1
|
Bacon EE, Myers KS, Iruegas-López R, Banta AB, Place M, Ebersberger I, Peters JM. Physiological roles of an Acinetobacter-specific σ factor. mBio 2025:e0096825. [PMID: 40387328 DOI: 10.1128/mbio.00968-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Accepted: 04/15/2025] [Indexed: 05/20/2025] Open
Abstract
The Gram-negative pathogen Acinetobacter baumannii is considered an "urgent threat" to human health due to its propensity to become antibiotic resistant. Understanding the distinct regulatory paradigms used by A. baumannii to mitigate cellular stresses may uncover new therapeutic targets. Many γ-proteobacteria use the extracytoplasmic function (ECF) σ factor, RpoE, to invoke envelope homeostasis networks in response to stress. Acinetobacter species contain the poorly characterized ECF "SigAb"; however, it is unclear if SigAb has the same physiological role as RpoE. Here, we show that SigAb is a metal stress-responsive ECF that appears unique to Acinetobacter species and distinct from RpoE-like ECFs. We combine promoter mutagenesis, motif scanning, and chromatin immunoprecipitation-sequencing (ChIP-seq) to define the direct SigAb regulon, which consists of genes encoding SigAb itself, the stringent response mediator, RelA, and the uncharacterized small RNA, "SabS." However, RNA-seq of strains overexpressing SigAb revealed a large, indirect regulon containing hundreds of genes. Metal resistance genes are key elements of the indirect regulon, as CRISPRi knockdown of sigAb or sabS resulted in increased copper sensitivity and excess copper-induced SigAb-dependent transcription. Furthermore, we found that two uncharacterized genes in the sigAb operon, "aabA" and "aabB," have anti-SigAb activity. Finally, employing a targeted Tn-seq approach that uses CRISPR-associated transposons, we show that sigAb, aabA, and aabB are important for fitness even during optimal growth conditions. Our work reveals new physiological roles for SigAb and SabS, provides a novel approach for assessing gene fitness, and highlights the distinct regulatory architecture of A. baumannii. IMPORTANCE Acinetobacter baumannii is a hospital-acquired pathogen, and many strains are resistant to multiple antibiotics. Understanding how A. baumannii senses and responds to stress may uncover novel routes to treat infections. Here, we examine how the Acinetobacter-specific transcription factor, SigAb, mitigates stress. We find that SigAb directly regulates only a small number of genes, but indirectly controls hundreds of genes that have substantial impacts on cell physiology. We show that SigAb is required for maximal growth, even during optimal conditions, and is acutely required during growth in the presence of elevated copper. Given that copper toxicity plays roles in pathogenesis and on copper-containing surfaces in hospitals, we speculate that SigAb function may be important in clinically relevant contexts.
Collapse
Affiliation(s)
- Emily E Bacon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kevin S Myers
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Rubén Iruegas-López
- Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt, Hesse, Germany
| | - Amy B Banta
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Michael Place
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Ingo Ebersberger
- Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Frankfurt, Hesse, Germany
- Senckenberg Biodiversity and Climate Research Centre (S-BIKF), Frankfurt, Hesse, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Frankfurt, Hesse, Germany
| | - Jason M Peters
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, Wisconsin, USA
| |
Collapse
|
2
|
Wolella EK, Cheng Z, Li M, Xia D, Zhang J, Duan L, Liu L, Li Z, Zhang J. Large-Scale Rice Mutant Establishment and High-Throughput Mutant Manipulation Help Advance Rice Functional Genomics. PLANTS (BASEL, SWITZERLAND) 2025; 14:1492. [PMID: 40431057 PMCID: PMC12114927 DOI: 10.3390/plants14101492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 05/08/2025] [Accepted: 05/13/2025] [Indexed: 05/29/2025]
Abstract
Rice (Oryza sativa L.) is a stable food for over half of the world population, contributing 50-80% of the daily calorie intake. The completion of rice genome sequencing marks a significant milestone in understanding functional genomics, yet the systematic identification of gene functions remains a bottleneck for rice improvement. Large-scale mutant libraries in which the functions of genes are lost or gained (e.g., through chemical/physical treatments, T-DNA, transposons, RNAi, CRISPR/Cas9) have proven to be powerful tools for the systematic linking of genotypes to phenotypes. So far, using different mutagenesis approaches, a million mutant lines have been established and about 5-10% of the predicted rice gene functions have been identified due to the high demands of labor and low-throughput utilization. DNA-barcoding-based large-scale mutagenesis offers unprecedented precision and scalability in functional genomics. This review summarizes large-scale loss-of-function and gain-of-function mutant library development approaches and emphasizes the integration of DNA barcoding for pooled analysis. Unique DNA barcodes can be tagged to transposons/retrotransposons, DNA constructs, miRNA/siRNA, gRNA, and cDNA, allowing for pooling analysis and the assignment of functions to genes that cause phenotype alterations. In addition, the integration of high-throughput phenotyping and OMICS technologies can accelerate the identification of gene functions.
