1
|
Han W, Liang X, Yao H, Zhang M, Chen Q, Xie Y, Liu Y, Cai H, Zhang C, Zhang Y. Rational design of a dual-bacterial system for synchronous removal of antibiotics and Pb(Ⅱ)/Cd(Ⅱ) from water. JOURNAL OF HAZARDOUS MATERIALS 2025; 485:136773. [PMID: 39657493 DOI: 10.1016/j.jhazmat.2024.136773] [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: 09/03/2024] [Revised: 11/25/2024] [Accepted: 12/03/2024] [Indexed: 12/12/2024]
Abstract
Facing the combined pollution of antibiotics and heavy metals caused by livestock excrement and industrial effluents, how to use microbial technology to remove these pollutants simultaneously is an important research topic in environmental remediation. In addition, quick separation of the bacteria-water after remediation is also an urgent problem. In this study, we gradually developed a dual-bacteria microbial treatment technology capable of removing Pb(Ⅱ), Cd(Ⅱ) and common antibiotics, as well as self-settling after treatment. The key technology in this study mainly includes modifying the bacterial membrane proteins using Pb-binding protein PbrR, Cd-binding protein CadR and bacterial laccase CotA via surface-display technology to maximize the removal of Pb(Ⅱ), Cd(Ⅱ) and antibiotics, separately. Besides, the introduction of nanobody-antigen adhesion facilitated the self-settling in dual-bacterial system. Then, we studied its effectiveness in removing single pollutants, analyzed the influence of different heavy metal ions, and conducted detailed studies on the kinetics. Further characterization of heavy metal biosorption behavior was conducted using SEM, SEM-EDS, FTIR, and XPS techniques. Via protein fusion and dual vector expression, we constructed a dual-bacteria treatment system that could achieve rapid, selective removal of combined pollutants at a wide pH range temperature range, ultimately precipitating at bottom. Finally, molecular dynamics simulation was employed to elucidate the molecular mechanism underlying the selective biosorption by metal-binding proteins. The findings in this study hold significant implications for achieving selective pollutant removal using engineering bacteria in complex water environments.
Collapse
Affiliation(s)
- Wei Han
- School of Resources and Environment, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Xinying Liang
- School of Resources and Environment, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Hongkai Yao
- School of Resources and Environment, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Meng Zhang
- School of Resources and Environment, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Qi Chen
- School of Resources and Environment, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Yuzhu Xie
- School of Resources and Environment, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Yuan Liu
- School of Resources and Environment, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Hongguang Cai
- Jilin Academy of Agricultural Sciences, Changchun, Jilin 130033, PR China
| | - Congyu Zhang
- School of Resources and Environment, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China
| | - Ying Zhang
- School of Resources and Environment, Northeast Agricultural University, Harbin, Heilongjiang 150030, PR China.
| |
Collapse
|
2
|
Liu J, Wu L, Xie A, Liu W, He Z, Wan Y, Mao W. Unveiling the new chapter in nanobody engineering: advances in traditional construction and AI-driven optimization. J Nanobiotechnology 2025; 23:87. [PMID: 39915791 PMCID: PMC11800653 DOI: 10.1186/s12951-025-03169-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/27/2025] [Indexed: 02/11/2025] Open
Abstract
Nanobodies (Nbs), miniature antibodies consisting solely of the variable region of heavy chains, exhibit unique properties such as small size, high stability, and strong specificity, making them highly promising for disease diagnosis and treatment. The engineering production of Nbs has evolved into a mature process, involving library construction, screening, and expression purification. Different library types, including immune, naïve, and synthetic/semi-synthetic libraries, offer diverse options for various applications, while display platforms like phage display, cell surface display, and non-surface display provide efficient screening of target Nbs. Recent advancements in artificial intelligence (AI) have opened new avenues in Nb engineering. AI's exceptional performance in protein structure prediction and molecular interaction simulation has introduced novel perspectives and tools for Nb design and optimization. Integrating AI with traditional experimental methods is anticipated to enhance the efficiency and precision of Nb development, expediting the transition from basic research to clinical applications. This review comprehensively examines the latest progress in Nb engineering, emphasizing library construction strategies, display platform technologies, and AI applications. It evaluates the strengths and weaknesses of various libraries and display platforms and explores the potential and challenges of AI in predicting Nb structure, antigen-antibody interactions, and optimizing physicochemical properties.
Collapse
Affiliation(s)
- Jiwei Liu
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
- Wuxi College of Clinical Medicine, Nanjing Medical University, Wuxi, 214023, China
| | - Lei Wu
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
- Wuxi College of Clinical Medicine, Nanjing Medical University, Wuxi, 214023, China
| | - Anqi Xie
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
| | - Weici Liu
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
- Wuxi College of Clinical Medicine, Nanjing Medical University, Wuxi, 214023, China
| | - Zhao He
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China
- Wuxi College of Clinical Medicine, Nanjing Medical University, Wuxi, 214023, China
| | - Yuan Wan
- The Pq Laboratory of BiomeDx/Rx, Department of Biomedical Engineering, Binghamton University, Binghamton, 13850, USA.
- Department of Biomedical Engineering, The Pq Laboratory of BiomeDx/Rx, Binghamton University, Binghamton, NY, 13902, USA.
| | - Wenjun Mao
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Wuxi, 214023, China.
- Wuxi College of Clinical Medicine, Nanjing Medical University, Wuxi, 214023, China.
- Department of Thoracic Surgery, Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, No. 299 Qingyang Rd., Wuxi, 214023, China.
| |
Collapse
|
3
|
Ullrich T, Klimenkova O, Pollmann C, Lasram A, Hatskovska V, Maksymenko K, Milijaš-Jotić M, Schenk L, Lengerke C, Hartmann MD, Piehler J, Skokowa J, ElGamacy M. A strategy to design protein-based antagonists against type I cytokine receptors. PLoS Biol 2024; 22:e3002883. [PMID: 39591631 PMCID: PMC11596305 DOI: 10.1371/journal.pbio.3002883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 10/06/2024] [Indexed: 11/28/2024] Open
Abstract
Excessive cytokine signaling resulting from dysregulation of a cytokine or its receptor can be a main driver of cancer, autoimmune, or hematopoietic disorders. Here, we leverage protein design to create tailored cytokine receptor blockers with idealized properties. Specifically, we aimed to tackle the granulocyte-colony stimulating factor receptor (G-CSFR), a mediator of different types of leukemia and autoinflammatory diseases. By modifying designed G-CSFR binders, we engineered hyper-stable proteins that function as nanomolar signaling antagonists. X-ray crystallography showed atomic-level agreement with the experimental structure of an exemplary design. Furthermore, the most potent design blocks G-CSFR in acute myeloid leukemia cells and primary human hematopoietic stem cells. Thus, the resulting designs can be used for inhibiting or homing to G-CSFR-expressing cells. Our results also demonstrate that similarly designed cytokine mimics can be used to derive antagonists to tackle other type I cytokine receptors.
Collapse
Affiliation(s)
- Timo Ullrich
- Max Planck Institute for Biology, Department of Protein Evolution, Tübingen, Germany
| | - Olga Klimenkova
- Translational Oncology, Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Christoph Pollmann
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Asma Lasram
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Valeriia Hatskovska
- Max Planck Institute for Biology, Department of Protein Evolution, Tübingen, Germany
- Translational Oncology, Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Kateryna Maksymenko
- Max Planck Institute for Biology, Department of Protein Evolution, Tübingen, Germany
| | - Matej Milijaš-Jotić
- Max Planck Institute for Biology, Department of Protein Evolution, Tübingen, Germany
| | - Lukas Schenk
- Translational Oncology, Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Claudia Lengerke
- Translational Oncology, Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Marcus D. Hartmann
- Max Planck Institute for Biology, Department of Protein Evolution, Tübingen, Germany
- Interfaculty Institute of Biochemistry, University of Tübingen, Tübingen, Germany
| | - Jacob Piehler
- Department of Biology/Chemistry and Center for Cellular Nanoanalytics, Osnabrück University, Osnabrück, Germany
| | - Julia Skokowa
- Translational Oncology, Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| | - Mohammad ElGamacy
- Max Planck Institute for Biology, Department of Protein Evolution, Tübingen, Germany
- Translational Oncology, Internal Medicine II, University Hospital Tübingen, Tübingen, Germany
| |
Collapse
|
4
|
Schreiber S, Zaayenga A, Jose J. The Assembly of the Inverse Autotransporter Protein YeeJ is Driven by its C-terminal β-strand. J Mol Biol 2024; 436:168749. [PMID: 39173735 DOI: 10.1016/j.jmb.2024.168749] [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: 05/08/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 08/24/2024]
Abstract
Autotransporter proteins are bacterial outer membrane proteins that display passenger domains with various functions through a β-barrel shaped translocation domain. YeeJ is an autotransporter protein from E. coli MG1655. In contrast to most other autotransporter proteins, its passenger domain is located at the C-terminus of the translocation domain. Due to this inverted domain organization, YeeJ belongs to autotransporter proteins of type Ve. To investigate the assembly of YeeJ, the fluorescence of a heterologous mCherry passenger domain was measured to quantify its assembly. Based on AlphaFold2 models of 119 sequences similar to YeeJ, a sequence conservation logo for the β1- and the β12-strand of type Ve autotransporter proteins was generated. Then, the effect of mutations in these strands on the assembly of YeeJ were analyzed. Mutations of the N-terminal aromatic amino acid of the β1-strand did not affect the assembly of the translocation domain and the display of the passenger domain. Likewise, exchange of the β1-strand with the β3-strand did not impair the assembly of the autotransporter fusion protein. Mutation of the C-terminal aromatic amino acid of the β12-strand strongly impaired surface display of the mCherry passenger domain. This amino acid has been shown before as an essential feature of the β-signals of classical autotransporter proteins and outer membrane β-barrel proteins in general. We therefore propose that the β12-strand of YeeJ acts as its β-signal and that the assembly of the YeeJ β-barrel is driven by its C-terminal β-strand, like in most other autotransporter proteins, despite its inverted domain organization.
Collapse
Affiliation(s)
- Sebastian Schreiber
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, PharmaCampus, Corrensstr. 48, 48149 Münster, Germany
| | - Annika Zaayenga
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, PharmaCampus, Corrensstr. 48, 48149 Münster, Germany
| | - Joachim Jose
- University of Münster, Institute of Pharmaceutical and Medicinal Chemistry, PharmaCampus, Corrensstr. 48, 48149 Münster, Germany.
| |
Collapse
|
5
|
Chen PY, Chen YC, Chen PP, Lin KT, Sargsyan K, Hsu CP, Wang WL, Hsia KC, Ting SY. A whole-cell platform for discovering synthetic cell adhesion molecules in bacteria. Nat Commun 2024; 15:6568. [PMID: 39095377 PMCID: PMC11297345 DOI: 10.1038/s41467-024-51017-1] [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: 12/14/2023] [Accepted: 07/26/2024] [Indexed: 08/04/2024] Open
Abstract
Developing programmable bacterial cell-cell adhesion is of significant interest due to its versatile applications. Current methods that rely on presenting cell adhesion molecules (CAMs) on bacterial surfaces are limited by the lack of a generalizable strategy to identify such molecules targeting bacterial membrane proteins in their natural states. Here, we introduce a whole-cell screening platform designed to discover CAMs targeting bacterial membrane proteins within a synthetic bacteria-displayed nanobody library. Leveraging the potency of the bacterial type IV secretion system-a contact-dependent DNA delivery nanomachine-we have established a positive feedback mechanism to selectively enrich for bacteria displaying nanobodies that target antigen-expressing cells. Our platform successfully identified functional CAMs capable of recognizing three distinct outer membrane proteins (TraN, OmpA, OmpC), demonstrating its efficacy in CAM discovery. This approach holds promise for engineering bacterial cell-cell adhesion, such as directing the antibacterial activity of programmed inhibitor cells toward target bacteria in mixed populations.
Collapse
Affiliation(s)
- Po-Yin Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, Taiwan
| | - Yung-Chih Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
| | - Po-Pang Chen
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Kuan-Ting Lin
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Program in Molecular Medicine, National Yang-Ming Chao-Tung University and Academia Sinica, Taipei, Taiwan
| | | | - Chao-Ping Hsu
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Physics Division, National Center for Theoretical Sciences, Taipei, Taiwan
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan
| | - Wei-Le Wang
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, Taiwan
- Program in Molecular Medicine, National Yang-Ming Chao-Tung University and Academia Sinica, Taipei, Taiwan
| | - Kuo-Chiang Hsia
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, Taiwan
- Institute of Biochemistry and Molecular Biology, College of Life Sciences, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - See-Yeun Ting
- Institute of Molecular Biology, Academia Sinica, Taipei, Taiwan.
- Molecular and Cell Biology, Taiwan International Graduate Program, Academia Sinica and National Defense Medical Center, Taipei, Taiwan.
