Download (12Mb)

Total Page:16

File Type:pdf, Size:1020Kb

Download (12Mb) A Thesis Submitted for the Degree of PhD at the University of Warwick Permanent WRAP URL: http://wrap.warwick.ac.uk/110352 Copyright and reuse: This thesis is made available online and is protected by original copyright. Please scroll down to view the document itself. Please refer to the repository record for this item for information to help you to cite it. Our policy information is available from the repository home page. For more information, please contact the WRAP Team at: [email protected] warwick.ac.uk/lib-publications THE BRITISH LIBRARY BRITISH THESIS SERVICE THE DISTRIBUTION OF PHENOTYPIC AND GENOTYPIC CHARACTERS WITHIN STREPTOMYCETES AND THEIR RELATIONSHIP TITLE . TO ANTIBIOTIC PRODUCTION. AUTHOR........ Lesley Phillips, DEGREE.................................................... AWARDING BODY _ TI. .. The University of Warwick, THESIS NUMBER THIS THESIS HAS BEEN MICROFILMED EXACTLY AS RECEIVED The quality of this reproduction is dependent upon the quality of the original thesis submitted for microfilming. Every effort has been made to ensure the highest quality of reproduction. Some pages may have indistinct print, especially if the original papers were poorly produced or if the awarding body sent an inferior copy. If pages are missing, please contact the awarding body which granted the degree. Previously copyrighted materials (journal articles, published texts, etc.) are not filmed. This copy of the thesis has been supplied on condition that anyone who consults it is understood to recognise that Its copyright rests with its author and that no information derived from it may be published without the author's prior written consent. Reproduction of this thesis, other than as permitted under the United Kingdom Copyright Designs and Patents Act 1988, or under specific agreement with the copyright holder, is prohibited. 1 1 i T i j •7 Is 1 6 cms REDUCTION X j C A M ERA 3 No. of pages / THE DISTRIBUTION OF PHENOTYPIC AND GENOTYPIC CHARACTERS WITHIN STREPTOMYCETES AND THEIR RELATIONSHIP TO ANTIBIOTIC PRODUCTION. Lesley Phillips, Thesis submitted for the degree of Doctor of Philosophy (PhD), To The University of Warwick, Department of Biological Sciences, April, 1992. I Table of Contents, Page No. Chapter 1. Introduction 1 1.1. The Life-cycle and Ecology of the Streptomycetes. 1 1.2. The Taxonomy of Streptomycetes. 2 1.2.1. The Development of the genus Streptomyces. 2 1.2.2. Streptomycete Classifications Based on Chemotaxonomy. 4 1.2.3. Systematic Methods Used in Taxonomic Studies. 6 1.3. The Biosynthesis, Evolution and Natural Role of 10 Antibiotics in Streptomycetes. 1.3.1. Interactions Between Primary and Secondary 10 Metabolism in Streptomycetes. 1.3.2. The Diversity of Secondary Metabolites from 14 Streptomycetes. 1.3.3. The Evolution o f Antibiotics and its Relationship to 16 Antibiotic Function. 1.3.4. Theories Proposed For The Function of Secondary 18 Metabolites. 1.4. The Regulation and Control of Secondary Metabolism. 23 1.4.1. Carbon Source Regulation. 24 1.4.2. Nitrogen Source Regulation. 23 1.4.3. Regulation by Inorganic Phosphate (Pi). 26 1.4.4. Other Bioregulators. 