniae regrew post exposure, 6 with emergence of HLR (proportion = 0.01%-100%). The three isolates without regrowth did not have a detectable HLR subpopulation after dynamic drug-free incubation. All K. pneumoniae had MPC >1024 mg/L and were fosA positive. WGS analysis and fosA expression failed to predict fosfomycin efficacy. CONCLUSIONS E. coli and K. pneumoniae isolates demonstrate discrepant responses to a single fosfomycin dose in a dynamic bladder infection in vitro model. Treatment failure against E. coli was related to an HLR subpopulation, not identified by standard MIC testing. Activity against K. pneumoniae appeared limited, regardless of MIC testing, due to universal baseline heteroresistance.
Collapse
Affiliation(s)
- Iain J Abbott
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Jordy Dekker
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Elke van Gorp
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Rixt A Wijma
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Hospital Pharmacy, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Merel N Raaphorst
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Corné H W Klaassen
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Joseph Meletiadis
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Haidari, Athens, Greece
| | - Johan W Mouton
- Department of Medical Microbiology and Infectious Diseases, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Anton Y Peleg
- Department of Infectious Diseases, Alfred Hospital and Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
33
|
Emerging Prospects for Combating Fungal Infections by Targeting Phosphatidylinositol Transfer Proteins. Int J Mol Sci 2021; 22:ijms22136754. [PMID: 34201733 PMCID: PMC8269425 DOI: 10.3390/ijms22136754] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/11/2021] [Accepted: 06/11/2021] [Indexed: 12/27/2022] Open
Abstract
The emergence of fungal “superbugs” resistant to the limited cohort of anti-fungal agents available to clinicians is eroding our ability to effectively treat infections by these virulent pathogens. As the threat of fungal infection is escalating worldwide, this dwindling response capacity is fueling concerns of impending global health emergencies. These developments underscore the urgent need for new classes of anti-fungal drugs and, therefore, the identification of new targets. Phosphoinositide signaling does not immediately appear to offer attractive targets due to its evolutionary conservation across the Eukaryota. However, recent evidence argues otherwise. Herein, we discuss the evidence identifying Sec14-like phosphatidylinositol transfer proteins (PITPs) as unexplored portals through which phosphoinositide signaling in virulent fungi can be chemically disrupted with exquisite selectivity. Recent identification of lead compounds that target fungal Sec14 proteins, derived from several distinct chemical scaffolds, reveals exciting inroads into the rational design of next generation Sec14 inhibitors. Development of appropriately refined next generation Sec14-directed inhibitors promises to expand the chemical weaponry available for deployment in the shifting field of engagement between fungal pathogens and their human hosts.
Collapse
|
34
|
Singh SK, Kumar U, Guleria A, Kumar D. A brief overview about the use of different bioactive liposome-based drug delivery systems in Peritoneal Dialysis and some other diseases. NANO EXPRESS 2021. [DOI: 10.1088/2632-959x/abfdd1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Abstract
Peritoneal dialysis (PD) is a promising way of treatment used for patients suffering from End-Stage Renal Failure (ESRF). Liposomes are nanocarriers comprised of lipid bilayers encapsulating an aqueous core. Liposomes are extensively used as drug delivery systems and several liposomal nanomedicines have been approved for clinical applications. Nanomedicine constitutes a new direction in peritonitis prevention using peritoneal dialysis (PD). In case of PD; there is a more risk of bacterial infection in the peritoneal cavity along with subcutaneous tunnel and catheter existing site. These infections are the most common complications associated with prolonged peritoneal dialysis (PD) therapy. To prevent such complications, patients used to treat with suitable antibiotic. Nanocarriers consist of assembly of nano-sized vehicles planned to deliver encapsulated/loaded bioactive(s) to the specific target (tissues or organs) and have provided prominent improved therapeutic efficacy for PD patients. The advantage of bioactive loaded nanocarrier has the efficient capacity to deliver at target specific site in PD. This review focuses mainly on the current use of different liposomal encapsulated bioactive compounds in drug delivery systems in the case of PD and other human diseases and briefly highlights the importance and use of different liposomal encapsulated antimicrobial agents to improve the PD technique.
