1
|
Weingard B, Becker SL, Schneitler S, Trudzinski FC, Bals R, Wilkens H, Langer F. Risk factors for survival after lung transplantation in cystic fibrosis: impact of colonization with multidrug-resistant strains of Pseudomonas aeruginosa. Infection 2025:10.1007/s15010-025-02478-z. [PMID: 39883262 DOI: 10.1007/s15010-025-02478-z] [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: 11/15/2024] [Accepted: 01/20/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Lung transplantation is the ultimate treatment option for patients with advanced cystic fibrosis. Chronic colonization of these recipients with multidrug-resistant (MDR) pathogens may constitute a risk factor for an adverse outcome. We sought to analyze whether colonization with MDR pathogens, as outlined in the German classification of multiresistant Gram-negative bacteria (MRGN), was associated with the success of lung transplantation. METHODS We performed a monocentric retrospective analysis of 361 lung transplantations performed in Homburg, Germany, between 1995 and 2020. All recipients with a main diagnosis of cystic fibrosis (n = 69) were stratified into two groups based on colonization with Pseudomonas aeruginosa in view of MRGN before transplantation: no colonization and colonization without (n = 23) or with (n = 46) resistance to three or four antibiotic groups (3MRGN/4MRGN). Multivariable analyses were performed including various clinical parameters (preoperative data, postoperative data). RESULTS CF patients colonized with multidrug-resistant pathogens (Pseudomonas aeruginosa) classified as 3MRGN/4MRGN had poorer survival (median survival 16 years (without MRGN) versus 8 years (with MRGN), P = 0.048). Extracorporeal support (P = 0.014, HR = 2.929), re-transplantation (P = 0.023, HR = 2.303), female sex (P = 0.019, HR = 2.244) and 3MRGN/4MRGN (P = 0.036, HR = 2.376) were predictors of poor outcomes in the multivariate analysis. Co-colonization with the mold Aspergillus fumigatus was further associated with mortality risk in the 3MRGN/4MRGN group (P = 0.037, HR = 2.150). CONCLUSION Patients with cystic fibrosis and MDR colonization (Pseudomonas aeruginosa) are risk candidates for lung transplantation, targeted diagnostics and tailored anti-infective strategies are essential for survival after surgery. MDR colonization as expressed by MRGN may help to identify patients at increased risk to improve the organ allocation process.
Collapse
Affiliation(s)
- Bettina Weingard
- Internal Medicine V, Saarland University, 66421, Homburg/Saar, Germany
| | - Sören L Becker
- Institute of Medical Microbiology and Hygiene, Saarland University, 66421, Homburg/Saar, Germany
| | - Sophie Schneitler
- Institute of Medical Microbiology and Hygiene, Saarland University, 66421, Homburg/Saar, Germany
| | | | - Robert Bals
- Internal Medicine V, Saarland University, 66421, Homburg/Saar, Germany
| | - Heinrike Wilkens
- Internal Medicine V, Saarland University, 66421, Homburg/Saar, Germany
| | - Frank Langer
- Department of Thoracic Surgery, Saarland University, 66421, Homburg/Saar, Germany.
| |
Collapse
|
2
|
Ali SM, Soor TAH, Ahmed GA, Mhdin GA, Othman GA, Faiq SM. Distribution and Molecular Characterization of Antibiotic-Resistant Pseudomonas aeruginosa in Hospital Settings of Sulaymaniyah, Iraq. Pol J Microbiol 2024; 73:467-473. [PMID: 39465905 PMCID: PMC11639386 DOI: 10.33073/pjm-2024-037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/08/2024] [Indexed: 10/29/2024] Open
Abstract
Pseudomonas aeruginosa is a significant pathogen in hospital settings, notorious for its role in hospital-acquired infections and its ability to develop resistance to multiple antibiotics. This study investigates the prevalence, distribution, and antibiotic resistance gene profiles of P. aeruginosa in seven hospitals in Sulaymaniyah City. A total of 300 samples were collected from various hospital surfaces including mops, sinks, medical equipment, beds, desks, and floors. Using bacteriological, biochemical, and molecular methods, 66 isolates were confirmed as Pseudomonas species, with 26 identified as P. aeruginosa. Antibiotic susceptibility testing revealed resistance rates of 23.3% to streptomycin, 13.6% to tobramycin, 22.7% to moxifloxacin, 21.2% to levofloxacin, and 22.7% to norfloxacin. Furthermore, the antibiotic resistance gene detection showed the presence of the bla CTX-M, bla SHV, qnrB, and bla ACC-1 genes among the isolates. The study highlights a 22% contamination rate of hospital surfaces with Pseudomonas species, emphasizing the urgent need for enhanced infection control measures and targeted antimicrobial stewardship to manage and reduce the spread of multidrug-resistant P. aeruginosa.
Collapse
Affiliation(s)
- Seenaa Muhammed Ali
- Medical Laboratory Department, Technical College of Health and Medical Technology, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Taib Ahmed Hama Soor
- Medical Laboratory Department, Technical College of Health and Medical Technology, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Gashin Awat Ahmed
- Medical Laboratory Department, Technical College of Health and Medical Technology, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Glena Aziz Mhdin
- Medical Laboratory Department, Technical College of Health and Medical Technology, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Gulabakh Ali Othman
- Medical Laboratory Department, Technical College of Health and Medical Technology, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| | - Sarkhel Mhamad Faiq
- Medical Laboratory Department, Technical College of Health and Medical Technology, Sulaimani Polytechnic University, Sulaymaniyah, Iraq
| |
Collapse
|
3
|
Yamada Y, Miyazaki M, Kushima H, Hirata H, Ogawa A, Komiya Y, Hagiwara C, Nakashima A, Ishii H, Imakyure O. Effects of Prospective Audit and Feedback in Patients with Extended-Spectrum β-Lactamase-Producing Escherichia coli Bacteremia. Microorganisms 2024; 12:2275. [PMID: 39597664 PMCID: PMC11596308 DOI: 10.3390/microorganisms12112275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
Antimicrobial stewardship (AS) Guidelines by the Infectious Diseases Society of America recommend employing prospective audit and feedback (PAF) as an effective intervention in AS programs. Since July 2022, our hospital has implemented PAF for all patients with positive blood cultures, including those with extended-spectrum β-lactamase (ESBL)-producing Escherichia coli (EC) bacteremia. Our study examined the effect of PAF on clinical outcomes in patients with ESBL-EC bacteremia. We enrolled 62 patients diagnosed with ESBL-EC via blood culture who were undergoing antibiotic treatment. The patients were divided into the pre-PAF and post-PAF implementation groups. The rate of antibiotic de-escalation from broad-spectrum antibiotics to narrow-spectrum cefmetazole was significantly higher in the post-PAF group than in the pre-PAF group (80.7% vs. 32.4%, p = 0.0003). The treatment failure rate in the pre-PAF group was higher than that in the post-PAF group (38.7% vs. 12.9%, p = 0.04). The results of this study indicate that the implementation of PAF is advantageous not only in terms of process indicators but also in improved clinical outcomes, including reduced treatment failure rates. We hope that this study will encourage the implementation of PAF in more facilities to instigate a collective effort to reduce the incidence of antimicrobial resistance.
Collapse
Affiliation(s)
- Yota Yamada
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (M.M.); (H.H.); (A.O.); (C.H.); (A.N.); (O.I.)
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (H.K.); (Y.K.); (H.I.)
| | - Motoyasu Miyazaki
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (M.M.); (H.H.); (A.O.); (C.H.); (A.N.); (O.I.)
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (H.K.); (Y.K.); (H.I.)
- Department of Hospital Pharmacy, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan
| | - Hisako Kushima
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (H.K.); (Y.K.); (H.I.)
- Department of Respiratory Medicine, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan
| | - Hitomi Hirata
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (M.M.); (H.H.); (A.O.); (C.H.); (A.N.); (O.I.)
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (H.K.); (Y.K.); (H.I.)
| | - Arata Ogawa
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (M.M.); (H.H.); (A.O.); (C.H.); (A.N.); (O.I.)
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (H.K.); (Y.K.); (H.I.)
| | - Yukie Komiya
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (H.K.); (Y.K.); (H.I.)
- Department of Clinical Laboratory, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan
| | - Chika Hagiwara
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (M.M.); (H.H.); (A.O.); (C.H.); (A.N.); (O.I.)
| | - Akio Nakashima
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (M.M.); (H.H.); (A.O.); (C.H.); (A.N.); (O.I.)
- Department of Hospital Pharmacy, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan
| | - Hiroshi Ishii
- Department of Infection Control and Prevention, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (H.K.); (Y.K.); (H.I.)
- Department of Respiratory Medicine, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan
| | - Osamu Imakyure
- Department of Pharmacy, Fukuoka University Chikushi Hospital, Fukuoka 818-8502, Japan; (M.M.); (H.H.); (A.O.); (C.H.); (A.N.); (O.I.)
- Department of Hospital Pharmacy, Faculty of Pharmaceutical Sciences, Fukuoka University, Fukuoka 814-0180, Japan
| |
Collapse
|
4
|
Met CM, Hofstaedter CE, O'Keefe IP, Yang H, Moustafa DA, Sherman ME, Doi Y, Rasko DA, Sweet CR, Goldberg JB, Ernst RK. Characterization of Pseudomonas aeruginosa from subjects with diffuse panbronchiolitis. Microbiol Spectr 2024; 12:e0053024. [PMID: 39377602 PMCID: PMC11537112 DOI: 10.1128/spectrum.00530-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 08/26/2024] [Indexed: 10/09/2024] Open
Abstract
Diffuse panbronchiolitis (DPB) is a rare, idiopathic inflammatory disease primarily diagnosed in East Asian populations. DPB is characterized by diffuse pulmonary lesions, inflammation of the respiratory bronchioles, and bacterial infections of the airway. Historically, sputum cultures reveal Pseudomonas aeruginosa in 22% of DPB patients, increasing to 60% after 4 years from disease onset. Although DPB patients have a known susceptibility to respiratory P. aeruginosa infections, as is observed in other chronic lung diseases such as cystic fibrosis (CF), the characterization of DPB P. aeruginosa strains is limited. In this study, we characterized 24 strains obtained from a cohort of DPB patients for traits previously associated with virulence, including growth, motility, antibiotic susceptibility, lipopolysaccharide structure, and genomic diversity. Our cohort of DPB P. aeruginosa strains exhibits considerable genomic variability when compared with isolates from people with cystic fibrosis chronically colonized with P. aeruginosa and acute P. aeruginosa infection isolates. Similar to CF, DPB P. aeruginosa strains produce a diverse array of modified lipid A structures. Antibiotic susceptibility testing revealed increased resistance to erythromycin, a representative agent of the macrolide antibiotics used to manage DPB patients. Differences in the O-antigen type among P. aeruginosa strains collected from these different backgrounds were also observed. Ultimately, the characterization of DPB P. aeruginosa strains highlights several unique qualities of P. aeruginosa strains collected from chronically diseased airways, underscoring the challenges in treating DPB, CF, and other obstructive respiratory disease patients with P. aeruginosa infections. IMPORTANCE Diffuse panbronchiolitis (DPB), a chronic lung disease characterized by persistent P. aeruginosa infection, serves as an informative comparator to more common chronic lung diseases, such as cystic fibrosis (CF). This study aimed to better address the interplay between P. aeruginosa and chronically compromised airway environments through the examination of DPB P. aeruginosa strains, as existing literature regarding DPB is limited to case reports, case series, and clinical treatment guidelines. The evaluation of these features in the context of DPB, in tandem with prevailing knowledge of P. aeruginosa strains collected from more common chronic lung diseases (e.g., CF), can aid in the development of more effective strategies to combat respiratory P. aeruginosa infections in patients with chronic lung diseases.
Collapse
Affiliation(s)
- Charles M. Met
- Department of Microbial Pathogenesis, University of Maryland – Baltimore, Baltimore, Maryland, USA
| | - Casey E. Hofstaedter
- Department of Microbial Pathogenesis, University of Maryland – Baltimore, Baltimore, Maryland, USA
- Medical Scientist Training Program, University of Maryland – Baltimore, Baltimore, Maryland, USA
| | - Ian P. O'Keefe
- Department of Microbial Pathogenesis, University of Maryland – Baltimore, Baltimore, Maryland, USA
- Department of Biochemistry and Molecular Biology, University of Maryland – Baltimore, Baltimore, Maryland, USA
| | - Hyojik Yang
- Department of Microbial Pathogenesis, University of Maryland – Baltimore, Baltimore, Maryland, USA
| | - Dina A. Moustafa
- Department of Pediatrics, Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Matthew E. Sherman
- Department of Microbial Pathogenesis, University of Maryland – Baltimore, Baltimore, Maryland, USA
| | - Yohei Doi
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - David A. Rasko
- Department of Microbial Pathogenesis, University of Maryland – Baltimore, Baltimore, Maryland, USA
- Department of Microbiology and Immunology, Institute for Genome Sciences, University of Maryland - Baltimore, Baltimore, Maryland, USA
| | - Charles R. Sweet
- Chemistry Department, USA Naval Academy, Annapolis, Maryland, USA
| | - Joanna B. Goldberg
- Department of Pediatrics, Division of Pulmonary, Asthma, Cystic Fibrosis, and Sleep, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Robert K. Ernst
- Department of Microbial Pathogenesis, University of Maryland – Baltimore, Baltimore, Maryland, USA
| |
Collapse
|
5
|
Monroy-Pérez E, Herrera-Gabriel JP, Olvera-Navarro E, Ugalde-Tecillo L, García-Cortés LR, Moreno-Noguez M, Martínez-Gregorio H, Vaca-Paniagua F, Paniagua-Contreras GL. Molecular Properties of Virulence and Antibiotic Resistance of Pseudomonas aeruginosa Causing Clinically Critical Infections. Pathogens 2024; 13:868. [PMID: 39452738 PMCID: PMC11510431 DOI: 10.3390/pathogens13100868] [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: 09/05/2024] [Revised: 09/29/2024] [Accepted: 10/02/2024] [Indexed: 10/26/2024] Open
Abstract
The increase in the number of hospital strains of hypervirulent and multidrug resistant (MDR) Pseudomonas aeruginosa is a major health problem that reduces medical treatment options and increases mortality. The molecular profiles of virulence and multidrug resistance of P. aeruginosa-associated hospital and community infections in Mexico have been poorly studied. In this study, we analyzed the different molecular profiles associated with the virulence genotypes related to multidrug resistance and the genotypes of multidrug efflux pumps (mex) in P. aeruginosa causing clinically critical infections isolated from Mexican patients with community- and hospital-acquired infections. Susceptibility to 12 antibiotics was determined using the Kirby-Bauer method. The identification of P. aeruginosa and the detection of virulence and efflux pump system genes were performed using conventional PCR. All strains isolated from patients with hospital-acquired (n = 67) and community-acquired infections (n = 57) were multidrug resistant, mainly to beta-lactams (ampicillin [96.7%], carbenicillin [98.3%], cefalotin [97.5%], and cefotaxime [87%]), quinolones (norfloxacin [78.2%]), phenicols (chloramphenicol [91.9%]), nitrofurans (nitrofurantoin [70.9%]), aminoglycosides (gentamicin [75%]), and sulfonamide/trimethoprim (96.7%). Most strains (95.5%) isolated from patients with hospital- and community-acquired infections carried the adhesion (pilA) and biofilm formation (ndvB) genes. Outer membrane proteins (oprI and oprL) were present in 100% of cases, elastases (lasA and lasB) in 100% and 98.3%, respectively, alkaline protease (apr) and alginate (algD) in 99.1% and 97.5%, respectively, and chaperone (groEL) and epoxide hydrolase (cif) in 100% and 97.5%, respectively. Overall, 99.1% of the strains isolated from patients with hospital- and community-acquired infections carried the efflux pump system genes mexB and mexY, while 98.3% of the strains carried mexF and mexZ. These findings show a wide distribution of the virulome related to the genotypic and phenotypic profiles of antibiotic resistance and the origin of the strains isolated from patients with hospital- and community-acquired infections, demonstrating that these molecular mechanisms may play an important role in high-pathogenicity infections caused by P. aeruginosa.
