1
|
Hartlage W, Imlay H, Spivak ES. The role of empiric atypical antibiotic coverage in non-severe community-acquired pneumonia. ANTIMICROBIAL STEWARDSHIP & HEALTHCARE EPIDEMIOLOGY : ASHE 2024; 4:e214. [PMID: 39758879 PMCID: PMC11696604 DOI: 10.1017/ash.2024.453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/16/2024] [Accepted: 09/17/2024] [Indexed: 01/07/2025]
Abstract
A β-lactam plus a macrolide or a respiratory fluoroquinolone alone is recommended as standard empiric antibacterial therapy for non-severe adults hospitalized with community-acquired pneumonia (CAP) per Infectious Diseases Society of America guidelines. However, the evidence in support of adding empiric atypical antibacterial therapy, and specifically the addition of a macrolide, is conflicting and should be balanced with additional factors: the necessity of covering atypical organisms, benefits of macrolide-associated immunomodulation, harms associated with antibiotic use, and selection for antibiotic-resistant organisms. In this review, we examine the role of atypical coverage in standard treatment regimens for patients admitted with non-severe CAP and specifically focus on the addition of macrolides to β-lactams. We conclude that a subset of patients should not be given atypical coverage as part of their regimen.
Collapse
Affiliation(s)
- Whitney Hartlage
- Division of Infectious Diseases, Veteran’s Affairs Salt Lake City Health Care System, Salt Lake City, UT, USA
| | - Hannah Imlay
- Division of Infectious Diseases, Veteran’s Affairs Salt Lake City Health Care System, Salt Lake City, UT, USA
- Division of Infectious Diseases, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| | - Emily S. Spivak
- Division of Infectious Diseases, Veteran’s Affairs Salt Lake City Health Care System, Salt Lake City, UT, USA
- Division of Infectious Diseases, Department of Internal Medicine, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
2
|
Yu P, Guo X, Wang W, Wang L, Zhang H, Deng L, Yang H, He T, Wu P, Zhang Y. Distribution and driving mechanisms of antibiotic resistance genes in urbanized watersheds. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 954:176387. [PMID: 39317254 DOI: 10.1016/j.scitotenv.2024.176387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/12/2024] [Accepted: 09/17/2024] [Indexed: 09/26/2024]
Abstract
Antibiotic resistance genes (ARGs) have emerged as a global concern, posing significant threats to human health and safety. Understanding the contamination levels and driving mechanisms behind ARG proliferation is urgently needed. Urban watersheds, influenced by human activities, serve as critical reservoirs for ARGs; however, the impact of urbanization on ARG spread of and the underlying driving mechanisms remain unclear. This study evaluates the diversity and abundance of ARGs in water and sediment samples from the Jialing River watershed in Chongqing City, China. The obtained results indicate that aminoglycoside and multidrug ARGs are the primary contributors to ARG presence in both sediments and water. Additionally, the diversity and abundance of ARGs are higher in water than in sediments. ARGs in watershed show a significant positive correlation with mobile genetic elements (MGEs). While environmental factors in urbanized watersheds affect ARG abundance and distribution to some extent, they are not the primary drivers. Urbanization itself emerges as a prominent factor influencing ARG diversity and abundance in river basins. Specifically, livestock, healthcare, and agriculture are identified as the main social factors influencing ARG proliferation in the highly urbanized areas of the Jialing River watershed. Further investigation into other contributing social factors, such as industrial development, is warranted. This study reveals the factors driving ARG distribution in urbanized watersheds, providing a foundation for future efforts to maintain ecological health in these environments.
Collapse
Affiliation(s)
- Ping Yu
- College of Resources and Environment, Chengdu University of Information Technology, No. 24 Block 1, Xuefu Road, Chengdu 610225, PR China; Biogas Institute of Ministry of Agriculture and Rural Affairs, No. 13, Section 4, Renmin South Road, Chengdu 610041, PR China; Key Laboratory of Development and Application of Rural Renewable Energy, Ministry of Agriculture and Rural Affairs, Chengdu 610041, PR China
| | - Xujing Guo
- College of Resources and Environment, Chengdu University of Information Technology, No. 24 Block 1, Xuefu Road, Chengdu 610225, PR China
| | - Wenguo Wang
- Biogas Institute of Ministry of Agriculture and Rural Affairs, No. 13, Section 4, Renmin South Road, Chengdu 610041, PR China; Key Laboratory of Development and Application of Rural Renewable Energy, Ministry of Agriculture and Rural Affairs, Chengdu 610041, PR China
| | - Lan Wang
- Biogas Institute of Ministry of Agriculture and Rural Affairs, No. 13, Section 4, Renmin South Road, Chengdu 610041, PR China; Key Laboratory of Development and Application of Rural Renewable Energy, Ministry of Agriculture and Rural Affairs, Chengdu 610041, PR China
| | - Hongwei Zhang
- Biogas Institute of Ministry of Agriculture and Rural Affairs, No. 13, Section 4, Renmin South Road, Chengdu 610041, PR China; Key Laboratory of Development and Application of Rural Renewable Energy, Ministry of Agriculture and Rural Affairs, Chengdu 610041, PR China
| | - Liangwei Deng
- Biogas Institute of Ministry of Agriculture and Rural Affairs, No. 13, Section 4, Renmin South Road, Chengdu 610041, PR China; Key Laboratory of Development and Application of Rural Renewable Energy, Ministry of Agriculture and Rural Affairs, Chengdu 610041, PR China
| | - Hongnan Yang
- Biogas Institute of Ministry of Agriculture and Rural Affairs, No. 13, Section 4, Renmin South Road, Chengdu 610041, PR China; Key Laboratory of Development and Application of Rural Renewable Energy, Ministry of Agriculture and Rural Affairs, Chengdu 610041, PR China
| | - Ting He
- Biogas Institute of Ministry of Agriculture and Rural Affairs, No. 13, Section 4, Renmin South Road, Chengdu 610041, PR China; Key Laboratory of Development and Application of Rural Renewable Energy, Ministry of Agriculture and Rural Affairs, Chengdu 610041, PR China
| | - Peike Wu
- Biogas Institute of Ministry of Agriculture and Rural Affairs, No. 13, Section 4, Renmin South Road, Chengdu 610041, PR China; Key Laboratory of Development and Application of Rural Renewable Energy, Ministry of Agriculture and Rural Affairs, Chengdu 610041, PR China
| | - Yunhong Zhang
- Biogas Institute of Ministry of Agriculture and Rural Affairs, No. 13, Section 4, Renmin South Road, Chengdu 610041, PR China; Key Laboratory of Development and Application of Rural Renewable Energy, Ministry of Agriculture and Rural Affairs, Chengdu 610041, PR China.
| |
Collapse
|
3
|
Rodrigues SO, Santiago FR, Silva MS, Lima ASG, Godoy LE, De Waard M, Fouad D, Batiha GE, Santos TL, Pagnossa JP. Macrolide resistance outcomes after the Covid-19 pandemic: A one health approach investigation. Biomed Pharmacother 2024; 180:117437. [PMID: 39303450 DOI: 10.1016/j.biopha.2024.117437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/22/2024] Open
Abstract
During the Covid-19 pandemic period, the indiscriminate use of macrolide-class antibiotics was frequent among the Brazilian population due to the lack of knowledge and information with a scientific basis. Thus, the class of drugs that includes azithromycin, clarithromycin, and erythromycin, which alter metabolic reactions in the body and act on the immune system, was widely used without medical prescription. Samples of bacterial strains from hospital environments were obtained during the most extensive spread of Covid-19 and studied in the present article, emphasizing the investigation for macrolide resistance genes (erm and msr) and bacteria of the genus Staphylococcus isolated from urinary tract infections. In addition, the physiological, genetic, immunological, and socio-epidemiological aspects were highlighted with a focus on the One Health approach and implications on the gut-brain axis in this integrative research, revealing that the inappropriate use of antibiotics directly affects entire communities, representing a significant concern for public and environmental health.
Collapse
Affiliation(s)
- Sarah O Rodrigues
- Department of Health and Biological Sciences, Pontifical Catholic University, Minas Gerais, Brazil.
| | - Felipe R Santiago
- Department of Health and Biological Sciences, Pontifical Catholic University, Minas Gerais, Brazil.
| | | | | | | | - Michel De Waard
- Smartox Biotechnology, 6 rue des Platanes, Saint-Egrève 38120, France; L'institut du thorax, INSERM, CNRS, UNIV NANTES, Nantes F-44007, France; Université de Nice Sophia-Antipolis, LabEx (Ion Channels, Science & Therapeutics), Valbonne F-06560, France.
| | - Dalia Fouad
- Department of Zoology, College of Science, King Saud University, PO Box 22452, Riyadh 11495, Saudi Arabia.
| | - Gaber E Batiha
- Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, AlBeheira, Egypt.
| | - Tamara L Santos
- Department of Health and Biological Sciences, Pontifical Catholic University, Minas Gerais, Brazil.
| | - Jorge P Pagnossa
- Department of Health and Biological Sciences, Pontifical Catholic University, Minas Gerais, Brazil.
| |
Collapse
|
4
|
Lomakin IB, Devarkar SC, Grada A, Bunick CG. Mechanistic Basis for the Translation Inhibition of Cutibacterium acnes by Clindamycin. J Invest Dermatol 2024; 144:2553-2561.e3. [PMID: 39122144 DOI: 10.1016/j.jid.2024.07.013] [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: 06/06/2024] [Revised: 07/11/2024] [Accepted: 07/13/2024] [Indexed: 08/12/2024]
Abstract
Inflammation and the Gram-positive anaerobic bacterium Cutibacterium acnes, which is implicated in acne pathogenesis and pilosebaceous-unit inflammation, are the main targets of antibiotic-based therapy against acne vulgaris (acne). The most widely used antibiotics in acne therapy are tetracyclines, macrolides, and lincosamides. Unfortunately, C. acnes bacteria over the past several decades have demonstrated increased resistance to these antibiotics, particularly to clindamycin. The precise knowledge of how antibiotics interact with their clinical target is needed to overcome this problem. Toward this goal, we determined the structure of clindamycin in complex with the ribosome of C. acnes at 2.53 Å resolution using cryogenic electron microscopy. The galactose sugar moiety of clindamycin interacts with nucleotides of the 23S ribosomal RNA directly or through a conserved network of water-mediated interactions. Its propyl pyrrolidinyl group interacts with the 23S ribosomal RNA through van der Waals forces. Clindamycin binding to the C. acnes ribosome interferes with both: proper orientation of the aminoacyl group of the A-site bound transfer RNA that is needed for peptide bond formation and with the extension of the nascent peptide. Our data are important for advancing the understanding of antibiotic resistance and development of narrow-spectrum antibacterial drugs, which is an urgent need for contemporary antibiotic stewardship.
Collapse
Affiliation(s)
- Ivan B Lomakin
- Department of Dermatology, Yale University, New Haven, Connecticut, USA.
| | - Swapnil C Devarkar
- Department of Molecular Biophysics and Biochemistry, Yale University, New Haven, Connecticut, USA
| | - Ayman Grada
- Department of Dermatology, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Christopher G Bunick
- Department of Dermatology, Yale University, New Haven, Connecticut, USA; Program in Translational Biomedicine, Yale University School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
5
|
Nor Amdan NA, Shahrulzamri NA, Hashim R, Mohamad Jamil N. Understanding the evolution of macrolides resistance: A mini review. J Glob Antimicrob Resist 2024; 38:368-375. [PMID: 39117142 DOI: 10.1016/j.jgar.2024.07.016] [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: 11/28/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Macrolides inhibit the growth of bacterial cells by preventing the elongation of polypeptides during protein biosynthesis and include natural, synthetic, and semi-synthetic products. Elongation prevention occurs by blocking the passage of the polypeptide chain as the macrolides bind at the nascent peptide exit tunnel. OBJECTIVE Recent data of ribosome profiling via ribo-seq further proves that, other than blocking the polypeptide chain, macrolides are also able to affect the synthesis of individual proteins. Thus, this shows that the mode of action of macrolides is more complex than we initially thought. Since the discovery of macrolides in the 1950s, they have been widely used in veterinary practice, agriculture, and medicine. Due to misuse and overuse of antibiotics, bacteria have acquired resistance against them. Hence, it is of utmost importance for us to fully understand the mode of action of macrolides as well as the mechanisms of resistance against macrolides in order to mitigate antibiotic-resistance issues. RESULTS Chemical modifications can be performed to improve macrolide potency if we have a better understanding of their mode of action. Furthermore, a complete and detailed understanding of the mode of action of macrolides has remained vague, as new findings have challenged theories that are already in existence-due to this obscurity, research into macrolide modes of action continues to this day. CONCLUSION In this review, we present an overview of macrolide antibiotics, with an emphasis on the latest knowledge regarding the mode of action of macrolides as well as the mechanisms of resistance employed by bacteria against macrolides.