Collapse
|
35
|
Filipska I, Winiarska A, Knysak M, Stompór T. Contribution of Gut Microbiota-Derived Uremic Toxins to the Cardiovascular System Mineralization. Toxins (Basel) 2021; 13:toxins13040274. [PMID: 33920096 PMCID: PMC8070663 DOI: 10.3390/toxins13040274] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 02/07/2023] Open
Abstract
Chronic kidney disease (CKD) affects more than 10% of the world population and leads to excess morbidity and mortality (with cardiovascular disease as a leading cause of death). Vascular calcification (VC) is a phenomenon of disseminated deposition of mineral content within the media layer of arteries preceded by phenotypic changes in vascular smooth muscle cells (VSMC) and/or accumulation of mineral content within the atherosclerotic lesions. Medial VC results in vascular stiffness and significantly contributes to increased cardio-vascular (CV) morbidity, whereas VC of plaques may rather increase their stability. Mineral and bone disorders of CKD (CKD-MBD) contribute to VC, which is further aggravated by accumulation of uremic toxins. Both CKD-MBD and uremic toxin accumulation affect not only patients with advanced CKD (glomerular filtration rate (GFR) less than 15 mL/min/1.72 m2, end-stage kidney disease) but also those on earlier stages of a disease. The key uremic toxins that contribute to VC, i.e., p-cresyl sulphate (PCS), indoxyl sulphate (IS) and trimethylamine-N-oxide (TMAO) originate from bacterial metabolism of gut microbiota. All mentioned toxins promote VC by several mechanisms, including: Transdifferentiation and apoptosis of VSMC, dysfunction of endothelial cells, oxidative stress, interaction with local renin–angiotensin–aldosterone system or miRNA profile modification. Several attractive methods of gut microbiota manipulations have been proposed in order to modify their metabolism and to limit vascular damage (and VC) triggered by uremic toxins. Unfortunately, to date no such method was demonstrated to be effective at the level of “hard” patient-oriented or even clinically relevant surrogate endpoints.
Collapse
|
36
|
Full pathogen characterisation: species identification including the detection of virulence factors and antibiotic resistance genes via multiplex DNA-assays. Sci Rep 2021; 11:6001. [PMID: 33727586 PMCID: PMC7966752 DOI: 10.1038/s41598-021-85438-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 02/19/2021] [Indexed: 11/08/2022] Open
Abstract
Antibiotic resistances progressively cause treatment failures, and their spreading dynamics reached an alarming level. Some strains have already been classified as highly critical, e.g. the ones summarised by the acronym ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter spp.). To restrain this trend and enable effective medication, as much information as possible must be obtained in the least possible time. Here, we present a DNA microarray-based assay that screens for the most important sepsis-relevant 44 pathogenic species, 360 virulence factors (mediate pathogenicity in otherwise non-pathogenic strains), and 409 antibiotic resistance genes in parallel. The assay was evaluated with 14 multidrug resistant strains, including all ESKAPE pathogens, mainly obtained from clinical isolates. We used a cost-efficient ligation-based detection platform designed to emulate the highly specific multiplex detection of padlock probes. Results could be obtained within one day, requiring approximately 4 h for amplification, application to the microarray, and detection.
Collapse
|
37
|
Abbott IJ, Roberts JA, Meletiadis J, Peleg AY. Antimicrobial pharmacokinetics and preclinical in vitro models to support optimized treatment approaches for uncomplicated lower urinary tract infections. Expert Rev Anti Infect Ther 2020; 19:271-295. [PMID: 32820686 DOI: 10.1080/14787210.2020.1813567] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
INTRODUCTION Urinary tract infections (UTIs) are extremely common. Millions of people, particularly healthy women, are affected worldwide every year. One-in-two women will have a recurrence within 12-months of an initial UTI. Inadequate treatment risks worsening infection leading to acute pyelonephritis, bacteremia and sepsis. In an era of increasing antimicrobial resistance, it is critical to provide optimized antimicrobial treatment. AREAS COVERED Literature was searched using PubMed and Google Scholar (up to 06/2020), examining the etiology, diagnosis and oral antimicrobial therapy for uncomplicated UTIs, with emphasis on urinary antimicrobial pharmacokinetics (PK) and the application of dynamic in vitro models for the pharmacodynamic (PD) profiling of pathogen response. EXPERT OPINION The majority of antimicrobial agents included in international guidelines were developed decades ago without well-described dose-response relationships. Microbiology laboratories still apply standard diagnostic methodology that has essentially remained unchanged for decades. Furthermore, it is uncertain how relevant standard in vitro susceptibility is for predicting antimicrobial efficacy in urine. In order to optimize UTI treatments, clinicians must exploit the urine-specific PK of antimicrobial agents. Dynamic in vitro models are valuable tools to examine the PK/PD and urodynamic variables associated with UTIs, while informing uropathogen susceptibility reporting, optimized dosing schedules, clinical trials and treatment guidelines.
