1
|
Nakamura S, Tanimura Y, Nomura R, Suzuki H, Nishikawa K, Kamegawa A, Numoto N, Tanaka A, Kawabata S, Sakaguchi S, Emi A, Suzuki Y, Fujiyoshi Y. Structure-guided engineering of a mutation-tolerant inhibitor peptide against variable SARS-CoV-2 spikes. Proc Natl Acad Sci U S A 2025; 122:e2413465122. [PMID: 39854234 PMCID: PMC11789008 DOI: 10.1073/pnas.2413465122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/26/2024] [Indexed: 01/26/2025] Open
Abstract
Pathogen mutations present an inevitable and challenging problem for therapeutics and the development of mutation-tolerant anti-infective drugs to strengthen global health and combat evolving pathogens is urgently needed. While spike proteins on viral surfaces are attractive targets for preventing viral entry, they mutate frequently, making it difficult to develop effective therapeutics. Here, we used a structure-guided strategy to engineer an inhibitor peptide against the SARS-CoV-2 spike, called CeSPIACE, with mutation-tolerant and potent binding ability against all variants to enhance affinity for the invariant architecture of the receptor-binding domain (RBD). High-resolution structures of the peptide complexed with mutant RBDs revealed a mechanism of mutation-tolerant inhibition. CeSPIACE bound major mutant RBDs with picomolar affinity and inhibited infection by SARS-CoV-2 variants in VeroE6/TMPRSS2 cells (IC50 4 pM to 13 nM) and demonstrated potent in vivo efficacy by inhalation administration in hamsters. Mutagenesis analyses to address mutation risks confirmed tolerance against existing and/or potential future mutations of the RBD. Our strategy of engineering mutation-tolerant inhibitors may be applicable to other infectious diseases.
Collapse
Affiliation(s)
- Shun Nakamura
- Cellular and Structural Physiology Laboratory, Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
- CeSPIA Inc., Taisei Otemachi, Chiyoda-ku, Tokyo100-0004, Japan
| | - Yukihiro Tanimura
- Cellular and Structural Physiology Laboratory, Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
| | - Risa Nomura
- Cellular and Structural Physiology Laboratory, Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
| | - Hiroshi Suzuki
- Cellular and Structural Physiology Laboratory, Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
| | - Kouki Nishikawa
- CeSPIA Inc., Taisei Otemachi, Chiyoda-ku, Tokyo100-0004, Japan
| | - Akiko Kamegawa
- Cellular and Structural Physiology Laboratory, Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
- CeSPIA Inc., Taisei Otemachi, Chiyoda-ku, Tokyo100-0004, Japan
| | - Nobutaka Numoto
- International Center for Structural Biology, Research Institute for Interdisciplinary Science, Okayama University, Kita-ku, Okayama700-8530, Japan
| | - Atsushi Tanaka
- Division of Research Animal Laboratory and Translational Medicine, Research and Development Center, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka569-8686, Japan
| | - Shigeru Kawabata
- Department of Pathology, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka569-8686, Japan
| | - Shoichi Sakaguchi
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka569-8686, Japan
| | - Akino Emi
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka569-8686, Japan
| | - Youichi Suzuki
- Department of Microbiology and Infection Control, Faculty of Medicine, Osaka Medical and Pharmaceutical University, Takatsuki, Osaka569-8686, Japan
| | - Yoshinori Fujiyoshi
- Cellular and Structural Physiology Laboratory, Advanced Research Initiative, Institute of Integrated Research, Institute of Science Tokyo, Bunkyo-ku, Tokyo113-8510, Japan
- CeSPIA Inc., Taisei Otemachi, Chiyoda-ku, Tokyo100-0004, Japan
| |
Collapse
|
2
|
Hao S, Su J. Whispering gallery mode optical resonators for biological and chemical detection: current practices, future perspectives, and challenges. REPORTS ON PROGRESS IN PHYSICS. PHYSICAL SOCIETY (GREAT BRITAIN) 2024; 88:016402. [PMID: 39626318 DOI: 10.1088/1361-6633/ad99e7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 12/03/2024] [Indexed: 12/24/2024]
Abstract
Sensors are important for a wide variety of applications include medical diagnostics and environmental monitoring. Due to their long photon confinement times, whispering gallery mode (WGM) sensors are among the most sensitive sensors currently in existence. We briefly discuss what are WGM sensors, the principles of WGM sensing, and the history of the field, beginning with Mie theory. We discuss recent work in the field on using these WGM resonators as sensors, focusing particularly on biological and chemical sensing applications. We discuss how sensorgrams are acquired and fundamental measurement limits. In addition, we discuss how to interpret binding curves and extract physical parameters such as binding affinity constants. We discuss the controversy surrounding single-molecule detection and discuss hybrid WGM nanoparticle sensors. In addition, we place these sensors in context with others sensing technologies both labeled and label-free. Finally, we discuss what we believe are the most promising applications for these devices, outline remaining challenges, and provide an outlook for the future.