Collapse
Affiliation(s)
- Eyob Kassaye Wolella
- State Key Laboratory of Rice Biology and Breeding, China National Rice Research Institute, Hangzhou 311400, China; (E.K.W.); (Z.C.)
- Department of Biology, College of Natural and Computational Sciences, Debre Tabor University, Debre Tabor P.O. Box 272, Ethiopia
| | - Zhen Cheng
- State Key Laboratory of Rice Biology and Breeding, China National Rice Research Institute, Hangzhou 311400, China; (E.K.W.); (Z.C.)
- School of Life Sciences, Hubei University, Wuhan 430062, China; (L.D.); (L.L.)
| | - Mengyuan Li
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.L.); (D.X.); (J.Z.)
| | - Dandan Xia
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.L.); (D.X.); (J.Z.)
| | - Jianwei Zhang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan 430070, China; (M.L.); (D.X.); (J.Z.)
| | - Liu Duan
- School of Life Sciences, Hubei University, Wuhan 430062, China; (L.D.); (L.L.)
| | - Li Liu
- School of Life Sciences, Hubei University, Wuhan 430062, China; (L.D.); (L.L.)
| | - Zhiyong Li
- State Key Laboratory of Rice Biology and Breeding, China National Rice Research Institute, Hangzhou 311400, China; (E.K.W.); (Z.C.)
- National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya 572024, China
| | - Jian Zhang
- State Key Laboratory of Rice Biology and Breeding, China National Rice Research Institute, Hangzhou 311400, China; (E.K.W.); (Z.C.)
- National Nanfan Research Institute (Sanya), Chinese Academy of Agricultural Sciences, Sanya 572024, China
| |
Collapse
|
3
|
Clavel T, Faber F, Groussin M, Haller D, Overmann J, Pauvert C, Poyet M, Selkrig J, Stecher B, Typas A, Vehreschild MJGT, Westermann AJ, Wylensek D, Maier L. Enabling next-generation anaerobic cultivation through biotechnology to advance functional microbiome research. Nat Biotechnol 2025:10.1038/s41587-025-02660-6. [PMID: 40301656 DOI: 10.1038/s41587-025-02660-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Accepted: 03/31/2025] [Indexed: 05/01/2025]
Abstract
Microbiomes are complex communities of microorganisms that are essential for biochemical processes on Earth and for the health of humans, animals and plants. Many environmental and host-associated microbiomes are dominated by anaerobic microbes, some of which cannot tolerate oxygen. Anaerobic microbial communities have been extensively studied over the last 20 years using molecular techniques, especially next-generation sequencing. However, there is a renewed interest in microbial cultivation because isolates provide the basis for understanding the taxonomic and functional units of biodiversity, elucidating novel biochemical pathways and the mechanisms underlying microbe-microbe and microbe-host interactions and opening new avenues for biotechnological and clinical applications. In this Perspective, we present areas of research and applications that will benefit from advancement in anaerobic microbial cultivation. We highlight key technical and infrastructural hurdles associated with the development and deployment of sophisticated cultivation workflows. Improving the performance of cultivation techniques will set new trends in functional microbiome research in the coming years.