Collapse
Affiliation(s)
- Iain J Abbott
- Department of Infectious Diseases, the Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia
| | - Jason A Roberts
- University of Queensland Centre for Clinical Research, Faculty of Medicine, The University of Queensland, Brisbane, Australia.,School of Pharmacy, Centre for Translational Anti-infective Pharmacodynamics, The University of Queensland, Brisbane, Australia.,Department of Intensive Care Medicine, Royal Brisbane and Women's Hospital, Brisbane, Australia.,Division of Anaesthesiology Critical Care Emergency and Pain Medicine, Nîmes University Hospital, University of Montpellier, Nîmes, France
| | - Joseph Meletiadis
- Clinical Microbiology Laboratory, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, Haidari, Greece
| | - Anton Y Peleg
- Department of Infectious Diseases, the Alfred Hospital and Central Clinical School, Monash University, Melbourne, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, Australia
| |
Collapse
|
38
|
Zhou Y, Lv X, Chen M, Guo Y, Ding R, Liu B, Deng X, Wang J. Characterization of Corosolic Acid as a KPC-2 Inhibitor That Increases the Susceptibility of KPC-2-Positive Bacteria to Carbapenems. Front Pharmacol 2020; 11:1047. [PMID: 32733256 PMCID: PMC7363806 DOI: 10.3389/fphar.2020.01047] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/26/2020] [Indexed: 12/18/2022] Open
Abstract
The emergence of KPC-producing Gram-negative bacteria in clinical practice highlights the need to search for novel antimicrobials and new anti-infection strategies. In this study, we constructed a laboratory KPC-2-positive strain, E. coli BL21(DE3) (pET28a-KPC-2) and identified the activity of KPC-2 in this strain. Using enzyme inhibition assays, checkerboard MIC assays, growth curves, time-killing assays and combined disk test, we found that the natural compound corosolic acid (CA) significantly inhibited the activity of the class A β-lactamase KPC-2, which is common among clinical isolates. CA treatment increased the antibacterial or bactericidal activity of imipenem and meropenem against E. coli BL21(DE3) (pET28a-KPC-2) in vitro (FIC index = 0.17 ± 0.03 for both carbapenems). In addition, the mouse intraperitoneal infection model confirmed that the combination therapy significantly reduced the bacterial load in the livers and spleens following subcutaneous administration. Our results showed that CA can be used to extend the life of carbapenems, providing a viable strategy for severe infections caused by KPC-2-positive bacteria.
Collapse
Affiliation(s)
- Yonglin Zhou
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Xiaohong Lv
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Meishan Chen
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Yan Guo
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Rui Ding
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Bin Liu
- Jilin Institute for Food Control, Changchun, China
| | - Xuming Deng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| | - Jianfeng Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Institute of Zoonosis, Jilin University, Changchun, China
| |
Collapse
|
39
|
Fodor A, Abate BA, Deák P, Fodor L, Gyenge E, Klein MG, Koncz Z, Muvevi J, Ötvös L, Székely G, Vozik D, Makrai L. Multidrug Resistance (MDR) and Collateral Sensitivity in Bacteria, with Special Attention to Genetic and Evolutionary Aspects and to the Perspectives of Antimicrobial Peptides-A Review. Pathogens 2020; 9:pathogens9070522. [PMID: 32610480 PMCID: PMC7399985 DOI: 10.3390/pathogens9070522] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/23/2020] [Accepted: 06/23/2020] [Indexed: 12/18/2022] Open
Abstract
Antibiotic poly-resistance (multidrug-, extreme-, and pan-drug resistance) is controlled by adaptive evolution. Darwinian and Lamarckian interpretations of resistance evolution are discussed. Arguments for, and against, pessimistic forecasts on a fatal “post-antibiotic era” are evaluated. In commensal niches, the appearance of a new antibiotic resistance often reduces fitness, but compensatory mutations may counteract this tendency. The appearance of new antibiotic resistance is frequently accompanied by a collateral sensitivity to other resistances. Organisms with an expanding open pan-genome, such as Acinetobacter baumannii, Pseudomonas aeruginosa, and Klebsiella pneumoniae, can withstand an increased number of resistances by exploiting their evolutionary plasticity and disseminating clonally or poly-clonally. Multidrug-resistant pathogen clones can become predominant under antibiotic stress conditions but, under the influence of negative frequency-dependent selection, are prevented from rising to dominance in a population in a commensal niche. Antimicrobial peptides have a great potential to combat multidrug resistance, since antibiotic-resistant bacteria have shown a high frequency of collateral sensitivity to antimicrobial peptides. In addition, the mobility patterns of antibiotic resistance, and antimicrobial peptide resistance, genes are completely different. The integron trade in commensal niches is fortunately limited by the species-specificity of resistance genes. Hence, we theorize that the suggested post-antibiotic era has not yet come, and indeed might never come.