- Genome and Systems Biology Degree Program, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
6
|
Xu X, Ding Y, Dong Y, Yuan H, Xia P, Qu C, Ma J, Wang H, Zhang X, Zhao L, Li Z, Liang Z, Wang J. Nanobody-Engineered Biohybrid Bacteria Targeting Gastrointestinal Cancers Induce Robust STING-Mediated Anti-Tumor Immunity. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2401905. [PMID: 38888519 PMCID: PMC11336900 DOI: 10.1002/advs.202401905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 05/22/2024] [Indexed: 06/20/2024]
Abstract
Bacteria can be utilized for cancer therapy owing to their preferential colonization at tumor sites. However, unmodified non-pathogenic bacteria carry potential risks due to their non-specific targeting effects, and their anti-tumor activity is limited when used as monotherapy. In this study, a biohybrid-engineered bacterial system comprising non-pathogenic MG1655 bacteria modified with CDH17 nanobodies on their surface and conjugated with photosensitizer croconium (CR) molecules is developed. The resultant biohybrid bacteria can efficiently home to CDH17-positive tumors, including gastric, pancreatic, and colorectal cancers, and significantly suppress tumor growth upon irradiation. More importantly, biohybrid bacteria-mediated photothermal therapy (PTT) induced abundant macrophage infiltration in a syngeneic murine colorectal model. Further, that the STING pathway is activated in tumor macrophages by the released bacterial nucleic acid after PTT is revealed, leading to the production of type I interferons. The addition of CD47 nanobody but not PD-1 antibody to the PTT regimen can eradicate the tumors and extend survival. This results indicate that bacteria endowed with tumor-specific selectivity and coupled with photothermal payloads can serve as an innovative strategy for low-immunogenicity cancers. This strategy can potentially reprogram the tumor microenvironment by inducing macrophage infiltration and enhancing the efficacy of immunotherapy targeting macrophages.
Collapse
Affiliation(s)
- Xiaolong Xu
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Integrated Chinese and Western Medicine Postdoctoral Research StationJinan UniversityGuangzhou510632China
| | - Youbin Ding
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Department of Medical ImagingThe Third Affiliated HospitalSouthern Medical University (Academy of Orthopedics Guangdong Province)Guangzhou510515China
| | - Yafang Dong
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Department of Medical ImagingThe Third Affiliated HospitalSouthern Medical University (Academy of Orthopedics Guangdong Province)Guangzhou510515China
| | - Haitao Yuan
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Peng Xia
- Department of Hepatobiliary & Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanHubei430071China
| | - Chengming Qu
- Department of Hepatobiliary & Pancreatic SurgeryZhongnan Hospital of Wuhan UniversityWuhanHubei430071China
| | - Jingbo Ma
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Huifang Wang
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Xiaodong Zhang
- Department of Medical ImagingThe Third Affiliated HospitalSouthern Medical University (Academy of Orthopedics Guangdong Province)Guangzhou510515China
| | - Liang Zhao
- Department of PathologyShunde Hospital, Southern Medical University (The First People's Hospital of Shunde)Foshan528308China
- Department of Pathology & Guangdong Province Key Laboratory of Molecular Tumor Pathology, School of Basic Medical SciencesSouthern Medical UniversityGuangzhou510515China
| | - Zhijie Li
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Zhen Liang
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
| | - Jigang Wang
- Department of Geriatrics and Shenzhen Clinical Research Centre for Geriatrics, Department of UrologyShenzhen People's Hospital (The First Affiliated Hospital, Southern University of Science and TechnologyThe Second Clinical Medical CollegeJinan University)ShenzhenGuangdong518020China
- Department of OncologyThe Affiliated Hospital of Southwest Medical UniversityLuzhouSichuan646000China
- Department of Traditional Chinese Medicine and School of Pharmaceutical SciencesSouthern Medical UniversityGuangzhou510515China
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao‐di Herbs, Artemisinin Research Center, and Institute of Chinese Materia MedicaChina Academy of Chinese Medical SciencesBeijing100700China
- State Key Laboratory of Antiviral DrugsSchool of PharmacyHenan UniversityKaifeng475004China
| |
Collapse
|
7
|
Prieto A, Miró L, Margolles Y, Bernabeu M, Salguero D, Merino S, Tomas J, Corbera JA, Perez-Bosque A, Huttener M, Fernández LÁ, Juarez A. Targeting plasmid-encoded proteins that contain immunoglobulin-like domains to combat antimicrobial resistance. eLife 2024; 13:RP95328. [PMID: 39046772 PMCID: PMC11268884 DOI: 10.7554/elife.95328] [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] [Indexed: 07/25/2024] Open
Abstract
Antimicrobial resistance (AMR) poses a significant threat to human health. Although vaccines have been developed to combat AMR, it has proven challenging to associate specific vaccine antigens with AMR. Bacterial plasmids play a crucial role in the transmission of AMR. Our recent research has identified a group of bacterial plasmids (specifically, IncHI plasmids) that encode large molecular mass proteins containing bacterial immunoglobulin-like domains. These proteins are found on the external surface of the bacterial cells, such as in the flagella or conjugative pili. In this study, we show that these proteins are antigenic and can protect mice from infection caused by an AMR Salmonella strain harboring one of these plasmids. Furthermore, we successfully generated nanobodies targeting these proteins, that were shown to interfere with the conjugative transfer of IncHI plasmids. Considering that these proteins are also encoded in other groups of plasmids, such as IncA/C and IncP2, targeting them could be a valuable strategy in combating AMR infections caused by bacteria harboring different groups of AMR plasmids. Since the selected antigens are directly linked to AMR itself, the protective effect extends beyond specific microorganisms to include all those carrying the corresponding resistance plasmids.
Collapse
Affiliation(s)
- Alejandro Prieto
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Luïsa Miró
- Department of Biochemistry and Physiology, Universitat de BarcelonaBarcelonaSpain
- Institut de Nutrició i Seguretat Alimentària, Universitat de BarcelonaBarcelonaSpain
| | - Yago Margolles
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC)MadridSpain
| | - Manuel Bernabeu
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - David Salguero
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Susana Merino
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Joan Tomas
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Juan Alberto Corbera
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Facultad de Veterinaria, Universidad de Las Palmas de Gran Canaria (ULPGC), Campus Universitario de ArucasLas PalmasSpain
| | - Anna Perez-Bosque
- Department of Biochemistry and Physiology, Universitat de BarcelonaBarcelonaSpain
- Institut de Nutrició i Seguretat Alimentària, Universitat de BarcelonaBarcelonaSpain
| | - Mario Huttener
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC)MadridSpain
| | - Antonio Juarez
- Department of Genetics, Microbiology and Statistics, University of BarcelonaBarcelonaSpain
- Institute for Bioengineering of Catalonia, The Barcelona Institute of Science and TechnologyBarcelonaSpain
| |
Collapse
|
8
|
Costan SA, Ryan PM, Kim H, Wolgemuth CW, Riedel-Kruse IH. Biophysical characterization of synthetic adhesins for predicting and tuning engineered living material properties. MATTER 2024; 7:2125-2143. [PMID: 39165662 PMCID: PMC11335339 DOI: 10.1016/j.matt.2024.03.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Bacterial synthetic multicellular systems are promising platforms for engineered living materials (ELMs) for medical, biosynthesis, environmental, and smart materials applications. Recent advancements in genetically encoded adhesion toolkits have enabled precise manipulation of cell-cell adhesion and the design and patterning of self-assembled multicellular materials. However, in contrast to gene regulation in synthetic biology, the characterization and control of synthetic adhesins remains limited. Here, we demonstrate the quantitative characterization of a bacterial synthetic adhesion toolbox through various biophysical methods. We determine key parameters, including number of adhesins per cell, in-membrane diffusion constant, production and decay rates, and bond-breaking force between adhesins. With these parameters, we demonstrate the bottom-up prediction and quantitative tuning of macroscopic ELM properties (tensile strength) and, furthermore, that cells inside ELMs are connected only by a small fraction of available adhesins. These results enable the rational engineering, characterization, and modeling of other synthetic and natural adhesins and multicellular consortia.
Collapse
Affiliation(s)
- Stefana A. Costan
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85721, USA
| | - Paul M. Ryan
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
- Department of Physics, University of Arizona, Tucson, AZ 85721, USA
| | - Honesty Kim
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
| | - Charles W. Wolgemuth
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
- Department of Physics, University of Arizona, Tucson, AZ 85721, USA
- Department of Applied Mathematics, University of Arizona, Tucson, AZ 85721, USA
| | - Ingmar H. Riedel-Kruse
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85721, USA
- Department of Biomedical Engineering, University of Arizona, Tucson, AZ 85721, USA
- Department of Physics, University of Arizona, Tucson, AZ 85721, USA
- Department of Applied Mathematics, University of Arizona, Tucson, AZ 85721, USA
- Lead contact
| |
Collapse
|
9
|
Hinmon JA, King JM, Mayo LJ, Faries CR, Lockett YT, Crawford DW, Beardslee PC, Hendricks A, McNaughton BR. Cell surface β-lactamase recruitment: A facile selection to identify protein-protein interactions. Protein Sci 2024; 33:e4919. [PMID: 38501433 PMCID: PMC10949332 DOI: 10.1002/pro.4919] [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: 09/14/2023] [Revised: 12/17/2023] [Accepted: 01/25/2024] [Indexed: 03/20/2024]
Abstract
Protein-protein interactions (PPIs) are central to many cellular processes, and the identification of novel PPIs is a critical step in the discovery of protein therapeutics. Simple methods to identify naturally existing or laboratory evolved PPIs are therefore valuable research tools. We have developed a facile selection that links PPI-dependent β-lactamase recruitment on the surface of Escherichia coli with resistance to ampicillin. Bacteria displaying a protein that forms a complex with a specific protein-β-lactamase fusion are protected from ampicillin-dependent cell death. In contrast, bacteria that do not recruit β-lactamase to the cell surface are killed by ampicillin. Given its simplicity and tunability, we anticipate this selection will be a valuable addition to the palette of methods for illuminating and interrogating PPIs.
Collapse
Affiliation(s)
- Jordan A. Hinmon
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
| | - Jade M. King
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
| | - Latrina J. Mayo
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
| | - Cierra R. Faries
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
| | - Ya'hnis T. Lockett
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
| | - David W. Crawford
- Department of ChemistryColorado State UniversityFort CollinsColoradoUSA
| | | | | | - Brian R. McNaughton
- Department of Biological SciencesDelaware State UniversityDoverDelawareUSA
- Department of ChemistryColorado State UniversityFort CollinsColoradoUSA
| |
Collapse
|
10
|
Parks L, Ek M, Ståhl S, Löfblom J. Investigation of an AIDA-I based expression system for display of various affinity proteins on Escherichia coli. Biochem Biophys Res Commun 2024; 696:149534. [PMID: 38241810 DOI: 10.1016/j.bbrc.2024.149534] [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: 01/12/2024] [Accepted: 01/12/2024] [Indexed: 01/21/2024]
Abstract
Autotransporters constitute a large family of natural proteins that are essential for delivering many types of proteins and peptides across the outer membrane in Gram-negative bacteria. In biotechnology, autotransporters have been explored for display of recombinant proteins and peptides on the surface of Escherichia coli and have potential as tools for directed evolution of affinity proteins. Here, we investigate conditions for AIDA-I autotransporter-mediated display of recombinant proteins. A new expression vector was designed and engineered for this purpose, and a panel of proteins from different affinity-protein classes were subcloned to the vector, followed by evaluation of expression, surface display and functionality. Surface expression was explored in ten different E. coli strains together with assessment of transformation efficiencies. Furthermore, the most promising strain and expression vector combination was used in mock library selections for evaluation of magnetic-assisted cell sortings (MACS). The results demonstrated dramatically different performances depending on the type of affinity protein and choice of E. coli strain. The optimized MACS protocol showed efficient enrichment, and thus potential for the new AIDA-I display system to be used in methods for directed evolution of affinity proteins.
Collapse
Affiliation(s)
- Luke Parks
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - Moira Ek
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - Stefan Ståhl
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology, SE-106 91, Stockholm, Sweden.
| |
Collapse
|
11
|
Vázquez‐Arias A, Vázquez‐Iglesias L, Pérez‐Juste I, Pérez‐Juste J, Pastoriza‐Santos I, Bodelon G. Bacterial surface display of human lectins in Escherichia coli. Microb Biotechnol 2024; 17:e14409. [PMID: 38380565 PMCID: PMC10884992 DOI: 10.1111/1751-7915.14409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 01/02/2024] [Indexed: 02/22/2024] Open
Abstract
Lectin-glycan interactions sustain fundamental biological processes involved in development and disease. Owing to their unique sugar-binding properties, lectins have great potential in glycobiology and biomedicine. However, their relatively low affinities and broad specificities pose a significant challenge when used as analytical reagents. New approaches for expression and engineering of lectins are in demand to overcome current limitations. Herein, we report the application of bacterial display for the expression of human galectin-3 and mannose-binding lectin in Escherichia coli. The analysis of the cell surface expression and binding activity of the surface-displayed lectins, including point and deletion mutants, in combination with molecular dynamics simulation, demonstrate the robustness and suitability of this approach. Furthermore, the display of functional mannose-binding lectin in the bacterial surface proved the feasibility of this method for disulfide bond-containing lectins. This work establishes for the first time bacterial display as an efficient means for the expression and engineering of human lectins, thereby increasing the available toolbox for glycobiology research.
