Characterization of antibiotic resistomes by reprogrammed bacteriophage-enabled functional metagenomics in clinical strains

Functional metagenomics is a powerful experimental tool to identify antibiotic resistance genes (ARGs) in the environment, but the range of suitable host bacterial species is limited. This limitation affects both the scope of the identified ARGs and the interpretation of their clinical relevance. He...

Full description

Saved in:
Bibliographic Details
Published inNature microbiology Vol. 8; no. 3; pp. 410 - 423
Main Authors Apjok, Gábor, Számel, Mónika, Christodoulou, Chryso, Seregi, Viktória, Vásárhelyi, Bálint Márk, Stirling, Tamás, Eszenyi, Bálint, Sári, Tóbiás, Vidovics, Fanni, Nagrand, Erika, Kovács, Dorina, Szili, Petra, Lantos, Ildikó Ilona, Méhi, Orsolya, Jangir, Pramod K, Herczeg, Róbert, Gálik, Bence, Urbán, Péter, Gyenesei, Attila, Draskovits, Gábor, Nyerges, Ákos, Fekete, Gergely, Bodai, László, Zsindely, Nóra, Dénes, Béla, Yosef, Ido, Qimron, Udi, Papp, Balázs, Pál, Csaba, Kintses, Bálint
Format Journal Article
LanguageEnglish
Published England Nature Publishing Group 01.03.2023
Nature Publishing Group UK
Subjects
Online AccessGet full text

Cover

Loading…
Abstract Functional metagenomics is a powerful experimental tool to identify antibiotic resistance genes (ARGs) in the environment, but the range of suitable host bacterial species is limited. This limitation affects both the scope of the identified ARGs and the interpretation of their clinical relevance. Here we present a functional metagenomics pipeline called Reprogrammed Bacteriophage Particle Assisted Multi-species Functional Metagenomics (DEEPMINE). This approach combines and improves the use of T7 bacteriophage with exchanged tail fibres and targeted mutagenesis to expand phage host-specificity and efficiency for functional metagenomics. These modified phage particles were used to introduce large metagenomic plasmid libraries into clinically relevant bacterial pathogens. By screening for ARGs in soil and gut microbiomes and clinical genomes against 13 antibiotics, we demonstrate that this approach substantially expands the list of identified ARGs. Many ARGs have species-specific effects on resistance; they provide a high level of resistance in one bacterial species but yield very limited resistance in a related species. Finally, we identified mobile ARGs against antibiotics that are currently under clinical development or have recently been approved. Overall, DEEPMINE expands the functional metagenomics toolbox for studying microbial communities.
AbstractList Functional metagenomics is a powerful experimental tool to identify antibiotic resistance genes (ARGs) in the environment, but the range of suitable host bacterial species is limited. This limitation affects both the scope of the identified ARGs and the interpretation of their clinical relevance. Here we present a functional metagenomics pipeline called Reprogrammed Bacteriophage Particle Assisted Multi-species Functional Metagenomics (DEEPMINE). This approach combines and improves the use of T7 bacteriophage with exchanged tail fibres and targeted mutagenesis to expand phage host-specificity and efficiency for functional metagenomics. These modified phage particles were used to introduce large metagenomic plasmid libraries into clinically relevant bacterial pathogens. By screening for ARGs in soil and gut microbiomes and clinical genomes against 13 antibiotics, we demonstrate that this approach substantially expands the list of identified ARGs. Many ARGs have species-specific effects on resistance; they provide a high level of resistance in one bacterial species but yield very limited resistance in a related species. Finally, we identified mobile ARGs against antibiotics that are currently under clinical development or have recently been approved. Overall, DEEPMINE expands the functional metagenomics toolbox for studying microbial communities. Application of reprogrammed bacteriophage to functional metagenomics in clinically relevant bacterial strains improves identification of antibiotic resistance genes, including those against recently developed or approved antibiotics.
Functional metagenomics is a powerful experimental tool to identify antibiotic resistance genes (ARGs) in the environment, but the range of suitable host bacterial species is limited. This limitation affects both the scope of the identified ARGs and the interpretation of their clinical relevance. Here we present a functional metagenomics pipeline called Reprogrammed Bacteriophage Particle Assisted Multi-species Functional Metagenomics (DEEPMINE). This approach combines and improves the use of T7 bacteriophage with exchanged tail fibres and targeted mutagenesis to expand phage host-specificity and efficiency for functional metagenomics. These modified phage particles were used to introduce large metagenomic plasmid libraries into clinically relevant bacterial pathogens. By screening for ARGs in soil and gut microbiomes and clinical genomes against 13 antibiotics, we demonstrate that this approach substantially expands the list of identified ARGs. Many ARGs have species-specific effects on resistance; they provide a high level of resistance in one bacterial species but yield very limited resistance in a related species. Finally, we identified mobile ARGs against antibiotics that are currently under clinical development or have recently been approved. Overall, DEEPMINE expands the functional metagenomics toolbox for studying microbial communities.Application of reprogrammed bacteriophage to functional metagenomics in clinically relevant bacterial strains improves identification of antibiotic resistance genes, including those against recently developed or approved antibiotics.
Functional metagenomics is a powerful experimental tool to identify antibiotic resistance genes (ARGs) in the environment, but the range of suitable host bacterial species is limited. This limitation affects both the scope of the identified ARGs and the interpretation of their clinical relevance. Here we present a functional metagenomics pipeline called Reprogrammed Bacteriophage Particle Assisted Multi-species Functional Metagenomics (DEEPMINE). This approach combines and improves the use of T7 bacteriophage with exchanged tail fibres and targeted mutagenesis to expand phage host-specificity and efficiency for functional metagenomics. These modified phage particles were used to introduce large metagenomic plasmid libraries into clinically relevant bacterial pathogens. By screening for ARGs in soil and gut microbiomes and clinical genomes against 13 antibiotics, we demonstrate that this approach substantially expands the list of identified ARGs. Many ARGs have species-specific effects on resistance; they provide a high level of resistance in one bacterial species but yield very limited resistance in a related species. Finally, we identified mobile ARGs against antibiotics that are currently under clinical development or have recently been approved. Overall, DEEPMINE expands the functional metagenomics toolbox for studying microbial communities.
