1
|
Beizman-Magen Y, Orevi T, Kashtan N. Hydration conditions as a critical factor in antibiotic-mediated bacterial competition outcomes. Appl Environ Microbiol 2025; 91:e0200424. [PMID: 39714150 PMCID: PMC11784440 DOI: 10.1128/aem.02004-24] [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: 10/10/2024] [Accepted: 11/27/2024] [Indexed: 12/24/2024] Open
Abstract
Antibiotic secretion plays a pivotal role in bacterial interference competition; yet, the impact of environmental hydration conditions on such competition is not well understood. Here, we investigate how hydration conditions affect interference competition among bacteria, studying the interactions between the antibiotic-producing Bacillus velezensis FZB42 and two bacterial strains susceptible to its antibiotics: Xanthomonas euvesicatoria 85-10 and Pseudomonas syringae DC3000. Our results show that wet-dry cycles significantly modify the response of the susceptible bacteria to both the supernatant and cells of the antibiotic-producing bacteria, compared to constantly wet conditions. Notably, X. euvesicatoria shows increased protection against both the cells and supernatants of B. velezensis under wet-dry cycles, while P. syringae cells become more susceptible under wet-dry cycles. In addition, we observed a reciprocal interaction between P. syringae and B. velezensis, where P. syringae inhibits B. velezensis under wet conditions. Our findings highlight the important role of hydration conditions in shaping bacterial interference competition, providing valuable insights into the microbial ecology of water-unsaturated surfaces, with implications for applications such as biological control of plant pathogens and mitigating antibiotic resistance.IMPORTANCEOur study reveals that hydration conditions, particularly wet-dry cycles, significantly influence antibiotic-mediated competition between bacterial species. We revealed that the effectiveness of antibiotics produced by Bacillus velezensis against two susceptible bacterial species: Xanthomonas and Pseudomonas varies based on these hydration conditions. Unlike traditional laboratory environments, many real-world habitats, such as soil, plant surfaces, and even animal skin, undergo frequent wet-dry cycles. These conditions affect bacterial competition dynamics and outcomes, with wet-dry cycles providing increased protection for some bacteria while making others more susceptible. Our findings highlight the importance of considering environmental hydration when studying microbial interactions and developing biological control strategies. This research has important implications for improving agricultural practices and understanding natural microbial ecosystems.
Collapse
Affiliation(s)
- Yana Beizman-Magen
- Institute of Environmental Sciences, Department of Plant Pathology and Microbiology, Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University, Rehovot, Israel
| | - Tomer Orevi
- Institute of Environmental Sciences, Department of Plant Pathology and Microbiology, Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University, Rehovot, Israel
| | - Nadav Kashtan
- Institute of Environmental Sciences, Department of Plant Pathology and Microbiology, Robert H. Smith Faculty of Agriculture, Food, and Environment, Hebrew University, Rehovot, Israel
| |
Collapse
|
2
|
Niedźwiedzka A, Micallef MP, Biazzo M, Podrini C. The Role of the Skin Microbiome in Acne: Challenges and Future Therapeutic Opportunities. Int J Mol Sci 2024; 25:11422. [PMID: 39518974 PMCID: PMC11546345 DOI: 10.3390/ijms252111422] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
Acne vulgaris is a widespread dermatological condition that significantly affects the quality of life of adolescents and adults. Traditionally, acne pathogenesis has been linked to factors such as excess sebum production, follicular hyperkeratinization, and the presence of Cutibacterium acnes (C. acnes). However, recent studies have highlighted the role of the skin microbiome, shifting focus from individual pathogens to microbial community dynamics. This review critically evaluates existing research on the skin microbiome and its relationship to acne, focusing on microbial diversity, C. acnes strain variability, and emerging therapies targeting the microbiome. While certain studies associate C. acnes with acne severity, others show this bacterium's presence in healthy skin, suggesting that strain-specific differences and overall microbial balance play crucial roles. Emerging therapeutic approaches, such as probiotics and bacteriophage therapy, aim to restore microbial equilibrium or selectively target pathogenic strains without disturbing the broader microbiome. However, the lack of standardized methodologies, limited longitudinal studies, and the narrow focus on bacterial communities are major limitations in current research. Future research should explore the broader skin microbiome, including fungi and viruses, use consistent methodologies, and focus on longitudinal studies to better understand microbial fluctuations over time. Addressing these gaps will enable the development of more effective microbiome-based treatments for acne. In conclusion, while microbiome-targeted therapies hold promise, further investigation is needed to validate their efficacy and safety, paving the way for innovative, personalized acne management strategies.
Collapse
Affiliation(s)
| | | | | | - Christine Podrini
- The BioArte Ltd., Malta Life Science Park, Triq San Giljan, SGN 3000 San Gwann, Malta
| |
Collapse
|
3
|
Del Rosso JQ, Armillei MK, Lomakin IB, Grada A, Bunick CG. Clindamycin: A Comprehensive Status Report with Emphasis on Use in Dermatology. THE JOURNAL OF CLINICAL AND AESTHETIC DERMATOLOGY 2024; 17:29-40. [PMID: 39148960 PMCID: PMC11324192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Clindamycin is a lincosamide antibiotic that has been used as a topical, oral, or injectable formulation for over five decades. It exhibits a narrow spectrum of microbiologic activity, primarily against gram-positive and anaerobic bacteria. In dermatology, clindamycin has been used primarily as a topical agent, usually for the treatment of acne vulgaris. Despite questions surrounding antibiotic resistance and/or its relative contribution to antibiotic treatment efficacy, a large body of data support the therapeutic value of topical clindamycin for acne vulgaris. As a systemic agent, clindamycin is used orally to treat a variety of cutaneous bacterial infections, and sometimes for acne vulgaris, with oral treatment for the latter less common in more recent years. The modes of action of clindamycin are supported by data showing both its anti-inflammatory and antibiotic mechanisms, which are discussed here along with pharmacokinetic profiles and structure-activity relationships. The diverse applications of clindamycin for multiple disease states, its efficacy, and safety considerations are also reviewed here, including for both topical and systemic formulations. Emphasis is placed on uses in dermatology, but other information on clindamycin relevant to clinicians is also discussed.
Collapse
Affiliation(s)
- James Q. Del Rosso
- Dr. Del Rosso is Adjunct Clinical Faculty in Dermatology at Touro University Nevada in Henderson, Nevada, JDR Dermatology Research in Las Vegas, Nevada, and Advanced Dermatology and Cosmetic Surgery in Maitland, Florida
| | - Maria K. Armillei
- Ms. Armillei is with the Translational Biomedicine Program at the Yale University School of Medicine in New Haven, Connecticut
| | - Ivan B. Lomakin
- Dr. Lomakin is with the Department of Dermatology at Yale University in New Haven, Connecticut
| | - Ayman Grada
- Dr. Grada is with the Department of Dermatology at Case Western Reserve University School of Medicine in Cleveland, Ohio
| | - Christopher G. Bunick
- Dr. Bunick is with the Translational Biomedicine Program at the Yale University School of Medicine in New Haven, Connecticut, and the Department of Dermatology at Yale University in New Haven, Connecticut
| |
Collapse
|
4
|
Bava R, Castagna F, Lupia C, Poerio G, Liguori G, Lombardi R, Naturale MD, Mercuri C, Bulotta RM, Britti D, Palma E. Antimicrobial Resistance in Livestock: A Serious Threat to Public Health. Antibiotics (Basel) 2024; 13:551. [PMID: 38927217 PMCID: PMC11200672 DOI: 10.3390/antibiotics13060551] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/04/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial resistance represents an alarming public health problem; its importance is related to the significant clinical implications (increased morbidity, mortality, disease duration, development of comorbidities, and epidemics), as well as its economic effects on the healthcare sector. In fact, therapeutic options are severely limited by the advent and spread of germs resistant to many antibiotics. The situation worldwide is worrying, especially in light of the prevalence of Gram-negative bacteria-Klebsiella pneumoniae and Acinetobacter baumannii-which are frequently isolated in hospital environments and, more specifically, in intensive care units. The problem is compounded by the ineffective treatment of infections by patients who often self-prescribe therapy. Resistant bacteria also show resistance to the latest generation antibiotics, such as carbapenems. In fact, superbacteria, grouped under the acronym extended-spectrum betalactamase (ESBL), are becoming common. Antibiotic resistance is also found in the livestock sector, with serious repercussions on animal production. In general, this phenomenon affects all members of the biosphere and can only be addressed by adopting a holistic "One Health" approach. In this literature overview, a stock is taken of what has been learned about antibiotic resistance, and suggestions are proposed to stem its advance.
Collapse
Affiliation(s)
- Roberto Bava
- Department of Health Sciences, University of Catanzaro Magna Græcia, 88100 Catanzaro, Italy; (R.B.); (C.L.); (R.M.B.); (D.B.); (E.P.)
| | - Fabio Castagna
- Department of Health Sciences, University of Catanzaro Magna Græcia, 88100 Catanzaro, Italy; (R.B.); (C.L.); (R.M.B.); (D.B.); (E.P.)
- Mediterranean Ethnobotanical Conservatory, Sersale (CZ), 88054 Catanzaro, Italy
| | - Carmine Lupia
- Department of Health Sciences, University of Catanzaro Magna Græcia, 88100 Catanzaro, Italy; (R.B.); (C.L.); (R.M.B.); (D.B.); (E.P.)
- Mediterranean Ethnobotanical Conservatory, Sersale (CZ), 88054 Catanzaro, Italy
| | - Giusi Poerio
- ATS Val Padana, Via dei Toscani, 46100 Mantova, Italy;
| | | | - Renato Lombardi
- IRCCS Casa Sollievo Della Sofferenza, San Giovanni Rotondo (FG), 71013 Foggia, Italy;
| | - Maria Diana Naturale
- Ministry of Health, Directorate General for Health Programming, 00144 Rome, Italy;
| | - Caterina Mercuri
- Department of Experimental and Clinical Medicine, University “Magna Graecia”, 88100 Catanzaro, Italy;
| | - Rosa Maria Bulotta
- Department of Health Sciences, University of Catanzaro Magna Græcia, 88100 Catanzaro, Italy; (R.B.); (C.L.); (R.M.B.); (D.B.); (E.P.)
| | - Domenico Britti
- Department of Health Sciences, University of Catanzaro Magna Græcia, 88100 Catanzaro, Italy; (R.B.); (C.L.); (R.M.B.); (D.B.); (E.P.)
| | - Ernesto Palma
- Department of Health Sciences, University of Catanzaro Magna Græcia, 88100 Catanzaro, Italy; (R.B.); (C.L.); (R.M.B.); (D.B.); (E.P.)
- Center for Pharmacological Research, Food Safety, High Tech and Health (IRC-FSH), University of Catanzaro Magna Græcia, 88100 Catanzaro, Italy
| |
Collapse
|
5
|
Baranchyk Y, Gestels Z, Van den Bossche D, Abdellati S, Britto Xavier B, Manoharan-Basil SS, Kenyon C. Effect of erythromycin residuals in food on the development of resistance in Streptococcus pneumoniae: an in vivo study in Galleria mellonella. PeerJ 2024; 12:e17463. [PMID: 38827315 PMCID: PMC11141549 DOI: 10.7717/peerj.17463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 05/05/2024] [Indexed: 06/04/2024] Open
Abstract
Background The use of antimicrobials to treat food animals may result in antimicrobial residues in foodstuffs of animal origin. The European Medicines Association (EMA) and World Health Organization (WHO) define safe antimicrobial concentrations in food based on acceptable daily intakes (ADIs). It is unknown if ADI doses of antimicrobials in food could influence the antimicrobial susceptibility of human-associated bacteria. Objectives This aim of this study was to evaluate if the consumption of ADI doses of erythromycin could select for erythromycin resistance in a Galleria mellonella model of Streptococcus pneumoniae infection. Methods A chronic model of S. pneumoniae infection in G. mellonella larvae was used for the experiment. Inoculation of larvae with S. pneumoniae was followed by injections of erythromycin ADI doses (0.0875 and 0.012 μg/ml according to EMA and WHO, respectively). Isolation of S. pneumoniae colonies was then performed on selective agar plates. Minimum inhibitory concentrations (MICs) of resistant colonies were measured, and whole genome sequencing (WGS) was performed followed by variant calling to determine the genetic modifications. Results Exposure to single doses of both EMA and WHO ADI doses of erythromycin resulted in the emergence of erythromycin resistance in S. pneumoniae. Emergent resistance to erythromycin was associated with a mutation in rplA, which codes for the L1 ribosomal protein and has been linked to macrolide resistance in previous studies. Conclusion In our in vivo model, even single doses of erythromycin that are classified as acceptable by the WHO and EMA induced significant increases in erythromycin MICs in S. pneumoniae. These results suggest the need to include the induction of antimicrobial resistance (AMR) as a significant criterion for determining ADIs.