Collapse
Affiliation(s)
- Alba Vázquez‐Arias
- CINBIOUniversidade de VigoVigoSpain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS‐UVIGOVigoSpain
| | - Lorena Vázquez‐Iglesias
- CINBIOUniversidade de VigoVigoSpain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS‐UVIGOVigoSpain
| | | | - Jorge Pérez‐Juste
- CINBIOUniversidade de VigoVigoSpain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS‐UVIGOVigoSpain
- Departamento de Química FísicaUniversidade de VigoVigoSpain
| | - Isabel Pastoriza‐Santos
- CINBIOUniversidade de VigoVigoSpain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS‐UVIGOVigoSpain
- Departamento de Química FísicaUniversidade de VigoVigoSpain
| | - Gustavo Bodelon
- CINBIOUniversidade de VigoVigoSpain
- Galicia Sur Health Research Institute (IIS Galicia Sur), SERGAS‐UVIGOVigoSpain
- Departamento de Biología Funcional y Ciencias de la SaludUniversidade de VigoVigoSpain
| |
Collapse
|
12
|
Cerdán L, Álvarez B, Fernández LÁ. Massive integration of large gene libraries in the chromosome of Escherichia coli. Microb Biotechnol 2024; 17:e14367. [PMID: 37971317 PMCID: PMC10832519 DOI: 10.1111/1751-7915.14367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/21/2023] [Accepted: 10/22/2023] [Indexed: 11/19/2023] Open
Abstract
Large gene libraries are frequently created in Escherichia coli plasmids, which can induce cell toxicity and expression instability due to the high gene dosage. To address these limitations, gene libraries can be integrated in a single copy into the bacterial chromosome. Here, we describe an efficient system for the massive integration (MAIN) of large gene libraries in the E. coli chromosome that generates in-frame gene fusions that are expressed stably. MAIN uses a thermosensitive integrative plasmid that is linearized in vivo to promote extensive integration of the gene library via homologous recombination. Positive and negative selections efficiently remove bacteria lacking gene integration in the target site. We tested MAIN with a library of 107 VHH genes that encode nanobodies (Nbs). The integration of VHH genes into a custom target locus of the E. coli chromosome enabled stable expression and surface display of the Nbs. Next-generation DNA sequencing confirmed that MAIN preserved the diversity of the gene library after integration. Finally, we screened the integrated library to select Nbs that bind a specific antigen using magnetic and fluorescence-activated cell sorting. This allowed us to identify Nbs binding the epidermal growth factor receptor that were not previously isolated in a similar screening of a multicopy plasmid library. Our results demonstrate that MAIN enables large gene library integration into the E. coli chromosome, creating stably expressed in-frame fusions for functional screening.
Collapse
Affiliation(s)
- Lidia Cerdán
- Department of Microbial BiotechnologyCentro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
| | - Beatriz Álvarez
- Department of Microbial BiotechnologyCentro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
| | - Luis Ángel Fernández
- Department of Microbial BiotechnologyCentro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CSIC)MadridSpain
| |
Collapse
|
13
|
Tanniche I, Behkam B. Engineered live bacteria as disease detection and diagnosis tools. J Biol Eng 2023; 17:65. [PMID: 37875910 PMCID: PMC10598922 DOI: 10.1186/s13036-023-00379-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/18/2023] [Indexed: 10/26/2023] Open
Abstract
Sensitive and minimally invasive medical diagnostics are essential to the early detection of diseases, monitoring their progression and response to treatment. Engineered bacteria as live sensors are being developed as a new class of biosensors for sensitive, robust, noninvasive, and in situ detection of disease onset at low cost. Akin to microrobotic systems, a combination of simple genetic rules, basic logic gates, and complex synthetic bioengineering principles are used to program bacterial vectors as living machines for detecting biomarkers of diseases, some of which cannot be detected with other sensing technologies. Bacterial whole-cell biosensors (BWCBs) can have wide-ranging functions from detection only, to detection and recording, to closed-loop detection-regulated treatment. In this review article, we first summarize the unique benefits of bacteria as living sensors. We then describe the different bacteria-based diagnosis approaches and provide examples of diagnosing various diseases and disorders. We also discuss the use of bacteria as imaging vectors for disease detection and image-guided surgery. We conclude by highlighting current challenges and opportunities for further exploration toward clinical translation of these bacteria-based systems.
Collapse
Affiliation(s)
- Imen Tanniche
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, 24061, USA
| | - Bahareh Behkam
- Department of Mechanical Engineering, Virginia Tech, Blacksburg, VA, 24061, USA.
- School of Biomedical Engineered and Sciences, Virginia Tech, Blacksburg, VA, 24061, USA.
- Center for Engineered Health, Institute for Critical Technology and Applied Science, Virginia Tech, Blacksburg, VA, 24061, USA.
| |
Collapse
|
14
|
Gasse C, Srivastava P, Schepers G, Jose J, Hollenstein M, Marlière P, Herdewijn P. Controlled E. coli Aggregation Mediated by DNA and XNA Hybridization. Chembiochem 2023; 24:e202300191. [PMID: 37119472 DOI: 10.1002/cbic.202300191] [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: 03/10/2023] [Revised: 04/26/2023] [Accepted: 04/27/2023] [Indexed: 05/01/2023]
Abstract
Chemical cell surface modification is a fast-growing field of research, due to its enormous potential in tissue engineering, cell-based immunotherapy, and regenerative medicine. However, engineering of bacterial tissues by chemical cell surface modification has been vastly underexplored and the identification of suitable molecular handles is in dire need. We present here, an orthogonal nucleic acid-protein conjugation strategy to promote artificial bacterial aggregation. This system gathers the high selectivity and stability of linkage to a protein Tag expressed at the cell surface and the modularity and reversibility of aggregation due to oligonucleotide hybridization. For the first time, XNA (xeno nucleic acids in the form of 1,5-anhydrohexitol nucleic acids) were immobilized via covalent, SNAP-tag-mediated interactions on cell surfaces to induce bacterial aggregation.
Collapse
Affiliation(s)
- Cécile Gasse
- Génomique Métabolique, Genoscope Institut François Jacob, CEA, CNRS Univ Evry, Université Paris-Saclay, 2 Rue Gaston Crémieux, 91057, Evry, France
| | - Puneet Srivastava
- Laboratory of Medicinal Chemistry, Rega Institute for Biomedical Research, KU Leuven, Herestraat 49, Box 1041, 3000, Leuven, Belgium
| | - Guy Schepers
- Laboratory of Medicinal Chemistry, Rega Institute for Biomedical Research, KU Leuven, Herestraat 49, Box 1041, 3000, Leuven, Belgium
| | - Joachim Jose
- Institute of Pharmaceutical and Medicinal Chemistry, University of Münster, Corrensstr. 48, D-48149, Münster, Germany
| | - Marcel Hollenstein
- Institut Pasteur, Université Paris Cité, Department of Structural Biology and Chemistry, Laboratory for Bioorganic Chemistry of Nucleic Acids, CNRS UMR3523, 28, rue du Docteur Roux, 75724, Paris Cedex 15, France
| | - Philippe Marlière
- The European Syndicate of Synthetic Scientists and Industrialists (TESSSI), 81 rue Réaumur, 75002, Paris, France
| | - Piet Herdewijn
- Laboratory of Medicinal Chemistry, Rega Institute for Biomedical Research, KU Leuven, Herestraat 49, Box 1041, 3000, Leuven, Belgium
| |
Collapse
|
15
|
Chekli Y, Stevick RJ, Kornobis E, Briolat V, Ghigo JM, Beloin C. Escherichia coli Aggregates Mediated by Native or Synthetic Adhesins Exhibit Both Core and Adhesin-Specific Transcriptional Responses. Microbiol Spectr 2023; 11:e0069023. [PMID: 37039668 PMCID: PMC10269875 DOI: 10.1128/spectrum.00690-23] [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: 02/17/2023] [Accepted: 03/20/2023] [Indexed: 04/12/2023] Open
Abstract
Bacteria can rapidly tune their physiology and metabolism to adapt to environmental fluctuations. In particular, they can adapt their lifestyle to the close proximity of other bacteria or the presence of different surfaces. However, whether these interactions trigger transcriptomic responses is poorly understood. We used a specific setup of E. coli strains expressing native or synthetic adhesins mediating bacterial aggregation to study the transcriptomic changes of aggregated compared to nonaggregated bacteria. Our results show that, following aggregation, bacteria exhibit a core response independent of the adhesin type, with differential expression of 56.9% of the coding genome, including genes involved in stress response and anaerobic lifestyle. Moreover, when aggregates were formed via a naturally expressed E. coli adhesin (antigen 43), the transcriptomic response of the bacteria was more exaggerated than that of aggregates formed via a synthetic adhesin. This suggests that the response to aggregation induced by native E. coli adhesins could have been finely tuned during bacterial evolution. Our study therefore provides insights into the effect of self-interaction in bacteria and allows a better understanding of why bacterial aggregates exhibit increased stress tolerance. IMPORTANCE The formation of bacterial aggregates has an important role in both clinical and ecological contexts. Although these structures have been previously shown to be more resistant to stressful conditions, the genetic basis of this stress tolerance associated with the aggregate lifestyle is poorly understood. Surface sensing mediated by different adhesins can result in various changes in bacterial physiology. However, whether adhesin-adhesin interactions, as well as the type of adhesin mediating aggregation, affect bacterial cell physiology is unknown. By sequencing the transcriptomes of aggregated and nonaggregated cells expressing native or synthetic adhesins, we characterized the effects of aggregation and adhesin type on E. coli physiology.
Collapse
Affiliation(s)
- Yankel Chekli
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris, France
| | - Rebecca J. Stevick
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris, France
| | - Etienne Kornobis
- Hub de Bioinformatique et Biostatistique-Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, Paris, France
- Plate-forme Technologique Biomics—Centre de Ressources et Recherches Technologiques, Institut Pasteur, Paris, France
| | - Valérie Briolat
- Hub de Bioinformatique et Biostatistique-Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, Paris, France
- Plate-forme Technologique Biomics—Centre de Ressources et Recherches Technologiques, Institut Pasteur, Paris, France
| | - Jean-Marc Ghigo
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris, France
| | - Christophe Beloin
- Institut Pasteur, Université Paris Cité, CNRS UMR 6047, Genetics of Biofilms Laboratory, Paris, France
| |
Collapse
|
16
|
Alonso Villela SM, Kraïem-Ghezal H, Bouhaouala-Zahar B, Bideaux C, Aceves Lara CA, Fillaudeau L. Production of recombinant scorpion antivenoms in E. coli: current state and perspectives. Appl Microbiol Biotechnol 2023:10.1007/s00253-023-12578-1. [PMID: 37199752 DOI: 10.1007/s00253-023-12578-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/05/2023] [Accepted: 05/09/2023] [Indexed: 05/19/2023]
Abstract
Scorpion envenomation is a serious health problem in tropical and subtropical zones. The access to scorpion antivenom is sometimes limited in availability and specificity. The classical production process is cumbersome, from the hyper-immunization of the horses to the IgG digestion and purification of the F(ab)'2 antibody fragments. The production of recombinant antibody fragments in Escherichia coli is a popular trend due to the ability of this microbial host to produce correctly folded proteins. Small recombinant antibody fragments, such as single-chain variable fragments (scFv) and nanobodies (VHH), have been constructed to recognize and neutralize the neurotoxins responsible for the envenomation symptoms in humans. They are the focus of interest of the most recent studies and are proposed as potentially new generation of pharmaceuticals for their use in immunotherapy against scorpion stings of the Buthidae family. This literature review comprises the current status on the scorpion antivenom market and the analyses of cross-reactivity of commercial scorpion anti-serum against non-specific scorpion venoms. Recent studies on the production of new recombinant scFv and nanobodies will be presented, with a focus on the Androctonus and Centruroides scorpion species. Protein engineering-based technology could be the key to obtaining the next generation of therapeutics capable of neutralizing and cross-reacting against several types of scorpion venoms. KEY POINTS: • Commercial antivenoms consist of predominantly purified equine F(ab)'2fragments. • Nanobody-based antivenom can neutralize Androctonus venoms and have a low immunogenicity. • Affinity maturation and directed evolution are used to obtain potent scFv families against Centruroides scorpions.
Collapse
Affiliation(s)
| | - Hazar Kraïem-Ghezal
- Laboratoire Des Venins Et Molécules Thérapeutiques, Institut Pasteur de Tunis, Université de Tunis El Manar, 13 Place Pasteur BP74, 1002, Tunis, Tunisia
| | - Balkiss Bouhaouala-Zahar
- Laboratoire Des Venins Et Molécules Thérapeutiques, Institut Pasteur de Tunis, Université de Tunis El Manar, 13 Place Pasteur BP74, 1002, Tunis, Tunisia.
- Faculté de Médecine de Tunis, Université de Tunis El Manar, Tunis, Tunisia.
| | - Carine Bideaux
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| | | | - Luc Fillaudeau
- TBI, Université de Toulouse, CNRS, INRAE, INSA, Toulouse, France
| |
Collapse
|
17
|
Lei M, Trivedi VD, Nair NU, Lee K, Van Deventer JA. Flow cytometric evaluation of yeast-bacterial cell-cell interactions. Biotechnol Bioeng 2023; 120:399-408. [PMID: 36259110 PMCID: PMC10072783 DOI: 10.1002/bit.28253] [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: 06/10/2022] [Revised: 09/23/2022] [Accepted: 10/09/2022] [Indexed: 01/13/2023]
Abstract
Synthetic cell-cell interaction systems can be useful for understanding multicellular communities or for screening binding molecules. We adapt a previously characterized set of synthetic cognate nanobody-antigen pairs to a yeast-bacteria coincubation format and use flow cytometry to evaluate cell-cell interactions mediated by binding between surface-displayed molecules. We further use fluorescence-activated cell sorting to enrich a specific yeast-displayed nanobody within a mixed yeast-display population. Finally, we demonstrate that this system supports the characterization of a therapeutically relevant nanobody-antigen interaction: a previously discovered nanobody that binds to the intimin protein expressed on the surface of enterohemorrhagic Escherichia coli. Overall, our findings indicate that the yeast-bacteria format supports efficient evaluation of ligand-target interactions. With further development, this format may facilitate systematic characterization and high-throughput discovery of bacterial surface-binding molecules.
