1
|
Kaur N, Sahoo J, De M. Development of Nanomaterials-Based Agents for Selective Antibacterial Activity. Chembiochem 2025; 26:e202400693. [PMID: 39632741 DOI: 10.1002/cbic.202400693] [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: 08/20/2024] [Revised: 10/26/2024] [Indexed: 12/07/2024]
Abstract
Bacterial infections continue to threaten public health due to limitations in rapid and accurate diagnostic techniques. While broad-spectrum antibiotics offer empirical treatment, their overuse has fuelled the emergence of antimicrobial resistance (AMR) pathogens, posing a critical global public health challenge. In this critical scenario, nanomaterial-based antibacterial agents emerge as a promising solution to combat bacteria and inhibit their proliferation. However, selective elimination of pathogenic bacteria is paramount. This review highlights recent advancements in developing nanomaterials for selective antibacterial activity. We categorize these agents based on their mode of action, exploring how they selectively interact with bacteria and their potential antibacterial mechanisms. This review offers crucial insights for researchers exploring the potential of nanotechnology to address the growing threat of AMR.
Collapse
Affiliation(s)
- Navjot Kaur
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Jagabandhu Sahoo
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India
| | - Mrinmoy De
- Department of Organic Chemistry, Indian Institute of Science, Bangalore, 560012, India
| |
Collapse
|
2
|
Wei T, Lai W, Chen Q, Sun C. Engineered Phage Enables Efficient Control of Gene Expression upon Infection of the Host Cell. Int J Mol Sci 2024; 26:250. [PMID: 39796105 PMCID: PMC11720261 DOI: 10.3390/ijms26010250] [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: 12/08/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Recently, we developed a spatial phage-assisted continuous evolution (SPACE) system. This system utilizes chemotaxis coupled with the growth of motile bacteria during their spatial range expansion in soft agar to provide fresh host cells for iterative phage infection and selection pressure for preserving evolved genes of interest carried by phage mutants. Controllable mutagenesis activated only in a subpopulation of the migrating cells is essential in this system to efficiently generate mutated progeny phages from which desired individuals are selected during the directed evolution process. But, the widely adopted small molecule-dependent inducible system could hardly fulfill this purpose because it always affects all cells homogeneously. In this study, we developed a phage infection-induced gene expression system using modified Escherichia coli (E. coli) phage shock protein operon or sigma factors from Bacillus subtilis. Results showed that this system enabled efficient control of gene expression upon phage infection with dynamic output ranges from small to large using combinations of different engineered phages and corresponding promoters. This system was incorporated into SPACE to function as a phage infection-induced mutagenesis module and successfully facilitated the evolution of T7 RNA polymerase, which generated diverse mutants with altered promoter recognition specificity. We expect that phage infection-induced gene expression system could be further extended to more applications involving partial induction in a portion of a population and targeted induction in specific strains among a mixed bacterial community, which provides an important complement to small molecule-dependent inducible systems.
Collapse
Affiliation(s)
- Ting Wei
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.L.); (Q.C.); (C.S.)
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wangsheng Lai
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.L.); (Q.C.); (C.S.)
| | - Qian Chen
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.L.); (Q.C.); (C.S.)
| | - Chenjian Sun
- CAS Key Laboratory for Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; (W.L.); (Q.C.); (C.S.)
| |
Collapse
|
3
|
Pujar A, Pathania A, Hopper C, Pandi A, Calderón CR, Függer M, Nowak T, Kushwaha M. Phage-mediated intercellular CRISPRi for biocomputation in bacterial consortia. Nucleic Acids Res 2024:gkae1256. [PMID: 39727169 DOI: 10.1093/nar/gkae1256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 11/30/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024] Open
Abstract
Coordinated actions of cells in microbial communities and multicellular organisms enable them to perform complex tasks otherwise difficult for single cells. This has inspired biological engineers to build cellular consortia for larger circuits with improved functionalities while implementing communication systems for coordination among cells. Here, we investigate the signalling dynamics of a phage-mediated synthetic DNA messaging system and couple it with CRISPR interference to build distributed circuits that perform logic gate operations in multicellular bacterial consortia. We find that growth phases of both sender and receiver cells, as well as resource competition between them, shape communication outcomes. Leveraging the easy programmability of DNA messages, we build eight orthogonal signals and demonstrate that intercellular CRISPRi (i-CRISPRi) regulates gene expression across cells. Finally, we multiplex the i-CRISPRi system to implement several multicellular logic gates that involve up to seven cells and take up to three inputs simultaneously, with single- and dual-rail encoding: NOT, YES, AND and AND-AND-NOT. The communication system developed here lays the groundwork for implementing complex biological circuits in engineered bacterial communities, using phage signals for communication.
Collapse
Affiliation(s)
- Abhinav Pujar
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Amit Pathania
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Corbin Hopper
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
- Université Paris-Saclay, CNRS, ENS Paris-Saclay, Laboratoire Méthodes Formelles, 91190 Gif-sur-Yvette, France
- Université Paris-Saclay, CNRS, Laboratoire Interdisciplinaire des Sciences du Numérique, 91405 Orsay, France
| | - Amir Pandi
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Cristian Ruiz Calderón
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| | - Matthias Függer
- Université Paris-Saclay, CNRS, ENS Paris-Saclay, Laboratoire Méthodes Formelles, 91190 Gif-sur-Yvette, France
| | - Thomas Nowak
- Université Paris-Saclay, CNRS, ENS Paris-Saclay, Laboratoire Méthodes Formelles, 91190 Gif-sur-Yvette, France
- Institut Universitaire de France, Paris, France
| | - Manish Kushwaha
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350 Jouy-en-Josas, France
| |
Collapse
|
4
|
Guo Y, Dong X, Li H, Tong Y, Liu Z, Jin J. Cyanophage Engineering for Algal Blooms Control. Viruses 2024; 16:1745. [PMID: 39599859 PMCID: PMC11598953 DOI: 10.3390/v16111745] [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: 09/26/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/29/2024] Open
Abstract
Cyanobacteria represent a prevalent category of photosynthetic autotrophs capable of generating deleterious algal blooms, commonly known as cyanobacteria harmful algal blooms (cyanoHABs). These blooms often produce cyanotoxins, which pose risks to public health and ecosystems by contaminating surface waters and drinking water sources. Traditional treatment methods have limited effectiveness. Therefore, there is an urgent need for a new approach to effectively manage cyanoHABs. One promising approach is the use of cyanophages, which are viruses that specifically target cyanobacteria. Cyanophages serve as an effective biological control method for reducing cyanoHABs in aquatic systems. By engineering cyanophages, it is possible to develop a highly specific control strategy that minimally impacts non-target species and their propagation in the environment. This review explores the potential application of cyanophages as a strategy for controlling cyanoHABs. It includes the identification and isolation of broad-spectrum and novel cyanophages, with a specific focus on freshwater Microcystis cyanophages, highlighting their broad spectrum and high efficiency. Additionally, recent advancements in cyanophage engineering are discussed, including genome modification, functional gene identification, and the construction of artificial cyanophages. Furthermore, the current state of application is addressed. Cyanophage is a promising control strategy for effectively managing cyanoHABs in aquatic environments.
Collapse
Affiliation(s)
- Yujing Guo
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaoxiao Dong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Huiying Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yigang Tong
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zihe Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
- State Key Laboratory of Chemical Resource Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jin Jin
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
5
|
Li FY, Tan XE, Shimamori Y, Kiga K, Veeranarayanan S, Watanabe S, Nishikawa Y, Aiba Y, Sato'o Y, Miyanaga K, Sasahara T, Hossain S, Thitiananpakorn K, Kawaguchi T, Nguyen HM, Yeo Syin Lian A, Sultana S, Alessa O, Kumwenda G, Sarangi J, Revilleza JEC, Baranwal P, Faruk MO, Hidaka Y, Thu M, Arbaah M, Batbold A, Maniruzzaman, Liu Y, Duyen HTM, Sugano T, Tergel N, Shimojyo T, Cui L. Phagemid-based capsid system for CRISPR-Cas13a antimicrobials targeting methicillin-resistant Staphylococcus aureus. Commun Biol 2024; 7:1129. [PMID: 39271957 PMCID: PMC11399120 DOI: 10.1038/s42003-024-06754-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: 12/06/2023] [Accepted: 08/20/2024] [Indexed: 09/15/2024] Open
Abstract
In response to the escalating antibiotic resistance in multidrug-resistant pathogens, we propose an innovative phagemid-based capsid system to generate CRISPR-Cas13a-loaded antibacterial capsids (AB-capsids) for targeted therapy against multidrug-resistant Staphylococcus aureus. Our optimized phagemid system maximizes AB-capsid yield and purity, showing a positive correlation with phagemid copy number. Notably, an 8.65-fold increase in copy number results in a 2.54-fold rise in AB-capsid generation. Phagemids carrying terL-terS-rinA-rinB (prophage-encoded packaging site genes) consistently exhibit high packaging efficiency, and the generation of AB-capsids using lysogenized hosts with terL-terS deletion resulted in comparatively lower level of wild-type phage contamination, with minimal compromise on AB-capsid yield. These generated AB-capsids selectively eliminate S. aureus strains carrying the target gene while sparing non-target strains. In conclusion, our phagemid-based capsid system stands as a promising avenue for developing sequence-specific bactericidal agents, offering a streamlined approach to combat antibiotic-resistant pathogens within the constraints of efficient production and targeted efficacy.
Collapse
Affiliation(s)
- Feng-Yu Li
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Xin-Ee Tan
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Yuzuki Shimamori
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Kotaro Kiga
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Srivani Veeranarayanan
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Shinya Watanabe
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Yutaro Nishikawa
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
- EIKEN CHEMICAL CO.,LTD., Shimotsuga District, Tochigi, 329-0114, Japan
| | - Yoshifumi Aiba
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Yusuke Sato'o
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Kazuhiko Miyanaga
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Teppei Sasahara
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Sarah Hossain
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Kanate Thitiananpakorn
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Tomofumi Kawaguchi
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Huong Minh Nguyen
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Adeline Yeo Syin Lian
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Sharmin Sultana
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Ola Alessa
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Geoffrey Kumwenda
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Jayathilake Sarangi
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Jastin Edrian Cocuangco Revilleza
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Priyanka Baranwal
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Mohammad Omar Faruk
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Yuya Hidaka
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Myat Thu
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Mahmoud Arbaah
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Anujin Batbold
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Maniruzzaman
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Yi Liu
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Ho Thi My Duyen
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Takashi Sugano
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Nayanjin Tergel
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan
| | - Takayuki Shimojyo
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo, 162-8640, Japan
| | - Longzhu Cui
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke city, Tochigi, 329-0498, Japan.
| |
Collapse
|
6
|
Cui L, Watanabe S, Miyanaga K, Kiga K, Sasahara T, Aiba Y, Tan XE, Veeranarayanan S, Thitiananpakorn K, Nguyen HM, Wannigama DL. A Comprehensive Review on Phage Therapy and Phage-Based Drug Development. Antibiotics (Basel) 2024; 13:870. [PMID: 39335043 PMCID: PMC11428490 DOI: 10.3390/antibiotics13090870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 09/06/2024] [Accepted: 09/08/2024] [Indexed: 09/30/2024] Open
Abstract
Phage therapy, the use of bacteriophages (phages) to treat bacterial infections, is regaining momentum as a promising weapon against the rising threat of multidrug-resistant (MDR) bacteria. This comprehensive review explores the historical context, the modern resurgence of phage therapy, and phage-facilitated advancements in medical and technological fields. It details the mechanisms of action and applications of phages in treating MDR bacterial infections, particularly those associated with biofilms and intracellular pathogens. The review further highlights innovative uses of phages in vaccine development, cancer therapy, and as gene delivery vectors. Despite its targeted and efficient approach, phage therapy faces challenges related to phage stability, immune response, and regulatory approval. By examining these areas in detail, this review underscores the immense potential and remaining hurdles in integrating phage-based therapies into modern medical practices.