Abstract Functional metagenomics is a powerful experimental tool to identify antibiotic resistance genes (ARGs) in the environment, but the range of suitable host bacterial species is limited. This limitation affects both the scope of the identified ARGs and the interpretation of their clinical relevance. Here we present a functional metagenomics pipeline called Reprogrammed Bacteriophage Particle Assisted Multi-species Functional Metagenomics (DEEPMINE). This approach combines and improves the use of T7 bacteriophage with exchanged tail fibres and targeted mutagenesis to expand phage host-specificity and efficiency for functional metagenomics. These modified phage particles were used to introduce large metagenomic plasmid libraries into clinically relevant bacterial pathogens. By screening for ARGs in soil and gut microbiomes and clinical genomes against 13 antibiotics, we demonstrate that this approach substantially expands the list of identified ARGs. Many ARGs have species-specific effects on resistance; they provide a high level of resistance in one bacterial species but yield very limited resistance in a related species. Finally, we identified mobile ARGs against antibiotics that are currently under clinical development or have recently been approved. Overall, DEEPMINE expands the functional metagenomics toolbox for studying microbial communities.
Author Kovács, Dorina
Méhi, Orsolya
Qimron, Udi
Fekete, Gergely
Stirling, Tamás
Számel, Mónika
Nagrand, Erika
Herczeg, Róbert
Nyerges, Ákos
Christodoulou, Chryso
Urbán, Péter
Dénes, Béla
Kintses, Bálint
Zsindely, Nóra
Vidovics, Fanni
Szili, Petra
Jangir, Pramod K
Gálik, Bence
Pál, Csaba
Seregi, Viktória
Gyenesei, Attila
Sári, Tóbiás
Papp, Balázs
Eszenyi, Bálint
Draskovits, Gábor
Lantos, Ildikó Ilona
Vásárhelyi, Bálint Márk
Apjok, Gábor
Yosef, Ido
Bodai, László
Author_xml – sequence: 1
  givenname: Gábor
  orcidid: 0000-0002-8627-2378
  surname: Apjok
  fullname: Apjok, Gábor
  organization: Doctoral School of Biology, University of Szeged, Szeged, Hungary
– sequence: 2
  givenname: Mónika
  surname: Számel
  fullname: Számel, Mónika
  organization: Doctoral School of Biology, University of Szeged, Szeged, Hungary
– sequence: 3
  givenname: Chryso
  surname: Christodoulou
  fullname: Christodoulou, Chryso
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 4
  givenname: Viktória
  surname: Seregi
  fullname: Seregi, Viktória
  organization: HCEMM-BRC Translational Microbiology Research Group, Szeged, Hungary
– sequence: 5
  givenname: Bálint Márk
  surname: Vásárhelyi
  fullname: Vásárhelyi, Bálint Márk
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 6
  givenname: Tamás
  orcidid: 0000-0002-8964-6443
  surname: Stirling
  fullname: Stirling, Tamás
  organization: Institute of Biochemistry, Biological Research Centre, National Laboratory for Health Security, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 7
  givenname: Bálint
  surname: Eszenyi
  fullname: Eszenyi, Bálint
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 8
  givenname: Tóbiás
  surname: Sári
  fullname: Sári, Tóbiás
  organization: Doctoral School of Biology, University of Szeged, Szeged, Hungary
– sequence: 9
  givenname: Fanni
  surname: Vidovics
  fullname: Vidovics, Fanni
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 10
  givenname: Erika
  surname: Nagrand
  fullname: Nagrand, Erika
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 11
  givenname: Dorina
  surname: Kovács
  fullname: Kovács, Dorina
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 12
  givenname: Petra
  surname: Szili
  fullname: Szili, Petra
  organization: Doctoral School of Multidisciplinary Medical Sciences, University of Szeged, Szeged, Hungary
– sequence: 13
  givenname: Ildikó Ilona
  surname: Lantos
  fullname: Lantos, Ildikó Ilona
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 14
  givenname: Orsolya
  surname: Méhi
  fullname: Méhi, Orsolya
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 15
  givenname: Pramod K
  orcidid: 0000-0001-8330-0655
  surname: Jangir
  fullname: Jangir, Pramod K
  organization: Department of Zoology, University of Oxford, Oxford, UK
– sequence: 16
  givenname: Róbert
  surname: Herczeg
  fullname: Herczeg, Róbert
  organization: Bioinformatics Research Group, Genomics and Bioinformatics Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
– sequence: 17
  givenname: Bence
  orcidid: 0000-0002-3949-7005
  surname: Gálik
  fullname: Gálik, Bence
  organization: Department of Clinical Molecular Biology, Medical University of Bialystok, Bialystok, Poland
– sequence: 18
  givenname: Péter
  surname: Urbán
  fullname: Urbán, Péter
  organization: Bioinformatics Research Group, Genomics and Bioinformatics Core Facility, Szentágothai Research Centre, University of Pécs, Pécs, Hungary
– sequence: 19
  givenname: Attila
  surname: Gyenesei