Collapse
Affiliation(s)
- Shuang Hao
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, United States of America
| | - Judith Su
- Wyant College of Optical Sciences, The University of Arizona, Tucson, AZ, United States of America
- Department of Biomedical Engineering, The University of Arizona, Tucson, AZ, United States of America
| |
Collapse
|
3
|
Mori S, Shionyu M, Shimamoto K, Nomura K. Bacterial Glycolipid Acting on Protein Transport Across Membranes. Chembiochem 2024; 25:e202300808. [PMID: 38400776 DOI: 10.1002/cbic.202300808] [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: 11/30/2023] [Revised: 01/11/2024] [Accepted: 02/22/2024] [Indexed: 02/26/2024]
Abstract
The process of protein transport across membranes involves a variety of factors and has been extensively investigated. Traditionally, proteinaceous translocons and chaperones have been recognized as crucial factors in this process. However, recent studies have highlighted the significant roles played by lipids and a glycolipid present in biological membranes in membrane protein transport. Membrane lipids can influence transport efficiency by altering the physicochemical properties of membranes. Notably, our studies have revealed that diacylglycerol (DAG) attenuates mobility in the membrane core region, leading to a dramatic suppression of membrane protein integration. Conversely, a glycolipid in Escherichia coli inner membranes, named membrane protein integrase (MPIase), enhances integration not only through the alteration of membrane properties but also via direct interactions with membrane proteins. This review explores the mechanisms of membrane protein integration mediated by membrane lipids, specifically DAG, and MPIase. Our results, along with the employed physicochemical analysis methods such as fluorescence measurements, nuclear magnetic resonance, surface plasmon resonance, and docking simulation, are presented to elucidate these mechanisms.
Collapse
Affiliation(s)
- Shoko Mori
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto, 619-0284, Japan
| | - Masafumi Shionyu
- Department of Frontier Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura-cho, Nagahama, Shiga, 526-0829, Japan
| | - Keiko Shimamoto
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto, 619-0284, Japan
- Department of Chemistry Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka, 560-0043, Japan
| | - Kaoru Nomura
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto, 619-0284, Japan
| |
Collapse
|
4
|
Huang YK, Cheng WC, Kuo TT, Yang JC, Wu YC, Wu HH, Lo CC, Hsieh CY, Wong SC, Lu CH, Wu WL, Liu SJ, Li YC, Lin CC, Shen CN, Hung MC, Lin JT, Yeh CC, Sher YP. Inhibition of ADAM9 promotes the selective degradation of KRAS and sensitizes pancreatic cancers to chemotherapy. NATURE CANCER 2024; 5:400-419. [PMID: 38267627 DOI: 10.1038/s43018-023-00720-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Accepted: 12/19/2023] [Indexed: 01/26/2024]
Abstract
Kirsten rat sarcoma virus (KRAS) signaling drives pancreatic ductal adenocarcinoma (PDAC) malignancy, which is an unmet clinical need. Here, we identify a disintegrin and metalloproteinase domain (ADAM)9 as a modulator of PDAC progression via stabilization of wild-type and mutant KRAS proteins. Mechanistically, ADAM9 loss increases the interaction of KRAS with plasminogen activator inhibitor 1 (PAI-1), which functions as a selective autophagy receptor in conjunction with light chain 3 (LC3), triggering lysosomal degradation of KRAS. Suppression of ADAM9 by a small-molecule inhibitor restricts disease progression in spontaneous models, and combination with gemcitabine elicits dramatic regression of patient-derived tumors. Our findings provide a promising strategy to target the KRAS signaling cascade and demonstrate a potential modality to enhance sensitivity to chemotherapy in PDAC.
Collapse
Affiliation(s)
- Yu-Kai Huang
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Wei-Chung Cheng
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan
| | - Ting-Ting Kuo
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Juan-Cheng Yang
- School of Chinese Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Yang-Chang Wu
- Graduate Institute of Integrated Medicine, College of Chinese Medicine, China Medical University, Taichung, Taiwan
- Department of Medical Laboratory Science and Biotechnology, College of Medical and Health Science, Asia University, Taichung, Taiwan
| | - Heng-Hsiung Wu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chia-Chien Lo
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Chih-Ying Hsieh
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Sze-Ching Wong
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
| | - Chih-Hao Lu
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wan-Ling Wu
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
| | - Shih-Jen Liu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli, Taiwan
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yi-Chuan Li
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Ching-Chan Lin
- Division of Hematology and Oncology, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Ning Shen
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Mien-Chie Hung
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jaw-Town Lin
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan
| | - Chun-Chieh Yeh
- Department of Medicine, School of Medicine, China Medical University, Taichung, Taiwan.