Collapse
Affiliation(s)
- Longzhu Cui
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Shinya Watanabe
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Kazuhiko Miyanaga
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Kotaro Kiga
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
- Research Center for Drug and Vaccine Development, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| | - Teppei Sasahara
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Yoshifumi Aiba
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Xin-Ee Tan
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Srivani Veeranarayanan
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Kanate Thitiananpakorn
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Huong Minh Nguyen
- Division of Bacteriology, Department of Infection and Immunity, School of Medicine, Jichi Medical University, Shimotsuke City 329-0498, Japan
| | - Dhammika Leshan Wannigama
- Department of Infectious Diseases and Infection Control, Yamagata Prefectural Central Hospital, Yamagata 990-2292, Japan
| |
Collapse
|
7
|
Guo Y, Dong X, Li H, Lin W, Cao L, Li D, Zhang Y, Jin J, Tong Y, Liu Z. Efficient Broad-Spectrum Cyanophage Function Module Mining. Microorganisms 2024; 12:1578. [PMID: 39203420 PMCID: PMC11356776 DOI: 10.3390/microorganisms12081578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 07/28/2024] [Accepted: 07/30/2024] [Indexed: 09/03/2024] Open
Abstract
Cyanobacterial harmful algal blooms (CyanoHABs) cause health and environmental effects worldwide. Cyanophage is a virus that exclusively infects cyanobacteria. Using cyanophages to control blooms is the latest biological control method. However, little research on the genomics of cyanophages and the presence of numerous proteins with unidentified functions in cyanophage genomes pose challenges for their practical application and comprehensive investigation. We selected the broad-spectrum and efficient cyanophage YongM for our study. On the one hand, through rational analysis, we analyze essential genes, establish the minimal cyanophage genome and single essential gene modules, and examine the impact of essential modules on growth. Additionally, we conducted ultraviolet mutagenesis on YongM to generate more efficient cyanophages' critical modules through random mutagenesis. Then, we sequenced and analyzed the functionality of the mutational gene modules. These findings highlight several gene modules that contribute to a deeper understanding of the functional components within cyanophage genomes.
Collapse
Affiliation(s)
- Yujing Guo
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Xiaoxiao Dong
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Huiying Li
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wei Lin
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Lei Cao
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Dengfeng Li
- Key Laboratory of Marine Biotechnology, School of Marine Sciences, Ningbo University, Ningbo 315211, China;
| | - Yiming Zhang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jin Jin
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yigang Tong
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Zihe Liu
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing University of Chemical Technology, Beijing 100029, China
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
8
|
Lewis JM, Williams J, Sagona AP. Making the leap from technique to treatment - genetic engineering is paving the way for more efficient phage therapy. Biochem Soc Trans 2024; 52:1373-1384. [PMID: 38716972 PMCID: PMC11346441 DOI: 10.1042/bst20231289] [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: 01/16/2024] [Revised: 03/30/2024] [Accepted: 04/29/2024] [Indexed: 06/27/2024]
Abstract
Bacteriophages (phages) are viruses specific to bacteria that target them with great efficiency and specificity. Phages were first studied for their antibacterial potential in the early twentieth century; however, their use was largely eclipsed by the popularity of antibiotics. Given the surge of antimicrobial-resistant strains worldwide, there has been a renaissance in harnessing phages as therapeutics once more. One of the key advantages of phages is their amenability to modification, allowing the generation of numerous derivatives optimised for specific functions depending on the modification. These enhanced derivatives could display higher infectivity, expanded host range or greater affinity to human tissues, where some bacterial species exert their pathogenesis. Despite this, there has been a noticeable discrepancy between the generation of derivatives in vitro and their clinical application in vivo. In most instances, phage therapy is only used on a compassionate-use basis, where all other treatment options have been exhausted. A lack of clinical trials and numerous regulatory hurdles hamper the progress of phage therapy and in turn, the engineered variants, in becoming widely used in the clinic. In this review, we outline the various types of modifications enacted upon phages and how these modifications contribute to their enhanced bactericidal function compared with wild-type phages. We also discuss the nascent progress of genetically modified phages in clinical trials along with the current issues these are confronted with, to validate it as a therapy in the clinic.
Collapse
Affiliation(s)
| | - Joshua Williams
- School of Life Sciences, University of Warwick, Coventry, U.K
| | | |
Collapse
|
9
|
Fu Y, Li J, Cai W, Huang Y, Liu X, Ma Z, Tang Z, Bian X, Zheng J, Jiang J, Li C. The emerging tumor microbe microenvironment: From delineation to multidisciplinary approach-based interventions. Acta Pharm Sin B 2024; 14:1560-1591. [PMID: 38572104 PMCID: PMC10985043 DOI: 10.1016/j.apsb.2023.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 10/20/2023] [Accepted: 11/03/2023] [Indexed: 04/05/2024] Open
Abstract
Intratumoral microbiota has become research hotspots, and emerges as a non-negligent new component of tumor microenvironments (TME), due to its powerful influence on tumor initiation, metastasis, immunosurveillance and prognosis despite in low-biomass. The accumulations of microbes, and their related components and metabolites within tumor tissues, endow TME with additional pluralistic features which are distinct from the conventional one. Therefore, it's definitely necessary to comprehensively delineate the sophisticated landscapes of tumor microbe microenvironment, as well as their functions and related underlying mechanisms. Herein, in this review, we focused on the fields of tumor microbe microenvironment, including the heterogeneity of intratumor microbiota in different types of tumors, the controversial roles of intratumoral microbiota, the basic features of tumor microbe microenvironment (i.e., pathogen-associated molecular patterns (PAMPs), typical microbial metabolites, autophagy, inflammation, multi-faceted immunomodulation and chemoresistance), as well as the multidisciplinary approach-based intervention of tumor microbiome for cancer therapy by applying wild-type or engineered live microbes, microbiota metabolites, antibiotics, synthetic biology and rationally designed biomaterials. We hope our work will provide valuable insight to deeply understand the interplay of cancer-immune-microbial, and facilitate the development of microbes-based tumor-specific treatments.
Collapse
Affiliation(s)
- Yu Fu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Jia Li
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Wenyun Cai
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Yulan Huang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xinlong Liu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongyi Ma
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Zhongjie Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Xufei Bian
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Jiayun Jiang
- Institute of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| |
Collapse
|
10
|
Mahdizade Ari M, Dadgar L, Elahi Z, Ghanavati R, Taheri B. Genetically Engineered Microorganisms and Their Impact on Human Health. Int J Clin Pract 2024; 2024:6638269. [PMID: 38495751 PMCID: PMC10944348 DOI: 10.1155/2024/6638269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 11/20/2023] [Accepted: 02/12/2024] [Indexed: 03/19/2024] Open
Abstract
The emergence of antibiotic-resistant strains, the decreased effectiveness of conventional therapies, and the side effects have led researchers to seek a safer, more cost-effective, patient-friendly, and effective method that does not develop antibiotic resistance. With progress in synthetic biology and genetic engineering, genetically engineered microorganisms effective in treatment, prophylaxis, drug delivery, and diagnosis have been developed. The present study reviews the types of genetically engineered bacteria and phages, their impacts on diseases, cancer, and metabolic and inflammatory disorders, the biosynthesis of these modified strains, the route of administration, and their effects on the environment. We conclude that genetically engineered microorganisms can be considered promising candidates for adjunctive treatment of diseases and cancers.
Collapse
Affiliation(s)
- Marzie Mahdizade Ari
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Leila Dadgar
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Elahi
- Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran
| | | | - Behrouz Taheri
- Department of Biotechnology, School of Medicine, Ahvaz Jundishapour University of medical Sciences, Ahvaz, Iran
| |
Collapse
|
11
|
Ipoutcha T, Racharaks R, Huttelmaier S, Wilson CJ, Ozer EA, Hartmann EM. A synthetic biology approach to assemble and reboot clinically relevant Pseudomonas aeruginosa tailed phages. Microbiol Spectr 2024; 12:e0289723. [PMID: 38294230 PMCID: PMC10913387 DOI: 10.1128/spectrum.02897-23] [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: 08/04/2023] [Accepted: 12/17/2023] [Indexed: 02/01/2024] Open
Abstract
The rise in the frequency of antibiotic resistance has made bacterial infections, specifically Pseudomonas aeruginosa, a cause for greater concern. Phage therapy is a promising solution that uses naturally isolated phages to treat bacterial infections. Ecological limitations, which stipulate a discrete host range and the inevitable evolution of resistance, may be overcome through a better understanding of phage biology and the utilization of engineered phages. In this study, we developed a synthetic biology approach to construct tailed phages that naturally target clinically relevant strains of Pseudomonas aeruginosa. As proof of concept, we successfully cloned and assembled the JG024 and DMS3 phage genomes in yeast using transformation-associated recombination cloning and rebooted these two phage genomes in two different strains of P. aeruginosa. We identified factors that affected phage reboot efficiency like the phage species or the presence of antiviral defense systems in the bacterial strain. We have successfully extended this method to two other phage species and observed that the method enables the reboot of phages that are naturally unable to infect the strain used for reboot. This research represents a critical step toward the construction of clinically relevant, engineered P. aeruginosa phages.IMPORTANCEPseudomonas aeruginosa is a bacterium responsible for severe infections and a common major complication in cystic fibrosis. The use of antibiotics to treat bacterial infections has become increasingly difficult as antibiotic resistance has become more prevalent. Phage therapy is an alternative solution that is already being used in some European countries, but its use is limited by the narrow host range due to the phage receptor specificity, the presence of antiviral defense systems in the bacterial strain, and the possible emergence of phage resistance. In this study, we demonstrate the use of a synthetic biology approach to construct and reboot clinically relevant P. aeruginosa tailed phages. This method enables a significant expansion of possibilities through the construction of engineered phages for therapy applications.
Collapse
Affiliation(s)
- Thomas Ipoutcha
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Ratanachat Racharaks
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Stefanie Huttelmaier
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Cole J. Wilson
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
| | - Egon A. Ozer
- Division of Infectious Diseases, Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - Erica M. Hartmann
- Department of Civil and Environmental Engineering, Northwestern University, Evanston, Illinois, USA
- Center for Synthetic Biology, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
12
|
Kang D, Bagchi D, Chen IA. Pharmacokinetics and Biodistribution of Phages and their Current Applications in Antimicrobial Therapy. ADVANCED THERAPEUTICS 2024; 7:2300355. [PMID: 38933919 PMCID: PMC11198966 DOI: 10.1002/adtp.202300355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Indexed: 06/28/2024]
Abstract
Antimicrobial resistance remains a critical global health concern, necessitating the investigation of alternative therapeutic approaches. With the diminished efficacy of conventional small molecule drugs due to the emergence of highly resilient bacterial strains, there is growing interest in the potential for alternative therapeutic modalities. As naturally occurring viruses of bacteria, bacteriophage (or phage) are being re-envisioned as a platform to engineer properties that can be tailored to target specific bacterial strains and employ diverse antibacterial mechanisms. However, limited understanding of key pharmacological properties of phage is a major challenge to translating its use from preclinical to clinical settings. Here, we review modern advancements in phage-based antimicrobial therapy and discuss the in vivo pharmacokinetics and biodistribution of phage, addressing critical challenges in their application that must be overcome for successful clinical implementation.