Collapse
Affiliation(s)
- Thomas Clavel
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany.
| | - Franziska Faber
- Institute for Hygiene and Microbiology, Faculty of Medicine, University of Würzburg, Würzburg, Germany
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
| | - Mathieu Groussin
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - Dirk Haller
- Chair of Nutrition and Immunology, School of Life Sciences, Technical University of Munich, Freising, Germany
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, Germany
| | - Jörg Overmann
- Leibniz-Institut DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
- Microbiology, Technical University of Braunschweig, Braunschweig, Germany
- German Centre for Infection Research (DZIF), Partner Site Hannover-Braunschweig, Braunschweig, Germany
| | - Charlie Pauvert
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Mathilde Poyet
- Institute of Experimental Medicine, Kiel University, Kiel, Germany
| | - Joel Selkrig
- Host-Microbe Interactomics Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Bärbel Stecher
- ZIEL - Institute for Food and Health, Technical University of Munich, Freising, Germany
- Chair of Intestinal Microbiome, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Athanasios Typas
- European Molecular Biology Laboratory, Molecular Systems Biology Unit, Heidelberg, Germany
| | - Maria J G T Vehreschild
- Goethe University Frankfurt, University Hospital Frankfurt, Department II of Internal Medicine, Infectious Diseases, Frankfurt am Main, Germany
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, Frankfurt am Main, Germany
| | - Alexander J Westermann
- Helmholtz Institute for RNA-Based Infection Research (HIRI), Helmholtz Centre for Infection Research (HZI), Würzburg, Germany
- Department of Microbiology, Biocentre, University of Würzburg, Würzburg, Germany
| | - David Wylensek
- Functional Microbiome Research Group, Institute of Medical Microbiology, RWTH University Hospital, Aachen, Germany
| | - Lisa Maier
- Interfaculty Institute for Microbiology and Infection Medicine Tübingen, Tübingen, Germany
- Cluster of Excellence EXC 2124 Controlling Microbes to Fight Infections, Tübingen, Germany
- M3-Research Center for Malignome, Metabolome and Microbiome, University of Tübingen, Tübingen, Germany
| |
Collapse
|
4
|
Lin HC, Hsiao WC, Hsu YC, Lin MC, Hsu CC, Zhang MM. Highly efficient CRISPR-Cas9 base editing in Bifidobacterium with bypass of restriction modification systems. Appl Environ Microbiol 2025; 91:e0198524. [PMID: 40062897 PMCID: PMC12016496 DOI: 10.1128/aem.01985-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 02/10/2025] [Indexed: 04/24/2025] Open
Abstract
Intestinal microbiota members of the Bifidobacterium genus are increasingly explored as probiotics and therapeutics. However, the paucity of genetic tools and the widespread restriction modification (RM) systems in Bifidobacterium limit our ability to genetically manipulate these bacteria. Here we established a CRISPR-Cas9 cytosine base editor system (cBEST) for portable genome editing in bifidobacteria. Harboring different promoters characterized in this study, these cBEST plasmids showed a range of editing efficiencies in different strains and genomic contexts, highlighting the importance of fine-tuning base editor and sgRNA expression. Additionally, we showed that disruption or bypass of RM systems dramatically improved editing efficiencies in otherwise hard-to-edit genomic loci and Bifidobacterium strains. Notably, we demonstrated the use of RM-disrupted Bifidobacterium longum strains for simultaneous assembly, amplification, and methylation of the all-in-one editing plasmids, greatly streamlining the workflow for high-efficiency base editing. Last but not least, we showed the portability of cBESTs using the same editing construct to disrupt a conserved metabolic gene in multiple Bifidobacterium species. Looking ahead, the ability to efficiently edit and engineer bifidobacterial genomes will give rise to new opportunities for research and applications toward improving human health.IMPORTANCEThe ability to genetically manipulate specific genes and biological pathways in Bifidobacterium is essential to unlocking their probiotic and therapeutic potential in human health applications. The DNA double-strand break-free CRISPR-Cas9 cytosine base editor system established in this work allows portable and efficient base editing in Bifidobacterium spp. We further showed that bypass of restriction modification systems significantly improved base editing efficiency, especially for hard-to-edit genomic loci and strains. This expanded Bifidobacterium genome editing toolbox should facilitate mechanistic investigations into the roles of Bifidobacterium in host physiology and disease.
