CRISPR-Cas-Based Antimicrobials: Design, Challenges, and Bacterial Mechanisms of Resistance

The emergence of antibiotic-resistant bacterial strains is a source of public health concern across the globe. As the discovery of new conventional antibiotics has stalled significantly over the past decade, there is an urgency to develop novel approaches to address drug resistance in infectious diseases. The use of a CRISPR-Cas-based system for the precise elimination of targeted bacterial populations holds promise as an innovative approach for new antimicrobial agent design. The CRISPR-Cas targeting system is celebrated for its high versatility and specificity, offering an excellent opportunity to fight antibiotic resistance in pathogens by selectively inactivating genes involved in antibiotic resistance, biofilm formation, pathogenicity, virulence, or bacterial viability. The CRISPR-Cas strategy can enact antimicrobial effects by two approaches: inactivation of chromosomal genes or curing of plasmids encoding antibiotic resistance. In this Review, we provide an overview of the main CRISPR-Cas systems utilized for the creation of these antimicrobials, as well as highlighting promising studies in the field. We also offer a detailed discussion about the most commonly used mechanisms for CRISPR-Cas delivery: bacteriophages, nanoparticles, and conjugative plasmids. Lastly, we address possible mechanisms of interference that should be considered during the intelligent design of these novel approaches.

[1]  M. Zakrzewska,et al.  Mechanisms regulating the CRISPR-Cas systems , 2023, Frontiers in Microbiology.

[2]  Rajeshwari Kundar,et al.  CRISPR-Cas System: A Tool to Eliminate Drug-Resistant Gram-Negative Bacteria , 2022, Pharmaceuticals.

[3]  M. Webber,et al.  Molecular mechanisms of antibiotic resistance revisited , 2022, Nature Reviews Microbiology.

[4]  M. Suar,et al.  Synergy of Nanocarriers with CRISPR-Cas9 in an Emerging Technology Platform for Biomedical Appliances: Current Insights and Perspectives , 2022, Materials & Design.

[5]  Chao Wang,et al.  Efficient Suppression of Natural Plasmid-Borne Gene Expression in Carbapenem-Resistant Klebsiella pneumoniae Using a Compact CRISPR Interference System , 2022, Antimicrobial agents and chemotherapy.

[6]  P. C. Fineran,et al.  Widespread repression of anti-CRISPR production by anti-CRISPR-associated proteins , 2022, Nucleic acids research.

[7]  A. Patil,et al.  CRISPR in Modulating Antibiotic Resistance of ESKAPE Pathogens , 2022, Molecular Biotechnology.

[8]  Wei Liang,et al.  The Application of the CRISPR-Cas System in Antibiotic Resistance , 2022, Infection and drug resistance.

[9]  Min Wu,et al.  CRISPR-Cas systems target endogenous genes to impact bacterial physiology and alter mammalian immune responses , 2022, Molecular Biomedicine.

[10]  Lei Shi,et al.  Engineering a CRISPR interference system targeting AcrAB-TolC efflux pump to prevent multidrug resistance development in Escherichia coli. , 2022, The Journal of antimicrobial chemotherapy.

[11]  Pier-Luc Plante,et al.  A truncated anti-CRISPR protein prevents spacer acquisition but not interference , 2022, Nature Communications.

[12]  T. Mogensen,et al.  CRISPR-Cas in diagnostics and therapy of infectious diseases. , 2022, Journal of Infectious Diseases.

[13]  J. Bell,et al.  Advances in oncolytic virotherapy , 2022, Communications Medicine.

[14]  Z. Zeng,et al.  Targeted Elimination of blaNDM-5 Gene in Escherichia coli by Conjugative CRISPR-Cas9 System , 2022, Infection and drug resistance.

[15]  G. Paliouras,et al.  CRISPR–Cas9 gRNA efficiency prediction: an overview of predictive tools and the role of deep learning , 2022, Nucleic acids research.

[16]  Dalin Rifat,et al.  CRISPR Inhibition of Essential Peptidoglycan Biosynthesis Genes in Mycobacterium abscessus and Its Impact on β-Lactam Susceptibility , 2022, Antimicrobial agents and chemotherapy.

[17]  A. Kaushik,et al.  Exploring nano-enabled CRISPR-Cas-powered strategies for efficient diagnostics and treatment of infectious diseases , 2022, Journal of Nanostructure in Chemistry.

[18]  N. Høiby,et al.  Tolerance and resistance of microbial biofilms , 2022, Nature Reviews Microbiology.

[19]  M. Loessner,et al.  Engineering therapeutic phages for enhanced antibacterial efficacy , 2022, Current Opinion in Virology.

[20]  H. Schulenburg,et al.  The ESKAPE mobilome contributes to the spread of antimicrobial resistance and CRISPR-mediated conflict between mobile genetic elements , 2022, bioRxiv.

[21]  E. Rocha,et al.  Microbial defenses against mobile genetic elements and viruses: Who defends whom from what? , 2022, PLoS biology.