27 1.4.3. Genetic Control and Organisation in Secondary 28 Metabolism. 1.4.6. Antibiotic Resistance Mechanisms in Streptomycetes. 32 1.4.7. The Relationship o f Antibiotic Resistance and 39 II Table of Contents, Page No. Biosynthesis to Phenotype and Genotype. l.S. The Exploitation of Antibiotics from Streptomycetes. 42 1.5.1. The History of Natural Product Screening. 42 1.5.2. Modern Antibiotic Discovery. 43 1.5.3. The Selection of Organisms for Industrial Screens. 44 1.5.4. The Design o f Media for Antibiotic Screening Programs. 45 1.5.5. Extraction Procedures and Chemical Identification Systems. 47 1.5.6. Target Directed Screening. 48 1.5.7. Screens used in the Agrochemical Industry. 49 1.5.8. Screens used in the Pharmacological Industry. 50 1.5.9. Beneficial Products Derived from the Pharmaceutical and 51 Agrochemical Industries. 1.5.10. Target Directed Isolation and Selection. 52 1.5.11. Other Approaches to Antibiotic Discovery. 53 1.6. Rationale and Aims for this Research. 55 Chapter 2. Materials and Methods. 56 2.1. Materials. 56 2.2. Strain Maintenance. 75 2.2.1. Routine Maintenance of Strains. 75 2.2.2. Culture Storage. 75 2.2.3. Strain Resuscitation. 75 2.2.4. Standardization of Streptomycete Inoculae. 76 2.3. Characterization of Streptomycetes. 76 2.3.1. Extraction of Bioactive Compounds. 76 2.3.2. Thin Layer Chromatography (TLC). 76 III Table of Contents, Page No. 2.3.3. Bioautography. 78 2.3.4. In vivo Agrochemical Screens. 78 2.3.3. Taxonomical Identification. 80 2.3.6. Scanning Electron Microscopy. 84 2.4. Fermentation Work. 86 2.4.1. Comparison of Antibiotic Production on Different Media. 86 2.4.2. Timecourses of Antibiotic Production by D133. 86 2.4.3. Oxygen Limitation Experiment. 87 2.4.4. Dry Weight Measurements. 87 2.4.5. Nutrient Gradients. 87 2.3. Fatty Acid Analysis. 88 2.5.1. Extraction Procedure. 88 2.5.2. Combined Capillary Gas Chromatography Mass. 89 Spectroscopy (GCMS). 2.5.3. Timecourses. 90 2.6. Metabolite Profiles. 90 2.7. Resistance Profiles. 90 2.7.1. The Use of Antibiotic Gradient Plates to Determine 90 Phenetic Resistance in Streptomyces Strains. 2.7.2. Determination of the Relationship Between Antibiotic 92 Concentrations Extrapolated from Gradient Plates and Corresponding Concentrations from Pour Plates. 2.8. DNA Isolation and Probing. 92 2.8.1. Small Scale Alkaline Lysis for Plasmid Preparation of 92 E.coli. 2.8.2. Maxi Preparation of Plasmid from E.coli. 93 2.8.3. Density Gradient Centrifugation. 93 IV Table of Contents. Page No. 2.8.4. Large Scale Isolation of Plasmid DNA from 94 Streptomycetes. 2.8.5. Isolation of Streptomyces " Total" DNA. 94 2.8.6. Phenol Extraction. 95 2.8.7. Quantitation of DNA. 95 2.8.8. Restriction Endonuclease Digestion of DNA. 95 2.8.9. Dot Blots. 96 2.8.12. Agarose Gel Electrophoresis. 96 2.8.13. Ethanol Precipitation. 96 2.8.14. Isopropanol Precipitation. 97 2.8.15. Electroelution. 97 2.8.16. Prehybridization. 97 2.8.17. Preparation of the Probe. 98 2.8.18. Removal of Unincorporated Nucleotides. 98 2.8.19. Hybridization. 99 2.8.20. Stringency. 99 2.8.21. Cerenkov Counting. 99 2.8.22. Detection of Radioactivity. 100 2.9. Cluster Analysis. 100 Chapter 3. The Distribution and Expression of Antibiotic 103 Production in Natural Streptomyces Isolates. 