Collapse
Affiliation(s)
- Hung-Chun Lin
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Wan-Chi Hsiao
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| | - Ya-Chen Hsu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Meng-Chieh Lin
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Cheng-Chih Hsu
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
- Leeuwenhoek Laboratories Co. Ltd, Taipei, Taiwan
| | - Mingzi M. Zhang
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
5
|
Shiver AL, Sun J, Culver R, Violette A, Wynter C, Nieckarz M, Mattiello SP, Sekhon PK, Bottacini F, Friess L, Carlson HK, Wong DPGH, Higginbottom S, Weglarz M, Wang W, Knapp BD, Guiberson E, Sanchez J, Huang PH, Garcia PA, Buie CR, Good BH, DeFelice B, Cava F, Scaria J, Sonnenburg JL, Van Sinderen D, Deutschbauer AM, Huang KC. Genome-scale resources in the infant gut symbiont Bifidobacterium breve reveal genetic determinants of colonization and host-microbe interactions. Cell 2025; 188:2003-2021.e19. [PMID: 40068681 DOI: 10.1016/j.cell.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 08/08/2024] [Accepted: 02/13/2025] [Indexed: 03/27/2025]
Abstract
Bifidobacteria represent a dominant constituent of human gut microbiomes during infancy, influencing nutrition, immune development, and resistance to infection. Despite interest in bifidobacteria as a live biotic therapy, our understanding of colonization, host-microbe interactions, and the health-promoting effects of bifidobacteria is limited. To address these major knowledge gaps, we used a large-scale genetic approach to create a mutant fitness compendium in Bifidobacterium breve. First, we generated a high-density randomly barcoded transposon insertion pool and used it to determine fitness requirements during colonization of germ-free mice and chickens with multiple diets and in response to hundreds of in vitro perturbations. Second, to enable mechanistic investigation, we constructed an ordered collection of insertion strains covering 1,462 genes. We leveraged these tools to reveal community- and diet-specific requirements for colonization and to connect the production of immunomodulatory molecules to growth benefits. These resources will catalyze future investigations of this important beneficial microbe.
Collapse
Affiliation(s)
- Anthony L Shiver
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Jiawei Sun
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Rebecca Culver
- Department of Genetics, Stanford University, Stanford, CA 94305, USA
| | - Arvie Violette
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Char Wynter
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Marta Nieckarz
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Science for Life Laboratory (SciLifeLab), Umeå University, Umeå 90187, Sweden
| | - Samara Paula Mattiello
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA; College of Mathematics and Science, The University of Tennessee Southern, Pulaski, TN 38478, USA
| | - Prabhjot Kaur Sekhon
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA; Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74074, USA; Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN 55905, USA
| | - Francesca Bottacini
- School of Microbiology, University College Cork, Cork, Ireland; Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Lisa Friess
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Hans K Carlson
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA
| | - Daniel P G H Wong
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA
| | - Steven Higginbottom
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Meredith Weglarz
- Stanford Shared FACS Facility, Center for Molecular and Genetic Medicine, Stanford University, Stanford, CA 94305, USA
| | - Weigao Wang
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Benjamin D Knapp
- Biophysics Program, Stanford University, Stanford, CA 94305, USA
| | - Emma Guiberson
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Chemistry and Biochemistry, Middlebury College, Middlebury, VT 05753, USA
| | - Juan Sanchez
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Po-Hsun Huang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Paulo A Garcia
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Cullen R Buie
- Department of Mechanical Engineering, Massachusetts Institute of Technology, 77 Massachusetts Avenue, Cambridge, MA 02139, USA
| | - Benjamin H Good
- Department of Applied Physics, Stanford University, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | | | - Felipe Cava
- Department of Molecular Biology and Laboratory for Molecular Infection Medicine Sweden (MIMS), Umeå Centre for Microbial Research (UCMR), Science for Life Laboratory (SciLifeLab), Umeå University, Umeå 90187, Sweden
| | - Joy Scaria
- Department of Veterinary and Biomedical Sciences, South Dakota State University, Brookings, SD 57007, USA; Department of Veterinary Pathobiology, Oklahoma State University, Stillwater, OK 74074, USA
| | - Justin L Sonnenburg
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Douwe Van Sinderen
- School of Microbiology, University College Cork, Cork, Ireland; APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Adam M Deutschbauer
- Environmental Genomics and Systems Biology Division, Lawrence Berkeley National Laboratory, Berkeley, CA 94720, USA; Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kerwyn Casey Huang
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA; Chan Zuckerberg Biohub, San Francisco, CA 94158, USA.