[22]  J. Lawrence,et al.  Environmental Biofilms as Reservoirs for Antimicrobial Resistance , 2021, Frontiers in Microbiology.

[23]  A. Eldar,et al.  Dormant phages communicate via arbitrium to control exit from lysogeny , 2021, Nature Microbiology.

[24]  Benjamin E. Rubin,et al.  Species- and site-specific genome editing in complex bacterial communities , 2021, Nature Microbiology.

[25]  Mohamed S. Draz,et al.  Engineered CRISPR-Cas systems for the detection and control of antibiotic-resistant infections , 2021, Journal of Nanobiotechnology.

[26]  David A. Baltrus,et al.  Introduction: the secret lives of microbial mobile genetic elements , 2021, Philosophical Transactions of the Royal Society B.

[27]  W. Hanage,et al.  Horizontal gene transfer and adaptive evolution in bacteria , 2021, Nature Reviews Microbiology.

[28]  Carl-Fredrik Flach,et al.  Antibiotic resistance in the environment , 2021, Nature Reviews Microbiology.

[29]  Jian Sun,et al.  Re-engineering a mobile-CRISPR/Cas9 system for antimicrobial resistance gene curing and immunization in Escherichia coli. , 2021, The Journal of antimicrobial chemotherapy.

[30]  Frédéric Grenier,et al.  High‐efficiency delivery of CRISPR‐Cas9 by engineered probiotics enables precise microbiome editing , 2021, Molecular systems biology.

[31]  F. F. de Melo,et al.  Oncolytic virus therapy in cancer: A current review , 2021, World journal of virology.

[32]  Joshua W. Modell,et al.  Viral recombination systems limit CRISPR-Cas targeting through the generation of escape mutations. , 2021, Cell host & microbe.

[33]  P. Devi,et al.  Targeting of Uropathogenic Escherichia coli papG gene using CRISPR-dot nanocomplex reduced virulence of UPEC , 2021, Scientific Reports.

[34]  M. Ellabaan,et al.  Bacterial resistance to CRISPR-Cas antimicrobials , 2021, Scientific reports.

[35]  H. Cao,et al.  Harnessing the CRISPR-Cas Systems to Combat Antimicrobial Resistance , 2021, Frontiers in Microbiology.

[36]  H. Petković,et al.  Towards the sustainable discovery and development of new antibiotics , 2021, Nature Reviews Chemistry.

[37]  Jian Sun,et al.  A Transposon-Associated CRISPR/Cas9 System Specifically Eliminates both Chromosomal and Plasmid-Borne mcr-1 in Escherichia coli , 2021, Antimicrobial agents and chemotherapy.

[38]  K. Palmer,et al.  CRISPR-based antimicrobials to obstruct antibiotic-resistant and pathogenic bacteria , 2021, PLoS pathogens.

[39]  P. Leiman,et al.  Reprogramming bacteriophage host range: design principles and strategies for engineering receptor binding proteins. , 2021, Current opinion in biotechnology.

[40]  Mohamed S. Draz,et al.  Novel Strategy to Combat Antibiotic Resistance: A Sight into the Combination of CRISPR/Cas9 and Nanoparticles , 2021, Pharmaceutics.

[41]  A. Uhlemann,et al.  Conjugation dynamics depend on both the plasmid acquisition cost and the fitness cost , 2021, Molecular systems biology.

[42]  Clément Fage,et al.  Delivery of CRISPR-Cas systems using phage-based vectors. , 2020, Current opinion in biotechnology.

[43]  T. Lu,et al.  Enhancing phage therapy through synthetic biology and genome engineering. , 2020, Current opinion in biotechnology.

[44]  Wenjun Li,et al.  Anti-quorum sensing and anti-biofilm activity of nickel oxide nanoparticles against Pseudomonas aeruginosa , 2020 .

[45]  Chris M. Brown,et al.  Discovery of multiple anti-CRISPRs highlights anti-defense gene clustering in mobile genetic elements , 2020, Nature Communications.

[46]  T. Kwok,et al.  Horizontal gene transfer potentiates adaptation by reducing selective constraints on the spread of genetic variation , 2020, Proceedings of the National Academy of Sciences.

[47]  A. Grossman,et al.  A mobile genetic element increases bacterial host fitness by manipulating development , 2020, bioRxiv.

[48]  Frédéric Grenier,et al.  Highly efficient gene transfer in the mouse gut microbiota is enabled by the Incl2 conjugative plasmid TP114 , 2020, Communications Biology.

[49]  Dawei Zhang,et al.  Application of different types of CRISPR/Cas-based systems in bacteria , 2020, Microbial Cell Factories.

[50]  P. Gabant,et al.  In the Age of Synthetic Biology, Will Antimicrobial Peptides be the Next Generation of Antibiotics? , 2020, Antibiotics.

[51]  Yang Wang,et al.  Promising Therapeutic Strategies Against Microbial Biofilm Challenges , 2020, Frontiers in Cellular and Infection Microbiology.