3.1. Introduction. 103 3.2. Bioactivity as Measured under Standard Conditions. 104 3.2.1. Antibacterial Activities. 104 3.2.2. Antifungal Activities. 107 V Table of Contents. Page No. 3.2.3. Specific Biological Activities. 109 3.2.4. Agrochemical Activities. 109 3.3. Factors Affecting the Expression of Bioactivity. 112 3.3.1. Choice of Medium. 112 3.3.2. Effects of Nutrient Gradients on Differentiation. 114 3.3.3. Effects of Nutrient Gradients on Antibiotic 118 Production in a Selection of Streptomycetes. 3.4. The Characterization of Antibiotic Production in D1S3. 121 3.4.1. Timecourses. 121 3.4.2. Repression of Nigericin Biosynthesis. 123 3.3. Discussion. 124 3.3.1. The Distribution of Antibiotic Production in 124 Streptomyces Isolates. 3.3.2. Factors Affecting Differentiation and Antibiotic 127 Production. Chapter 4. The Use of Fatty Add Profiles for Grouping 130 Streptomyces Species. 4.1. Introduction. 130 4.1.2. Choice and Characterization of Strains. 132 4.2. The Distributions of Fatty Acid Methyl Esters Within 133 Strep tomycetes. 4.2.1. The Distribution o f Individual FAMEs and FAME 133 Classes Within Representatives of the Genus Streptomyces. 4.2.2. The Diversity of FAME Patterns Within Streptomycetes. 137 4.3. Hierarchical Clustering to Determine Inter-relationships 140 VI Table of Contents, Page No. Between Streptomycetes Based on their Fatty Acid Profiles. 4.4. Distributions of FAMEs Within Sub-groups of the 145 Population. 4.4.1. The Distribution of FAMEs in Streptomyces Species 145 Groups. 4.4.2. The Distribution of FAMEs Within Producers of Specific 147 Classes of Antibiotics. 4.4.3. Distribution o f FAMEs Amongst Isolates from Specific 147 Geographical Locations and Isolation Procedures. 4.5. Alterations in the FAME Profile of D153 During Batch 151 Culture and its Relationship to Secondary Metabolite Biosynthesis. 4.5.1. Choice of Medium. 151 4.5.2. Timecourse o f Fatty Acid Biosynthesis Versus Antibiotic 154 Production by D1S3. 4.6. Discussion. 160 Chapter 5. The Relationship Between Chemical Profiles, 162 Taxonomic Status and Biological Activity. 5.1. Introduction. 162 5.1.1. Characteristics of Strains Chosen for this Study. 163 5.2. The Development of Methodology. 164 5.2.1. Scheme for the Numbering and Scoring of TLC Spots. 164 5.3. The Frequency of Occurrence of Different Spots. 166 5.3.1. Distribution of Metabolites. 166 VII Table o f Contents Page No. 5.4. The Estimation of Test Error Involved in Scoring 168 Metabolite Patterns. 5.4.1. The Effect of Operator Error in Scoring Metabolite 168 Patterns. 5.4.2. Reproducibility of Samples. 171 5.5. Cluster Analysis. 175 5.5.1. The Distribution of Streptomycetes Based on Chemical 175 Profiles, Using Hierarchical Methods. 5.5.2. Ordination of Streptomycete Chemical Profiles. 180 5.6. Recognising Interesting Strains using TLC. 184 5.7. Conclusions. 185 Chapter 6. Relationship Between Antibiotic Resistance 186 Phenotype and Biological Activities. 6.1.1. Introduction. 186 6.1.2. Choice of Strains. 188 6.2. The Distribution of Antibiotic Resistance in 190 Streptomycetes. 6.2.1. The Distribution of Antibiotic Resistance in Natural 190 Isolates. 6.2.2. The Diversity of Antibiotic Resistance Profiles. 193 6.2.3.