Collapse
Affiliation(s)
- Eric Monroy-Pérez
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.P.H.-G.); (E.O.-N.); (L.U.-T.)
| | - Jennefer Paloma Herrera-Gabriel
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.P.H.-G.); (E.O.-N.); (L.U.-T.)
| | - Elizabeth Olvera-Navarro
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.P.H.-G.); (E.O.-N.); (L.U.-T.)
| | - Lorena Ugalde-Tecillo
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.P.H.-G.); (E.O.-N.); (L.U.-T.)
| | - Luis Rey García-Cortés
- Coordinación de Investigación del Estado de México Oriente, Insitituto Mexicano del Seguro Social, Tlalnepantla de Baz 50090, Mexico;
| | - Moisés Moreno-Noguez
- Coordinación Clínica de Educación e Investigación en Salud, Unidad de Medicina Familiar No. 55, Insitituto Mexicano del Seguro Social Estado de México Oriente, Zumpango 55600, Mexico;
| | - Héctor Martínez-Gregorio
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (H.M.-G.); (F.V.-P.)
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Felipe Vaca-Paniagua
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (H.M.-G.); (F.V.-P.)
- Laboratorio Nacional en Salud, Diagnóstico Molecular y Efecto Ambiental en Enfermedades Crónico-Degenerativas, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico
| | - Gloria Luz Paniagua-Contreras
- Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla 54090, Mexico; (J.P.H.-G.); (E.O.-N.); (L.U.-T.)
| |
Collapse
|
6
|
Yang X, Zhang H, Zhao Q, Li Q, Li T, Gao J. Total Synthesis of the Repeating Units of Highly Functionalized O-Antigens of Pseudomonas aeruginosa ATCC 27577, O10, and O19. JACS AU 2024; 4:2351-2362. [PMID: 38938791 PMCID: PMC11200240 DOI: 10.1021/jacsau.4c00321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/24/2024] [Accepted: 05/29/2024] [Indexed: 06/29/2024]
Abstract
The first total synthesis of the repeating units of the O-antigens of Pseudomonas aeruginosa ATCC 27577, O10, and O19 was achieved via a linear glycosylation strategy. This also represents the first synthesis of an oligosaccharide containing an α-linked N-acetyl-l-galactosaminuronic acid (l-GalpNAcA) unit. All of the glycosyl linkages, including three challenging 1,2-cis-glycosidic bonds of amino sugars, were effectively constructed with high to exclusive stereoselectivity, while orthogonal protection tactics were employed to facilitate regioselective glycosylations and the introduction of a variety of functionalities. An acetyl group migration phenomenon was found during the synthesis of the O-acylated repeating unit of the P. aeruginosa ATCC 27577 antigen. All synthetic targets carried an amino functional group in the linker at the reducing end, thus facilitating further regioselective elaboration and biological studies. The synthetic strategy established here should be useful for the preparation of other similar oligosaccharides.
Collapse
Affiliation(s)
- Xiaoyu Yang
- National
Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate
Chemistry and Glycobiology, Shandong University, Qingdao ,Shandong 266237, China
- NMPA
Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based
Medicine, Shandong University, Qingdao ,Shandong 266237, China
| | - Han Zhang
- Department
of Pharmacy, Shandong University of Traditional
Chinese Medicine, Jinan ,Shandong 250355, China
| | - Qingpeng Zhao
- National
Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate
Chemistry and Glycobiology, Shandong University, Qingdao ,Shandong 266237, China
- NMPA
Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based
Medicine, Shandong University, Qingdao ,Shandong 266237, China
| | - Qingjiang Li
- Department
of Chemistry, University of Massachusetts
Boston, 100 Morrissey Boulevard, Boston, Massachusetts 02125, United States
| | - Tiehai Li
- State
Key Laboratory of Chemical Biology, Shanghai Institute of Materia
Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Jian Gao
- National
Glycoengineering Research Center, Shandong Key Laboratory of Carbohydrate
Chemistry and Glycobiology, Shandong University, Qingdao ,Shandong 266237, China
- NMPA
Key Laboratory for Quality Research and Evaluation of Carbohydrate-Based
Medicine, Shandong University, Qingdao ,Shandong 266237, China
| |
Collapse
|
7
|
Agyeman AA, López-Causapé C, Rogers KE, Lucas DD, Cortés-Lara S, Gomis-Font MA, Fraile-Ribot P, Figuerola J, Lang Y, Franklyn ERT, Lee WL, Zhou J, Zhang Y, Bulitta JB, Boyce JD, Nation RL, Oliver A, Landersdorfer CB. Ceftolozane/tazobactam plus tobramycin against free-floating and biofilm bacteria of hypermutable Pseudomonas aeruginosa epidemic strains: Resistance mechanisms and synergistic activity. Int J Antimicrob Agents 2023; 62:106887. [PMID: 37315906 DOI: 10.1016/j.ijantimicag.2023.106887] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 05/26/2023] [Accepted: 06/07/2023] [Indexed: 06/16/2023]
Abstract
OBJECTIVE Acute exacerbations of biofilm-associated Pseudomonas aeruginosa infections in cystic fibrosis (CF) have limited treatment options. Ceftolozane/tazobactam (alone and with a second antibiotic) has not yet been investigated against hypermutable clinical P. aeruginosa isolates in biofilm growth. This study aimed to evaluate, using an in vitro dynamic biofilm model, ceftolozane/tazobactam alone and in combination with tobramycin at simulated representative lung fluid pharmacokinetics against free-floating (planktonic) and biofilm states of two hypermutable P. aeruginosa epidemic strains (LES-1 and CC274) from adolescents with CF. METHODS Regimens were intravenous ceftolozane/tazobactam 4.5 g/day continuous infusion, inhaled tobramycin 300 mg 12-hourly, intravenous tobramycin 10 mg/kg 24-hourly, and both ceftolozane/tazobactam-tobramycin combinations. The isolates were susceptible to both antibiotics. Total and less-susceptible free-floating and biofilm bacteria were quantified over 120-168 h. Ceftolozane/tazobactam resistance mechanisms were investigated by whole-genome sequencing. Mechanism-based modelling of bacterial viable counts was performed. RESULTS Monotherapies of ceftolozane/tazobactam and tobramycin did not sufficiently suppress emergence of less-susceptible subpopulations, although inhaled tobramycin was more effective than intravenous tobramycin. Ceftolozane/tazobactam resistance development was associated with classical (AmpC overexpression plus structural modification) and novel (CpxR mutations) mechanisms depending on the strain. Against both isolates, combination regimens demonstrated synergy and completely suppressed the emergence of ceftolozane/tazobactam and tobramycin less-susceptible free-floating and biofilm bacterial subpopulations. CONCLUSION Mechanism-based modelling incorporating subpopulation and mechanistic synergy well described the antibacterial effects of all regimens against free-floating and biofilm bacterial states. These findings support further investigation of ceftolozane/tazobactam in combination with tobramycin against biofilm-associated P. aeruginosa infections in adolescents with CF.
Collapse
Affiliation(s)
- Akosua A Agyeman
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Carla López-Causapé
- Servicio de Microbiología, Hospital Universitario Son Espases-IdISBa, Palma de Mallorca, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Kate E Rogers
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Deanna Deveson Lucas
- Monash Bioinformatics Platform, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Sara Cortés-Lara
- Servicio de Microbiología, Hospital Universitario Son Espases-IdISBa, Palma de Mallorca, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Maria A Gomis-Font
- Servicio de Microbiología, Hospital Universitario Son Espases-IdISBa, Palma de Mallorca, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Pablo Fraile-Ribot
- Servicio de Microbiología, Hospital Universitario Son Espases-IdISBa, Palma de Mallorca, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
| | - Joan Figuerola
- Servicio de Pediatría, Hospital Universitario Son Espases-IdISBa, Palma de Mallorca, Spain
| | - Yinzhi Lang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Eva R T Franklyn
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Wee Leng Lee
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Jieqiang Zhou
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Yongzhen Zhang
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - Jurgen B Bulitta
- Department of Pharmacotherapy and Translational Research, College of Pharmacy, University of Florida, Orlando, Florida, USA
| | - John D Boyce
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, Victoria, Australia
| | - Roger L Nation
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Antonio Oliver
- Servicio de Microbiología, Hospital Universitario Son Espases-IdISBa, Palma de Mallorca, Spain; CIBER Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain.
| | - Cornelia B Landersdorfer
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia; Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
| |
Collapse
|
8
|
Boaro A, Ageitos L, Torres MDT, Blasco EB, Oztekin S, de la Fuente-Nunez C. Structure-function-guided design of synthetic peptides with anti-infective activity derived from wasp venom. CELL REPORTS. PHYSICAL SCIENCE 2023; 4:101459. [PMID: 38239869 PMCID: PMC10795512 DOI: 10.1016/j.xcrp.2023.101459] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/22/2024]
Abstract
Antimicrobial peptides (AMPs) derived from natural toxins and venoms offer a promising alternative source of antibiotics. Here, through structure-function-guided design, we convert two natural AMPs derived from the venom of the solitary eumenine wasp Eumenes micado into α-helical AMPs with reduced toxicity that kill Gram-negative bacteria in vitro and in a preclinical mouse model. To identify the sequence determinants conferring antimicrobial activity, an alanine scan screen and strategic single lysine substitutions are made to the amino acid sequence of these natural peptides. These efforts yield a total of 34 synthetic derivatives, including alanine substituted and lysine-substituted sequences with stabilized α-helical structures and increased net positive charge. The resulting lead synthetic peptides kill the Gram-negative pathogens Escherichia coli and Pseudomonas aeruginosa (PAO1 and PA14) by rapidly permeabilizing both their outer and cytoplasmic membranes, exhibit anti-infective efficacy in a mouse model by reducing bacterial loads by up to three orders of magnitude, and do not readily select for bacterial resistance.
Collapse
Affiliation(s)
- Andreia Boaro
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Present address: Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, São Paulo 09210-580, Brazil
- These authors contributed equally
| | - Lucía Ageitos
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Present address: CICA - Centro Interdisciplinar de Química e Bioloxía, Departamento de Química, Facultade de Ciencias, Universidade da Coruña, 15008 A Coruña, Spain
- These authors contributed equally
| | - Marcelo Der Torossian Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Esther Broset Blasco
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sebahat Oztekin
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Present address: Faculty of Engineering, Department of Food Engineering, Bayburt University, Bayburt 69000, Turkey
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA 19104, USA
- Lead contact
| |
Collapse
|
9
|
Quaye JA, Gadda G. The Pseudomonas aeruginosa PAO1 metallo flavoprotein d-2-hydroxyglutarate dehydrogenase requires Zn 2+ for substrate orientation and activation. J Biol Chem 2023; 299:103008. [PMID: 36775127 PMCID: PMC10034468 DOI: 10.1016/j.jbc.2023.103008] [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: 12/14/2022] [Revised: 01/31/2023] [Accepted: 02/03/2023] [Indexed: 02/12/2023] Open
Abstract
Pseudomonas aeruginosa PAO1 d-2-hydroxyglutarate (D2HG) dehydrogenase (PaD2HGDH) oxidizes D2HG to 2-ketoglutarate during the vital l-serine biosynthesis and is a potential therapeutic target against P. aeruginosa. PaD2HGDH, which oxidizes d-malate as an alternative substrate, has been demonstrated to be a metallo flavoprotein that requires Zn2+ for activity. However, the role of Zn2+ in the enzyme has not been elucidated, making it difficult to rationalize why nature employs both a redox center and a metal ion for catalysis in PaD2HGDH and other metallo flavoenzymes. In this study, recombinant His-tagged PaD2HGDH was purified to high levels in the presence of Zn2+ or Co2+ to investigate the metal's role in catalysis. We found that the flavin reduction step was reversible and partially rate limiting for the enzyme's turnover at pH 7.4 with either D2HG or d-malate with similar rate constants for both substrates, irrespective of whether Zn2+ or Co2+ was bound to the enzyme. The steady-state pL profiles of the kcat and kcat/Km values with d-malate demonstrate that Zn2+ mediates the activation of water coordinated to the metal. Our data are consistent with a dual role for the metal, which orients the hydroxy acid substrate in the enzyme's active site and rapidly deprotonates the substrate to yield an alkoxide species for hydride transfer to the flavin. Thus, we propose a catalytic mechanism for PaD2HGDH oxidation that establishes Zn2+ as a cofactor required for substrate orientation and activation during enzymatic turnover.
Collapse
Affiliation(s)
- Joanna A Quaye
- Department of Chemistry, Georgia State University, Atlanta, Georgia, USA
| | - Giovanni Gadda
- Department of Chemistry, Georgia State University, Atlanta, Georgia, USA; Department of Biology, Georgia State University, Atlanta, Georgia, USA; Department of The Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, Georgia, USA.
| |
Collapse
|
10
|
Jones JD, Varghese D, Pabary R, Langley RJ. The potential of bacteriophage therapy in the treatment of paediatric respiratory infections. Paediatr Respir Rev 2022; 44:70-77. [PMID: 35241371 DOI: 10.1016/j.prrv.2022.02.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 02/08/2022] [Accepted: 02/09/2022] [Indexed: 12/14/2022]
Abstract
The looming antibiotic resistance crisis is forcing clinicians to consider alternative approaches to treating bacterial infections. As the window of use for current antimicrobial agents becomes ever narrower, we consider if looking back will now be the way forward. Conceptually, phage therapy is simple and specific; a targeted treatment to control bacterial overgrowth. In this article we discuss bacteriophage and potential use in future therapy.
Collapse
Affiliation(s)
- J D Jones
- Infection Medicine, University of Edinburgh, United Kingdom
| | - D Varghese
- Department of Paediatric Respiratory and Sleep Medicine, Royal Hospital for Children, Glasgow, United Kingdom
| | - R Pabary
- Department of Paediatric Respiratory and Sleep Medicine, Royal Brompton Hospital, London, United Kingdom
| | - R J Langley
- Department of Paediatric Respiratory and Sleep Medicine, Royal Hospital for Children, Glasgow, United Kingdom; School of Medicine, Dentistry & Nursing, University of Glasgow, United Kingdom.
| |
Collapse
|
11
|
Almutairi MBF, Alrouji M, Almuhanna Y, Asad M, Joseph B. In-Vitro and In-Vivo Antibacterial Effects of Frankincense Oil and Its Interaction with Some Antibiotics against Multidrug-Resistant Pathogens. Antibiotics (Basel) 2022; 11:1591. [PMID: 36358246 PMCID: PMC9686721 DOI: 10.3390/antibiotics11111591] [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: 10/10/2022] [Revised: 11/05/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Frankincense (Boswellia sacra oleo gum resin) is reported to possess antimicrobial activity against several pathogens in-vitro. The antimicrobial effects of frankincense oil and its interaction with imipenem and gentamicin against methicillin-resistant Staphylococcus aureus (MRSA) and multidrug-resistant P. aeruginosa were determined through in-vitro methods and an in-vivo study using a rat pneumonia model. Frankincense oil was subjected to GC-MS analysis to determine the different volatile components. Antibacterial effects against MRSA and MDR-P. aeruginosa was evaluated and its MIC and MBC were determined. For the rat pneumonia model (in-vivo), oil was administered at a dose of 500 mg/kg and 1000 mg/kg followed by determination of CFU in lung tissue and histological studies. Frankincense oil did not show a very potent inhibitory effect against MRSA or MDR-P. aeruginosa; the oil did not affect the zone of inhibition or FIC when combined with imipenem or gentamicin indicating a lack of interaction between the oil and the antibiotics. Furthermore, there was no interaction between the antibiotics and the frankincense oil in the in-vivo model. The result of the study revealed that frankincense oil has a weak inhibitory effect against MRSA and MDR-P. aeruginosa, and it did not show any interaction with imipenem or gentamicin.