Collapse
Affiliation(s)
- Nur Asyura Nor Amdan
- Bacteriology Unit, Infectious Disease Research Centre (IDRC), Institute for Medical Research (IMR), National Institutes of Health, Ministry of Health Malaysia, 40170, Shah Alam, Selangor, Malaysia
| | - Nur Atikah Shahrulzamri
- Faculty of Applied Sciences, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor, Malaysia
| | - Rohaidah Hashim
- Bacteriology Unit, Infectious Disease Research Centre (IDRC), Institute for Medical Research (IMR), National Institutes of Health, Ministry of Health Malaysia, 40170, Shah Alam, Selangor, Malaysia
| | - Norashirene Mohamad Jamil
- Faculty of Applied Sciences, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor, Malaysia; Molecular Microbial Pathogenicity Research Group, Pharmaceutical and Life Sciences Community of Research, Faculty of Applied Sciences, Universiti Teknologi MARA (UiTM), 40450 Shah Alam, Selangor, Malaysia.
| |
Collapse
|
6
|
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
|
7
|
Mullally CA, Fahriani M, Mowlaboccus S, Coombs GW. Non- faecium non- faecalis enterococci: a review of clinical manifestations, virulence factors, and antimicrobial resistance. Clin Microbiol Rev 2024; 37:e0012123. [PMID: 38466110 PMCID: PMC11237509 DOI: 10.1128/cmr.00121-23] [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] [Indexed: 03/12/2024] Open
Abstract
SUMMARYEnterococci are a diverse group of Gram-positive bacteria that are typically found as commensals in humans, animals, and the environment. Occasionally, they may cause clinically relevant diseases such as endocarditis, septicemia, urinary tract infections, and wound infections. The majority of clinical infections in humans are caused by two species: Enterococcus faecium and Enterococcus faecalis. However, there is an increasing number of clinical infections caused by non-faecium non-faecalis (NFF) enterococci. Although NFF enterococcal species are often overlooked, studies have shown that they may harbor antimicrobial resistance (AMR) genes and virulence factors that are found in E. faecium and E. faecalis. In this review, we present an overview of the NFF enterococci with a particular focus on human clinical manifestations, epidemiology, virulence genes, and AMR genes.
Collapse
Affiliation(s)
- Christopher A Mullally
- Antimicrobial Resistance and Infectious Diseases (AMRID) Research Laboratory, Murdoch University, Murdoch, Western Australia, Australia
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Marhami Fahriani
- Antimicrobial Resistance and Infectious Diseases (AMRID) Research Laboratory, Murdoch University, Murdoch, Western Australia, Australia
| | - Shakeel Mowlaboccus
- Antimicrobial Resistance and Infectious Diseases (AMRID) Research Laboratory, Murdoch University, Murdoch, Western Australia, Australia
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
- PathWest Laboratory Medicine-WA, Department of Microbiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| | - Geoffrey W Coombs
- Antimicrobial Resistance and Infectious Diseases (AMRID) Research Laboratory, Murdoch University, Murdoch, Western Australia, Australia
- The Marshall Centre for Infectious Diseases Research and Training, School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
- PathWest Laboratory Medicine-WA, Department of Microbiology, Fiona Stanley Hospital, Murdoch, Western Australia, Australia
| |
Collapse
|
8
|
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
|
9
|
Tabanelli G, Barbieri F, Baños A, Madero JMG, Daza MVB, Cortimiglia C, Milani G, Bassi D, Gardini F, Montanari C. Companilactobacillus alimentarius: An extensive characterization of strains isolated from spontaneous fermented sausages. Int J Food Microbiol 2024; 410:110489. [PMID: 38039926 DOI: 10.1016/j.ijfoodmicro.2023.110489] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/07/2023] [Accepted: 11/11/2023] [Indexed: 12/03/2023]
Abstract
Companilactobacillus alimentarius is a facultatively heterofermentative lactic acid bacterium (LAB) that is a significant constituent within the microbiota of various traditional fermented foods exerting several functions in fermentative or ripening processes. This species has been isolated from Spanish fermented sausages, where its frequency of isolation was comparable to those of Latilactobacillus sakei and Latilactobacillus curvatus. Despite to its presence in several niches, ecological information on this species is still scarce and only few publications report information about its safety features (i.e. antibiotic resistance). Since studies on C. alimentarius concern the analysis of a few individual traits regarding this species, a more extensive work on a larger number of isolates from the same matrix have been performed to allow a clearer interpretation of their phenotypic and technological characteristics. Specifically, 14 strains of C. alimentarius isolated from Mediterranean spontaneously fermented sausages, have been screened for their safety and technological characteristics (such as antibiotic resistance, biogenic amine production, inhibiting potential, growth at different temperatures and NaCl concentrations) and with phenotype microarrays with the aim to elucidate their potential role and contribution to sausage fermentation and ripening. In general, a wide variability was observed in relation to the parameters considered. Several of the tested strains were able to produce histamine, tyramine and putrescine while the antibiotic resistance greatly varied according to the strains, with the exception of vancomycin. In addition, C. alimentarius strains showed a relevant potential to grow in conditions of salt and temperature mimicking those found in fermented foods. In particular, the growth at 10 °C and in the presence of salt can explain the presence of C. alimentarius in sausages and its adaptation to fermented meat environment in which low temperature can be applied during ripening. The differentiation of the phenotypic profile reflected the environmental conditions that influenced the isolation source, including those derived by the raw materials. Given the species frequent association with spontaneous fermentations or the ripening microbiota of various products, despite not being intentionally used as starter cultures, the data presented in this study contribute to a deeper comprehension of their role, both advantageous and detrimental, in numerous significant fermented foods.
Collapse
Affiliation(s)
- Giulia Tabanelli
- Department of Agricultural and Food Sciences, University of Bologna, 40127 Bologna, Italy; Interdepartmental Center for Industrial Agri-Food Research, University of Bologna, 47521 Cesena, Italy
| | - Federica Barbieri
- Department of Agricultural and Food Sciences, University of Bologna, 40127 Cesena, Italy.
| | - Alberto Baños
- Department of Microbiology, DOMCA S.A.U., 18620 Alhendín, Spain
| | | | - Mireya Viviana Belloso Daza
- Department for Sustainable Food Process (DISTAS), Università Cattolica del Sacro Cuore, 26100 Cremona, Italy
| | - Claudia Cortimiglia
- Department for Sustainable Food Process (DISTAS), Università Cattolica del Sacro Cuore, 26100 Cremona, Italy
| | - Giovanni Milani
- Department for Sustainable Food Process (DISTAS), Università Cattolica del Sacro Cuore, 26100 Cremona, Italy
| | - Daniela Bassi
- Department for Sustainable Food Process (DISTAS), Università Cattolica del Sacro Cuore, 26100 Cremona, Italy
| | - Fausto Gardini
- Department of Agricultural and Food Sciences, University of Bologna, 40127 Cesena, Italy
| | - Chiara Montanari
- Department of Agricultural and Food Sciences, University of Bologna, 40127 Cesena, Italy
| |
Collapse
|
10
|
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
|
11
|
Yang Z, Yang X, Wang M, Jia R, Chen S, Liu M, Zhao X, Yang Q, Wu Y, Zhang S, Huang J, Ou X, Mao S, Gao Q, Sun D, Tian B, Zhu D, Cheng A. Genome-wide association study reveals serovar-associated genetic loci in Riemerella anatipestifer. BMC Genomics 2024; 25:57. [PMID: 38216873 PMCID: PMC10787497 DOI: 10.1186/s12864-024-09988-4] [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: 04/26/2023] [Accepted: 01/08/2024] [Indexed: 01/14/2024] Open
Abstract
BACKGROUND The disease caused by Riemerella anatipestifer (R. anatipestifer, RA) results in large economic losses to the global duck industry every year. Serovar-related genomic variation, such as the O-antigen and capsular polysaccharide (CPS) gene clusters, has been widely used for serotyping in many gram-negative bacteria. RA has been classified into at least 21 serovars based on slide agglutination, but the molecular basis of serotyping is unknown. In this study, we performed a pan-genome-wide association study (Pan-GWAS) to identify the genetic loci associated with RA serovars. RESULTS The results revealed a significant association between the putative CPS synthesis gene locus and the serological phenotype. Further characterization of the CPS gene clusters in 11 representative serovar strains indicated that they were highly diverse and serovar-specific. The CPS gene cluster contained the key genes wzx and wzy, which are involved in the Wzx/Wzy-dependent pathway of CPS synthesis. Similar CPS loci have been found in some other species within the family Weeksellaceae. We have also shown that deletion of the wzy gene in RA results in capsular defects and cross-agglutination. CONCLUSIONS This study indicates that the CPS synthesis gene cluster of R. anatipestifer is a serotype-specific genetic locus. Importantly, our finding provides a new perspective for the systematic analysis of the genetic basis of the R anatipestifer serovars and a potential target for establishing a complete molecular serotyping scheme.
Collapse
Affiliation(s)
- Zhishuang Yang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
| | - Xueqin Yang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
| | - Mingshu Wang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Renyong Jia
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Shun Chen
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Mafeng Liu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Xinxin Zhao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Qiao Yang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Ying Wu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Shaqiu Zhang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Juan Huang
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Xumin Ou
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Sai Mao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Qun Gao
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Di Sun
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Bin Tian
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China
| | - Dekang Zhu
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China.
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China.
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China.
| | - Anchun Cheng
- Research Center of Avian Diseases, College of Veterinary Medicine, Sichuan Agricultural University, Chengdu, Sichuan, China.
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Chengdu, Sichuan, China.
- International Joint Research Center for Animal Disease Prevention and Control of Sichuan Province, Chengdu, Sichuan, China.
- Engineering Research Center of Southwest Animal Disease Prevention and Control Technology, Ministry of Education of the People's Republic of China, Chengdu, Sichuan, China.
| |
Collapse
|
12
|
Clark JA, Curran MD, Gouliouris T, Conway Morris A, Bousfield R, Navapurkar V, Kean IRL, Daubney E, White D, Baker S, Pathan N. Rapid Detection of Antimicrobial Resistance Genes in Critically Ill Children Using a Custom TaqMan Array Card. Antibiotics (Basel) 2023; 12:1701. [PMID: 38136735 PMCID: PMC10740637 DOI: 10.3390/antibiotics12121701] [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: 11/01/2023] [Revised: 11/28/2023] [Accepted: 12/04/2023] [Indexed: 12/24/2023] Open
Abstract
Bacteria are identified in only 22% of critically ill children with respiratory infections treated with antimicrobial therapy. Once an organism is isolated, antimicrobial susceptibility results (phenotypic testing) can take another day. A rapid diagnostic test identifying antimicrobial resistance (AMR) genes could help clinicians make earlier, informed antimicrobial decisions. Here we aimed to validate a custom AMR gene TaqMan Array Card (AMR-TAC) for the first time and assess its feasibility as a screening tool in critically ill children. An AMR-TAC was developed using a combination of commercial and bespoke targets capable of detecting 23 AMR genes. This was validated using isolates with known phenotypic resistance. The card was then tested on lower respiratory tract and faecal samples obtained from mechanically ventilated children in a single-centre observational study of respiratory infection. There were 82 children with samples available, with a median age of 1.2 years. Major comorbidity was present in 29 (35%) children. A bacterial respiratory pathogen was identified in 13/82 (16%) of children, of which 4/13 (31%) had phenotypic AMR. One AMR gene was detected in 49/82 (60%), and multiple AMR genes were detected in 14/82 (17%) children. Most AMR gene detections were not associated with the identification of phenotypic AMR. AMR genes are commonly detected in samples collected from mechanically ventilated children with suspected respiratory infections. AMR-TAC may have a role as an adjunct test in selected children in whom there is a high suspicion of antimicrobial treatment failure.
Collapse
Affiliation(s)
- John A. Clark
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK; (I.R.L.K.); (E.D.); (D.W.); (N.P.)
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK; (T.G.); (A.C.M.); (R.B.); (V.N.)
| | - Martin D. Curran
- Clinical Microbiology and Public Health Laboratory, United Kingdom Health Security Agency, Cambridge CB2 0QQ, UK;
| | - Theodore Gouliouris
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK; (T.G.); (A.C.M.); (R.B.); (V.N.)
- Clinical Microbiology and Public Health Laboratory, United Kingdom Health Security Agency, Cambridge CB2 0QQ, UK;
| | - Andrew Conway Morris
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK; (T.G.); (A.C.M.); (R.B.); (V.N.)
- Division of Anaesthesia, Department of Medicine, University of Cambridge, Cambridge CB2 2QQ, UK
- Division of Immunology, Department of Pathology, University of Cambridge, Cambridge CB2 1QP, UK
| | - Rachel Bousfield
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK; (T.G.); (A.C.M.); (R.B.); (V.N.)
- Clinical Microbiology and Public Health Laboratory, United Kingdom Health Security Agency, Cambridge CB2 0QQ, UK;
| | - Vilas Navapurkar
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK; (T.G.); (A.C.M.); (R.B.); (V.N.)
| | - Iain R. L. Kean
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK; (I.R.L.K.); (E.D.); (D.W.); (N.P.)
| | - Esther Daubney
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK; (I.R.L.K.); (E.D.); (D.W.); (N.P.)
| | - Deborah White
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK; (I.R.L.K.); (E.D.); (D.W.); (N.P.)
| | - Stephen Baker
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, University of Cambridge, Cambridge CB2 0AW, UK;
| | - Nazima Pathan
- Department of Paediatrics, University of Cambridge, Cambridge CB2 0QQ, UK; (I.R.L.K.); (E.D.); (D.W.); (N.P.)
- Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK; (T.G.); (A.C.M.); (R.B.); (V.N.)
| |
Collapse
|
13
|
Obana N, Takada H, Crowe-McAuliffe C, Iwamoto M, Egorov AA, Wu KJY, Chiba S, Murina V, Paternoga H, Tresco BIC, Nomura N, Myers AG, Atkinson G, Wilson DN, Hauryliuk V. Genome-encoded ABCF factors implicated in intrinsic antibiotic resistance in Gram-positive bacteria: VmlR2, Ard1 and CplR. Nucleic Acids Res 2023; 51:4536-4554. [PMID: 36951104 PMCID: PMC10201436 DOI: 10.1093/nar/gkad193] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 02/17/2023] [Accepted: 03/06/2023] [Indexed: 03/24/2023] Open
Abstract
Genome-encoded antibiotic resistance (ARE) ATP-binding cassette (ABC) proteins of the F subfamily (ARE-ABCFs) mediate intrinsic resistance in diverse Gram-positive bacteria. The diversity of chromosomally-encoded ARE-ABCFs is far from being fully experimentally explored. Here we characterise phylogenetically diverse genome-encoded ABCFs from Actinomycetia (Ard1 from Streptomyces capreolus, producer of the nucleoside antibiotic A201A), Bacilli (VmlR2 from soil bacterium Neobacillus vireti) and Clostridia (CplR from Clostridium perfringens, Clostridium sporogenes and Clostridioides difficile). We demonstrate that Ard1 is a narrow spectrum ARE-ABCF that specifically mediates self-resistance against nucleoside antibiotics. The single-particle cryo-EM structure of a VmlR2-ribosome complex allows us to rationalise the resistance spectrum of this ARE-ABCF that is equipped with an unusually long antibiotic resistance determinant (ARD) subdomain. We show that CplR contributes to intrinsic pleuromutilin, lincosamide and streptogramin A resistance in Clostridioides, and demonstrate that C. difficile CplR (CDIF630_02847) synergises with the transposon-encoded 23S ribosomal RNA methyltransferase Erm to grant high levels of antibiotic resistance to the C. difficile 630 clinical isolate. Finally, assisted by uORF4u, our novel tool for detection of upstream open reading frames, we dissect the translational attenuation mechanism that controls the induction of cplR expression upon an antibiotic challenge.
Collapse
Affiliation(s)
- Nozomu Obana
- Transborder Medical Research Center, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
- Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, Tsukuba, Japan
| | - Hiraku Takada
- Faculty of Life Sciences, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-Ku, Kyoto 603-8555, Japan
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Caillan Crowe-McAuliffe
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Mizuki Iwamoto
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Artyom A Egorov
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Kelvin J Y Wu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Shinobu Chiba
- Faculty of Life Sciences, Kyoto Sangyo University, Motoyama, Kamigamo, Kita-Ku, Kyoto 603-8555, Japan
- Institute for Protein Dynamics, Kyoto Sangyo University, Japan
| | | | - Helge Paternoga
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Ben I C Tresco
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Nobuhiko Nomura
- Microbiology Research Center for Sustainability (MiCS), University of Tsukuba, Tsukuba, Japan
- Faculty of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan
| | - Andrew G Myers
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Gemma C Atkinson
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Daniel N Wilson
- Institute for Biochemistry and Molecular Biology, University of Hamburg, Martin-Luther-King-Platz 6, 20146 Hamburg, Germany
| | - Vasili Hauryliuk
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- University of Tartu, Institute of Technology, Tartu, Estonia
- Science for Life Laboratory, Lund, Sweden
| |
Collapse
|
14
|
Zaher HA, El Baz S, Alothaim AS, Alsalamah SA, Alghonaim MI, Alawam AS, Eraqi MM. Molecular Basis of Methicillin and Vancomycin Resistance in Staphylococcus aureus from Cattle, Sheep Carcasses and Slaughterhouse Workers. Antibiotics (Basel) 2023; 12:antibiotics12020205. [PMID: 36830115 PMCID: PMC9952529 DOI: 10.3390/antibiotics12020205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/13/2023] [Accepted: 01/14/2023] [Indexed: 01/21/2023] Open
Abstract
Staphylococcus aureus (S. aureus) is a serious infection-causing pathogen in humans and animal. In particular, methicillin-resistant S. aureus (MRSA) is considered one of the major life-threatening pathogens due to its rapid resistance to several antibiotics in clinical practice. MRSA strains have recently been isolated in a number of animals utilized in food production processes, and these species are thought to be the important sources of the spread of infection and disease in both humans and animals. The main objective of the current study was to assess the prevalence of drug-resistant S. aureus, particularly vancomycin-resistant S. aureus (VRSA) and MRSA, by molecular methods. To address this issue, a total of three hundred samples (200 meat samples from cattle and sheep carcasses (100 of each), 50 hand swabs, and 50 stool samples from abattoir workers) were obtained from slaughterhouses in Egypt provinces. In total, 19% S. aureus was isolated by standard culture techniques, and the antibiotic resistance was confirmed genotypically by amplification nucA gen. Characteristic resistance genes were identified by PCR with incidence of 31.5%, 19.3%, 8.7%, and 7% for the mecA, VanA, ermA, and tet L genes, respectively, while the aac6-aph gene was not found in any of the isolates. In this study, the virulence genes responsible for S. aureus' resistance to antibiotics had the highest potential for infection or disease transmission to animal carcasses, slaughterhouse workers, and meat products.
Collapse
Affiliation(s)
- Hanan A. Zaher
- Food Hygiene and Control Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Shimaa El Baz
- Department of Hygiene and Zoonoses, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Abdulaziz S. Alothaim
- Department of Biology, College of Science in Zulfi, Majmaah University, Majmaah 11952, Saudi Arabia
| | - Sulaiman A. Alsalamah
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 11623, Saudi Arabia
| | - Mohammed Ibrahim Alghonaim
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 11623, Saudi Arabia
| | - Abdullah S. Alawam
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University, Riyadh 11623, Saudi Arabia
| | - Mostafa M. Eraqi
- Department of Biology, College of Science in Zulfi, Majmaah University, Majmaah 11952, Saudi Arabia
- Microbiology and Immunology Department, Veterinary Research Institute, National Research Centre, Dokki, Giza 12622, Egypt
- Correspondence: ; Tel.: +966-565709849
| |
Collapse
|
15
|
Lepczyński A, Herosimczyk A, Bucław M, Adaszyńska-Skwirzyńska M. Antibiotics in avian care and husbandry-status and alternative antimicrobials. PHYSICAL SCIENCES REVIEWS 2023. [DOI: 10.1515/psr-2021-0123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Abstract
Undoubtedly, the discovery of antibiotics was one of the greatest milestones in the treatment of human and animal diseases. Due to their over-use mainly as antibiotic growth promoters (AGP) in livestock farming, antimicrobial resistance has been reported with increasing intensity, especially in the last decades. In order to reduce the scale of this phenomenon, initially in the Scandinavian countries and then throughout the entire European Union, a total ban on the use of AGP was introduced, moreover, a significant limitation in the use of these feed additives is now observed almost all over the world. The withdrawal of AGP from widespread use has prompted investigators to search for alternative strategies to maintain and stabilize the composition of the gut microbiota. These strategies include substances that are used in an attempt to stimulate the growth and activity of symbiotic bacteria living in the digestive tract of animals, as well as living microorganisms capable of colonizing the host’s gastrointestinal tract, which can positively affect the composition of the intestinal microbiota by exerting a number of pro-health effects, i.e., prebiotics and probiotics, respectively. In this review we also focused on plants/herbs derived products that are collectively known as phytobiotic.
Collapse
Affiliation(s)
- Adam Lepczyński
- Department of Physiology, Cytobiology and Proteomics , West Pomeranian University of Technology , Szczecin , Poland
| | - Agnieszka Herosimczyk
- Department of Physiology, Cytobiology and Proteomics , West Pomeranian University of Technology , Szczecin , Poland
| | - Mateusz Bucław
- Department of Monogastric Animal Sciences , West Pomeranian University of Technology , Szczecin , Poland
| | | |
Collapse
|
16
|
Sparbrod M, Gager Y, Koehler AK, Jentsch H, Stingu CS. Relationship between Phenotypic and Genotypic Resistance of Subgingival Biofilm Samples in Patients with Periodontitis. Antibiotics (Basel) 2022; 12:antibiotics12010068. [PMID: 36671269 PMCID: PMC9854985 DOI: 10.3390/antibiotics12010068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 12/20/2022] [Accepted: 12/23/2022] [Indexed: 01/03/2023] Open
Abstract
The phenotypic expression of antibiotic resistance genes (ARGs) can hamper the use of antibiotics as adjuncts to subgingival instrumentation in the treatment of periodontitis patients. The aim of the study was to analyze the relationship between the phenotypic and genotypic resistance against ampicillin-sulbactam, clindamycin, doxycycline and metronidazole of subgingival biofilm samples from 19 periodontitis patients. Samples were analyzed with shotgun sequencing and cultivated anaerobically for 7 days on microbiological culture media incorporating antibiotics. All growing isolates were identified to the species level using MALDI-TOF-MS and sequence analysis of the 16S ribosomal RNA (rRNA) gene. Phenotypic resistance was determined using EUCAST-breakpoints. The genetic profile of eight patients matched completely with phenotypical resistance to the tested antibiotics. The positive predictive values varied from 1.00 for clindamycin to 0.57 for doxycycline and 0.25 for ampicillin-sulbactam. No sample contained the nimI gene. It can be concluded that antibiotic resistance may be polygenetic and genes may be silent. Every biofilm sample harboring erm genes was phenotypic resistant. The absence of cfx and tet genes correlated to 100%, respectively, to 75%, with the absence of phenotypic resistance. The absence of nimI genes leads to the assumption that constitutive resistance among several species could explain the resistance to metronidazole.
Collapse
Affiliation(s)
- Moritz Sparbrod
- Institute for Medical Microbiology and Virology, University Hospital Leipzig, 04103 Leipzig, Germany
- Correspondence: (M.S.); (C.-S.S.)
| | | | | | - Holger Jentsch
- Center of Periodontology, University Hospital Leipzig, 04103 Leipzig, Germany
| | - Catalina-Suzana Stingu
- Institute for Medical Microbiology and Virology, University Hospital Leipzig, 04103 Leipzig, Germany
- Correspondence: (M.S.); (C.-S.S.)
| |
Collapse
|
17
|
Li G, Walker MJ, De Oliveira DMP. Vancomycin Resistance in Enterococcus and Staphylococcus aureus. Microorganisms 2022; 11:microorganisms11010024. [PMID: 36677316 PMCID: PMC9866002 DOI: 10.3390/microorganisms11010024] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/24/2022] Open
Abstract
Enterococcus faecalis, Enterococcus faecium and Staphylococcus aureus are both common commensals and major opportunistic human pathogens. In recent decades, these bacteria have acquired broad resistance to several major classes of antibiotics, including commonly employed glycopeptides. Exemplified by resistance to vancomycin, glycopeptide resistance is mediated through intrinsic gene mutations, and/or transferrable van resistance gene cassette-carrying mobile genetic elements. Here, this review will discuss the epidemiology of vancomycin-resistant Enterococcus and S. aureus in healthcare, community, and agricultural settings, explore vancomycin resistance in the context of van and non-van mediated resistance development and provide insights into alternative therapeutic approaches aimed at treating drug-resistant Enterococcus and S. aureus infections.
Collapse
|
18
|
Immunomodulatory Effects of Macrolides Considering Evidence from Human and Veterinary Medicine. Microorganisms 2022; 10:microorganisms10122438. [PMID: 36557690 PMCID: PMC9784682 DOI: 10.3390/microorganisms10122438] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 11/17/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
Macrolide antimicrobial agents have been in clinical use for more than 60 years in both human and veterinary medicine. The discovery of the non-antimicrobial properties of macrolides and the effect of immunomodulation of the inflammatory response has benefited patients with chronic airway diseases and impacted morbidity and mortality. This review examines the evidence of antimicrobial and non-antimicrobial properties of macrolides in human and veterinary medicine with a focus toward veterinary macrolides but including important and relevant evidence from the human literature. The complete story for these complex and important molecules is continuing to be written.