Recommended publications
  • B-Lactams: Chemical Structure, Mode of Action and Mechanisms of Resistance
    b-Lactams: chemical structure, mode of action and mechanisms of resistance Ru´ben Fernandes, Paula Amador and Cristina Prudeˆncio This synopsis summarizes the key chemical and bacteriological characteristics of b-lactams, penicillins, cephalosporins, carbanpenems, monobactams and others. Particular notice is given to first-generation to fifth-generation cephalosporins. This review also summarizes the main resistance mechanism to antibiotics, focusing particular attention to those conferring resistance to broad-spectrum cephalosporins by means of production of emerging cephalosporinases (extended-spectrum b-lactamases and AmpC b-lactamases), target alteration (penicillin-binding proteins from methicillin-resistant Staphylococcus aureus) and membrane transporters that pump b-lactams out of the bacterial cell. Keywords: b-lactams, chemical structure, mechanisms of resistance, mode of action Historical perspective Alexander Fleming first noticed the antibacterial nature of penicillin in 1928. When working with Antimicrobials must be understood as any kind of agent another bacteriological problem, Fleming observed with inhibitory or killing properties to a microorganism. a contaminated culture of Staphylococcus aureus with Antibiotic is a more restrictive term, which implies the the mold Penicillium notatum. Fleming remarkably saw natural source of the antimicrobial agent. Similarly, under- the potential of this unfortunate event. He dis- lying the term chemotherapeutic is the artificial origin of continued the work that he was dealing with and was an antimicrobial agent by chemical synthesis [1]. Initially, able to describe the compound around the mold antibiotics were considered as small molecular weight and isolates it. He named it penicillin and published organic molecules or metabolites used in response of his findings along with some applications of penicillin some microorganisms against others that inhabit the same [4].
    [Show full text]
  • Antibiotic Discovery
    ANTIBIOTIC DISCOVERY RESISTANCE PROFILING OF MICROBIAL GENOMES TO REVEAL NOVEL ANTIBIOTIC NATURAL PRODUCTS By CHELSEA WALKER, H. BSc. A Thesis Submitted to the School of Graduate Studies in Partial Fulfilment of the Requirements for the Degree Master of Science McMaster University © Copyright by Chelsea Walker, May 2017 McMaster University MASTER OF SCIENCE (2017) Hamilton, Ontario (Biochemistry and Biomedical Sciences) TITLE: Resistance Profiling of Microbial Genomes to Reveal Novel Antibiotic Natural Products. AUTHOR: Chelsea Walker, H. BSc. (McMaster University) SUPERVISOR: Dr. Nathan A. Magarvey. COMMITTEE MEMBERS: Dr. Eric Brown and Dr. Michael G. Surette. NUMBER OF PAGES: xvii, 168 ii Lay Abstract It would be hard to imagine a world where we could no longer use the antibiotics we are routinely being prescribed for common bacterial infections. Currently, we are in an era where this thought could become a reality. Although we have been able to discover antibiotics in the past from soil dwelling microbes, this approach to discovery is being constantly challenged. At the same time, the bacteria are getting smarter in their ways to evade antibiotics, in the form of resistance, or self-protection mechanisms. As such is it essential to devise methods which can predict the potential for resistance to the antibiotics we use early in the discovery and isolation process. By using what we have learned in the past about how bacteria protect themselves for antibiotics, we can to stay one step ahead of them as we continue to search for new sources of antibiotics from bacteria. iii Abstract Microbial natural products have been an invaluable resource for providing clinically relevant therapeutics for almost a century, including most of the commonly used antibiotics that are still in medical use today.
    [Show full text]
  • General Prescription
    GENERAL PRESCRIPTION LESSON 1. INTRODUCTION. PRESCRIPTION. SOLID MEDICINAL FORMS Objective: To study the structure of the prescription, learn the rules and get practical skills in writing out solid medicinal forms in prescription. To carry out practical tasks on prescriptions it is recommended to use Appendix 1. Key questions: 1. Pharmacology as a science and the basis of therapy. Main development milestones of modern pharmacology. Sections of Pharmacology. 2. The concept of medicinal substance, medicinal agent (medicinal drug, drug), medicinal form. 3. The concept of the pharmacological action and types of the action of drugs. 4. The sources of obtaining drugs. 5. International and national pharmacopeia, its content and purpose. 6. Pharmacy. Rules of drug storage and dispensing. 7. Prescription and its structure. Prescription forms. General rules for writing out a prescription. State regulation of writing out and dispensing drugs. 8. Name of medicinal products (international non-proprietary name - INN, trade name). 9. Peculiarities of writing out narcotic, poisonous and potent substances in prescription. 10. Drugs under control. Drugs prohibited for prescribing. 11. Solid medicinal forms: tablets, dragee (pills), powders, capsules. Their characteristics, advantages and disadvantages. Rules of prescribing. Write out prescriptions for: 1. 5 powders of Codeine 0.015 g. 1 powder orally twice a day. 2. 10 powders of Didanosine 0.25 g in sachets to prepare solution for internal use. Accept inside twice a day one sachet powder after dissolution in a glass of boiled water. 3. 50 mg of Alteplase powder in the bottle. Dissolve the content of the bottle in 50 ml of saline. First 15 ml introduce intravenously streamly, then intravenous drip.