Collapse
Affiliation(s)
- Megren Bin Faisal Almutairi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
- Department of Clinical Laboratory, King Saud Hospital, Unaizah 56215, Saudi Arabia
| | - Mohammed Alrouji
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Yasir Almuhanna
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Mohammed Asad
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| | - Babu Joseph
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
12
|
Whole-Genome Sequencing Reveals Diversity of Carbapenem-Resistant Pseudomonas aeruginosa Collected through CDC's Emerging Infections Program, United States, 2016-2018. Antimicrob Agents Chemother 2022; 66:e0049622. [PMID: 36066241 PMCID: PMC9487505 DOI: 10.1128/aac.00496-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The CDC's Emerging Infections Program (EIP) conducted population- and laboratory-based surveillance of US carbapenem-resistant Pseudomonas aeruginosa (CRPA) from 2016 through 2018. To characterize the pathotype, 1,019 isolates collected through this project underwent antimicrobial susceptibility testing and whole-genome sequencing. Sequenced genomes were classified using the seven-gene multilocus sequence typing (MLST) scheme and a core genome (cg)MLST scheme was used to determine phylogeny. Both chromosomal and horizontally transmitted mechanisms of carbapenem resistance were assessed. There were 336 sequence types (STs) among the 1,019 sequenced genomes, and the genomes varied by an average of 84.7% of the cgMLST alleles used. Mutations associated with dysfunction of the porin OprD were found in 888 (87.1%) of the genomes and were correlated with carbapenem resistance, and a machine learning model incorporating hundreds of genetic variations among the chromosomal mechanisms of resistance was able to classify resistant genomes. While only 7 (0.1%) isolates harbored carbapenemase genes, 66 (6.5%) had acquired non-carbapenemase β-lactamase genes, and these were more likely to have OprD dysfunction and be resistant to all carbapenems tested. The genetic diversity demonstrates that the pathotype includes a variety of strains, and clones previously identified as high-risk make up only a minority of CRPA strains in the United States. The increased carbapenem resistance in isolates with acquired non-carbapenemase β-lactamase genes suggests that horizontally transmitted mechanisms aside from carbapenemases themselves may be important drivers of the spread of carbapenem resistance in P. aeruginosa.
Collapse
|
13
|
Dahibhate NL, Shukla SK, Kumar K. A Cyclic Disulfide Diastereomer From Bioactive Fraction of Bruguiera gymnorhiza Shows Anti- Pseudomonas aeruginosa Activity. Front Pharmacol 2022; 13:890790. [PMID: 35721160 PMCID: PMC9201687 DOI: 10.3389/fphar.2022.890790] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Accepted: 04/14/2022] [Indexed: 01/08/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic pathogen that commonly causes hospital-acquired infection and is of great concern in immunocompromised patients. The quorum sensing (QS) mechanism of P. aeruginosa is well studied and known to be responsible for pathogenicity and virulence. The QS inhibitor derived from the natural product can be an important therapeutic agent for pathogen control. The present study reports the role of Bruguiera gymnorhiza purified fraction (BG138) in inhibiting virulence factor production, biofilm formation, quorum sensing molecules, and expression of QS-related genes of P. aeruginosa. Structural characterization of BG138 by high resolution mass spectrometry, Fourier transform infrared spectroscopy, 1D (1H and 13C NMR) and 2D NMR reveals that the fraction is a mixture of already known cyclic disulfide diastereomer, namely, brugierol and isobrugierol. The minimum inhibitory concentration (MIC) of BG138 against P. aeruginosa was 32 μg/ml. Biofilm formation was significantly reduced at sub-MIC concentrations of BG138. Scanning electron microscopy analysis reports the concentration-dependent biofilm inhibition and morphological changes of P. aeruginosa. Flow cytometry–based cell viability assay showed that P. aeruginosa cells exhibit increased propidium iodide uptake on treatment with 32 and 64 μg/ml of BG138. At sub-MIC concentrations, BG138 exhibited significant inhibition of virulence factors and reduced swimming and swarming motility of P. aeruginosa. Furthermore, the effect of BG138 on the expression of QS-related genes was investigated by qRT-PCR. Taken together, our study reports the isolation and structural characterization of bioactive fraction BG138 from B. gymnorhiza and its anti-biofilm, anti-virulence, anti-quorum sensing, and cell-damaging activities against P. aeruginosa.
Collapse
Affiliation(s)
- Nilesh Lakshman Dahibhate
- Department of Biological Sciences, Birla Institute of Technology & Science Pilani, K. K. Birla Goa Campus, Goa, India
| | - Sanjeev K Shukla
- Sophisticated Analytical Instrument Facility, CSIR-Central Drug Research Institute, Lucknow, India
| | - Kundan Kumar
- Department of Biological Sciences, Birla Institute of Technology & Science Pilani, K. K. Birla Goa Campus, Goa, India
| |
Collapse
|
14
|
Rojas LJ, Yasmin M, Benjamino J, Marshall SM, DeRonde KJ, Krishnan NP, Perez F, Colin AA, Cardenas M, Martinez O, Pérez-Cardona A, Rhoads DD, Jacobs MR, LiPuma JJ, Konstan MW, Vila AJ, Smania A, Mack AR, Scott JG, Adams MD, Abbo LM, Bonomo RA. Genomic heterogeneity underlies multidrug resistance in Pseudomonas aeruginosa: A population-level analysis beyond susceptibility testing. PLoS One 2022; 17:e0265129. [PMID: 35358221 PMCID: PMC8970513 DOI: 10.1371/journal.pone.0265129] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/23/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Pseudomonas aeruginosa is a persistent and difficult-to-treat pathogen in many patients, especially those with Cystic Fibrosis (CF). Herein, we describe a longitudinal analysis of a series of multidrug resistant (MDR) P. aeruginosa isolates recovered in a 17-month period, from a young female CF patient who underwent double lung transplantation. Our goal was to understand the genetic basis of the observed resistance phenotypes, establish the genomic population diversity, and define the nature of sequence evolution over time. METHODS Twenty-two sequential P. aeruginosa isolates were obtained within a 17-month period, before and after a double-lung transplant. At the end of the study period, antimicrobial susceptibility testing, whole genome sequencing (WGS), phylogenetic analyses and RNAseq were performed in order to understand the genetic basis of the observed resistance phenotypes, establish the genomic population diversity, and define the nature of sequence changes over time. RESULTS The majority of isolates were resistant to almost all tested antibiotics. A phylogenetic reconstruction revealed 3 major clades representing a genotypically and phenotypically heterogeneous population. The pattern of mutation accumulation and variation of gene expression suggested that a group of closely related strains was present in the patient prior to transplantation and continued to change throughout the course of treatment. A trend toward accumulation of mutations over time was observed. Different mutations in the DNA mismatch repair gene mutL consistent with a hypermutator phenotype were observed in two clades. RNAseq performed on 12 representative isolates revealed substantial differences in the expression of genes associated with antibiotic resistance and virulence traits. CONCLUSIONS The overwhelming current practice in the clinical laboratories setting relies on obtaining a pure culture and reporting the antibiogram from a few isolated colonies to inform therapy decisions. Our analyses revealed significant underlying genomic heterogeneity and unpredictable evolutionary patterns that were independent of prior antibiotic treatment, highlighting the need for comprehensive sampling and population-level analysis when gathering microbiological data in the context of CF P. aeruginosa chronic infection. Our findings challenge the applicability of antimicrobial stewardship programs based on single-isolate resistance profiles for the selection of antibiotic regimens in chronic infections such as CF.
Collapse
Affiliation(s)
- Laura J. Rojas
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio, United States of America
| | - Mohamad Yasmin
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Jacquelynn Benjamino
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, United States of America
| | - Steven M. Marshall
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Kailynn J. DeRonde
- Jackson Memorial Hospital, Jackson Health System, Miami, Florida, United States of America
| | - Nikhil P. Krishnan
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Departments of Translational Hematology and Oncology Research and Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Federico Perez
- Medical Service, Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, United States of America
- CONICET, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), Córdoba, Argentina
- Division of Infectious Diseases and HIV Medicine, Cleveland, Ohio, United States of America
- GRECC Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Andrew A. Colin
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Monica Cardenas
- Department of Pediatrics, University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Octavio Martinez
- Jackson Memorial Hospital, Jackson Health System, Miami, Florida, United States of America
- Division of Pulmonology, Department of Pathology University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Armando Pérez-Cardona
- Jackson Memorial Hospital, Jackson Health System, Miami, Florida, United States of America
| | - Daniel D. Rhoads
- Department of Laboratory Medicine and Infection Biology Program, Cleveland Clinic, Cleveland, Ohio, United States of America
- Department of Pathology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University Cleveland, Ohio, United States of America
| | - Michael R. Jacobs
- Department of Pathology, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University Cleveland, Ohio, United States of America
| | - John J. LiPuma
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Michael W. Konstan
- Department of Pediatrics, Case Western Reserve University School of Medicine and Rainbow Babies and Children’s Hospital, Cleveland, Ohio, United States of America
| | - Alejandro J. Vila
- Instituto de Biología Molecular y Celular de Rosario (IBR, CONICET-UNR), Rosario, Argentina
| | - Andrea Smania
- CONICET, Centro de Investigaciones en Química Biológica de Córdoba (CIQUIBIC), Córdoba, Argentina
- Universidad Nacional de Córdoba, Facultad de Ciencias Químicas, Departamento de Química Biológica, Córdoba, Argentina
| | - Andrew R. Mack
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
| | - Jacob G. Scott
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Departments of Translational Hematology and Oncology Research and Radiation Oncology, Cleveland Clinic, Cleveland, Ohio, United States of America
| | - Mark D. Adams
- The Jackson Laboratory for Genomic Medicine, Farmington, Connecticut, United States of America
| | - Lilian M. Abbo
- Jackson Memorial Hospital, Jackson Health System, Miami, Florida, United States of America
- Division of Infectious Diseases Department of Medicine University of Miami Miller School of Medicine, Miami, Florida, United States of America
| | - Robert A. Bonomo
- Department of Molecular Biology and Microbiology, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Research Service, Louis Stokes Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
- CWRU-Cleveland VAMC Center for Antimicrobial Resistance and Epidemiology (Case VA CARES), Cleveland, Ohio, United States of America
- Center for Proteomics and Bioinformatics, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
- Medical Service, Louis Stokes Cleveland VA Medical Center, Cleveland, Ohio, United States of America
- Division of Infectious Diseases and HIV Medicine, Cleveland, Ohio, United States of America
- GRECC Louis Stokes Cleveland Veterans Affairs Medical Center, Cleveland, Ohio, United States of America
- Department of Pharmacology, Cleveland, Ohio, United States of America
- Department of Biochemistry Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| |
Collapse
|
15
|
Lynch JP, Zhanel GG. Pseudomonas aeruginosa Pneumonia: Evolution of Antimicrobial Resistance and Implications for Therapy. Semin Respir Crit Care Med 2022; 43:191-218. [PMID: 35062038 DOI: 10.1055/s-0041-1740109] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Pseudomonas aeruginosa (PA), a non-lactose-fermenting gram-negative bacillus, is a common cause of nosocomial infections in critically ill or debilitated patients, particularly ventilator-associated pneumonia (VAP), and infections of urinary tract, intra-abdominal, wounds, skin/soft tissue, and bloodstream. PA rarely affects healthy individuals, but may cause serious infections in patients with chronic structural lung disease, comorbidities, advanced age, impaired immune defenses, or with medical devices (e.g., urinary or intravascular catheters, foreign bodies). Treatment of pseudomonal infections is difficult, as PA is intrinsically resistant to multiple antimicrobials, and may acquire new resistance determinants even while on antimicrobial therapy. Mortality associated with pseudomonal VAP or bacteremias is high (> 35%) and optimal therapy is controversial. Over the past three decades, antimicrobial resistance (AMR) among PA has escalated globally, via dissemination of several international multidrug resistant "epidemic" clones. We discuss the importance of PA as a cause of pneumonia including health care-associated pneumonia, hospital-acquired pneumonia, VAP, the emergence of AMR to this pathogen, and approaches to therapy (both empirical and definitive).
Collapse
Affiliation(s)
- Joseph P Lynch
- Division of Pulmonary, Critical Care Medicine, Allergy, and Clinical Immunology, Department of Medicine, The David Geffen School of Medicine at UCLA, Los Angeles, California
| | - George G Zhanel
- Department of Medical Microbiology/Infectious Diseases, University of Manitoba, Max Rady College of Medicine, Winnipeg, Manitoba, Canada
| |
Collapse
|
16
|
Freire CMADS, Taunay-Rodrigues A, Gonzatti MB, Fonseca FMP, Freire JEDC. New insights about the EptA protein and its correlation with the pmrC gene in polymyxin resistance in Pseudomonas aeruginosa. CURRENT RESEARCH IN MICROBIAL SCIENCES 2021; 2:100042. [PMID: 34841333 PMCID: PMC8610356 DOI: 10.1016/j.crmicr.2021.100042] [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: 01/26/2021] [Revised: 04/30/2021] [Accepted: 06/06/2021] [Indexed: 11/19/2022] Open
Abstract
Computational biology. Bacterial resistance. Pseudomonas aeruginosa. Gram-negative bacteria. Polymyxin.
Nowadays, clinical and scientific interest in antibiotics, as polymyxin, has increased due to the large number of reports of multiresistant Gram-negative bacteria, as Pseudomonas aeruginosa. The aim of this study was to investigate a related group of proteins for resistance to polymyxins, encoded by P. aeruginosa genome, through in silico analysis. The mobilized colistin resistance 1 (MCR1) protein from Escherichia coli was used for comparison. Similar sequences to the protein MCR1 in P. aeruginosa were analysed for physicochemical properties. 31 protein isoforms in P. aeruginosa (EptA) were found able to confer resistance to polymyxin showing protein lengths between 551 and 572 amino acids, with molecular mass values between 61.36 - 62. 80 kDa, isoelectric point between 6.10 to 7.17, instability index between 33.76 to 41.87, aliphatic index between 98.67 to 102.63 and the hydropathyindex between - 0.008 to 0.094. These proteins belong to the DUF1705 superfamily with bit-score values between 559.81 and 629.78. A high degree of similarity between EpTAs in P. aeruginosa was observed in relation to other proteins that confer resistance to polymyxins, present in Gram-negative bacteria species of clinical interest. Although, further studies are needed to identify the actual contribution of EptAs in P. aeruginosa species.