  fullname: Gyenesei, Attila
  organization: Department of Clinical Molecular Biology, Medical University of Bialystok, Bialystok, Poland
– sequence: 20
  givenname: Gábor
  surname: Draskovits
  fullname: Draskovits, Gábor
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 21
  givenname: Ákos
  orcidid: 0000-0002-1581-490X
  surname: Nyerges
  fullname: Nyerges, Ákos
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 22
  givenname: Gergely
  surname: Fekete
  fullname: Fekete, Gergely
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary
– sequence: 23
  givenname: László
  orcidid: 0000-0001-8411-626X
  surname: Bodai
  fullname: Bodai, László
  organization: Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
– sequence: 24
  givenname: Nóra
  orcidid: 0000-0002-6189-3100
  surname: Zsindely
  fullname: Zsindely, Nóra
  organization: Department of Genetics, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary
– sequence: 25
  givenname: Béla
  orcidid: 0000-0002-9889-529X
  surname: Dénes
  fullname: Dénes, Béla
  organization: Veterinary Diagnostic Directorate, National Food Chain Safety Office, Budapest, Hungary
– sequence: 26
  givenname: Ido
  surname: Yosef
  fullname: Yosef, Ido
  organization: Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
– sequence: 27
  givenname: Udi
  surname: Qimron
  fullname: Qimron, Udi
  organization: Department of Clinical Microbiology and Immunology, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
– sequence: 28
  givenname: Balázs
  surname: Papp
  fullname: Papp, Balázs
  organization: HCEMM-BRC Metabolic Systems Biology Lab, Szeged, Hungary
– sequence: 29
  givenname: Csaba
  orcidid: 0000-0002-5187-9903
  surname: Pál
  fullname: Pál, Csaba
  email: csaba.pal@brc.hu
  organization: Synthetic and System Biology Unit, Institute of Biochemistry, Biological Research Centre, National Laboratory of Biotechnology, Eötvös Loránd Research Network (ELKH), Szeged, Hungary. csaba.pal@brc.hu
– sequence: 30
  givenname: Bálint
  orcidid: 0000-0003-0844-0310
  surname: Kintses
  fullname: Kintses, Bálint
  email: kintses.balint@brc.hu, kintses.balint@brc.hu, kintses.balint@brc.hu
  organization: Department of Biochemistry and Molecular Biology, Faculty of Science and Informatics, University of Szeged, Szeged, Hungary. kintses.balint@brc.hu
BackLink https://www.ncbi.nlm.nih.gov/pubmed/36759752$$D View this record in MEDLINE/PubMed
BookMark eNpdUUtLHTEYDaL4_gNdlIFuuhnNYyaPTaFcWhUEN7oOSfzm3shMcptkCtY_b65XxboIeZxHTnKO0G6IARD6QvAZwUye5470vGsxZS0mjOKW7qBDinvZ9lTw3Q_rA3Sa8wPGmHDKueT76IBx0SvR00P0tFiZZFyB5P-Z4mNo4tCYULz1sXjXJMg-lzhBbuxj3a1TXCYzTXDf2K0srldmCS0EY8d6OszBbXzM2ExQKhLi5F1ufGjc6IN3FcglGR_yCdobzJjh9HU-Rne_f90uLtvrm4urxc_r1nUMlxZohwfVMSnqUBIrwiV2ijJ5T4XqB0s5M2CpAEYZJtJZrjrVOy6Ys7aKjtGPre96tjW4g1DvH_U6-cmkRx2N1_8jwa_0Mv7VSknScVINvr8apPhnhlz05LODcTQB4pw1FaLnNSXbUL99oj7EOdXf2LCqG5FSyMqiW5ZLMecEw3sYgvWmXr2tV9d69Uu9mlbR14_PeJe8lcmeAQWbpHw
CitedBy_id crossref_primary_10_1002_ppul_26766
crossref_primary_10_1093_nar_gkae281
crossref_primary_10_1038_s41598_024_55776_1
crossref_primary_10_1093_femsml_uqad045
crossref_primary_10_1007_s11356_024_32535_3
crossref_primary_10_1128_spectrum_03162_23
crossref_primary_10_1038_s41467_024_45024_5
crossref_primary_10_3390_biology13010028
Cites_doi 10.1016/j.gene.2010.11.004
10.1007/s00248-016-0866-x
10.1007/978-1-4939-6634-9_19
10.1128/mSystems.00195-17
10.1016/j.gpb.2018.01.002
10.4161/viru.1.4.12010
10.1038/s41467-021-22757-1
10.1038/nature10571
10.1016/S0022-2836(05)80360-2
10.3389/fmicb.2014.00648
10.1128/AAC.00218-07
10.1128/am.14.3.470-471.1966
10.1126/science.1176950
10.1101/gr.214270.116
10.2147/DDDT.S56616
10.1038/s41467-018-08177-8
10.1016/j.vetmic.2014.02.017
10.1128/AEM.01802-16
10.2307/1411
10.1093/nar/gky1050
10.1002/cpt.2104
10.1093/bioinformatics/bty149
10.1093/jac/dkab131
10.1093/jac/dks261
10.1038/s41467-021-25096-3
10.1093/nar/gkz1136
10.1093/bioinformatics/btp352
10.1128/AAC.02465-14
10.1126/science.aax3879
10.7554/eLife.63775
10.1073/pnas.0609428103
10.1111/j.1574-6968.2012.02647.x
10.1038/s41598-019-57221-0
10.1038/nrmicro1160
10.1093/bioinformatics/bty191
10.1016/j.cell.2019.09.015
10.1016/j.molcel.2017.04.025
10.1016/j.mib.2019.06.005
10.1016/j.copbio.2009.09.010
10.1038/nmeth.3176
10.1016/j.copbio.2011.02.010
10.3389/fmicb.2015.00672
10.1038/s41467-018-02944-3
10.1038/nrg1709
10.3390/antibiotics10020170
10.1021/acssynbio.0c00467
10.1038/s41564-020-00839-y
10.1093/bioinformatics/bts565
10.1038/nrmicro.2017.28
10.1093/molbev/msz109
10.1101/gr.108332.110
10.1897/09-073.1
10.1186/1471-2105-11-119
10.1038/s41589-018-0100-x
10.1038/nature17672
10.1007/s00792-008-0193-3
10.1038/s41467-020-19124-x
10.1073/pnas.1801646115
10.1007/s15010-017-1007-2
10.1038/nrmicro1161
10.1038/ncomms10008
10.7554/eLife.46540
10.1093/bioinformatics/btl158
ContentType Journal Article
Copyright 2023. The Author(s).