    [Show full text]
  • Anew Drug Design Strategy in the Liht of Molecular Hybridization Concept
    www.ijcrt.org © 2020 IJCRT | Volume 8, Issue 12 December 2020 | ISSN: 2320-2882 “Drug Design strategy and chemical process maximization in the light of Molecular Hybridization Concept.” Subhasis Basu, Ph D Registration No: VB 1198 of 2018-2019. Department Of Chemistry, Visva-Bharati University A Draft Thesis is submitted for the partial fulfilment of PhD in Chemistry Thesis/Degree proceeding. DECLARATION I Certify that a. The Work contained in this thesis is original and has been done by me under the guidance of my supervisor. b. The work has not been submitted to any other Institute for any degree or diploma. c. I have followed the guidelines provided by the Institute in preparing the thesis. d. I have conformed to the norms and guidelines given in the Ethical Code of Conduct of the Institute. e. Whenever I have used materials (data, theoretical analysis, figures and text) from other sources, I have given due credit to them by citing them in the text of the thesis and giving their details in the references. Further, I have taken permission from the copyright owners of the sources, whenever necessary. IJCRT2012039 International Journal of Creative Research Thoughts (IJCRT) www.ijcrt.org 284 www.ijcrt.org © 2020 IJCRT | Volume 8, Issue 12 December 2020 | ISSN: 2320-2882 f. Whenever I have quoted written materials from other sources I have put them under quotation marks and given due credit to the sources by citing them and giving required details in the references. (Subhasis Basu) ACKNOWLEDGEMENT This preface is to extend an appreciation to all those individuals who with their generous co- operation guided us in every aspect to make this design and drawing successful.
    [Show full text]
  • Β-Lactam/Β-Lactamase Inhibitors for the Treatment of Infections Caused by Extended-Spectrum Β-Lactamase (ESBL)-Producing Enterobacteriaceae
    β-lactam/β-lactamase Inhibitors for the Treatment of Infections Caused by Extended-Spectrum β-Lactamase (ESBL)-producing Enterobacteriaceae Alireza FakhriRavari, Pharm.D. PGY-2 Pharmacotherapy Resident Controversies in Clinical Therapeutics University of the Incarnate Word Feik School of Pharmacy San Antonio, Texas November 13, 2015 Learning Objectives At the completion of this activity, the participant will be able to: 1. Describe different classes of β-lactamases produced by gram-negative bacteria. 2. Identify β-lactamase inhibitors and their spectrum of inhibition of β-lactamases. 3. Evaluate the evidence for use of β-lactam/β-lactamase inhibitors compared to carbapenems for treatment of ESBL infections. β-lactam/β-lactamase inhibitors for the treatment of infections caused by ESBL-producing Enterobacteriaceae 1 1. A Brief History of the Universe A. Timeline: 1940s a. β-lactams and β-lactamases i. Sir Alexander Fleming discovered penicillin from Penicillium notatum (now Penicillium chrysogenum) in 1928.1,2 ii. Chain, Florey, et al isolated penicillin in 1940, leading to its commercial production.3 iii. First β-lactamase was described as a penicillinase in Escherichia coli in 1940.4 iv. Giuseppe Brotzu discovered cephalosporin C from the mold Cephalosporin acremonium (now Acremonium chrysogenum) in 1945, but cephalosporins were not clinically used for another 2 decades.2,5 b. What are β-lactamases? i. β-lactamases are enzymes that hydrolyze the amide bond of the β-lactam ring, thereby inactivating them.6 Figure 1: Mechanism of action of β-lactamases ii. β-lactamase production is the principal mechanism by which gram-negative bacteria resist β-lactam antibiotics.6 iii.