Collapse
|
17
|
Interplay of OpdP Porin and Chromosomal Carbapenemases in the Determination of Carbapenem Resistance/Susceptibility in Pseudomonas aeruginosa. Microbiol Spectr 2021; 9:e0118621. [PMID: 34585948 PMCID: PMC8557820 DOI: 10.1128/spectrum.01186-21] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Carbapenem resistance in Pseudomonas aeruginosa strains responsible for chronic lung infections in cystic fibrosis (CF) patients is mainly due to loss of the OprD protein and, limited to meropenem and doripenem, to overexpression of efflux pumps. However, recent reports of isolates showing inconsistent genotype-phenotype combinations (e.g., susceptibility in the presence of resistance determinants and vice versa) suggest the involvement of additional factors whose role is not yet fully elucidated. Among them, the OpdP porin as an alternative route of entry for carbapenems other than OprD and the overexpression of two chromosomal carbapenemases, the Pseudomonas-derived cephalosporinase (PDC) and the PoxB oxacillinase, have recently been reconsidered and studied in specific model strains. Here, the contribution of these factors was investigated by comparing different phenotypic variants of three strains collected from the sputum of colonized CF patients. Carbapenem uptake through OpdP was investigated both at the functional level, by assessing the competition exerted by glycine-glutamate, the OpdP’s natural substrate, against imipenem uptake, and at the molecular level, by comparing the expression levels of opdP genes by quantitative real-time PCR (qRT-PCR). Moreover, overexpression of the chromosomal carbapenemases in some of the isolates was also investigated by qRT-PCR. The results showed that, even if OprD inactivation remains the most important determinant of carbapenem resistance in strains infecting the CF lung, the interplay of other determinants might have a nonnegligible impact on bacterial susceptibility, being able to modify the phenotype of part of the population and consequently complicating the choice of an appropriate therapy. IMPORTANCE This study examines the interplay of multiple factors in determining a pattern of resistance or susceptibility to carbapenems in clinical isolates of Pseudomonas aeruginosa, focusing on the role of previously poorly understood determinants. In particular, the impact of carbapenem permeability through OprD and OpdP porins was analyzed, as well as the activity of the chromosomal carbapenemases AmpC and PoxB, going beyond the simple identification of resistance determinants encoded by each isolate. Indeed, analysis of the expression levels of these determinants provides a new approach to determine the contribution of each factor, both individually and in coexistence with the other factors. The study contributes to understanding some phenotype-genotype discordances closely related to the heteroresistance frequently detected in P. aeruginosa isolates responsible for pulmonary infections in cystic fibrosis patients, which complicates the choice of an appropriate patient-specific therapy.
Collapse
|
18
|
Cogen JD, Onchiri FM, Hamblett NM, Gibson RL, Morgan WJ, Rosenfeld M. Association of Intensity of Antipseudomonal Antibiotic Therapy With Risk of Treatment-Emergent Organisms in Children With Cystic Fibrosis and Newly Acquired Pseudomonas Aeruginosa. Clin Infect Dis 2021; 73:987-993. [PMID: 33693586 DOI: 10.1093/cid/ciab208] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND While Pseudomonas aeruginosa (Pa) eradication regimens have contributed to a decline in Pa prevalence in people with cystic fibrosis (CF), this antibiotic exposure might increase the risk of acquisition of drug-resistant organisms. This study evaluated the association between antipseudomonal antibiotic exposure intensity and acquisition risk of drug-resistant organisms among children with CF and new Pa infection. METHODS We utilized data from the Early Pseudomonas Infection Control Clinical Trial (EPIC CT), a randomized controlled trial comparing Pa eradication strategies in children with CF and new Pa. The exposure was the number of weeks of oral or inhaled antipseudomonal antibiotics or ever versus never treatment with intravenous antipseudomonal antibiotics during the 18 months of EPIC CT participation. Primary outcomes were risks of acquisition of several respiratory organisms during 5 years of follow-up after EPIC CT estimated using Cox proportional hazards models separately for each specific organism. RESULTS Among 249 participants, there was no increased acquisition risk of any organism associated with greater inhaled antibiotic exposure. With each additional week of oral antibiotics, there was an increased hazard of Achromobacter xylosoxidans acquisition (HR, 1.24; 95% CI: 1.02-1.50; P = .03). Treatment with intravenous antibiotics was associated with an increased hazard of acquisition of multidrug-resistant Pa (HR, 2.47; 95% CI: 1.28-4.78; P = .01) and MRSA (HR, 1.57; 95% CI: 1.03-2.40; P = .04). CONCLUSIONS Results from this study illustrate the importance of making careful antibiotic choices to balance the benefits of antibiotics in people with CF while minimizing risk of acquisition of drug-resistant organisms.
Collapse
Affiliation(s)
- Jonathan D Cogen
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
- Seattle Children's Research Institute, Seattle, Washington, USA
| | | | - Nicole Mayer Hamblett
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
- Seattle Children's Research Institute, Seattle, Washington, USA
- Department of Biostatistics, University of Washington School of Medicine, Seattle, Washington, USA
| | - Ronald L Gibson
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
- Seattle Children's Research Institute, Seattle, Washington, USA
| | - Wayne J Morgan
- Department of Pediatrics, The University of Arizona, Tucson, Arizona, USA
| | - Margaret Rosenfeld
- Department of Pediatrics, University of Washington School of Medicine, Seattle, Washington, USA
- Seattle Children's Research Institute, Seattle, Washington, USA
| |
Collapse
|
19
|
Deatherage DE, Barrick JE. High-throughput characterization of mutations in genes that drive clonal evolution using multiplex adaptome capture sequencing. Cell Syst 2021; 12:1187-1200.e4. [PMID: 34536379 DOI: 10.1016/j.cels.2021.08.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 07/14/2021] [Accepted: 08/20/2021] [Indexed: 11/17/2022]
Abstract
Understanding how cells are likely to evolve can guide medical interventions and bioengineering efforts that must contend with unwanted mutations. The adaptome of a cell-the neighborhood of genetic changes that are most likely to drive adaptation in a given environment-can be mapped by tracking rare beneficial variants during the early stages of clonal evolution. We used multiplex adaptome capture sequencing (mAdCap-seq), a procedure that combines unique molecular identifiers and hybridization-based enrichment, to characterize mutations in eight Escherichia coli genes known to be under selection in a laboratory environment. We tracked 301 mutations at frequencies as low as 0.01% and inferred the fitness effects of 240 of these mutations. There were distinct molecular signatures of selection on protein structure and function for the three genes with the most beneficial mutations. Our results demonstrate how mAdCap-seq can be used to deeply profile a targeted portion of a cell's adaptome.
Collapse
Affiliation(s)
- Daniel E Deatherage
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX 78712, USA
| | - Jeffrey E Barrick
- Department of Molecular Biosciences, Center for Systems and Synthetic Biology, The University of Texas at Austin, Austin, TX 78712, USA.
| |
Collapse
|
20
|
Antimicrobial Synergy Testing: Comparing the Tobramycin and Ceftazidime Gradient Diffusion Methodology Used in Assessing Synergy in Cystic Fibrosis-Derived Multidrug-Resistant Pseudomonas aeruginosa. Antibiotics (Basel) 2021; 10:antibiotics10080967. [PMID: 34439017 PMCID: PMC8388873 DOI: 10.3390/antibiotics10080967] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/26/2021] [Accepted: 08/04/2021] [Indexed: 11/17/2022] Open
Abstract
The need for synergy testing is driven by the necessity to extend the antimicrobial spectrum, reducing drug dosage/toxicity and the development of resistance. Despite the abundance of synergy testing methods, there is the absence of a gold standard and a lack of synergy correlation among methods. The most popular method (checkerboard) is labor-intensive and is not practical for clinical use. Most clinical laboratories use several gradient synergy methods which are quicker/easier to use. This study sought to evaluate three gradient synergy methods (direct overlay, cross, MIC:MIC ratio) with the checkerboard, and compare two interpretative criteria (the fractional inhibitory concentration index (FICI) and susceptibility breakpoint index (SBPI)) regarding these methods. We tested 70 multidrug-resistant Pseudomonas aeruginosa, using a tobramycin and ceftazidime combination. The agreement between the checkerboard and gradient methods was 60 to 77% for FICI, while agreements for SBPI that ranged between 67 and 82.86% were statistically significant (p ≤ 0.001). High kappa agreements were observed using SBPI (Ƙ > 0.356) compared to FICI (Ƙ < 0.291) criteria, and the MIC:MIC method demonstrated the highest, albeit moderate, intraclass correlation coefficient (ICC = 0.542) estimate. Isolate resistance profiles suggest method-dependent synergism for isolates, with ceftazidime susceptibility after increased exposure. The results show that when interpretative criteria are considered, gradient diffusion (especially MIC:MIC) is a valuable and practical method that can inform the treatment of cystic fibrosis patients who are chronically infected with P. aeruginosa.
Collapse
|
21
|
Fidan E, Alci G, Koldaş SS, Karadag B, Gökdemir Y, Erdem Eralp E, Karahasan Yagcı A. Cumulative Antimicrobial Susceptibility Data of Pseudomonas Aeruginosa Isolates from Cystic Fibrosis Patients: 4-Year Experience. J PEDIAT INF DIS-GER 2021. [DOI: 10.1055/s-0041-1731344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Abstract
Objective Pseudomonas aeruginosa is the most important cause of lung infection among cystic fibrosis (CF) patients, and to reduce the severity of the infection, facility-specific cumulative antibiograms could help clinicians in empirical treatment.
Methods Respiratory samples of CF patients between January 2015 and December 2018 were scanned through Laboratory Operating System retrospectively. Demographical data of patients, culture results, and antibiotic susceptibilities are recorded using Microsoft Excel 2010. Cumulative antibiogram data were obtained according to the CLSI M39A4 document.
Results The number of registered patients increased in 4 years from 154 to 253. The mean age of patients varied from 9 to 11.7 (range, 2–42). The ratio of patients with a positive culture for P. aeruginosa increased from 32 to 40%, and the mean patients' age decreased from 16.6 to 11.1 (p <0.05). A total number of 4,146 respiratory samples were analyzed. Sputum samples consisted of 42.5% (n: 1,767) of the samples with a 58.4% isolation rate of P. aeruginosa (n: 1,034). A notable increase of resistance was seen almost all antimicrobials tested by years. The ratio of multidrug-resistant (MDR) P. aeruginosa was 4.1, 10.2, 4.5, and 8.6% in 2015, 2016, 2017, and 2018.
Conclusion Antimicrobial resistance is a challenging problem in CF patients, and surveillance should be done regularly.
Collapse
Affiliation(s)
- Ebru Fidan
- Medical Microbiology Department, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Gamze Alci
- Medical Microbiology Department, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Seda Sevilay Koldaş
- Medical Microbiology Department, Faculty of Medicine, Marmara University, Istanbul, Turkey
| | - Bülent Karadag
- Division of Pediatric Pulmonology, Marmara University Hospital, Istanbul, Turkey
| | - Yasemin Gökdemir
- Division of Pediatric Pulmonology, Marmara University Hospital, Istanbul, Turkey
| | - Ela Erdem Eralp
- Division of Pediatric Pulmonology, Marmara University Hospital, Istanbul, Turkey
| | | |
Collapse
|
22
|
Tait JR, Bilal H, Kim TH, Oh A, Peleg AY, Boyce JD, Oliver A, Bergen PJ, Nation RL, Landersdorfer CB. Pharmacodynamics of ceftazidime plus tobramycin combination dosage regimens against hypermutable Pseudomonas aeruginosa isolates at simulated epithelial lining fluid concentrations in a dynamic in vitro infection model. J Glob Antimicrob Resist 2021; 26:55-63. [PMID: 34023531 DOI: 10.1016/j.jgar.2021.04.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 03/12/2021] [Accepted: 04/16/2021] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES Hypermutable Pseudomonas aeruginosa strains are a major challenge in cystic fibrosis. We investigated bacterial killing and resistance emergence for approved ceftazidime and tobramycin regimens, alone and in combination. METHODS Pseudomonas aeruginosa PAOΔmutS and six hypermutable clinical isolates were examined using 48-h static concentration time-kill (SCTK) studies (inoculum ~107.5 CFU/mL); four strains were also studied in a dynamic in vitro model (IVM) (inoculum ~108 CFU/mL). The IVM simulated concentration-time profiles in epithelial lining fluid following intravenous administration of ceftazidime (3 g/day and 9 g/day continuous infusion), tobramycin (5 mg/kg and 10 mg/kg via 30-min infusion 24-hourly; half-life 3.5 h), and their combinations. Time courses of total and less-susceptible populations were determined. RESULTS Ceftazidime plus tobramycin demonstrated synergistic killing in SCTK studies for all strains, although to a lesser extent for ceftazidime-resistant strains. In the IVM, ceftazidime and tobramycin monotherapies provided ≤5.4 and ≤3.4 log10 initial killing, respectively; however, re-growth with resistance occurred by 72 h. Against strains susceptible to one or both antibiotics, high-dose combination regimens provided >6 log10 initial killing, which was generally synergistic from 8-24 h, and marked suppression of re-growth and resistance at 72 h. The time course of bacterial density in the IVM was well described by mechanism-based models, enabling Monte Carlo simulations (MCSs) to predict likely effectiveness of the combination in patients. CONCLUSION Results of the IVM and MCS suggested antibacterial effect depends both on the strain's susceptibility and hypermutability. Further investigation of the combination against hypermutable P. aeruginosa strains is warranted.
Collapse
Affiliation(s)
- Jessica R Tait
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia; Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Hajira Bilal
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Tae Hwan Kim
- College of Pharmacy, Daegu Catholic University, Daegu, South Korea
| | - Abigail Oh
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Anton Y Peleg
- Department of Infectious Diseases, The Alfred Hospital and Central Clinical School, Monash University, Melbourne, VIC, Australia; Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - John D Boyce
- Biomedicine Discovery Institute, Department of Microbiology, Monash University, Melbourne, VIC, Australia
| | - Antonio Oliver
- Servicio de Microbiología, Hospital Universitario Son Espases, Instituto de Investigación Sanitaria de Palma, Palma de Mallorca, Spain
| | - Phillip J Bergen
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Roger L Nation
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia
| | - Cornelia B Landersdorfer
- Centre for Medicine Use and Safety, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, VIC, Australia.
| |
Collapse
|
23
|
Hyperinflammation and airway surface liquid dehydration in cystic fibrosis: purinergic system as therapeutic target. Inflamm Res 2021; 70:633-649. [PMID: 33904934 DOI: 10.1007/s00011-021-01464-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 04/06/2021] [Accepted: 04/19/2021] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVE AND DESIGN The exacerbate inflammatory response contributes to the progressive loss of lung function in cystic fibrosis (CF), a genetic disease that affects the osmotic balance of mucus and mucociliary clearance, resulting in a microenvironment that favors infection and inflammation. The purinergic system, an extracellular signaling pathway characterized by nucleotides, enzymes and receptors, may have a protective role in the disease, through its action in airway surface liquid (ASL) and anti-inflammatory response. MATERIALS AND METHODS To make up this review, studies covering topics of CF, inflammation, ASL and purinergic system were selected from the main medical databases, such as Pubmed and ScienceDirect. CONCLUSION We propose several ways to modulate the purinergic system as a potential therapy for CF, like inhibition of P2X7, activation of P2Y2, A2A and A2B receptors and blocking of adenosine deaminase. Among them, we postulate that the most suitable strategy is to block the action of adenosine deaminase, which culminates in the increase of Ado levels that presents anti-inflammatory actions and improves mucociliary clearance. Furthermore, it is possible to maintain the physiological levels of ATP to control the hydration of ASL. These therapies could correct the main mechanisms that contribute to the progression of CF.