Collapse
Affiliation(s)
- Dayeon Kang
- Department of Chemical and Biomolecular Engineering, Department of Chemistry and Biochemistry, University of California, Los Angeles, 90024 USA
| | - Damayanti Bagchi
- Department of Chemical and Biomolecular Engineering, Department of Chemistry and Biochemistry, University of California, Los Angeles, 90024 USA
| | - Irene A. Chen
- Department of Chemical and Biomolecular Engineering, Department of Chemistry and Biochemistry, University of California, Los Angeles, 90024 USA
| |
Collapse
|
13
|
Chen C, Chen L, Mao C, Jin L, Wu S, Zheng Y, Cui Z, Li Z, Zhang Y, Zhu S, Jiang H, Liu X. Natural Extracts for Antibacterial Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306553. [PMID: 37847896 DOI: 10.1002/smll.202306553] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/23/2023] [Indexed: 10/19/2023]
Abstract
Bacteria-induced epidemics and infectious diseases are seriously threatening the health of people around the world. In addition, antibiotic therapy has been inducing increasingly more serious bacterial resistance, which makes it urgent to develop new treatment strategies to combat bacteria, including multidrug-resistant bacteria. Natural extracts displaying antibacterial activity and good biocompatibility have attracted much attention due to greater concerns about the safety of synthetic chemicals and emerging drug resistance. These antibacterial components can be isolated and utilized as antimicrobials, as well as transformed, combined, or wrapped with other substances by using modern assistive technologies to fight bacteria synergistically. This review summarizes recent advances in natural extracts from three kinds of sources-plants, animals, and microorganisms-for antibacterial applications. This work discusses the corresponding antibacterial mechanisms and the future development of natural extracts in antibacterial fields.
Collapse
Affiliation(s)
- Cuihong Chen
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Lin Chen
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Congyang Mao
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
| | - Liguo Jin
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Shuilin Wu
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Yufeng Zheng
- School of Materials Science & Engineering, Peking University, Yiheyuan Road 5#, Beijing, 100871, China
| | - Zhenduo Cui
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Zhaoyang Li
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Yu Zhang
- Department of Orthopedics, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Shengli Zhu
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Hui Jiang
- School of Materials Science & Engineering, the Key Laboratory of Advanced Ceramics and Machining Technology by the Ministry of Education of China, Tianjin University, Yaguan Road 135#, Tianjin, 300072, China
| | - Xiangmei Liu
- Biomedical Materials Engineering Research Center, Hubei Key Laboratory of Polymer Materials, Ministry-of-Education Key Laboratory for the Green Preparation and Application of Functional Materials, School of Materials Science & Engineering, State Key Laboratory of Biocatalysis and Enzyme Engineering, Hubei University, Wuhan, 430062, China
- School of Health Science & Biomedical Engineering, Hebei University of Technology, Xiping Avenue 5340#, Tianjin, 300401, China
| |
Collapse
|
14
|
Schmitt DS, Siegel SD, Selle K. Applications of designer phage encoding recombinant gene payloads. Trends Biotechnol 2024; 42:326-338. [PMID: 37833198 DOI: 10.1016/j.tibtech.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/17/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023]
Abstract
Advances in genetic engineering, synthetic biology, and DNA sequencing have transformed the re-emergent therapeutic bacteriophage field. The increasing rate of multidrug resistant (MDR) infections and the speed at which new bacteriophages can be isolated, sequenced, characterized, and engineered has reinvigorated phage therapy and unlocked new applications of phages for modulating bacteria. The methods used to genetically engineer bacteriophages are undergoing significant development, but identification of heterologous gene payloads with desirable activity and determination of their impact on bacteria or human cells in translationally relevant applications remain underexplored areas. Here, we discuss and categorize recombinant gene payloads for their potential outcome on phage-bacteria interactions when genetically engineered into phage genomes for expression in their bacterial hosts.
Collapse
Affiliation(s)
- Daniel S Schmitt
- Biomanufacturing Training and Education Center, North Carolina State University, Raleigh, NC, USA
| | - Sara D Siegel
- Biomanufacturing Training and Education Center, North Carolina State University, Raleigh, NC, USA
| | - Kurt Selle
- Biomanufacturing Training and Education Center, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
15
|
Boroujeni MB, Mohebi S, Malekian A, Shahraeini SS, Gharagheizi Z, Shahkolahi S, Sadeghi RV, Naderifar M, Akbarizadeh MR, Soltaninejad S, Moghadam ZT, Moghadam MT, Mirzadeh F. The therapeutic effect of engineered phage, derived protein and enzymes against superbug bacteria. Biotechnol Bioeng 2024; 121:82-99. [PMID: 37881139 DOI: 10.1002/bit.28581] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 09/18/2023] [Accepted: 10/15/2023] [Indexed: 10/27/2023]
Abstract
Defending against antibiotic-resistant infections is similar to fighting a war with limited ammunition. As the new century unfolded, antibiotic resistance became a significant concern. In spite of the fact that phage treatment has been used as an effective means of fighting infections for more than a century, researchers have had to overcome many challenges of superbug bacteria by manipulating phages and producing engineered enzymes. New enzymes and phages with enhanced properties have a significant impact on the ability to fight antibiotic-resistant infections, which is considered a window of hope for the future. This review, therefore, illustrates not only the challenges caused by antibiotic resistance and superbug bacteria but also the engineered enzymes and phages that are being developed to solve these issues. Our study found that engineered phages, phage proteins, and enzymes can be effective in treating superbug bacteria and destroying the biofilm caused by them. Combining these engineered compounds with other antimicrobial substances can increase their effectiveness against antibiotic-resistant bacteria. Therefore, engineered phages, proteins, and enzymes can be used as a substitute for antibiotics or in combination with antibiotics to treat patients with superbug infections in the future.
Collapse
Affiliation(s)
| | - Samane Mohebi
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Azam Malekian
- Department of Parasitology, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Sadegh Shahraeini
- Department of Medical Biotechnology, Drug Design and Bioinformatics Unit, Biotechnology Research Centre, Pasteur Institute of Iran, Tehran, Iran
| | - Zahra Gharagheizi
- Department of Biology, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Shaghayegh Shahkolahi
- Department of Microbiology, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Rezvaneh Vahedian Sadeghi
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahin Naderifar
- School of Nursing & Midwifery, Zabol University of Medical Sciences, Zabol, Iran
| | | | | | - Zahra Taati Moghadam
- School of Nursing and Midwifery, Guilan University of Medical Sciences, Rasht, Iran
| | | | | |
Collapse
|
16
|
Fadaie M, Dianat-Moghadam H, Ghafouri E, Naderi S, Darvishali MH, Ghovvati M, Khanahmad H, Boshtam M, Makvandi P. Unraveling the potential of M13 phages in biomedicine: Advancing drug nanodelivery and gene therapy. ENVIRONMENTAL RESEARCH 2023; 238:117132. [PMID: 37714365 DOI: 10.1016/j.envres.2023.117132] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 09/05/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023]
Abstract
M13 phages possessing filamentous phage genomes offer the benefits of selective display of molecular moieties and delivery of therapeutic agent payloads with a tolerable safety profile. M13 phage-displayed technology for resembling antigen portions led to the discovery of mimetic epitopes that applied to antibody-based therapy and could be useful in the design of anticancer vaccines. To date, the excremental experiences have engaged the M13 phage in the development of innovative biosensors for detecting biospecies, biomolecules, and human cells with an acceptable limit of detection. Addressing the emergence of antibiotic-resistant bacteria, M13 phages are potent for packaging the programmed gene editing tools, such as CRISPR/Cas, to target multiple antimicrobial genes. Moreover, their display potential in combination with nanoparticles inspires new approaches for engineering targeted theragnostic platforms targeting multiple cellular biomarkers in vivo. In this review, we present the available data on optimizing the use of bacteriophages with a focus on the to date experiences with M13 phages, either as monoagent or as part of combination regimens in the practices of biosensors, vaccines, bactericidal, modeling of specific antigen epitopes, and phage-guided nanoparticles for drug delivery systems. Despite increasing research interest, a deep understanding of the underlying biological and genetic behaviors of M13 phages is needed to enable the full potential of these bioagents in biomedicine, as discussed here. We also discuss some of the challenges that have thus far limited the development and practical marketing of M13 phages.
Collapse
Affiliation(s)
- Mahmood Fadaie
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Pediatric Inherited Diseases Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Elham Ghafouri
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shamsi Naderi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mohammad Hossein Darvishali
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mahsa Ghovvati
- Department of Radiological Sciences, David Geffen School of Medicine, University of California - Los Angeles, Los Angeles, CA 90095, USA
| | - Hossein Khanahmad
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China.
| |
Collapse
|
17
|
Lv S, Wang Y, Jiang K, Guo X, Zhang J, Zhou F, Li Q, Jiang Y, Yang C, Teng T. Genetic Engineering and Biosynthesis Technology: Keys to Unlocking the Chains of Phage Therapy. Viruses 2023; 15:1736. [PMID: 37632078 PMCID: PMC10457950 DOI: 10.3390/v15081736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 07/21/2023] [Accepted: 08/12/2023] [Indexed: 08/27/2023] Open
Abstract
Phages possess the ability to selectively eliminate pathogenic bacteria by recognizing bacterial surface receptors. Since their discovery, phages have been recognized for their potent bactericidal properties, making them a promising alternative to antibiotics in the context of rising antibiotic resistance. However, the rapid emergence of phage-resistant strains (generally involving temperature phage) and the limited host range of most phage strains have hindered their antibacterial efficacy, impeding their full potential. In recent years, advancements in genetic engineering and biosynthesis technology have facilitated the precise engineering of phages, thereby unleashing their potential as a novel source of antibacterial agents. In this review, we present a comprehensive overview of the diverse strategies employed for phage genetic engineering, as well as discuss their benefits and drawbacks in terms of bactericidal effect.