[52]  Trevor C. Charles,et al.  Microbiome definition re-visited: old concepts and new challenges , 2020, Microbiome.

[53]  Allyson E. Park,et al.  Mobile element warfare via CRISPR and anti-CRISPR in Pseudomonas aeruginosa , 2020, bioRxiv.

[54]  P. Turner,et al.  Pleiotropy complicates a trade-off between phage resistance and antibiotic resistance , 2020, Proceedings of the National Academy of Sciences.

[55]  T. Mendes,et al.  Exploring the Potential of CRISPR-Cas9 Under Challenging Conditions: Facing High-Copy Plasmids and Counteracting Beta-Lactam Resistance in Clinical Strains of Enterobacteriaceae , 2020, Frontiers in Microbiology.

[56]  Jordan J. Green,et al.  Poly(Beta-Amino Ester) Nanoparticles Enable Nonviral Delivery of CRISPR-Cas9 Plasmids for Gene Knockout and Gene Deletion , 2020, Molecular therapy. Nucleic acids.

[57]  K. Ganbarov,et al.  How CRISPR-Cas System Could Be Used to Combat Antimicrobial Resistance , 2020, Infection and drug resistance.

[58]  A. Davidson,et al.  Anti-CRISPRs: Protein Inhibitors of CRISPR-Cas Systems. , 2020, Annual review of biochemistry.

[59]  Bálint Csörgő,et al.  Anti-CRISPR protein applications: natural brakes for CRISPR-Cas technologies , 2020, Nature Methods.

[60]  R. Barrangou,et al.  In Vivo Targeting of Clostridioides difficile Using Phage-Delivered CRISPR-Cas3 Antimicrobials , 2020, mBio.

[61]  Rodrigo Ledesma-Amaro,et al.  Multiplexed CRISPR technologies for gene editing and transcriptional regulation , 2020, Nature Communications.

[62]  Qiang Cheng,et al.  Selective ORgan Targeting (SORT) nanoparticles for tissue specific mRNA delivery and CRISPR/Cas gene editing , 2020, Nature Nanotechnology.

[63]  Weilin Wang,et al.  Inflammation-targeting polymeric nanoparticles deliver sparfloxacin and tacrolimus for combating acute lung sepsis. , 2020, Journal of controlled release : official journal of the Controlled Release Society.

[64]  A. Krawczyk-Balska,et al.  RNA-Targeting CRISPR–Cas Systems and Their Applications , 2020, International journal of molecular sciences.

[65]  Rodolphe Barrangou,et al.  Characterization and applications of Type I CRISPR-Cas systems. , 2020, Biochemical Society transactions.

[66]  Xiaoxue Wang,et al.  Colistin Resistance Gene mcr-1 Mediates Cell Permeability and Resistance to Hydrophobic Antibiotics , 2020, Frontiers in Microbiology.

[67]  Qi Liu,et al.  Strategies for nonviral nanoparticle-based delivery of CRISPR/Cas9 therapeutics. , 2019, Wiley interdisciplinary reviews. Nanomedicine and nanobiotechnology.

[68]  V. Nizet,et al.  A bacterial gene-drive system efficiently edits and inactivates a high copy number antibiotic resistance locus , 2019, Nature Communications.

[69]  J. Dordick,et al.  Reducing Staphylococcus aureus resistance to lysostaphin using CRISPR‐dCas9 , 2019, Biotechnology and bioengineering.

[70]  R. Willems,et al.  CRISPR-Cas9-mediated genome editing in vancomycin-resistant Enterococcus faecium , 2019, FEMS microbiology letters.

[71]  Lei Tang Anti-anti-CRISPR , 2019, Nature Methods.

[72]  L. Cui,et al.  Development of CRISPR-Cas13a-based antimicrobials capable of sequence-specific killing of target bacteria , 2019, Nature Communications.

[73]  Xiaoyu Li,et al.  Efficacy of a phage cocktail in controlling phage resistance development in multidrug resistant Acinetobacter baumannii. , 2019, Virus research.

[74]  Bogumil J. Karas,et al.  Efficient inter-species conjugative transfer of a CRISPR nuclease for targeted bacterial killing , 2019, Nature Communications.

[75]  Mohamed S. Draz,et al.  Nanoparticle delivery systems for DNA/RNA and their potential applications in nanomedicine. , 2019, Current topics in medicinal chemistry.

[76]  Kwang-sun Kim,et al.  Nanomaterials as Delivery Vehicles and Components of New Strategies to Combat Bacterial Infections: Advantages and Limitations , 2019, Microorganisms.

[77]  Adair L. Borges,et al.  Anti-CRISPR-Associated Proteins Are Crucial Repressors of Anti-CRISPR Transcription , 2019, Cell.

[78]  A. Górski,et al.  Factors determining phage stability/activity: challenges in practical phage application , 2019, Expert review of anti-infective therapy.