| |
Collapse
|
6
|
Wang H, Huang J, Fang X, Liu M, Fan X, Li Y. Advances in next-generation sequencing (NGS) applications in drug discovery and development. Expert Opin Drug Discov 2025; 20:537-550. [PMID: 40099494 DOI: 10.1080/17460441.2025.2481262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/27/2025] [Accepted: 03/14/2025] [Indexed: 03/20/2025]
Abstract
INTRODUCTION Drug discovery is a complex and multifaceted process driven by scientific innovation and advanced technologies. Next-Generation Sequencing (NGS) platforms, encompassing both short-read and long-read technologies, have revolutionized the field by enabling the high-throughput and cost-effective analysis of DNA and RNA molecules. Continuous advancements in NGS-based technologies have enabled their seamless integration across preclinical and clinical workflows in drug discovery, encompassing early-stage drug target identification, candidate selection, genetically stratified clinical trials, and pharmacogenetic studies. AREA COVERED This review provides an overview of the current and potential applications of NGS-based technologies in drug discovery and development process, including their roles in novel drug target identification, high-throughput screening, clinical trials, and clinical medication studies. The review is based on literature retrieval from the PubMed and Web of Science databases between 2018 and 2024. EXPERT OPINION As technologies advance rapidly, NGS enhances accuracy and generates vast datasets. These datasets are extensively integrated with other heterogeneous data in systems biology and are mined using machine learning to extract significant insights, thereby driving progress in drug discovery.
Collapse
Affiliation(s)
- Huihong Wang
- Pharmaceutical Department, Chongqing University Three Gorges Hospital, Chongqing University, Chongqing, P. R. China
| | - Jiale Huang
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xianfu Fang
- Pharmaceutical Department, Chongqing University Three Gorges Hospital, Chongqing University, Chongqing, P. R. China
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Mengyao Liu
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xiaohong Fan
- Pharmaceutical Department, Chongqing University Three Gorges Hospital, Chongqing University, Chongqing, P. R. China
| | - Yizhou Li
- Chongqing Key Laboratory of Natural Product Synthesis and Drug Research, Innovative Drug Research Center, School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
7
|
Federici F, Luppino F, Aguilar-Vilar C, Mazaraki ME, Petersen LB, Ahonen L, Nikel PI. CIFR (Clone-Integrate-Flip-out-Repeat): A toolset for iterative genome and pathway engineering of Gram-negative bacteria. Metab Eng 2025; 88:180-195. [PMID: 39778677 DOI: 10.1016/j.ymben.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/01/2025] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Advanced genome engineering enables precise and customizable modifications of bacterial species, and toolsets that exhibit broad-host compatibility are particularly valued owing to their portability. Tn5 transposon vectors have been widely used to establish random integrations of desired DNA sequences into bacterial genomes. However, the iteration of the procedure remains challenging because of the limited availability and reusability of selection markers. We addressed this challenge with CIFR, a mini-Tn5 integration system tailored for iterative genome engineering. The pCIFR vectors incorporate attP and attB sites flanking an antibiotic resistance marker used to select for the insertion. Subsequent removal of antibiotic determinants is facilitated by the Bxb1 integrase paired to a user-friendly counter-selection marker, both encoded in auxiliary plasmids. CIFR delivers engineered strains harboring stable DNA insertions and free of any antibiotic resistance cassette, allowing for the reusability of the tool. The system was validated in Pseudomonas putida, Escherichia coli, and Cupriavidus necator, underscoring its portability across diverse industrially relevant hosts. The CIFR toolbox was calibrated through combinatorial integrations of chromoprotein genes in P. putida, generating strains displaying a diverse color palette. We also introduced a carotenoid biosynthesis pathway in P. putida in a two-step engineering process, showcasing the potential of the tool for pathway balancing. The broad utility of the CIFR toolbox expands the toolkit for metabolic engineering, allowing for the construction of complex phenotypes while opening new possibilities in bacterial genetic manipulations.