Collapse
Affiliation(s)
- Sixuan Lv
- School of Nursing and Health, Henan University, Kaifeng 475004, China
- Institute of Biomedical Informatics, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yuhan Wang
- Institute of Biomedical Informatics, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Kaixin Jiang
- School of Nursing and Health, Henan University, Kaifeng 475004, China
- Institute of Biomedical Informatics, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Xinge Guo
- School of Nursing and Health, Henan University, Kaifeng 475004, China
- Institute of Biomedical Informatics, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Jing Zhang
- School of Nursing and Health, Henan University, Kaifeng 475004, China
- Institute of Biomedical Informatics, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Fang Zhou
- School of Nursing and Health, Henan University, Kaifeng 475004, China
- Institute of Biomedical Informatics, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Qiming Li
- School of Nursing and Health, Henan University, Kaifeng 475004, China
- Institute of Biomedical Informatics, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yuan Jiang
- Institute of Biomedical Informatics, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Changyong Yang
- School of Nursing and Health, Henan University, Kaifeng 475004, China
| | - Tieshan Teng
- School of Nursing and Health, Henan University, Kaifeng 475004, China
- Institute of Biomedical Informatics, School of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| |
Collapse
|
18
|
Borg RE, Ozbakir HF, Xu B, Li E, Fang X, Peng H, Chen IA, Mukherjee A. Genetically engineered filamentous phage for bacterial detection using magnetic resonance imaging. SENSORS & DIAGNOSTICS 2023; 2:948-955. [PMID: 38405385 PMCID: PMC10888512 DOI: 10.1039/d3sd00026e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/27/2024]
Abstract
Detecting bacterial cells with high specificity in deep tissues is challenging. Optical probes provide specificity, but are limited by the scattering and absorption of light in biological tissues. Conversely, magnetic resonance imaging (MRI) allows unfettered access to deep tissues, but lacks contrast agents for detecting specific bacterial strains. Here, we introduce a biomolecular platform that combines both capabilities by exploiting the modularity of M13 phage to target bacteria with tunable specificity and allow deep-tissue imaging using T1-weighted MRI. We engineered two types of phage probes: one for detecting the phage's natural host, viz., F-pilus expressing E. coli; and the other for detecting a different (F-negative) bacterial target, V. cholerae. We show that these phage sensors generate 3-9-fold stronger T1 relaxation upon recognizing target cells relative to non-target bacteria. We further establish a preliminary proof-of-concept for in vivo applications, by demonstrating that phage-labeled bacteria can be detected in mice using MRI. The framework developed in this study may have potential utility in a broad range of applications, from basic biomedical research to in situ diagnostics, which require methods to detect and track specific bacteria in the context of intact living systems.
Collapse
Affiliation(s)
- Raymond E Borg
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
| | - Harun F Ozbakir
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Binzhi Xu
- Biomolecular Science and Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Eugene Li
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Xiwen Fang
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
| | - Huan Peng
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA
| | - Irene A Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, CA 90095, USA
| | - Arnab Mukherjee
- Department of Chemistry, University of California, Santa Barbara, CA 93106, USA
- Department of Chemical Engineering, University of California, Santa Barbara, CA 93106, USA
- Biological Engineering, University of California, Santa Barbara, CA 93106, USA
- Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA
| |
Collapse
|
19
|
Zhu X, Li Z, Tong Y, Chen L, Sun T, Zhang W. From natural to artificial cyanophages: Current progress and application prospects. ENVIRONMENTAL RESEARCH 2023; 223:115428. [PMID: 36746205 DOI: 10.1016/j.envres.2023.115428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/01/2023] [Accepted: 02/03/2023] [Indexed: 06/18/2023]
Abstract
The over proliferation of harmful cyanobacteria and their cyanotoxins resulted in damaged aquatic ecosystem, polluted drinking water and threatened human health. Cyanophages are a kind of viruses that exclusively infect cyanobacteria, which is considered as a potential strategy to deal with cyanobacterial blooms. Nevertheless, the infecting host range and/or lysis efficiency of natural cyanophages is limited, rising the necessity of constructing non-natural cyanophages via artificial modification, design and synthesis to expand their host range and/or efficiency. The paper firstly reviewed representative cyanophages such as P60 with a short latent period of 1.5 h and S-CBS1 having a burst size up to 200 PFU/cell. To explore the in-silico design principles, we critically summarized the interactions between cyanophages and the hosts, indicating modifying the recognized receptors, enhancing the adsorption ability, changing the lysogeny and excluding the defense of hosts are important for artificial cyanophages. The research progress of synthesizing artificial cyanophages were summarized subsequently, raising the importance of developing genetic manipulation technologies and their rescue strategies in the future. Meanwhile, Large-scale preparation of cyanophages for bloom control is a big challenge. The application prospects of artificial cyanophages besides cyanobacteria bloom control like adaptive evolution and phage therapy were discussed at last. The review will promote the design, synthesis and application of cyanophages for cyanobacteria blooms, which may provide new insights for the related water pollution control and ensuring hydrosphere security.
Collapse
Affiliation(s)
- Xiaofei Zhu
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin, 300072, PR China; Frontier Science Center for Synthetic Biology & Key Laboratory of Systems Bioengineering, Ministry of Education of China, Tianjin, 300072, PR China
| | - Zipeng Li
- School of Environmental Science and Engineering, Tianjin University, Tianjin, 300072, China
| | - Yindong Tong
- School of Environmental Science and Engineering, Tianjin University, Tianjin, 300072, China
| | - Lei Chen
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin, 300072, PR China; Frontier Science Center for Synthetic Biology & Key Laboratory of Systems Bioengineering, Ministry of Education of China, Tianjin, 300072, PR China.
| | - Tao Sun
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin, 300072, PR China; Frontier Science Center for Synthetic Biology & Key Laboratory of Systems Bioengineering, Ministry of Education of China, Tianjin, 300072, PR China; Center for Biosafety Research and Strategy, Tianjin University, Tianjin, 300072, PR China.
| | - Weiwen Zhang
- Laboratory of Synthetic Microbiology, School of Chemical Engineering & Technology, Tianjin University, Tianjin, 300072, PR China; Frontier Science Center for Synthetic Biology & Key Laboratory of Systems Bioengineering, Ministry of Education of China, Tianjin, 300072, PR China; Center for Biosafety Research and Strategy, Tianjin University, Tianjin, 300072, PR China
| |
Collapse
|
20
|
Yin L, Li H, Shi L, Chen K, Pan H, Han W. Research advances in nanomedicine applied to the systemic treatment of colorectal cancer. Int J Cancer 2023; 152:807-821. [PMID: 35984398 DOI: 10.1002/ijc.34256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 01/06/2023]
Abstract
The systematic treatment of colorectal cancer (CRC) still has room for improvement. The efficacy of chemotherapy, that of anti-vascular therapy, and that of immunotherapy have been unsatisfactory. In recent years, nanomaterials have been used as carriers to improve the bioavailability of anticancer drugs. For the treatment of colorectal cancer, nanodrugs increase the possibility of more precise targeted delivery. However, the actual benefits may cover more aspects. Nanocarriers can produce synergistic effects with anticancer drugs, including the scavenging of reactive oxygen species and co-delivery of a variety of drugs. Currently, immunotherapy has very limited clinical applications in CRC. Modified nanocarriers can activate the immune microenvironment, which can be used for staging antigen recognition or the immune response. Cancer vaccines based on nanomaterials and modified immune checkpoint inhibitors have shown therapeutic potential in animal models. Considering the direct or indirect relationship between the intestinal microflora and CRC, a variety of nanodrugs that regulate microbial function have been explored as an anticancer strategy, and the special structure of microorganisms can also be used as a basis for improving the delivery of traditional nanoparticles (NPs). This review summarizes recent research performed on nanocarriers in in vivo and in vitro models and the synergistic anticancer effects of nanocarriers, focusing on the interaction between NPs and the body, resulting in enhanced efficacy and immune activation. Furthermore, this review describes the current trend of NPs used in the treatment of CRC.
Collapse
Affiliation(s)
- Luxi Yin
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Haozhe Li
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Linlin Shi
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Keda Chen
- Shulan International Medical College, Zhejiang Shuren University, Hangzhou, Zhejiang, China
| | - Hongming Pan
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Weidong Han
- Department of Medical Oncology, Sir Run Run Shaw Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
21
|
Sun Q, Shen L, Zhang BL, Yu J, Wei F, Sun Y, Chen W, Wang S. Advance on Engineering of Bacteriophages by Synthetic Biology. Infect Drug Resist 2023; 16:1941-1953. [PMID: 37025193 PMCID: PMC10072152 DOI: 10.2147/idr.s402962] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Accepted: 03/16/2023] [Indexed: 04/03/2023] Open
Abstract
Since bacteriophages (phages) were firstly reported at the beginning of the 20th century, the study on them experiences booming-fading-emerging with discovery and overuse of antibiotics. Although they are the hotspots for therapy of antibiotic-resistant strains nowadays, natural phage applications encounter some challenges such as limited host range and bacterial resistance to phages. Synthetic biology, one of the most dramatic directions in the recent 20-years study of microbiology, has generated numerous methods and tools and has contributed a lot to understanding phage evolution, engineering modification, and controlling phage-bacteria interactions. In order to better modify and apply phages by using synthetic biology techniques in the future, in this review, we comprehensively introduce various strategies on engineering or modification of phage genome and rebooting of recombinant phages, summarize the recent researches and potential directions of phage synthetic biology, and outline the current application of engineered phages in practice.
Collapse
Affiliation(s)
- Qingqing Sun
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, the College of Life Sciences, Northwest University, Xi’an, 710069, People’s Republic of China
| | - Lixin Shen
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, the College of Life Sciences, Northwest University, Xi’an, 710069, People’s Republic of China
| | - Bai-Ling Zhang
- Department of Laboratory Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, 330006, People’s Republic of China
| | - Jiaoyang Yu
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, the College of Life Sciences, Northwest University, Xi’an, 710069, People’s Republic of China
- Clinical Research Center, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, People’s Republic of China
| | - Fu Wei
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, the College of Life Sciences, Northwest University, Xi’an, 710069, People’s Republic of China
| | - Yanmei Sun
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, the College of Life Sciences, Northwest University, Xi’an, 710069, People’s Republic of China
| | - Wei Chen
- Clinical Research Center, the Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, People’s Republic of China
- The Clinical Infectious Disease Center of Nanjing, Nanjing, 210003, People’s Republic of China
- Correspondence: Wei Chen; Shiwei Wang, Email ;
| | - Shiwei Wang
- Key Laboratory of Resources Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology of Shaanxi Province, the College of Life Sciences, Northwest University, Xi’an, 710069, People’s Republic of China
| |
Collapse
|
22
|
Makvandi P, Song H, Yiu CKY, Sartorius R, Zare EN, Rabiee N, Wu WX, Paiva-Santos AC, Wang XD, Yu CZ, Tay FR. Bioengineered materials with selective antimicrobial toxicity in biomedicine. Mil Med Res 2023; 10:8. [PMID: 36829246 PMCID: PMC9951506 DOI: 10.1186/s40779-023-00443-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Accepted: 01/31/2023] [Indexed: 02/26/2023] Open
Abstract
Fungi and bacteria afflict humans with innumerous pathogen-related infections and ailments. Most of the commonly employed microbicidal agents target commensal and pathogenic microorganisms without discrimination. To distinguish and fight the pathogenic species out of the microflora, novel antimicrobials have been developed that selectively target specific bacteria and fungi. The cell wall features and antimicrobial mechanisms that these microorganisms involved in are highlighted in the present review. This is followed by reviewing the design of antimicrobials that selectively combat a specific community of microbes including Gram-positive and Gram-negative bacterial strains as well as fungi. Finally, recent advances in the antimicrobial immunomodulation strategy that enables treating microorganism infections with high specificity are reviewed. These basic tenets will enable the avid reader to design novel approaches and compounds for antibacterial and antifungal applications.