Collapse
|
19
|
Santos RAND, Abdel-Nour J, McAuley C, Moore SC, Fegan N, Fox EM. Clostridium perfringens associated with dairy farm systems show diverse genotypes. Int J Food Microbiol 2022; 382:109933. [PMID: 36166891 DOI: 10.1016/j.ijfoodmicro.2022.109933] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/08/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
Abstract
Clostridium perfringens is a bacterial species of importance to both public and animal health. Frequently found in food system environments, it presents a risk to food animal health such as dairy herds, and may cross contaminate associated ingredients or food products, with potential to cause sporadic and outbreaks of disease in human populations, including gastroenteric illness. In this study, we characterized C. perfringens isolated from bovine, caprine, and ovine dairy farm systems (n = 8, 11 and 4, respectively). Isolates were phenotypically screened for antimicrobial sensitivity profiling, and subjected to whole genome sequencing to elucidate related genetic markers, as well as examine virulence gene markers, mobile genetic elements, and other features. Both toxin type A and type D isolates were identified (78 % and 22 % of isolates, respectively), including 20 novel sequence types. Resistance to clindamycin was most prevalent among antibiotics screened (30 %), followed by erythromycin (13 %), then penicillin and tetracycline (4 %), although an additional 3 isolates were non-susceptible to tetracycline. Most isolates harboured plasmids, which mobilised virulence markers such as etx, cpb2, and resistance markers tetA(P), tetB(P), and erm(Q), on conjugative plasmids. The presence of type D isolates on caprine farms emphasizes the need for control efforts to prevent infection and potential enterotoxemia. Clostridium perfringens enterotoxin (cpe) was not identified, suggesting lower risk of gastrointestinal illness from contaminated foods, the presence of other virulence and antimicrobial resistance markers suggests farm hygiene remains an important consideration to help ensure food safety of associated dairy foods produced.
Collapse
Affiliation(s)
| | | | - Cathy McAuley
- CSIRO Agriculture and Food, Werribee, VIC 3030, Australia
| | - Sean C Moore
- CSIRO Agriculture and Food, Cooper Plains, QLD 4108, Australia
| | - Narelle Fegan
- CSIRO Agriculture and Food, Cooper Plains, QLD 4108, Australia
| | - Edward M Fox
- Department of Applied Sciences, Northumbria University, Newcastle upon Tyne, UK.
| |
Collapse
|
20
|
Zhu J, Wang J, Chen YP, Qing T, Zhang P, Feng B. Quantitative proteomics and phosphoproteomics elucidate the molecular mechanism of nanostructured TiO 2-stimulated biofilm formation. JOURNAL OF HAZARDOUS MATERIALS 2022; 432:128709. [PMID: 35325859 DOI: 10.1016/j.jhazmat.2022.128709] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/08/2022] [Accepted: 03/12/2022] [Indexed: 06/14/2023]
Abstract
With the increasing concerns regarding bacterial adaption to nanomaterials, it is critical to explore the main mechanism behind the adaptive morphogenesis of microorganisms. In this work, the biofilms formed from activated sludge exposed to 5 and 50 mg/L nTiO2 in the dark had increased biomass and selectively enriched pathogens. To further elaborate adaptive mechanism of biofilm formation induced by nTiO2, the protein response and protein phosphorylation modification of Escherichia coli K12 were determined using integrative systems biology analyses of proteomics and phosphoproteomics. Results identified that E. coli cultivated with nTiO2 considerably upregulated iron acquisition, and regulated protein phosphorylation states associated with of transcription and translation and biofilm formation relative to unexposed controls. Accordingly, bacteria increased siderophores and exopolysaccharide content (increased by about 57% and 231%, respectively), and enhanced resistance to transcriptional inhibitory antibiotics. Moreover, a dose of an iron chelator, i.e., deferoxamine mesylate salt, effectively retarded the biofilm development of bacteria exposed to 50 mg/L nTiO2. Overall, this work will provide a new insight for biofouling control, and contribute to an improved understanding of microbial adaption to nanomaterials.
Collapse
Affiliation(s)
- Jing Zhu
- College of Environment and Resources, Xiangtan University, Xiangtan 411105, China
| | - Jingyu Wang
- College of Environment and Resources, Xiangtan University, Xiangtan 411105, China
| | - You-Peng Chen
- Key Laboratory of the Three Gorges Reservoir Region's Eco-Environments of MOE, Chongqing University, Chongqing 400045, China
| | - Taiping Qing
- College of Environment and Resources, Xiangtan University, Xiangtan 411105, China
| | - Peng Zhang
- College of Environment and Resources, Xiangtan University, Xiangtan 411105, China.
| | - Bo Feng
- College of Environment and Resources, Xiangtan University, Xiangtan 411105, China
| |
Collapse
|
21
|
Molecular docking and pharmacokinetic prediction of phytochemicals from Syzygium cumini in interaction with penicillin-binding protein 2a and erythromycin ribosomal methylase of Staphylococcus aureus. BIOTECHNOLOGIA 2022; 103:5-18. [PMID: 36605380 PMCID: PMC9642940 DOI: 10.5114/bta.2022.113910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 08/23/2021] [Accepted: 10/12/2021] [Indexed: 01/09/2023] Open
Abstract
Background MRSA and MLSB resistant S. aureus are known as important pathogens, which are responsible for many cases of both hospital and community-acquired infections worldwide. Studying drug discovery from plant sources is regarded as an important prevention strategy regarding these types of infections. Material and methods Agar well diffusion method was performed for antimicrobial evaluation, LCMS technique used for identification of different compounds, molecular docking performed by application of i GEMDOCK for PBP2a and ERM to plant compounds, and its pharmacokinetic evaluation of ADMET through use of AdmetSAR. Results Water extract was the most effective against resistant strains of Staphylococcus aureus. Twenty compounds belonging to phenols, flavonoids, organic acids, terpenoids groups were reported. Eighteen plant compounds passed in Lipinski's rule of five. i GEMDOCK revealed diferulic acid has the least binding energy -102.37 kcal/mole to penicillin-binding protein 2a and taxifolin has the least binding energy of -103.12 kcal/mole to erythromycin ribosomal methylase in comparison to control linezolid. These compounds raise the potential for developing potent inhibitors of penicillin-binding protein 2a and erythromycin ribosomal methylase for drug development. ADMET properties revealed that eighteen studied compounds were found in category III and IV with non-toxic properties except two butin and taxifolin found in category II with toxic properties. Conclusions It can be concluded that diferulic acid and taxifolin compounds provide the best inhibitor effect to PBP2a and ERM protein for inhibition of MRSA and MLSB resistant strains of S. aureus through the application of molecular docking, leading to a lead drug candidate for the treatment of diseases.
Collapse
|
22
|
Lee AR, Park SB, Kim SW, Jung JW, Chun JH, Kim J, Kim YR, Lazarte JMS, Jang HB, Thompson KD, Jung M, Ha MW, Jung TS. Membrane vesicles (MVs) from antibiotic-resistant Staphylococcus aureus transfer antibiotic-resistance to antibiotic-susceptible Escherichia coli. J Appl Microbiol 2022; 132:2746-2759. [PMID: 35019198 PMCID: PMC9306644 DOI: 10.1111/jam.15449] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 01/03/2022] [Accepted: 01/08/2022] [Indexed: 11/03/2022]
Abstract
AIM Bacteria naturally produce membrane vesicles (MVs), which have been shown to contribute to the spread of multi-drug resistant bacteria (MDR) by delivering antibiotic-resistant substances to antibiotic-susceptible bacteria. Here, we aim to show that MVs from Gram-positive bacteria are capable of transferring β-lactam antibiotic-resistant substances to antibiotic-sensitive Gram-negative bacteria. MATERIALS AND METHODS MVs were collected from a methicillin-resistant strain of Staphylococcus aureus (MRSA) and vesicle-mediated fusion with antimicrobial-sensitive Escherichia coli (RC85). It was performed by exposing the bacteria to the MVs to develop antimicrobial-resistant E. coli (RC85-T). RESULTS The RC85-T exhibited a higher resistance to β-lactam antibiotics compared to the parent strain. Although the secretion rates of the MVs from RC85-T and the parent strain were nearly equal, the β-lactamase activity of the MVs from RC85-T was 12-times higher than that of MVs from the parent strain, based on equivalent protein concentrations. Moreover, MVs secreted by RC85-T were able to protect β-lactam-susceptible E. coli from β-lactam antibiotic-induced growth inhibition in a dose-dependent manner. CONCLUSION MVs play a role in transferring substances from Gram-positive to Gram-negative bacteria, shown by the release of MVs from RC85-T that were able to protect β-lactam-susceptible bacteria from β-lactam antibiotics. SIGNIFICANCE AND IMPACT OF STUDY MVs are involved in the emergence of antibiotic resistant strains in a mixed bacterial culture, helping us to understand how the spread of multidrug resistant bacteria could be reduced.
Collapse
Affiliation(s)
- Ae Rin Lee
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do, Republic of Korea, 52828
| | - Seong Bin Park
- Coastal Research Extension Center, Mississippi State University, Mississippi State, MS, 39762, USA
| | - Si Won Kim
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do, Republic of Korea, 52828
| | - Jae Wook Jung
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do, Republic of Korea, 52828
| | - Jin Hong Chun
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do, Republic of Korea, 52828
| | - Jaesung Kim
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do, Republic of Korea, 52828
| | - Young Rim Kim
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do, Republic of Korea, 52828
| | - Jassy Mary S Lazarte
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do, Republic of Korea, 52828
| | - Ho Bin Jang
- Department of Microbiology, Institute for Viral Diseases, College of Medicine, Korea University, Seoul, Republic of Korea, 02841
| | - Kim D Thompson
- Moredun Research Institute, Pentlands Science Park, Bush Loan, Penicuik, Midlothian, EH26 0PZ, UK
| | - Myunghwan Jung
- Department of Microbiology and Department of Convergence Medical Science, College of Medicine, Gyeongsang National University, Jinju, Republic of Korea
| | - Min Woo Ha
- College of Pharmacy, Jeju National University, Jeju, 63243, Republic of Korea
| | - Tae Sung Jung
- Laboratory of Aquatic Animal Diseases, Research Institute of Natural Science, College of Veterinary Medicine, Gyeongsang National University, 501-201, 501, Jinju-daero, Jinju-si, Gyeongsangnam-do, Republic of Korea, 52828.,Centre for Marine Bioproducts Development, Flinders University, Bedford Park, SA, 5042, Australia
| |
Collapse
|
23
|
Mitcheltree MJ, Pisipati A, Syroegin EA, Silvestre KJ, Klepacki D, Mason JD, Terwilliger DW, Testolin G, Pote AR, Wu KJY, Ladley RP, Chatman K, Mankin AS, Polikanov YS, Myers AG. A synthetic antibiotic class overcoming bacterial multidrug resistance. Nature 2021; 599:507-512. [PMID: 34707295 PMCID: PMC8549432 DOI: 10.1038/s41586-021-04045-6] [Citation(s) in RCA: 107] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 09/21/2021] [Indexed: 02/08/2023]
Abstract
The dearth of new medicines effective against antibiotic-resistant bacteria presents a growing global public health concern1. For more than five decades, the search for new antibiotics has relied heavily on the chemical modification of natural products (semisynthesis), a method ill-equipped to combat rapidly evolving resistance threats. Semisynthetic modifications are typically of limited scope within polyfunctional antibiotics, usually increase molecular weight, and seldom permit modifications of the underlying scaffold. When properly designed, fully synthetic routes can easily address these shortcomings2. Here we report the structure-guided design and component-based synthesis of a rigid oxepanoproline scaffold which, when linked to the aminooctose residue of clindamycin, produces an antibiotic of exceptional potency and spectrum of activity, which we name iboxamycin. Iboxamycin is effective against ESKAPE pathogens including strains expressing Erm and Cfr ribosomal RNA methyltransferase enzymes, products of genes that confer resistance to all clinically relevant antibiotics targeting the large ribosomal subunit, namely macrolides, lincosamides, phenicols, oxazolidinones, pleuromutilins and streptogramins. X-ray crystallographic studies of iboxamycin in complex with the native bacterial ribosome, as well as with the Erm-methylated ribosome, uncover the structural basis for this enhanced activity, including a displacement of the [Formula: see text] nucleotide upon antibiotic binding. Iboxamycin is orally bioavailable, safe and effective in treating both Gram-positive and Gram-negative bacterial infections in mice, attesting to the capacity for chemical synthesis to provide new antibiotics in an era of increasing resistance.