Collapse
Affiliation(s)
- Ming Lei
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155
| | - Vikas D. Trivedi
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155
| | - Nikhil U. Nair
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155
| | - Kyongbum Lee
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155
| | - James A. Van Deventer
- Department of Chemical and Biological Engineering, Tufts University, Medford, Massachusetts 02155
- Department of Biomedical Engineering, Tufts University, Medford, Massachusetts 02155
| |
Collapse
|
18
|
Robledo M, Álvarez B, Cuevas A, González S, Ruano-Gallego D, Fernández L, de la Cruz F. Targeted bacterial conjugation mediated by synthetic cell-to-cell adhesions. Nucleic Acids Res 2022; 50:12938-12950. [PMID: 36511856 PMCID: PMC9825185 DOI: 10.1093/nar/gkac1164] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 12/15/2022] Open
Abstract
Genetic interventions on microbiomes, for clinical or biotechnological purposes, remain challenging. Conjugation-based delivery of genetic cargo is still unspecific and limited by low conjugation rates. Here we report an approach to overcome these problems, based on a synthetic bacterial adhesion system. Mating assemblers consist on a synthetic adhesion formed by the expression on the surface of donor and target cells of specific nanobodies (Nb) and their cognate antigen (Ag). The Nb-Ag bridge increased 1-3 logs transfer of a variety of plasmids, especially in liquid media, confirming that cell-cell docking is a main determinant limiting mating efficiency. Synthetic cell-to-cell adhesion allows efficient conjugation to targeted recipients, enhancing delivery of desired genes to a predefined subset of prey species, or even specific pathogenic strains such as enterohemorrhagic Escherichia coli (EHEC), within a bacterial community. The synthetic conjugation enhancer presented here optimizes plasmid delivery by selecting the target hosts with high selectivity.
Collapse
Affiliation(s)
- Marta Robledo
- Correspondence may also be addressed to Marta Robledo.
| | - Beatriz Álvarez
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Campus UAM Cantoblanco, 28049 Madrid, Spain
| | - Ana Cuevas
- Intergenomics Group, Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria, 39011, Santander, Spain
| | - Sheila González
- Intergenomics Group, Instituto de Biomedicina y Biotecnología de Cantabria (IBBTEC), CSIC-Universidad de Cantabria, 39011, Santander, Spain
| | - David Ruano-Gallego
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Campus UAM Cantoblanco, 28049 Madrid, Spain
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Campus UAM Cantoblanco, 28049 Madrid, Spain
| | | |
Collapse
|
19
|
A de novo matrix for macroscopic living materials from bacteria. Nat Commun 2022; 13:5544. [PMID: 36130968 PMCID: PMC9492681 DOI: 10.1038/s41467-022-33191-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 09/08/2022] [Indexed: 11/08/2022] Open
Abstract
Engineered living materials (ELMs) embed living cells in a biopolymer matrix to create materials with tailored functions. While bottom-up assembly of macroscopic ELMs with a de novo matrix would offer the greatest control over material properties, we lack the ability to genetically encode a protein matrix that leads to collective self-organization. Here we report growth of ELMs from Caulobacter crescentus cells that display and secrete a self-interacting protein. This protein formed a de novo matrix and assembled cells into centimeter-scale ELMs. Discovery of design and assembly principles allowed us to tune the composition, mechanical properties, and catalytic function of these ELMs. This work provides genetic tools, design and assembly rules, and a platform for growing ELMs with control over both matrix and cellular structure and function. Engineered living materials (ELMs) embed living cells in a biopolymer matrix to create novel materials with tailored functions. In this work, the authors engineered bacteria to grow novel macroscopic materials that can be reshaped, functionalized, and used to filter contaminated water while also showing that the stiffness of these materials can be tuned through genetic changes.
Collapse
|
20
|
Robinson CM, Short NE, Riglar DT. Achieving spatially precise diagnosis and therapy in the mammalian gut using synthetic microbial gene circuits. Front Bioeng Biotechnol 2022; 10:959441. [PMID: 36118573 PMCID: PMC9478464 DOI: 10.3389/fbioe.2022.959441] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
The mammalian gut and its microbiome form a temporally dynamic and spatially heterogeneous environment. The inaccessibility of the gut and the spatially restricted nature of many gut diseases translate into difficulties in diagnosis and therapy for which novel tools are needed. Engineered bacterial whole-cell biosensors and therapeutics have shown early promise at addressing these challenges. Natural and engineered sensing systems can be repurposed in synthetic genetic circuits to detect spatially specific biomarkers during health and disease. Heat, light, and magnetic signals can also activate gene circuit function with externally directed spatial precision. The resulting engineered bacteria can report on conditions in situ within the complex gut environment or produce biotherapeutics that specifically target host or microbiome activity. Here, we review the current approaches to engineering spatial precision for in vivo bacterial diagnostics and therapeutics using synthetic circuits, and the challenges and opportunities this technology presents.
Collapse
Affiliation(s)
| | | | - David T. Riglar
- Section of Structural and Synthetic Biology, Department of Infectious Disease, Imperial College London, London, United Kingdom
| |
Collapse
|
21
|
Álvarez B, Muñoz-Abad V, Asensio-Calavia A, Fernández LÁ. Enhanced protein translocation to mammalian cells by expression of EtgA transglycosylase in a synthetic injector E. coli strain. Microb Cell Fact 2022; 21:133. [PMID: 35780105 PMCID: PMC9250224 DOI: 10.1186/s12934-022-01860-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Accepted: 06/23/2022] [Indexed: 12/29/2022] Open
Abstract
Background Bacterial type III secretion systems (T3SSs) assemble a multiprotein complex termed the injectisome, which acts as a molecular syringe for translocation of specific effector proteins into the cytoplasm of host cells. The use of injectisomes for delivery of therapeutic proteins into mammalian cells is attractive for biomedical applications. With that aim, we previously generated a non-pathogenic Escherichia coli strain, called Synthetic Injector E. coli (SIEC), which assembles functional injectisomes from enteropathogenic E. coli (EPEC). The assembly of injectisomes in EPEC is assisted by the lytic transglycosylase EtgA, which degrades the peptidoglycan layer. As SIEC lacks EtgA, we investigated whether expression of this transglycosylase enhances the protein translocation capacity of the engineered bacterium. Results The etgA gene from EPEC was integrated into the SIEC chromosome under the control of the inducible tac promoter, generating the strain SIEC-eEtgA. The controlled expression of EtgA had no effect on the growth or viability of bacteria. Upon induction, injectisome assembly was ~ 30% greater in SIEC-eEtgA than in the parental strain, as determined by the level of T3SS translocon proteins, the hemolytic activity of the bacterial strain, and the impairment in flagellar motility. The functionality of SIEC-eEtgA injectisomes was evaluated in a derivative strain carrying a synthetic operon (eLEE5), which was capable of delivering Tir effector protein into the cytoplasm of HeLa cells triggering F-actin polymerization beneath the attached bacterium. Lastly, using β-lactamase as a reporter of T3SS-protein injection, we determined that the protein translocation capacity was ~ 65% higher in the SIEC-EtgA strain than in the parental SIEC strain. Conclusions We demonstrate that EtgA enhances the assembly of functional injectisomes in a synthetic injector E. coli strain, enabling the translocation of greater amounts of proteins into the cytoplasm of mammalian cells. Accordingly, EtgA expression may boost the protein translocation of SIEC strains programmed as living biotherapeutics.
Collapse
Affiliation(s)
- Beatriz Álvarez
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, Campus Cantoblanco, 28049, Madrid, Spain
| | - Víctor Muñoz-Abad
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, Campus Cantoblanco, 28049, Madrid, Spain.,Programa de Doctorado en Biociencias Moleculares, Universidad Autónoma de Madrid (UAM), Campus Cantoblanco, 28049, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" (Consejo Superior de Investigaciones Científicas - Universidad Autónoma de Madrid), Nicolas Cabrera 1, Campus Cantoblanco, 28049, Madrid, Spain
| | - Alejandro Asensio-Calavia
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, Campus Cantoblanco, 28049, Madrid, Spain.,Programa de Doctorado en Biociencias Moleculares, Universidad Autónoma de Madrid (UAM), Campus Cantoblanco, 28049, Madrid, Spain
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Darwin 3, Campus Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
22
|
Mahdavi SZB, Oroojalian F, Eyvazi S, Hejazi M, Baradaran B, Pouladi N, Tohidkia MR, Mokhtarzadeh A, Muyldermans S. An overview on display systems (phage, bacterial, and yeast display) for production of anticancer antibodies; advantages and disadvantages. Int J Biol Macromol 2022; 208:421-442. [PMID: 35339499 DOI: 10.1016/j.ijbiomac.2022.03.113] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/12/2021] [Accepted: 03/17/2022] [Indexed: 11/05/2022]
Abstract
Antibodies as ideal therapeutic and diagnostic molecules are among the top-selling drugs providing considerable efficacy in disease treatment, especially in cancer therapy. Limitations of the hybridoma technology as routine antibody generation method in conjunction with numerous developments in molecular biology led to the development of alternative approaches for the streamlined identification of most effective antibodies. In this regard, display selection technologies such as phage display, bacterial display, and yeast display have been widely promoted over the past three decades as ideal alternatives to traditional methods. The display of antibodies on phages is probably the most widespread of these methods, although surface display on bacteria or yeast have been employed successfully, as well. These methods using various sizes of combinatorial antibody libraries and different selection strategies possessing benefits in screening potency, generating, and isolation of high affinity antibodies with low risk of immunogenicity. Knowing the basics of each method assists in the design and retrieval process of antibodies suitable for different diseases, including cancer. In this review, we aim to outline the basics of each library construction and its display method, screening and selection steps. The advantages and disadvantages in comparison to alternative methods, and their applications in antibody engineering will be explained. Finally, we will review approved or non-approved therapeutic antibodies developed by employing these methods, which may serve as therapeutic antibodies in cancer therapy.
Collapse
Affiliation(s)
| | - Fatemeh Oroojalian
- Department of Advanced Sciences and Technologies in Medicine, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Shirin Eyvazi
- Department of Biology, Tabriz Branch, Islamic Azad University, Tabriz, Iran; Biotechnology Research Center, Tabriz Branch, Islamic Azad University, Tabriz, Iran
| | - Maryam Hejazi
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nasser Pouladi
- Department of Biology, Faculty of Basic Sciences, Azarbaijan Shahid Madani University, Tabriz, Iran
| | - Mohammad Reza Tohidkia
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Serge Muyldermans
- Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, Dalian, China..
| |
Collapse
|
23
|
Casasnovas JM, Margolles Y, Noriega MA, Guzmán M, Arranz R, Melero R, Casanova M, Corbera JA, Jiménez-de-Oya N, Gastaminza P, Garaigorta U, Saiz JC, Martín-Acebes MÁ, Fernández LÁ. Nanobodies Protecting From Lethal SARS-CoV-2 Infection Target Receptor Binding Epitopes Preserved in Virus Variants Other Than Omicron. Front Immunol 2022; 13:863831. [PMID: 35547740 PMCID: PMC9082315 DOI: 10.3389/fimmu.2022.863831] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/28/2022] [Indexed: 11/28/2022] Open
Abstract
The emergence of SARS-CoV-2 variants that escape from immune neutralization are challenging vaccines and antibodies developed to stop the COVID-19 pandemic. Thus, it is important to establish therapeutics directed toward multiple or specific SARS-CoV-2 variants. The envelope spike (S) glycoprotein of SARS-CoV-2 is the key target of neutralizing antibodies (Abs). We selected a panel of nine nanobodies (Nbs) from dromedary camels immunized with the receptor-binding domain (RBD) of the S, and engineered Nb fusions as humanized heavy chain Abs (hcAbs). Nbs and derived hcAbs bound with subnanomolar or picomolar affinities to the S and its RBD, and S-binding cross-competition clustered them in two different groups. Most of the hcAbs hindered RBD binding to its human ACE2 (hACE2) receptor, blocked cell entry of viruses pseudotyped with the S protein and neutralized SARS-CoV-2 infection in cell cultures. Four potent neutralizing hcAbs prevented the progression to lethal SARS-CoV-2 infection in hACE2-transgenic mice, demonstrating their therapeutic potential. Cryo-electron microscopy identified Nb binding epitopes in and out the receptor binding motif (RBM), and showed different ways to prevent virus binding to its cell entry receptor. The Nb binding modes were consistent with its recognition of SARS-CoV-2 RBD variants; mono and bispecific hcAbs efficiently bound all variants of concern except omicron, which emphasized the immune escape capacity of this latest variant.