Collapse
Affiliation(s)
- Pooyan Makvandi
- Istituto Italiano di Tecnologia, Centre for Materials Interfaces, Pontedera, 56025, Italy. .,The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China.
| | - Hao Song
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Cynthia K Y Yiu
- Paediatric Dentistry and Orthodontics, Faculty of Dentistry, The University of Hong Kong, Prince Philip Dental Hospital, Hong Kong SAR, China
| | - Rossella Sartorius
- Institute of Biochemistry and Cell Biology (IBBC), National Research Council (CNR), 80131, Naples, Italy
| | | | - Navid Rabiee
- School of Engineering, Macquarie University, Sydney, NSW, 2109, Australia.,Centre for Molecular Medicine and Innovative Therapeutics, Murdoch University, Perth, WA, 6150, Australia
| | - Wei-Xi Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal.,REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy of the University of Coimbra, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Xiang-Dong Wang
- Department of Pulmonary and Critical Care Medicine, Zhongshan Hospital, Fudan University Shanghai Medical College, Shanghai, 200032, China
| | - Cheng-Zhong Yu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD, 4072, Australia.,School of Chemistry and Molecular Engineering, East China Normal University, Shanghai, 200241, China
| | - Franklin R Tay
- The Graduate School, Augusta University, Augusta, GA, 30912, USA.
| |
Collapse
|
23
|
Behler KL, Honemann MN, Rita Silva‐Santos A, Dietz H, Weuster‐Botz D. Phage‐free production of artificial ssDNA with
Escherichia coli. Biotechnol Bioeng 2022; 119:2878-2889. [DOI: 10.1002/bit.28171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 05/09/2022] [Accepted: 06/19/2022] [Indexed: 11/07/2022]
Affiliation(s)
- Karl L. Behler
- Technical University of Munich, School of Engineering and Design, Chair of Biochemical EngineeringGarching nearMunichGermany
| | - Maximilian N. Honemann
- Technical University of MunichDepartment of PhysicsGarching nearMunichGermany
- Technical University of MunichMunich Institute of Biomedical EngineeringGarching nearMunichGermany
| | | | - Hendrik Dietz
- Technical University of MunichDepartment of PhysicsGarching nearMunichGermany
- Technical University of MunichMunich Institute of Biomedical EngineeringGarching nearMunichGermany
| | - Dirk Weuster‐Botz
- Technical University of Munich, School of Engineering and Design, Chair of Biochemical EngineeringGarching nearMunichGermany
| |
Collapse
|
24
|
Nath A, Bhattacharjee R, Nandi A, Sinha A, Kar S, Manoharan N, Mitra S, Mojumdar A, Panda PK, Patro S, Dutt A, Ahuja R, Verma SK, Suar M. Phage delivered CRISPR-Cas system to combat multidrug-resistant pathogens in gut microbiome. Biomed Pharmacother 2022; 151:113122. [PMID: 35594718 DOI: 10.1016/j.biopha.2022.113122] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 11/02/2022] Open
Abstract
The Host-microbiome interactions that exist inside the gut microbiota operate in a synergistic and abnormal manner. Additionally, the normal homeostasis and functioning of gut microbiota are frequently disrupted by the intervention of Multi-Drug Resistant (MDR) pathogens. CRISPR-Cas (CRISPR-associated protein with clustered regularly interspersed short palindromic repeats) recognized as a prokaryotic immune system has emerged as an effective genome-editing tool to edit and delete specific microbial genes for the expulsion of bacteria through bactericidal action. In this review, we demonstrate many functioning CRISPR-Cas systems against the anti-microbial resistance of multiple pathogens, which infiltrate the gastrointestinal tract. Moreover, we discuss the advancement in the development of a phage-delivered CRISPR-Cas system for killing a gut MDR pathogen. We also discuss a combinatorial approach to use bacteriophage as a delivery system for the CRISPR-Cas gene for targeting a pathogenic community in the gut microbiome to resensitize the drug sensitivity. Finally, we discuss engineered phage as a plausible potential option for the CRISPR-Cas system for pathogenic killing and improvement of the efficacy of the system.
Collapse
Affiliation(s)
- Arijit Nath
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Rahul Bhattacharjee
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Aditya Nandi
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Adrija Sinha
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Sulagna Kar
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | | | - Shirsajit Mitra
- KaviKrishna Laboratory, Indian Institute of Technology, Guwahati, Assam, India
| | - Abhik Mojumdar
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Pritam Kumar Panda
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Swadheena Patro
- KIIT School of Dental Sciences, KIIT University. Bhubaneswar 751024, Odisha
| | - Ateet Dutt
- Instituto de Investigaciones en Materiales, UNAM, CDMX, Mexico
| | - Rajeev Ahuja
- Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden.
| | - Suresh K Verma
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden.
| | - Mrutyunjay Suar
- KIIT School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India.
| |
Collapse
|
25
|
Greve JM, Cowan JA. Tackling antimicrobial stewardship through synergy and antimicrobial peptides. RSC Med Chem 2022; 13:511-521. [PMID: 35694695 PMCID: PMC9132191 DOI: 10.1039/d2md00048b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 04/04/2022] [Indexed: 11/21/2022] Open
Abstract
The unrestricted use of antibiotics has led to rapid development of antibiotic resistance (AR) and renewed calls to address this serious problem. This review summarizes the most common mechanisms of antibiotic action, and in turn antibiotic resistance, as well as pathways to mitigate the harm. Focus is then turned to emerging antibiotic strategies, including antimicrobial peptides (AMPs), with a discussion of their modes of action, biochemical features, and potential challenges for their use as antibiotics. The role of synergy in antimicrobials is also examined, with a focus on the synergy of AMPs and other emerging interactions with synergistic potential.
Collapse
Affiliation(s)
- Jenna M Greve
- Department of Chemistry and Biochemistry, The Ohio State University 100 West 18th Avenue Columbus Ohio 43210 USA +1 614 292 2703
| | - James A Cowan
- Department of Chemistry and Biochemistry, The Ohio State University 100 West 18th Avenue Columbus Ohio 43210 USA +1 614 292 2703
| |
Collapse
|
26
|
Abstract
Antimicrobial peptides (AMPs) have recently become widely publicized because they have the potential to function in alternative therapies as “natural” antibiotics, with their main advantage being a broad spectrum of activity. The potential for antimicrobial peptides to treat diabetes mellitus (DM) has been reported. In diabetes mellitus type I (T1D), cathelicidin-related antimicrobial peptide (CRAMP), cathelicidin antimicrobial peptide (CAMP) and mouse-β- defensin 14 (mBD14) are positively affected. Decreased levels of LL-37 and human neutrophil peptide 1-3 (HNP1-3) have been reported in diabetes mellitus type II (T2D) relative to healthy patients. Moreover, AMPs from amphibians and social wasps have antidiabetic effects. In infections occurring in patients with tuberculosis-diabetes or diabetic foot, granulysin, HNP1, HNP2, HNP3, human beta-defensin 2 (HBD2), and cathelicidins are responsible for pathogen clearance. An interesting alternative is also the use of modified M13 bacteriophages containing encapsulated AMPs genes or phagemids.
Collapse
|
27
|
Ma J, Li K, Gu S. Selective strategies for antibacterial regulation of nanomaterials. RSC Adv 2022; 12:4852-4864. [PMID: 35425473 PMCID: PMC8981418 DOI: 10.1039/d1ra08996j] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/25/2022] [Indexed: 12/14/2022] Open
Abstract
Recalcitrant bacterial infection, as a worldwide challenge, causes large problems for human health and is attracting great attention. The excessive antibiotic-dependent treatment of infections is prone to induce antibiotic resistance. A variety of unique nanomaterials provide an excellent toolkit for killing bacteria and preventing drug resistance. It is of great importance to summarize the design rules of nanomaterials for inhibiting the growth of pathogenic bacteria. We completed a review involving the strategies for regulating antibacterial nanomaterials. First, we discuss the antibacterial manipulation of nanomaterials, including the interaction between the nanomaterial and the bacteria, the damage of the bacterial structure, and the inactivation of biomolecules. Next, we identify six main factors for controlling the antibacterial activity of nanomaterials, including their element composition, size dimensions, surface charge, surface topography, shape selection and modification density. Every factor possesses a preferable standard for maximizing antibacterial activity, providing universal rules for antibacterial regulation of nanomaterials. We hope this comprehensive review will help researchers to precisely design and synthesize nanomaterials, developing intelligent antibacterial agents to address bacterial infections.
Collapse
Affiliation(s)
- Jinliang Ma
- College of Food and Bioengineering, Henan University of Science and Technology Luoyang Henan 471023 China
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University Shanghai 200127 China
| | - Kexin Li
- College of Food and Bioengineering, Henan University of Science and Technology Luoyang Henan 471023 China
| | - Shaobin Gu
- College of Food and Bioengineering, Henan University of Science and Technology Luoyang Henan 471023 China
| |
Collapse
|
28
|
Yu H, Feng C, Raza SHA, Zhang L, Chi T, Qi Y, Jia K, Zhang Y, Wei J, Qian A, Sun W, Shan X, Zhang L. Characterization and genome analysis of two new Aeromonas hydrophila phages, PZL-Ah1and PZL-Ah8. Arch Virol 2022; 167:669-673. [DOI: 10.1007/s00705-021-05345-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 11/11/2021] [Indexed: 11/02/2022]
|
29
|
Ma J, Li K, Gu S, Wu Y, Zhang J, Wu J, Zhao L, Li X. Antimicrobial carbon-dot–stabilized silver nanoparticles. NEW J CHEM 2022. [DOI: 10.1039/d1nj05798g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbon-dot–stabilized silver nanoparticles (CD–AgNPs) with high stability and low toxicity exhibit good antibacterial activity and broad-spectrum performance.
Collapse
Affiliation(s)
- Jinliang Ma
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan 471023, China
- Institute of Molecular Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Kexin Li
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Shaobin Gu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Ying Wu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Jing Zhang
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Jiafa Wu
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Lina Zhao
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan 471023, China
| | - Xuan Li
- College of Food and Bioengineering, Henan University of Science and Technology, Luoyang, Henan 471023, China
| |
Collapse
|
30
|
Meile S, Du J, Dunne M, Kilcher S, Loessner MJ. Engineering therapeutic phages for enhanced antibacterial efficacy. Curr Opin Virol 2021; 52:182-191. [PMID: 34952266 DOI: 10.1016/j.coviro.2021.12.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/22/2021] [Accepted: 12/04/2021] [Indexed: 12/18/2022]
Abstract
The alarming rise in antimicrobial resistance coupled with a lack of innovation in antibiotics has renewed interest in the development of alternative therapies to combat bacterial infections. Despite phage therapy demonstrating success in various individual cases, a comprehensive and unequivocal demonstration of the therapeutic potential of phages remains to be shown. The co-evolution of phages and their bacterial hosts resulted in several inherent limitations for the use of natural phages as therapeutics such as restricted host range, moderate antibacterial efficacy, and frequent emergence of phage-resistance. However, these constraints can be overcome by leveraging recent advances in synthetic biology and genetic engineering to provide phages with additional therapeutic capabilities, improved safety profiles, and adaptable host ranges. Here, we examine different ways phages can be engineered to deliver heterologous therapeutic payloads to enhance their antibacterial efficacy and discuss their versatile applicability to combat bacterial pathogens.
Collapse
Affiliation(s)
- Susanne Meile
- Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 7, 8092 Zurich, Switzerland
| | - Jiemin Du
- Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 7, 8092 Zurich, Switzerland
| | - Matthew Dunne
- Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 7, 8092 Zurich, Switzerland
| | - Samuel Kilcher
- Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 7, 8092 Zurich, Switzerland
| | - Martin J Loessner
- Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 7, 8092 Zurich, Switzerland.
| |
Collapse
|
31
|
Ibarra-Chávez R, Hansen MF, Pinilla-Redondo R, Seed KD, Trivedi U. Phage satellites and their emerging applications in biotechnology. FEMS Microbiol Rev 2021; 45:fuab031. [PMID: 34104956 PMCID: PMC8632786 DOI: 10.1093/femsre/fuab031] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The arms race between (bacterio)phages and their hosts is a recognised hot spot for genome evolution. Indeed, phages and their components have historically paved the way for many molecular biology techniques and biotech applications. Further exploration into their complex lifestyles has revealed that phages are often parasitised by distinct types of hyperparasitic mobile genetic elements. These so-called phage satellites exploit phages to ensure their own propagation and horizontal transfer into new bacterial hosts, and their prevalence and peculiar lifestyle has caught the attention of many researchers. Here, we review the parasite-host dynamics of the known phage satellites, their genomic organisation and their hijacking mechanisms. Finally, we discuss how these elements can be repurposed for diverse biotech applications, kindling a new catalogue of exciting tools for microbiology and synthetic biology.