Collapse
Affiliation(s)
- Matthew J Mitcheltree
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Amarnath Pisipati
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Egor A Syroegin
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Katherine J Silvestre
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Dorota Klepacki
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Jeremy D Mason
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Daniel W Terwilliger
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Giambattista Testolin
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Aditya R Pote
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Kelvin J Y Wu
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Richard Porter Ladley
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Kelly Chatman
- Harvard Center for Mass Spectrometry, Harvard University, Cambridge, MA, USA
| | - Alexander S Mankin
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, USA
| | - Yury S Polikanov
- Department of Biological Sciences, University of Illinois at Chicago, Chicago, IL, USA.
| | - Andrew G Myers
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
24
|
Andolfo G, Schuster C, Gharsa HB, Ruocco M, Leclerque A. Genomic analysis of the nomenclatural type strain of the nematode-associated entomopathogenic bacterium Providencia vermicola. BMC Genomics 2021; 22:708. [PMID: 34598677 PMCID: PMC8487129 DOI: 10.1186/s12864-021-08027-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 09/20/2021] [Indexed: 12/18/2022] Open
Abstract
Background Enterobacteria of the genus Providencia are mainly known as opportunistic human pathogens but have been isolated from highly diverse natural environments. The species Providencia vermicola comprises insect pathogenic bacteria carried by entomoparasitic nematodes and is investigated as a possible insect biocontrol agent. The recent publication of several genome sequences from bacteria assigned to this species has given rise to inconsistent preliminary results. Results The genome of the nematode-derived P. vermicola type strain DSM_17385 has been assembled into a 4.2 Mb sequence comprising 5 scaffolds and 13 contigs. A total of 3969 protein-encoding genes were identified. Multilocus sequence typing with different marker sets revealed that none of the previously published presumed P. vermicola genomes represents this taxonomic species. Comparative genomic analysis has confirmed a close phylogenetic relationship of P. vermicola to the P. rettgeri species complex. P. vermicola DSM_17385 carries a type III secretion system (T3SS-1) with probable function in host cell invasion or intracellular survival. Potentially antibiotic resistance-associated genes comprising numerous efflux pumps and point-mutated house-keeping genes, have been identified across the P. vermicola genome. A single small (3.7 kb) plasmid identified, pPVER1, structurally belongs to the qnrD-type family of fluoroquinolone resistance conferring plasmids that is prominent in Providencia and Proteus bacteria, but lacks the qnrD resistance gene. Conclusions The sequence reported represents the first well-supported published genome for the taxonomic species P. vermicola to be used as reference in further comparative genomics studies on Providencia bacteria. Due to a striking difference in the type of injectisome encoded by the respective genomes, P. vermicola might operate a fundamentally different mechanism of entomopathogenicity when compared to insect-pathogenic Providencia sneebia or Providencia burhodogranariea. The complete absence of antibiotic resistance gene carrying plasmids or mobile genetic elements as those causing multi drug resistance phenomena in clinical Providencia strains, is consistent with the invertebrate pathogen P. vermicola being in its natural environment efficiently excluded from the propagation routes of multidrug resistance (MDR) carrying genetic elements operating between human pathogens. Susceptibility to MDR plasmid acquisition will likely become a major criterion in the evaluation of P. vermicola for potential applications in biological pest control. Supplementary Information The online version contains supplementary material available at 10.1186/s12864-021-08027-w.
Collapse
Affiliation(s)
- Giuseppe Andolfo
- Department of Agricultural Sciences, University of Naples "Federico II", Via Università 100, 80055, Portici, Italy.
| | - Christina Schuster
- Department of Biology, Technische Universität Darmstadt, Schnittspahnstraße 10, 64287, Darmstadt, Germany
| | - Haifa Ben Gharsa
- Department of Biology, Technische Universität Darmstadt, Schnittspahnstraße 10, 64287, Darmstadt, Germany
| | - Michelina Ruocco
- Istituto per la Protezione Sostenibile delle Piante (IPSP), Consiglio Nazionale delle Ricerche (CNR), Piazzale Enrico Fermi 1, 80055, Portici, Italy
| | - Andreas Leclerque
- Department of Biology, Technische Universität Darmstadt, Schnittspahnstraße 10, 64287, Darmstadt, Germany. .,Istituto per la Protezione Sostenibile delle Piante (IPSP), Consiglio Nazionale delle Ricerche (CNR), Piazzale Enrico Fermi 1, 80055, Portici, Italy.
| |
Collapse
|
25
|
Crowe HM, Wesselink AK, Wise LA, Wang TR, Horsburgh CR, Mikkelsen EM, Hatch EE. Antibiotics and fecundability among female pregnancy planners: a prospective cohort study. Hum Reprod 2021; 36:2761-2768. [PMID: 34269389 DOI: 10.1093/humrep/deab173] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/22/2021] [Indexed: 11/13/2022] Open
Abstract
STUDY QUESTION To what extent is female preconception antibiotic use associated with fecundability? SUMMARY ANSWER Preconception antibiotic use overall was not appreciably associated with fecundability. WHAT IS KNOWN ALREADY Antibiotics are commonly used by women and are generally thought to be safe for use during pregnancy. However, little is known about possible effects of antibiotic use on fecundability, the per-cycle probability of conception. Previous research on this question has been limited to occupational rather than therapeutic exposure. STUDY DESIGN, SIZE, DURATION We analyzed data from an Internet-based preconception cohort study of 9524 female pregnancy planners aged 21-45 years residing in the USA and Canada who had been attempting to conceive for six or fewer cycles at study entry. Participants enrolled between June 2013 and September 2020 and completed baseline and bimonthly follow-up questionnaires for up to 12 months or until a reported pregnancy, whichever came first. The questions pertaining to antibiotic type and indication were added to the PRESTO questionnaires in March 2016. PARTICIPANTS/MATERIALS, SETTING, METHODS We assessed antibiotic use in the previous 4 weeks at baseline and on each follow-up questionnaire. Participants provided the name of the specific antibiotic and the indication for use. Antibiotics were classified based on active ingredient (penicillins, macrolides, nitrofurantoin, nitroimidazole, cephalosporins, sulfonamides, quinolones, tetracyclines, lincosamides), and indications were classified by type of infection (respiratory, urinary tract, skin, vaginal, pelvic, and surgical). Participants reported pregnancy status on follow-up questionnaires. We used proportional probabilities regression to estimate fecundability ratios (FR), the per-cycle probability of conception comparing exposed with unexposed individuals, and 95% CI, adjusting for sociodemographics, lifestyle factors, and reproductive history. MAIN RESULTS AND THE ROLE OF CHANCE Overall, women who used antibiotics in the past 4 weeks at baseline had similar fecundability to those who had not used antibiotics (FR: 0.98, 95% CI: 0.89-1.07). Sulfonamides and lincosamides were associated with slightly increased fecundability (FR: 1.39, 95% CI: 0.90-2.15, and FR: 1.58 95% CI: 0.96-2.60, respectively), while macrolides were associated with slightly reduced fecundability (FR: 0.70, 95% CI: 0.47-1.04). Analyses of the indication for antibiotic use suggest that there is likely some confounding by indication. LIMITATIONS, REASONS FOR CAUTION Findings were imprecise for some antibiotic classes and indications for use owing to small numbers of antibiotic users in these categories. There are likely heterogeneous effects of different combinations of indications and treatments, which may be obscured in the overall null results, but cannot be further elucidated in this analysis. WIDER IMPLICATIONS OF THE FINDINGS There is little evidence that most antibiotics are associated with reduced fecundability. Antibiotics and the infections they treat are likely associated with fecundability through differing mechanisms, resulting in their association with increased fecundability in some circumstances and decreased fecundability in others. STUDY FUNDING/COMPETING INTEREST(S) This study was supported through funds provided by the National Institute of Child Health and Human Development, National Institutes of Health (R01-HD086742, R21-HD072326). L.A.W. has received in-kind donations from Swiss Precision Diagnostics, Sandstone Diagnostics, Fertility Friend, and Kindara for primary data collection in PRESTO. The other authors have no conflicts of interest to disclose. TRIAL REGISTRATION NUMBER N/A.
Collapse
Affiliation(s)
- Holly Michelle Crowe
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Amelia Kent Wesselink
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Lauren Anne Wise
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | - Tanran R Wang
- Department of Epidemiology, Boston University School of Public Health, Boston, MA, USA
| | | | - Ellen Margrethe Mikkelsen
- Department of Clinical Epidemiology, Aarhus University Hospital, Aarhus N, Denmark.,Department of Clinical Medicine, Aarhus University Hospital, Aarhus N, Denmark
| | | |
Collapse
|
26
|
Seabra G, Ventura Mendes RF, Dos Santos Amorim LFV, Peregrino IV, Branquinha MH, Dos Santos ALS, Nunes APF. Azithromycin Use in COVID-19 Patients: Implications on the Antimicrobial Resistance. Curr Top Med Chem 2021; 21:677-683. [PMID: 34028347 DOI: 10.2174/156802662108210319145317] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Gabriela Seabra
- Laboratorio de Resistencia Bacteriana (RESBAC), Departamento de Microbiologia, Centro de Ciencias da Saude (CCS), Universidade Federal do Espirito Santo (UFES), Vitoria, Brazil
| | - Roberta Ferreira Ventura Mendes
- Laboratorio de Resistencia Bacteriana (RESBAC), Departamento de Microbiologia, Centro de Ciencias da Saude (CCS), Universidade Federal do Espirito Santo (UFES), Vitoria, Brazil
| | - Luiz Felipe Vieira Dos Santos Amorim
- Laboratorio de Resistencia Bacteriana (RESBAC), Departamento de Microbiologia, Centro de Ciencias da Saude (CCS), Universidade Federal do Espirito Santo (UFES), Vitoria, Brazil
| | - Ingrid Vianez Peregrino
- Laboratorio de Resistencia Bacteriana (RESBAC), Departamento de Microbiologia, Centro de Ciencias da Saude (CCS), Universidade Federal do Espirito Santo (UFES), Vitoria, Brazil
| | - Marta Helena Branquinha
- Laboratorio de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - André Luis Souza Dos Santos
- Laboratorio de Estudos Avancados de Microrganismos Emergentes e Resistentes (LEAMER), Departamento de Microbiologia Geral, Instituto de Microbiologia Paulo de Goes (IMPG), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, Brazil
| | - Ana Paula Ferreira Nunes
- Laboratorio de Resistencia Bacteriana (RESBAC), Departamento de Microbiologia, Centro de Ciencias da Saude (CCS), Universidade Federal do Espirito Santo (UFES), Vitoria, Brazil
| |
Collapse
|
27
|
Liu J, Chen F, Wang X, Peng H, Zhang H, Wang KJ. The Synergistic Effect of Mud Crab Antimicrobial Peptides Sphistin and Sph 12-38 With Antibiotics Azithromycin and Rifampicin Enhances Bactericidal Activity Against Pseudomonas Aeruginosa. Front Cell Infect Microbiol 2020; 10:572849. [PMID: 33194811 PMCID: PMC7645104 DOI: 10.3389/fcimb.2020.572849] [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: 06/15/2020] [Accepted: 09/02/2020] [Indexed: 12/16/2022] Open
Abstract
Overuse or abuse of antibiotics has undoubtedly accelerated the increasing prevalence of global antibiotic resistance crisis, and thus, people have been trying to explore approaches to decrease dosage of antibiotics or find new antibacterial agents for many years. Antimicrobial peptides (AMPs) are the ideal candidates that could kill pathogens and multidrug-resistant bacteria either alone or in combination with conventional antibiotics. In the study, the antimicrobial efficacy of mud crab Scylla paramamosain AMPs Sphistin and Sph12−38 in combination with eight selected antibiotics was evaluated using a clinical pathogen, Pseudomonas aeruginosa. It was interesting to note that the in vitro combination of rifampicin and azithromycin with Sphistin and Sph12−38 showed significant synergistic activity against P. aeruginosa. Moreover, an in vivo study was carried out using a mouse model challenged with P. aeruginosa, and the result showed that the combination of Sph12−38 with either rifampicin or azithromycin could significantly promote the healing of wounds and had the healing time shortened to 4–5 days compared with 7–8 days in control. The underlying mechanism might be due to the binding of Sphistin and Sph12−38 with P. aeruginosa lipopolysaccharides (LPS) and subsequent promotion of the intracellular uptake of rifampicin and azithromycin. Taken together, the significant synergistic antibacterial effect on P. aeruginosa in vitro and in vivo conferred by the combination of low dose of Sphistin and Sph12−38 with low dose of rifampicin and azithromycin would be beneficial for the control of antibiotic resistance and effective treatment of P. aeruginosa-infected diseases in the future.
Collapse
Affiliation(s)
- Jie Liu
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| | - Fangyi Chen
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| | - Xiaofei Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| | - Hui Peng
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| | - Hua Zhang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| | - Ke-Jian Wang
- State Key Laboratory of Marine Environmental Science, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China.,State-Province Joint Engineering Laboratory of Marine Bioproducts and Technology, College of Ocean & Earth Sciences, Xiamen University, Xiamen, China
| |
Collapse
|
28
|
Abstract
Streptogramins are antibiotics produced by several species of Streptomyces bacteria that are used in both human and veterinary medicine. Group A streptogramins comprise 23-membered macrocyclic polyketide/nonribosomal peptide hybrids for which several innovative, fully synthetic routes have been developed. Herein we describe in detail our scalable routes to natural group A streptogramins and compare these routes to other reported syntheses.
Collapse
Affiliation(s)
- Qi Li
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California 94158, United States
| | - Ian B Seiple
- Department of Pharmaceutical Chemistry and Cardiovascular Research Institute, University of California, San Francisco, San Francisco, California 94158, United States
| |
Collapse
|
29
|
Synthetic group A streptogramin antibiotics that overcome Vat resistance. Nature 2020; 586:145-150. [PMID: 32968273 PMCID: PMC7546582 DOI: 10.1038/s41586-020-2761-3] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 08/19/2020] [Indexed: 11/08/2022]
Abstract
Natural products serve as chemical blueprints for the majority of antibiotics in our clinical arsenal. The evolutionary process by which these molecules arise is inherently accompanied by the co-evolution of resistance mechanisms that shorten the clinical lifetime of any given class1. Virginiamycin acetyltransferases (Vats) are resistance proteins that provide protection against streptogramins2, potent Gram-positive antibiotics that inhibit the bacterial ribosome3. Due to the challenge of selectively modifying the chemically complex, 23-membered macrocyclic scaffold of group A streptogramins, analogs that overcome Vat resistance have not been previously accessed2. Here we report the design, synthesis, and antibacterial evaluation of group A streptogramin antibiotics with unprecedented structural variability. Using cryo-electron microscopy and forcefield-based refinement, we characterize the binding of eight analogs to the bacterial ribosome at high resolution, revealing new binding interactions that extend into the peptidyl tRNA binding site and towards synergistic binders that occupy the nascent peptide exit tunnel (NPET). One of these analogs has excellent activity against several streptogramin-resistant strains of S. aureus, exhibits decreased acetylation rates in vitro, and is effective at lowering bacterial load in a mouse model of infection. Our results demonstrate that the combination of rational design and modular chemical synthesis can revitalize classes of antibiotics that are limited by naturally arising resistance mechanisms.