Collapse
Affiliation(s)
- Yuliia Baranchyk
- UnivLyon, Université Claude Bernard Lyon 1, Lyon, France
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Zina Gestels
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | | | - Saïd Abdellati
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Basil Britto Xavier
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Hospital Outbreak Support Team-HOST, Ziekenhuis Netwerk Antwerpen Middelheim, Antwerp, Belgium
| | | | - Chris Kenyon
- Institute of Tropical Medicine Antwerp, Antwerp, Belgium
- Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
6
|
Armillei MK, Lomakin IB, Del Rosso JQ, Grada A, Bunick CG. Scientific Rationale and Clinical Basis for Clindamycin Use in the Treatment of Dermatologic Disease. Antibiotics (Basel) 2024; 13:270. [PMID: 38534705 PMCID: PMC10967556 DOI: 10.3390/antibiotics13030270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Revised: 02/27/2024] [Accepted: 03/07/2024] [Indexed: 03/28/2024] Open
Abstract
Clindamycin is a highly effective antibiotic of the lincosamide class. It has been widely used for decades to treat a range of skin and soft tissue infections in dermatology and medicine. Clindamycin is commonly prescribed for acne vulgaris, with current practice standards utilizing fixed-combination topicals containing clindamycin that prevent Cutibacterium acnes growth and reduce inflammation associated with acne lesion formation. Certain clinical presentations of folliculitis, rosacea, staphylococcal infections, and hidradenitis suppurativa are also responsive to clindamycin, demonstrating its suitability and versatility as a treatment option. This review describes the use of clindamycin in dermatological practice, the mechanism of protein synthesis inhibition by clindamycin at the level of the bacterial ribosome, and clindamycin's anti-inflammatory properties with a focus on its ability to ameliorate inflammation in acne. A comparison of the dermatologic indications for similarly utilized antibiotics, like the tetracycline class antibiotics, is also presented. Finally, this review addresses both the trends and mechanisms for clindamycin and antibiotic resistance, as well as the current clinical evidence in support of the continued, targeted use of clindamycin in dermatology.
Collapse
Affiliation(s)
- Maria K. Armillei
- Program in Translational Biomedicine, Yale School of Medicine, Yale University, New Haven, CT 06511, USA;
| | - Ivan B. Lomakin
- Department of Dermatology, Yale University, New Haven, CT 06520, USA;
| | - James Q. Del Rosso
- College of Osteopathic Medicine, Touro University Nevada, Henderson, NV 89014, USA;
- JDR Dermatology Research, Las Vegas, NV 89148, USA
- Clinical Research and Strategic Development, Advanced Dermatology and Cosmetic Surgery, Maitland, FL 32751, USA
| | - Ayman Grada
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA;
| | - Christopher G. Bunick
- Program in Translational Biomedicine, Yale School of Medicine, Yale University, New Haven, CT 06511, USA;
- Department of Dermatology, Yale University, New Haven, CT 06520, USA;
| |
Collapse
|
7
|
El-Banna TES, Sonbol FI, Kamer AMA, Badr SAMM. Genetic diversity of macrolides resistant Staphylococcus aureus clinical isolates and the potential synergistic effect of vitamins, C and K 3. BMC Microbiol 2024; 24:30. [PMID: 38245680 PMCID: PMC10799532 DOI: 10.1186/s12866-023-03169-1] [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: 05/27/2023] [Accepted: 12/22/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Macrolide antibiotics have been extensively used for the treatment of Staphylococcus aureus infections. However, the emergence of macrolide-resistant strains of S. aureus has become a major concern for public health. The molecular mechanisms underlying macrolide resistance in S. aureus are complex and diverse, involving both target site modification and efflux pump systems. In this study, we aim to overcome the molecular diversity of macrolide resistance mechanisms in S. aureus by identifying common molecular targets that could be exploited for the development of novel therapeutics. METHODS About 300 Staphylococcus aureus different isolates were recovered and purified from 921 clinical specimen including urine (88), blood (156), sputum (264), nasal swabs (168), pus (181) and bone (39) collected from different departments in Tanta University Hospital. Macrolide resistant isolates were detected and tested for Multi Drug Resistant (MDR). Gel electrophoresis was performed after the D test and PCR reaction for erm(A), (B), (C), msr(A), and mph(C) genes. Finally, we tried different combinations of Erythromycin or Azithromycin antibiotics with either vitamin K3 or vitamin C. RESULTS Macrolide resistance S. aureus isolates exhibited 7 major resistance patterns according to number of resistance markers and each pattern included sub patterns or subgroups. The PCR amplified products of different erm genes; analysis recorded different phenotypes of the Staphylococcus aureus isolates according to their different genotypes. In addition, our new tested combinations of Erythromycin and vitamin C, Erythromycin, and vitamin K3, Azithromycin and vitamin C and Azithromycin and vitamin K3 showed significant antibacterial effect when using every antibiotic alone. Our findings provide new insights into the molecular mechanisms of macrolide resistance in S. aureus and offer potential strategies for the development of novel protocols to overcome this emerging public health threat.
Collapse
|
8
|
Peela SM, Basu S, Sharma J, AlAsmari AF, AlAsmari F, Alalmaee S, Ramaiah S, Sistla S, Livingstone P, Anbarasu A. Structure Elucidation and Interaction Dynamics of MefA-MsrD Efflux Proteins in Streptococcus pneumoniae: Impact on Macrolide Susceptibility. ACS OMEGA 2023; 8:39454-39467. [PMID: 37901543 PMCID: PMC10601061 DOI: 10.1021/acsomega.3c05210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 09/28/2023] [Indexed: 10/31/2023]
Abstract
Macrolides are empirically used to treat bacterial community-acquired pneumonia (CAP). Streptococcus pneumoniae, being the major pathogen responsible for bacterial CAP with high mortality rates, express MefA-MsrD efflux pumps to hinder macrolide susceptibility. Despite its importance, the structural features of the efflux-protein complex and its impact on macrolide susceptibility have not yet been elucidated explicitly. Therefore, in the present study, combining homology, threading, and dynamics approaches, MefA and MsrD proteins in pathogenic S. pneumoniae were modeled. Both membrane (lipid-bilayer) and cytoplasmic (aqueous) environments were considered to simulate the MefA and MsrD proteins in their ideal cellular conditions followed by dynamics analyses. The simulated MefA structure represented a typical major facilitator superfamily protein structure with 13 transmembrane helices. MefA-MsrD interaction via clustering-based docking revealed low-energy conformers with stable intermolecular interactions. The higher clinical MIC value of azithromycin over erythromycin was reflected upon erythromycin eliciting stronger interactions (dissociation constant or ki = ∼52 μM) with the cytoplasmic ATP-binding MsrD than azithromycin (ki = ∼112 μM). The strong (binding energy = -132.1 ± 9.5 kcal/mol) and highly stable (root-mean-square fluctuation < 1.0 Å) physical association between MefA with MsrD was validated and was found to be unaffected by the antibiotic binding. Higher propensity of the macrolides to interact with MsrD than MefA established the importance of the former in macrolide susceptibility. Ours is probably the first report on the structural arrangements in the MefA-MsrD efflux complex and the macrolide susceptibility in S. pneumoniae. This study provides a novel lead for experimental explorations and efflux-pump inhibitor designs.
Collapse
Affiliation(s)
- Sreeram
Chandra Murthy Peela
- Department
of Microbiology, Jawaharlal Institute of
Postgraduate Medical Education and Research (JIPMER), Puducherry 605006, India
| | - Soumya Basu
- Medical
and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Jyoti Sharma
- Department
of Bioscience and Bioengineering, Indian
Institute of Technology (IIT), Jodhpur342011, Rajasthan, India
| | - Abdullah F. AlAsmari
- Department
of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fawaz AlAsmari
- Department
of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | | | - Sudha Ramaiah
- Department
of Biosciences, Vellore Institute of Technology
(VIT), Vellore 632014, Tamil Nadu, India
| | - Sujatha Sistla
- Department
of Microbiology, Jawaharlal Institute of
Postgraduate Medical Education and Research (JIPMER), Puducherry 605006, India
| | - Paul Livingstone
- Department
of Sports and Health Sciences, Cardiff Metropolitan
University, Cardiff CF5 2YB, U.K.
| | - Anand Anbarasu
- Medical
and Biological Computing Laboratory, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| |
Collapse
|
9
|
Gingras H, Peillard-Fiorente F, Godin C, Patron K, Leprohon P, Ouellette M. New Resistance Mutations Linked to Decreased Susceptibility to Solithromycin in Streptococcus pneumoniae Revealed by Chemogenomic Screens. Antimicrob Agents Chemother 2023; 67:e0039523. [PMID: 37409958 PMCID: PMC10433811 DOI: 10.1128/aac.00395-23] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/18/2023] [Indexed: 07/07/2023] Open
Abstract
Two strains of Streptococcus pneumoniae, one expressing the methyltransferase Erm(B) and the other negative for erm(B), were selected for solithromycin resistance in vitro either with direct drug selection or with chemical mutagenesis followed by drug selection. We obtained a series of mutants that we characterized by next-generation sequencing. We found mutations in various ribosomal proteins (L3, L4, L22, L32, and S4) and in the 23S rRNA. We also found mutations in subunits of the phosphate transporter, in the DEAD box helicase CshB, and in the erm(B)L leader peptide. All mutations were shown to decrease solithromycin susceptibility when transformed into sensitive isolates. Some of the genes derived from our in vitro screens were found to be mutated also in clinical isolates with decreased susceptibility to solithromycin. While many mutations were in coding sequences, some were found in regulatory regions. These included novel phenotypic mutations in the intergenic regions of the macrolide resistance locus mef(E)/mel and in the vicinity of the ribosome binding site of erm(B). Our screens highlighted that macrolide-resistant S. pneumoniae can easily acquire resistance to solithromycin, and they revealed many new phenotypic mutations.
Collapse
Affiliation(s)
- Hélène Gingras
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Flora Peillard-Fiorente
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Chantal Godin
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Kevin Patron
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Philippe Leprohon
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| | - Marc Ouellette
- Centre de Recherche en Infectiologie du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec City, Québec, Canada
| |
Collapse
|
10
|
Rozman V, Mohar Lorbeg P, Treven P, Accetto T, Janežič S, Rupnik M, Bogovič Matijašić B. Genomic insights into antibiotic resistance and mobilome of lactic acid bacteria and bifidobacteria. Life Sci Alliance 2023; 6:e202201637. [PMID: 36781180 PMCID: PMC9930590 DOI: 10.26508/lsa.202201637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 01/30/2023] [Accepted: 01/30/2023] [Indexed: 02/15/2023] Open
Abstract
Lactic acid bacteria (LAB) and Bifidobacterium sp. (bifidobacteria) can carry antimicrobial resistance genes (ARGs), yet data on resistance mechanisms in these bacteria are limited. The aim of our study was to identify the underlying genetic mechanisms of phenotypic resistance in 103 LAB and bifidobacteria using whole-genome sequencing. Sequencing data not only confirmed the presence of 36 acquired ARGs in genomes of 18 strains, but also revealed wide dissemination of intrinsic ARGs. The presence of acquired ARGs on known and novel mobile genetic elements raises the possibility of their horizontal spread. In addition, our data suggest that mutations may be a common mechanism of resistance. Several novel candidate resistance mechanisms were uncovered, providing a basis for further in vitro studies. Overall, 1,314 minimum inhibitory concentrations matched with genotypes in 92.4% of the cases; however, prediction of phenotype based on genotypic data was only partially efficient, especially with respect to aminoglycosides and chloramphenicol. Our study sheds light on resistance mechanisms and their transferability potential in LAB and bifidobacteria, which will be useful for risk assessment analysis.
Collapse
Affiliation(s)
- Vita Rozman
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Institute of Dairy Science and Probiotics, Domžale, Slovenia
| | - Petra Mohar Lorbeg
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Institute of Dairy Science and Probiotics, Domžale, Slovenia
| | - Primož Treven
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Institute of Dairy Science and Probiotics, Domžale, Slovenia
| | - Tomaž Accetto
- University of Ljubljana, Biotechnical Faculty, Department of Microbiology, Chair of Microbial Diversity, Microbiomics and Biotechnology, Ljubljana, Slovenia
| | - Sandra Janežič
- National Laboratory of Health, Environment and Food, Maribor, Slovenia
- University of Maribor, Faculty of Medicine, Maribor, Slovenia
| | - Maja Rupnik
- National Laboratory of Health, Environment and Food, Maribor, Slovenia
- University of Maribor, Faculty of Medicine, Maribor, Slovenia
| | - Bojana Bogovič Matijašić
- University of Ljubljana, Biotechnical Faculty, Department of Animal Science, Institute of Dairy Science and Probiotics, Domžale, Slovenia
| |
Collapse
|
11
|
Brar B, Marwaha S, Poonia AK, Koul B, Kajla S, Rajput VD. Nanotechnology: a contemporary therapeutic approach in combating infections from multidrug-resistant bacteria. Arch Microbiol 2023; 205:62. [PMID: 36629918 DOI: 10.1007/s00203-023-03404-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/24/2022] [Accepted: 01/02/2023] [Indexed: 01/12/2023]
Abstract
In the 20th century, the discovery of antibiotics played an essential role in the fight against infectious diseases, including meningitis, typhoid fever, pneumonia and Mycobacterium tuberculosis. The development of multidrug resistance in microflora due to improper antibiotic use created significant public health issues. Antibiotic resistance has increased at an alarming rate in the past few decades. Multidrug-resistant bacteria (superbugs) such as methicillin-resistant Staphylococcus aureus (MRSA) as well as drug-resistant tuberculosis pose serious health implications. Despite the continuous increase in resistant microbes, the discovery of novel antibiotics is constrained by the cost and complexities of discovery of drugs. The nanotechnology has given new hope in combating this problem. In the present review, recent developments in therapeutics utilizing nanotechnology for novel antimicrobial drug development are discussed. The nanoparticles of silver, gold and zinc oxide have proved to be efficient antimicrobial agents against multidrug-resistant Klebsiella, Pseudomonas, Escherichia Coli and MRSA. Using nanostructures as carriers for antimicrobial agents provides better bioavailability, less chances of sub-therapeutic drug accumulation and less drug-related toxicity. Nanophotothermal therapy using fullerene and antibody functionalized nanostructures are other strategies that can prove to be helpful.