The Author(s) 2023. This work is published under http://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
The Author(s) 2023
Copyright_xml – notice: 2023. The Author(s).
– notice: The Author(s) 2023. This work is published under http://creativecommons.org/licenses/by/4.0/ (the “License”). Notwithstanding the ProQuest Terms and Conditions, you may use this content in accordance with the terms of the License.
– notice: The Author(s) 2023
DBID CGR
CUY
CVF
ECM
EIF
NPM
AAYXX
CITATION
8FE
8FH
AFKRA
AZQEC
BBNVY
BENPR
BHPHI
CCPQU
DWQXO
GNUQQ
HCIFZ
LK8
M7P
PQEST
PQQKQ
PQUKI
PRINS
7X8
5PM
DOI 10.1038/s41564-023-01320-2
DatabaseName Medline
MEDLINE
MEDLINE (Ovid)
MEDLINE
MEDLINE
PubMed
CrossRef
ProQuest SciTech Collection
ProQuest Natural Science Collection
ProQuest Central
ProQuest Central Essentials
Biological Science Collection
ProQuest Central
Natural Science Collection
ProQuest One Community College
ProQuest Central Korea
ProQuest Central Student
SciTech Premium Collection (Proquest) (PQ_SDU_P3)
ProQuest Biological Science Collection
Biological Science Database
ProQuest One Academic Eastern Edition (DO NOT USE)
ProQuest One Academic
ProQuest One Academic UKI Edition
ProQuest Central China
MEDLINE - Academic
PubMed Central (Full Participant titles)
DatabaseTitle MEDLINE
Medline Complete
MEDLINE with Full Text
PubMed
MEDLINE (Ovid)
CrossRef
ProQuest Central Student
ProQuest Biological Science Collection
ProQuest Central Essentials
ProQuest One Academic Eastern Edition
SciTech Premium Collection
ProQuest One Community College
ProQuest Natural Science Collection
Biological Science Database
ProQuest SciTech Collection
ProQuest Central China
ProQuest Central
ProQuest One Academic UKI Edition
Natural Science Collection
ProQuest Central Korea
Biological Science Collection
ProQuest One Academic
MEDLINE - Academic
DatabaseTitleList
ProQuest Central Student
MEDLINE
CrossRef
Database_xml – sequence: 1
  dbid: NPM
  name: PubMed
  url: https://proxy.k.utb.cz/login?url=http://www.ncbi.nlm.nih.gov/entrez/query.fcgi?db=PubMed
  sourceTypes: Index Database
– sequence: 2
  dbid: EIF
  name: MEDLINE
  url: https://proxy.k.utb.cz/login?url=https://www.webofscience.com/wos/medline/basic-search
  sourceTypes: Index Database
– sequence: 3
  dbid: BENPR
  name: ProQuest Central
  url: https://www.proquest.com/central
  sourceTypes: Aggregation Database
DeliveryMethod fulltext_linktorsrc
Discipline Biology
EISSN 2058-5276
EndPage 423
ExternalDocumentID 10_1038_s41564_023_01320_2
36759752
Genre Research Support, Non-U.S. Gov't
Journal Article
GrantInformation_xml – fundername: ;
– fundername: ;
  grantid: BO/00303/19/8
GroupedDBID 0R~
53G
8FE
8FH
AAEEF
AAHBH
AARCD
AAZLF
ABLJU
ABVXF
ACGFS
ADBBV
AFBBN
AFKRA
AFSHS
AFWHJ
AHSBF
AIBTJ
ALFFA
ALMA_UNASSIGNED_HOLDINGS
ARMCB
AXYYD
BBNVY
BENPR
BHPHI
BKKNO
CCPQU
CGR
CUY
CVF
EBS
ECM
EIF
EJD
FSGXE
FZEXT
HCIFZ
HZ~
LK8
M7P
NNMJJ
NPM
O9-
ODYON
R9-
RNT
SHXYY
SIXXV
SNYQT
TAOOD
TBHMF
TDRGL
TSG
AAYXX
CITATION
AZQEC
DWQXO
GNUQQ
PQEST
PQQKQ
PQUKI
PRINS
7X8
5PM
ID FETCH-LOGICAL-c430t-e240f9438743898091680c9238d2795fb263aeb27e323018cb69495c673cbb743
IEDL.DBID BENPR
ISSN 2058-5276
IngestDate Tue Sep 17 21:30:02 EDT 2024
Sat Oct 05 06:11:53 EDT 2024
Fri Sep 13 10:16:48 EDT 2024
Thu Sep 12 21:19:19 EDT 2024
Sat Sep 28 08:12:24 EDT 2024
IsDoiOpenAccess true
IsOpenAccess true
IsPeerReviewed true
IsScholarly true
Issue 3
Language English
License 2023. The Author(s).
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.