Collapse
Affiliation(s)
- Rodrigo Ibarra-Chávez
- Section of Microbiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Mads Frederik Hansen
- Section of Microbiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
- Max Planck Institute for Terrestrial Microbiology, 35043 Marburg, Germany
| | - Rafael Pinilla-Redondo
- Section of Microbiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Kimberley D Seed
- Department of Plant and Microbial Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Chan Zuckerberg Biohub, San Francisco, CA 94158, USA
| | - Urvish Trivedi
- Section of Microbiology, Department of Biology, University of Copenhagen, 2100 Copenhagen, Denmark
| |
Collapse
|
32
|
CRISPR-Cas Technology: Emerging Applications in Clinical Microbiology and Infectious Diseases. Pharmaceuticals (Basel) 2021; 14:ph14111171. [PMID: 34832953 PMCID: PMC8625472 DOI: 10.3390/ph14111171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 11/06/2021] [Accepted: 11/09/2021] [Indexed: 12/26/2022] Open
Abstract
Through the years, many promising tools for gene editing have been developed including zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), CRISPR-associated protein 9 (Cas9), and homing endonucleases (HEs). These novel technologies are now leading new scientific advancements and practical applications at an inimitable speed. While most work has been performed in eukaryotes, CRISPR systems also enable tools to understand and engineer bacteria. The increase in the number of multi-drug resistant strains highlights a necessity for more innovative approaches to the diagnosis and treatment of infections. CRISPR has given scientists a glimmer of hope in this area that can provide a novel tool to fight against antimicrobial resistance. This system can provide useful information about the functions of genes and aid us to find potential targets for antimicrobials. This paper discusses the emerging use of CRISPR-Cas systems in the fields of clinical microbiology and infectious diseases with a particular emphasis on future prospects.
Collapse
|
33
|
Novick RP. Antibacterial particles and predatory bacteria as alternatives to antibacterial chemicals in the era of antibiotic resistance. Curr Opin Microbiol 2021; 64:109-116. [PMID: 34688038 DOI: 10.1016/j.mib.2021.09.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/26/2022]
Abstract
This review is focused on the subset of antibacterial agents whose action involves one-on-one targeting of infecting bacteria. These agents target individual bacteria and their efficacy is based on particle numbers in contrast to chemical agents such as antibiotics, whose efficacy is based on minimal inhibitory concentrations. Four extant members of this class are predatory bacteria, functional (plaque-forming) phages, and engineered particulate systems, phagemids (plasmids that contain a phage packaging signal) and antibacterial drones (ABDs) that package chromosomal island DNA carrying antibacterial genes. We differentiate the natural predators, phages and predatory bacteria, from the engineered delivery vehicles, phagemids and ABDs, because the latter are much more versatile and can largely bypass the historical warfare that informs the predator-prey interactions.
Collapse
|
34
|
Manufacturing Bacteriophages (Part 1 of 2): Cell Line Development, Upstream, and Downstream Considerations. Pharmaceuticals (Basel) 2021; 14:ph14090934. [PMID: 34577634 PMCID: PMC8471501 DOI: 10.3390/ph14090934] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/06/2021] [Accepted: 09/15/2021] [Indexed: 01/21/2023] Open
Abstract
Within this first part of the two-part series on phage manufacturing, we will give an overview of the process leading to bacteriophages as a drug substance, before covering the formulation into a drug product in the second part. The principal goal is to provide the reader with a comprehensive framework of the challenges and opportunities that present themselves when developing manufacturing processes for bacteriophage-based products. We will examine cell line development for manufacture, upstream and downstream processes, while also covering the additional opportunities that engineered bacteriophages present.
Collapse
|
35
|
Li X, He Y, Wang Z, Wei J, Hu T, Si J, Tao G, Zhang L, Xie L, Abdalla AE, Wang G, Li Y, Teng T. A combination therapy of Phages and Antibiotics: Two is better than one. Int J Biol Sci 2021; 17:3573-3582. [PMID: 34512166 PMCID: PMC8416725 DOI: 10.7150/ijbs.60551] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Accepted: 08/01/2021] [Indexed: 12/15/2022] Open
Abstract
Emergence of antibiotic resistance presents a major setback to global health, and shortage of antibiotic pipelines has created an urgent need for development of alternative therapeutic strategies. Bacteriophage (phage) therapy is considered as a potential approach for treatment of the increasing number of antibiotic-resistant pathogens. Phage-antibiotic synergy (PAS) refers to sublethal concentrations of certain antibiotics that enhance release of progeny phages from bacterial cells. A combination of phages and antibiotics is a promising strategy to reduce the dose of antibiotics and the development of antibiotic resistance during treatment. In this review, we highlight the state-of-the-art advancements of PAS studies, including the analysis of bacterial-killing enhancement, bacterial resistance reduction, and anti-biofilm effect, at both in vitro and in vivo levels. A comprehensive review of the genetic and molecular mechanisms of phage antibiotic synergy is provided, and synthetic biology approaches used to engineer phages, and design novel therapies and diagnostic tools are discussed. In addition, the role of engineered phages in reducing pathogenicity of bacteria is explored.
Collapse
Affiliation(s)
- Xianghui Li
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yuhua He
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Zhili Wang
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Jiacun Wei
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Tongxin Hu
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Jiangzhe Si
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Guangzhao Tao
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Lei Zhang
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Longxiang Xie
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Abualgasim Elgaili Abdalla
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 2014, Saudi Arabia
| | - Guoying Wang
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Yanzhang Li
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| | - Tieshan Teng
- Institute of Biomedical Informatics, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China.,Henan International Joint Laboratory of Nuclear Protein Regulation, school of Basic Medical Sciences, Henan University, Kaifeng 475004, China
| |
Collapse
|
36
|
Lam CN, Mehta-Kolte MG, Martins-Sorenson N, Eckert B, Lin PH, Chu K, Moghaddasi A, Goldman D, Nguyen H, Chan R, Nukala L, Suko S, Hanson B, Yuan R, Cady KC. A Tail Fiber Engineering Platform for Improved Bacterial Transduction-Based Diagnostic Reagents. ACS Synth Biol 2021; 10:1292-1299. [PMID: 33983709 DOI: 10.1021/acssynbio.1c00036] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Bacterial transduction particles were critical to early advances in molecular biology and are currently experiencing a resurgence in interest within the diagnostic and therapeutic fields. The difficulty of developing a robust and specific transduction reagent capable of delivering a genetic payload to the diversity of strains constituting a given bacterial species or genus is a major impediment to their expanded utility as commercial products. While recent advances in engineering the reactivity of these reagents have made them more attractive for product development, considerable improvements are still needed. Here, we demonstrate a synthetic biology platform derived from bacteriophage P1 as a chassis to target transduction reagents against four clinically prevalent species within the Enterobacterales order. Bacteriophage P1 requires only a single receptor binding protein to enable attachment and injection into a target bacterium. By engineering and screening particles displaying a diverse array of chimeric receptor binding proteins, we generated a potential transduction reagent for a future rapid phenotypic carbapenem-resistant Enterobacterales diagnostic assay.
Collapse
Affiliation(s)
- Colin N. Lam
- Roche Molecular Systems, Santa Clara, California 95050, United States
| | | | | | - Barbara Eckert
- Roche Molecular Systems, Pleasanton, California 94588, United States
| | - Patrick H. Lin
- Roche Molecular Systems, Santa Clara, California 95050, United States
| | - Kristina Chu
- Roche Molecular Systems, Pleasanton, California 94588, United States
| | - Arrash Moghaddasi
- Roche Molecular Systems, Santa Clara, California 95050, United States
| | - Dylan Goldman
- Roche Molecular Systems, Santa Clara, California 95050, United States
| | - Hai Nguyen
- Roche Molecular Systems, Santa Clara, California 95050, United States
| | - Ryan Chan
- Roche Molecular Systems, Santa Clara, California 95050, United States
| | - Laxmi Nukala
- Roche Molecular Systems, Santa Clara, California 95050, United States
| | - Shawn Suko
- Roche Molecular Systems, Pleasanton, California 94588, United States
| | - Brett Hanson
- Roche Molecular Systems, Santa Clara, California 95050, United States
| | - Richard Yuan
- Roche Molecular Systems, Santa Clara, California 95050, United States
| | - Kyle C. Cady
- Roche Molecular Systems, Santa Clara, California 95050, United States
| |
Collapse
|
37
|
Chen L, Hong W, Ren W, Xu T, Qian Z, He Z. Recent progress in targeted delivery vectors based on biomimetic nanoparticles. Signal Transduct Target Ther 2021; 6:225. [PMID: 34099630 PMCID: PMC8182741 DOI: 10.1038/s41392-021-00631-2] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 04/28/2021] [Accepted: 04/29/2021] [Indexed: 02/05/2023] Open
Abstract
Over the past decades, great interest has been given to biomimetic nanoparticles (BNPs) since the rise of targeted drug delivery systems and biomimetic nanotechnology. Biological vectors including cell membranes, extracellular vesicles (EVs), and viruses are considered promising candidates for targeted delivery owing to their biocompatibility and biodegradability. BNPs, the integration of biological vectors and functional agents, are anticipated to load cargos or camouflage synthetic nanoparticles to achieve targeted delivery. Despite their excellent intrinsic properties, natural vectors are deliberately modified to endow multiple functions such as good permeability, improved loading capability, and high specificity. Through structural modification and transformation of the vectors, they are pervasively utilized as more effective vehicles that can deliver contrast agents, chemotherapy drugs, nucleic acids, and genes to target sites for refractory disease therapy. This review summarizes recent advances in targeted delivery vectors based on cell membranes, EVs, and viruses, highlighting the potential applications of BNPs in the fields of biomedical imaging and therapy industry, as well as discussing the possibility of clinical translation and exploitation trend of these BNPs.
Collapse
Affiliation(s)
- Li Chen
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weiqi Hong
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wenyan Ren
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Xu
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyong Qian
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyao He
- Department of Pharmacy, State Key Laboratory of Biotherapy and Cancer Center, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
38
|
Abd-Allah IM, El-Housseiny GS, Yahia IS, Aboshanab KM, Hassouna NA. Rekindling of a Masterful Precedent; Bacteriophage: Reappraisal and Future Pursuits. Front Cell Infect Microbiol 2021; 11:635597. [PMID: 34136415 PMCID: PMC8201069 DOI: 10.3389/fcimb.2021.635597] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/13/2021] [Indexed: 12/30/2022] Open
Abstract
Antibiotic resistance is exuberantly becoming a deleterious health problem world-wide. Seeking innovative approaches is necessary in order to circumvent such a hazard. An unconventional fill-in to antibiotics is bacteriophage. Bacteriophages are viruses capable of pervading bacterial cells and disrupting their natural activity, ultimately resulting in their defeat. In this article, we will run-through the historical record of bacteriophage and its correlation with bacteria. We will also delineate the potential of bacteriophage as a therapeutic antibacterial agent, its supremacy over antibiotics in multiple aspects and the challenges that could arise on the way to its utilization in reality. Pharmacodynamics, pharmacokinetics and genetic engineering of bacteriophages and its proteins will be briefly discussed as well. In addition, we will highlight some of the in-use applications of bacteriophages, and set an outlook for their future ones. We will also overview some of the miscellaneous abilities of these tiny viruses in several fields other than the clinical one. This is an attempt to encourage tackling a long-forgotten hive. Perhaps, one day, the smallest of the creatures would be of the greatest help.