Collapse
Affiliation(s)
- András Fodor
- Department of Genetics, University of Szeged, H-6726 Szeged, Hungary;
- Correspondence: or (A.F.); (L.M.); Tel.: +36-(30)-490-9294 (A.F.); +36-(30)-271-2513 (L.M.)
| | - Birhan Addisie Abate
- Ethiopian Biotechnology Institute, Agricultural Biotechnology Directorate, Addis Ababa 5954, Ethiopia;
| | - Péter Deák
- Department of Genetics, University of Szeged, H-6726 Szeged, Hungary;
- Institute of Biochemistry, Biological Research Centre, H-6726 Szeged, Hungary
| | - László Fodor
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine, P.O. Box 22, H-1581 Budapest, Hungary;
| | - Ervin Gyenge
- Hungarian Department of Biology and Ecology, Faculty of Biology and Geology, Babeș-Bolyai University, 5-7 Clinicilor St., 400006 Cluj-Napoca, Romania; (E.G.); (G.S.)
- Institute for Research-Development-Innovation in Applied Natural Sciences, Babeș-Bolyai University, 30 Fântânele St., 400294 Cluj-Napoca, Romania
| | - Michael G. Klein
- Department of Entomology, The Ohio State University, 1680 Madison Ave., Wooster, OH 44691, USA;
| | - Zsuzsanna Koncz
- Max-Planck Institut für Pflanzenzüchtungsforschung, Carl-von-Linné-Weg 10, D-50829 Köln, Germany;
| | | | - László Ötvös
- OLPE, LLC, Audubon, PA 19403-1965, USA;
- Institute of Medical Microbiology, Semmelweis University, H-1085 Budapest, Hungary
- Arrevus, Inc., Raleigh, NC 27612, USA
| | - Gyöngyi Székely
- Hungarian Department of Biology and Ecology, Faculty of Biology and Geology, Babeș-Bolyai University, 5-7 Clinicilor St., 400006 Cluj-Napoca, Romania; (E.G.); (G.S.)
- Institute for Research-Development-Innovation in Applied Natural Sciences, Babeș-Bolyai University, 30 Fântânele St., 400294 Cluj-Napoca, Romania
- Centre for Systems Biology, Biodiversity and Bioresources, Babeș-Bolyai University, 5-7 Clinicilor St., 400006 Cluj-Napoca, Romania
| | - Dávid Vozik
- Research Institute on Bioengineering, Membrane Technology and Energetics, Faculty of Engineering, University of Veszprem, H-8200 Veszprém, Hungary; or or
| | - László Makrai
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine, P.O. Box 22, H-1581 Budapest, Hungary;
- Correspondence: or (A.F.); (L.M.); Tel.: +36-(30)-490-9294 (A.F.); +36-(30)-271-2513 (L.M.)
| |
Collapse
|
40
|
De Oliveira DMP, Forde BM, Kidd TJ, Harris PNA, Schembri MA, Beatson SA, Paterson DL, Walker MJ. Antimicrobial Resistance in ESKAPE Pathogens. Clin Microbiol Rev 2020; 23:788-99. [PMID: 32404435 DOI: 10.1111/imb.12124] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2023] Open
Abstract
Antimicrobial-resistant ESKAPE ( Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) pathogens represent a global threat to human health. The acquisition of antimicrobial resistance genes by ESKAPE pathogens has reduced the treatment options for serious infections, increased the burden of disease, and increased death rates due to treatment failure and requires a coordinated global response for antimicrobial resistance surveillance. This looming health threat has restimulated interest in the development of new antimicrobial therapies, has demanded the need for better patient care, and has facilitated heightened governance over stewardship practices.
Collapse
Affiliation(s)
- David M P De Oliveira
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| | - Brian M Forde
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| | - Timothy J Kidd
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| | - Patrick N A Harris
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
- UQ Centre for Clinical Research, The University of Queensland, QLD, Australia
| | - Mark A Schembri
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| | - Scott A Beatson
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| | - David L Paterson
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
- UQ Centre for Clinical Research, The University of Queensland, QLD, Australia
| | - Mark J Walker
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| |
Collapse
|
41
|
De Oliveira DMP, Forde BM, Kidd TJ, Harris PNA, Schembri MA, Beatson SA, Paterson DL, Walker MJ. Antimicrobial Resistance in ESKAPE Pathogens. Clin Microbiol Rev 2020; 33:e00181-19. [PMID: 32404435 PMCID: PMC7227449 DOI: 10.1128/cmr.00181-19] [Citation(s) in RCA: 958] [Impact Index Per Article: 191.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Antimicrobial-resistant ESKAPE ( Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) pathogens represent a global threat to human health. The acquisition of antimicrobial resistance genes by ESKAPE pathogens has reduced the treatment options for serious infections, increased the burden of disease, and increased death rates due to treatment failure and requires a coordinated global response for antimicrobial resistance surveillance. This looming health threat has restimulated interest in the development of new antimicrobial therapies, has demanded the need for better patient care, and has facilitated heightened governance over stewardship practices.