Collapse
Affiliation(s)
- Basanti Brar
- HABITAT, Genome Improvement Primary Producer Company Ltd. Centre of Biofertilizer Production and Technology, HAU, Hisar, 125004, India
| | - Sumnil Marwaha
- ICAR-National Research Centre On Camel, Bikaner, 334001, Rajasthan, India
| | - Anil Kumar Poonia
- Department of Botany, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, Punjab, India. .,Department of Molecular Biology &Biotechnology, CCSHAU, Hisar, 125004, Haryana, India.
| | - Bhupendra Koul
- Department of Botany, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, 144411, Punjab, India.
| | - Subhash Kajla
- Department of Molecular Biology &Biotechnology, CCSHAU, Hisar, 125004, Haryana, India.
| | - Vishnu D Rajput
- Academy of Biology and Biotechnology, Southern Federal University, Rostov-On-Don, 344090, Russia.
| |
Collapse
|
12
|
Wang Y, Taylor SL, Choo JM, Papanicolas LE, Keating R, Hindmarsh K, Thomson RM, Morgan L, Rogers GB, Burr LD. Carriage and Transmission of Macrolide Resistance Genes in Patients With Chronic Respiratory Conditions and Their Close Contacts. Chest 2022; 162:56-65. [DOI: 10.1016/j.chest.2022.01.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 12/17/2021] [Accepted: 01/21/2022] [Indexed: 11/16/2022] Open
|
13
|
Sionov RV, Steinberg D. Targeting the Holy Triangle of Quorum Sensing, Biofilm Formation, and Antibiotic Resistance in Pathogenic Bacteria. Microorganisms 2022; 10:1239. [PMID: 35744757 PMCID: PMC9228545 DOI: 10.3390/microorganisms10061239] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Revised: 06/12/2022] [Accepted: 06/14/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic and recurrent bacterial infections are frequently associated with the formation of biofilms on biotic or abiotic materials that are composed of mono- or multi-species cultures of bacteria/fungi embedded in an extracellular matrix produced by the microorganisms. Biofilm formation is, among others, regulated by quorum sensing (QS) which is an interbacterial communication system usually composed of two-component systems (TCSs) of secreted autoinducer compounds that activate signal transduction pathways through interaction with their respective receptors. Embedded in the biofilms, the bacteria are protected from environmental stress stimuli, and they often show reduced responses to antibiotics, making it difficult to eradicate the bacterial infection. Besides reduced penetration of antibiotics through the intricate structure of the biofilms, the sessile biofilm-embedded bacteria show reduced metabolic activity making them intrinsically less sensitive to antibiotics. Moreover, they frequently express elevated levels of efflux pumps that extrude antibiotics, thereby reducing their intracellular levels. Some efflux pumps are involved in the secretion of QS compounds and biofilm-related materials, besides being important for removing toxic substances from the bacteria. Some efflux pump inhibitors (EPIs) have been shown to both prevent biofilm formation and sensitize the bacteria to antibiotics, suggesting a relationship between these processes. Additionally, QS inhibitors or quenchers may affect antibiotic susceptibility. Thus, targeting elements that regulate QS and biofilm formation might be a promising approach to combat antibiotic-resistant biofilm-related bacterial infections.
Collapse
Affiliation(s)
- Ronit Vogt Sionov
- The Biofilm Research Laboratory, The Institute of Biomedical and Oral Research, The Faculty of Dental Medicine, Hadassah Medical School, The Hebrew University, Jerusalem 9112102, Israel;
| | | |
Collapse
|
14
|
Li G, Lou M, Qi X. A brief overview of classical natural product drug synthesis and bioactivity. Org Chem Front 2022. [DOI: 10.1039/d1qo01341f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This manuscript briefly overviewed the total synthesis and structure–activity relationship studies of eight classical natural products, which emphasizes the important role of total synthesis in natural product-based drug development.
Collapse
Affiliation(s)
- Gen Li
- National Institute of Biological Sciences (NIBS), 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | - Mingliang Lou
- National Institute of Biological Sciences (NIBS), 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
| | - Xiangbing Qi
- National Institute of Biological Sciences (NIBS), 7 Science Park Road ZGC Life Science Park, Beijing 102206, China
- Tsinghua Institute of Multidisciplinary Biomedical Research, Tsinghua University, Beijing 100084, China
| |
Collapse
|
15
|
Varghese R, Daniel JL, Neeravi A, Baskar P, Manoharan A, Sundaram B, Manchanda V, Saigal K, Yesudhasan BL, Veeraraghavan B. Multicentric Analysis of Erythromycin Resistance Determinants in Invasive Streptococcus pneumoniae; Associated Serotypes and Sequence Types in India. Curr Microbiol 2021; 78:3239-3245. [PMID: 34223923 DOI: 10.1007/s00284-021-02594-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 06/25/2021] [Indexed: 11/24/2022]
Abstract
Streptococcus pneumoniae is the major cause of childhood pneumonia and related deaths in India. Widespread use of erythromycin for the treatment of pneumonia has led to the emergence of erythromycin resistance. Despite this increase in erythromycin resistance, there are very little data on resistance determinants from India. Hence, we aimed to perform the molecular characterization of erythromycin-resistant invasive pneumococcal isolates in India. In this study, 250 erythromycin-resistant invasive isolates obtained from four Indian hospitals between 2014 and 2019 were included. The isolates were reconfirmed by standard CDC protocols, followed by detection of erm(B), mef(A/E) genes, and screening for mutations in 23S rRNA, ribosomal proteins L4 and L22. Among the 250 erythromycin-resistant isolates, 46% (n = 114) and 35% (n = 87) carried the mef(A/E) gene and erm(B) gene, respectively; both genes were present in 8% (n = 20) of the isolates and 12% (n = 29) of the studied strains did not bear any of them. The major mutations associated with erythromycin resistance in 23S rRNA, such as A2060C, A2061G, and C2613G, were absent. The predominant serotypes were 19F, 14, 23F, 6A, 6B, 19A, and 9V. The major clonal complexes were CC320, followed by CC230 and CC63. The predominant gene was mef(A/E), and most of the serotypes were PCV13 (54%). This study contributes to the baseline understanding of the erythromycin resistance determinants associated with the serotypes and sequence types (ST) of Indian invasive S. pneumoniae.
Collapse
Affiliation(s)
- Rosemol Varghese
- Department of Clinical Microbiology, Christian Medical College, Vellore, 632004, TN, India
| | - Jones Lionel Daniel
- Department of Clinical Microbiology, Christian Medical College, Vellore, 632004, TN, India
| | - Ayyanraj Neeravi
- Department of Clinical Microbiology, Christian Medical College, Vellore, 632004, TN, India
| | - Pavithra Baskar
- Department of Clinical Microbiology, Christian Medical College, Vellore, 632004, TN, India
| | - Anand Manoharan
- The CHILDS Trust Medical Research Foundation, Chennai, TN, India
| | | | - Vikas Manchanda
- Department of Microbiology, Maulana Azad Medical College, New Delhi, Delhi, India
| | - Karnika Saigal
- Department of Microbiology, Chacha Nehru Bal Chikitsalaya, New Delhi, Delhi, India
| | - Binesh Lal Yesudhasan
- Department of Clinical Microbiology, Christian Medical College, Vellore, 632004, TN, India
| | - Balaji Veeraraghavan
- Department of Clinical Microbiology, Christian Medical College, Vellore, 632004, TN, India.
| |
Collapse
|
16
|
Bacterial Resistance to Antimicrobial Agents. Antibiotics (Basel) 2021; 10:antibiotics10050593. [PMID: 34067579 PMCID: PMC8157006 DOI: 10.3390/antibiotics10050593] [Citation(s) in RCA: 96] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/12/2021] [Accepted: 05/14/2021] [Indexed: 12/26/2022] Open
Abstract
Bacterial pathogens as causative agents of infection constitute an alarming concern in the public health sector. In particular, bacteria with resistance to multiple antimicrobial agents can confound chemotherapeutic efficacy towards infectious diseases. Multidrug-resistant bacteria harbor various molecular and cellular mechanisms for antimicrobial resistance. These antimicrobial resistance mechanisms include active antimicrobial efflux, reduced drug entry into cells of pathogens, enzymatic metabolism of antimicrobial agents to inactive products, biofilm formation, altered drug targets, and protection of antimicrobial targets. These microbial systems represent suitable focuses for investigation to establish the means for their circumvention and to reestablish therapeutic effectiveness. This review briefly summarizes the various antimicrobial resistance mechanisms that are harbored within infectious bacteria.
Collapse
|
17
|
Gingras H, Patron K, Leprohon P, Ouellette M. Azithromycin resistance mutations in Streptococcus pneumoniae as revealed by a chemogenomic screen. Microb Genom 2020; 6. [PMID: 33074087 PMCID: PMC7725334 DOI: 10.1099/mgen.0.000454] [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] [Indexed: 12/02/2022] Open
Abstract
We report on the combination of chemical mutagenesis, azithromycin selection and next-generation sequencing (Mut-Seq) for the identification of small nucleotide variants that decrease the susceptibility of Streptococcus pneumoniae to the macrolide antibiotic azithromycin. Mutations in the 23S ribosomal RNA or in ribosomal proteins can confer resistance to macrolides and these were detected by Mut-Seq. By concentrating on recurrent variants, we could associate mutations in genes implicated in the metabolism of glutamine with decreased azithromycin susceptibility among S. pneumoniae mutants. Glutamine synthetase catalyses the transformation of glutamate and ammonium into glutamine and its chemical inhibition is shown to sensitize S. pneumoniae to antibiotics. A mutation affecting the ribosomal-binding site of a putative ribonuclease J2 is also shown to confer low-level resistance. Mut-Seq has the potential to reveal chromosomal changes enabling high resistance as well as novel events conferring more subtle phenotypes.
Collapse
Affiliation(s)
- Hélène Gingras
- Axe des Maladies Infectieuses et Immunitaires du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Kévin Patron
- Axe des Maladies Infectieuses et Immunitaires du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Philippe Leprohon
- Axe des Maladies Infectieuses et Immunitaires du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec, Canada
| | - Marc Ouellette
- Axe des Maladies Infectieuses et Immunitaires du Centre de Recherche du CHU de Québec and Département de Microbiologie, Infectiologie et Immunologie, Faculté de Médecine, Université Laval, Québec, Québec, Canada
| |
Collapse
|
18
|
Investigation of Macrolide Resistance Genotypes in Mycoplasma bovis Isolates from Canadian Feedlot Cattle. Pathogens 2020; 9:pathogens9080622. [PMID: 32751555 PMCID: PMC7459582 DOI: 10.3390/pathogens9080622] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/27/2020] [Accepted: 07/28/2020] [Indexed: 12/27/2022] Open
Abstract
Mycoplasma bovis is associated with bovine respiratory disease (BRD) and chronic pneumonia and polyarthritis syndrome (CPPS) in feedlot cattle. No efficacious vaccines for M. bovis exist; hence, macrolides are commonly used to control mycoplasmosis. Whole genome sequences of 126 M. bovis isolates, derived from 96 feedlot cattle over 12 production years, were determined. Antimicrobial susceptibility testing (AST) of five macrolides (gamithromycin, tildipirosin, tilmicosin, tulathromycin, tylosin) was conducted using a microbroth dilution method. The AST phenotypes were compared to the genotypes generated for 23S rRNA and the L4 and L22 ribosomal proteins. Mutations in domains II (nucleotide 748; E. coli numbering) and V (nucleotide 2059 and 2060) of the 23S rRNA (rrl) gene alleles were associated with resistance. All isolates with a single mutation at Δ748 were susceptible to tulathromycin, but resistant to tilmicosin and tildipirosin. Isolates with mutations in both domain II and V (Δ748Δ2059 or Δ748Δ2060) were resistant to all five macrolides. However, >99% of isolates were resistant to tildipirosin and tilmicosin, regardless of the number and positions of the mutations. Isolates with a Δ748 mutation in the 23S rRNA gene and mutations in L4 and L22 were resistant to all macrolides except for tulathromycin.