Collapse
Affiliation(s)
- José M Casasnovas
- Departments of Macromolecule Structure, Microbial Biotechnology, and Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Yago Margolles
- Departments of Macromolecule Structure, Microbial Biotechnology, and Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - María A Noriega
- Departments of Macromolecule Structure, Microbial Biotechnology, and Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - María Guzmán
- Departments of Macromolecule Structure, Microbial Biotechnology, and Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Rocío Arranz
- Departments of Macromolecule Structure, Microbial Biotechnology, and Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Roberto Melero
- Departments of Macromolecule Structure, Microbial Biotechnology, and Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Mercedes Casanova
- Departments of Macromolecule Structure, Microbial Biotechnology, and Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Juan Alberto Corbera
- Instituto Universitario de Investigaciones Biomédicas y Sanitarias (IUIBS), Facultad de Veterinaria, Universidad de Las Palmas de Gran Canaria (ULPGC), Campus Universitario de Arucas, Arucas, Spain
| | - Nereida Jiménez-de-Oya
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA, CSIC), Madrid, Spain
| | - Pablo Gastaminza
- Departments of Macromolecule Structure, Microbial Biotechnology, and Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Urtzi Garaigorta
- Departments of Macromolecule Structure, Microbial Biotechnology, and Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| | - Juan Carlos Saiz
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA, CSIC), Madrid, Spain
| | - Miguel Ángel Martín-Acebes
- Department of Biotechnology, Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Consejo Superior de Investigaciones Científicas (INIA, CSIC), Madrid, Spain
| | - Luis Ángel Fernández
- Departments of Macromolecule Structure, Microbial Biotechnology, and Cellular and Molecular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid, Spain
| |
Collapse
|
24
|
Martí M, Merwaiss F, Butković A, Daròs JA. Production of Potyvirus-Derived Nanoparticles Decorated with a Nanobody in Biofactory Plants. Front Bioeng Biotechnol 2022; 10:877363. [PMID: 35433643 PMCID: PMC9008781 DOI: 10.3389/fbioe.2022.877363] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Accepted: 03/14/2022] [Indexed: 01/10/2023] Open
Abstract
Viral nanoparticles (VNPs) have recently attracted attention for their use as building blocks for novel materials to support a range of functions of potential interest in nanotechnology and medicine. Viral capsids are ideal for presenting small epitopes by inserting them at an appropriate site on the selected coat protein (CP). VNPs presenting antibodies on their surfaces are considered highly promising tools for therapeutic and diagnostic purposes. Due to their size, nanobodies are an interesting alternative to classic antibodies for surface presentation. Nanobodies are the variable domains of heavy-chain (VHH) antibodies from animals belonging to the family Camelidae, which have several properties that make them attractive therapeutic molecules, such as their small size, simple structure, and high affinity and specificity. In this work, we have produced genetically encoded VNPs derived from two different potyviruses—the largest group of RNA viruses that infect plants—decorated with nanobodies. We have created a VNP derived from zucchini yellow mosaic virus (ZYMV) decorated with a nanobody against the green fluorescent protein (GFP) in zucchini (Cucurbita pepo) plants. As reported for other viruses, the expression of ZYMV-derived VNPs decorated with this nanobody was only made possible by including a picornavirus 2A splicing peptide between the fused proteins, which resulted in a mixed population of unmodified and decorated CPs. We have also produced tobacco etch virus (TEV)-derived VNPs in Nicotiana benthamiana plants decorated with the same nanobody against GFP. Strikingly, in this case, VNPs could be assembled by direct fusion of the nanobody to the viral CP with no 2A splicing involved, likely resulting in fully decorated VNPs. For both expression systems, correct assembly and purification of the recombinant VNPs was confirmed by transmission electron microscope; the functionality of the CP-fused nanobody was assessed by western blot and binding assays. In sum, here we report the production of genetically encoded plant-derived VNPs decorated with a nanobody. This system may be an attractive alternative for the sustainable production in plants of nanobody-containing nanomaterials for diagnostic and therapeutic purposes.
Collapse
|
25
|
Lim B, Yin Y, Ye H, Cui Z, Papachristodoulou A, Huang WE. Reprogramming Synthetic Cells for Targeted Cancer Therapy. ACS Synth Biol 2022; 11:1349-1360. [PMID: 35255684 PMCID: PMC9084601 DOI: 10.1021/acssynbio.1c00631] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
![]()
Advances
in synthetic biology enable the reprogramming of bacteria
as smart agents to specifically target tumors and locally release
anticancer drugs in a highly controlled manner. However, the bench-to-bedside
translation of engineered bacteria is often impeded by genetic instability
and the potential risk of uncontrollable replication of engineered
bacteria inside the patient. SimCells (simple cells) are chromosome-free
bacteria controlled by designed gene circuits, which can bypass the
interference of the native gene network in bacteria and eliminate
the risk of bacterial uncontrolled growth. Here, we describe the reprogramming
of SimCells and mini-SimCells to serve as “safe and live drugs”
for targeted cancer therapy. We engineer SimCells to display nanobodies
on the surface for the binding of carcinoembryonic antigen (CEA),
which is an important biomarker found commonly in colorectal cancer
cells. We show that SimCells and mini-SimCells with surface display
of anti-CEA nanobody can specifically bind CEA-expressing Caco2 cancer
cells in vitro while leaving the non-CEA-expressing
SW80 cancer cells untouched. These cancer-targeting SimCells and mini-SimCells
induced cancer cell death in vitro by compromising
the plasma membrane of cancer cells. The cancer-killing effect can
be further enhanced by an aspirin/salicylate inducible gene circuit
that converts salicylate into catechol, a potent anticancer. This
work highlights the potential of SimCells and mini-SimCells for targeted
cancer therapy and lays the foundation for the application of synthetic
biology to medicine.
Collapse
Affiliation(s)
- Boon Lim
- Department of Engineering Science, University of Oxford, Parks Road, OX1 3PJ Oxford, U.K
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, U.K
| | - Yutong Yin
- Department of Engineering Science, University of Oxford, Parks Road, OX1 3PJ Oxford, U.K
| | - Hua Ye
- Department of Engineering Science, University of Oxford, Parks Road, OX1 3PJ Oxford, U.K
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, U.K
| | - Zhanfeng Cui
- Department of Engineering Science, University of Oxford, Parks Road, OX1 3PJ Oxford, U.K
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford, OX3 7DQ Oxford, U.K
| | | | - Wei E. Huang
- Department of Engineering Science, University of Oxford, Parks Road, OX1 3PJ Oxford, U.K
| |
Collapse
|
26
|
Liu B, Yang D. Easily Established and Multifunctional Synthetic Nanobody Libraries as Research Tools. Int J Mol Sci 2022; 23:ijms23031482. [PMID: 35163405 PMCID: PMC8835997 DOI: 10.3390/ijms23031482] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 01/19/2022] [Accepted: 01/25/2022] [Indexed: 02/06/2023] Open
Abstract
Nanobodies, or VHHs, refer to the antigen-binding domain of heavy-chain antibodies (HCAbs) from camelids. They have been widely used as research tools for protein purification and structure determination due to their small size, high specificity, and high stability, overcoming limitations with conventional antibody fragments. However, animal immunization and subsequent retrieval of antigen-specific nanobodies are expensive and complicated. Construction of synthetic nanobody libraries using DNA oligonucleotides is a cost-effective alternative for immunization libraries and shows great potential in identifying antigen-specific or even conformation-specific nanobodies. This review summarizes and analyses synthetic nanobody libraries in the current literature, including library design and biopanning methods, and further discusses applications of antigen-specific nanobodies obtained from synthetic libraries to research.
Collapse
|
27
|
Al-Ramahi Y, Nyerges A, Margolles Y, Cerdán L, Ferenc G, Pál C, Fernández LÁ, de Lorenzo V. ssDNA recombineering boosts in vivo evolution of nanobodies displayed on bacterial surfaces. Commun Biol 2021; 4:1169. [PMID: 34621006 PMCID: PMC8497518 DOI: 10.1038/s42003-021-02702-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 09/21/2021] [Indexed: 12/11/2022] Open
Abstract
ssDNA recombineering has been exploited to hyperdiversify genomically-encoded nanobodies displayed on the surface of Escherichia coli for originating new binding properties. As a proof-of-principle a nanobody recognizing the antigen TirM from enterohaemorrhagic E. coli (EHEC) was evolved towards the otherwise not recognized TirM antigen from enteropathogenic E. coli (EPEC). To this end, E. coli cells displaying this nanobody fused to the intimin outer membrane-bound domain were subjected to multiple rounds of mutagenic oligonucleotide recombineering targeting the complementarity determining regions (CDRs) of the cognate VHH gene sequence. Binders to the EPEC-TirM were selected upon immunomagnetic capture of bacteria bearing active variants and nanobodies identified with a new ability to strongly bind the new antigen. The results highlight the power of combining evolutionary properties of bacteria in vivo with oligonucleotide synthesis in vitro for the sake of focusing diversification to specific segments of a gene (or protein thereof) of interest.
Collapse
Affiliation(s)
- Yamal Al-Ramahi
- Systems and Synthetic Biology Department, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, 28049, Spain
| | - Akos Nyerges
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, H-6726, Hungary
- Department of Genetics, Harvard Medical School, Boston, MA, 02115, USA
| | - Yago Margolles
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, 28049, Spain
| | - Lidia Cerdán
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, 28049, Spain
| | - Gyorgyi Ferenc
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, H-6726, Hungary
| | - Csaba Pál
- Synthetic and Systems Biology Unit, Institute of Biochemistry, Biological Research Centre, Szeged, H-6726, Hungary
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, 28049, Spain.
| | - Víctor de Lorenzo
- Systems and Synthetic Biology Department, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, Madrid, 28049, Spain.
| |
Collapse
|
28
|
Phan TH, Kuijl C, Huynh DT, Jong WSP, Luirink J, van Ulsen P. Overproducing the BAM complex improves secretion of difficult-to-secrete recombinant autotransporter chimeras. Microb Cell Fact 2021; 20:176. [PMID: 34488755 PMCID: PMC8419823 DOI: 10.1186/s12934-021-01668-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 08/26/2021] [Indexed: 11/14/2022] Open
Abstract
Monomeric autotransporters have been used extensively to transport recombinant proteins or protein domains to the cell surface of Gram-negative bacteria amongst others for antigen display. Genetic fusion of such antigens into autotransporters has yielded chimeras that can be used for vaccination purposes. However, not every fusion construct is transported efficiently across the cell envelope. Problems occur in particular when the fused antigen attains a relatively complex structure in the periplasm, prior to its translocation across the outer membrane. The latter step requires the interaction with periplasmic chaperones and the BAM (β-barrel assembly machinery) complex in the outer membrane. This complex catalyzes insertion and folding of β-barrel outer membrane proteins, including the β-barrel domain of autotransporters. Here, we investigated whether the availability of periplasmic chaperones or the BAM complex is a limiting factor for the surface localization of difficult-to-secrete chimeric autotransporter constructs. Indeed, we found that overproduction of in particular the BAM complex, increases surface display of difficult-to-secrete chimeras. Importantly, this beneficial effect appeared to be generic not only for a number of monomeric autotransporter fusions but also for fusions to trimeric autotransporters. Therefore, overproduction of BAM might be an attractive strategy to improve the production of recombinant autotransporter constructs.
Collapse
Affiliation(s)
- Trang H Phan
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Coen Kuijl
- Medical Microbiology and Infection Control, Amsterdam Institute of Infection & Immunity, Amsterdam UMC, Amsterdam, The Netherlands
| | - Dung T Huynh
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - Joen Luirink
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.,Abera Bioscience AB, Solna, Sweden
| | - Peter van Ulsen
- Department of Molecular Microbiology, Amsterdam Institute of Molecular and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
29
|
Zakri AM, Al-Doss AA, Ali AA, Samara EM, Ahmed BS, Al-Saleh MA, Idris AM, Abdalla OA, Sack M. Generation and Characterization of Nanobodies Against Tomato Leaf Curl Sudan Virus. PLANT DISEASE 2021; 105:2410-2417. [PMID: 33599515 DOI: 10.1094/pdis-11-20-2407-re] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Begomoviruses infect food, fiber, and vegetable crop plants, including tomato, potato, bean, cotton, cucumber, and pumpkin, and damage many economically important crop plants worldwide. Tomato leaf curl Sudan virus (ToLCSDV) is the most widespread tomato-infecting begomovirus in Saudi Arabia. Using phage display technology, this study isolated two camel-derived nanobodies against purified ToLCSDV virions from a library of antigen-binding fragments (VHH or nanobody) of heavy-chain antibodies built from an immunized camel. The isolated nanobodies also cross-reacted with purified tomato yellow leaf curl virus virions and showed significant enzyme-linked immunosorbent assay reactivity with extracts from plants with typical begomovirus infection symptoms. The results can pave the way to developing diagnostics for begomovirus detection, design, and characterization of novel nanomaterials based on virus-like particles, in addition to nanobody-mediated begomovirus resistance in economically important crops, such as tomato, potato, and cucumber.
Collapse
Affiliation(s)
- Adel M Zakri
- Department of Plant Production, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah A Al-Doss
- Department of Plant Production, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | - Ahmed A Ali
- Department of Plant Production, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | - Emad M Samara
- Department of Animal Production, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | - Basem S Ahmed
- Department of Plant Production, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | - Mohammed A Al-Saleh
- Department of Plant Protection, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | - Ali M Idris
- School of Plant Sciences, University of Arizona, Tucson, AZ 85721, U.S.A
| | - Omar A Abdalla
- Department of Plant Protection, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia
| | | |
Collapse
|
30
|
Fraile S, Briones M, Revenga-Parra M, de Lorenzo V, Lorenzo E, Martínez-García E. Engineering Tropism of Pseudomonas putida toward Target Surfaces through Ectopic Display of Recombinant Nanobodies. ACS Synth Biol 2021; 10:2049-2059. [PMID: 34337948 PMCID: PMC8397431 DOI: 10.1021/acssynbio.1c00227] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Indexed: 12/15/2022]
Abstract
Gram-negative bacteria are endowed with complex outer membrane (OM) structures that allow them to both interact with other organisms and attach to different physical structures. However, the design of reliable bacterial coatings of solid surfaces is still a considerable challenge. In this work, we report that ectopic expression of a fibrinogen-specific nanobody on the envelope of Pseudomonas putida cells enables controllable formation of a bacterial monolayer strongly bound to an antigen-coated support. To this end, either the wild type or a surface-naked derivative of P. putida was engineered to express a hybrid between the β-barrel of an intimin-type autotransporter inserted in the outer membrane and a nanobody (VHH) moiety that targets fibrinogen as its cognate interaction partner. The functionality of the thereby presented VHH and the strength of the resulting cell attachment to a solid surface covered with the cognate antigen were tested and parametrized with Quartz Crystal Microbalance technology. The results not only demonstrated the value of using bacteria with reduced OM complexity for efficient display of artificial adhesins, but also the potential of this approach to engineer specific bacterial coverings of predetermined target surfaces.