Collapse
|
24
|
Tekintas Y, Demir-Dora D, Erac B, Erac Y, Yilmaz O, Aydemir SS, Kocagoz ZT, Hosgor-Limoncu M. Silencing acpP gene via antisense oligonucleotide-niosome complex in clinical Pseudomonas aeruginosa isolates. Res Microbiol 2021; 172:103834. [PMID: 33894336 DOI: 10.1016/j.resmic.2021.103834] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
Pseudomonas aeruginosa, an opportunistic Gram-negative pathogen, is one of the major causes of nosocomial infections. In addition to its physiological adaptation capacity, it can develop resistance to disinfectants and antibiotics through various mechanisms. Recently, new eradication methods are gaining attention. Therefore, in this study, an LNA-2'-O-methyl hybrid antisense oligonucleotide targeting the acyl carrier protein P (acpP) gene was introduced into P. aeruginosa isolates. The design was determined through sequence analysis and prediction of the secondary structure of mRNA by software. Niosomes were used for enhancing cellular uptake. The control of the binding and transfection ability of the sequence was determined fluorometrically by labeling with 6-Fam. The effects were determined with broth microdilution method and qPCR studies. Eight different formulations were prepared. Among these, one formulation has shown to have ASO complexation ability whose composition was 312 μl Span 80 + 69.5 mg Cholesterol+ 36.4 mg CTAB+1 ml Chloroform and 5 ml dH2O. Thus this formulation was determined as the delivery system for the next stages. Significant gene inhibition was detected at the six isolates. Results of this study suggested that niosomes can be used as a delivery system for cellular uptake of ASO and could eliminate bacterial growth.
Collapse
Affiliation(s)
- Yamac Tekintas
- Izmir Kâtip Celebi University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Izmir, Turkey
| | - Devrim Demir-Dora
- Akdeniz University, Faculty of Medicine, Department of Pharmacology, Antalya, Turkey; Akdeniz University, Health Sciences Institute, Department of Medical Biotechnology, Antalya, Turkey; Akdeniz University, Health Sciences Institute, Department of Gene and Cell Therapy, Antalya, Turkey
| | - Bayrı Erac
- Ege University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Izmir, Turkey
| | - Yasemin Erac
- Ege University, Faculty of Pharmacy, Department of Pharmacology, Izmir, Turkey
| | - Ozlem Yilmaz
- Akdeniz University, Faculty of Medicine, Department of Gene and Cell Therapy Research and Application Centre, Antalya, Turkey; Akdeniz University, Faculty of Medicine, Department of Medical Biology and Genetics, Antalya, Turkey
| | - Sabire Sohret Aydemir
- Ege University, Faculty of Medicine, Department of Medical Microbiology, Izmir, Turkey
| | - Zuhtu Tanil Kocagoz
- Acıbadem Mehmet Ali Aydinlar University, School of Medicine, Department of Medical Microbiology, Istanbul, Turkey
| | - Mine Hosgor-Limoncu
- Ege University, Faculty of Pharmacy, Department of Pharmaceutical Microbiology, Izmir, Turkey.
| |
Collapse
|
25
|
Datar R, Coello Pelegrin A, Orenga S, Chalansonnet V, Mirande C, Dombrecht J, Perry JD, Perry A, Goossens H, van Belkum A. Phenotypic and Genomic Variability of Serial Peri-Lung Transplantation Pseudomonas aeruginosa Isolates From Cystic Fibrosis Patients. Front Microbiol 2021; 12:604555. [PMID: 33897629 PMCID: PMC8058383 DOI: 10.3389/fmicb.2021.604555] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Accepted: 03/15/2021] [Indexed: 12/21/2022] Open
Abstract
Cystic fibrosis (CF) represents one of the major genetic and chronic lung diseases affecting Caucasians of European descent. Patients with CF suffer from recurring infections that lead to further damage of the lungs. Pulmonary infection due to Pseudomonas aeruginosa is most prevalent, further increasing CF-related mortality. The present study describes the phenotypic and genotypic variations among 36 P. aeruginosa isolates obtained serially from a non-CF and five CF patients before, during and after lung transplantation (LTx). The classical and genomic investigation of these isolates revealed a common mucoid phenotype and only subtle differences in the genomes. Isolates originating from an individual patient shared ≥98.7% average nucleotide identity (ANI). However, when considering isolates from different patients, substantial variations in terms of sequence type (ST), virulence factors and antimicrobial resistance (AMR) genes were observed. Whole genome multi-locus sequence typing (MLST) confirmed the presence of unique STs per patient regardless of the time from LTx. It was supported by the monophyletic clustering found in the genome-wide phylogeny. The antibiogram shows that ≥91.6% of the isolates were susceptible to amikacin, colistin and tobramycin. For other antibiotics from the panel, isolates frequently showed resistance. Alternatively, a comparative analysis of the 36 P. aeruginosa isolates with 672 strains isolated from diverse ecologies demonstrated clustering of the CF isolates according to the LTx patients from whom they were isolated. We observed that despite LTx and associated measures, all patients remained persistently colonized with similar isolates. The present study shows how whole genome sequencing (WGS) along with phenotypic analysis can help us understand the evolution of P. aeruginosa over time especially its antibiotic resistance.
Collapse
Affiliation(s)
| | - Andreu Coello Pelegrin
- BioMérieux, La Balme les Grottes, France
- Laboratory of Medical Microbiology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | | | | | | | | | - John D. Perry
- Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Audrey Perry
- Freeman Hospital, Newcastle upon Tyne, United Kingdom
| | - Herman Goossens
- Laboratory of Medical Microbiology, Faculty of Medicine and Health Sciences, Vaccine and Infectious Disease Institute, University of Antwerp, Antwerp, Belgium
| | | |
Collapse
|
26
|
Recchiuti A, Patruno S, Mattoscio D, Isopi E, Pomilio A, Lamolinara A, Iezzi M, Pecce R, Romano M. Resolvin D1 and D2 reduce SARS-CoV-2-induced inflammatory responses in cystic fibrosis macrophages. FASEB J 2021; 35:e21441. [PMID: 33749902 PMCID: PMC8250053 DOI: 10.1096/fj.202001952r] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/29/2021] [Accepted: 01/29/2021] [Indexed: 02/06/2023]
Abstract
An excessive, non‐resolving inflammatory response underlies severe COVID‐19 that may have fatal outcomes. Therefore, the investigation of endogenous pathways leading to resolution of inflammation is of interest to uncover strategies for mitigating inflammation in people with SARS‐CoV‐2 infection. This becomes particularly urgent in individuals with preexisting pathologies characterized by chronic respiratory inflammation and prone to bacterial infection, such as cystic fibrosis (CF). Here, we analyzed the immune responses to SARS‐CoV‐2 virion spike 1 glycoprotein (S1) of macrophages (MΦ) from volunteers with and without CF and tested the efficacy of resolvins (Rv) D1 and D2 in regulating the inflammatory and antimicrobial functions of MΦ exposed to S1. S1 significantly increased chemokine release, including interleukin (IL)‐8, in CF and non‐CF MΦ, while it enhanced IL‐6 and tumor necrosis factor (TNF)‐α in non‐CF MΦ, but not in CF cells. S1 also triggered the biosynthesis of RvD1 and modulated microRNAs miR‐16, miR‐29a, and miR‐103, known to control the inflammatory responses. RvD1 and RvD2 treatment abated S1‐induced inflammatory responses in CF and non‐CF MΦ, significantly reducing the release of select chemokines and cytokines including IL‐8 and TNF‐α. RvD1 and RvD2 both restored the expression of miR‐16 and miR‐29a, while selectively increasing miR‐223 and miR‐125a, which are involved in NF‐κB activation and MΦ inflammatory polarization. During Pseudomonas aeruginosa infection, S1 stimulated the MΦ phagocytic activity that was further enhanced by RvD1 and RvD2. These results provide a map of molecular responses to SARS‐CoV‐2 in MΦ, key determinants of COVID‐19‐related inflammation, unveiling some peculiarity in the response of cells from individuals with CF. They also demonstrate beneficial, regulatory actions of RvD1 and RvD2 on SARS‐CoV‐2‐induced inflammation.
Collapse
Affiliation(s)
- Antonio Recchiuti
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Sara Patruno
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Domenico Mattoscio
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Elisa Isopi
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Antonella Pomilio
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Alessia Lamolinara
- Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Department of Neurosciences, Imaging and Clinical Sciences, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Manuela Iezzi
- Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Department of Neurosciences, Imaging and Clinical Sciences, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Romina Pecce
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| | - Mario Romano
- Department of Medical, Oral, and Biotechnology Science, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy.,Center for Advanced Studies and Technology, Università "G. d'Annunzio" Chieti - Pescara, Chieti, Italy
| |
Collapse
|
27
|
Martin I, Waters V, Grasemann H. Approaches to Targeting Bacterial Biofilms in Cystic Fibrosis Airways. Int J Mol Sci 2021; 22:ijms22042155. [PMID: 33671516 PMCID: PMC7926955 DOI: 10.3390/ijms22042155] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/14/2021] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
The treatment of lung infection in the context of cystic fibrosis (CF) is limited by a biofilm mode of growth of pathogenic organisms. When compared to planktonically grown bacteria, bacterial biofilms can survive extremely high levels of antimicrobials. Within the lung, bacterial biofilms are aggregates of microorganisms suspended in a matrix of self-secreted proteins within the sputum. These structures offer both physical protection from antibiotics as well as a heterogeneous population of metabolically and phenotypically distinct bacteria. The bacteria themselves and the components of the extracellular matrix, in addition to the signaling pathways that direct their behaviour, are all potential targets for therapeutic intervention discussed in this review. This review touches on the successes and failures of current anti-biofilm strategies, before looking at emerging therapies and the mechanisms by which it is hoped they will overcome current limitations.
Collapse
Affiliation(s)
- Isaac Martin
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada;
- Correspondence:
| | - Valerie Waters
- Division of Infectious Diseases, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada;
- Department of Paediatrics and Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 0A4, Canada
| | - Hartmut Grasemann
- Division of Respiratory Medicine, Department of Paediatrics, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 1X8, Canada;
- Department of Paediatrics and Translational Medicine, Research Institute, The Hospital for Sick Children, University of Toronto, Toronto, ON M5G 0A4, Canada
| |
Collapse
|
28
|
Evolution of Antibiotic Tolerance Shapes Resistance Development in Chronic Pseudomonas aeruginosa Infections. mBio 2021; 12:mBio.03482-20. [PMID: 33563834 PMCID: PMC7885114 DOI: 10.1128/mbio.03482-20] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Over the past decades, pan-resistant strains of major bacterial pathogens have emerged and have rendered clinically available antibiotics ineffective, putting at risk many of the major achievements of modern medicine, including surgery, cancer therapy, and organ transplantation. A thorough understanding of processes leading to the development of antibiotic resistance in human patients is thus urgently needed. The widespread use of antibiotics promotes the evolution and dissemination of resistance and tolerance mechanisms. To assess the relevance of tolerance and its implications for resistance development, we used in vitro evolution and analyzed the inpatient microevolution of Pseudomonas aeruginosa, an important human pathogen causing acute and chronic infections. We show that the development of tolerance precedes and promotes the acquisition of resistance in vitro, and we present evidence that similar processes shape antibiotic exposure in human patients. Our data suggest that during chronic infections, P. aeruginosa first acquires moderate drug tolerance before following distinct evolutionary trajectories that lead to high-level multidrug tolerance or to antibiotic resistance. Our studies propose that the development of antibiotic tolerance predisposes bacteria for the acquisition of resistance at early stages of infection and that both mechanisms independently promote bacterial survival during antibiotic treatment at later stages of chronic infections.
Collapse
|
29
|
Silveira GGOS, Torres MDT, Ribeiro CFA, Meneguetti BT, Carvalho CME, de la Fuente-Nunez C, Franco OL, Cardoso MH. Antibiofilm Peptides: Relevant Preclinical Animal Infection Models and Translational Potential. ACS Pharmacol Transl Sci 2021; 4:55-73. [PMID: 33615161 DOI: 10.1021/acsptsci.0c00191] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Indexed: 12/21/2022]
Abstract
Biofilm-forming bacteria may be 10-1000 times more resistant to antibiotics than planktonic bacteria and represent about 75% of bacterial infections in humans. Antibiofilm treatments are scarce, and no effective therapies have been reported so far. In this context, antibiofilm peptides (ABPs) represent an exciting class of agents with potent activity against biofilms both in vitro and in vivo. Moreover, murine models of bacterial biofilm infections have been used to evaluate the in vivo effectiveness of ABPs. Therefore, here we highlight the translational potential of ABPs and provide an overview of the different clinically relevant murine models to assess ABP efficacy, including wound, foreign body, chronic lung, and oral models of infection. We discuss key challenges to translate ABPs to the clinic and the pros and cons of the existing murine biofilm models for reliable assessment of the efficacy of ABPs.
Collapse
Affiliation(s)
- Gislaine G O S Silveira
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil
| | - Marcelo D T Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Camila F A Ribeiro
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil
| | - Beatriz T Meneguetti
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil
| | - Cristiano M E Carvalho
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States.,Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, Pennsylvania 19104, United States
| | - Octávio L Franco
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal 71966-700, Brazil
| | - Marlon H Cardoso
- S-Inova Biotech, Programa de Pós-Graduação Stricto Sensu em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-010, Brazil.,Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Distrito Federal 71966-700, Brazil
| |
Collapse
|
30
|
Lasko MJ, Huse HK, Nicolau DP, Kuti JL. Contemporary analysis of ETEST for antibiotic susceptibility and minimum inhibitory concentration agreement against Pseudomonas aeruginosa from patients with cystic fibrosis. Ann Clin Microbiol Antimicrob 2021; 20:9. [PMID: 33468149 PMCID: PMC7816365 DOI: 10.1186/s12941-021-00415-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Objectives Cystic fibrosis (CF) acute pulmonary exacerbations are often caused by Pseudomonas aeruginosa, including multi-drug resistant strains. Optimal antibiotic therapy is required to return lung function and should be guided by in vitro susceptibility results. There are sparse data describing ETEST performance for CF isolates using contemporary isolates, methods and interpretation, as well as novel antibiotics, such as ceftazidime–avibactam and ceftolozane–tazobactam. Methods Pseudomonas aeruginosa (n = 105) isolated during pulmonary exacerbation from patients with CF were acquired from 3 US hospitals. Minimum inhibitory concentrations (MICs) were assessed by reference broth microdilution (BMD) and ETEST for aztreonam, cefepime, ceftazidime, ceftazidime–avibactam, ceftolozane–tazobactam, ciprofloxacin, levofloxacin, meropenem, piperacillin–tazobactam, and tobramycin. Broth microdilution was conducted in concordance with the Clinical and Laboratory Standards Institute M100. ETEST methodology reflected package insert recommendations. Performance of ETEST strips was evaluated using the Food and Drug Administration (FDA) and Susceptibility Testing Manufacturers Association (STMA) guidance. Results Of the 105 P. aeruginosa included, 46% had a mucoid phenotype. ETEST MICs typically read 0–1 dilution higher than BMD for all drugs. Categorical agreement and essential agreement ranged from 64 to 93% and 63 to 86%, respectively. The majority of observed errors were minor. A single very major error occurred with ceftazidime (4.2%). For ceftazidime–vibactam, 2 very major errors were observed and both were within essential agreement. Major errors occurred for aztreonam (3.3%), cefepime (9.4%), ceftazidime–avibactam (5.3%, adjusted 2.1%), ceftolozane–tazobactam (1%), meropenem (3.3%), piperacillin–tazobactam (2.9%), and tobramycin (1.5%). Conclusions ETEST methods performed conservatively for most antibiotics against this challenging collection of P. aeruginosa from patients with CF.