Collapse
|
19
|
Midouni Ayadi B, Mehiri E, Draoui H, Ghariani A, Essalah L, Raoult D, Fournier PE, Slim-Saidi LN. Phenotypic and molecular characterization of macrolide resistance mechanisms among Streptococcus pneumoniae isolated in Tunisia. J Med Microbiol 2020; 69:505-520. [PMID: 32159507 DOI: 10.1099/jmm.0.001151] [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] [Indexed: 01/29/2023] Open
Abstract
Introduction. Streptococcus pneumoniae is responsible for many community infections, with the main ones being pneumonia and meningitis. Pneumococcus has developed increased resistance to multiple classes of antibiotics. The evolution of antibiotic resistance in pneumococcus was influenced by changes in serotype distribution under vaccine selection pressure.Aim. The aim of this study was to determine the genes involved in macrolide resistance, the antimicrobial susceptibility, the serotype distribution and the spread of international antibiotic-resistant clones among clinical isolates of S. pneumoniae.Methodology. We investigated 86 erythromycin-resistant S. pneumoniae strains isolated from respiratory (n=74) or non-respiratory (n=12) samples in Tunisia. Antimicrobial susceptibility was tested using the disk diffusion method. Macrolide-resistant strains were analysed by polymerase chain reaction (PCR) for ermA, ermB, mefA and msrD. We also investigated the macrolide resistance mechanisms in eight isolates (9.3%) by sequencing the L4 and L22 riboprotein-coding genes, plus relevant segments of the three 23S rRNA genes. Capsular serotypes were detected by multiplex PCR. Sequence types (STs) were explored using multilocus sequence typing (MLST).Results. Among the 86 studied strains, 70 (81.4 %) were resistant to penicillin G. The prevalent serotypes were 19F, 14, 19A and 23F. We observed that the cMLSB phenotype (66/86, 76.7%) was the most common in these pneumococci. In addition, ermB was the most frequent resistance gene. No mutation in ribosomal protein L22 or L4 or 23S rRNA was detected. Overall, 44 STs were identified in this study, including 16 that were described for the first time. Resistance to lincomycin, tetracycline and trimethoprim/sulfamethoxazole was observed in 55 (64 %), 34 (39.5 %) and 31 (36 %) isolates, respectively. Furthermore, an increase in fluoroquinolone use in particular may lead to the emergence of levofloxacin-resistant strains. Multidrug resistance was observed in 83 isolates (96.5%). Three global antibiotic-resistant clones were identified: Denmark14 ST230, Portugal19F ST177 and Spain9V ST156.Conclusion. This study shows that macrolide resistance among S. pneumoniae isolated in Tunisia is mainly related to target site modification. Our observations demonstrate a high degree of genetic diversity and capsular types among strains resistant to macrolides.
Collapse
Affiliation(s)
- B Midouni Ayadi
- Aix Marseille University, IRD, SSA, Vitrome, IHU Mediterranee Infection, 19-21 Bd Jean Moulin, 13005 Marseille, France.,Microbiology Laboratory, A. Mami Hospital of Pneumology, UR12/SP18, Ariana, Tunisia.,Faculty of Sciences of Tunis - University of Tunis El Manar, Ariana, Tunisia
| | - E Mehiri
- Microbiology Laboratory, A. Mami Hospital of Pneumology, UR12/SP18, Ariana, Tunisia
| | - H Draoui
- Microbiology Laboratory, A. Mami Hospital of Pneumology, UR12/SP18, Ariana, Tunisia
| | - A Ghariani
- Microbiology Laboratory, A. Mami Hospital of Pneumology, UR12/SP18, Ariana, Tunisia
| | - L Essalah
- Microbiology Laboratory, A. Mami Hospital of Pneumology, UR12/SP18, Ariana, Tunisia
| | - D Raoult
- Aix Marseille University, IRD, Mephi, IHU Mediterranee Infection, 19-21 Bd Jean Moulin, 13005 Marseille, France
| | - P E Fournier
- Aix Marseille University, IRD, SSA, Vitrome, IHU Mediterranee Infection, 19-21 Bd Jean Moulin, 13005 Marseille, France
| | - L N Slim-Saidi
- Microbiology Laboratory, A. Mami Hospital of Pneumology, UR12/SP18, Ariana, Tunisia
| |
Collapse
|
20
|
Yao W, Xu G, Bai B, Wang H, Deng M, Zheng J, Li D, Deng X, Liu X, Lin Z, Chen Z, Li G, Deng Q, Yu Z. In vitro-induced erythromycin resistance facilitates cross-resistance to the novel fluoroketolide, solithromycin, in Staphylococcus aureus. FEMS Microbiol Lett 2019; 365:4992303. [PMID: 29733362 DOI: 10.1093/femsle/fny116] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 05/02/2018] [Indexed: 12/31/2022] Open
Abstract
The aim of this study was to determine whether in vitro induced erythromycin resistance facilitates the cross-resistance to the novel fluoroketolide, solithromycin, in Staphylococcus aureus. Four strains of methicillin-susceptible S. aureus strains S2, S3, S5 and S7 were successfully induced to establish erythromycin-resistant strains by continuous in vitro culture with erythromycin. Mutations at drug binding sites were shown to increase the minimal inhibitory concentrations for ketolides, including telithromycin and the novel compound solithromycin, but did not increase for lincosamides, chloramphenicols or oxazolidinones. In S2-, S5- and S7-derived strains, L22 protein mutations occurred first, resulting in a low level of cross-resistance to ketolides (≤4 μg/mL). The L4 protein mutations were dependent on the L22 protein, resulting in high-level cross-resistance to ketolides (≥8 μg/mL). In S3-derived strains, high levels of cross-resistance occurred concurrently in the 23S rRNA domains II/V and the L22 protein. Hence, long-term exposure of erythromycin results in resistance to ketolides in S. aureus through drug binding site mutations. These results demonstrate that since erythromycin has been used clinically for a long time, it is necessary to carefully evaluate the rewards and risks when prescribing solithromycin for the treatment of infectious diseases.
Collapse
Affiliation(s)
- Weiming Yao
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Guangjian Xu
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Bing Bai
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Hongyan Wang
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Minggui Deng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Jinxin Zheng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, No 130, Dongan road, Xuhui District, Shanghai 200032, China
| | - Duoyun Li
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Xiangbin Deng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Xiaojun Liu
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Zhiwei Lin
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, No 130, Dongan road, Xuhui District, Shanghai 200032, China
| | - Zhong Chen
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Guiqiu Li
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Qiwen Deng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| | - Zhijian Yu
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China.,Quality Control Center of Hospital Infection Management of Shenzhen, No 89, Taoyuan Road, Nanshan District, Shenzhen 518052, China
| |
Collapse
|
21
|
Ruddaraju LK, Pammi SVN, Guntuku GS, Padavala VS, Kolapalli VRM. A review on anti-bacterials to combat resistance: From ancient era of plants and metals to present and future perspectives of green nano technological combinations. Asian J Pharm Sci 2019; 15:42-59. [PMID: 32175017 PMCID: PMC7066045 DOI: 10.1016/j.ajps.2019.03.002] [Citation(s) in RCA: 106] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Revised: 02/03/2019] [Accepted: 03/06/2019] [Indexed: 12/29/2022] Open
Abstract
In the primitive era, humans benefited partially from plants and metals to treat microbial infections. Later these infections were cured with antibiotics but further suffered from resistance issues. In searching of an alternative, researchers developed an adjuvant therapy but were hampered by spreading resistance. Subsequently, nanoparticles (NPs) were proposed to cease the multi-drug resistant bacteria but were hindered due to toxicity issues. Recently, a novel adjuvant therapy employed metals and botanicals into innovative nanotechnology as nano-antibiotics. The combination of green synthesized metallic NPs with antibiotics seems to be a viable platform to combat against MDR bacteria by alleviating resistance and toxicity. This review focuses on the primitive to present era dealings with bacterial resistance mechanisms, newer innovations of nanotechnology and their multiple mechanisms to combat resistance. In addition, special focus is paid on greener NPs as antibiotic carriers, and their future prospects of controlled release and toxicity study.
Collapse
|
22
|
Yao W, Xu G, Li D, Bai B, Wang H, Cheng H, Zheng J, Sun X, Lin Z, Deng Q, Yu Z. Staphylococcus aureus with an erm-mediated constitutive macrolide-lincosamide-streptogramin B resistance phenotype has reduced susceptibility to the new ketolide, solithromycin. BMC Infect Dis 2019; 19:175. [PMID: 30782125 PMCID: PMC6381629 DOI: 10.1186/s12879-019-3779-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Accepted: 02/04/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Solithromycin, the fourth generation of ketolides, has been demonstrated potent antibacterial effect against commonly-isolated gram-positive strains. However, Staphylococcus aureus (S. aureus) strains with a higher solithromycin MIC have already been emerged, the mechanism of which is unknown. METHODS Antimicrobial susceptibility test was performed on 266 strains of S. aureus. The antibiotic resistance phenotype of erm-positive strain was determined by D-zone test. Spontaneous mutation frequency analysis was performed to compare the risk levels for solithromycin resistance among different strains. Efflux pumps and mutational analysis of ribosomal fragments as well as erm(B) gene domains were detected. Quantitative reverse transcription polymerase chain reaction was conducted to compare the transcriptional expression of the erm gene between the constitutive macrolide-lincosamide-streptogramin B (cMLSB)- and inducible MLSB (iMLSB)-phenotypes. RESULTS In the erm-positive S. aureus strains, the minimum inhibitory concentration (MIC)50/90 of solithromycin (2/> 16 mg/L) was significantly higher than that in the erm-negative strains (0.125/0.25 mg/L). Of note, the MIC50 value of the strains with iMLSB (0.25 mg/L) was significantly lower than that of the strains with cMLSB (4 mg/L). A comparison among strains demonstrated that the median mutational frequency in isolates with cMLSB (> 1.2 × 10- 4) was approximately > 57-fold and > 3333-fold higher than that in iMLSB strains (2.1 × 10- 6) and in erythromycin-sensitive strains (3.6 × 10- 8), respectively. The differential antibiotic in vitro activity against strains between cMLSB and iMLSB could not be explained by efflux pump carriers or genetic mutations in the test genes. The expression of the erm genes in strains with cMLSB did not differ from that in strains with iMLSB. CONCLUSIONS The reduced susceptibility to solithromycin by S. aureus was associated with the cMLSB resistance phenotype mediated by erm.
Collapse
Affiliation(s)
- Weiming Yao
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Guangjian Xu
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Duoyun Li
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Bing Bai
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Hongyan Wang
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Hang Cheng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Jinxin Zheng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, No.130, Dongan road, Xuhui District, Shanghai, 200032, China
| | - Xiang Sun
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China
| | - Zhiwei Lin
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.,Key Laboratory of Medical Molecular Virology of Ministries of Education and Health, School of Basic Medical Science and Institutes of Biomedical Sciences, Shanghai Medical College of Fudan University, No.130, Dongan road, Xuhui District, Shanghai, 200032, China
| | - Qiwen Deng
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
| | - Zhijian Yu
- Department of Infectious Diseases and Shenzhen Key Lab for Endogenous Infection, Shenzhen Nanshan Hospital of Shenzhen University, No. 89, Taoyuan Road, Nanshan District, Shenzhen, 518052, China.
| |
Collapse
|
23
|
Zhou P, Chen Y, Lu Q, Qin H, Ou H, He B, Ye J. Cellular metabolism network of Bacillus thuringiensis related to erythromycin stress and degradation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2018; 160:328-341. [PMID: 29857237 DOI: 10.1016/j.ecoenv.2018.05.048] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 05/15/2018] [Accepted: 05/20/2018] [Indexed: 06/08/2023]
Abstract
Erythromycin is one of the most widely used macrolide antibiotics. To present a system-level understanding of erythromycin stress and degradation, proteome, phospholipids and membrane potentials were investigated after the erythromycin degradation. Bacillus thuringiensis could effectively remove 77% and degrade 53% of 1 µM erythromycin within 24 h. The 36 up-regulated and 22 down-regulated proteins were mainly involved in spore germination, chaperone and nucleic acid binding. Up-regulated ribose-phosphate pyrophosphokinase and ribosomal proteins confirmed that the synthesis of protein, DNA and RNA were enhanced after the erythromycin degradation. The reaction network of glycolysis/gluconeogenesis was activated, whereas, the activity of spore germination was decreased. The increased synthesis of phospholipids, especially, palmitoleic acid and oleic acid, altered the membrane permeability for erythromycin transport. Ribose-phosphate pyrophosphokinase and palmitoleic acid could be biomarkers to reflect erythromycin exposure. Lipids, disease, pyruvate metabolism and citrate cycle in human cells could be the target pathways influenced by erythromycin. The findings presented novel insights to the interaction among erythromycin stress, protein interaction and metabolism network, and provided a useful protocol for investigating cellular metabolism responses under pollutant stress.