Collapse
Affiliation(s)
- Sofía Fraile
- Systems Biology Department, Centro Nacional
de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - María Briones
- Departamento de Química Analítica y Análisis
Instrumental, Universidad Autónoma
de Madrid, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Mónica Revenga-Parra
- Departamento de Química Analítica y Análisis
Instrumental, Universidad Autónoma
de Madrid, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Víctor de Lorenzo
- Systems Biology Department, Centro Nacional
de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Encarnación Lorenzo
- Departamento de Química Analítica y Análisis
Instrumental, Universidad Autónoma
de Madrid, Campus de Cantoblanco, 28049 Madrid, Spain
| | - Esteban Martínez-García
- Systems Biology Department, Centro Nacional
de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
31
|
Abstract
Pathogen attachment to host tissue is critical in the progress of many infections. Bacteria use adhesion in vivo to stabilize colonization and subsequently regulate the deployment of contact-dependent virulence traits. To specifically target host cells, they decorate themselves with adhesins, proteins that bind to mammalian cell surface receptors. One common assumption is that adhesin-receptor interactions entirely govern bacterial attachment. However, how adhesins engage with their receptors in an in vivo-like context remains unclear, in particular under the influence of a heterogeneous mechanical microenvironment. We here investigate the biophysical processes governing bacterial adhesion to host cells using a tunable adhesin-receptor system. By dynamically visualizing attachment, we found that bacterial adhesion to host cell surface, unlike adhesion to inert surfaces, involves two consecutive steps. Bacteria initially attach to their host without engaging adhesins. This step lasts about 1 min, during which bacteria can easily detach. We found that at this stage, the glycocalyx, a layer of glycosylated proteins and lipids, shields the host cell by keeping adhesins away from their receptor ligand. In a second step, adhesins engage with their target receptors to strengthen attachment for minutes to hours. The active properties of the membrane, endowed by the actin cytoskeleton, strengthen specific adhesion. Altogether, our results demonstrate that adhesin-ligand binding is not the sole regulator of bacterial adhesion. In fact, the host cell’s surface mechanical microenvironment mediates the physical interactions between host and bacteria, thereby playing an essential role in the onset of infection.
Collapse
|
32
|
Birnbaum DP, Manjula‐Basavanna A, Kan A, Tardy BL, Joshi NS. Hybrid Living Capsules Autonomously Produced by Engineered Bacteria. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2004699. [PMID: 34141524 PMCID: PMC8188213 DOI: 10.1002/advs.202004699] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/10/2021] [Indexed: 06/12/2023]
Abstract
Bacterial cellulose (BC) has excellent material properties and can be produced sustainably through simple bacterial culture, but BC-producing bacteria lack the extensive genetic toolkits of model organisms such as Escherichia coli (E. coli). Here, a simple approach is reported for producing highly programmable BC materials through incorporation of engineered E. coli. The acetic acid bacterium Gluconacetobacter hansenii is cocultured with engineered E. coli in droplets of glucose-rich media to produce robust cellulose capsules, which are then colonized by the E. coli upon transfer to selective lysogeny broth media. It is shown that the encapsulated E. coli can produce engineered protein nanofibers within the cellulose matrix, yielding hybrid capsules capable of sequestering specific biomolecules from the environment and enzymatic catalysis. Furthermore, capsules are produced which can alter their own bulk physical properties through enzyme-induced biomineralization. This novel system uses a simple fabrication process, based on the autonomous activity of two bacteria, to significantly expand the functionality of BC-based living materials.
Collapse
Affiliation(s)
- Daniel P. Birnbaum
- John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02138USA
- Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMA02115USA
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Avinash Manjula‐Basavanna
- Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMA02115USA
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Anton Kan
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| | - Blaise L. Tardy
- Department of Bioproducts and BiosystemsAalto UniversityEspooFI‐00076Finland
| | - Neel S. Joshi
- Department of Chemistry and Chemical BiologyNortheastern UniversityBostonMA02115USA
- Wyss Institute for Biologically Inspired EngineeringHarvard UniversityBostonMA02115USA
| |
Collapse
|
33
|
Valenzuela Nieto G, Jara R, Watterson D, Modhiran N, Amarilla AA, Himelreichs J, Khromykh AA, Salinas-Rebolledo C, Pinto T, Cheuquemilla Y, Margolles Y, López González Del Rey N, Miranda-Chacon Z, Cuevas A, Berking A, Deride C, González-Moraga S, Mancilla H, Maturana D, Langer A, Toledo JP, Müller A, Uberti B, Krall P, Ehrenfeld P, Blesa J, Chana-Cuevas P, Rehren G, Schwefel D, Fernandez LÁ, Rojas-Fernandez A. Potent neutralization of clinical isolates of SARS-CoV-2 D614 and G614 variants by a monomeric, sub-nanomolar affinity nanobody. Sci Rep 2021; 11:3318. [PMID: 33558635 PMCID: PMC7870875 DOI: 10.1038/s41598-021-82833-w] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 01/25/2021] [Indexed: 02/06/2023] Open
Abstract
Despite unprecedented global efforts to rapidly develop SARS-CoV-2 treatments, in order to reduce the burden placed on health systems, the situation remains critical. Effective diagnosis, treatment, and prophylactic measures are urgently required to meet global demand: recombinant antibodies fulfill these requirements and have marked clinical potential. Here, we describe the fast-tracked development of an alpaca Nanobody specific for the receptor-binding-domain (RBD) of the SARS-CoV-2 Spike protein with potential therapeutic applicability. We present a rapid method for nanobody isolation that includes an optimized immunization regimen coupled with VHH library E. coli surface display, which allows single-step selection of Nanobodies using a simple density gradient centrifugation of the bacterial library. The selected single and monomeric Nanobody, W25, binds to the SARS-CoV-2 S RBD with sub-nanomolar affinity and efficiently competes with ACE-2 receptor binding. Furthermore, W25 potently neutralizes SARS-CoV-2 wild type and the D614G variant with IC50 values in the nanomolar range, demonstrating its potential as antiviral agent.
Collapse
Affiliation(s)
| | - Ronald Jara
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Institute of Biochemistry and Microbiology, Faculty of Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Daniel Watterson
- School of Chemistry and Molecular Bioscience, The University of Queensland, Brisbane, Australia
- The Australian Institute for Biotechnology and Nanotechnology, The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| | - Naphak Modhiran
- School of Chemistry and Molecular Bioscience, The University of Queensland, Brisbane, Australia
- The Australian Institute for Biotechnology and Nanotechnology, The University of Queensland, Brisbane, Australia
| | - Alberto A Amarilla
- School of Chemistry and Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Johanna Himelreichs
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Alexander A Khromykh
- School of Chemistry and Molecular Bioscience, The University of Queensland, Brisbane, Australia
- Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane, Australia
| | | | - Teresa Pinto
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Yorka Cheuquemilla
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Berking Biotechnology, Valdivia, Chile
| | - Yago Margolles
- Department of Microbial Biotechnology, National Biotechnology Center, Superior Council of Scientific Research, Madrid, Spain
| | | | - Zaray Miranda-Chacon
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Alexei Cuevas
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | | | - Camila Deride
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Institute of Veterinary Clinical Sciences, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | | | - Héctor Mancilla
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Daniel Maturana
- NanoTemper Technologies GmbH, Floessergasse 4, 81369, Munich, Germany
| | - Andreas Langer
- NanoTemper Technologies GmbH, Floessergasse 4, 81369, Munich, Germany
| | - Juan Pablo Toledo
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
| | - Ananda Müller
- Ross University School of Veterinary Medicine, Basseterre, Saint Kitts and Nevis
- Institute of Veterinary Clinical Sciences, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Benjamín Uberti
- Institute of Veterinary Clinical Sciences, Faculty of Veterinary Sciences, Universidad Austral de Chile, Valdivia, Chile
| | - Paola Krall
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Department of Pediatrics and Children's Surgery Oriente, Universidad de Chile, Valdivia, Chile
| | - Pamela Ehrenfeld
- Institute of Anatomy, Histology, and Pathology, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile
- Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile
| | - Javier Blesa
- HM CINAC, Hospital Universitario HM Puerta del Sur, Mostoles, 28938, Madrid, Spain
| | - Pedro Chana-Cuevas
- CETRAM & Faculty of Medical Science, Universidad de Santiago de Chile, Santiago, Chile
| | - German Rehren
- Technology Transfer and Licensing Office, Universidad Austral de Chile, Valdivia, Chile
| | - David Schwefel
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin Institute of Health, Berlin, Germany
| | - Luis Ángel Fernandez
- Department of Microbial Biotechnology, National Biotechnology Center, Superior Council of Scientific Research, Madrid, Spain
| | - Alejandro Rojas-Fernandez
- Institute of Medicine, Faculty of Medicine, Universidad Austral de Chile, Valdivia, Chile.
- Berking Biotechnology, Valdivia, Chile.
- Center for Interdisciplinary Studies on the Nervous System, CISNE, Universidad Austral de Chile, Valdivia, Chile.
- Institute of Philosophy and Complexity Sciences, Santiago, Chile.
| |
Collapse
|
34
|
Effect of temperature on the production of a recombinant antivenom in fed-batch mode. Appl Microbiol Biotechnol 2021; 105:1017-1030. [PMID: 33443635 DOI: 10.1007/s00253-021-11093-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 12/19/2020] [Accepted: 01/03/2021] [Indexed: 10/22/2022]
Abstract
In the pharmaceutical industry, nanobodies show promising properties for its application in serotherapy targeting the highly diffusible scorpion toxins. The production of recombinant nanobodies in Escherichia coli has been widely studied in shake flask cultures in rich medium. However, there are no upstream bioprocess studies of nanobody production in defined minimal medium and the effect of the induction temperature on the production kinetics. In this work, the effect of the temperature during the expression of the chimeric bispecific nanobody CH10-12 form, showing high scorpion antivenom potential, was studied in bioreactor cultures of E. coli. High biomass concentrations (25 g cdw/L) were achieved in fed-batch mode, and the expression of the CH10-12 nanobody was induced at temperatures 28, 29, 30, 33, and 37°C with a constant glucose feed. For the bispecific form NbF12-10, the induction was performed at 29°C. Biomass and carbon dioxide yields were reported for each culture phase, and the maintenance coefficient was obtained for each strain. Nanobody production in the CH10-12 strain was higher at low temperatures (lower than 30°C) and declined with the increase of the temperature. At 29°C, the CH10-12, NbF12-10, and WK6 strains were compared. Strains CH10-12 and NbF12-10 had a productivity of 0.052 and 0.021 mg/L/h of nanobody, respectively, after 13 h of induction. The specific productivity of the nanobodies was modeled as a function of the induction temperature and the specific growth rates. Experimental results confirm that low temperatures increase the productivity of the nanobody.Key points• Nanobodies with scorpion antivenom activity produced using two recombinant strains.• Nanobodies production was achieved in fed-batch cultures at different induction temperatures.• Low induction temperatures result in high volumetric productivities of the nanobody CH10-12.
Collapse
|
35
|
Abstract
Ever since the discovery of antibodies, they have been generated by complicated multi-step procedures. Typically, these involve sequencing, cloning, and screening after expression of the antibodies in a suitable organism and format. Here, a staphylococcal nanobody display is described that omits many the abovementioned intermediate steps and allows for simultaneous screening of multiple targets without prior knowledge nor expression of the binders. This paper reports a detailed, general step-by-step protocol to achieve nanobodies of high affinity. Apart from its focus on radioactive and fluorescent targets, it gives options for various other target formats and additional applications for the staphylococcal library; including flow cytometry and immunoprecipitation. This provides a system for antibody engineers that can be easily adopted to their specific needs.
Collapse
Affiliation(s)
- Marco Cavallari
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
36
|
Yu S, Xiong G, Zhao S, Tang Y, Tang H, Wang K, Liu H, Lan K, Bi X, Duan S. Nanobodies targeting immune checkpoint molecules for tumor immunotherapy and immunoimaging (Review). Int J Mol Med 2020; 47:444-454. [PMID: 33416134 PMCID: PMC7797440 DOI: 10.3892/ijmm.2020.4817] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 10/29/2020] [Indexed: 12/21/2022] Open
Abstract
The immune checkpoint blockade is an effective strategy to enhance the anti-tumor T cell effector activity, thus becoming one of the most promising immunotherapeutic strategies in the history of cancer treatment. Several immune checkpoint inhibitor have been approved by the FDA, such as anti-CTLA-4, anti-PD-1, anti-PD-L1 monoclonal antibodies. Most tumor patients benefitted from these antibodies, but some of the patients did not respond to them. To increase the effectiveness of immunotherapy, including immune checkpoint blockade therapies, miniaturization of antibodies has been introduced. A single-domain antibody, also known as nanobody, is an attractive reagent for immunotherapy and immunoimaging thanks to its unique structural characteristic consisting of a variable region of a single heavy chain antibody. This structure confers to the nanobody a light molecular weight, making it smaller than conventional antibodies, although remaining able to bind to a specific antigen. Therefore, this review summarizes the production of nanobodies targeting immune checkpoint molecules and the application of nanobodies targeting immune checkpoint molecules in immunotherapy and immunoimaging.