Collapse
|
30
|
Djamin RS, Talman S, Schrauwen EJA, von Wintersdorff CJH, Wolffs PF, Savelkoul PHM, Uzun S, Kerstens R, van der Eerden MM, Kluytmans JAJW. Prevalence and abundance of selected genes conferring macrolide resistance genes in COPD patients during maintenance treatment with azithromycin. Antimicrob Resist Infect Control 2020; 9:116. [PMID: 32723393 PMCID: PMC7389634 DOI: 10.1186/s13756-020-00783-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/14/2020] [Indexed: 11/16/2022] Open
Abstract
Objectives Maintenance treatment with macrolide antibiotics has shown to be effective in reducing exacerbations in COPD patients. A major concern with prolonged treatment with antibiotics is the development of bacterial resistance. In this study we determined the effect of azithromycin on the development and acquisition of resistance to macrolides in the nasopharyngeal flora in COPD patients. Methods This study was part of the COLUMBUS trial, a randomised, double-blind, placebo-controlled trial to measure the effect of maintenance treatment with azithromycin in 92 COPD patients on the exacerbation rates during a 12-month period. In order to determine resistance to macrolides, we used a targeted metagenomic approach to measure the presence and relative abundance of specific macrolide resistance genes ermB, ermF and mefA in throat samples collected at different time-points during this 12-month period. Results There was no increased risk for acquisition of macrolide resistance genes in the azithromycin group compared to the placebo group in COPD patients. However, loss of the macrolide resistance gene ermB was increased overtime in the placebo treated group compared to the azithromycin group (n = 5 for the placebo group versus n = 0 for the azithromycin group at 12 months; p = 0.012). The change in relative abundance of the three macrolide-resistance genes showed that all but one (ermF) increased during treatment with azithromycin. Conclusions The acquisition rate of macrolide resistance genes in COPD patients treated with azithromycin maintenance therapy was limited, but the relative abundance of macrolide resistance genes increased significantly over time compared to placebo. This study was part of the COLUMBUS trial (Clinicaltrials.gov, NCT00985244).
Collapse
Affiliation(s)
- Remco S Djamin
- Department t of Respiratory Medicine, Amphia Hospital, Molengracht 21, 4818 CK, Breda, The Netherlands
| | - Sander Talman
- Department t of Respiratory Medicine, Amphia Hospital, Molengracht 21, 4818 CK, Breda, The Netherlands.
| | - Eefje J A Schrauwen
- Laboratory for Microbiology and Infection Control, Amphia Hospital, Breda, The Netherlands.,Academy for Technology and Environmental Health, Avans University of Applied Sciences, Breda, the Netherlands
| | | | - Petra F Wolffs
- Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Paul H M Savelkoul
- Department of Medical Microbiology, Maastricht University Medical Center+, Maastricht, The Netherlands
| | - Sevim Uzun
- Department t of Respiratory Medicine, Amphia Hospital, Molengracht 21, 4818 CK, Breda, The Netherlands
| | - René Kerstens
- Orion Statistical Consulting BV, Hilvarenbeek, The Netherlands
| | | | - Jan A J W Kluytmans
- Laboratory for Microbiology and Infection Control, Amphia Hospital, Breda, The Netherlands.,Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, The Netherlands
| |
Collapse
|
31
|
Nahm JH, Kim WK, Kwon Y, Kim H. Detection of Helicobacter pylori with clarithromycin resistance-associated mutations using peptide nucleic acid probe-based melting point analysis. Helicobacter 2019; 24:e12634. [PMID: 31304664 DOI: 10.1111/hel.12634] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 06/12/2019] [Accepted: 06/13/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND Detection of Helicobacter pylori in gastric biopsy is important for appropriate treatment and prevention of gastric carcinoma and lymphoma. A novel peptide nucleic acid probe (PNA)-based real-time polymerase chain reaction (PCR) method was developed for detection of H pylori and A2142G/A2143G mutation of the 23S rRNA gene, which is associated with clarithromycin resistance. METHODS To evaluate the performance of the PNA probe-based PCR method, a total of 409 gastric biopsy samples were analyzed by PNA probe-based PCR and compared with other H pylori detection methods, including hematoxylin and eosin (HE) and Warthin-Starry (WS) staining, immunohistochemistry (IHC). A2142G/A2143G mutation of the 23S rRNA gene was tested by dual priming oligonucleotide (DPO)-based PCR and Sanger sequencing to evaluate PNA probe-based PCR. RESULTS Among 271 cases that were positive for H pylori on IHC which was considered as a standard method, 264 cases (97.4%) and 259 cases (95.6%) were positively detected by HE/WS and PNA probe-based qPCR, respectively. Of 100 H pylori-positive patients tested by IHC, H pylori was detected in 93 cases (93.0%) by PNA probe-based PCR, 86 cases (86.0%) by DPO-based PCR, and 93 cases (93.0%) by conventional PCR. The sensitivity, specificity, accuracy, positive predictive value, and negative predictive value of PNA probe-based qPCR were 93.0%, 94.9%, 93.9%, 94.9%, and 93.0%, respectively, which were all higher than those of DPO-based PCR. When Sanger sequencing was determined as a standard method to detect A2142G/A2143G mutations, the sensitivity of the PNA- and DPO-based methods was 100% and 94.4%, respectively, and the specificity was 100% for both methods. CONCLUSION PNA probe-based qPCR is an appropriate method for detecting H pylori and the clarithromycin resistance-associated mutation type.
Collapse
Affiliation(s)
- Ji Hae Nahm
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| | - Won Kyu Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Projects for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Yujin Kwon
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea.,Brain Korea 21 PLUS Projects for Medical Science, Yonsei University College of Medicine, Seoul, South Korea
| | - Hyunki Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
32
|
Lopatto E, Choi J, Colina A, Ma L, Howe A, Hinsa-Leasure S. Characterizing the soil microbiome and quantifying antibiotic resistance gene dynamics in agricultural soil following swine CAFO manure application. PLoS One 2019; 14:e0220770. [PMID: 31425534 PMCID: PMC6699696 DOI: 10.1371/journal.pone.0220770] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Accepted: 07/23/2019] [Indexed: 12/16/2022] Open
Abstract
As agriculture industrializes, concentrated animal feeding operations (CAFOs) are becoming more common. Feces from CAFOs is often used as fertilizer on fields. However, little is known about the effects manure has on the soil microbiome, which is an important aspect of soil health and fertility. In addition, due to the subtherapeutic levels of antibiotics necessary to keep the animals healthy, CAFO manure has elevated levels of antibiotic resistant bacteria. Using 16s rRNA high-throughput sequencing and qPCR, this study sought to determine the impact of swine CAFO manure application on both the soil microbiome and abundance of select antibiotic resistance genes (ARGs) and mobile element genes (erm(B), erm(C), sul1, str(B), intI1, IncW repA) in agricultural soil over the fall and spring seasons. We found the manure community to be distinct from the soil community, with a majority of bacteria belonging to Bacteroidetes and Firmicutes. The soil samples had more diverse communities dominated by Acidobacteria, Actinobacteria, Proteobacteria, Verrucomicrobia, and unclassified bacteria. We observed significant differences in the soil microbiome between all time points, except between the spring samples. However, by tracking manure associated taxa, we found the addition of the manure microbiome to be a minor driver of the shift. Of the measured genes, manure application only significantly increased the abundance of erm(B) and erm(C) which remained elevated in the spring. These results suggest bacteria in the manure do not survive well in soil and that ARG dynamics in soil following manure application vary by resistance gene.
Collapse
Affiliation(s)
- Edward Lopatto
- Department of Biology, Grinnell College, Grinnell, Iowa, United States of America
| | - Jinlyung Choi
- Department of Agricultural and Biosystems Engineering, Iowa State University, Ames, Iowa, United States of America
| | - Alfredo Colina
- Department of Biology, Grinnell College, Grinnell, Iowa, United States of America
| | - Lanying Ma
- Department of Agricultural and Biosystems Engineering, Iowa State University, Ames, Iowa, United States of America
| | - Adina Howe
- Department of Agricultural and Biosystems Engineering, Iowa State University, Ames, Iowa, United States of America
| | - Shannon Hinsa-Leasure
- Department of Biology, Grinnell College, Grinnell, Iowa, United States of America
- * E-mail:
| |
Collapse
|
33
|
Ding W, Baumdicker F, Neher RA. panX: pan-genome analysis and exploration. Nucleic Acids Res 2019; 46:e5. [PMID: 29077859 PMCID: PMC5758898 DOI: 10.1093/nar/gkx977] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2016] [Accepted: 10/10/2017] [Indexed: 11/24/2022] Open
Abstract
Horizontal transfer, gene loss, and duplication result in dynamic bacterial genomes shaped by a complex mixture of different modes of evolution. Closely related strains can differ in the presence or absence of many genes, and the total number of distinct genes found in a set of related isolates—the pan-genome—is often many times larger than the genome of individual isolates. We have developed a pipeline that efficiently identifies orthologous gene clusters in the pan-genome. This pipeline is coupled to a powerful yet easy-to-use web-based visualization for interactive exploration of the pan-genome. The visualization consists of connected components that allow rapid filtering and searching of genes and inspection of their evolutionary history. For each gene cluster, panX displays an alignment, a phylogenetic tree, maps mutations within that cluster to the branches of the tree and infers gain and loss of genes on the core-genome phylogeny. PanX is available at pangenome.de. Custom pan-genomes can be visualized either using a web server or by serving panX locally as a browser-based application.
Collapse
Affiliation(s)
- Wei Ding
- Max Planck Institute for Developmental Biology, 72076 Tübingen, Germany
| | - Franz Baumdicker
- Mathematisches Institut, Albert-Ludwigs University of Freiburg, 79104 Freiburg, Germany
| | - Richard A Neher
- Max Planck Institute for Developmental Biology, 72076 Tübingen, Germany.,Biozentrum and SIB Swiss Institute of Bioinformatics, University of Basel, 4056 Basel, Switzerland
| |
Collapse
|
34
|
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: 98] [Impact Index Per Article: 16.3] [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
|
35
|
Mikhaleva TV, Zakharova OI, Ilyasov PV. Antibiotic Resistance: Modern Approaches and Ways to Overcome It (Review). APPL BIOCHEM MICRO+ 2019. [DOI: 10.1134/s000368381902011x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
36
|
Intracellular Penetration and Effects of Antibiotics on Staphylococcus aureus Inside Human Neutrophils: A Comprehensive Review. Antibiotics (Basel) 2019; 8:antibiotics8020054. [PMID: 31060222 PMCID: PMC6628357 DOI: 10.3390/antibiotics8020054] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2019] [Revised: 04/25/2019] [Accepted: 05/02/2019] [Indexed: 12/13/2022] Open
Abstract
Neutrophils are important assets in defense against invading bacteria like staphylococci. However, (dysfunctioning) neutrophils can also serve as reservoir for pathogens that are able to survive inside the cellular environment. Staphylococcus aureus is a notorious facultative intracellular pathogen. Most vulnerable for neutrophil dysfunction and intracellular infection are immune-deficient patients or, as has recently been described, severely injured patients. These dysfunctional neutrophils can become hide-out spots or “Trojan horses” for S. aureus. This location offers protection to bacteria from most antibiotics and allows transportation of bacteria throughout the body inside moving neutrophils. When neutrophils die, these bacteria are released at different locations. In this review, we therefore focus on the capacity of several groups of antibiotics to enter human neutrophils, kill intracellular S. aureus and affect neutrophil function. We provide an overview of intracellular capacity of available antibiotics to aid in clinical decision making. In conclusion, quinolones, rifamycins and sulfamethoxazole-trimethoprim seem very effective against intracellular S. aureus in human neutrophils. Oxazolidinones, macrolides and lincosamides also exert intracellular antibiotic activity. Despite that the reviewed data are predominantly of in vitro origin, these findings should be taken into account when intracellular infection is suspected, as can be the case in severely injured patients.