Collapse
Affiliation(s)
- Israa M. Abd-Allah
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ghadir S. El-Housseiny
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Ibrahim S. Yahia
- Research Center for Advanced Materials Science (RCAMS), Advanced Functional Materials & Optoelectronic Laboratory (AFMOL), Department of Physics, Faculty of Science, King Khalid University, Abha, Saudi Arabia
- Nanoscience Laboratory for Environmental and Bio-Medical Applications (NLEBA), Semiconductor Lab., Metallurgical Lab, Physics Department, Faculty of Education, Ain Shams University, Cairo, Egypt
| | - Khaled M. Aboshanab
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| | - Nadia A. Hassouna
- Department of Microbiology and Immunology, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
39
|
Łobocka M, Dąbrowska K, Górski A. Engineered Bacteriophage Therapeutics: Rationale, Challenges and Future. BioDrugs 2021; 35:255-280. [PMID: 33881767 PMCID: PMC8084836 DOI: 10.1007/s40259-021-00480-z] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/29/2021] [Indexed: 12/20/2022]
Abstract
The current problems with increasing bacterial resistance to antibacterial therapies, resulting in a growing frequency of incurable bacterial infections, necessitates the acceleration of studies on antibacterials of a new generation that could offer an alternative to antibiotics or support their action. Bacteriophages (phages) can kill antibiotic-sensitive as well as antibiotic-resistant bacteria, and thus are a major subject of such studies. Their efficacy in curing bacterial infections has been demonstrated in in vivo experiments and in the clinic. Unlike antibiotics, phages have a narrow range of specificity, which makes them safe for commensal microbiota. However, targeting even only the most clinically relevant strains of pathogenic bacteria requires large collections of well characterized phages, whose specificity would cover all such strains. The environment is a rich source of diverse phages, but due to their complex relationships with bacteria and safety concerns, only some naturally occurring phages can be considered for therapeutic applications. Still, their number and diversity make a detailed characterization of all potentially promising phages virtually impossible. Moreover, no single phage combines all the features required of an ideal therapeutic agent. Additionally, the rapid acquisition of phage resistance by bacteria may make phages already approved for therapy ineffective and turn the search for environmental phages of better efficacy and new specificity into an endless race. An alternative strategy for acquiring phages with desired properties in a short time with minimal cost regarding their acquisition, characterization, and approval for therapy could be based on targeted genome modifications of phage isolates with known properties. The first example demonstrating the potential of this strategy in curing bacterial diseases resistant to traditional therapy is the recent successful treatment of a progressing disseminated Mycobacterium abscessus infection in a teenage patient with the use of an engineered phage. In this review, we briefly present current methods of phage genetic engineering, highlighting their advantages and disadvantages, and provide examples of genetically engineered phages with a modified host range, improved safety or antibacterial activity, and proven therapeutic efficacy. We also summarize novel uses of engineered phages not only for killing pathogenic bacteria, but also for in situ modification of human microbiota to attenuate symptoms of certain bacterial diseases and metabolic, immune, or mental disorders.
Collapse
Affiliation(s)
- Małgorzata Łobocka
- Institute of Biochemistry and Biophysics of the Polish Academy of Sciences, Warsaw, Poland
| | - Krystyna Dąbrowska
- Institute of Immunology and Experimental Therapy of the Polish Academy of Sciences, Wrocław, Poland
| | - Andrzej Górski
- Institute of Immunology and Experimental Therapy of the Polish Academy of Sciences, Wrocław, Poland
| |
Collapse
|
40
|
Gafur A, Sukamdani GY, Kristi N, Maruf A, Xu J, Chen X, Wang G, Ye Z. From bulk to nano-delivery of essential phytochemicals: recent progress and strategies for antibacterial resistance. J Mater Chem B 2021; 8:9825-9835. [PMID: 33000844 DOI: 10.1039/d0tb01671c] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Bacterial biofilms caused by antibiotic resistance are a severe cause of infection threatening human health nowadays. The primary causes of this emerging threat are poor penetration of conventional antibiotics and the growing number of varied strains of resistant bacteria. Recently, bulk phytochemical oils have been widely explored for their potential as antibacterial agents. However, due to their poor solubility, low stability, and highly volatile properties, essential oils are not effective for in vitro and in vivo antibacterial applications and require further preparation. In this review, we discuss the recent progress and strategies to overcome the drawbacks of bulk phytochemical oils using nano-delivery, as well as the current challenges and future outlook of these nano-delivery systems against bacterial resistance.
Collapse
Affiliation(s)
- Alidha Gafur
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Faculty of Medicine, Chongqing University, Chongqing, 400030, China.
| | - Gerry Yusuf Sukamdani
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Faculty of Medicine, Chongqing University, Chongqing, 400030, China.
| | - Natalia Kristi
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Faculty of Medicine, Chongqing University, Chongqing, 400030, China.
| | - Ali Maruf
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Faculty of Medicine, Chongqing University, Chongqing, 400030, China.
| | - Jing Xu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Faculty of Medicine, Chongqing University, Chongqing, 400030, China.
| | - Xue Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Faculty of Medicine, Chongqing University, Chongqing, 400030, China.
| | - Guixue Wang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Faculty of Medicine, Chongqing University, Chongqing, 400030, China.
| | - Zhiyi Ye
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College, Faculty of Medicine, Chongqing University, Chongqing, 400030, China.
| |
Collapse
|
41
|
Fage C, Lemire N, Moineau S. Delivery of CRISPR-Cas systems using phage-based vectors. Curr Opin Biotechnol 2020; 68:174-180. [PMID: 33360715 DOI: 10.1016/j.copbio.2020.11.012] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Revised: 11/15/2020] [Accepted: 11/23/2020] [Indexed: 12/14/2022]
Abstract
Antimicrobial resistance has spread quickly on a worldwide scale, reducing therapeutic options for bacterial infections. CRISPR-Cas is an adaptive immune system found in many prokaryotes that can be designed to target bacterial genomes, leading to cell death. Repurposing the CRISPR-Cas system as a therapeutic strategy offers an attractive way to overcome antimicrobial resistance. However, this strategy requires efficient vectors for the CRISPR-Cas system to reach the bacterial genomes. Engineered phages offer an attractive option as cargo delivery vectors. In this review, we discuss the production of phage-based vectors and the relevance of using repurposed CRISPR-Cas systems as antimicrobials. We also discuss recent progress in phage engineering that can potentially overcome the limitations and increase the efficiency of CRISPR-Cas delivery.
Collapse
Affiliation(s)
- Clément Fage
- Département de biochimie, de microbiologie, et de bio-informatique, Faculté des sciences et de génie, Université Laval, Québec City, QC, Canada; Groupe de recherche en écologie buccale, Faculté de médecine dentaire, Université Laval, Québec City, QC, Canada
| | - Nicolas Lemire
- Département de biochimie, de microbiologie, et de bio-informatique, Faculté des sciences et de génie, Université Laval, Québec City, QC, Canada; Groupe de recherche en écologie buccale, Faculté de médecine dentaire, Université Laval, Québec City, QC, Canada
| | - Sylvain Moineau
- Département de biochimie, de microbiologie, et de bio-informatique, Faculté des sciences et de génie, Université Laval, Québec City, QC, Canada; Groupe de recherche en écologie buccale, Faculté de médecine dentaire, Université Laval, Québec City, QC, Canada; Félix d'Hérelle Reference Center for Bacterial Viruses, Faculté de médecine dentaire, Université Laval, Québec City, QC, Canada.
| |
Collapse
|
42
|
Wu Y, Deng G, Jiang K, Wang H, Song Z, Han H. Photothermally triggered nitric oxide nanogenerator targeting type IV pili for precise therapy of bacterial infections. Biomaterials 2020; 268:120588. [PMID: 33307370 DOI: 10.1016/j.biomaterials.2020.120588] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 10/27/2020] [Accepted: 11/27/2020] [Indexed: 12/24/2022]
Abstract
Nitric oxide (NO) is an important biological messenger involved in the treatment of bacterial infections, but its controlled and targeted release in bacterial infections remains a major challenge. Herein, an intelligent NO nanogenerator triggered by near-infrared (NIR) light is constructed for targeted treatment of P. aeruginosa bacterial infection. Since maleimide can recognize and attach to the pilus of T4P of P. aeruginosa, we adopt this strategy to achieve the accurate release of therapeutic drugs at the infection site, i.e., after maleimide targets Gram-negative bacteria, the SNP@MOF@Au-Mal nanogenerator will release NO and generate ROS in situ from the inorganic photosensitizer gold nanoparticles under NIR irradiation to achieve synergistic antibacterial effect. In vivo experiments proved that the bacterial burden on the wound was reduced by 97.7%. Additionally, the nanogenerator was shown to promote the secretion of growth factors, which play a key role in regulating inflammation and inducing angiogenesis. This strategy has the advantage of generating a high concentration of NO in situ to promote the transfer of more NO and its derivatives (N2O3, ONOO-) to bacteria, thereby significantly improving the antibacterial effect. The multifunctional antibacterial platform has been demonstrated as a good carrier for gas therapy because of its simple and efficient gas release performance, indicating its great potential for the treatment of drug-resistant bacterial infections.
Collapse
Affiliation(s)
- Yang Wu
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Guiyun Deng
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Kai Jiang
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, 430070, China
| | - Huajuan Wang
- State Key Laboratory of Agricultural Microbiology, College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China
| | - Zhiyong Song
- State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Heyou Han
- State Key Laboratory of Agricultural Microbiology, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China; State Key Laboratory of Agricultural Microbiology, College of Science, Huazhong Agricultural University, Wuhan, 430070, China; State Key Laboratory of Agricultural Microbiology, College of Food Science and Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
43
|
Kumar P, Sinha R, Shukla P. Artificial intelligence and synthetic biology approaches for human gut microbiome. Crit Rev Food Sci Nutr 2020; 62:2103-2121. [PMID: 33249867 DOI: 10.1080/10408398.2020.1850415] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The gut microbiome comprises a variety of microorganisms whose genes encode proteins to carry out crucial metabolic functions that are responsible for the majority of health-related issues in human beings. The advent of the technological revolution in artificial intelligence (AI) assisted synthetic biology (SB) approaches will play a vital role in the modulating the therapeutic and nutritive potential of probiotics. This can turn human gut as a reservoir of beneficial bacterial colonies having an immense role in immunity, digestion, brain function, and other health benefits. Hence, in the present review, we have discussed the role of several gene editing tools and approaches in synthetic biology that have equipped us with novel tools like Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR-Cas) systems to precisely engineer probiotics for diagnostic, therapeutic and nutritive value. A brief discussion over the AI techniques to understand the metagenomic data from the healthy and diseased gut microbiome is also presented. Further, the role of AI in potentially impacting the pace of developments in SB and its current challenges is also discussed. The review also describes the health benefits conferred by engineered microbes through the production of biochemicals, nutraceuticals, drugs or biotherapeutics molecules etc. Finally, the review concludes with the challenges and regulatory concerns in adopting synthetic biology engineered microbes for clinical applications. Thus, the review presents a synergistic approach of AI and SB toward human gut microbiome for better health which will provide interesting clues to researchers working in the area of rapidly evolving food and nutrition science.