Collapse
Affiliation(s)
- David M P De Oliveira
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| | - Brian M Forde
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| | - Timothy J Kidd
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| | - Patrick N A Harris
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
- UQ Centre for Clinical Research, The University of Queensland, QLD, Australia
| | - Mark A Schembri
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| | - Scott A Beatson
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| | - David L Paterson
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
- UQ Centre for Clinical Research, The University of Queensland, QLD, Australia
| | - Mark J Walker
- School of Chemistry and Molecular Biosciences, The University of Queensland, QLD, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, QLD, Australia
| |
Collapse
|
42
|
Wang M, Gao R, Sang P, Odom T, Zheng M, Shi Y, Xu H, Cao C, Cai J. Dimeric γ-AApeptides With Potent and Selective Antibacterial Activity. Front Chem 2020; 8:441. [PMID: 32596202 PMCID: PMC7304243 DOI: 10.3389/fchem.2020.00441] [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: 03/26/2020] [Accepted: 04/27/2020] [Indexed: 12/30/2022] Open
Abstract
Over the past few decades, the emergence of antibiotic resistance developed by life-threatening bacteria has become increasingly prevalent. Thus, there is an urgent demand to develop novel antibiotics capable of mitigating this trend. Herein, we report a series of dimeric γ-AApeptide derivatives as potential antibiotic agents with limited toxicity and excellent selectivity against Gram-positive strains. Among them, compound 2 was identified to have the best MICs without inducing drug resistance, even after exposure to MRSA for 20 passages. Time-kill kinetics and mechanistic studies suggested that 2 could mimic host-defense peptides (HDPs) and rapidly eradicate MRSA within 2 hours through disturbing the bacteria membrane. Meanwhile, biofilm formation was successfully inhibited even at a low concentration. Taken together, these results suggested the great potential of dimeric γ-AApeptide derivatives as antibacterial agents.
Collapse
Affiliation(s)
- Minghui Wang
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Ruixuan Gao
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Peng Sang
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Timothy Odom
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Mengmeng Zheng
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Yan Shi
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| | - Hai Xu
- College of Chemistry and Chemical Engineering, Central South University, Changsha, China
| | - Chuanhai Cao
- College of Pharmacy, University of South Florida, Tampa, FL, United States
| | - Jianfeng Cai
- Department of Chemistry, University of South Florida, Tampa, FL, United States
| |
Collapse
|
43
|
Zhou Y, Guo Y, Sun X, Ding R, Wang Y, Niu X, Wang J, Deng X. Application of Oleanolic Acid and Its Analogues in Combating Pathogenic Bacteria In Vitro/ Vivo by a Two-Pronged Strategy of β-Lactamases and Hemolysins. ACS OMEGA 2020; 5:11424-11438. [PMID: 32478231 PMCID: PMC7254530 DOI: 10.1021/acsomega.0c00460] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 04/30/2020] [Indexed: 05/06/2023]
Abstract
The rapid spread of β-lactamase-producing bacteria in clinical practice has increasingly deteriorated the performance of β-lactam antibiotics against such resistant strains. Thus, novel agents or strategies for the war against β-lactamase-producing bacteria, especially hypervirulent resistant bacteria (such as toxin-secreting Staphylococcus aureus) carrying complex β-lactamases, are urgently needed. In this study, we found that the natural compound oleanolic acid (OA) and its analogues (especially corosolic acid (CA)) significantly inhibited the activity of important β-lactamases (NDM-1, KPC-2, and VIM-1) in Enterobacteriaceae and β-lactamases (β-lactamase N1) in S. aureus. The results showed significant synergy with β-lactams against β-lactamase-positive bacteria (fractional inhibitory concentration (FIC) index <0.5). Additionally, OA treatment significantly inhibited the activity of hemolysin from various bacteria. In the mouse infection models, the combined therapy with OA and β-lactams exhibited a significant synergistic effect in the treatment of β-lactamase-producing bacteria, as evidenced by the survival rate of S. aureus- or Escherichia coli-infected mice, which increased from 25.0 to 75.0% or from 44.4 to 61.1% (CA increased to 77.8%), respectively, compared to treatment with individual β-lactams. Although OA treatment alone led to systemic protection against S. aureus-infected mice by directly targeting α-hemolysin (Hla), a relatively better therapeutic effect was observed for the combined therapy. To the best of our knowledge, this study is the first to find effective inhibitors against resistant bacterial infections with a two-pronged strategy by simultaneously targeting resistance enzymes and toxins, which may provide a promising therapeutic strategy for drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Yonglin Zhou