    [Show full text]
  • Mechanisms of Β- Lactamase Inhibition And
    MECHANISMS OF β- LACTAMASE INHIBITION AND HETEROTROPIC ALLOSTERIC REGULATION OF AN ENGINEERED β- LACTAMASE-MBP FUSION PROTEIN By WEI KE Submitted in partial fulfillment of the requirements For the degree of Doctor of Philosophy. Dissertation Advisor: Dr. Focco ven den Akker Department of Biochemistry CASE WESTERN RESERVE UNIVERSITY May, 2011 CASE WESTERN RESERVE UNVERISTY SCHOOL OF GRADUATE STUDIES We hereby approve the thesis/dissertation of Wei Ke . candidate for the Ph.D degree*. (signed)Paul Carey . (chair of the committee) Focco van den Akker . Menachem Shoham . Robert A. Bonomo . Marion Skalweit . ___________________________________________ (date) 23 March, 2011 *We also certify that written approval has been obtained for any proprietary material contained therein. TABLE OF CONTENTS LIST OF TABLES ………………………………………………………………………8 LIST OF FIGURES ………………………………...…………………………………9 ACKNOWLEDGEMENTS ...………………………………………………………..13 LIST OF ABBREVIATIONS …………………...……………………………………15 ABSTRACT ……………………………….………………………………………...17 CHAPTER 1 Background and Significance …………………………………………....18 1.1 Antibiotic Resistance Crisis……………….…………………………….…...18 1.2 β-lactamases overview……………………………………………………….19 1.3 β-Lactam antibiotics and β-lactamase inhibitors ……………………………23 1.4 Structures of class A β –lactamases …………………………………………25 CHAPTER 2 Crystal Structures of SHV-1 β-Lactamase in Complex with Boronic Acid Transition State Inhibitors …………………………………………………...…….32 2.1 Introduction ………………………………………………………………….32 2.2 Materials and Methods ………………………………………………….…...34 2.2.1.
    [Show full text]
  • BMJ Open Is Committed to Open Peer Review. As Part of This Commitment We Make the Peer Review History of Every Article We Publish Publicly Available
    BMJ Open: first published as 10.1136/bmjopen-2018-027935 on 5 May 2019. Downloaded from BMJ Open is committed to open peer review. As part of this commitment we make the peer review history of every article we publish publicly available. When an article is published we post the peer reviewers’ comments and the authors’ responses online. We also post the versions of the paper that were used during peer review. These are the versions that the peer review comments apply to. The versions of the paper that follow are the versions that were submitted during the peer review process. They are not the versions of record or the final published versions. They should not be cited or distributed as the published version of this manuscript. BMJ Open is an open access journal and the full, final, typeset and author-corrected version of record of the manuscript is available on our site with no access controls, subscription charges or pay-per-view fees (http://bmjopen.bmj.com). If you have any questions on BMJ Open’s open peer review process please email [email protected] http://bmjopen.bmj.com/ on September 26, 2021 by guest. Protected copyright. BMJ Open BMJ Open: first published as 10.1136/bmjopen-2018-027935 on 5 May 2019. Downloaded from Treatment of stable chronic obstructive pulmonary disease: a protocol for a systematic review and evidence map Journal: BMJ Open ManuscriptFor ID peerbmjopen-2018-027935 review only Article Type: Protocol Date Submitted by the 15-Nov-2018 Author: Complete List of Authors: Dobler, Claudia; Mayo Clinic, Evidence-Based Practice Center, Robert D.