[79]  L. Priddy,et al.  CRISPR-Cas9 modified bacteriophage for treatment of Staphylococcus aureus induced osteomyelitis and soft tissue infection , 2019, bioRxiv.

[80]  E. Nogales,et al.  Target preference of Type III-A CRISPR-Cas complexes at the transcription bubble , 2019, Nature Communications.

[81]  Karthik Hullahalli,et al.  Conjugative Delivery of CRISPR-Cas9 for the Selective Depletion of Antibiotic-Resistant Enterococci , 2019, Antimicrobial Agents and Chemotherapy.

[82]  Qiaobing Xu,et al.  Fast and Efficient CRISPR/Cas9 Genome Editing In Vivo Enabled by Bioreducible Lipid and Messenger RNA Nanoparticles , 2019, Advanced materials.

[83]  Munazza Gull,et al.  Plasmids as Genetic Tools and Their Applications in Ecology and Evolution , 2019, Plasmid.

[84]  D. Ling,et al.  Responsive Assembly of Silver Nanoclusters with a Biofilm Locally Amplified Bactericidal Effect to Enhance Treatments against Multi-Drug-Resistant Bacterial Infections , 2019, ACS central science.

[85]  Abdallah S. Abdelsattar,et al.  Encapsulation of E. coli phage ZCEC5 in chitosan–alginate beads as a delivery system in phage therapy , 2019, AMB Express.

[86]  C. Gersbach,et al.  The next generation of CRISPR–Cas technologies and applications , 2019, Nature Reviews Molecular Cell Biology.

[87]  M. Soliman,et al.  Biocompatibility, biodegradation and biomedical applications of poly(lactic acid)/poly(lactic-co-glycolic acid) micro and nanoparticles , 2019, Journal of Pharmaceutical Investigation.

[88]  Alfonso Rodríguez-Patón,et al.  Engineered toxin–intein antimicrobials can selectively target and kill antibiotic-resistant bacteria in mixed populations , 2019, Nature Biotechnology.

[89]  Sari Mattila,et al.  Midbiotics: conjugative plasmids for genetic engineering of natural gut flora , 2019, Gut microbes.

[90]  M. Loessner,et al.  Engineering Bacteriophages as Versatile Biologics. , 2019, Trends in microbiology.

[91]  D. Guo,et al.  Application of CRISPR/Cas9-Based Gene Editing in HIV-1/AIDS Therapy , 2019, Front. Cell. Infect. Microbiol..

[92]  V. Burrus,et al.  Entry Exclusion of Conjugative Plasmids of the IncA, IncC, and Related Untyped Incompatibility Groups , 2019, Journal of bacteriology.

[93]  Barry L. Stoddard,et al.  Functional metagenomics-guided discovery of potent Cas9 inhibitors in the human microbiome , 2019, bioRxiv.

[94]  Stefan Kol,et al.  Discovery and Characterization of Cas9 Inhibitors Disseminated across Seven Bacterial Phyla. , 2019, Cell host & microbe.

[95]  M. O’Connell,et al.  Molecular Mechanisms of RNA Targeting by Cas13-containing Type VI CRISPR-Cas Systems. , 2019, Journal of molecular biology.

[96]  Harris H. Wang,et al.  Metagenomic engineering of the mammalian gut microbiome in situ , 2018, Nature Methods.

[97]  J. Keith Joung,et al.  Discovery of widespread type I and type V CRISPR-Cas inhibitors , 2018, Science.

[98]  R. Novick,et al.  Conversion of staphylococcal pathogenicity islands to CRISPR-Cas9-based antibacterial drones that cure staph infections in mice , 2018, Nature Biotechnology.

[99]  M. Javed,et al.  CRISPR-Cas System: History and Prospects as a Genome Editing Tool in Microorganisms , 2018, Current Microbiology.

[100]  S. Partridge,et al.  Mobile Genetic Elements Associated with Antimicrobial Resistance , 2018, Clinical Microbiology Reviews.

[101]  A. Borman,et al.  Transcriptome Assembly and Profiling of Candida auris Reveals Novel Insights into Biofilm-Mediated Resistance , 2018, mSphere.

[102]  G. Zhu,et al.  CRISPR/Cas9/sgRNA-mediated targeted gene modification confirms the cause-effect relationship between gyrA mutation and quinolone resistance in Escherichia coli , 2018, FEMS microbiology letters.

[103]  E. Westra,et al.  CRISPR-Cas antimicrobials: Challenges and future prospects , 2018, PLoS pathogens.

[104]  Karthik Hullahalli,et al.  Enterococcus faecalis CRISPR-Cas Is a Robust Barrier to Conjugative Antibiotic Resistance Dissemination in the Murine Intestine , 2019, mSphere.

[105]  Jennifer A. Doudna,et al.  CRISPR-Cas12a target binding unleashes indiscriminate single-stranded DNase activity , 2018, Science.

[106]  R. Tjian,et al.  Genomes in Focus: Development and Applications of CRISPR-Cas9 Imaging Technologies. , 2018, Angewandte Chemie.