Collapse
Affiliation(s)
- Filippo Federici
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Francesco Luppino
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Clara Aguilar-Vilar
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Maria Eleni Mazaraki
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lars Boje Petersen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Linda Ahonen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Pablo I Nikel
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
8
|
Sakenova N, Cacace E, Orakov A, Huber F, Varik V, Kritikos G, Michiels J, Bork P, Cossart P, Goemans CV, Typas A. Systematic mapping of antibiotic cross-resistance and collateral sensitivity with chemical genetics. Nat Microbiol 2025; 10:202-216. [PMID: 39623067 PMCID: PMC11726442 DOI: 10.1038/s41564-024-01857-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 10/13/2024] [Indexed: 01/12/2025]
Abstract
By acquiring or evolving resistance to one antibiotic, bacteria can become cross-resistant to a second antibiotic, which further limits therapeutic choices. In the opposite scenario, initial resistance leads to collateral sensitivity to a second antibiotic, which can inform cycling or combinatorial treatments. Despite their clinical relevance, our knowledge of both interactions is limited. We used published chemical genetics data of the Escherichia coli single-gene deletion library in 40 antibiotics and devised a metric that discriminates between known cross-resistance and collateral-sensitivity antibiotic interactions. Thereby we inferred 404 cases of cross-resistance and 267 of collateral-sensitivity, expanding the number of known interactions by over threefold. We further validated 64/70 inferred interactions using experimental evolution. By identifying mutants driving these interactions in chemical genetics, we demonstrated that a drug pair can exhibit both interactions depending on the resistance mechanism. Finally, we applied collateral-sensitive drug pairs in combination to reduce antibiotic-resistance development in vitro.
Collapse
Affiliation(s)
- Nazgul Sakenova
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Center for Microbiology, VIB-KU Leuven, Leuven, Belgium
- Center of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
| | - Elisabetta Cacace
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Institute of Microbiology and Swiss Institute of Bioinformatics, ETH Zürich, Zürich, Switzerland
| | - Askarbek Orakov
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Florian Huber
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Vallo Varik
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - George Kritikos
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- European Food Safety Authority, Parma, Italy
| | - Jan Michiels
- Center for Microbiology, VIB-KU Leuven, Leuven, Belgium
- Center of Microbial and Plant Genetics, KU Leuven, Leuven, Belgium
| | - Peer Bork
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Department of Bioinformatics, University of Würzburg, Würzburg, Germany
- Max Delbrück Centre for Molecular Medicine, Berlin, Germany
| | - Pascale Cossart
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
- Department of Cell Biology and Infection, Institut Pasteur, Paris, France
| | - Camille V Goemans
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
- Global Health Institute, School of Life Sciences, École Polytechnique Federale de Lausanne, Lausanne, Switzerland.
| | - Athanasios Typas
- Genome Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| |
Collapse
|
9
|
Russell BJ, Verma M, Maier NK, Jost M. Dissecting host-microbe interactions with modern functional genomics. Curr Opin Microbiol 2024; 82:102554. [PMID: 39368241 PMCID: PMC11609025 DOI: 10.1016/j.mib.2024.102554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 10/07/2024]
Abstract
Interrogation of host-microbe interactions has long been a source of both basic discoveries and benefits to human health. Here, we review the role that functional genomics approaches have played in such efforts, with an emphasis on recent examples that have harnessed technological advances to provide mechanistic insight at increased scale and resolution. Finally, we discuss how concurrent innovations in model systems and genetic tools have afforded opportunities to interrogate additional types of host-microbe relationships, such as those in the mammalian gut. Bringing these innovations together promises many exciting discoveries ahead.