    [Show full text]
  • An Alternative Methodology for Determination of Cephamycin C
    aphy & S gr ep to a a ra t m i o o r n h T C e de Baptista Neto et al., J Chromat Separation Techniq 2012, 3:3 f c o h Chromatography l n a i DOI: 10.4172/2157-7064.1000130 q n u r e u s o J Separation Techniques ISSN: 2157-7064 Research Article OpenOpen Access Access An Alternative Methodology for Determination of Cephamycin C from Fermentation Broth Álvaro de Baptista Neto1, Liliane Maciel de Oliveira2, Carolina Bellão2, Alberto Colli Badino Jr2, Marlei Barboza2* and Carlos Osamu Hokka2 1Verdartis- Desenvolvimento Biotecnológico Ltda – ME, Brazil 2Departamento de Engenharia Química - Universidade Federal de São Carlos, Brazil Abstract Cephamycin C is a β-lactam antibiotic used as a raw material in several commercial antibiotics. The production and purification process of this antibiotic requires a fast, simple and accurate method to quantify it. In this paper, it was developed a high-performance liquid chromatography method to determine cephamycin C concentration, in order to offer an option for the traditional, but laborious, bioassay method normally employed. A method to obtain a calibration curve using the bioassay and cephalosporin C as standard was also proposed. The method showed more efficiency in determining cephamycin C than the bioassay, and it was simpler and faster to execute. Keywords: Cephamycin C; Bioassay method; Quantification; In a patent filled by Kamogashira et al. [10], the authors describe the Chromatography; Mass spectrometry determination of cephamycin C concentrations without derivatization of the antibiotic, using an acetic acid solution as the mobile phase Introduction and a Waters C-18 µ-bondapak column.
    [Show full text]
  • Escherichia Coli, Nova Scotia, Canada Brian Clarke,* Margot Hiltz,* Heather Musgrave,* and Kevin R
    RESEARCH Cephamycin Resistance in Clinical Isolates and Laboratory-derived Strains of Escherichia coli, Nova Scotia, Canada Brian Clarke,* Margot Hiltz,* Heather Musgrave,* and Kevin R. Forward* AmpC β-lactamase, altered porins, or both are usually E. coli processed from urine samples, 0.4% were responsible for cefoxitin resistance in Escherichia coli. We cephamycin resistant. examined the relative importance of each. We studied 18 All strains of E. coli possess a gene that encodes an strains of clinical isolates with reduced cefoxitin susceptibil- AmpC β-lactamase. Usually, almost no β-lactamase is pro- ity and 10 initially-susceptible strains passaged through duced because the gene is preceded by a weak promoter cefoxitin-gradient plates. Of 18 wild-resistant strains, 9 had identical promoter mutations (including creation of a con- and a strong attenuator (4). Surveys of resistance mecha- sensus 17-bp spacer) and related pulsed-field gel elec- nisms in cephamycin-resistant strains have most often trophoresis patterns; the other 9 strains were unrelated. identified promoter or attenuator mutations, which results Nine strains had attenuator mutations; two strains did not in an up-regulation of AmpC β-lactamase production express OmpC or OmpF. After serial passage, 8 of 10 (5–7). Occasionally, cephamycin-resistant strains bear strains developed cefoxitin resistance, none developed mobilized β-lactamases derived from bacteria such as promoter or attenuator mutations, 6 lost both the OmpC Citrobacter feundii (8). In addition, mutation or altered and OmpF porin proteins, and 1 showed decreased pro- expression of outer membrane proteins constituting porins duction of both. One strain had neither porin alteration or can also contribute to cephamycin resistance.
    [Show full text]
  • Caution Is Warranted in Using Cephamycin Antibiotics Against Recurrent Clostridioides Difficile Infection
    This is a repository copy of Caution is warranted in using cephamycin antibiotics against recurrent Clostridioides difficile infection. White Rose Research Online URL for this paper: http://eprints.whiterose.ac.uk/156451/ Version: Accepted Version Article: Wilcox, MH orcid.org/0000-0002-4565-2868 (2020) Caution is warranted in using cephamycin antibiotics against recurrent Clostridioides difficile infection. Nature Microbiology, 5 (2). p. 236. https://doi.org/10.1038/s41564-019-0661-9 © The Author(s), under exclusive licence to Springer Nature Limited 2020. This is an author produced version of an editorial comment published in Nature Microbiology. Uploaded in accordance with the publisher's self-archiving policy. Reuse Items deposited in White Rose Research Online are protected by copyright, with all rights reserved unless indicated otherwise. They may be downloaded and/or printed for private study, or other acts as permitted by national copyright laws. The publisher or other rights holders may allow further reproduction and re-use of the full text version. This is indicated by the licence information on the White Rose Research Online record for the item. Takedown If you consider content in White Rose Research Online to be in breach of UK law, please notify us by emailing [email protected] including the URL of the record and the reason for the withdrawal request. [email protected] https://eprints.whiterose.ac.uk/ Caution is warranted for using cephamycin antibiotics against recurrent Clostridioides difficile infection Mark H. Wilcox 1 1 Department of Microbiology, and UK Clostridium difficile Reference Laboratory, Leeds Teaching Hospitals and University of Leeds, Leeds, West Yorkshire, UK.