Collapse
Affiliation(s)
- Pulin Zhou
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Ya Chen
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Qiying Lu
- College of Biology and Food Engineering, Guangdong University of Education, Guangzhou 510303, Guangdong, China
| | - Huaming Qin
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Huase Ou
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Baoyan He
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China
| | - Jinshao Ye
- School of Environment, Guangdong Key Laboratory of Environmental Pollution and Health, Jinan University, Guangzhou 510632, Guangdong, China.
| |
Collapse
|
24
|
Wei B, Kang M. Molecular Basis of Macrolide Resistance in Campylobacter Strains Isolated from Poultry in South Korea. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4526576. [PMID: 30069469 PMCID: PMC6057423 DOI: 10.1155/2018/4526576] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/11/2018] [Accepted: 06/19/2018] [Indexed: 12/31/2022]
Abstract
We investigated the molecular mechanisms underlying macrolide resistance in 38 strains of Campylobacter isolated from poultry. Twenty-seven strains were resistant to azithromycin and erythromycin, five showed intermediate azithromycin resistance and erythromycin susceptibility, and six showed azithromycin resistance and erythromycin susceptibility. Four Campylobacter jejuni and six Campylobacter coli strains had azithromycin MICs which were 8-16 and 2-8-fold greater than those of erythromycin, respectively. The A2075G mutation in the 23S rRNA gene was detected in 11 resistant strains with MICs ranging from 64 to ≥ 512 μg/mL. Mutations including V137A, V137S, and a six-amino acid insertion (114-VAKKAP-115) in ribosomal protein L22 were detected in the C. jejuni strains. Erythromycin ribosome methylase B-erm(B) was not detected in any strain. All strains except three showed increased susceptibility to erythromycin with twofold to 256-fold MIC change in the presence of phenylalanine arginine ß-naphthylamide (PAßN); the effects of PAßN on azithromycin MICs were limited in comparison to those on erythromycin MICs, and 13 strains showed no azithromycin MIC change in the presence of PAßN. Differences between azithromycin and erythromycin resistance and macrolide resistance phenotypes and genotypes were observed even in highly resistant strains. Further studies are required to better understand macrolide resistance in Campylobacter.
Collapse
Affiliation(s)
- Bai Wei
- Department of Veterinary Infectious Diseases and Avian Diseases, College of Veterinary Medicine and Center for Poultry Diseases Control, Chonbuk National University, Jeonju, Republic of Korea
| | - Min Kang
- Department of Veterinary Infectious Diseases and Avian Diseases, College of Veterinary Medicine and Center for Poultry Diseases Control, Chonbuk National University, Jeonju, Republic of Korea
| |
Collapse
|
25
|
Kulczycka-Mierzejewska K, Sadlej J, Trylska J. Molecular dynamics simulations suggest why the A2058G mutation in 23S RNA results in bacterial resistance against clindamycin. J Mol Model 2018; 24:191. [PMID: 29971530 PMCID: PMC6028897 DOI: 10.1007/s00894-018-3689-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 05/22/2018] [Indexed: 01/15/2023]
Abstract
Clindamycin, a lincosamide antibiotic, binds to 23S ribosomal RNA and inhibits protein synthesis. The A2058G mutation in 23S RNA results in bacterial resistance to clindamycin. To understand the influence of this mutation on short-range interactions of clindamycin with 23S RNA, we carried out full-atom molecular dynamics simulations of a ribosome fragment containing clindamycin binding site. We compared the dynamical behavior of this fragment simulated with and without the A2058G mutation. Molecular dynamics simulations suggest that clindamycin in the native ribosomal binding site is more internally flexible than in the A2058G mutant. Only in the native ribosome fragment did we observe intramolecular conformational change of clindamycin around its C7-N1-C10-C11 dihedral. In the mutant, G2058 makes more stable hydrogen bonds with clindamycin hindering its conformational freedom in the ribosome-bound state. Clindamycin binding site is located in the entrance to the tunnel through which the newly synthesized polypeptide leaves the ribosome. We observed that in the native ribosome fragment, clindamycin blocks the passage in the tunnel entrance, whereas in the mutated fragment the aperture is undisturbed due to a different mode of binding of clindamycin in the mutant. Restricted conformational freedom of clindamycin in a position not blocking the tunnel entrance in the A2058G mutant could explain the molecular mechanism of bacterial resistance against clindamycin occurring in this mutant.
Collapse
Affiliation(s)
- Katarzyna Kulczycka-Mierzejewska
- Interdisciplinary Centre for Mathematical and Computational Modelling, University of Warsaw, Pawinskiego 5A, 02-106 Warsaw, Poland
| | - Joanna Sadlej
- Faculty of Chemistry, University of Warsaw, Pasteura 1, 02-093 Warsaw, Poland
| | - Joanna Trylska
- Centre of New Technologies, University of Warsaw, Banacha 2c, 02-093 Warsaw, Poland
| |
Collapse
|
26
|
Wan C, Li Y, Le WJ, Liu YR, Li S, Wang BX, Rice PA, Su XH. Increasing Resistance to Azithromycin in Neisseria gonorrhoeae in Eastern Chinese Cities: Resistance Mechanisms and Genetic Diversity among Isolates from Nanjing. Antimicrob Agents Chemother 2018; 62:e02499-17. [PMID: 29530847 PMCID: PMC5923098 DOI: 10.1128/aac.02499-17] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2017] [Accepted: 02/23/2018] [Indexed: 12/20/2022] Open
Abstract
Azithromycin resistance (AZM-R) of Neisseria gonorrhoeae is emerging as a clinical and public health challenge. We determined molecular characteristics of recent AZM-R Nanjing gonococcal isolates and tracked the emergence of AZM-R isolates in eastern Chinese cities in recent years. A total of 384 N. gonorrhoeae isolates from Nanjing collected from 2013 to 2014 were tested for susceptibility to AZM and six additional antibiotics; all AZM-R strains were characterized genetically for resistance determinants by sequencing and were genotyped using N. gonorrhoeae multiantigen sequence typing (NG-MAST). Among the 384 isolates, 124 (32.3%) were AZM-R. High-level resistance (MIC, ≥256 mg/liter) was present in 10.4% (40/384) of isolates, all of which possessed the A2143G mutation in all four 23S rRNA alleles. Low- to mid-level resistance (MIC, 1 to 64 mg/liter) was present in 21.9% (84/384) of isolates, 59.5% of which possessed the C2599T mutation in all four 23S rRNA alleles. The 124 AZM-R isolates were distributed in 71 different NG-MAST sequence types (STs). ST1866 was the most prevalent type in high-level AZM-R (HL-AZM-R) isolates (45% [18/40]). This study, together with previous reports, revealed that the prevalence of AZM-R in N. gonorrhoeae isolates in certain eastern Chinese cities has risen >4-fold (7% to 32%) from 2008 to 2014. The principal mechanisms of AZM resistance in recent Nanjing isolates were A2143G mutations (high-level resistance) and C2599T mutations (low- to mid-level resistance) in the 23S rRNA alleles. Characterization of NG-MAST STs and phylogenetic analysis indicated the genetic diversity of N. gonorrhoeae in Nanjing; however, ST1866 was the dominant genotype associated with HL-AZM-R isolates.
Collapse
Affiliation(s)
- Chuan Wan
- STD Clinic, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Yang Li
- STD Clinic, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Wen-Jing Le
- STD Clinic, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Yu-Rong Liu
- STD Clinic, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Sai Li
- STD Clinic, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Bao-Xi Wang
- STD Clinic, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| | - Peter A Rice
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Xiao-Hong Su
- STD Clinic, Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, Jiangsu, China
| |
Collapse
|
27
|
Abstract
Natural products have served as powerful therapeutics against pathogenic bacteria since the golden age of antibiotics of the mid-20th century. However, the increasing frequency of antibiotic-resistant infections clearly demonstrates that new antibiotics are critical for modern medicine. Because combinatorial approaches have not yielded effective drugs, we propose that the development of new antibiotics around proven natural scaffolds is the best short-term solution to the rising crisis of antibiotic resistance. We analyze herein synthetic approaches aiming to reengineer natural products into potent antibiotics. Furthermore, we discuss approaches in modulating quorum sensing and biofilm formation as a nonlethal method, as well as narrow-spectrum pathogen-specific antibiotics, which are of interest given new insights into the implications of disrupting the microbiome.
Collapse
Affiliation(s)
- Sean E. Rossiter
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| | - Madison H. Fletcher
- Department of Chemistry, Temple University, 1901 North 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - William M. Wuest
- Department of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
| |
Collapse
|
28
|
Dinos GP. The macrolide antibiotic renaissance. Br J Pharmacol 2017; 174:2967-2983. [PMID: 28664582 DOI: 10.1111/bph.13936] [Citation(s) in RCA: 260] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/29/2017] [Accepted: 06/20/2017] [Indexed: 12/19/2022] Open
Abstract
Macrolides represent a large family of protein synthesis inhibitors of great clinical interest due to their applicability to human medicine. Macrolides are composed of a macrocyclic lactone of different ring sizes, to which one or more deoxy-sugar or amino sugar residues are attached. Macrolides act as antibiotics by binding to bacterial 50S ribosomal subunit and interfering with protein synthesis. The high affinity of macrolides for bacterial ribosomes, together with the highly conserved structure of ribosomes across virtually all of the bacterial species, is consistent with their broad-spectrum activity. Since the discovery of the progenitor macrolide, erythromycin, in 1950, many derivatives have been synthesised, leading to compounds with better bioavailability and acid stability and improved pharmacokinetics. These efforts led to the second generation of macrolides, including well-known members such as azithromycin and clarithromycin. Subsequently, in order to address increasing antibiotic resistance, a third generation of macrolides displaying improved activity against many macrolide resistant strains was developed. However, these improvements were accompanied with serious side effects, leading to disappointment and causing many researchers to stop working on macrolide derivatives, assuming that this procedure had reached the end. In contrast, a recent published breakthrough introduced a new chemical platform for synthesis and discovery of a wide range of diverse macrolide antibiotics. This chemical synthesis revolution, in combination with reduction in the side effects, namely, 'Ketek effects', has led to a macrolide renaissance, increasing the hope for novel and safe therapeutic agents to combat serious human infectious diseases.
Collapse
Affiliation(s)
- George P Dinos
- Department of Biochemistry, School of Medicine, University of Patras, Patras, Greece
| |
Collapse
|
29
|
Wekselman I, Zimmerman E, Davidovich C, Belousoff M, Matzov D, Krupkin M, Rozenberg H, Bashan A, Friedlander G, Kjeldgaard J, Ingmer H, Lindahl L, Zengel JM, Yonath A. The Ribosomal Protein uL22 Modulates the Shape of the Protein Exit Tunnel. Structure 2017; 25:1233-1241.e3. [PMID: 28689968 DOI: 10.1016/j.str.2017.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2016] [Revised: 05/08/2017] [Accepted: 06/02/2017] [Indexed: 10/19/2022]
Abstract
Erythromycin is a clinically useful antibiotic that binds to an rRNA pocket in the ribosomal exit tunnel. Commonly, resistance to erythromycin is acquired by alterations of rRNA nucleotides that interact with the drug. Mutations in the β hairpin of ribosomal protein uL22, which is rather distal to the erythromycin binding site, also generate resistance to the antibiotic. We have determined the crystal structure of the large ribosomal subunit from Deinococcus radiodurans with a three amino acid insertion within the β hairpin of uL22 that renders resistance to erythromycin. The structure reveals a shift of the β hairpin of the mutated uL22 toward the interior of the exit tunnel, triggering a cascade of structural alterations of rRNA nucleotides that propagate to the erythromycin binding pocket. Our findings support recent studies showing that the interactions between uL22 and specific sequences within nascent chains trigger conformational rearrangements in the exit tunnel.
Collapse
Affiliation(s)
- Itai Wekselman
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ella Zimmerman
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Chen Davidovich
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Matthew Belousoff
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Donna Matzov
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Miri Krupkin
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Haim Rozenberg
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Anat Bashan
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Gilgi Friedlander
- The Ilana and Pascal Mantoux Institute for Bioinformatics, The Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Jette Kjeldgaard
- Department of Veterinary Disease Biology, University of Copenhagen, 1870 Frederiksbergc, Denmark
| | - Hanne Ingmer
- Department of Veterinary Disease Biology, University of Copenhagen, 1870 Frederiksbergc, Denmark
| | - Lasse Lindahl
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Janice M Zengel
- Department of Biological Sciences, University of Maryland, Baltimore County, Baltimore, MD 21250, USA
| | - Ada Yonath
- Department of Structural Biology, The Weizmann Institute of Science, Rehovot 7610001, Israel.
| |
Collapse
|
30
|
Pavlova A, Parks JM, Oyelere AK, Gumbart JC. Toward the rational design of macrolide antibiotics to combat resistance. Chem Biol Drug Des 2017; 90:641-652. [DOI: 10.1111/cbdd.13004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/03/2017] [Accepted: 04/08/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Anna Pavlova
- School of Physics Georgia Institute of Technology Atlanta GA USA
| | - Jerry M. Parks
- Biosciences Division Oak Ridge National Laboratory Oak Ridge TN USA
| | - Adegboyega K. Oyelere
- School of Chemistry and Biochemistry Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology Atlanta GA USA
| | - James C. Gumbart
- School of Physics Georgia Institute of Technology Atlanta GA USA
- School of Chemistry and Biochemistry Parker H. Petit Institute for Bioengineering and Bioscience Georgia Institute of Technology Atlanta GA USA
| |
Collapse
|
31
|
Ribosomal Mutations Conferring Macrolide Resistance in Legionella pneumophila. Antimicrob Agents Chemother 2017; 61:AAC.02188-16. [PMID: 28069647 DOI: 10.1128/aac.02188-16] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 01/04/2017] [Indexed: 01/15/2023] Open
Abstract
Monitoring the emergence of antibiotic resistance is a recent issue in the treatment of Legionnaires' disease. Macrolides are recommended as first-line therapy, but resistance mechanisms have not been studied in Legionella species. Our aim was to determine the molecular basis of macrolide resistance in L. pneumophila Twelve independent lineages from a common susceptible L. pneumophila ancestral strain were propagated under conditions of erythromycin or azithromycin pressure to produce high-level macrolide resistance. Whole-genome sequencing was performed on 12 selected clones, and we investigated mutations common to all lineages. We reconstructed the dynamics of mutation for each lineage and demonstrated their involvement in decreased susceptibility to macrolides. The resistant mutants were produced in a limited number of passages to obtain a 4,096-fold increase in erythromycin MICs. Mutations affected highly conserved 5-amino-acid regions of L4 and L22 ribosomal proteins and of domain V of 23S rRNA (G2057, A2058, A2059, and C2611 nucleotides). The early mechanisms mainly affected L4 and L22 proteins and induced a 32-fold increase in the MICs of the selector drug. Additional mutations related to 23S rRNA mostly occurred later and were responsible for a major increase of macrolide MICs, depending on the mutated nucleotide, the substitution, and the number of mutated genes among the three rrl copies. The major mechanisms of the decreased susceptibility to macrolides in L. pneumophila and their dynamics were determined. The results showed that macrolide resistance could be easily selected in L. pneumophila and warrant further investigations in both clinical and environmental settings.