- Key
Laboratory of Zoonosis Research, Ministry of Education, Institute
of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
- Department
of Respiratory Medicine, The First Hospital
of Jilin University, Changchun 130021, Jilin, China
| | - Yan Guo
- Key
Laboratory of Zoonosis Research, Ministry of Education, Institute
of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
- Department
of Respiratory Medicine, The First Hospital
of Jilin University, Changchun 130021, Jilin, China
| | - Xiaodi Sun
- Key
Laboratory of Zoonosis Research, Ministry of Education, Institute
of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Rui Ding
- Key
Laboratory of Zoonosis Research, Ministry of Education, Institute
of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
| | - Yanling Wang
- Key
Laboratory of Zoonosis Research, Ministry of Education, Institute
of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
- Qingdao
Vland Biological Limited Co., LTD, Qingdao 266102, Shandong, China
| | - Xiaodi Niu
- Department
of Food Quality and Safety, Jilin University, Changchun 130062, China
| | - Jianfeng Wang
- Key
Laboratory of Zoonosis Research, Ministry of Education, Institute
of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
- Department
of Respiratory Medicine, The First Hospital
of Jilin University, Changchun 130021, Jilin, China
| | - Xuming Deng
- Key
Laboratory of Zoonosis Research, Ministry of Education, Institute
of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China
- Department
of Respiratory Medicine, The First Hospital
of Jilin University, Changchun 130021, Jilin, China
| |
Collapse
|
44
|
Zhang P, Zhu L, Cai J, Lei F, Qin JJ, Xie J, Liu YM, Zhao YC, Huang X, Lin L, Xia M, Chen MM, Cheng X, Zhang X, Guo D, Peng Y, Ji YX, Chen J, She ZG, Wang Y, Xu Q, Tan R, Wang H, Lin J, Luo P, Fu S, Cai H, Ye P, Xiao B, Mao W, Liu L, Yan Y, Liu M, Chen M, Zhang XJ, Wang X, Touyz RM, Xia J, Zhang BH, Huang X, Yuan Y, Loomba R, Liu PP, Li H. Association of Inpatient Use of Angiotensin-Converting Enzyme Inhibitors and Angiotensin II Receptor Blockers With Mortality Among Patients With Hypertension Hospitalized With COVID-19. Circ Res 2020; 126:1671-1681. [PMID: 32302265 PMCID: PMC7265882 DOI: 10.1161/circresaha.120.317134] [Citation(s) in RCA: 882] [Impact Index Per Article: 176.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Supplemental Digital Content is available in the text. Use of ACEIs (angiotensin-converting enzyme inhibitors) and ARBs (angiotensin II receptor blockers) is a major concern for clinicians treating coronavirus disease 2019 (COVID-19) in patients with hypertension.
Collapse
Affiliation(s)
- Peng Zhang
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Medical Science Research Center (P.Z., Y.-X.J., H.L.), Zhongnan Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.).,Basic Medical School, Wuhan University (P.Z., L. Lin, H.L.)
| | - Lihua Zhu
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Jingjing Cai
- Cardiology, The Third Xiangya Hospital, Central South University, Changsha, China (J. Cai)
| | - Fang Lei
- Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Juan-Juan Qin
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Jing Xie
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University
| | - Ye-Mao Liu
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Yan-Ci Zhao
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Xuewei Huang
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Lijin Lin
- Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.).,Basic Medical School, Wuhan University (P.Z., L. Lin, H.L.)
| | - Meng Xia
- Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Ming-Ming Chen
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Xu Cheng
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Xiao Zhang
- Eye Center (X.Z.), Renmin Hospital of Wuhan University
| | - Deliang Guo
- Hepatobiliary and Pancreatic Surgery (D.G., H.W., Y. Yuan), Zhongnan Hospital of Wuhan University
| | - Yuanyuan Peng
- Cardiology (Y.P.), Zhongnan Hospital of Wuhan University
| | - Yan-Xiao Ji
- Medical Science Research Center (P.Z., Y.-X.J., H.L.), Zhongnan Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Jing Chen
- Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Zhi-Gang She
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Yibin Wang
- Anesthesiology, Cardiovascular Research Laboratories, David Geffen School of Medicine, University of California, Los Angeles (Y.W.)
| | - Qingbo Xu
- Centre for Clinic Pharmacology, The William Harvey Research Institute, Queen Mary University of London, United Kingdom (Q.X.)