Collapse
Affiliation(s)
- Maxwell J Lasko
- Center for Anti-Infective Research and Development, Hartford Hospital, 80 Seymour Street, Hartford, 06102, CT, USA
| | - Holly K Huse
- Department of Clinical Microbiology, Huntington Hospital, Pasadena, CA, USA.,Department of Clinical Microbiology, Harbor-UCLA Medical Center, Torrance, CA, USA
| | - David P Nicolau
- Center for Anti-Infective Research and Development, Hartford Hospital, 80 Seymour Street, Hartford, 06102, CT, USA.,Division of Infectious Diseases, Hartford Hospital, Hartford, CT, USA
| | - Joseph L Kuti
- Center for Anti-Infective Research and Development, Hartford Hospital, 80 Seymour Street, Hartford, 06102, CT, USA.
| |
Collapse
|
31
|
Behzadi P, Baráth Z, Gajdács M. It's Not Easy Being Green: A Narrative Review on the Microbiology, Virulence and Therapeutic Prospects of Multidrug-Resistant Pseudomonas aeruginosa. Antibiotics (Basel) 2021; 10:42. [PMID: 33406652 PMCID: PMC7823828 DOI: 10.3390/antibiotics10010042] [Citation(s) in RCA: 122] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 12/11/2022] Open
Abstract
Pseudomonas aeruginosa is the most frequent cause of infection among non-fermenting Gram-negative bacteria, predominantly affecting immunocompromised patients, but its pathogenic role should not be disregarded in immunocompetent patients. These pathogens present a concerning therapeutic challenge to clinicians, both in community and in hospital settings, due to their increasing prevalence of resistance, and this may lead to prolonged therapy, sequelae, and excess mortality in the affected patient population. The resistance mechanisms of P. aeruginosa may be classified into intrinsic and acquired resistance mechanisms. These mechanisms lead to occurrence of resistant strains against important antibiotics-relevant in the treatment of P. aeruginosa infections-such as β-lactams, quinolones, aminoglycosides, and colistin. The occurrence of a specific resistotype of P. aeruginosa, namely the emergence of carbapenem-resistant but cephalosporin-susceptible (Car-R/Ceph-S) strains, has received substantial attention from clinical microbiologists and infection control specialists; nevertheless, the available literature on this topic is still scarce. The aim of this present review paper is to provide a concise summary on the adaptability, virulence, and antibiotic resistance of P. aeruginosa to a readership of basic scientists and clinicians.
Collapse
Affiliation(s)
- Payam Behzadi
- Department of Microbiology, College of Basic Sciences, Shahr-e-Qods Branch, Islamic Azad University, Tehran 37541-374, Iran;
| | - Zoltán Baráth
- Department of Prosthodontics, Faculty of Dentistry, University of Szeged, Tisza Lajos körút 62-64, 6720 Szeged, Hungary;
| | - Márió Gajdács
- Institute of Medical Microbiology, Faculty of Medicine, Semmelweis University, 1089 Budapest, Hungary
- Department of Pharmacodynamics and Biopharmacy, Faculty of Pharmacy, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
32
|
Sörensen M, Khakimov B, Nurjadi D, Boutin S, Yi B, Dalpke AH, Eigenbrod T. Comparative evaluation of the effect of different growth media on in vitro sensitivity to azithromycin in multi-drug resistant Pseudomonas aeruginosa isolated from cystic fibrosis patients. Antimicrob Resist Infect Control 2020; 9:197. [PMID: 33298147 PMCID: PMC7724801 DOI: 10.1186/s13756-020-00859-7] [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: 07/21/2020] [Accepted: 11/20/2020] [Indexed: 12/26/2022] Open
Abstract
Long-term treatment with azithromycin is a therapeutic option in Cystic Fibrosis (CF) patients chronically infected with P. aeruginosa. It was recently shown that azithromycin has direct antimicrobial activity when P. aeruginosa isolates are tested in Roswell Park Memorial Institute medium supplemented with fetal calf serum (RPMI 1640/FCS) by broth microdilution. We now investigated whether (i) azithromycin might also be active against multidrug resistant (MDR) P. aeruginosa isolated from CF patients and (ii) how in vitro sensitivity assays perform in synthetic cystic fibrosis sputum medium (SCFM), a medium that mimics the particular CF airway environment. In 17 (59%) out of 29 MDR P. aeruginosa CF isolates MICs for azithromycin ranged between 0.25 and 8 μg/ml and 12 isolates (41%) showed a MIC ≥512 μg/ml when measured in RPMI/FCS. In contrast, MICs were ≥ 256 μg/ml for all P. aeruginosa MDR isolates when tested in either SCFM or in conventional cation-adjusted Mueller Hinton Broth. High MIC values observed in CF adapted medium SCFM for both PAO1 and MDR P. aeruginosa CF isolates, as opposed to findings in RPMI, argue against routine azithromycin MIC testing of CF isolates.
Collapse
Affiliation(s)
- Michael Sörensen
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany. .,Laboratory Enders and Partners, 70193, Stuttgart, Germany.
| | - Bakhodur Khakimov
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Dennis Nurjadi
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Sébastien Boutin
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany
| | - Buqing Yi
- Institute of Medical Microbiology and Hygiene, Medical Faculty, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany
| | - Alexander H Dalpke
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany. .,Institute of Medical Microbiology and Hygiene, Medical Faculty, Technische Universität Dresden, Fetscherstraße 74, 01307, Dresden, Germany.
| | - Tatjana Eigenbrod
- Department of Infectious Diseases, Medical Microbiology and Hygiene, Heidelberg University Hospital, 69120, Heidelberg, Germany. .,SLK-Kliniken Heilbronn, 74080, Heilbronn, Germany.
| |
Collapse
|
33
|
Mordue J, O'Boyle N, Gadegaard N, Roe AJ. The force awakens: The dark side of mechanosensing in bacterial pathogens. Cell Signal 2020; 78:109867. [PMID: 33279672 DOI: 10.1016/j.cellsig.2020.109867] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 11/27/2020] [Accepted: 11/29/2020] [Indexed: 02/01/2023]
Abstract
For many bacteria, the ability to sense physical stimuli such as contact with a surface or a potential host cell is vital for survival and proliferation. This ability, and subsequent attachment, confers a wide range of benefits to bacteria and many species have evolved to take advantage of this. Despite the impressive diversity of bacterial pathogens and their virulence factors, mechanosensory mechanisms are often conserved. These include sensing impedance of flagellar rotation and resistance to type IV pili retraction. There are additional mechanisms that rely on the use of specific membrane-bound adhesins to sense either surface proximity or shear forces. This review aims to examine these mechanosensors, and how they are used by pathogenic bacteria to sense physical features in their environment. We will explore how these sensors generate and transmit signals which can trigger modulation of virulence-associated gene expression in some of the most common bacterial pathogens: Pseudomonas aeruginosa, Proteus mirabilis, Escherichia coli and Vibrio species.
Collapse
Affiliation(s)
- James Mordue
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Nicky O'Boyle
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK
| | - Nikolaj Gadegaard
- School of Engineering, Rankine Building, University of Glasgow, Glasgow G12 8LT, UK
| | - Andrew J Roe
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow G12 8TA, UK.
| |
Collapse
|
34
|
Pappa O, Kefala AM, Tryfinopoulou K, Dimitriou M, Kostoulas K, Dioli C, Moraitou E, Panopoulou M, Vogiatzakis E, Mavridou A, Galanis A, Beloukas A. Molecular Epidemiology of Multi-Drug Resistant Pseudomonas aeruginosa Isolates from Hospitalized Patients in Greece. Microorganisms 2020; 8:microorganisms8111652. [PMID: 33114400 PMCID: PMC7693957 DOI: 10.3390/microorganisms8111652] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 12/13/2022] Open
Abstract
Resistant Pseudomonas aeruginosa isolates are one of the major causes of both hospital-acquired infections (HAIs) and community-acquired infections (CAIs). However, management of P. aeruginosa infections is difficult as the bacterium is inherently resistant to many antibiotics. In this study, a collection of 75 P. aeruginosa clinical isolates from two tertiary hospitals from Athens and Alexnadroupolis in Greece was studied to assess antimicrobial sensitivity and molecular epidemiology. All P. aeruginosa isolates were tested for susceptibility to 11 commonly used antibiotics, and the newly introduced Double Locus Sequence Typing (DLST) scheme was implemented to elucidate the predominant clones. The tested P. aeruginosa isolates presented various resistant phenotypes, with Verona Integron-Mediated Metallo-β-lactamase (VIM-2) mechanisms being the majority, and a new phenotype, FEPR-CAZS, being reported for the first time in Greek isolates. DLST revealed two predominant types, 32-39 and 8-37, and provided evidence for intra-hospital transmission of the 32-39 clone in one of the hospitals. The results indicate that DLST can be a valuable tool when local outbreaks demand immediate tracking investigation with limited time and financial resources.
Collapse
Affiliation(s)
- Olga Pappa
- Department of Biomedical Sciences, University of West Attica, 12243 Egaleo, Greece; (A.M.K.); (M.D.); (C.D.); (A.M.)
- Central Public Health Laboratory, National Public Health Organization, 16672 Athens, Greece;
- Correspondence: or (O.P.); or (A.B.)
| | - Anastasia Maria Kefala
- Department of Biomedical Sciences, University of West Attica, 12243 Egaleo, Greece; (A.M.K.); (M.D.); (C.D.); (A.M.)
| | - Kyriaki Tryfinopoulou
- Central Public Health Laboratory, National Public Health Organization, 16672 Athens, Greece;
| | - Marios Dimitriou
- Department of Biomedical Sciences, University of West Attica, 12243 Egaleo, Greece; (A.M.K.); (M.D.); (C.D.); (A.M.)
| | - Kostas Kostoulas
- Laboratory of Microbiology, ‘Sotiria’ General Hospital, 152 Mesogeion Avenue, 11527 Athens, Greece; (K.K.); (E.M.); (E.V.)
| | - Chrysa Dioli
- Department of Biomedical Sciences, University of West Attica, 12243 Egaleo, Greece; (A.M.K.); (M.D.); (C.D.); (A.M.)
| | - Eleni Moraitou
- Laboratory of Microbiology, ‘Sotiria’ General Hospital, 152 Mesogeion Avenue, 11527 Athens, Greece; (K.K.); (E.M.); (E.V.)
| | - Maria Panopoulou
- Laboratory of Microbiology, School of Medicine, Democritus University of Thrace, Dragana, 68100 Alexandroupolis, Greece;
| | - Evaggelos Vogiatzakis
- Laboratory of Microbiology, ‘Sotiria’ General Hospital, 152 Mesogeion Avenue, 11527 Athens, Greece; (K.K.); (E.M.); (E.V.)
| | - Athena Mavridou
- Department of Biomedical Sciences, University of West Attica, 12243 Egaleo, Greece; (A.M.K.); (M.D.); (C.D.); (A.M.)
| | - Alex Galanis
- Department of Molecular Biology and Genetics, Health Science School, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Apostolos Beloukas
- Department of Biomedical Sciences, University of West Attica, 12243 Egaleo, Greece; (A.M.K.); (M.D.); (C.D.); (A.M.)
- Institute of Infection & Global Health, University of Liverpool, Liverpool L69 7BE, UK
- Correspondence: or (O.P.); or (A.B.)
| |
Collapse
|
35
|
Assessment of Bacterial Contamination of Air at the Museum of King John III’s Palace at Wilanow (Warsaw, Poland): Selection of an Optimal Growth Medium for Analyzing Airborne Bacteria Diversity. APPLIED SCIENCES-BASEL 2020. [DOI: 10.3390/app10207128] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
There is no standardized protocol for the assessment of microbial air contamination in museums and other cultural heritage sites. Therefore, most museums conduct such assessments based on their own guidelines or good practices. Usually, microbial air contamination is assessed using only classical microbiology methods with the application of a single growth medium. Therefore, this medium should be carefully selected to limit any selective cultivation bias. Metabarcoding, i.e., a next-generation sequencing (NGS)-based method, combined with classical microbiological culturing was used to assess the effectiveness of various media applications in microbiological screening at the Museum of King John III’s Palace at Wilanow (Warsaw, Poland). The obtained results indicated that when using a classical microbiology approach to assess the microbial air contamination at the museum, the selection of a proper growth medium was critical. It was shown that the use of rich media (commonly applied by museum conservators) introduced significant bias by severely underreporting putative human pathogens and the bacterial species involved in biodeterioration. Therefore, we recommend the use of other media, such as Frazier or Reasoner’s 2A (R2A) medium, as they could yield more diverse communities and recovered the highest number of genera containing human pathogens, which may be suitable for public health assessments.
Collapse
|
36
|
The Antibacterial and Anti-biofilm Activity of Metal Complexes Incorporating 3,6,9-Trioxaundecanedioate and 1,10-Phenanthroline Ligands in Clinical Isolates of Pseudomonas Aeruginosa from Irish Cystic Fibrosis Patients. Antibiotics (Basel) 2020; 9:antibiotics9100674. [PMID: 33027987 PMCID: PMC7600655 DOI: 10.3390/antibiotics9100674] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 09/29/2020] [Accepted: 10/01/2020] [Indexed: 12/31/2022] Open
Abstract
Chronic infections of Pseudomonas aeruginosa in the lungs of cystic fibrosis (CF) patients are problematic in Ireland where inherited CF is prevalent. The bacteria’s capacity to form a biofilm in its pathogenesis is highly virulent and leads to decreased susceptibility to most antibiotic treatments. Herein, we present the activity profiles of the Cu(II), Mn(II) and Ag(I) tdda-phen chelate complexes {[Cu(3,6,9-tdda)(phen)2]·3H2O·EtOH}n (Cu-tdda-phen), {[Mn(3,6,9-tdda)(phen)2]·3H2O·EtOH}n (Mn-tdda-phen) and [Ag2(3,6,9-tdda)(phen)4]·EtOH (Ag-tdda-phen) (tddaH2 = 3,6,9-trioxaundecanedioic acid; phen = 1,10-phenanthroline) towards clinical isolates of P. aeruginosa derived from Irish CF patients in comparison to two reference laboratory strains (ATCC 27853 and PAO1). The effects of the metal-tdda-phen complexes and gentamicin on planktonic growth, biofilm formation (pre-treatment) and mature biofilm (post-treatment) alone and in combination were investigated. The effects of the metal-tdda-phen complexes on the individual biofilm components; exopolysaccharide, extracellular DNA (eDNA), pyocyanin and pyoverdine are also presented. All three metal-tdda-phen complexes showed comparable and often superior activity to gentamicin in the CF strains, compared to their activities in the laboratory strains, with respect to both biofilm formation and established biofilms. Combination studies presented synergistic activity between all three complexes and gentamicin, particularly for the post-treatment of established mature biofilms, and was supported by the reduction of the individual biofilm components examined.