Collapse
|
32
|
In vitro antibacterial activity of α-methoxyimino acylide derivatives against macrolide-resistant pathogens and mutation analysis in 23S rRNA. J Antibiot (Tokyo) 2017; 70:264-271. [DOI: 10.1038/ja.2016.148] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Revised: 10/27/2016] [Accepted: 11/13/2016] [Indexed: 11/08/2022]
|
33
|
Selection of Resistance to Clarithromycin in Mycobacterium abscessus Subspecies. Antimicrob Agents Chemother 2016; 61:AAC.00943-16. [PMID: 27799212 DOI: 10.1128/aac.00943-16] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2016] [Accepted: 10/25/2016] [Indexed: 11/20/2022] Open
Abstract
Mycobacterium abscessus is an emerging pathogen against which clarithromycin is the main drug used. Clinical failures are commonly observed and were first attributed to acquired mutations in rrl encoding 23S rRNA but were then attributed to the intrinsic production of the erm(41) 23S RNA methylase. Since strains of M. abscessus were recently distributed into subspecies and erm(41) sequevars, we investigated acquired clarithromycin resistance mechanisms in mutants selected in vitro from four representative strains. Mutants were sequenced for rrl, erm(41), whiB, rpIV, and rplD and studied for seven antibiotic MICs. For mutants obtained from strain M. abscessus subsp. abscessus erm(41) T28 sequevar and strain M. abscessus subsp. bolletii, which are both known to produce effective methylase, rrl was mutated in only 19% (4/21) and 32.5% (13/40) of mutants, respectively, at position 2058 (A2058C, A2058G) or position 2059 (A2059C, A2059G). No mutations were observed in any of the other genes studied, and resistance to other antibiotics (amikacin, cefoxitin, imipenem, tigecycline, linezolid, and ciprofloxacin) was mainly unchanged. For M. abscessus subsp. abscessus erm(41) C28 sequevar and M. abscessus subsp. massiliense, not producing effective methylase, 100% (26/26) and 97.5% (39/40) of mutants had rrl mutations at position 2058 (A2058C, A2058G, A2058T) or position 2059 (A2059C, A2059G). The remaining M. abscessus subsp. massiliense mutant showed an 18-bp repeat insertion in rpIV, encoding the L22 protein. Our results showed that acquisition of clarithromycin resistance is 100% mediated by structural 50S ribosomal subunit mutations for M. abscessus subsp. abscessus erm(41) C28 and M. abscessus subsp. massiliense, whereas it is less common for M. abscessus subsp. abscessus erm(41) T28 sequevar and M. abscessus subsp. bolletii, where other mechanisms may be responsible for failure.
Collapse
|
34
|
Swedan SF, Hayajneh WA, Bshara GN. Genotyping and serotyping of macrolide and multidrug resistant Streptococcus pneumoniae isolated from carrier children. Indian J Med Microbiol 2016; 34:159-65. [PMID: 27080766 DOI: 10.4103/0255-0857.176840] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
AIMS Streptococcus pneumoniae, an opportunistic pathogen commonly carried asymptomatically in the nasopharynx of children, is associated with increasing rates of treatment failures due to a worldwide increase in drug resistance. We investigated the carriage of S. pneumoniae in children 5 years or younger, the identity of prevalent serotypes, the rates of resistance to macrolides and other antimicrobial agents and the genotypes responsible for macrolide resistance. MATERIALS AND METHODS Nasopharyngeal swabs were collected from 157 children under 5 years for cultural isolation of S. pneumoniae. Antibiogram of isolates was determined using the disk diffusion test, and the minimal inhibitory concentration to macrolides was determined using the E-test. Isolate serotypes and macrolide resistance genes, erm(B) and mef(E), were identified using multiplex polymerase chain reactions. RESULTS S. pneumoniae was recovered from 33.8% of children; 41.9% among males and 21.9% among females (P = 0.009). The highest carriage rate occurred among age groups 7-12 months and 49-60 months. Most frequent serotypes were 19F, 6A/B, 11A, 19A, 14 and 15B/C. Resistance to macrolides was 60.4%. Resistance to oxacillin, trimethoprim/sulfamethoxazole and clindamycin was present among 90.6%, 54.7% and 32.1% of isolates, respectively. All isolates were susceptible to chloramphenicol, levofloxacin and vancomycin. Isolates resistant to one or more macrolide drugs were more likely to be multidrug resistant. Resistance to clindamycin or oxacillin coexisted with macrolide resistance. Among the erythromycin-resistant isolates, erm(B), mef(E) and erm(B) and mef(E) genes were present at rates of 43.8%, 37.5% and 6.3%, respectively. Erm(B) and mef(E) were associated with very high level and moderate-to-high level resistance to macrolides, respectively. CONCLUSION A significant proportion of children harboured macrolide and multidrug-resistant S. pneumoniae.
Collapse
Affiliation(s)
- S F Swedan
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid 22110, Jordan
| | | | | |
Collapse
|
35
|
Gomes C, Martínez-Puchol S, Palma N, Horna G, Ruiz-Roldán L, Pons MJ, Ruiz J. Macrolide resistance mechanisms in Enterobacteriaceae: Focus on azithromycin. Crit Rev Microbiol 2016; 43:1-30. [DOI: 10.3109/1040841x.2015.1136261] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Cláudia Gomes
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic ? Universitat de Barcelona, Spain
| | - Sandra Martínez-Puchol
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic ? Universitat de Barcelona, Spain
| | - Noemí Palma
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic ? Universitat de Barcelona, Spain
| | - Gertrudis Horna
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic ? Universitat de Barcelona, Spain
- Universidad Peruana Cayetano Heredia, Lima, Peru
| | | | - Maria J Pons
- Universidad Peruana de Ciencias Aplicadas, Lima, Peru
| | - Joaquim Ruiz
- ISGlobal, Barcelona Ctr. Int. Health Res. (CRESIB), Hospital Clínic ? Universitat de Barcelona, Spain
| |
Collapse
|
36
|
Fyfe C, Grossman TH, Kerstein K, Sutcliffe J. Resistance to Macrolide Antibiotics in Public Health Pathogens. Cold Spring Harb Perspect Med 2016; 6:a025395. [PMID: 27527699 PMCID: PMC5046686 DOI: 10.1101/cshperspect.a025395] [Citation(s) in RCA: 142] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Macrolide resistance mechanisms can be target-based with a change in a 23S ribosomal RNA (rRNA) residue or a mutation in ribosomal protein L4 or L22 affecting the ribosome's interaction with the antibiotic. Alternatively, mono- or dimethylation of A2058 in domain V of the 23S rRNA by an acquired rRNA methyltransferase, the product of an erm (erythromycin ribosome methylation) gene, can interfere with antibiotic binding. Acquired genes encoding efflux pumps, most predominantly mef(A) + msr(D) in pneumococci/streptococci and msr(A/B) in staphylococci, also mediate resistance. Drug-inactivating mechanisms include phosphorylation of the 2'-hydroxyl of the amino sugar found at position C5 by phosphotransferases and hydrolysis of the macrocyclic lactone by esterases. These acquired genes are regulated by either translation or transcription attenuation, largely because cells are less fit when these genes, especially the rRNA methyltransferases, are highly induced or constitutively expressed. The induction of gene expression is cleverly tied to the mechanism of action of macrolides, relying on antibiotic-bound ribosomes stalled at specific sequences of nascent polypeptides to promote transcription or translation of downstream sequences.
Collapse
Affiliation(s)
- Corey Fyfe
- Tetraphase Pharmaceuticals, Watertown, Massachusetts 02472
| | | | - Kathy Kerstein
- Tetraphase Pharmaceuticals, Watertown, Massachusetts 02472
| | | |
Collapse
|
37
|
Dayao DAE, Seddon JM, Gibson JS, Blackall PJ, Turni C. Whole Genome Sequence Analysis of Pig Respiratory Bacterial Pathogens with Elevated Minimum Inhibitory Concentrations for Macrolides. Microb Drug Resist 2016; 22:531-537. [DOI: 10.1089/mdr.2015.0214] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
| | - Jennifer M. Seddon
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - Justine S. Gibson
- School of Veterinary Science, The University of Queensland, Gatton, Queensland, Australia
| | - Patrick J. Blackall
- Queensland Alliance for Agriculture and Food Innovation, EcoSciences Precinct, The University of Queensland, Dutton Park, Queensland, Australia
| | - Conny Turni
- Queensland Alliance for Agriculture and Food Innovation, EcoSciences Precinct, The University of Queensland, Dutton Park, Queensland, Australia
| |
Collapse
|
38
|
Macrolones Are a Novel Class of Macrolide Antibiotics Active against Key Resistant Respiratory Pathogens In Vitro and In Vivo. Antimicrob Agents Chemother 2016; 60:5337-48. [PMID: 27353268 DOI: 10.1128/aac.00524-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 06/14/2016] [Indexed: 11/20/2022] Open
Abstract
As we face an alarming increase in bacterial resistance to current antibacterial chemotherapeutics, expanding the available therapeutic arsenal in the fight against resistant bacterial pathogens causing respiratory tract infections is of high importance. The antibacterial potency of macrolones, a novel class of macrolide antibiotics, against key respiratory pathogens was evaluated in vitro and in vivo MIC values against Streptococcus pneumoniae, Streptococcus pyogenes, Staphylococcus aureus, and Haemophilus influenzae strains sensitive to macrolide antibiotics and with defined macrolide resistance mechanisms were determined. The propensity of macrolones to induce the expression of inducible erm genes was tested by the triple-disk method and incubation in the presence of subinhibitory concentrations of compounds. In vivo efficacy was assessed in a murine model of S. pneumoniae-induced pneumonia, and pharmacokinetic (PK) profiles in mice were determined. The in vitro antibacterial profiles of macrolones were superior to those of marketed macrolide antibiotics, including the ketolide telithromycin, and the compounds did not induce the expression of inducible erm genes. They acted as typical protein synthesis inhibitors in an Escherichia coli transcription/translation assay. Macrolones were characterized by low to moderate systemic clearance, a large volume of distribution, a long half-life, and low oral bioavailability. They were highly efficacious in a murine model of pneumonia after intraperitoneal application even against an S. pneumoniae strain with constitutive resistance to macrolide-lincosamide-streptogramin B antibiotics. Macrolones are the class of macrolide antibiotics with an outstanding antibacterial profile and reasonable PK parameters resulting in good in vivo efficacy.
Collapse
|
39
|
Hu D, Sun Z, Luo X, Liu S, Yu L, Qu Y, Yang J, Yu J, Li X, Zhang J. Drug Resistance Characteristics and Macrolide-Resistant Mechanisms of Streptococcus pneumoniae in Wenzhou City, China. Med Sci Monit 2016; 22:2731-5. [PMID: 27483416 PMCID: PMC4973796 DOI: 10.12659/msm.896766] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Background Streptococcus pneumoniae (SP) is a Gram-positive, alpha-hemolytic, facultative anaerobic member of the genus Streptococcus. The erythromycin-resistant methylase (erm) gene and macrolide efflux (mef) gene are the 2 main genes that can mediate SP. Transposon (Tn) also plays an important role in the collection and metastasis of the gene. In the present study we investigated the drug resistance characteristics and the macrolide-resistant mechanisms of SP in Wenzhou City, China. Material/Methods Sixty-eight strains of SP were isolated from sputum samples of hospitalized children in the Second Affiliated Hospital of Wenzhou Medical University. These strains were analyzed using antimicrobial susceptibility tests to determine their drug resistance to 10 kinds of antibacterials. Macrolide-resistant phenotypes were identified using K-B method. PCR method was used to analyze the erm B gene, mef A gene, and int Tn gene. Results Drug resistance rates of 68 strains of SP were 98.5%, 100.0%, 63.2%, 52.9%, 94.1%, 89.7%, 0.0%, 0.0%, 16.2%, and 14.7% for clindamycin, erythromycin, penicillin G, cefotaxime, tetracycline, sulfamethoxazole/trimethoprim, levofloxacin, vancomycin, chloramphenicol, and amoxicillin, respectively. Total detection rates of the erm B gene, mef A gene, and int Tn gene were 98.5%, 91.2%, and 100.0%, respectively. Conclusions SP shows significant multi-drug resistance in Wenzhou City, whereas there is no clinical value of macrolides antibiotics for SP. cMLSB mediated by erm B gene is the most predominant phenotype among macrolide-resistant SP. The int Tn gene may play an important role in horizontal transfer and clonal dissemination of SP drug resistance genes in Wenzhou City.