LinkModel DirectLink
MergedId FETCHMERGED-LOGICAL-c430t-e240f9438743898091680c9238d2795fb263aeb27e323018cb69495c673cbb743
Notes ObjectType-Article-1
SourceType-Scholarly Journals-1
ObjectType-Feature-2
content type line 23
ORCID 0000-0002-1581-490X
0000-0002-3949-7005
0000-0003-0844-0310
0000-0002-6189-3100
0000-0002-9889-529X
0000-0002-8964-6443
0000-0001-8411-626X
0000-0001-8330-0655
0000-0002-5187-9903
0000-0002-8627-2378
OpenAccessLink https://pubmed.ncbi.nlm.nih.gov/PMC9981461
PMID 36759752
PQID 2781418878
PQPubID 2069616
PageCount 14
ParticipantIDs pubmedcentral_primary_oai_pubmedcentral_nih_gov_9981461
proquest_miscellaneous_2775624031
proquest_journals_2781418878
crossref_primary_10_1038_s41564_023_01320_2
pubmed_primary_36759752
PublicationCentury 2000
PublicationDate 2023-03-01
PublicationDateYYYYMMDD 2023-03-01
PublicationDate_xml – month: 03
  year: 2023
  text: 2023-03-01
  day: 01
PublicationDecade 2020
PublicationPlace England
PublicationPlace_xml – name: England
– name: London
PublicationTitle Nature microbiology
PublicationTitleAlternate Nat Microbiol
PublicationYear 2023
Publisher Nature Publishing Group
Nature Publishing Group UK
Publisher_xml – name: Nature Publishing Group
– name: Nature Publishing Group UK
References C Lübbert (1320_CR25) 2017; 45
MOA Sommer (1320_CR18) 2009; 325
CS Smillie (1320_CR40) 2011; 480
M Tridgett (1320_CR27) 2020; 10
MMH Ellabaan (1320_CR37) 2021; 12
W De Coster (1320_CR55) 2018; 34
EC Pehrsson (1320_CR17) 2016; 533
K Yehl (1320_CR29) 2019; 179
GD Wright (1320_CR23) 2019; 51
E Zankari (1320_CR36) 2012; 67
V Galata (1320_CR41) 2019; 47
U Qimron (1320_CR31) 2006; 103
RC MacLean (1320_CR50) 2019; 365
D Hyatt (1320_CR61) 2010; 11
W Li (1320_CR64) 2006; 22
Y Hu (1320_CR39) 2016; 82
H Li (1320_CR56) 2018; 34
P Huss (1320_CR28) 2021; 10
R Vaser (1320_CR59) 2017; 27
P Lorenz (1320_CR4) 2005; 3
A Porse (1320_CR20) 2018; 9
TS Crofts (1320_CR6) 2017; 15
T Sou (1320_CR33) 2021; 109
KN Lam (1320_CR11) 2018; 3
EM Lammens (1320_CR15) 2020; 11
BP Alcock (1320_CR35) 2020; 48
F Rousset (1320_CR49) 2021; 6
G Apjok (1320_CR19) 2019; 36
L Fu (1320_CR63) 2012; 28
J Bengtsson-Palme (1320_CR24) 2014; 5
L Tasse (1320_CR47) 2010; 20
1320_CR66
KS Kakirde (1320_CR48) 2011; 475
DFK dos Santos (1320_CR10) 2017; 73
AM Queenan (1320_CR44) 2007; 51
1320_CR60
M Hao (1320_CR46) 2021; 76
T Uchiyama (1320_CR14) 2009; 20
Á Nyerges (1320_CR22) 2018; 115
KJ Forsberg (1320_CR7) 2019; 8
M Taupp (1320_CR12) 2011; 22
VK Mutalik (1320_CR34) 2019; 10
1320_CR9
P-Y Colin (1320_CR5) 2015; 6
B Buchfink (1320_CR65) 2014; 12
SG Tringe (1320_CR1) 2005; 6
T Lupia (1320_CR32) 2021; 10
SF Altschul (1320_CR62) 1990; 215
1320_CR54
LM Coughlan (1320_CR2) 2015; 6
G Cheng (1320_CR52) 2012; 336
GCA Amos (1320_CR51) 2014; 171
DJ Farrell (1320_CR45) 2014; 58
AN Zhang (1320_CR38) 2021; 12
C Cillóniz (1320_CR42) 2019; 32
E van der Helm (1320_CR8) 2018; 14
J Fick (1320_CR53) 2009; 28
A Torres (1320_CR43) 2015; 9
R Daniel (1320_CR3) 2005; 3
C Bakermans (1320_CR26) 2009; 13
MOA Sommer (1320_CR16) 2010; 1
TR Ngara (1320_CR13) 2018; 16
T Holtzman (1320_CR30) 2020; 10
H Li (1320_CR57) 2009; 25
I Yosef (1320_CR21) 2017; 66
1320_CR58
References_xml – volume: 32
  start-page: 17
  year: 2019
  ident: 1320_CR42
  publication-title: Rev. Esp. Quimioter
  contributor:
    fullname: C Cillóniz
– volume: 475
  start-page: 57
  year: 2011
  ident: 1320_CR48
  publication-title: Gene
  doi: 10.1016/j.gene.2010.11.004
  contributor:
    fullname: KS Kakirde
– volume: 73
  start-page: 479
  year: 2017
  ident: 1320_CR10
  publication-title: Microb. Ecol.
  doi: 10.1007/s00248-016-0866-x
  contributor:
    fullname: DFK dos Santos
– ident: 1320_CR58
– ident: 1320_CR9
  doi: 10.1007/978-1-4939-6634-9_19
– volume: 3
  start-page: e00195-
  year: 2018
  ident: 1320_CR11
  publication-title: mSystems
  doi: 10.1128/mSystems.00195-17
  contributor:
    fullname: KN Lam
– volume: 16
  start-page: 405
  year: 2018
  ident: 1320_CR13
  publication-title: Genom. Proteom. Bioinform.
  doi: 10.1016/j.gpb.2018.01.002
  contributor:
    fullname: TR Ngara
– volume: 1
  start-page: 299
  year: 2010
  ident: 1320_CR16
  publication-title: Virulence
  doi: 10.4161/viru.1.4.12010
  contributor:
    fullname: MOA Sommer
– volume: 12
  start-page: 2435
  year: 2021
  ident: 1320_CR37
  publication-title: Nat Commun.
  doi: 10.1038/s41467-021-22757-1
  contributor:
    fullname: MMH Ellabaan
– volume: 480
  start-page: 241
  year: 2011
  ident: 1320_CR40
  publication-title: Nature
  doi: 10.1038/nature10571
  contributor:
    fullname: CS Smillie
– volume: 215
  start-page: 403
  year: 1990
  ident: 1320_CR62
  publication-title: J. Mol. Biol.