Collapse
Affiliation(s)
- Baylee J Russell
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Manasvi Verma
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Nolan K Maier
- Department of Microbiology, Harvard Medical School, Boston, MA, USA
| | - Marco Jost
- Department of Microbiology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
10
|
Bacon EE, Myers KS, Iruegas-López R, Banta AB, Place M, Ebersberger I, Peters JM. Physiological Roles of an Acinetobacter-specific σ Factor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.08.602572. [PMID: 39026751 PMCID: PMC11257525 DOI: 10.1101/2024.07.08.602572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
The Gram-negative pathogen Acinetobacter baumannii is considered an "urgent threat" to human health due to its propensity to become antibiotic resistant. Understanding the distinct regulatory paradigms used by A. baumannii to mitigate cellular stresses may uncover new therapeutic targets. Many γ-proteobacteria use the extracytoplasmic function (ECF) σ factor, RpoE, to invoke envelope homeostasis networks in response to stress. Acinetobacter species contain the poorly characterized ECF "SigAb;" however, it is unclear if SigAb has the same physiological role as RpoE. Here, we show that SigAb is a metal stress-responsive ECF that appears unique to Acinetobacter species and distinct from RpoE. We combine promoter mutagenesis, motif scanning, and ChIP-seq to define the direct SigAb regulon, which consists of sigAb itself, the stringent response mediator, relA, and the uncharacterized small RNA, "sabS." However, RNA-seq of strains overexpressing SigAb revealed a large, indirect regulon containing hundreds of genes. Metal resistance genes are key elements of the indirect regulon, as CRISPRi knockdown of sigAb or sabS resulted in increased copper sensitivity and excess copper induced SigAb-dependent transcription. Further, we found that two uncharacterized genes in the sigAb operon, "aabA" and "aabB", have anti-SigAb activity. Finally, employing a targeted Tn-seq approach that uses CRISPR-associated transposons, we show that sigAb, aabA, and aabB are important for fitness even during optimal growth conditions. Our work reveals new physiological roles for SigAb and SabS, provides a novel approach for assessing gene fitness, and highlights the distinct regulatory architecture of A. baumannii.
Collapse
Affiliation(s)
- Emily E. Bacon
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
- Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI, USA
| | - Kevin S. Myers
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Rubén Iruegas-López
- Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Germany
| | - Amy B. Banta
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Michael Place
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, USA
| | - Ingo Ebersberger
- Institute of Cell Biology and Neuroscience, Goethe University Frankfurt, Germany
- Senckenberg Biodiversity and Climate Research Centre (S-BIKF), Frankfurt, Germany
- LOEWE Center for Translational Biodiversity Genomics (TBG), Frankfurt, Germany
| | - Jason M. Peters
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, USA
- Great Lakes Bioenergy Research Center, University of Wisconsin-Madison, Madison, WI, USA
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, WI, USA
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
11
|
Wang S, Gong X, Xiao F, Yang Y. Recent advances in host-focused molecular tools for investigating host-gut microbiome interactions. Front Microbiol 2024; 15:1335036. [PMID: 38605718 PMCID: PMC11007152 DOI: 10.3389/fmicb.2024.1335036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 03/18/2024] [Indexed: 04/13/2024] Open
Abstract
Microbial communities in the human gut play a significant role in regulating host gene expression, influencing a variety of biological processes. To understand the molecular mechanisms underlying host-microbe interactions, tools that can dissect signaling networks are required. In this review, we discuss recent advances in molecular tools used to study this interplay, with a focus on those that explore how the microbiome regulates host gene expression. These tools include CRISPR-based whole-body genetic tools for deciphering host-specific genes involved in the interaction process, Cre-loxP based tissue/cell-specific gene editing approaches, and in vitro models of host-derived organoids. Overall, the application of these molecular tools is revolutionizing our understanding of how host-microbiome interactions contribute to health and disease, paving the way for improved therapies and interventions that target microbial influences on the host.
Collapse
Affiliation(s)
- Siyao Wang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| | - Xu Gong
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| | - Fei Xiao
- Department of Thoracic Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Yun Yang
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, Beijing Advanced Innovation Centre for Biomedical Engineering, School of Engineering Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Ministry of Industry and Information Technology, Beihang University, Beijing, China
| |
Collapse
|