Collapse
Affiliation(s)
- Dakang Hu
- Department of Laboratory Medicine, Taizhou Municipal Hospital, Taizhou, Zhejiang, China (mainland)
| | - Zheng Sun
- Department of Clinical Laboratory, Shengli Oil Field Shengli Hospital, Dongying, Shandong, China (mainland)
| | - Xinhua Luo
- Department of Laboratory Medicine, Taizhou Municipal Hospital, Taizhou, Zhejiang, China (mainland)
| | - Shuangchun Liu
- Department of Blood Transfusion, Taizhou Municipal Hospital, Taizhou, Zhejiang, China (mainland)
| | - Lianhua Yu
- Department of Laboratory Medicine, Taizhou Municipal Hospital, Taizhou, Zhejiang, China (mainland)
| | - Ying Qu
- Department of Laboratory Medicine, Taizhou Municipal Hospital, Taizhou, Zhejiang, China (mainland)
| | - Jinhong Yang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Jian Yu
- Department of Laboratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Xiangyang Li
- Department of Laboratory Medicine, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China (mainland)
| | - Jin Zhang
- Department of Laboratory Medicine, Taizhou Municipal Hospital, Taizhou, Zhejiang, China (mainland)
| |
Collapse
|
40
|
Belkacem A, Jacquier H, Goubard A, Mougari F, La Ruche G, Patey O, Micaëlo M, Semaille C, Cambau E, Bercot B. Molecular epidemiology and mechanisms of resistance of azithromycin-resistant Neisseria gonorrhoeae isolated in France during 2013-14. J Antimicrob Chemother 2016; 71:2471-8. [PMID: 27301565 DOI: 10.1093/jac/dkw182] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 04/21/2016] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVES The objective of this study was to determine the prevalence and mechanisms of azithromycin resistance of Neisseria gonorrhoeae French isolates from 2013 to 2014. METHODS N. gonorrhoeae samples isolated in a network of laboratories were tested for susceptibility to azithromycin between April 2013 and March 2014. Fifty-four isolates that were non-susceptible to azithromycin and 18 susceptible isolates were characterized for molecular mechanisms of resistance by PCR/sequencing and genotyped using N. gonorrhoeae multiantigen sequence typing (NG-MAST). RESULTS Among the 970 N. gonorrhoeae isolates, 54 (5.56%) were non-susceptible to azithromycin, 9 (1%) were resistant and 45 (4.6%) showed intermediate resistance. Azithromycin-non-susceptible isolates harboured a C2599T mutation in the rrl gene encoding the 23S rRNA alleles (5.5%), a C substitution in the mtrR promoter (5.5%), an A deletion in the mtrR promoter (53.7%) and mutations in the L4 ribosomal protein (14.8%) and in the MtrR repressor (25.9%). No isolates showed an L22 mutation or carried an erm, ere, mef(A)/(E) or mphA gene. Thirty different STs were highlighted using the NG-MAST technique. The predominant genogroups non-susceptible to azithromycin were G21 (31%), G1407 (20%) and G2400 (15%). Genogroup G2400 (15%) was revealed to be a novel cluster prevalent in the south of France and resistant to azithromycin, ciprofloxacin and tetracycline. CONCLUSIONS Our study highlights that the prevalence of resistance of N. gonorrhoeae to azithromycin in France is low and essentially due to multiple genetic mutations. Its dissemination occurs through three major genogroups including a novel one in France (G2400).
Collapse
Affiliation(s)
- Anna Belkacem
- APHP, Lariboisière-Fernand Widal Hospital, Laboratory of Bacteriology and Associated Laboratory for the National Reference Centre for Gonococci, F-75010 Paris, France Department of Tropical and Infectious Diseases, CHI, Villeneuve Saint Georges, France
| | - Hervé Jacquier
- APHP, Lariboisière-Fernand Widal Hospital, Laboratory of Bacteriology and Associated Laboratory for the National Reference Centre for Gonococci, F-75010 Paris, France INSERM, IAME, UMR 1137, F-75018 Paris, France University Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France
| | - Agathe Goubard
- Alfred Fournier Institute, National Reference Centre for Gonococci, F-75014 Paris, France
| | - Faiza Mougari
- APHP, Lariboisière-Fernand Widal Hospital, Laboratory of Bacteriology and Associated Laboratory for the National Reference Centre for Gonococci, F-75010 Paris, France INSERM, IAME, UMR 1137, F-75018 Paris, France University Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France
| | - Guy La Ruche
- French Institute for Public Health Surveillance (InVS), Department of Infectious Diseases, Saint-Maurice, France
| | - Olivier Patey
- Department of Tropical and Infectious Diseases, CHI, Villeneuve Saint Georges, France
| | - Maïté Micaëlo
- APHP, Lariboisière-Fernand Widal Hospital, Laboratory of Bacteriology and Associated Laboratory for the National Reference Centre for Gonococci, F-75010 Paris, France INSERM, IAME, UMR 1137, F-75018 Paris, France University Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France
| | - Caroline Semaille
- French Institute for Public Health Surveillance (InVS), Department of Infectious Diseases, Saint-Maurice, France
| | - Emmanuelle Cambau
- APHP, Lariboisière-Fernand Widal Hospital, Laboratory of Bacteriology and Associated Laboratory for the National Reference Centre for Gonococci, F-75010 Paris, France INSERM, IAME, UMR 1137, F-75018 Paris, France University Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France
| | - Béatrice Bercot
- APHP, Lariboisière-Fernand Widal Hospital, Laboratory of Bacteriology and Associated Laboratory for the National Reference Centre for Gonococci, F-75010 Paris, France INSERM, IAME, UMR 1137, F-75018 Paris, France University Paris Diderot, IAME, UMR 1137, Sorbonne Paris Cité, F-75018 Paris, France
| |
Collapse
|
41
|
Discovery of Novel MLSB Resistance Methylase Genes and Their Associated Genetic Elements in Staphylococci. CURRENT CLINICAL MICROBIOLOGY REPORTS 2016. [DOI: 10.1007/s40588-016-0030-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
42
|
Eyal Z, Matzov D, Krupkin M, Wekselman I, Paukner S, Zimmerman E, Rozenberg H, Bashan A, Yonath A. Structural insights into species-specific features of the ribosome from the pathogen Staphylococcus aureus. Proc Natl Acad Sci U S A 2015; 112:E5805-14. [PMID: 26464510 PMCID: PMC4629319 DOI: 10.1073/pnas.1517952112] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The emergence of bacterial multidrug resistance to antibiotics threatens to cause regression to the preantibiotic era. Here we present the crystal structure of the large ribosomal subunit from Staphylococcus aureus, a versatile Gram-positive aggressive pathogen, and its complexes with the known antibiotics linezolid and telithromycin, as well as with a new, highly potent pleuromutilin derivative, BC-3205. These crystal structures shed light on specific structural motifs of the S. aureus ribosome and the binding modes of the aforementioned antibiotics. Moreover, by analyzing the ribosome structure and comparing it with those of nonpathogenic bacterial models, we identified some unique internal and peripheral structural motifs that may be potential candidates for improving known antibiotics and for use in the design of selective antibiotic drugs against S. aureus.
Collapse
Affiliation(s)
- Zohar Eyal
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Donna Matzov
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Miri Krupkin
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Itai Wekselman
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | | | - Ella Zimmerman
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Haim Rozenberg
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Anat Bashan
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| | - Ada Yonath
- Department of Structural Biology, Weizmann Institute of Science, Rehovot 7610001, Israel
| |
Collapse
|
43
|
Wu L, Wu H, Chen L, Yu X, Borriss R, Gao X. Difficidin and bacilysin from Bacillus amyloliquefaciens FZB42 have antibacterial activity against Xanthomonas oryzae rice pathogens. Sci Rep 2015; 5:12975. [PMID: 26268540 PMCID: PMC4534799 DOI: 10.1038/srep12975] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Accepted: 07/15/2015] [Indexed: 12/28/2022] Open
Abstract
Bacterial blight and bacterial leaf streak are serious, economically damaging, diseases of rice caused by the bacteria Xanthomonas oryzae pv. oryzae and X. oryzae pv. oryzicola. Bacillus amyloliquefaciens FZB42 was shown to possess biocontrol activity against these Xanthomonas strains by producing the antibiotic compounds difficidin and bacilysin. Analyses using fluorescence, scanning electron and transmission electron microscopy revealed difficidin and bacilysin caused changes in the cell wall and structure of Xanthomonas. Biological control experiments on rice plants demonstrated the ability of difficidin and bacilysin to suppress disease. Difficidin and bacilysin caused downregulated expression of genes involved in Xanthomonas virulence, cell division, and protein and cell wall synthesis. Taken together, our results highlight the potential of B. amyloliquefaciens FZB42 as a biocontrol agent against bacterial diseases of rice, and the utility of difficidin and bacilysin as antimicrobial compounds.
Collapse
Affiliation(s)
- Liming Wu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Key Laboratory of Monitoring and Management of Crop Diseases and Pest Insects, Ministry of Education, Nanjing, China
| | - Huijun Wu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Key Laboratory of Monitoring and Management of Crop Diseases and Pest Insects, Ministry of Education, Nanjing, China
| | - Lina Chen
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Key Laboratory of Monitoring and Management of Crop Diseases and Pest Insects, Ministry of Education, Nanjing, China
| | - Xinfang Yu
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Key Laboratory of Monitoring and Management of Crop Diseases and Pest Insects, Ministry of Education, Nanjing, China
| | | | - Xuewen Gao
- Department of Plant Pathology, College of Plant Protection, Nanjing Agricultural University, Key Laboratory of Monitoring and Management of Crop Diseases and Pest Insects, Ministry of Education, Nanjing, China
| |
Collapse
|
44
|
Rodriguez de Evgrafov M, Gumpert H, Munck C, Thomsen TT, Sommer MOA. Collateral Resistance and Sensitivity Modulate Evolution of High-Level Resistance to Drug Combination Treatment in Staphylococcus aureus. Mol Biol Evol 2015; 32:1175-85. [PMID: 25618457 DOI: 10.1093/molbev/msv006] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
As drug-resistant pathogens continue to emerge, combination therapy will increasingly be relied upon to treat infections and to help combat further development of multidrug resistance. At present a dichotomy exists between clinical practice, which favors therapeutically synergistic combinations, and the scientific model emerging from in vitro experimental work, which maintains that this interaction provides greater selective pressure toward resistance development than other interaction types. We sought to extend the current paradigm, based on work below or near minimum inhibitory concentration levels, to reflect drug concentrations more likely to be encountered during treatment. We performed a series of adaptive evolution experiments using Staphylococcus aureus. Interestingly, no relationship between drug interaction type and resistance evolution was found as resistance increased significantly beyond wild-type levels. All drug combinations, irrespective of interaction types, effectively limited resistance evolution compared with monotreatment. Cross-resistance and collateral sensitivity were found to be important factors in the extent of resistance evolution toward a combination. Comparative genomic analyses revealed that resistance to drug combinations was mediated largely by mutations in the same genes as single-drug-evolved lineages highlighting the importance of the component drugs in determining the rate of resistance evolution. Results of this work suggest that the mechanisms of resistance to constituent drugs should be the focus of future resistance evolution work.
Collapse
Affiliation(s)
| | - Heidi Gumpert
- Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Christian Munck
- Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Thomas T Thomsen
- Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark
| | - Morten O A Sommer
- Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Hørsholm, Denmark
| |
Collapse
|
45
|
Ou G, Liu Y, Tang Y, You X, Zeng Y, Xiao J, Chen L, Yu M, Wang M, Zhu C. In vitro subminimum inhibitory concentrations of macrolide antibiotics induce macrolide resistance in Mycoplasma pneumoniae. Hippokratia 2015; 19:57-62. [PMID: 26435649 PMCID: PMC4574589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
AIM This study aims to investigate the inducing effect of subminimum inhibitory concentrations of macrolide antibiotics on Mycoplasma pneumoniae (M. pneumoniae) resistance to drugs. MATERIALS AND METHODS One M. pneumoniae reference strain M129 (ATCC 29342) and 104 clinical isolates were incubated at 37C for 6-8 days. Genomic DNA of M. pneumoniae was extracted using TIANamp Bacteria DNA kit and amplified by polymerase chain reaction (PCR). RESULTS Ten sensitive isolates obtained from 104 M. pneumoniae clinical isolates were induced by subminimum inhibitory concentrations of macrolide antibiotics. Among them, three were found to possess mutations in L4 and L22 ribosomal proteins. Two cases carried simultaneously the C162A and A430G mutations of L4 and the T279C mutation of L22. In addition, one case had only the A209T mutation of L4. CONCLUSIONS Repeated in vitro exposure to subminimum inhibitory concentrations of macrolide antibiotics could induce selective mutations in ribosomal genes of M. pneumoniae clinical isolates that cause resistance to macrolide antibiotics. Hippokratia 2015, 19 (1): 57-62.