  doi: 10.1016/S0022-2836(05)80360-2
  contributor:
    fullname: SF Altschul
– volume: 5
  start-page: 648
  year: 2014
  ident: 1320_CR24
  publication-title: Front. Microbiol.
  doi: 10.3389/fmicb.2014.00648
  contributor:
    fullname: J Bengtsson-Palme
– volume: 51
  start-page: 3089
  year: 2007
  ident: 1320_CR44
  publication-title: Antimicrob. Agents Chemother.
  doi: 10.1128/AAC.00218-07
  contributor:
    fullname: AM Queenan
– ident: 1320_CR54
  doi: 10.1128/am.14.3.470-471.1966
– volume: 325
  start-page: 1128
  year: 2009
  ident: 1320_CR18
  publication-title: Science
  doi: 10.1126/science.1176950
  contributor:
    fullname: MOA Sommer
– volume: 27
  start-page: 737
  year: 2017
  ident: 1320_CR59
  publication-title: Genome Res.
  doi: 10.1101/gr.214270.116
  contributor:
    fullname: R Vaser
– volume: 9
  start-page: 4565
  year: 2015
  ident: 1320_CR43
  publication-title: Drug Des. Dev. Ther
  doi: 10.2147/DDDT.S56616
  contributor:
    fullname: A Torres
– volume: 10
  year: 2019
  ident: 1320_CR34
  publication-title: Nat. Commun.
  doi: 10.1038/s41467-018-08177-8
  contributor:
    fullname: VK Mutalik
– volume: 171
  start-page: 441
  year: 2014
  ident: 1320_CR51
  publication-title: Vet. Microbiol.
  doi: 10.1016/j.vetmic.2014.02.017
  contributor:
    fullname: GCA Amos
– volume: 82
  start-page: 6672
  year: 2016
  ident: 1320_CR39
  publication-title: Appl. Environ. Microbiol.
  doi: 10.1128/AEM.01802-16
  contributor:
    fullname: Y Hu
– ident: 1320_CR60
  doi: 10.2307/1411
– volume: 47
  start-page: D195
  year: 2019
  ident: 1320_CR41
  publication-title: Nucleic Acids Res.
  doi: 10.1093/nar/gky1050
  contributor:
    fullname: V Galata
– volume: 109
  start-page: 1063
  year: 2021
  ident: 1320_CR33
  publication-title: Clin. Pharmacol. Ther.
  doi: 10.1002/cpt.2104
  contributor:
    fullname: T Sou
– volume: 34
  start-page: 2666
  year: 2018
  ident: 1320_CR55
  publication-title: Bioinformatics
  doi: 10.1093/bioinformatics/bty149
  contributor:
    fullname: W De Coster
– volume: 76
  start-page: 2017
  year: 2021
  ident: 1320_CR46
  publication-title: J. Antimicrob. Chemother.
  doi: 10.1093/jac/dkab131
  contributor:
    fullname: M Hao
– volume: 67
  start-page: 2640
  year: 2012
  ident: 1320_CR36
  publication-title: J. Antimicrob. Chemother.
  doi: 10.1093/jac/dks261
  contributor:
    fullname: E Zankari
– volume: 12
  start-page: 4765
  year: 2021
  ident: 1320_CR38
  publication-title: Nat Commun.
  doi: 10.1038/s41467-021-25096-3
  contributor:
    fullname: AN Zhang
– volume: 48
  start-page: D517
  year: 2020
  ident: 1320_CR35
  publication-title: Nucleic Acids Res.
  doi: 10.1093/nar/gkz1136
  contributor:
    fullname: BP Alcock
– volume: 25
  start-page: 2078
  year: 2009
  ident: 1320_CR57
  publication-title: Bioinformatics
  doi: 10.1093/bioinformatics/btp352
  contributor:
    fullname: H Li
– volume: 58
  start-page: 3882
  year: 2014
  ident: 1320_CR45
  publication-title: Antimicrob. Agents Chemother.
  doi: 10.1128/AAC.02465-14
  contributor:
    fullname: DJ Farrell
– volume: 365
  start-page: 1082
  year: 2019
  ident: 1320_CR50
  publication-title: Science
  doi: 10.1126/science.aax3879
  contributor:
    fullname: RC MacLean
– volume: 10
  start-page: e63775
  year: 2021
  ident: 1320_CR28
  publication-title: eLife
  doi: 10.7554/eLife.63775
  contributor:
    fullname: P Huss
– volume: 103
  start-page: 19039
  year: 2006
  ident: 1320_CR31
  publication-title: Proc. Natl Acad. Sci. USA
  doi: 10.1073/pnas.0609428103
  contributor:
    fullname: U Qimron
– volume: 336
  start-page: 11
  year: 2012
  ident: 1320_CR52
  publication-title: FEMS Microbiol. Lett.
  doi: 10.1111/j.1574-6968.2012.02647.x
  contributor:
    fullname: G Cheng
– volume: 10
  start-page: 307
  year: 2020
  ident: 1320_CR30
  publication-title: Sci Rep.
  doi: 10.1038/s41598-019-57221-0
  contributor:
    fullname: T Holtzman
– volume: 3
  start-page: 470
  year: 2005
  ident: 1320_CR3
  publication-title: Nat. Rev. Microbiol.
  doi: 10.1038/nrmicro1160
  contributor:
    fullname: R Daniel
– volume: 34
  start-page: 3094
  year: 2018
  ident: 1320_CR56
  publication-title: Bioinformatics
  doi: 10.1093/bioinformatics/bty191
  contributor:
    fullname: H Li
– volume: 179
  start-page: 459
  year: 2019
  ident: 1320_CR29
  publication-title: Cell
  doi: 10.1016/j.cell.2019.09.015
  contributor:
    fullname: K Yehl
– volume: 66
  start-page: 721
  year: 2017
  ident: 1320_CR21
  publication-title: Mol. Cell
  doi: 10.1016/j.molcel.2017.04.025
  contributor:
    fullname: I Yosef
– volume: 51
  start-page: 57
  year: 2019
  ident: 1320_CR23
  publication-title: Curr. Opin. Microbiol.