Collapse
Affiliation(s)
- Sheng Yu
- Department of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Gui Xiong
- Department of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Shimei Zhao
- Department of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Yanbo Tang
- Department of Gastroenterology, The First Affiliated Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region 545001, P.R. China
| | - Hua Tang
- Department of Clinical Laboratory, The Second Clinical Medical College of Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region 545006, P.R. China
| | - Kaili Wang
- Department of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Hongjing Liu
- Department of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Ke Lan
- Department of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region 545005, P.R. China
| | - Xiongjie Bi
- Department of Clinical Laboratory, The First Affiliated Hospital of Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region 545001, P.R. China
| | - Siliang Duan
- Department of Medicine, Guangxi University of Science and Technology, Liuzhou, Guangxi Zhuang Autonomous Region 545005, P.R. China
| |
Collapse
|
37
|
Dvořák P, Bayer EA, de Lorenzo V. Surface Display of Designer Protein Scaffolds on Genome-Reduced Strains of Pseudomonas putida. ACS Synth Biol 2020; 9:2749-2764. [PMID: 32877604 DOI: 10.1021/acssynbio.0c00276] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The bacterium Pseudomonas putida KT2440 is gaining considerable interest as a microbial platform for biotechnological valorization of polymeric organic materials, such as lignocellulosic residues or plastics. However, P. putida on its own cannot make much use of such complex substrates, mainly because it lacks an efficient extracellular depolymerizing apparatus. We seek to address this limitation by adopting a recombinant cellulosome strategy for this host. In this work, we report an essential step in this endeavor-a display of designer enzyme-anchoring protein "scaffoldins", encompassing cohesin binding domains from divergent cellulolytic bacterial species on the P. putida surface. Two P. putida chassis strains, EM42 and EM371, with streamlined genomes and differences in the composition of the outer membrane were employed in this study. Scaffoldin variants were optimally delivered to their surface with one of four tested autotransporter systems (Ag43 from Escherichia coli), and the efficient display was confirmed by extracellular attachment of chimeric β-glucosidase and fluorescent proteins. Our results not only highlight the value of cell surface engineering for presentation of recombinant proteins on the envelope of Gram-negative bacteria but also pave the way toward designer cellulosome strategies tailored for P. putida.
Collapse
Affiliation(s)
- Pavel Dvořák
- Department of Experimental Biology (Section of Microbiology), Faculty of Science, Masaryk University, Kamenice 753/5, 62500 Brno, Czech Republic
| | - Edward A Bayer
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot 76100, Israel
| | - Víctor de Lorenzo
- Systems and Synthetic Biology Program, Centro Nacional de Biotecnología CNB-CSIC, Cantoblanco, Darwin 3, 28049 Madrid, Spain
| |
Collapse
|
38
|
Chekli Y, Peron-Cane C, Dell'Arciprete D, Allemand JF, Li C, Ghigo JM, Gautier A, Lebreton A, Desprat N, Beloin C. Visualizing the dynamics of exported bacterial proteins with the chemogenetic fluorescent reporter FAST. Sci Rep 2020; 10:15791. [PMID: 32978420 PMCID: PMC7519654 DOI: 10.1038/s41598-020-72498-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/27/2020] [Indexed: 01/09/2023] Open
Abstract
Bacterial proteins exported to the cell surface play key cellular functions. However, despite the interest to study the localisation of surface proteins such as adhesins, transporters or hydrolases, monitoring their dynamics in live imaging remains challenging, due to the limited availability of fluorescent probes remaining functional after secretion. In this work, we used the Escherichia coli intimin and the Listeria monocytogenes InlB invasin as surface exposed scaffolds fused with the recently developed chemogenetic fluorescent reporter protein FAST. Using both membrane permeant (HBR-3,5DM) and non-permeant (HBRAA-3E) fluorogens that fluoresce upon binding to FAST, we demonstrated that fully functional FAST can be exposed at the cell surface and used to specifically tag the external side of the bacterial envelop in both diderm and monoderm bacteria. Our work opens new avenues to study the organization and dynamics of the bacterial cell surface proteins.
Collapse
Affiliation(s)
- Yankel Chekli
- Genetics of Biofilms Laboratory, Institut Pasteur, UMR CNRS2001, 75015, Paris, France
- Université de Paris, Sorbonne Paris Cité, Paris, France
| | - Caroline Peron-Cane
- Laboratoire de Physique de L'ENS, École Normale Supérieure, Université PSL, CNRS, Sorbonne Université, Université de Paris, 75005, Paris, France
- Institut de Biologie de I'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Dario Dell'Arciprete
- Laboratoire de Physique de L'ENS, École Normale Supérieure, Université PSL, CNRS, Sorbonne Université, Université de Paris, 75005, Paris, France
| | - Jean-François Allemand
- Laboratoire de Physique de L'ENS, École Normale Supérieure, Université PSL, CNRS, Sorbonne Université, Université de Paris, 75005, Paris, France
- Institut de Biologie de I'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
| | - Chenge Li
- École Normale Supérieure, Université PSL, CNRS, Laboratoire Des Biomolécules (LBM), Sorbonne Université, 75005, Paris, France
| | - Jean-Marc Ghigo
- Genetics of Biofilms Laboratory, Institut Pasteur, UMR CNRS2001, 75015, Paris, France
| | - Arnaud Gautier
- École Normale Supérieure, Université PSL, CNRS, Laboratoire Des Biomolécules (LBM), Sorbonne Université, 75005, Paris, France
- PASTEUR, Department of Chemistry, École Normale Supérieure, PSL University, Sorbonne Université, CNRS, 75005, Paris, France
- Institut Universitaire de France, Paris, France
| | - Alice Lebreton
- Institut de Biologie de I'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France
- INRAE, IBENS, 75005, Paris, France
| | - Nicolas Desprat
- Laboratoire de Physique de L'ENS, École Normale Supérieure, Université PSL, CNRS, Sorbonne Université, Université de Paris, 75005, Paris, France.
- Institut de Biologie de I'ENS (IBENS), École Normale Supérieure, CNRS, INSERM, Université PSL, 75005, Paris, France.
| | - Christophe Beloin
- Genetics of Biofilms Laboratory, Institut Pasteur, UMR CNRS2001, 75015, Paris, France.
| |
Collapse
|
39
|
Martínez-García E, Fraile S, Rodríguez Espeso D, Vecchietti D, Bertoni G, de Lorenzo V. Naked Bacterium: Emerging Properties of a Surfome-Streamlined Pseudomonas putida Strain. ACS Synth Biol 2020; 9:2477-2492. [PMID: 32786355 DOI: 10.1021/acssynbio.0c00272] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Environmental bacteria are most often endowed with native surface-attachment programs that frequently conflict with efforts to engineer biofilms and synthetic communities with given tridimensional architectures. In this work, we report the editing of the genome of Pseudomonas putida KT2440 for stripping the cells of most outer-facing structures of the bacterial envelope that mediate motion, binding to surfaces, and biofilm formation. To this end, 23 segments of the P. putida chromosome encoding a suite of such functions were deleted, resulting in the surface-naked strain EM371, the physical properties of which changed dramatically in respect to the wild type counterpart. As a consequence, surface-edited P. putida cells were unable to form biofilms on solid supports and, because of the swimming deficiency and other alterations, showed a much faster sedimentation in liquid media. Surface-naked bacteria were then used as carriers of interacting partners (e.g., Jun-Fos domains) ectopically expressed by means of an autotransporter display system on the now easily accessible cell envelope. Abstraction of individual bacteria as adhesin-coated spherocylinders enabled rigorous quantitative description of the multicell interplay brought about by thereby engineered physical interactions. The model was then applied to parametrize the data extracted from automated analysis of confocal microscopy images of the experimentally assembled bacterial flocks for analyzing their structure and distribution. The resulting data not only corroborated the value of P. putida EM371 over the parental strain as a platform for display artificial adhesins but also provided a strategy for rational engineering of catalytic communities.
Collapse
Affiliation(s)
- Esteban Martínez-García
- Systems Biology Program, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Sofía Fraile
- Systems Biology Program, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - David Rodríguez Espeso
- Systems Biology Program, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| | - Davide Vecchietti
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Giovanni Bertoni
- Department of Biosciences, Università degli Studi di Milano, 20133 Milan, Italy
| | - Víctor de Lorenzo
- Systems Biology Program, Centro Nacional de Biotecnología (CNB-CSIC), Campus de Cantoblanco, 28049 Madrid, Spain
| |
Collapse
|
40
|
Muyldermans S. A guide to: generation and design of nanobodies. FEBS J 2020; 288:2084-2102. [PMID: 32780549 PMCID: PMC8048825 DOI: 10.1111/febs.15515] [Citation(s) in RCA: 175] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/03/2020] [Accepted: 08/07/2020] [Indexed: 01/09/2023]
Abstract
A nanobody (Nb) is a registered trademark of Ablynx, referring to the single antigen-binding domain of heavy chain-only antibodies (HCAbs) that are circulating in Camelidae. Nbs are produced recombinantly in micro-organisms and employed as research tools or for diagnostic and therapeutic applications. They were - and still are - also named single-domain antibodies (sdAbs) or variable domain of the heavy chain of HCAbs (VHH). A variety of methods are currently in use for the fast and efficient generation of target-specific Nbs. Such Nbs are produced at low cost and associate with high affinity to their cognate antigen. They are robust, strictly monomeric and easy to tailor into more complex entities to meet the requirements of their application. Here, we review the various sources and different strategies that have been developed to identify rapidly, target-specific Nbs. We further discuss a variety of engineering technologies that have been explored to broaden the application range of Nbs and summarise those applications where designed Nbs might offer a marked advantage over other affinity reagents.
Collapse
Affiliation(s)
- Serge Muyldermans
- Cellular and Molecular Immunology, Vrije Universiteit Brussel, Belgium.,Liaoning Key Laboratory of Molecular Recognition and Imaging, School of Bioengineering, Dalian University of Technology, China
| |
Collapse
|
41
|
Riangrungroj P, Bever CS, Hammock BD, Polizzi KM. A label-free optical whole-cell Escherichia coli biosensor for the detection of pyrethroid insecticide exposure. Sci Rep 2019; 9:12466. [PMID: 31462650 PMCID: PMC6713742 DOI: 10.1038/s41598-019-48907-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 08/13/2019] [Indexed: 11/15/2022] Open
Abstract
There is a growing need for low-cost, portable technologies for the detection of threats to the environment and human health. Here we propose a label-free, optical whole-cell Escherichia coli biosensor for the detection of 3-phenoxybenzoic acid (3-PBA), a biomarker for monitoring human exposure to synthetic pyrethroid insecticides. The biosensor functions like a competitive ELISA but uses whole-cells surface displaying an anti-3-PBA VHH as the detection element. When the engineered cells are mixed with 3-PBA-protein conjugate crosslinking that can be visually detected occurs. Free 3-PBA in samples competes with these crosslinks, leading to a detectable change in the output. The assay performance was improved by coloring the cells via expression of the purple-blue amilCP chromoprotein and the VHH expression level was reduced to obtain a limit of detection of 3 ng/mL. The optimized biosensor exhibited robust function in complex sample backgrounds such as synthetic urine and plasma. Furthermore, lyophilization enabled storage of biosensor cells for at least 90 days without loss of functionality. Our whole-cell biosensor is simple and low-cost and therefore has potential to be further developed as a screening tool for monitoring exposure to pyrethroids in low-resource environments.
Collapse
Affiliation(s)
- Pinpunya Riangrungroj
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
- Imperial College Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK
| | - Candace Spier Bever
- Foodborne Toxin Detection and Prevention Unit, Western Regional Research Center, Agricultural Research Service, United States Department of Agriculture, Albany, CA, 94710, USA
| | - Bruce D Hammock
- Department of Entomology and UCD Cancer Center, University of California, Davis, California, 95616, United States
| | - Karen M Polizzi
- Imperial College Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK.