Collapse
|
37
|
Poerio N, De Santis F, Rossi A, Ranucci S, De Fino I, Henriquez A, D’Andrea MM, Ciciriello F, Lucidi V, Nisini R, Bragonzi A, Fraziano M. Liposomes Loaded With Phosphatidylinositol 5-Phosphate Improve the Antimicrobial Response to Pseudomonas aeruginosa in Impaired Macrophages From Cystic Fibrosis Patients and Limit Airway Inflammatory Response. Front Immunol 2020; 11:532225. [PMID: 33117337 PMCID: PMC7562816 DOI: 10.3389/fimmu.2020.532225] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 09/10/2020] [Indexed: 01/02/2023] Open
Abstract
Despite intensive antimicrobial and anti-inflammatory therapies, cystic fibrosis (CF) patients are subjected to chronic infections due to opportunistic pathogens, including multidrug resistant (MDR) Pseudomonas aeruginosa. Macrophages from CF patients show many evidences of reduced phagocytosis in terms of internalization capability, phagosome maturation, and intracellular bacterial killing. In this study, we investigated if apoptotic body-like liposomes (ABLs) loaded with phosphatidylinositol 5-phosphate (PI5P), known to regulate actin dynamics and vesicular trafficking, could restore phagocytic machinery while limiting inflammatory response in in vitro and in vivo models of MDR P. aeruginosa infection. Our results show that the in vitro treatment with ABL carrying PI5P (ABL/PI5P) enhances bacterial uptake, ROS production, phagosome acidification, and intracellular bacterial killing in human monocyte-derived macrophages (MDMs) with pharmacologically inhibited cystic fibrosis transmembrane conductance regulator channel (CFTR), and improve uptake and intracellular killing of MDR P. aeruginosa in CF macrophages with impaired bactericidal activity. Moreover, ABL/PI5P stimulation of CFTR-inhibited MDM infected with MDR P. aeruginosa significantly reduces NF-κB activation and the production of TNF-α, IL-1β, and IL-6, while increasing IL-10 and TGF-β levels. The therapeutic efficacy of ABL/PI5P given by pulmonary administration was evaluated in a murine model of chronic infection with MDR P. aeruginosa. The treatment with ABL/PI5P significantly reduces pulmonary neutrophil infiltrate and the levels of KC and MCP-2 cytokines in the lungs, without affecting pulmonary bacterial load. Altogether, these results show that the ABL/PI5P treatment may represent a promising host-directed therapeutic approach to improve the impaired phagocytosis and to limit the potentially tissue-damaging inflammatory response in CF.
Collapse
Affiliation(s)
- Noemi Poerio
- Dipartimento di Biologia, Università degli Studi di Roma “Tor Vergata”, Roma, Italy
| | - Federica De Santis
- Dipartimento di Biologia, Università degli Studi di Roma “Tor Vergata”, Roma, Italy
| | - Alice Rossi
- Unità di Infezioni e Fibrosi Cistica, Istituto Scientifico San Raffaele, Milano, Italy
| | - Serena Ranucci
- Unità di Infezioni e Fibrosi Cistica, Istituto Scientifico San Raffaele, Milano, Italy
| | - Ida De Fino
- Unità di Infezioni e Fibrosi Cistica, Istituto Scientifico San Raffaele, Milano, Italy
| | - Ana Henriquez
- Dipartimento di Biologia, Università degli Studi di Roma “Tor Vergata”, Roma, Italy
| | - Marco M. D’Andrea
- Dipartimento di Biologia, Università degli Studi di Roma “Tor Vergata”, Roma, Italy
| | - Fabiana Ciciriello
- Unità Operativa Complessa Fibrosi Cistica, Dipartimento di Medicina Pediatrica, Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Vincenzina Lucidi
- Unità Operativa Complessa Fibrosi Cistica, Dipartimento di Medicina Pediatrica, Ospedale Pediatrico Bambino Gesù, Roma, Italy
| | - Roberto Nisini
- Dipartimento di Malattie Infettive, Istituto Superiore di Sanità, Roma, Italy
| | - Alessandra Bragonzi
- Unità di Infezioni e Fibrosi Cistica, Istituto Scientifico San Raffaele, Milano, Italy
| | - Maurizio Fraziano
- Dipartimento di Biologia, Università degli Studi di Roma “Tor Vergata”, Roma, Italy
| |
Collapse
|
38
|
Shao X, Xie Y, Zhang Y, Liu J, Ding Y, Wu M, Wang X, Deng X. Novel therapeutic strategies for treating Pseudomonas aeruginosa infection. Expert Opin Drug Discov 2020; 15:1403-1423. [PMID: 32880507 DOI: 10.1080/17460441.2020.1803274] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Persistent infections caused by the superbug Pseudomonas aeruginosa and its resistance to multiple antimicrobial agents are huge threats to patients with cystic fibrosis as well as those with compromised immune systems. Multidrug-resistant P. aeruginosa has posed a major challenge to conventional antibiotics and therapeutic approaches, which show limited efficacy and cause serious side effects. The public demand for new antibiotics is enormous; yet, drug development pipelines have started to run dry with limited targets available for inventing new antibacterial drugs. Consequently, it is important to uncover potential therapeutic targets. AREAS COVERED The authors review the current state of drug development strategies that are promising in terms of the development of novel and potent drugs to treat P. aeruginosa infection. EXPERT OPINION The prevention of P. aeruginosa infection is increasingly challenging. Furthermore, targeting key virulence regulators has great potential for developing novel anti-P. aeruginosa drugs. Additional promising strategies include bacteriophage therapy, immunotherapies, and antimicrobial peptides. Additionally, the authors believe that in the coming years, the overall network of molecular regulatory mechanism of P. aeruginosa virulence will be fully elucidated, which will provide more novel and promising drug targets for treating P. aeruginosa infections.
Collapse
Affiliation(s)
- Xiaolong Shao
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong SAR, China
| | - Yingpeng Xie
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong SAR, China
| | - Yingchao Zhang
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong SAR, China
| | - Jingui Liu
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong SAR, China
| | - Yiqing Ding
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong SAR, China
| | - Min Wu
- Department of Biomedical Sciences, University of North Dakota , Grand Forks, North Dakota, USA
| | - Xin Wang
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong SAR, China
| | - Xin Deng
- Department of Biomedical Sciences, City University of Hong Kong , Hong Kong SAR, China.,Shenzhen Research Institute, City University of Hong Kong , Shenzhen, China
| |
Collapse
|
39
|
Novel Aminoglycoside-Tolerant Phoenix Colony Variants of Pseudomonas aeruginosa. Antimicrob Agents Chemother 2020; 64:AAC.00623-20. [PMID: 32540981 DOI: 10.1128/aac.00623-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 06/09/2020] [Indexed: 01/13/2023] Open
Abstract
Pseudomonas aeruginosa is an opportunistic bacterial pathogen and is known to produce biofilms. We previously showed the emergence of colony variants in the presence of tobramycin-loaded calcium sulfate beads. In this study, we characterized the variant colonies, which survived the antibiotic treatment, and identified three distinct phenotypes-classically resistant colonies, viable but nonculturable colonies (VBNC), and phoenix colonies. Phoenix colonies, described here for the first time, grow out of the zone of clearance of antibiotic-loaded beads from lawn biofilms while there are still very high concentrations of antibiotic present, suggesting an antibiotic-resistant phenotype. However, upon subculturing of these isolates, phoenix colonies return to wild-type levels of antibiotic susceptibility. Compared with the wild type, phoenix colonies are morphologically similar aside from a deficiency in green pigmentation. Phoenix colonies do not recapitulate the phenotype of any previously described mechanisms of resistance, tolerance, or persistence and, thus, form a novel group with their own phenotype. Growth under anaerobic conditions suggests that an alternative metabolism could lead to the formation of phoenix colonies. These findings suggest that phoenix colonies could emerge in response to antibiotic therapies and lead to recurrent or persistent infections, particularly within biofilms where microaerobic or anaerobic environments are present.
Collapse
|
40
|
Clinically Relevant Epithelial Lining Fluid Concentrations of Meropenem with Ciprofloxacin Provide Synergistic Killing and Resistance Suppression of Hypermutable Pseudomonas aeruginosa in a Dynamic Biofilm Model. Antimicrob Agents Chemother 2020; 64:AAC.00469-20. [PMID: 32366710 DOI: 10.1128/aac.00469-20] [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] [Received: 03/11/2020] [Accepted: 04/20/2020] [Indexed: 01/18/2023] Open
Abstract
Treatment of exacerbations of chronic Pseudomonas aeruginosa infections in patients with cystic fibrosis (CF) is highly challenging due to hypermutability, biofilm formation, and an increased risk of resistance emergence. We evaluated the impact of ciprofloxacin and meropenem as monotherapy and in combination in the dynamic in vitro CDC biofilm reactor (CBR). Two hypermutable P. aeruginosa strains, PAOΔmutS (MIC of ciprofloxacin [MICciprofloxacin], 0.25 mg/liter; MICmeropenem, 2 mg/liter) and CW44 (MICciprofloxacin, 0.5 mg/liter; MICmeropenem, 4 mg/liter), were investigated for 120 h. Concentration-time profiles achievable in epithelial lining fluid (ELF) following FDA-approved doses were simulated in the CBR. Treatments were ciprofloxacin at 0.4 g every 8 h as 1-h infusions (80% ELF penetration), meropenem at 6 g/day as a continuous infusion (CI) (30% and 60% ELF penetration), and their combinations. Counts of total and less-susceptible planktonic and biofilm bacteria and MICs were determined. Antibiotic concentrations were quantified by an ultrahigh-performance liquid chromatography photodiode array (UHPLC-PDA) assay. For both strains, all monotherapies failed, with substantial regrowth and resistance of planktonic (≥8 log10 CFU/ml) and biofilm (>8 log10 CFU/cm2) bacteria at 120 h (MICciprofloxacin, up to 8 mg/liter; MICmeropenem, up to 64 mg/liter). Both combination treatments demonstrated synergistic bacterial killing of planktonic and biofilm bacteria of both strains from ∼48 h onwards and suppressed regrowth to ≤4 log10 CFU/ml and ≤6 log10 CFU/cm2 at 120 h. Overall, both combination treatments suppressed the amplification of resistance of planktonic bacteria for both strains and of biofilm bacteria for CW44. The combination with meropenem at 60% ELF penetration also suppressed the amplification of resistance of biofilm bacteria for PAOΔmutS Thus, combination treatment demonstrated synergistic bacterial killing and resistance suppression against difficult-to-treat hypermutable P. aeruginosa strains.
Collapse
|
41
|
Prevention of chronic infection with Pseudomonas aeruginosa infection in cystic fibrosis. Curr Opin Pulm Med 2020; 25:636-645. [PMID: 31397692 DOI: 10.1097/mcp.0000000000000616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
PURPOSE OF REVIEW This review provides an update on definitions of chronicity of infection, approaches to airway sampling to detect infection, strategies for Pseudomonas aeruginosa eradication, impact of cystic fibrosis transmembrane regulator protein (CFTR) modulators and future challenges for clinical trials. RECENT FINDINGS Rates of P. aeruginosa have decreased over the past two decades with establishment of effective eradication protocols. Definitions of chronic P. aeruginosa infection have required adaptation for healthier populations. Although molecular (PCR) approaches to early P. aeruginosa detection are sensitive, to date, earlier diagnosis has not impacted on clinical outcomes. Despite eradication regimens, some people with early P. aeruginosa fail to clear their infection. Most people also experience a recurrence and eventual transition to chronic infection. Several recent studies sought to address this gap. CFTR modulators (predominantly ivacaftor) demonstrated reduced P. aeruginosa density, although infection may persist or recur demonstrating the need for continued antiinfective therapies in the modulator era. SUMMARY Future studies of approaches to P. aeruginosa eradication will be complex due to expanded availability and ongoing competitive clinical trials of CFTR modulators. Studies to address optimal eradication therapy, particularly in adults, will be required, though adequate recruitment to power these studies may prove challenging.
Collapse
|
42
|
Li Y, Chen L, Zhang P, Bhagirath AY, Duan K. ClpV3 of the H3-Type VI Secretion System (H3-T6SS) Affects Multiple Virulence Factors in Pseudomonas aeruginosa. Front Microbiol 2020; 11:1096. [PMID: 32547522 PMCID: PMC7273116 DOI: 10.3389/fmicb.2020.01096] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2020] [Accepted: 05/01/2020] [Indexed: 11/13/2022] Open
Abstract
The type VI secretion system (T6SS) is a toxic effector delivery apparatus widely distributed in Gram-negative bacteria. The opportunistic pathogen Pseudomonas aeruginosa encodes three T6SSs, namely H1-, H2-, and H3-T6SS. Each T6SS possesses its own effectors and their roles are not yet fully understood. Here, we report that an H3-T6SS deletion mutant PAO1(ΔclpV3) significantly affected the virulence-related phenotypes including pyocyanin production, biofilm formation, proteolytic activity, and motilities. Most interestingly, the expression of T3SS genes was markedly affected, indicating a link between H3-T6SS and T3SS. RNA-Sequencing was performed to globally identify the genes differentially expressed when H3-T6SS was inactivated and the results obtained correlated well with the observed phenotypes. Interestingly, the expressions of T2SS, T3SS, H2-T6SS, and H3-T6SS were all significantly decreased, while H1-T6SS was increased in the PAO1(ΔclpV3) strain. We also observed that the intracellular concentration of secondary messenger cAMP was reduced in PAO1(ΔclpV3), and the c-di-GMP level was also decreased as indicated by the decreased cdrA reporter activity. Finally, by using a Galleria mellonella infection model, we show that H3-T6SS plays a key role in the pathogenicity of P. aeruginosa in vivo. Overall, our study highlights the unique connection of H3-T6SS in P. aeruginosa with T3SS, pyocyanin production, biofilm formation and in vivo pathogenicity.
Collapse
Affiliation(s)
- Yanqi Li
- Department of Oral Biology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Lin Chen
- College of Life Sciences, Northwest University, Xi’an, China
| | - Pansong Zhang
- College of Life Sciences, Northwest University, Xi’an, China
| | - Anjali Y. Bhagirath
- Department of Oral Biology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| | - Kangmin Duan
- Department of Oral Biology, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
- Department of Medical Microbiology & Infectious Diseases, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
43
|
Isopi E, Mattoscio D, Codagnone M, Mari VC, Lamolinara A, Patruno S, D'Aurora M, Cianci E, Nespoli A, Franchi S, Gatta V, Dubourdeau M, Moretti P, Di Sabatino M, Iezzi M, Romano M, Recchiuti A. Resolvin D1 Reduces Lung Infection and Inflammation Activating Resolution in Cystic Fibrosis. Front Immunol 2020; 11:581. [PMID: 32528461 PMCID: PMC7247852 DOI: 10.3389/fimmu.2020.00581] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Accepted: 03/12/2020] [Indexed: 12/13/2022] Open
Abstract
Non-resolving lung inflammation and Pseudomonas aeruginosa infections are the underlying cause of morbidity and mortality in cystic fibrosis (CF). The endogenous lipid mediator resolvin (Rv) D1 is a potent regulator of resolution, and its roles, actions, and therapeutic potential in CF are of interest. Here, we investigated actions and efficacy of RvD1 in preclinical models of cystic fibrosis. Cftr knockout mice with chronic P. aeruginosa lung infection were treated with RvD1 to assess differences in lung bacterial load, inflammation, and tissue damage. Cells from volunteers with CF were treated with RvD1 during ex vivo infection with P. aeruginosa, and effects on phagocytosis and inflammatory signaling were determined. In CF mice, RvD1 reduced bacterial burden, neutrophil infiltration, and histological signs of lung pathology, improving clinical scores of diseases. Mechanistically, RvD1 increased macrophage-mediated bacterial and leukocyte clearance in vivo. The clinical significance of these findings is supported by actions in primary leukocytes and epithelial cells from volunteers with CF where RvD1 enhanced P. aeruginosa phagocytosis and reduced genes and proteins associated to NF-κB activation and leukocyte infiltration. Concentration of RvD1 in sputum from patients with CF was also inversely correlated to those of cytokines and chemokines involved in CF lung pathology. These findings demonstrate efficacy of RvD1 in enhancing resolution of lung inflammation and infections and provide proof of concept for its potential as a prototypic novel pro-resolutive therapeutic approach for CF.