| | - Renfu Tan
- Wuhan Kanghuashuhai Technology Company (R.T.), Wuhan
| | - Haitao Wang
- Hepatobiliary and Pancreatic Surgery (D.G., H.W., Y. Yuan), Zhongnan Hospital of Wuhan University
| | - Jun Lin
- Gastroenterology (J.L.), Zhongnan Hospital of Wuhan University
| | - Pengcheng Luo
- Urology (P.L.), Wuhan Third Hospital & Tongren Hospital of Wuhan University
| | - Shouzhi Fu
- Intensive Care Unit (S.F.), Wuhan Third Hospital & Tongren Hospital of Wuhan University
| | | | - Ping Ye
- Cardiology, The Central Hospital of Wuhan (P.Y., M.C.)
| | - Bing Xiao
- Stomatology, Xiantao First People's Hospital (B.X.)
| | - Weiming Mao
- General Surgery, Huanggang Central Hospital, Wuhan, China (W.M.)
| | - Liming Liu
- General Surgery, Ezhou Central Hospital (L. Liu)
| | - Youqin Yan
- Infections Department, Wuhan Seventh Hospital (Y. Yan)
| | | | - Manhua Chen
- Cardiology, The Central Hospital of Wuhan (P.Y., M.C.)
| | - Xiao-Jing Zhang
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.)
| | - Xinghuan Wang
- Center for Evidence-Based and Translational Medicine (X.W.), Zhongnan Hospital of Wuhan University.,Urology (X.W.), Zhongnan Hospital of Wuhan University
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, BHF Glasgow Cardiovascular Research Centre, University of Glasgow, United Kingdom (R.M.T.)
| | - Jiahong Xia
- Cardiovascular Surgery, Union Hospital (J.Xia), Tongji Medical College, Huazhong University of Science and Technology
| | | | - Xiaodong Huang
- Gastroenterology (X.H.), Wuhan Third Hospital & Tongren Hospital of Wuhan University
| | - Yufeng Yuan
- Hepatobiliary and Pancreatic Surgery (D.G., H.W., Y. Yuan), Zhongnan Hospital of Wuhan University
| | - Rohit Loomba
- NAFLD Research Center, Division of Gastroenterology and Epidemiology, University of California San Diego, CA (L.R.)
| | - Peter P Liu
- Division of Cardiology, Department of Medicine, University of Ottawa Heart Institute, Ontario, Canada (P.P.L.)
| | - Hongliang Li
- From the Cardiology (P.Z., L.Z., J.-J.Q., J. Xie, Y.-M.L., Y.-C.Z., X. Huang, M.-M.C., X.C., Z.-G.S., X.-J.Z., H.L.), Renmin Hospital of Wuhan University.,Medical Science Research Center (P.Z., Y.-X.J., H.L.), Zhongnan Hospital of Wuhan University.,Institute of Model Animal of Wuhan University (P.Z., L.Z., F.L., J.-J.Q., Y.-M.L., Y.-C.Z., X. Huang, L. Lin, M.X., M.-M.C., X.C., Y.-X.J., J. Chen, Z.-G.S., X.-J.Z., H.L.).,Basic Medical School, Wuhan University (P.Z., L. Lin, H.L.)
| |
Collapse
|
45
|
Abstract
Bacteria are prime cell factories that can efficiently convert carbon and nitrogen sources into a large diversity of intracellular and extracellular biopolymers, such as polysaccharides, polyamides, polyesters, polyphosphates, extracellular DNA and proteinaceous components. Bacterial polymers have important roles in pathogenicity, and their varied chemical and material properties make them suitable for medical and industrial applications. The same biopolymers when produced by pathogenic bacteria function as major virulence factors, whereas when they are produced by non-pathogenic bacteria, they become food ingredients or biomaterials. Interdisciplinary research has shed light on the molecular mechanisms of bacterial polymer synthesis, identified new targets for antibacterial drugs and informed synthetic biology approaches to design and manufacture innovative materials. This Review summarizes the role of bacterial polymers in pathogenesis, their synthesis and their material properties as well as approaches to design cell factories for production of tailor-made bio-based materials suitable for high-value applications.
Collapse
Affiliation(s)
- M Fata Moradali
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL, USA
| | - Bernd H A Rehm
- Centre for Cell Factories and Biopolymers, Griffith Institute for Drug Discovery, Griffith University, Brisbane, QLD, Australia.