Collapse
Affiliation(s)
- Prasoon Kumar
- Department of Biotechnology and Medical Engineering, National Institute of Technology, Rourkela, India.,Department of Medical Devices, National Institute of Pharmaceutical Education and Research, Ahmedabad, India
| | | | - Pratyoosh Shukla
- School of Biotechnology, Institute of Science, Banaras Hindu University, Varanasi, India.,Enzyme Technology and Protein Bioinformatics Laboratory, Department of Microbiology, Maharshi Dayanand University, Rohtak, India
| |
Collapse
|
44
|
Ribeiro CFA, Silveira GGDOS, Cândido EDS, Cardoso MH, Espínola Carvalho CM, Franco OL. Effects of Antibiotic Treatment on Gut Microbiota and How to Overcome Its Negative Impacts on Human Health. ACS Infect Dis 2020; 6:2544-2559. [PMID: 32786282 DOI: 10.1021/acsinfecdis.0c00036] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The need for new antimicrobial therapies is evident, especially to reduce antimicrobial resistance and minimize deleterious effects on gut microbiota. However, although diverse studies discuss the adverse effects of broad-spectrum antibiotics on the microbiome ecology, targeted interventions that could solve this problem have often been overlooked. The impact of antibiotics on gut microbiota homeostasis is alarming, compromising its microbial community and leading to changes in host health. Recent studies have shown that these impacts can be transient or permanent, causing irreversible damage to gut microbiota. The responses to and changes in the gut microbial community arising from antibiotic treatment are related to its duration, the number of doses, antibiotic class, host age, genetic susceptibility, and lifestyle. In contrast, each individual's native microbiota can also affect the response to treatment as well as respond differently to antibiotic treatment. In this context, the current challenge is to promote the growth of potentially beneficial microorganisms and to reduce the proportion of microorganisms that cause dysbiosis, thus contributing to an improvement in the patient's health. An essential requirement for the development of novel antibiotics will be personalized medicinal strategies that recognize a patient's intestinal and biochemical individuality. Thus, this Review will address a new perspective on antimicrobial therapies through pathogen-selective antibiotics that minimize the impacts on human health due to changes in the gut microbiota from the use of antibiotics.
Collapse
Affiliation(s)
- Camila Fontoura Acosta Ribeiro
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-900, Brazil
| | | | - Elizabete de Souza Cândido
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-900, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Federal District 71966-700, Brazil
| | - Marlon Henrique Cardoso
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-900, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Federal District 71966-700, Brazil
| | - Cristiano Marcelo Espínola Carvalho
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-900, Brazil
| | - Octávio Luiz Franco
- S-Inova Biotech, Programa de Pós-Graduação em Biotecnologia, Universidade Católica Dom Bosco, Campo Grande, Mato Grosso do Sul 79117-900, Brazil
- Centro de Análises Proteômicas e Bioquímicas, Pós-Graduação em Ciências Genômicas e Biotecnologia, Universidade Católica de Brasília, Brasília, Federal District 71966-700, Brazil
| |
Collapse
|
45
|
Challenges & opportunities for phage-based in situ microbiome engineering in the gut. J Control Release 2020; 326:106-119. [DOI: 10.1016/j.jconrel.2020.06.016] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/14/2020] [Accepted: 06/15/2020] [Indexed: 12/16/2022]
|
46
|
Federici S, Nobs SP, Elinav E. Phages and their potential to modulate the microbiome and immunity. Cell Mol Immunol 2020; 18:889-904. [PMID: 32901128 DOI: 10.1038/s41423-020-00532-4] [Citation(s) in RCA: 99] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 08/06/2020] [Indexed: 02/07/2023] Open
Abstract
Bacteriophages (hence termed phages) are viruses that target bacteria and have long been considered as potential future treatments against antibiotic-resistant bacterial infection. However, the molecular nature of phage interactions with bacteria and the human host has remained elusive for decades, limiting their therapeutic application. While many phages and their functional repertoires remain unknown, the advent of next-generation sequencing has increasingly enabled researchers to decode new lytic and lysogenic mechanisms by which they attack and destroy bacteria. Furthermore, the last decade has witnessed a renewed interest in the utilization of phages as therapeutic vectors and as a means of targeting pathogenic or commensal bacteria or inducing immunomodulation. Importantly, the narrow host range, immense antibacterial repertoire, and ease of manipulating phages may potentially allow for their use as targeted modulators of pathogenic, commensal and pathobiont members of the microbiome, thereby impacting mammalian physiology and immunity along mucosal surfaces in health and in microbiome-associated diseases. In this review, we aim to highlight recent advances in phage biology and how a mechanistic understanding of phage-bacteria-host interactions may facilitate the development of novel phage-based therapeutics. We provide an overview of the challenges of the therapeutic use of phages and how these could be addressed for future use of phages as specific modulators of the human microbiome in a variety of infectious and noncommunicable human diseases.
Collapse
Affiliation(s)
- Sara Federici
- Immunology Department, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Samuel P Nobs
- Immunology Department, Weizmann Institute of Science, Rehovot, 7610001, Israel
| | - Eran Elinav
- Immunology Department, Weizmann Institute of Science, Rehovot, 7610001, Israel. .,Cancer-Microbiome Division Deutsches Krebsforschungszentrum (DKFZ), Neuenheimer Feld 280, 69120, Heidelberg, Germany.
| |
Collapse
|
47
|
Antimicrobial antisense RNA delivery to F-pili producing multidrug-resistant bacteria via a genetically engineered bacteriophage. Biochem Biophys Res Commun 2020; 530:533-540. [PMID: 32739024 DOI: 10.1016/j.bbrc.2020.06.088] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 06/18/2020] [Indexed: 12/16/2022]
Abstract
Multidrug-resistant bacteria are a growing issue worldwide. This study developed a convenient and effective method to downregulate the expression of a specific gene to produce a novel antimicrobial tool using a small (140 nucleotide) RNA with a 24-nucleotide antisense (as) region from an arabinose-inducible expression phagemid vector in Escherichia coli. Knockdown effects of rpoS encoding RNA polymerase sigma factor were observed using this inducible artificial asRNA approach. asRNAs targeting several essential E. coli genes produced significant growth defects, especially when targeted to acpP and ribosomal protein coding genes rplN, rplL, and rpsM. Growth inhibited phenotypes were facilitated in hfq- conditions. Phage lysates were prepared from cells harboring phagemids as a lethal-agent delivery tool. Targeting the rpsM gene by phagemid-derived M13 phage infection of E. coli containing a carbapenem-producing F-plasmid and multidrug-resistant Klebsiella pneumoniae containing an F-plasmid resulted in the death of over 99.99% of infected bacteria. This study provides a possible strategy for treating bacterial infection and can be applied to any F-pilus producing bacterial species.
Collapse
|
48
|
Dong X, Pan P, Zheng DW, Bao P, Zeng X, Zhang XZ. Bioinorganic hybrid bacteriophage for modulation of intestinal microbiota to remodel tumor-immune microenvironment against colorectal cancer. SCIENCE ADVANCES 2020; 6:eaba1590. [PMID: 32440552 PMCID: PMC7228756 DOI: 10.1126/sciadv.aba1590] [Citation(s) in RCA: 172] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Accepted: 03/03/2020] [Indexed: 05/17/2023]
Abstract
Mounting evidence suggests that the gut microbiota contribute to colorectal cancer (CRC) tumorigenesis, in which the symbiotic Fusobacterium nucleatum (Fn) selectively increases immunosuppressive myeloid-derived suppressor cells (MDSCs) to hamper the host's anticancer immune response. Here, a specifically Fn-binding M13 phage was screened by phage display technology. Then, silver nanoparticles (AgNP) were assembled electrostatically on its surface capsid protein (M13@Ag) to achieve specific clearance of Fn and remodel the tumor-immune microenvironment. Both in vitro and in vivo studies showed that of M13@Ag treatment could scavenge Fn in gut and lead to reduction in MDSC amplification in the tumor site. In addition, antigen-presenting cells (APCs) were activated by M13 phages to further awaken the host immune system for CRC suppression. M13@Ag combined with immune checkpoint inhibitors (α-PD1) or chemotherapeutics (FOLFIRI) significantly prolonged overall mouse survival in the orthotopic CRC model.
Collapse
Affiliation(s)
- Xue Dong
- Institute for Advanced Studies, Wuhan University, Wuhan 430072, P. R. China
| | - Pei Pan
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Di-Wei Zheng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Peng Bao
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xuan Zeng
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
| | - Xian-Zheng Zhang
- Institute for Advanced Studies, Wuhan University, Wuhan 430072, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education & Department of Chemistry, Wuhan University, Wuhan 430072, P. R. China
- Corresponding author.
| |
Collapse
|
49
|
Hellweger FL, Vick C, Rückbeil F, Bucci V. Fresh Ideas Bloom in Gut Healthcare to Cross-Fertilize Lake Management. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2019; 53:14099-14112. [PMID: 31647664 DOI: 10.1021/acs.est.9b04218] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Harmful bacteria may be the most significant threat to human gut and lake ecosystem health, and they are often managed using similar tools, like poisoning with antibiotics or algicides. Out-of-the-box thinking in human microbiome engineering is leading to novel methods, like engineering bacteria to kill pathogens, "persuade" them not to produce toxins, or "mop up" their toxins. The bacterial agent can be given a competitive edge via an exclusive nutrient, and they can be engineered to commit suicide once their work is done. Viruses can kill pathogens with specific DNA sequences or knock out their antibiotic resistance genes using CRISPR technology. Some of these ideas may work for lakes. We critically review novel methods for managing harmful bacteria in the gut from the perspective of managing toxic cyanobacteria in lakes, and discuss practical aspects such as modifying bacteria using genetic engineering or directed evolution, mass culturing and controlling the agents. A key knowledge gap is in the ecology of strains, like toxigenic vs nontoxigenic Microcystis, including allelopathic and Black Queen interactions. Some of the "gut methods" may have future potential for lakes, but there presently is no substitute for established management approaches, including reducing N and P nutrient inputs, and mitigating climate change.
Collapse
Affiliation(s)
- Ferdi L Hellweger
- Water Quality Engineering , Technical University of Berlin , Berlin 10623 , Germany
| | - Carsten Vick
- Water Quality Engineering , Technical University of Berlin , Berlin 10623 , Germany
| | - Fiona Rückbeil
- Water Quality Engineering , Technical University of Berlin , Berlin 10623 , Germany
| | - Vanni Bucci
- Department of Bioengineering , University of Massachusetts Dartmouth , North Dartmouth , Massachusetts 02747 , United States
| |
Collapse
|
50
|
Bacterial Heterogeneity and Antibiotic Survival: Understanding and Combatting Persistence and Heteroresistance. Mol Cell 2019; 76:255-267. [DOI: 10.1016/j.molcel.2019.09.028] [Citation(s) in RCA: 74] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Revised: 09/20/2019] [Accepted: 09/23/2019] [Indexed: 12/20/2022]
|