Collapse
Affiliation(s)
- Elisa Isopi
- Center for Advanced Studies and Technology, Department of Medical, Oral and Biotechnology Science, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Domenico Mattoscio
- Center for Advanced Studies and Technology, Department of Medical, Oral and Biotechnology Science, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Marilina Codagnone
- Center for Advanced Studies and Technology, Department of Medical, Oral and Biotechnology Science, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Veronica Cecilia Mari
- Center for Advanced Studies and Technology, Department of Medical, Oral and Biotechnology Science, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Alessia Lamolinara
- Center for Advanced Studies and Technology, Department of Medicine and Aging Sciences, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Sara Patruno
- Center for Advanced Studies and Technology, Department of Medical, Oral and Biotechnology Science, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Marco D'Aurora
- Center for Advanced Studies and Technology, Department of Psychological, Humanistic and Territorial Sciences, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Eleonora Cianci
- Center for Advanced Studies and Technology, Department of Medical, Oral and Biotechnology Science, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Annalisa Nespoli
- Center for Advanced Studies and Technology, Department of Medicine and Aging Sciences, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Sara Franchi
- Center for Advanced Studies and Technology, Department of Psychological, Humanistic and Territorial Sciences, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Valentina Gatta
- Center for Advanced Studies and Technology, Department of Psychological, Humanistic and Territorial Sciences, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | | | - Paolo Moretti
- Cystic Fibrosis Regional Center, Ospedale "San Liberatore," Atri, Italy
| | - Maria Di Sabatino
- Cystic Fibrosis Regional Center, Ospedale "San Liberatore," Atri, Italy
| | - Manuela Iezzi
- Center for Advanced Studies and Technology, Department of Medicine and Aging Sciences, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Mario Romano
- Center for Advanced Studies and Technology, Department of Medical, Oral and Biotechnology Science, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| | - Antonio Recchiuti
- Center for Advanced Studies and Technology, Department of Medical, Oral and Biotechnology Science, "G. d'Annunzio" University of Chieti - Pescara, Chieti, Italy
| |
Collapse
|
44
|
Leiter H, Toepfer S, Messner P, Rabensteiner M, Gostner JM, Lackner M, Hermann M, Nagl M. Microbicidal activity of N-chlorotaurine can be enhanced in the presence of lung epithelial cells. J Cyst Fibros 2020; 19:1011-1017. [PMID: 32201161 DOI: 10.1016/j.jcf.2020.03.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 03/07/2020] [Accepted: 03/07/2020] [Indexed: 01/24/2023]
Abstract
BACKGROUND N-chlorotaurine (NCT) is an endogenous active chlorine compound that can be used as an antiseptic and anti-infective in different body regions. Recently, tolerability of inhaled NCT has been demonstrated in humans so that it is of interest for future treatment of cystic fibrosis. In the present study, we tested the bactericidal and fungicidal activity of NCT in different lung cell culture models. METHODS Bacteria (Staphylococcus aureus, Pseudomonas aeruginosa) and fungi (Candida albicans, Exophiala dermatitidis) were co-incubated with lung epithelial cell cultures, and after 4 h NCT was added. After different incubation times, aliquots were removed and quantitative cultures were performed. RESULTS NCT at the therapeutically applied concentration of 1% (55 mM) completely killed the test pathogens within 15 - 30 min at 20 °C and at 37 °C. Killing by 0.3% NCT lasted up to 4 h dependent on the pathogen at 20 °C and up to 1 h at 37 °C. 0.1% NCT was the threshold concentration for killing since this amount of oxidation capacity was consumed by reactions with the organic compounds of the medium within 3 h (20 °C) and 0.5 h (37 °C). CONCLUSIONS NCT in therapeutic concentration demonstrated its microbicidal activity in the presence of lung epithelial cells. Remarkably, particularly the fungicidal activity was higher under these conditions than in phosphate buffer. This can be explained by formation of the stronger microbicidal monochloramine in equilibrium by transchlorination. The results suggest the suitability of NCT as inhalation medication in the lung.
Collapse
Affiliation(s)
- Hannes Leiter
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Schöpfstr. 41, A-6020 Innsbruck, Austria
| | - Stephanie Toepfer
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Schöpfstr. 41, A-6020 Innsbruck, Austria; Division of Medical Biochemistry, Medical University of Innsbruck, Austria
| | - Petra Messner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Schöpfstr. 41, A-6020 Innsbruck, Austria
| | - Marion Rabensteiner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Schöpfstr. 41, A-6020 Innsbruck, Austria
| | - Johanna M Gostner
- Division of Medical Biochemistry, Medical University of Innsbruck, Austria
| | - Michaela Lackner
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Schöpfstr. 41, A-6020 Innsbruck, Austria
| | - Martin Hermann
- Department of Anaesthesiology and Critical Care Medicine, Medical University of Innsbruck, Austria
| | - Markus Nagl
- Institute of Hygiene and Medical Microbiology, Medical University of Innsbruck, Schöpfstr. 41, A-6020 Innsbruck, Austria.
| |
Collapse
|
45
|
Kitao T. [Molecular biological studies toward controlling infectious diseases caused by multidrug-resistant Pseudomonas aeruginosa]. Nihon Saikingaku Zasshi 2020; 74:177-189. [PMID: 31902822 DOI: 10.3412/jsb.74.177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Pseudomonas aeruginosa is an opportunistic Gram-negative pathogen that causes serious acute, persistent, and relapsing infections. Recent year, the effectiveness of antibiotics for eliminating P. aeruginosa infections has been further complicated by the emergence of multidrug-resistant strains. Thus, new approaches for the rapid detection and novel antimicrobial drug discovery are urgently needed to control such intractable infections caused by the pathogen. Also, we do need deep understanding of the drug resistance mechanisms to overcome this issue. Here I describe a brief review on my biological studies toward controlling infectious diseases caused by multidrug-resistant P. aeruginosa.
Collapse
Affiliation(s)
- Tomoe Kitao
- Department of Microbiology, Graduate School of Medicine, Gifu University
| |
Collapse
|
46
|
Morrison JM, Chojnacki M, Fadrowski JJ, Bauza C, Dunman PM, Dudas RA, Goldenberg NA, Berman DM. Serum-Associated Antibiotic Tolerance in Pediatric Clinical Isolates of Pseudomonas aeruginosa. J Pediatric Infect Dis Soc 2019; 9:671-679. [PMID: 31886511 PMCID: PMC7974018 DOI: 10.1093/jpids/piz094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 12/05/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND When grown in human serum, laboratory isolates of Pseudomonas aeruginosa exhibit tolerance to antibiotics at inhibitory concentrations. This phenomenon, known as serum-associated antibiotic tolerance (SAT), could lead to clinical treatment failure of pseudomonal infections. Our purpose in this study was to determine the prevalence and clinical impact of SAT in Pseudomonas isolates in hospitalized children. METHODS The SAT phenotype was assessed in patients aged <18 years admitted with respiratory or blood cultures positive for P. aeruginosa. The SAT phenotype was a priori defined as a ≥2-log increase in colony-forming units when grown in human serum compared with Luria-Bertani medium in the presence of minocycline or tobramycin. RESULTS SAT was detected in 29 (64%) patients. Fourteen patients each (34%) had cystic fibrosis (CF) and tracheostomies. Patient demographics and comorbidities did not differ by SAT status. Among CF patients, SAT was associated with longer duration of intravenous antibiotics (10 days vs 5 days; P < .01). CONCLUSIONS This study establishes that SAT exists in P. aeruginosa from human serum and may be a novel factor that contributes to differences in clinical outcomes. Future research should investigate the mechanisms that contribute to SAT in order to identify novel targets for adjunctive antimicrobial therapies.
Collapse
Affiliation(s)
- John M Morrison
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Medicine, Division of Hospital Medicine, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA,Correspondence: John M. Morrison, 601 Fifth Street South Suite 501, Saint Petersburg, FL 33701 ()
| | - Michaelle Chojnacki
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Jeffrey J Fadrowski
- Department of Pediatrics, Division of Nephrology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Colleen Bauza
- Department of Health Informatics, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Paul M Dunman
- Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, New York, USA
| | - Robert A Dudas
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA,Department of Medicine, Division of Hospital Medicine, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - Neil A Goldenberg
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA, and All Children’s Research Institute, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| | - David M Berman
- Department of Medicine, Division of Infectious Diseases, Johns Hopkins All Children’s Hospital, St. Petersburg, Florida, USA
| |
Collapse
|
47
|
Synergistic Meropenem-Tobramycin Combination Dosage Regimens against Clinical Hypermutable Pseudomonas aeruginosa at Simulated Epithelial Lining Fluid Concentrations in a Dynamic Biofilm Model. Antimicrob Agents Chemother 2019; 63:AAC.01293-19. [PMID: 31427301 DOI: 10.1128/aac.01293-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2019] [Accepted: 08/12/2019] [Indexed: 12/19/2022] Open
Abstract
Exacerbations of chronic Pseudomonas aeruginosa infections are a major treatment challenge in cystic fibrosis due to biofilm formation and hypermutation. We aimed to evaluate different dosage regimens of meropenem and tobramycin as monotherapies and in combination against hypermutable carbapenem-resistant P. aeruginosa A hypermutable P. aeruginosa isolate (meropenem and tobramycin MICs, 8 mg/liter) was investigated in the dynamic CDC biofilm reactor over 120 h. Regimens were meropenem as the standard (2 g every 8 h, 30% epithelial lining fluid [ELF] penetration) and as a continuous infusion (CI; 6 g/day, 30% and 60% ELF penetration) and tobramycin at 10 mg/kg of body weight every 24 h (50% ELF penetration). The time courses of totally susceptible and less-susceptible bacteria and MICs were determined, and antibiotic concentrations were quantified by liquid chromatography-tandem mass spectrometry. All monotherapies failed, with the substantial regrowth of planktonic (>6 log10 CFU/ml) and biofilm (≥6 log10 CFU/cm2) bacteria occurring. Except for the meropenem CI (60% ELF penetration), all monotherapies amplified less-susceptible planktonic and biofilm bacteria by 120 h. The meropenem standard regimen with tobramycin caused initial killing followed by considerable regrowth with resistance (meropenem MIC, 64 mg/liter; tobramycin MIC, 32 mg/liter) for planktonic and biofilm bacteria. The combination containing the meropenem CI at both levels of ELF penetration synergistically suppressed the regrowth of total planktonic bacteria and the resistance of planktonic and biofilm bacteria. The combination with the meropenem CI at 60% ELF penetration, in addition, synergistically suppressed the regrowth of total biofilm bacteria. Standard regimens of meropenem and tobramycin were ineffective against planktonic and biofilm bacteria. The combination with meropenem CI exhibited enhanced bacterial killing and resistance suppression of carbapenem-resistant hypermutable P. aeruginosa.
Collapse
|
48
|
Buhl M, Kästle C, Geyer A, Autenrieth IB, Peter S, Willmann M. Molecular Evolution of Extensively Drug-Resistant (XDR) Pseudomonas aeruginosa Strains From Patients and Hospital Environment in a Prolonged Outbreak. Front Microbiol 2019; 10:1742. [PMID: 31440214 PMCID: PMC6694792 DOI: 10.3389/fmicb.2019.01742] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Accepted: 07/15/2019] [Indexed: 11/24/2022] Open
Abstract
In this study, we aimed to elucidate a prolonged outbreak of extensively drug-resistant (XDR) Pseudomonas aeruginosa, at two adjacent hospitals over a time course of 4 years. Since all strains exhibited a similar antibiotic susceptibility pattern and carried the carbapenemase gene blaVIM, a monoclonal outbreak was assumed. To shed light on the intra-hospital evolution of these strains over time, whole genome sequence (WGS) analysis of 100 clinical and environmental outbreak strains was employed. Phylogenetic analysis of the core genome revealed the outbreak to be polyclonal, rather than monoclonal as initially suggested. The vast majority of strains fell into one of two major clusters, composed of 27 and 59 strains, and their accessory genome each revealed over 400 and 600 accessory genes, respectively, thus indicating an unexpectedly high structural diversity among phylogenetically clustered strains. Further analyses focused on the cluster with 59 strains, representing the hospital from which both clinical and environmental strains were available. Our investigation clearly shows both accumulation and loss of genes occur very frequently over time, as reflected by analysis of protein enrichment as well as functional enrichment. In addition, we investigated adaptation through single nucleotide polymorphisms (SNPs). Among the genes affected by SNPs, there are a multidrug efflux pump (mexZ) and a mercury detoxification operon (merR) with deleterious mutations, potentially leading to loss of repression with resistance against antibiotics and disinfectants. Our results not only confirm WGS to be a powerful tool for epidemiologic analyses, but also provide insights into molecular evolution during an XDR P. aeruginosa hospital outbreak. Genome mutation unveiled a striking genetic plasticity on an unexpectedly high level, mostly driven by horizontal gene transfer. Our study adds valuable information to the molecular understanding of “real-world” Intra-hospital P. aeruginosa evolution and is a step forward toward more personalized medicine in infection control.
Collapse
Affiliation(s)
- Michael Buhl
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Christina Kästle
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
| | - André Geyer
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany
| | - Ingo B Autenrieth
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Silke Peter
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| | - Matthias Willmann
- Institute of Medical Microbiology and Hygiene, University of Tübingen, Tübingen, Germany.,German Center for Infection Research, Partner Site Tübingen, Tübingen, Germany
| |
Collapse
|
49
|
Lyons BJE, Strynadka NCJ. On the road to structure-based development of anti-virulence therapeutics targeting the type III secretion system injectisome. MEDCHEMCOMM 2019; 10:1273-1289. [PMID: 31534650 PMCID: PMC6748289 DOI: 10.1039/c9md00146h] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Accepted: 06/07/2019] [Indexed: 12/19/2022]
Abstract
The type III secretion system injectisome is a syringe-like multimembrane spanning nanomachine that is essential to the pathogenicity but not viability of many clinically relevant Gram-negative bacteria, such as enteropathogenic Escherichia coli, Salmonella enterica and Pseudomonas aeruginosa. Due to the rise in antibiotic resistance, new strategies must be developed to treat the growing spectre of drug resistant infections. Targeting the injectisome via an 'anti-virulence strategy' is a promising avenue to pursue as an alternative to the more commonly used bactericidal therapeutics, which have a high propensity for resulting resistance development and often more broad killing profile, including unwanted side effects in eliminating favourable members of the microbiome. Building on more than a decade of crystallographic work of truncated or isolated forms of the more than two dozen components of the secretion apparatus, recent advances in the field of single-particle cryo-electron microscopy have allowed for the elucidation of atomic resolution structures for many of the type III secretion system components in their assembled, oligomerized state including the needle complex, export apparatus and ATPase. Cryo-electron tomography studies have also advanced our understanding of the direct pathogen-host interaction between the type III secretion system translocon and host cell membrane. These new structural works that further our understanding of the myriad of protein-protein interactions that promote injectisome function will be highlighted in this review, with a focus on those that yield promise for future anti-virulence drug discovery and design. Recently developed inhibitors, including both synthetic, natural product and peptide inhibitors, as well as promising new developments of immunotherapeutics will be discussed. As our understanding of this intricate molecular machinery advances, the development of anti-virulence inhibitors can be enhanced through structure-guided drug design.
Collapse
Affiliation(s)
- Bronwyn J E Lyons
- Department of Biochemistry and Molecular Biology and Center for Blood Research , University of British Columbia , 2350 Health Sciences Mall , Vancouver , British Columbia V6T 1Z3 , Canada .
| | - Natalie C J Strynadka
- Department of Biochemistry and Molecular Biology and Center for Blood Research , University of British Columbia , 2350 Health Sciences Mall , Vancouver , British Columbia V6T 1Z3 , Canada .
| |
Collapse
|
50
|
Mottais A, Berchel M, Le Gall T, Sibiril Y, d'Arbonneau F, Laurent V, Jaffrès PA, Montier T. Antibacterial and transfection activities of nebulized formulations incorporating long n-alkyl chain silver N-heterocyclic carbene complexes. Int J Pharm 2019; 567:118500. [DOI: 10.1016/j.ijpharm.2019.118500] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 01/16/2023]
|