| |
Collapse
|
46
|
Specific NDM-1 Inhibitor of Isoliquiritin Enhances the Activity of Meropenem against NDM-1-positive Enterobacteriaceae in vitro. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17062162. [PMID: 32213926 PMCID: PMC7143545 DOI: 10.3390/ijerph17062162] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 12/14/2022]
Abstract
NDM-1-positive Enterobacteriaceae have caused serious clinical infections, with high mortality rates. Carbapenem was the ultimate expectation for the treatment of such infections in clinical practice. However, since the discovery of plasmid-mediated New Delhi metallo-β-lactamase-1 (NDM-1), the efficient therapeutic effects of carbapenems have been increasingly restricted. Here, we identified isoliquiritin, a novel specific inhibitor of the NDM-1 enzyme that restored the activity of carbapenem against NDM-1-producing E. coli isolates and K. pneumoniae isolates without affecting the growth of bacteria. A checkerboard test, growth curve assays and time-kill assays confirmed the significant synergistic effect of isoliquiritin combined with meropenem in vitro. It is worth noting that isoliquiritin only inhibited the activity of NDM-1 and had no obvious inhibitory effect on other class B metallo-β-lactamases (VIM-1) or NDM-1 mutants (NDM-5). The FIC indices of meropenem with isoliquiritin on NDM-1-positive E. coli and K. pneumoniae were all less than 0.5. Isoliquiritin had no influences on the expression of NDM-1-positive strains at concentrations below 64 µg/mL. Collectively, our results show that isoliquiritin is a potential adjuvant therapy drug that could enhance the antibacterial effect of carbapenems, such as meropenem, on NDM-1-positive Enterobacteria and lay the foundation for subsequent clinical trials.
Collapse
|
47
|
Zhou Y, Guo Y, Wen Z, Ci X, Xia L, Wang Y, Deng X, Wang J. Isoalantolactone Enhances the Antimicrobial Activity of Penicillin G against Staphylococcus aureus by Inactivating β-lactamase during Protein Translation. Pathogens 2020; 9:pathogens9030161. [PMID: 32110983 PMCID: PMC7157633 DOI: 10.3390/pathogens9030161] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 02/22/2020] [Accepted: 02/25/2020] [Indexed: 12/23/2022] Open
Abstract
β-Lactamase-positive Staphylococcus aureus is one of the most prevalent multidrug-resistant pathogens worldwide and is associated with increasing threats to clinical therapeutics and public health. Here, we showed that isoalantolactone (IAL), in combination with penicillin G, exhibited significant synergism against 21 β-lactamase-positive S. aureus strains (including methicillin resistant S. aureus). An enzyme inhibition assay, a checkerboard minimum inhibitory concentration (MIC) assay, a growth curve assay, a time-killing assay, a RT-PCR assay and Circular Dichroism (CD) spectroscopy were performed on different β-lactamases or β-lactamase-positive S. aureus strains, in vitro, to confirm the mechanism of inhibition of β-lactamase and the synergistic effects of the combination of penicillin G and IAL. All the fractional inhibitory concentration (FIC) indices of penicillin G, in combination with IAL, against β-lactamase-positive S. aureus, were less than 0.5, and ranged from 0.10 ± 0.02 to 0.38 ± 0.17. The survival rate of S. aureus-infected mice increased significantly from 35.29% to 88.24% within 144 h following multiple compound therapy approaches. Unlike sulbactam, IAL inactivated β-lactamase during protein translation, and the therapeutic effect of combination therapy with IAL and penicillin G was equivalent to that of sulbactam with penicillin G. Collectively, our results indicated that IAL is a promising and leading drug that can be used to restore the antibacterial effect of β-lactam antibiotics such as penicillin G and to address the inevitable infection caused by βlactamase-positive S. aureus.
Collapse
Affiliation(s)
- Yonglin Zhou
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun 130021, China; (Y.Z.); (Y.G.); (Z.W.); (X.C.)
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| | - Yan Guo
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun 130021, China; (Y.Z.); (Y.G.); (Z.W.); (X.C.)
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| | - Zhongmei Wen
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun 130021, China; (Y.Z.); (Y.G.); (Z.W.); (X.C.)
| | - Xinxin Ci
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun 130021, China; (Y.Z.); (Y.G.); (Z.W.); (X.C.)
| | - Lining Xia
- College of Veterinary Medicine, Xinjiang Agricultural University, Urmuqi 830052, China;
| | - Yanling Wang
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| | - Xuming Deng
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun 130021, China; (Y.Z.); (Y.G.); (Z.W.); (X.C.)
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China;
- Correspondence: (X.D.); (J.W.); Tel.: +86 431-87836161 (X.D.); +86 431-87835385 (J.W.); Fax: +86 431-87836160 (X.D.); +86 431-87836160 (J.W.)
| | - Jianfeng Wang
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun 130021, China; (Y.Z.); (Y.G.); (Z.W.); (X.C.)
- Key Laboratory of Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun 130062, China;
- Correspondence: (X.D.); (J.W.); Tel.: +86 431-87836161 (X.D.); +86 431-87835385 (J.W.); Fax: +86 431-87836160 (X.D.); +86 431-87836160 (J.W.)
| |
Collapse
|