[107]  J. Bondy-Denomy,et al.  How bacteria control the CRISPR-Cas arsenal. , 2018, Current opinion in microbiology.

[108]  Elleke F Bosma,et al.  Hijacking CRISPR-Cas for high-throughput bacterial metabolic engineering: advances and prospects. , 2018, Current opinion in biotechnology.

[109]  Karthik Hullahalli,et al.  An Attenuated CRISPR-Cas System in Enterococcus faecalis Permits DNA Acquisition , 2018, mBio.

[110]  A. Wong,et al.  Plasmid persistence: costs, benefits, and the plasmid paradox. , 2018, Canadian journal of microbiology.

[111]  E. Bae,et al.  Solution structure and dynamics of anti-CRISPR AcrIIA4, the Cas9 inhibitor , 2018, Scientific Reports.

[112]  David R. Liu,et al.  Evolved Cas9 variants with broad PAM compatibility and high DNA specificity , 2018, Nature.

[113]  Robin Patel,et al.  Enterococcus faecalis Sex Pheromone cCF10 Enhances Conjugative Plasmid Transfer In Vivo , 2018, mBio.

[114]  Zhen Gu,et al.  Macrophage-Specific in Vivo Gene Editing Using Cationic Lipid-Assisted Polymeric Nanoparticles. , 2018, ACS nano.

[115]  M. Ouellette,et al.  Discovery, research, and development of new antibiotics: the WHO priority list of antibiotic-resistant bacteria and tuberculosis. , 2017, The Lancet. Infectious diseases.

[116]  E. Bae,et al.  Crystal structure of an anti-CRISPR protein, AcrIIA1 , 2017, Nucleic acids research.

[117]  O. Katare,et al.  Liposome loaded phage cocktail: Enhanced therapeutic potential in resolving Klebsiella pneumoniae mediated burn wound infections. , 2017, Burns : journal of the International Society for Burn Injuries.

[118]  S. Kaushik,et al.  Antibacterial Synergy of Silver Nanoparticles with Gentamicin and Chloramphenicol against Enterococcus faecalis , 2017, Pharmacognosy magazine.

[119]  Gaurav Sahay,et al.  Boosting Intracellular Delivery of Lipid Nanoparticle-Encapsulated mRNA. , 2017, Nano letters.

[120]  Henry C. Lin,et al.  Phage therapy: An alternative to antibiotics in the age of multi-drug resistance , 2017, World journal of gastrointestinal pharmacology and therapeutics.

[121]  Morten H. H. Nørholm,et al.  A versatile one-step CRISPR-Cas9 based approach to plasmid-curing , 2017, Microbial Cell Factories.

[122]  Guyue Cheng,et al.  Antimicrobial Activity and Resistance: Influencing Factors , 2017, Front. Pharmacol..

[123]  R. Barrangou,et al.  Using CRISPR-Cas systems as antimicrobials. , 2017, Current opinion in microbiology.

[124]  J. Doudna,et al.  CRISPR-Cas9 Structures and Mechanisms. , 2017, Annual review of biophysics.

[125]  Anna Kirpichnikova,et al.  Formulation, stabilisation and encapsulation of bacteriophage for phage therapy. , 2017, Advances in colloid and interface science.

[126]  D. Burstein,et al.  RNA Targeting by Functionally Orthogonal Type VI-A CRISPR-Cas Enzymes. , 2017, Molecular cell.

[127]  R. Terns,et al.  Programmable type III-A CRISPR-Cas DNA targeting modules , 2017, PloS one.

[128]  G. Donelli,et al.  Strategies for combating bacterial biofilms: A focus on anti-biofilm agents and their mechanisms of action , 2017, Virulence.

[129]  S. Moineau,et al.  Genome Engineering of Virulent Lactococcal Phages Using CRISPR-Cas9. , 2017, ACS synthetic biology.

[130]  Juw Won Park,et al.  Genetic engineering of a temperate phage-based delivery system for CRISPR/Cas9 antimicrobials against Staphylococcus aureus , 2017, Scientific Reports.

[131]  Vincent M Rotello,et al.  Integrating recognition elements with nanomaterials for bacteria sensing. , 2017, Chemical Society reviews.

[132]  Christopher M Thomas,et al.  Plasmid interference for curing antibiotic resistance plasmids in vivo , 2017, PloS one.

[133]  Chankyu Park,et al.  Nonviral Genome Editing Based on a Polymer-Derivatized CRISPR Nanocomplex for Targeting Bacterial Pathogens and Antibiotic Resistance. , 2017, Bioconjugate chemistry.

[134]  T. Lu,et al.  CRISPR-Cas9 technology: applications in genome engineering, development of sequence-specific antimicrobials, and future prospects. , 2017, Integrative biology : quantitative biosciences from nano to macro.

[135]  Anirvan Ghosh,et al.  Efficient genome editing in the mouse brain by local delivery of engineered Cas9 ribonucleoprotein complexes , 2017, Nature Biotechnology.