  doi: 10.1016/j.mib.2019.06.005
  contributor:
    fullname: GD Wright
– volume: 20
  start-page: 616
  year: 2009
  ident: 1320_CR14
  publication-title: Curr Opin Biotechnol
  doi: 10.1016/j.copbio.2009.09.010
  contributor:
    fullname: T Uchiyama
– volume: 12
  start-page: 59
  year: 2014
  ident: 1320_CR65
  publication-title: Nat. Methods
  doi: 10.1038/nmeth.3176
  contributor:
    fullname: B Buchfink
– volume: 22
  start-page: 465
  year: 2011
  ident: 1320_CR12
  publication-title: Curr. Opin. Biotechnol.
  doi: 10.1016/j.copbio.2011.02.010
  contributor:
    fullname: M Taupp
– volume: 6
  start-page: 672
  year: 2015
  ident: 1320_CR2
  publication-title: Front. Microbiol.
  doi: 10.3389/fmicb.2015.00672
  contributor:
    fullname: LM Coughlan
– volume: 9
  start-page: 522
  year: 2018
  ident: 1320_CR20
  publication-title: Nat Commun.
  doi: 10.1038/s41467-018-02944-3
  contributor:
    fullname: A Porse
– volume: 6
  start-page: 805
  year: 2005
  ident: 1320_CR1
  publication-title: Nat. Rev. Genet.
  doi: 10.1038/nrg1709
  contributor:
    fullname: SG Tringe
– volume: 10
  start-page: 170
  year: 2021
  ident: 1320_CR32
  publication-title: Antibiotics
  doi: 10.3390/antibiotics10020170
  contributor:
    fullname: T Lupia
– ident: 1320_CR66
– volume: 10
  start-page: 107
  year: 2020
  ident: 1320_CR27
  publication-title: ACS Synth. Biol.
  doi: 10.1021/acssynbio.0c00467
  contributor:
    fullname: M Tridgett
– volume: 6
  start-page: 301
  year: 2021
  ident: 1320_CR49
  publication-title: Nat. Microbiol.
  doi: 10.1038/s41564-020-00839-y
  contributor:
    fullname: F Rousset
– volume: 28
  start-page: 3150
  year: 2012
  ident: 1320_CR63
  publication-title: Bioinformatics
  doi: 10.1093/bioinformatics/bts565
  contributor:
    fullname: L Fu
– volume: 15
  start-page: 422
  year: 2017
  ident: 1320_CR6
  publication-title: Nat. Rev. Microbiol.
  doi: 10.1038/nrmicro.2017.28
  contributor:
    fullname: TS Crofts
– volume: 36
  start-page: 1601
  year: 2019
  ident: 1320_CR19
  publication-title: Mol. Biol. Evol.
  doi: 10.1093/molbev/msz109
  contributor:
    fullname: G Apjok
– volume: 20
  start-page: 1605
  year: 2010
  ident: 1320_CR47
  publication-title: Genome Res.
  doi: 10.1101/gr.108332.110
  contributor:
    fullname: L Tasse
– volume: 28
  start-page: 2522
  year: 2009
  ident: 1320_CR53
  publication-title: Environ. Toxicol. Chem.
  doi: 10.1897/09-073.1
  contributor:
    fullname: J Fick
– volume: 11
  start-page: 119
  year: 2010
  ident: 1320_CR61
  publication-title: BMC Bioinformatics
  doi: 10.1186/1471-2105-11-119
  contributor:
    fullname: D Hyatt
– volume: 14
  start-page: 752
  year: 2018
  ident: 1320_CR8
  publication-title: Nat. Chem. Biol.
  doi: 10.1038/s41589-018-0100-x
  contributor:
    fullname: E van der Helm
– volume: 533
  start-page: 212
  year: 2016
  ident: 1320_CR17
  publication-title: Nature
  doi: 10.1038/nature17672
  contributor:
    fullname: EC Pehrsson
– volume: 13
  start-page: 21
  year: 2009
  ident: 1320_CR26
  publication-title: Extremophiles
  doi: 10.1007/s00792-008-0193-3
  contributor:
    fullname: C Bakermans
– volume: 11
  start-page: 5294
  year: 2020
  ident: 1320_CR15
  publication-title: Nat Commun.
  doi: 10.1038/s41467-020-19124-x
  contributor:
    fullname: EM Lammens
– volume: 115
  start-page: E5726
  year: 2018
  ident: 1320_CR22
  publication-title: Proc. Natl Acad. Sci. USA
  doi: 10.1073/pnas.1801646115
  contributor:
    fullname: Á Nyerges
– volume: 45
  start-page: 479
  year: 2017
  ident: 1320_CR25
  publication-title: Infection
  doi: 10.1007/s15010-017-1007-2
  contributor:
    fullname: C Lübbert
– volume: 3
  start-page: 510
  year: 2005
  ident: 1320_CR4
  publication-title: Nat Rev Microbiol
  doi: 10.1038/nrmicro1161
  contributor:
    fullname: P Lorenz
– volume: 6
  year: 2015
  ident: 1320_CR5
  publication-title: Nat. Commun.
  doi: 10.1038/ncomms10008
  contributor:
    fullname: P-Y Colin
– volume: 8
  start-page: e46540
  year: 2019
  ident: 1320_CR7
  publication-title: eLife
  doi: 10.7554/eLife.46540
  contributor:
    fullname: KJ Forsberg
– volume: 22
  start-page: 1658
  year: 2006
  ident: 1320_CR64
  publication-title: Bioinformatics
  doi: 10.1093/bioinformatics/btl158
  contributor:
    fullname: W Li
SSID ssj0001626686
Score 2.3199863
Snippet Functional metagenomics is a powerful experimental tool to identify antibiotic resistance genes (ARGs) in the environment, but the range of suitable host...