Collapse
|
37
|
Zarei-Baygi A, Harb M, Wang P, Stadler LB, Smith AL. Evaluating Antibiotic Resistance Gene Correlations with Antibiotic Exposure Conditions in Anaerobic Membrane Bioreactors. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:3599-3609. [PMID: 30810034 DOI: 10.1021/acs.est.9b00798] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Anaerobic membrane bioreactors (AnMBRs) are an emerging technology with potential to improve energy efficiency and effluent reuse in mainstream wastewater treatment. However, their contribution to the proliferation of contaminants of emerging concern, such as antibiotic resistance genes (ARGs), remains largely unknown. The purpose of this study was to determine the effect of select influent antibiotics at varying concentrations on the presence and abundance of ARGs in an AnMBR system and its effluent. Quantification of targeted ARGs revealed distinct profiles in biomass and effluent, with genes conferring resistance to different antibiotic classes dominating in biomass (macrolides) and effluent (sulfonamides). Effluent sul1 gene abundance was strongly correlated with abundance of intl1, signifying the potential importance of mobile genetic elements in ARG release from AnMBR systems. The addition of specific antibiotics also affected normalized abundances of their related ARGs, exemplifying the potential impact of selective pressures at both low (10 μg/L) and high (250 μg/L) influent antibiotic concentrations.
Collapse
Affiliation(s)
- Ali Zarei-Baygi
- Astani Department of Civil and Environmental Engineering , University of Southern California , 3620 South Vermont Avenue , Los Angeles , California 90089 , United States
| | - Moustapha Harb
- Astani Department of Civil and Environmental Engineering , University of Southern California , 3620 South Vermont Avenue , Los Angeles , California 90089 , United States
| | - Phillip Wang
- Astani Department of Civil and Environmental Engineering , University of Southern California , 3620 South Vermont Avenue , Los Angeles , California 90089 , United States
| | - Lauren B Stadler
- Department of Civil and Environmental Engineering , Rice University , 6100 Main Street , Houston , Texas 77005 , United States
| | - Adam L Smith
- Astani Department of Civil and Environmental Engineering , University of Southern California , 3620 South Vermont Avenue , Los Angeles , California 90089 , United States
| |
Collapse
|
38
|
Ardon CB, Prens EP, Tkadlec J, Fuursted K, Abourayale S, Jemec GBE, Jenssen H. Virulent Staphylococcus lugdunensis with limited genetic diversity in hidradenitis suppurativa lesions. J Eur Acad Dermatol Venereol 2019; 33:e248-e250. [PMID: 30801776 DOI: 10.1111/jdv.15523] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- C B Ardon
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands.,Department of Science and Environment, Roskilde University, Universitetsvej 1, Roskilde, DK-4000, Denmark
| | - E P Prens
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam, Dr. Molewaterplein 40, Rotterdam, 3015 GD, The Netherlands
| | - J Tkadlec
- Department of Medical Microbiology, 2nd Faculty of Medicine, Charles University and Motol University Hospital, Prague, V Úvalu 84, 150 06 Praha 5, Czech Republic
| | - K Fuursted
- Department of Microbiology and Infection Control, Staten Serum Institute, 5 Artillerivej, DK-2300, Copenhagen, Denmark
| | - S Abourayale
- Department of Science and Environment, Roskilde University, Universitetsvej 1, Roskilde, DK-4000, Denmark
| | - G B E Jemec
- Department of Dermatology, University Hospital Zealand, Sygehusvej 10, 4000 Roskilde, Denmark
| | - H Jenssen
- Department of Science and Environment, Roskilde University, Universitetsvej 1, Roskilde, DK-4000, Denmark
| |
Collapse
|
39
|
Choi J, Rieke EL, Moorman TB, Soupir ML, Allen HK, Smith SD, Howe A. Practical implications of erythromycin resistance gene diversity on surveillance and monitoring of resistance. FEMS Microbiol Ecol 2019; 94:4810543. [PMID: 29346541 PMCID: PMC5939627 DOI: 10.1093/femsec/fiy006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/12/2018] [Indexed: 12/29/2022] Open
Abstract
Use of antibiotics in human and animal medicine has applied selective pressure for the global dissemination of antibiotic-resistant bacteria. Therefore, it is of interest to develop strategies to mitigate the continued amplification and transmission of resistance genes in environmental reservoirs such as farms, hospitals and watersheds. However, the efficacy of mitigation strategies is difficult to evaluate because it is unclear which resistance genes are important to monitor, and which primers to use to detect those genes. Here, we evaluated the diversity of one type of macrolide antibiotic resistance gene (erm) in one type of environment (manure) to determine which primers would be most informative to use in a mitigation study of that environment. We analyzed all known erm genes and assessed the ability of previously published erm primers to detect the diversity. The results showed that all known erm resistance genes group into 66 clusters, and 25 of these clusters (40%) can be targeted with primers found in the literature. These primers can target 74%–85% of the erm gene diversity in the manures analyzed.
Collapse
Affiliation(s)
- Jinlyung Choi
- Department of Agricultural and Biosystems Engineering, Iowa State University, 1201 Sukup Hall, Ames, IA 50011, USA
| | - Elizabeth L Rieke
- Department of Agricultural and Biosystems Engineering, Iowa State University, 1201 Sukup Hall, Ames, IA 50011, USA
| | - Thomas B Moorman
- National Laboratory for Agriculture and the Environment, USDA-ARS, 2110 University Blvd, Ames, IA 50011, USA
| | - Michelle L Soupir
- Department of Agricultural and Biosystems Engineering, Iowa State University, 1201 Sukup Hall, Ames, IA 50011, USA
| | - Heather K Allen
- Food Safety and Enteric Pathogens Research Unit, National Animal Disease Center, USDA-ARS, 1920 Dayton Ave, Ames, IA, 50010, USA
| | - Schuyler D Smith
- Department of Bioinformatics and Computational Biology, Iowa State University, 2014 Molecular Biology Building, Ames, IA 50011, USA
| | - Adina Howe
- Department of Agricultural and Biosystems Engineering, Iowa State University, 1201 Sukup Hall, Ames, IA 50011, USA
| |
Collapse
|
40
|
[Antibiotic prophylaxis in preterm premature rupture of membranes: CNGOF preterm premature rupture of membranes guidelines]. ACTA ACUST UNITED AC 2018; 46:1043-1053. [PMID: 30392988 DOI: 10.1016/j.gofs.2018.10.017] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To analyse benefits and risks of antibiotic prophylaxis in the management of preterm premature rupture of membranes. METHODS PubMed and Cochrane Central databases search. RESULTS Streptoccoccus agalactiae (group B streptococcus) and Escherichia coli are the two main bacteria identified in early neonatal sepsis (EL3). Antibiotic prophylaxis at admission is associated with significant prolongation of pregnancy (EL2), reduction in neonatal morbidity (EL1) without impact on neonatal mortality (EL2). Co-amoxiclav could be associated with an increased risk for neonatal necrotising enterocolitis (EL2). Antibiotic prophylaxis at admission in women with preterm premature rupture of the membranes is recommended (Grade A). Monotherapy with amoxicillin, third generation cephalosporin and erythromycin can be used as well as combination of erythromycin and amoxicillin (Professional consensus) for 7 days (GradeC). Shorter treatment is possible when initial vaginal culture is negative (Professional consensus). Co-amxiclav, aminoglycosides, glycopeptides, first and second generation cephalosporin, clindamycin and metronidazole are not recommended (Professional consensus). CONCLUSIONS Antibiotic prophylaxis against Streptoccoccus agalactiae (group B streptococcus) and E. coli is recommended in women with preterm premature of the membranes (Grade A). Monotherapy with amoxicillin, third generation cephalosporin or erythromycin, as well as combination of erythromycin and amoxicillin are recommended (Professional consensus).
Collapse
|
41
|
Mechanism of Macrolide-Induced Inhibition of Pneumolysin Release Involves Impairment of Autolysin Release in Macrolide-Resistant Streptococcus pneumoniae. Antimicrob Agents Chemother 2018; 62:AAC.00161-18. [PMID: 30181369 DOI: 10.1128/aac.00161-18] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 08/27/2018] [Indexed: 11/20/2022] Open
Abstract
Streptococcus pneumoniae is a leading cause of community-acquired pneumonia. Over the past 2 decades, macrolide resistance among S. pneumoniae organisms has been increasing steadily and has escalated at an alarming rate worldwide. However, the use of macrolides in the treatment of community-acquired pneumonia has been reported to be effective regardless of the antibiotic susceptibility of the causative pneumococci. Although previous studies suggested that sub-MICs of macrolides inhibit the production of the pneumococcal pore-forming toxin pneumolysin by macrolide-resistant S. pneumoniae (MRSP), the underlying mechanisms of the inhibitory effect have not been fully elucidated. Here, we show that the release of pneumococcal autolysin, which promotes cell lysis and the release of pneumolysin, was inhibited by treatment with azithromycin and erythromycin, whereas replenishing with recombinant autolysin restored the release of pneumolysin from MRSP. Additionally, macrolides significantly downregulated ply transcription followed by a slight decrease of the intracellular pneumolysin level. These findings suggest the mechanisms involved in the inhibition of pneumolysin in MRSP, which may provide an additional explanation for the benefits of macrolides on the outcome of treatment for pneumococcal diseases.
Collapse
|
42
|
Golkar T, Zieliński M, Berghuis AM. Look and Outlook on Enzyme-Mediated Macrolide Resistance. Front Microbiol 2018; 9:1942. [PMID: 30177927 PMCID: PMC6109786 DOI: 10.3389/fmicb.2018.01942] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2018] [Accepted: 07/31/2018] [Indexed: 01/14/2023] Open
Abstract
Since their discovery in the early 1950s, macrolide antibiotics have been used in both agriculture and medicine. Specifically, macrolides such as erythromycin and azithromycin have found use as substitutes for β-lactam antibiotics in patients with penicillin allergies. Given the extensive use of this class of antibiotics it is no surprise that resistance has spread among pathogenic bacteria. In these bacteria different mechanisms of resistance have been observed. Frequently observed are alterations in the target of macrolides, i.e., the ribosome, as well as upregulation of efflux pumps. However, drug modification is also increasingly observed. Two classes of enzymes have been implicated in macrolide detoxification: macrolide phosphotransferases and macrolide esterases. In this review, we present a comprehensive overview on what is known about macrolide resistance with an emphasis on the macrolide phosphotransferase and esterase enzymes. Furthermore, we explore how this information can assist in addressing resistance to macrolide antibiotics.
Collapse
Affiliation(s)
- Tolou Golkar
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Michał Zieliński
- Department of Biochemistry, McGill University, Montreal, QC, Canada
| | - Albert M Berghuis
- Department of Biochemistry, McGill University, Montreal, QC, Canada.,Department of Microbiology & Immunology, McGill University, Montreal, QC, Canada
| |
Collapse
|
43
|
Iannelli F, Santoro F, Santagati M, Docquier JD, Lazzeri E, Pastore G, Cassone M, Oggioni MR, Rossolini GM, Stefani S, Pozzi G. Type M Resistance to Macrolides Is Due to a Two-Gene Efflux Transport System of the ATP-Binding Cassette (ABC) Superfamily. Front Microbiol 2018; 9:1670. [PMID: 30108557 PMCID: PMC6079230 DOI: 10.3389/fmicb.2018.01670] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 07/04/2018] [Indexed: 11/19/2022] Open
Abstract
The mef(A) gene was originally identified as the resistance determinant responsible for type M resistance to macrolides, a phenotype frequently found in clinical isolates of Streptococcus pneumoniae and Streptococcus pyogenes. MefA was defined as a secondary transporter of the major facilitator superfamily driven by proton-motive force. However, when characterizing the mef(A)-carrying elements Tn1207.1 and Φ1207.3, another macrolide resistance gene, msr(D), was found adjacent to mef(A). To define the respective contribution of mef(A) and msr(D) to macrolide resistance, three isogenic deletion mutants were constructed by transformation of a S. pneumoniae strain carrying Φ1207.3: (i) Δmef(A)–Δmsr(D); (ii) Δmef(A)–msr(D); and (iii) mef(A)–Δmsr(D). Susceptibility testing of mutants clearly showed that msr(D) is required for macrolide resistance, while deletion of mef(A) produced only a twofold reduction in the minimal inhibitory concentration (MIC) for erythromycin. The contribution of msr(D) to macrolide resistance was also studied in S. pyogenes, which is the original host of Φ1207.3. Two isogenic strains of S. pyogenes were constructed: (i) FR156, carrying Φ1207.3, and (ii) FR155, carrying Φ1207.3/Δmsr(D). FR155 was susceptible to erythromycin, whereas FR156 was resistant, with an MIC value of 8 μg/ml. Complementation experiments showed that reintroduction of the msr(D) gene could restore macrolide resistance in Δmsr(D) mutants. Radiolabeled erythromycin was retained by strains lacking msr(D), while msr(D)-carrying strains showed erythromycin efflux. Deletion of mef(A) did not affect erythromycin efflux. This data suggest that type M resistance to macrolides in streptococci is due to an efflux transport system of the ATP-binding cassette (ABC) superfamily, in which mef(A) encodes the transmembrane channel, and msr(D) the two ATP-binding domains.