- Department of Chemical Engineering, Imperial College London, London, SW7 2AZ, UK.
| |
Collapse
|
42
|
Riangrungroj P, Polizzi KM. BeQuIK (Biosensor Engineered Quorum Induced Killing): designer bacteria for destroying recalcitrant biofilms. Microb Biotechnol 2019; 13:311-314. [PMID: 31328393 PMCID: PMC7017806 DOI: 10.1111/1751-7915.13465] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 07/05/2019] [Accepted: 07/05/2019] [Indexed: 12/31/2022] Open
Affiliation(s)
- Pinpunya Riangrungroj
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK.,Imperial College Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK
| | - Karen M Polizzi
- Imperial College Centre for Synthetic Biology, Imperial College London, London, SW7 2AZ, UK.,Department of Chemical Engineering, Imperial College London, London, SW7 2AZ, UK
| |
Collapse
|
43
|
Kylilis N, Riangrungroj P, Lai HE, Salema V, Fernández LÁ, Stan GBV, Freemont PS, Polizzi KM. Whole-Cell Biosensor with Tunable Limit of Detection Enables Low-Cost Agglutination Assays for Medical Diagnostic Applications. ACS Sens 2019; 4:370-378. [PMID: 30623662 DOI: 10.1021/acssensors.8b01163] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Whole-cell biosensors can form the basis of affordable, easy-to-use diagnostic tests that can be readily deployed for point-of-care (POC) testing, but to date the detection of analytes such as proteins that cannot easily diffuse across the cell membrane has been challenging. Here we developed a novel biosensing platform based on cell agglutination using an E. coli whole-cell biosensor surface-displaying nanobodies which bind selectively to a target protein analyte. As a proof-of-concept, we show the feasibility of this design to detect a model analyte at nanomolar concentrations. Moreover, we show that the design architecture is flexible by building assays optimized to detect a range of model analyte concentrations using straightforward design rules and a mathematical model. Finally, we re-engineer our whole-cell biosensor for the detection of a medically relevant biomarker by the display of two different nanobodies against human fibrinogen and demonstrate a detection limit as low as 10 pM in diluted human plasma. Overall, we demonstrate that our agglutination technology fulfills the requirement of POC testing by combining low-cost nanobody production, customizable detection range and low detection limits. This technology has the potential to produce affordable diagnostics for field-testing in the developing world, emergency or disaster relief sites, as well as routine medical testing and personalized medicine.
Collapse
Affiliation(s)
- Nicolas Kylilis
- Imperial College Centre for Synthetic Biology, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Pinpunya Riangrungroj
- Imperial College Centre for Synthetic Biology, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Life Sciences, Imperial College London, London SW7 2AZ, United Kingdom
| | - Hung-En Lai
- Imperial College Centre for Synthetic Biology, Imperial College London, London SW7 2AZ, United Kingdom
- Section of Structural Biology, Department of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| | - Valencio Salema
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Cantoblanco UAM, 28049 Madrid, Spain
| | - Luis Ángel Fernández
- Department of Microbial Biotechnology, Centro Nacional de Biotecnología (CNB-CSIC), Darwin 3, Campus Cantoblanco UAM, 28049 Madrid, Spain
| | - Guy-Bart V. Stan
- Imperial College Centre for Synthetic Biology, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Bioengineering, Imperial College London, London SW7 2AZ, United Kingdom
| | - Paul S. Freemont
- Imperial College Centre for Synthetic Biology, Imperial College London, London SW7 2AZ, United Kingdom
- Section of Structural Biology, Department of Medicine, Imperial College London, London SW7 2AZ, United Kingdom
| | - Karen M. Polizzi
- Imperial College Centre for Synthetic Biology, Imperial College London, London SW7 2AZ, United Kingdom
- Department of Chemical Engineering, Imperial College London, London SW7 2AZ, United Kingdom
| |
Collapse
|
44
|
Timmis K, Timmis JK, Brüssow H, Fernández LÁ. Synthetic consortia of nanobody-coupled and formatted bacteria for prophylaxis and therapy interventions targeting microbiome dysbiosis-associated diseases and co-morbidities. Microb Biotechnol 2019; 12:58-65. [PMID: 30575298 PMCID: PMC6302794 DOI: 10.1111/1751-7915.13355] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Designed nanobody-linked synthetic consortia for microbiota dysbiosis therapies. A. Nanobodies (Nb) are selected for specific antigens on target bacteria destined for a synthetic therapy consortium that may consist of two (B) or multiple (C) members. For the treatment of dysbiosis co-morbidities requiring two functionally distinct consortia, these may be linked through a common member to generate a single bi-functional microbiota therapy (D).
Collapse
Affiliation(s)
- Kenneth Timmis
- Institute of MicrobiologyTechnical University BraunschweigBraunschweigGermany
| | | | - Harald Brüssow
- Division of Animal and Human Health EngineeringDepartment of BiosystemsKatholieke Universiteit LeuvenLeuvenBelgium
| | - Luis Ángel Fernández
- Department of Microbial BiotechnologyCentro Nacional de BiotecnologíaConsejo Superior de Investigaciones CientíficasMadridSpain
| |
Collapse
|
45
|
Andersson KG, Persson J, Ståhl S, Löfblom J. Autotransporter-Mediated Display of a Naïve Affibody Library on the Outer Membrane of Escherichia coli. Biotechnol J 2018; 14:e1800359. [PMID: 30179307 DOI: 10.1002/biot.201800359] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 08/23/2018] [Indexed: 12/14/2022]
Abstract
Development of new affinity proteins using combinatorial protein engineering is today established for generation of monoclonal antibodies and also essential for discovery of binders that are based on non-immunoglobulin proteins. Phage display is most frequently used, but yeast display is becoming increasingly popular, partly due to the option of utilizing fluorescence-activated cell sorting (FACS) for isolation of new candidates. Escherichia coli has several valuable properties for library applications and in particular the high transformation efficiency. The use of various autotransporters and intimins for secretion and anchoring on the outer membrane have shown promising results and particularly for directed evolution of different enzymes. Here, the authors report on display of a large naïve affibody library on the outer membrane of E. coli using the autotransporter Adhesin Involved in Diffuse Adherence (AIDA-I). The expression cassette is first engineered by removing non-essential sequences, followed by introduction of an affibody library, comprising more than 109 variants, into the new display vector. The quality of the library and general performance of the method is assessed by FACS against five different targets, which resulted in a panel of binders with down to nanomolar affinities, suggesting that the method has potential as a complement to phage display for generation of affibody molecules.
Collapse
Affiliation(s)
- Ken G Andersson
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - Jonas Persson
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - Stefan Ståhl
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, SE-106 91, Stockholm, Sweden
| |
Collapse
|
46
|
van Ulsen P, Zinner KM, Jong WSP, Luirink J. On display: autotransporter secretion and application. FEMS Microbiol Lett 2018; 365:5061625. [DOI: 10.1093/femsle/fny165] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 07/27/2018] [Indexed: 12/12/2022] Open
Affiliation(s)
- Peter van Ulsen
- Section Molecular Microbiology, Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | - Katinka M Zinner
- Section Molecular Microbiology, Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
| | | | - Joen Luirink
- Section Molecular Microbiology, Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit, 1081 HV Amsterdam, The Netherlands
- Abera Bioscience AB, SE-111 45 Stockholm, Sweden
| |
Collapse
|
47
|
A Synthetic Bacterial Cell-Cell Adhesion Toolbox for Programming Multicellular Morphologies and Patterns. Cell 2018; 174:649-658.e16. [DOI: 10.1016/j.cell.2018.06.041] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2018] [Revised: 05/18/2018] [Accepted: 06/22/2018] [Indexed: 01/08/2023]
|
48
|
Chen X, Yu X, Song X, Liu L, Yi Y, Yao W, Gao X. Selection, purification, and characterization of a HER2-targeting soluble designed ankyrin repeat protein by E. coli surface display using HER2-positive melanoma cells. Prep Biochem Biotechnol 2018; 48:144-150. [PMID: 29313422 DOI: 10.1080/10826068.2017.1407944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Human epidermal growth factor receptor 2 (HER2) is a powerful target for cancer immune therapy. The development of anti-HER2 monoclonal antibodies targeting different domains of HER2 is quite effective. However, the selection and production of multivalent antibodies are complicated. In this study, a mimivirus-based designed ankyrin repeat protein (DARPin) targeting HER2 was selected from an artificial library by bacteria surface display. The selection was performed on HER2-positive B16BL6/E2 melanoma cells and HER2-nagative cells. DARPin selected from the library could be expressed in soluble form with a yield of 70 mg/L. After purified by two continuous and easy steps, the purity of DARPin was 90% as established by SDS-PAGE and RP-HPLC. Selected DARPin showed significant HER2-targeting ability with an affinity of 1.05 ± 0.47 µM. MTT assay demonstrated that at the concentration of 640 nM, the selected DARPin dimer could inhibit the SK-BR-3 growth at a rate of 36.63 and 46.34% in 48 and 72 hr incubation separately, which was similar to trastuzumab (43.12 and 49.14% separately). These findings suggested that it was an effective method to select antibody mimetic DARPin by bacteria surface display combined with live cells sorting and provided a drug candidate for cancer therapy.
Collapse
Affiliation(s)
- Xiaofei Chen
- a Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines , School of Life Science and Technology, China Pharmaceutical University , Nanjing , PR China
| | - Xiaoxiao Yu
- a Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines , School of Life Science and Technology, China Pharmaceutical University , Nanjing , PR China
| | - Xiaoda Song
- a Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines , School of Life Science and Technology, China Pharmaceutical University , Nanjing , PR China
| | - Li Liu
- a Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines , School of Life Science and Technology, China Pharmaceutical University , Nanjing , PR China
| | - Yuting Yi
- a Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines , School of Life Science and Technology, China Pharmaceutical University , Nanjing , PR China
| | - Wenbing Yao
- a Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines , School of Life Science and Technology, China Pharmaceutical University , Nanjing , PR China
| | - Xiangdong Gao
- a Jiangsu Key Laboratory of Druggability of Biopharmaceuticals, State Key Laboratory of Natural Medicines , School of Life Science and Technology, China Pharmaceutical University , Nanjing , PR China
| |
Collapse
|
49
|
Jong WSP, Schillemans M, ten Hagen-Jongman CM, Luirink J, van Ulsen P. Comparing autotransporter β-domain configurations for their capacity to secrete heterologous proteins to the cell surface. PLoS One 2018; 13:e0191622. [PMID: 29415042 PMCID: PMC5802855 DOI: 10.1371/journal.pone.0191622] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Accepted: 01/08/2018] [Indexed: 01/11/2023] Open
Abstract
Monomeric autotransporters have been extensively used for export of recombinant proteins to the cell surface of Gram-negative bacteria. A bottleneck in the biosynthesis of such constructs is the passage of the outer membrane, which is facilitated by the β-domain at the C terminus of an autotransporter in conjunction with the Bam complex in the outer membrane. We have evaluated eight β-domain constructs for their capacity to secrete fused proteins to the cell surface. These constructs derive from the monomeric autotransporters Hbp, IgA protease, Ag43 and EstA and the trimeric autotransporter Hia, which all were selected because they have been previously used for secretion of recombinant proteins. We fused three different protein domains to the eight β-domain constructs, being a Myc-tag, the Hbp passenger and a nanobody or VHH domain, and assessed expression, membrane insertion and surface exposure. Our results show that expression levels differed considerably between the constructs tested. The constructs that included the β-domains of Hbp and IgA protease appeared the most efficient and resulted in expression levels that were detectable on Coomassie-stained SDS-PAGE gels. The VHH domain appeared the most difficult fusion partner to export, probably due to its complex immunoglobulin-like structure with a tertiary structure stabilized by an intramolecular disulfide bond. Overall, the Hbp β-domain compared favorably in exporting the fused recombinant proteins, because it showed in every instance tested a good level of expression, stable membrane insertion and clear surface exposure.
Collapse
Affiliation(s)
- Wouter S. P. Jong
- Section Molecular Microbiology, Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Abera Bioscience AB, Stockholm, Sweden
- * E-mail: ;
| | | | - Corinne M. ten Hagen-Jongman
- Section Molecular Microbiology, Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Joen Luirink
- Section Molecular Microbiology, Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- Abera Bioscience AB, Stockholm, Sweden
| | - Peter van Ulsen
- Section Molecular Microbiology, Department of Molecular Cell Biology, Amsterdam Institute for Molecules, Medicines and Systems (AIMMS), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- * E-mail: ;
| |
Collapse
|
50
|
Keeble AH, Banerjee A, Ferla MP, Reddington SC, Anuar INAK, Howarth M. Evolving Accelerated Amidation by SpyTag/SpyCatcher to Analyze Membrane Dynamics. Angew Chem Int Ed Engl 2017; 56:16521-16525. [PMID: 29024296 PMCID: PMC5814910 DOI: 10.1002/anie.201707623] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 09/15/2017] [Indexed: 12/19/2022]
Abstract
SpyTag is a peptide that forms a spontaneous amide bond with its protein partner SpyCatcher. This protein superglue is a broadly useful tool for molecular assembly, locking together biological building blocks efficiently and irreversibly in diverse architectures. We initially developed SpyTag and SpyCatcher by rational design, through splitting a domain from a Gram-positive bacterial adhesin. In this work, we established a phage-display platform to select for specific amidation, leading to an order of magnitude acceleration for interaction of the SpyTag002 variant with the SpyCatcher002 variant. We show that the 002 pair bonds rapidly under a wide range of conditions and at either protein terminus. SpyCatcher002 was fused to an intimin derived from enterohemorrhagic Escherichia coli. SpyTag002 reaction enabled specific and covalent decoration of intimin for live cell fluorescent imaging of the dynamics of the bacterial outer membrane as cells divide.
Collapse
Affiliation(s)
- Anthony H. Keeble
- Department of BiochemistryUniversity of OxfordSouth Parks RoadOxfordOX1 3QUUK
| | - Anusuya Banerjee
- Department of BiochemistryUniversity of OxfordSouth Parks RoadOxfordOX1 3QUUK
| | - Matteo P. Ferla
- Department of BiochemistryUniversity of OxfordSouth Parks RoadOxfordOX1 3QUUK
| | | | | | - Mark Howarth
- Department of BiochemistryUniversity of OxfordSouth Parks RoadOxfordOX1 3QUUK
| |
Collapse
|