    [Show full text]
  • (12) Patent Application Publication (10) Pub. No.: US 2014/0256616 A1 Hsu (43) Pub
    US 2014025,6616A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2014/0256616 A1 Hsu (43) Pub. Date: Sep. 11, 2014 (54) MODIFIED GREEN TEA POLYPHENOL Publication Classification FORMULATIONS (51) Int. Cl. (71) Applicant: Georgia Regents Research Institute, A613 L/353 (2006.01) Inc., Augusta, GA (US) A638/2 (2006.01) A63/546 (2006.01) (72) Inventor: Stephen D. Hsu, Evans, GA (US) A613 L/7036 (2006.01) A613 L/7048 (2006.01) A613 L/496 (2006.01) (73) Assignee: Georgia Regents Research Institute, A61E36/82 (2006.01) Inc., Augusta, GA (US) A613 L/43 (2006.01) A613 L/65 (2006.01) 52) U.S. C. (21) Appl. No.: 14/280,805 (52) CPC ............... A61 K3I/353 (2013.01); A61K 31/43 (2013.01); A61 K38/12 (2013.01); A61 K (22) Filed: May 19, 2014 3 1/546 (2013.01); A61 K3I/65 (2013.01); A6 IK3I/7048 (2013.01); A61 K3I/496 O O (2013.01); A61 K36/82 (2013.01); A61 K Related U.S. Application Data 3/7036 (2013.01) (63) Continuation of application No. 13/305,296, filed on USPC ............ 514/2.9; 514/456; 514/198: 514/202; Nov. 28, 2011, which is a continuation of application 514/154: 514/29: 514/254.11: 514/37 No. 12/063,139, filed on Feb. 7, 2008, now Pat No. (57) ABSTRACT 8,076.484, filed as application No. PCT/US06/31120 Modified green tea polyphenols and methods of their use are on Aug. 10, 2006. provided. One aspect provides compounds and compositions (60) Provisional application No.
    [Show full text]
  • Gene Clusters for Β-Lactam Antibiotics and Control of Their Expression
    RESEARCH REVIEW INTERNATIONAL MICROBIOLOGY (2006) 9:9-19 www.im.microbios.org Paloma Liras Gene clusters for β-lactam Juan F. Martín* antibiotics and control of Biotechnological Institute, their expression: why have Scientific Park of Leon, Spain, and Microbiology Area, clusters evolved, and from Faculty of Biological where did they originate? and Environmental Sciences, University of Leon, Spain Summary. While β-lactam compounds were discovered in filamentous fungi, actinomycetes and gram-negative bacteria are also known to produce different types of β-lactams. All β-lactam compounds contain a four-membered β-lactam ring. The structure of their second ring allows these compounds to be classified into peni- cillins, cephalosporins, clavams, carbapenens or monobactams. Most β-lactams inhibits bacterial cell wall biosynthesis but others behave as β-lactamase inhibitors (e.g., clavu- lanic acid) and even as antifungal agents (e.g., some clavams). Due to the nature of the second ring in β-lactam molecules, the precursors and biosynthetic pathways of cla- vams, carbapenems and monobactams differ from those of penicillins and cephalospo- rins. These last two groups, including cephamycins and cephabacins, are formed from three precursor amino acids that are linked into the α-aminoadipyl-L-cysteinyl-D-valine tripeptide. The first two steps of their biosynthetic pathways are common. The interme- diates of these pathways, the characteristics of the enzymes involved, the lack of introns in the genes and bioinformatic analysis suggest that all of them should have evolved from an ancestral gene cluster of bacterial origin, which was surely transferred horizon- tally in the soil from producer to non-producer microorganisms.
    [Show full text]