Abstract Functional metagenomics is a powerful experimental tool to identify antibiotic resistance genes (ARGs) in the environment, but the range of suitable...
SourceID pubmedcentral
proquest
crossref
pubmed
SourceType Open Access Repository
Aggregation Database
Index Database
StartPage 410
SubjectTerms Anti-Bacterial Agents - pharmacology
Antibiotic resistance
Antibiotics
Bacteria
Bacteria - genetics
Bacteriophages - genetics
Drug resistance
Genes, Bacterial
Genomes
Host specificity
Intestinal microflora
Metagenomics
Microbiomes
Phages
Site-directed mutagenesis
Species
Strains (organisms)
Title Characterization of antibiotic resistomes by reprogrammed bacteriophage-enabled functional metagenomics in clinical strains
URI https://www.ncbi.nlm.nih.gov/pubmed/36759752
https://www.proquest.com/docview/2781418878/abstract/
https://search.proquest.com/docview/2775624031
https://pubmed.ncbi.nlm.nih.gov/PMC9981461
Volume 8
hasFullText 1
inHoldings 1
isFullTextHit
isPrint
link http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwfV1La9wwEB7SLIVeQvrevFCgtyLWa8mWfArZkGUpdAmlgdyMJcvEh7W3sXMI-fOZke1NNoWcNWMZzWgekuYbgB9RoRx6bst1UiQc_XXOjZpGPAtyqZMY1dpS7fDvZby4lr9uopsdWAy1MPSscrCJ3lDntaUz8klI2ExT5NWTzNApgG0nZ-t_nPpH0T1r30zjHYxCpENdH80ul1d_ns9bMHKPddzXzQRCTxrKXSRHp8UDX0gcbvum_wLO1-8mXzii-T7s9REkO-9E_hF2XPUJ3nc9JR8-w-PFBoK5q7BkdcFw-UpT1sjBML0mNIGVa5h5YIRp6R9o4XzMdGz1-haNDHe-qipn5Pm6A0O2cm1GoK6r0jasrNhQVska32mi-QLX88u_Fwved1jgVoqg5Q79eZFIoRU1QdcYO8Q6sBjz6TxUSVSYMBYZ5t7KCUxVptqaOMGMysZKWGOQ6SvsVnXlvgNTwuSRyqQLtEUSZZIgcpnSOYaURko5hp_D2qbrDkgj9RfgQqedJFKUROolkYZjOBqWP-03VZM-q8AYTjfDuB3ojiOrXH1PNAojOommagzfOmltphOYHCUqwo-rLTluCAhqe3ukKm895DYmpdQA_eDt3zqED6FXJ9KoI9ht7-7dMcYsrTmB0fl8Nlue9Gr5BEIL78k
link.rule.ids 230,315,786,790,891,21416,27957,27958,33779,33780,43840
linkProvider ProQuest
linkToHtml http://utb.summon.serialssolutions.com/2.0.0/link/0/eLvHCXMwfV3db9MwED9BqwleJtgHBAozEm-TtTR2YudpgmlV2boKoU3qmxU7jpqHJmXpHqb985ydtKVD4tm-OPKd7-539t0BfI0LYdFyGyrTIqVor3OqxTCmWZhzmSYo1sblDt9Mk_Edv5rFsy7g1nTPKtc60SvqvDYuRn4WudpMQ6SV58vf1HWNcrerXQuNl9DnDKFKD_rfL6c_f22jLOivJzLpsmVCJs8ah1g4RVNFQ58-HO1apH_czOevJf8yP6M3sN_5jeRby-i38MJWB7DXdpJ8PISni03h5TavktQFwU0rdVkjBUFQ7WoILGxD9CNxlSz9syxcj-iWrF7OUbVQ63OpcuLsXRsmJAu7ylwp10VpGlJWZJ1MSRrfX6I5grvR5e3FmHZ9FajhLFxRi1a8SHHDhGt9LtFjSGRo0NOTeSTSuNBRwjJE3MIyBChDaXSSIo4yiWBGayQ6hl5VV_Y9EMF0HouM21AanCJ0GsY2EzJHR1JzzgM4Xe-tWrblM5S_9mZStZxQyAnlOaGiAAbr7VfdUWrUlvEBfNkM4yFwNxtZZesHN0egH8dRQQXwruXWZjmGkCgVMX5c7PBxM8EV2N4dqcq5L7SNUNS1Pf_w_986gVfj25uJmvyYXn-E15EXLSddA-it7h_sJ_RaVvpzJ5p_AMAz7YA
openUrl ctx_ver=Z39.88-2004&ctx_enc=info%3Aofi%2Fenc%3AUTF-8&rfr_id=info%3Asid%2Fsummon.serialssolutions.com&rft_val_fmt=info%3Aofi%2Ffmt%3Akev%3Amtx%3Ajournal&rft.genre=article&rft.atitle=Characterization+of+antibiotic+resistomes+by+reprogrammed+bacteriophage-enabled+functional+metagenomics+in+clinical+strains&rft.jtitle=Nature+microbiology&rft.au=Apjok%2C+G%C3%A1bor&rft.au=Sz%C3%A1mel%2C+M%C3%B3nika&rft.au=Christodoulou%2C+Chryso&rft.au=Seregi%2C+Vikt%C3%B3ria&rft.date=2023-03-01&rft.eissn=2058-5276&rft.volume=8&rft.issue=3&rft.spage=410&rft.epage=423&rft_id=info:doi/10.1038%2Fs41564-023-01320-2&rft.externalDBID=NO_FULL_TEXT
thumbnail_l http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/lc.gif&issn=2058-5276&client=summon
thumbnail_m http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/mc.gif&issn=2058-5276&client=summon
thumbnail_s http://covers-cdn.summon.serialssolutions.com/index.aspx?isbn=/sc.gif&issn=2058-5276&client=summon