[136]  J. Messer,et al.  Evolutionary and ecological forces that shape the bacterial communities of the human gut , 2017, Mucosal Immunology.

[137]  Yi-Wei Lee,et al.  Direct Cytosolic Delivery of CRISPR/Cas9-Ribonucleoprotein for Efficient Gene Editing. , 2017, ACS nano.

[138]  Jianzhong Du,et al.  Preparation and Antibacterial Mechanism Insight of Polypeptide-Based Micelles with Excellent Antibacterial Activities. , 2016, Biomacromolecules.

[139]  K. Gregorczyk,et al.  Phage Therapy in Bacterial Infections Treatment: One Hundred Years After the Discovery of Bacteriophages , 2016, Current Microbiology.

[140]  E. Top,et al.  Plasmid transfer in biofilms: a perspective on limitations and opportunities , 2016, npj Biofilms and Microbiomes.

[141]  H. Horz,et al.  Preliminary survey of local bacteriophages with lytic activity against multi‐drug resistant bacteria , 2016, Journal of basic microbiology.

[142]  Sara Mitri,et al.  The Ecology and Evolution of Microbial Competition. , 2016, Trends in microbiology.

[143]  Zonghui Yuan,et al.  Survival and Evolution of CRISPR–Cas System in Prokaryotes and Its Applications , 2016, Front. Immunol..

[144]  D. Sridhar,et al.  Achieving global targets for antimicrobial resistance , 2016, Science.

[145]  A. Górski,et al.  Bacteriophage Procurement for Therapeutic Purposes , 2016, Front. Microbiol..

[146]  M. Wagner,et al.  Influence of Environmental Factors on Phage–Bacteria Interaction and on the Efficacy and Infectivity of Phage P100 , 2016, Front. Microbiol..

[147]  K. Palmer,et al.  CRISPR-Cas and Restriction-Modification Act Additively against Conjugative Antibiotic Resistance Plasmid Transfer in Enterococcus faecalis , 2016, mSphere.

[148]  Eric S. Lander,et al.  C2c2 is a single-component programmable RNA-guided RNA-targeting CRISPR effector , 2016, Science.

[149]  M. Wiedmann,et al.  Temperature Significantly Affects the Plaquing and Adsorption Efficiencies of Listeria Phages , 2016, Front. Microbiol..

[150]  Chase L Beisel,et al.  Current and future prospects for CRISPR‐based tools in bacteria , 2016, Biotechnology and bioengineering.

[151]  Jinbao Xu,et al.  Cationic polycarbonate-grafted superparamagnetic nanoparticles with synergistic dual-modality antimicrobial activity. , 2016, Biomaterials science.

[152]  R. Langer,et al.  mRNA vaccine delivery using lipid nanoparticles. , 2016, Therapeutic delivery.

[153]  D. Bikard,et al.  Consequences of Cas9 cleavage in the chromosome of Escherichia coli , 2016, Nucleic acids research.

[154]  E. Rodríguez,et al.  Ethical Issues in Genome Editing using Crispr/Cas9 System , 2016 .

[155]  Jan Winter,et al.  CRISPR library designer (CLD): software for multispecies design of single guide RNA libraries , 2016, Genome Biology.

[156]  Fang-Ting Kuo,et al.  RNA-activated DNA cleavage by the Type III-B CRISPR–Cas effector complex , 2016, Genes & development.

[157]  P. Cotter,et al.  Faculty Opinions recommendation of Bacteriocin production augments niche competition by enterococci in the mammalian gastrointestinal tract. , 2015 .

[158]  Jean-Baptiste Veyrieras,et al.  Phylogenetic Distribution of CRISPR-Cas Systems in Antibiotic-Resistant Pseudomonas aeruginosa , 2015, mBio.

[159]  Peter C. Fineran,et al.  A century of the phage: past, present and future , 2015, Nature Reviews Microbiology.

[160]  Michael J. Shen,et al.  No time to waste—the ethical challenges created by CRISPR , 2015, EMBO reports.

[161]  Wenyan Jiang,et al.  CRISPR-Cas: New Tools for Genetic Manipulations from Bacterial Immunity Systems. , 2015, Annual review of microbiology.

[162]  Pavlos Bousounis,et al.  Bacteriocin production augments niche competition by enterococci in the mammalian GI tract , 2015, Nature.

[163]  Daniel H. Huson,et al.  Antibiotic Selection Pressure Determination through Sequence-Based Metagenomics , 2015, Antimicrobial Agents and Chemotherapy.

[164]  T. Takemoto,et al.  Electroporation enables the efficient mRNA delivery into the mouse zygotes and facilitates CRISPR/Cas9-based genome editing , 2015, Scientific Reports.

[165]  U. Qimron,et al.  Temperate and lytic bacteriophages programmed to sensitize and kill antibiotic-resistant bacteria , 2015, Proceedings of the National Academy of Sciences.

[166]  Jason R. Clark Bacteriophage therapy: history and future prospects , 2015 .