Collapse
Affiliation(s)
- Francesco Iannelli
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Francesco Santoro
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Maria Santagati
- Section of Microbiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | | | - Elisa Lazzeri
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gabiria Pastore
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Marco Cassone
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Marco R Oggioni
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Gian M Rossolini
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| | - Stefania Stefani
- Section of Microbiology, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Gianni Pozzi
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
44
|
Farrell LJ, Lo R, Wanford JJ, Jenkins A, Maxwell A, Piddock LJV. Revitalizing the drug pipeline: AntibioticDB, an open access database to aid antibacterial research and development. J Antimicrob Chemother 2018; 73:2284-2297. [DOI: 10.1093/jac/dky208] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- L J Farrell
- Institute of Microbiology & Infection, College of Medical & Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - R Lo
- School of Biological Sciences, University of East Anglia, Norwich Research Park, Norwich NR4 7TJ, UK
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, UK
| | - J J Wanford
- Institute of Microbiology & Infection, College of Medical & Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| | - A Jenkins
- British Society for Antimicrobial Chemotherapy, Griffin House, 53 Regent Place, Birmingham B1 3NJ, UK
| | - A Maxwell
- Department of Biological Chemistry, John Innes Centre, Norwich Research Park, Norwich NR4 7UH, UK
| | - L J V Piddock
- Institute of Microbiology & Infection, College of Medical & Dental Sciences, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
45
|
Ficellomycin: an aziridine alkaloid antibiotic with potential therapeutic capacity. Appl Microbiol Biotechnol 2018; 102:4345-4354. [DOI: 10.1007/s00253-018-8934-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 03/09/2018] [Accepted: 03/13/2018] [Indexed: 10/17/2022]
|
46
|
Shaw TD, Fairley DJ, Schneiders T, Pathiraja M, Hill RLR, Werner G, Elborn JS, McMullan R. The use of high-throughput sequencing to investigate an outbreak of glycopeptide-resistant Enterococcus faecium with a novel quinupristin-dalfopristin resistance mechanism. Eur J Clin Microbiol Infect Dis 2018; 37:959-967. [PMID: 29478197 PMCID: PMC5916979 DOI: 10.1007/s10096-018-3214-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 02/13/2018] [Indexed: 12/01/2022]
Abstract
High-throughput sequencing (HTS) has successfully identified novel resistance genes in enterococci and determined clonal relatedness in outbreak analysis. We report the use of HTS to investigate two concurrent outbreaks of glycopeptide-resistant Enterococcus faecium (GRE) with an uncharacterised resistance mechanism to quinupristin-dalfopristin (QD). Seven QD-resistant and five QD-susceptible GRE isolates from a two-centre outbreak were studied. HTS was performed to identify genes or predicted proteins that were associated with the QD-resistant phenotype. MLST and SNP typing on HTS data was used to determine clonal relatedness. Comparative genomic analysis confirmed this GRE outbreak involved two distinct clones (ST80 and ST192). HTS confirmed the absence of known QD resistance genes, suggesting a novel mechanism was conferring resistance. Genomic analysis identified two significant genetic determinants with explanatory power for the high level of QD resistance in the ST80 QD-resistant clone: an additional 56aa leader sequence at the N-terminus of the lsaE gene and a transposon containing seven genes encoding proteins with possible drug or drug-target modification activities. However, HTS was unable to conclusively determine the QD resistance mechanism and did not reveal any genetic basis for QD resistance in the ST192 clone. This study highlights the usefulness of HTS in deciphering the degree of relatedness in two concurrent GRE outbreaks. Although HTS was able to reveal some genetic candidates for uncharacterised QD resistance, this study demonstrates the limitations of HTS as a tool for identifying putative determinants of resistance to QD.
Collapse
Affiliation(s)
- Timothy D Shaw
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK. .,Department of Medical Microbiology, Kelvin Laboratory, Royal Victoria Hospital, Belfast, Northern Ireland, UK.
| | - D J Fairley
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK.,Department of Medical Microbiology, Kelvin Laboratory, Royal Victoria Hospital, Belfast, Northern Ireland, UK
| | - T Schneiders
- Division of Infection and Pathway Medicine, University of Edinburgh Medical School, Edinburgh, Scotland, UK
| | - M Pathiraja
- Department of Medical Microbiology, Kelvin Laboratory, Royal Victoria Hospital, Belfast, Northern Ireland, UK
| | - R L R Hill
- Antimicrobial Resistance & Healthcare-Associated Infection, Public Health England, London, UK
| | - G Werner
- National Reference Centre for Staphylococci and Enterococci, Robert Koch-Institute, Wernigerode Branch, Berlin, Germany
| | - J S Elborn
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK.,National Heart and Lung Institute, Imperial College London, London, UK
| | - R McMullan
- Centre for Experimental Medicine, Queen's University Belfast, Belfast, Northern Ireland, UK.,Department of Medical Microbiology, Kelvin Laboratory, Royal Victoria Hospital, Belfast, Northern Ireland, UK
| |
Collapse
|
47
|
Watteyn A, Devreese M, Plessers E, Wyns H, Garmyn A, Reddy VRAP, Pasmans F, Martel A, Haesebrouck F, De Backer P, Croubels S. Efficacy of gamithromycin against Ornithobacterium rhinotracheale in turkey poults pre-infected with avian metapneumovirus. Avian Pathol 2018; 45:545-51. [PMID: 27171615 DOI: 10.1080/03079457.2016.1183764] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Ornithobacterium rhinotracheale is an avian respiratory pathogen that affects turkeys. The objective of this study was to evaluate the clinical efficacy of gamithromycin (GAM) against O. rhinotracheale in turkeys. The birds were inoculated oculonasally with 10(8) colony-forming units (cfu) of O. rhinotracheale, preceded by infection with avian metapneumovirus. In addition to a negative (CONTR-) and a positive control group (CONTR+) there were two treated groups administered GAM (6 mg/kg) either subcutaneously (GAM SC) or orally (GAM PO) by administration as a single bolus at one-day post-bacterial infection (p.b.i.). From the start of the avian metapneumovirus infection until the end of the experiment, the turkeys were examined clinically and scored daily. In addition, tracheal swabs were collected at several days p.b.i. Necropsy was performed at 4, 8 and 12 days p.b.i. to evaluate the presence of gross lesions, and to collect trachea and lung tissue samples and air sac swabs for O. rhinotracheale quantification. The clinical score of the GAM SC group showed slightly lower values and birds recovered earlier than those in the GAM PO and CONTR+ groups. O. rhinotracheale cfus were significantly reduced in tracheal swabs of the SC group between 2 and 4 days p.b.i. At necropsy, CONTR+ showed higher O. rhinotracheale cfu in lung tissues compared to the treated groups. Moreover, at 8 days p.b.i. only the lung samples of CONTR+ were positive. In conclusion, the efficacy of GAM against O. rhinotracheale was demonstrated, especially in the lung tissue. However, the PO bolus administration of the commercially available product was not as efficacious as the SC bolus.
Collapse
Affiliation(s)
- Anneleen Watteyn
- a Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| | - Mathias Devreese
- a Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| | - Elke Plessers
- a Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| | - Heidi Wyns
- a Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| | - An Garmyn
- b Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| | - Vishwanatha R A P Reddy
- c Department of Virology, Parasitology and Immunology, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| | - Frank Pasmans
- b Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| | - An Martel
- b Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| | - Freddy Haesebrouck
- b Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| | - Patrick De Backer
- a Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| | - Siska Croubels
- a Department of Pharmacology, Toxicology and Biochemistry, Faculty of Veterinary Medicine , Ghent University , Merelbeke , Belgium
| |
Collapse
|
48
|
Rieke EL, Moorman TB, Douglass EL, Soupir ML. Seasonal variation of macrolide resistance gene abundances in the South Fork Iowa River Watershed. THE SCIENCE OF THE TOTAL ENVIRONMENT 2018; 610-611:1173-1179. [PMID: 28847138 DOI: 10.1016/j.scitotenv.2017.08.116] [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: 06/12/2017] [Revised: 08/11/2017] [Accepted: 08/12/2017] [Indexed: 06/07/2023]
Abstract
The Midwestern United States is dominated by agricultural production with high concentrations of swine, leading to application of swine manure onto lands with artificial subsurface drainage. Previous reports have indicated elevated levels of antibiotic resistance genes (ARGs) in surface water and groundwater around confined animal feeding operations which administer antimicrobials. While previous studies have examined the occurrence of ARGs around confined swine feeding operations, little information is known how their transport from tile-drained fields receiving swine manure application impacts downstream environments. To further our knowledge in this area, water samples were collected from five locations in the agriculturally dominated South Fork Iowa River Watershed with approximately 840,000 swine present in the 76,000ha basin. Samples were collected monthly from three stream sites and two main artificial subsurface drainage outlets. Samples were analyzed for macrolide resistance genes ermB, ermF and 16S rRNAgene abundance using qPCR. Abundance of erm genes ranged from below limits of quantification to >107 copies 100mL-1 water. Eighty-nine percent of stream water samples contained one of these two ARGs. Results indicate significantly more ermB and ermF in main drainage outlets than stream samples when normalized by 16S rRNA abundance (p<0.0001). Both artificial drainage locations revealed temporal trends for ermB and ermF abundance when normalized to 16S rRNA abundance. The higher resistance gene concentrations identified in artificial drainage samples occurring mid-Spring and late-Fall are likely due to manure application.
Collapse
Affiliation(s)
- Elizabeth Luby Rieke
- Department of Agricultural and Biosystems Engineering, Iowa State University, Ames, IA, USA.
| | - Thomas B Moorman
- National Laboratory for Agriculture and the Environment, USDA-ARS, Ames, IA, USA
| | - Elizabeth L Douglass
- National Laboratory for Agriculture and the Environment, USDA-ARS, Ames, IA, USA
| | - Michelle L Soupir
- Department of Agricultural and Biosystems Engineering, Iowa State University, Ames, IA, USA
| |
Collapse
|
49
|
van Duijkeren E, Schink AK, Roberts MC, Wang Y, Schwarz S. Mechanisms of Bacterial Resistance to Antimicrobial Agents. Microbiol Spectr 2018; 6:10.1128/microbiolspec.arba-0019-2017. [PMID: 29327680 PMCID: PMC11633570 DOI: 10.1128/microbiolspec.arba-0019-2017] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Indexed: 12/31/2022] Open
Abstract
During the past decades resistance to virtually all antimicrobial agents has been observed in bacteria of animal origin. This chapter describes in detail the mechanisms so far encountered for the various classes of antimicrobial agents. The main mechanisms include enzymatic inactivation by either disintegration or chemical modification of antimicrobial agents, reduced intracellular accumulation by either decreased influx or increased efflux of antimicrobial agents, and modifications at the cellular target sites (i.e., mutational changes, chemical modification, protection, or even replacement of the target sites). Often several mechanisms interact to enhance bacterial resistance to antimicrobial agents. This is a completely revised version of the corresponding chapter in the book Antimicrobial Resistance in Bacteria of Animal Origin published in 2006. New sections have been added for oxazolidinones, polypeptides, mupirocin, ansamycins, fosfomycin, fusidic acid, and streptomycins, and the chapters for the remaining classes of antimicrobial agents have been completely updated to cover the advances in knowledge gained since 2006.
Collapse
Affiliation(s)
- Engeline van Duijkeren
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), 3720 BA Bilthoven, The Netherlands
| | - Anne-Kathrin Schink
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Department of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| | - Marilyn C Roberts
- Department of Environmental and Occupational Health Sciences, University of Washington, Seattle, WA 98195-7234
| | - Yang Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Stefan Schwarz
- Institute of Microbiology and Epizootics, Centre of Infection Medicine, Department of Veterinary Medicine, Freie Universität Berlin, 14163 Berlin, Germany
| |
Collapse
|
50
|
Son MJ, Kim YE, Song YI, Kim YH. Herbal medicines for treating acute otitis media: A systematic review of randomised controlled trials. Complement Ther Med 2017; 35:133-139. [PMID: 29154058 DOI: 10.1016/j.ctim.2017.11.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 10/23/2017] [Accepted: 11/01/2017] [Indexed: 11/29/2022] Open
Abstract
INTRODUCTION This systematic review aimed to assess the clinical evidence for the widespread use of herbal medicines in treating acute otitis media. METHODS Eleven electronic databases, including MEDLINE, EMBASE, and the CENTRAL were searched, without language limitations. All randomised controlled trials involving the use of herbal medicines, alone or in combination with conventional therapies, for acute otitis media were included. RESULTS We identified 4956 studies, of which seven randomised clinical trials met the inclusion criteria. The overall risk of bias of the included trials was relatively high or unclear. Treatment with Longdan-xiegan decoction or Shenling-baizhu powder, combined with antibiotics, appeared to be more effective than treatment with antibiotics alone in terms of the proportion of patients with total symptom recovery. Moreover, combination treatment of Sinupret® and antibiotics facilitated the recovery of middle ear conditions and hearing acuity. CONCLUSIONS Despite some indications of potential symptom improvement, the evidence regarding the effectiveness and efficacy of herbal medicine for acute otitis media is inconclusive due to the poor quality of trials included. Moreover, we only analysed seven trials in this review. Therefore, to properly evaluate the effectiveness of herbal medicine for acute otitis media, systematic reviews based on more rigorously designed randomized trials are warranted in the future.
Collapse
Affiliation(s)
- Mi Ju Son
- Clinical Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Young-Eun Kim
- Mibyeong Research Center, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea
| | - Young Il Song
- Global doctor, Korea international cooperation agency, Gyeonggi-do, Republic of Korea
| | - Yun Hee Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon, Republic of Korea.
| |
Collapse
|