Collapse
Affiliation(s)
- G Ou
- Institution of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key, Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, P.R. China
| | - Y Liu
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Y Tang
- Shaoyang Medical College, Shaoyang City, Hunan Province, P.R. China
| | - X You
- Institution of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key, Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, P.R. China
| | - Y Zeng
- Institution of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key, Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, P.R. China
| | - J Xiao
- Institution of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key, Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, P.R. China
| | - L Chen
- Institution of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key, Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, P.R. China
| | - M Yu
- Institution of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key, Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, P.R. China
| | - M Wang
- Institute of Antibiotics, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - C Zhu
- Institution of Pathogenic Biology, Medical College, University of South China, Hunan Provincial Key, Laboratory for Special Pathogens Prevention and Control, Hengyang City, Hunan Province, P.R. China
| |
Collapse
|
46
|
Olsen AS, Warrass R, Douthwaite S. Macrolide resistance conferred by rRNA mutations in field isolates of Mannheimia haemolytica and Pasteurella multocida. J Antimicrob Chemother 2014; 70:420-3. [PMID: 25261417 DOI: 10.1093/jac/dku385] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES To determine how resistance to macrolides is conferred in field isolates of Pasteurella multocida and Mannheimia haemolytica that lack previously identified resistance determinants for rRNA methylation, efflux and macrolide-modifying enzymes. METHODS Isolates of P. multocida and M. haemolytica identified as being highly resistant (MICs >64 mg/L) to the macrolides erythromycin, gamithromycin, tilmicosin, tildipirosin and tulathromycin were screened by multiplex PCR for the previously identified resistance genes erm(42), msr(E) and mph(E). Strains lacking these determinants were analysed by genome sequencing and primer extension on the rRNAs. RESULTS Macrolide resistance in one M. haemolytica isolate was conferred by the 23S rRNA mutation A2058G; resistance in three P. multocida isolates were caused by mutations at the neighbouring nucleotide A2059G. In each strain, all six copies of the rrn operons encoded the respective mutations. There were no mutations in the ribosomal protein genes rplD or rplV, and no other macrolide resistance mechanism was evident. CONCLUSIONS High-level macrolide resistance can arise from 23S rRNA mutations in P. multocida and M. haemolytica despite their multiple copies of rrn. Selective pressures from exposure to different macrolide or lincosamide drugs presumably resulted in consolidation of either the A2058G or the A2059G mutation.
Collapse
Affiliation(s)
- Anders S Olsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| | - Ralf Warrass
- MSD Animal Health Innovation GmbH, Zur Propstei, D-55270 Schwabenheim, Germany
| | - Stephen Douthwaite
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, DK-5230 Odense M, Denmark
| |
Collapse
|
47
|
Fair RJ, Tor Y. Antibiotics and bacterial resistance in the 21st century. PERSPECTIVES IN MEDICINAL CHEMISTRY 2014; 6:25-64. [PMID: 25232278 PMCID: PMC4159373 DOI: 10.4137/pmc.s14459] [Citation(s) in RCA: 912] [Impact Index Per Article: 82.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Revised: 06/24/2014] [Accepted: 06/24/2014] [Indexed: 12/11/2022]
Abstract
Dangerous, antibiotic resistant bacteria have been observed with increasing frequency over the past several decades. In this review the factors that have been linked to this phenomenon are addressed. Profiles of bacterial species that are deemed to be particularly concerning at the present time are illustrated. Factors including economic impact, intrinsic and acquired drug resistance, morbidity and mortality rates, and means of infection are taken into account. Synchronously with the waxing of bacterial resistance there has been waning antibiotic development. The approaches that scientists are employing in the pursuit of new antibacterial agents are briefly described. The standings of established antibiotic classes as well as potentially emerging classes are assessed with an emphasis on molecules that have been clinically approved or are in advanced stages of development. Historical perspectives, mechanisms of action and resistance, spectrum of activity, and preeminent members of each class are discussed.
Collapse
Affiliation(s)
- Richard J Fair
- Department for Biomolecular Systems, Max Planck Institute of Colloids and Interfaces, Berlin, Germany
| | - Yitzhak Tor
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA, USA
| |
Collapse
|
48
|
Komiya K, Kurashima A, Ihi T, Nagai H, Matsumoto N, Mizunoe S, Ishii H, Takahashi O, Ohta K, Kudoh S, Kadota JI. Long-term, low-dose erythromycin monotherapy for Mycobacterium avium complex lung disease: a propensity score analysis. Int J Antimicrob Agents 2014; 44:131-5. [PMID: 24948577 DOI: 10.1016/j.ijantimicag.2014.03.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Revised: 02/13/2014] [Accepted: 03/30/2014] [Indexed: 10/25/2022]
Abstract
Multidrug regimens are initially withheld in mild cases of pulmonary Mycobacterium avium complex (MAC) disease. Based on the anti-inflammatory effects of macrolides, some patients are treated with erythromycin, which does not appear to exhibit cross-resistance with clarithromycin in MAC. The aim of this study was to evaluate the effects and adverse events of erythromycin monotherapy in patients with pulmonary MAC disease. This was a retrospective propensity score analysis consisting of 31 patients treated with erythromycin alone and 72 patients on conservative therapy, all of whom met the ATS/IDSA criteria for pulmonary MAC disease. The primary outcome was exacerbation requiring administration of a multidrug regimen. The secondary outcome was the rate of response to the multidrug regimens after exacerbation as a surrogate variable for cross-resistance to clarithromycin. As a result, erythromycin monotherapy was found to be likely to suppress exacerbation throughout the 7-year observation period after the diagnosis of pulmonary MAC disease (P=0.045, Breslow test). Multivariate analysis showed that erythromycin tended to prevent exacerbation, albeit statistically insignificantly (hazard ratio=0.495, 95% confidence interval 0.198-1.235; P=0.132). In addition, the rate of response to the multidrug regimens after exacerbation in the erythromycin group (56%; 5/9) was similar to that observed in the control group (62%; 13/21) (P=0.528). Erythromycin monotherapy for patients with pulmonary MAC disease may have the potential to suppress exacerbation without inducing cross-resistance to clarithromycin. However, further prospective studies are needed to microbiologically verify the effectiveness and potential for cross-resistance of these drugs.
Collapse
Affiliation(s)
- Kosaku Komiya
- Respiratory Medicine and Infectious Diseases, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan; Clinical Research Center of Respiratory Medicine, Tenshindo Hetsugi Hospital, 5956 Nihongi, Nakahetsugi, Oita 879-7761, Japan.
| | - Atsuyuki Kurashima
- Respiratory Medicine, Fukujuji Hospital, 1-24 Matsuyama, Kiyose, Tokyo 204-0022, Japan; Center of Respiratory Medicine, National Hospital Organization Tokyo Hospital, 3-1-1 Takeoka, Kiyose, Tokyo 204-0023, Japan
| | - Toshihiko Ihi
- Respiratory Medicine, National Hospital Organization Miyazaki Higashi Hospital, 4374-1, Tayoshi, Miyazaki 880-0911, Japan
| | - Hideaki Nagai
- Center of Respiratory Medicine, National Hospital Organization Tokyo Hospital, 3-1-1 Takeoka, Kiyose, Tokyo 204-0023, Japan
| | - Nobuhiro Matsumoto
- Respirology, Department of Internal Medicine, Faculty of Medicine, University of Miyazaki, 5200 Kihara, Kiyotake, Miyazaki 889-1692, Japan
| | - Shunji Mizunoe
- Respiratory Medicine, Oita Prefectural Hospital, 476 Bunyo, Oita 870-8511, Japan
| | - Hiroshi Ishii
- Respiratory Medicine and Infectious Diseases, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| | - Osamu Takahashi
- Center for Clinical Epidemiology, St Luke's Life Science Institute, 10-1 Akashi-machi, Chuo-ku, Tokyo 104-0044, Japan
| | - Ken Ohta
- Center of Respiratory Medicine, National Hospital Organization Tokyo Hospital, 3-1-1 Takeoka, Kiyose, Tokyo 204-0023, Japan
| | - Shoji Kudoh
- Respiratory Medicine, Fukujuji Hospital, 1-24 Matsuyama, Kiyose, Tokyo 204-0022, Japan
| | - Jun-Ichi Kadota
- Respiratory Medicine and Infectious Diseases, Oita University Faculty of Medicine, 1-1 Idaigaoka, Hasama, Yufu, Oita 879-5593, Japan
| |
Collapse
|
49
|
Hays C, Lienhard R, Auzou M, Barraud O, Guérin F, Ploy MC, Cattoir V. Erm(X)-mediated resistance to macrolides, lincosamides and streptogramins in Actinobaculum schaalii. J Antimicrob Chemother 2014; 69:2056-60. [PMID: 24710027 DOI: 10.1093/jac/dku099] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
OBJECTIVES Actinobaculum schaalii is a Gram-positive bacillus increasingly reported as a causative agent of urinary tract infections as well as invasive infections, mainly in the elderly and patients with underlying urological conditions. Since little is known about the molecular basis of antimicrobial resistance in A. schaalii, the aim of this study was to investigate resistance to macrolides, lincosamides and streptogramins (MLS) in this emerging pathogen. METHODS A total of 32 A. schaalii clinical isolates from France and Switzerland were studied. MICs of erythromycin, spiramycin, lincomycin, clindamycin and quinupristin/dalfopristin were determined by the agar dilution method. Resistance genes erm(A), erm(B), erm(C), erm(F), erm(G), erm(X), msr(A) and mef(A) were screened by PCR. The genetic environment was determined by random cloning and PCR mapping. RESULTS Out of 32 isolates tested, 21 were highly resistant to erythromycin, spiramycin, lincomycin and clindamycin (MICs >256 mg/L), whereas 11 exhibited low MICs (MICs < 0.12 mg/L). On the other hand, quinupristin/dalfopristin remained active against all the isolates. An inducible MLSB resistance phenotype was noted in all cases. The erm(X) gene was detected among all resistant strains, whereas none was detected in susceptible strains. Analysis of genetic support and environment revealed that erm(X) was probably part of the chromosome of A. schaalii. CONCLUSIONS This study is the first molecular characterization of MLS resistance in A. schaalii. In all cases, it was due to the presence of erm(X), a methylase gene previously identified in other clinically relevant Gram-positive bacilli.
Collapse
Affiliation(s)
| | | | - Michel Auzou
- CHU de Caen, Service de Microbiologie, Caen, France
| | - Olivier Barraud
- CHU de Limoges, Service de Bactériologie-Virologie-Hygiène, Limoges, France INSERM U1092, Limoges, France Université de Limoges, UMR-S1092, Limoges, France
| | - François Guérin
- CHU de Caen, Service de Microbiologie, Caen, France Université de Caen Basse-Normandie, EA4655 (équipe 'Antibio-résistance'), Caen, France
| | - Marie-Cécile Ploy
- CHU de Limoges, Service de Bactériologie-Virologie-Hygiène, Limoges, France INSERM U1092, Limoges, France Université de Limoges, UMR-S1092, Limoges, France
| | - Vincent Cattoir
- CHU de Caen, Service de Microbiologie, Caen, France Université de Caen Basse-Normandie, EA4655 (équipe 'Antibio-résistance'), Caen, France
| |
Collapse
|
50
|
Schito GC, Marchese A, Elkharrat D, Farrell DJ. Comparative Activity of Telithromycin Against Macrolide-Resistant Isolates ofStreptococcus pneumoniae:Results of Two Years of the PROTEKT Surveillance Study. J Chemother 2013; 16:13-22. [PMID: 15077994 DOI: 10.1179/joc.2004.16.1.13] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
UNLABELLED The increase in resistance to macrolides has been linked with increasing use of these agents as empirical therapy for community-acquired respiratory tract infections (CARTIs). As part of the ongoing PROTEKT (Prospective Resistant Organism Tracking and Epidemiology for the Ketolide Telithromycin) surveillance program, over 7600 Streptococcus pneumoniae isolates were collected worldwide from 1999-2001 and evaluated for macrolide resistance. Globally, macrolide resistance was 31-33%, with considerable inter-country variation (<10-88%) and particularly high prevalence in the Far East (>71%). In Europe, France had the most resistant isolates (>53%). The highest rates of macrolide resistance were seen in 0-2 year olds. Co-resistance to clindamycin (64%) and all beta-lactams (14-79%) was seen among macrolide-resistant isolates, but >99% of these isolates were susceptible to telithromycin, vancomycin and linezolid. There was considerable variation in the prevalence of erm(B) (56-64%) and mef(A) (30-35%): erm(B) was prevalent in Europe and mef(A) in North America. Globally, 5-7% isolates carried both mechanisms (erm(B)+mef(A)); of these, 47-65% were from South Korea. These double resistance isolates were >90% resistant to the beta-lactams, except amoxicillin-clavulanate. Clindamycin was active against >98% mef(A) but poorly active against erm(B) and erm(B)+mef(A) isolates. Telithromycin, vancomycin and linezolid were highly active (>99.5%) across all three genotypes. CONCLUSIONS In vitro, telithromycin, vancomycin and linezolid are highly active against antibiotic-resistant strains of S. pneumoniae. Telithromycin may be a useful therapeutic alternative to macrolides for the treatment of CARTIs.
Collapse
|