[167]  Nuria Quiles-Puchalt,et al.  Bacteriophage-mediated spread of bacterial virulence genes. , 2015, Current opinion in microbiology.

[168]  Eric C Keen,et al.  A century of phage research: Bacteriophages and the shaping of modern biology , 2015, BioEssays : news and reviews in molecular, cellular and developmental biology.

[169]  R. Hancock,et al.  Antibiotic resistance in Pseudomonas aeruginosa biofilms: towards the development of novel anti-biofilm therapies. , 2014, Journal of biotechnology.

[170]  J. Doudna,et al.  The new frontier of genome engineering with CRISPR-Cas9 , 2014, Science.

[171]  R. Barrangou,et al.  A CRISPR design for next-generation antimicrobials , 2014, Genome Biology.

[172]  Chad W. Euler,et al.  Exploiting CRISPR-Cas nucleases to produce sequence-specific antimicrobials , 2014, Nature Biotechnology.

[173]  B. B. Finlay,et al.  Effects of antibiotics on human microbiota and subsequent disease. , 2014, Annual review of microbiology.

[174]  Timothy K Lu,et al.  Sequence-specific antimicrobials using efficiently delivered RNA-guided nucleases , 2014, Nature Biotechnology.

[175]  E. Lander,et al.  Development and Applications of CRISPR-Cas9 for Genome Engineering , 2014, Cell.

[176]  L. Marraffini,et al.  Harnessing CRISPR-Cas9 immunity for genetic engineering. , 2014, Current opinion in microbiology.

[177]  P. Siguier,et al.  Bacterial insertion sequences: their genomic impact and diversity , 2014, FEMS microbiology reviews.

[178]  Ingolf F. Ness,et al.  Enterococcal Bacteriocins and Antimicrobial Proteins that Contribute to Niche Control , 2014 .

[179]  Chase L. Beisel,et al.  Programmable Removal of Bacterial Strains by Use of Genome-Targeting CRISPR-Cas Systems , 2014, mBio.

[180]  Luke A. Gilbert,et al.  CRISPR interference (CRISPRi) for sequence-specific control of gene expression , 2013, Nature Protocols.

[181]  Bruce R. Levin,et al.  Dealing with the Evolutionary Downside of CRISPR Immunity: Bacteria and Beneficial Plasmids , 2013, PLoS genetics.

[182]  Gang Bao,et al.  CRISPR/Cas9 systems targeting β-globin and CCR5 genes have substantial off-target activity , 2013, Nucleic acids research.

[183]  Alessandra Carattoli,et al.  Plasmids and the spread of resistance. , 2013, International journal of medical microbiology : IJMM.

[184]  L. Frost,et al.  F conjugation: back to the beginning. , 2013, Plasmid.

[185]  S. Abedon,et al.  Phage cocktails and the future of phage therapy. , 2013, Future microbiology.

[186]  Peter C. Fineran,et al.  Cytotoxic Chromosomal Targeting by CRISPR/Cas Systems Can Reshape Bacterial Genomes and Expel or Remodel Pathogenicity Islands , 2013, PLoS genetics.

[187]  C. Suttle,et al.  To kill or not to kill: The balance between lytic and lysogenic viral infection is driven by trophic status , 2013 .

[188]  James E. DiCarlo,et al.  RNA-Guided Human Genome Engineering via Cas9 , 2013, Science.

[189]  Le Cong,et al.  Multiplex Genome Engineering Using CRISPR/Cas Systems , 2013, Science.

[190]  D. O'sullivan,et al.  Developing an efficient and reproducible conjugation-based gene transfer system for bifidobacteria. , 2013, Microbiology.

[191]  N. Gough Bacterial Cooperation , 2012, Science Signaling.

[192]  R. Barrangou,et al.  Cas9–crRNA ribonucleoprotein complex mediates specific DNA cleavage for adaptive immunity in bacteria , 2012, Proceedings of the National Academy of Sciences.

[193]  J. Doudna,et al.  A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity , 2012, Science.

[194]  S. Sørensen,et al.  The interconnection between biofilm formation and horizontal gene transfer. , 2012, FEMS immunology and medical microbiology.

[195]  Brian Austin,et al.  Selective Pressure of Antibiotic Pollution on Bacteria of Importance to Public Health , 2012, Environmental health perspectives.

[196]  H. Wolinski,et al.  In situ monitoring of IncF plasmid transfer on semi-solid agar surfaces reveals a limited invasion of plasmids in recipient colonies , 2012, Plasmid.

[197]  P. Stewart,et al.  Heterogeneity in Pseudomonas aeruginosa Biofilms Includes Expression of Ribosome Hibernation Factors in the Antibiotic-Tolerant Subpopulation and Hypoxia-Induced Stress Response in the Metabolically Active Population , 2012, Journal of bacteriology.

[198]  P. Malfertheiner,et al.  Escherichia coli Nissle 1917 (Mutaflor): New Insights into an Old Probiotic Bacterium , 2011, Digestive Diseases.

[199]  Stephen M. Krone,et al.  Increased Transfer of a Multidrug Resistance Plasmid in Escherichia coli Biofilms at the Air-Liquid Interface , 2011, Applied and Environmental Microbiology.

[200]  Stan J. J. Brouns,et al.  Evolution and classification of the CRISPR–Cas systems , 2011, Nature Reviews Microbiology.

[201]  S. Abedon,et al.  Phage treatment of human infections , 2011, Bacteriophage.

[202]  Sam P. Brown,et al.  Horizontal Gene Transfer and The Evolution of Bacterial Cooperation , 2011, Evolution; international journal of organic evolution.

[203]  Philippe Horvath,et al.  The CRISPR/Cas bacterial immune system cleaves bacteriophage and plasmid DNA , 2010, Nature.

[204]  Fernando de la Cruz,et al.  Mobility of Plasmids , 2010, Microbiology and Molecular Biology Reviews.

[205]  Alessandra Carattoli,et al.  Resistance Plasmid Families in Enterobacteriaceae , 2009, Antimicrobial Agents and Chemotherapy.

[206]  W. Hardt,et al.  PEGylation of bacteriophages increases blood circulation time and reduces T‐helper type 1 immune response , 2008, Microbial biotechnology.

[207]  Stephen M. Krone,et al.  Modelling the spatial dynamics of plasmid transfer and persistence. , 2007, Microbiology.

[208]  R. Barrangou,et al.  CRISPR Provides Acquired Resistance Against Viruses in Prokaryotes , 2007, Science.

[209]  S. Levy,et al.  Molecular Mechanisms of Antibacterial Multidrug Resistance , 2007, Cell.

[210]  Didier Mazel,et al.  Integrons: agents of bacterial evolution , 2006, Nature Reviews Microbiology.

[211]  S. Levy,et al.  Antibacterial resistance worldwide: causes, challenges and responses , 2004, Nature Medicine.

[212]  Tim Tolker-Nielsen,et al.  Gene transfer occurs with enhanced efficiency in biofilms and induces enhanced stabilisation of the biofilm structure. , 2003, Current opinion in biotechnology.

[213]  S. Molin,et al.  In situ detection of horizontal transfer of mobile genetic elements. , 2002, FEMS microbiology ecology.

[214]  E. Lanka,et al.  The origin of conjugative IncP plasmid transfer: interaction with plasmid-encoded products and the nucleotide sequence at the relaxation site. , 1988, Biochimica et biophysica acta.

[215]  M. Demerec Origin of Bacterial Resistance to Antibiotics , 1948, Journal of bacteriology.

[216]  F. Twort AN INVESTIGATION ON THE NATURE OF ULTRA-MICROSCOPIC VIRUSES. , 1915 .

[217]  H. Xiang,et al.  Exploiting a conjugative CRISPR/Cas9 system to eliminate plasmid harbouring the mcr-1 gene from Escherichia coli. , 2019, International journal of antimicrobial agents.

[218]  Mohamed S. Draz,et al.  Functional nanomaterials for the detection and control of bacterial infections. , 2019, Current topics in medicinal chemistry.

[219]  Athanasios Kakasis,et al.  Bacteriophage therapy as an alternative treatment for human infections. A comprehensive review. , 2019, International journal of antimicrobial agents.

[220]  José Luis Balcázar,et al.  Exploring the contribution of bacteriophages to antibiotic resistance. , 2017, Environmental pollution.

[221]  R. Bonomo β-Lactamases: A Focus on Current Challenges. , 2017, Cold Spring Harbor perspectives in medicine.

[222]  Qun Sun,et al.  Association of CRISPR/Cas evolution with Vibrio parahaemolyticus virulence factors and genotypes. , 2015, Foodborne pathogens and disease.

[223]  Michael S. Gilmore,et al.  Enterococci: From Commensals to Leading Causes of Drug Resistant Infection , 2014 .

[224]  Sam P. Brown,et al.  What traits are carried on mobile genetic elements, and why? , 2011, Heredity.

[225]  S. Abedon Phage therapy pharmacology: calculating phage dosing. , 2011, Advances in applied microbiology.

[226]  J. Gill,et al.  Phage choice, isolation, and preparation for phage therapy. , 2010, Current pharmaceutical biotechnology.

[227]  C. Fuqua,et al.  Bacterial competition: surviving and thriving in the microbial jungle , 2010, Nature Reviews Microbiology.

[228]  S. Abedon,et al.  Phage therapy in clinical practice: treatment of human infections. , 2010, Current pharmaceutical biotechnology.

[229]  L. Goodridge Designing phage therapeutics. , 2010, Current pharmaceutical biotechnology.

[230]  S. Abedon,et al.  Bacteriophage host range and bacterial resistance. , 2010, Advances in applied microbiology.

[231]  D. Helinski,et al.  Plasmid Incompatibility and Replication Control , 1993 .