- Department of Surgery, Organ Transplantation Center, China Medical University Hospital, Taichung, Taiwan.
| | - Yuh-Pyng Sher
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan.
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, Taiwan.
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University and Academia Sinica, Taichung, Taiwan.
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan.
- Institute of Biochemistry and Molecular Biology, China Medical University, Taichung, Taiwan.
| |
Collapse
|
5
|
Gevertz JL, Kareva I. Minimally sufficient experimental design using identifiability analysis. NPJ Syst Biol Appl 2024; 10:2. [PMID: 38184643 PMCID: PMC10771435 DOI: 10.1038/s41540-023-00325-1] [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: 05/30/2023] [Accepted: 12/12/2023] [Indexed: 01/08/2024] Open
Abstract
Mathematical models are increasingly being developed and calibrated in tandem with data collection, empowering scientists to intervene in real time based on quantitative model predictions. Well-designed experiments can help augment the predictive power of a mathematical model but the question of when to collect data to maximize its utility for a model is non-trivial. Here we define data as model-informative if it results in a unique parametrization, assessed through the lens of practical identifiability. The framework we propose identifies an optimal experimental design (how much data to collect and when to collect it) that ensures parameter identifiability (permitting confidence in model predictions), while minimizing experimental time and costs. We demonstrate the power of the method by applying it to a modified version of a classic site-of-action pharmacokinetic/pharmacodynamic model that describes distribution of a drug into the tumor microenvironment (TME), where its efficacy is dependent on the level of target occupancy in the TME. In this context, we identify a minimal set of time points when data needs to be collected that robustly ensures practical identifiability of model parameters. The proposed methodology can be applied broadly to any mathematical model, allowing for the identification of a minimally sufficient experimental design that collects the most informative data.
Collapse
Affiliation(s)
- Jana L Gevertz
- Department of Mathematics and Statistics, The College of New Jersey, Ewing, NJ, USA.
| | - Irina Kareva
- Quantitative Pharmacology Department, EMD Serono, Merck KGaA, Billerica, MA, USA
| |
Collapse
|
6
|
Kulkarni MB, Ayachit NH, Aminabhavi TM. A Short Review on Miniaturized Biosensors for the Detection of Nucleic Acid Biomarkers. BIOSENSORS 2023; 13:412. [PMID: 36979624 PMCID: PMC10046286 DOI: 10.3390/bios13030412] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/17/2023] [Accepted: 03/20/2023] [Indexed: 06/18/2023]
Abstract
Even today, most biomarker testing is executed in centralized, dedicated laboratories using bulky instruments, automated analyzers, and increased analysis time and expenses. The development of miniaturized, faster, low-cost microdevices is immensely anticipated for substituting for these conventional laboratory-oriented assays and transferring diagnostic results directly onto the patient's smartphone using a cloud server. Pioneering biosensor-based approaches might make it possible to test biomarkers with reliability in a decentralized setting, but there are still a number of issues and restrictions that must be resolved before the development and use of several biosensors for the proper understanding of the measured biomarkers of numerous bioanalytes such as DNA, RNA, urine, and blood. One of the most promising processes to address some of the issues relating to the growing demand for susceptible, quick, and affordable analysis techniques in medical diagnostics is the creation of biosensors. This article critically discusses a short review of biosensors used for detecting nucleic acid biomarkers, and their use in biomedical prognostics will be addressed while considering several essential characteristics.
Collapse
Affiliation(s)
- Madhusudan B. Kulkarni
- School of Electronics and Communication Engineering, KLE Technological University, Vidyanagar, Hubballi 580023, Karnataka, India
- Medical Physics Department, Wisconsin Institutes for Medical Research, University of Wisconsin, Madison, WI 53705, USA
| | - Narasimha H. Ayachit
- School of Advanced Sciences, KLE Technological University, Hubballi 580031, Karnataka, India
| | - Tejraj M. Aminabhavi
- School of Advanced Sciences, KLE Technological University, Hubballi 580031, Karnataka, India
| |
Collapse
|
7
|
Herrera-Domínguez M, Morales-Luna G, Mahlknecht J, Cheng Q, Aguilar-Hernández I, Ornelas-Soto N. Optical Biosensors and Their Applications for the Detection of Water Pollutants. BIOSENSORS 2023; 13:bios13030370. [PMID: 36979582 PMCID: PMC10046542 DOI: 10.3390/bios13030370] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/02/2023] [Indexed: 05/14/2023]
Abstract
The correct detection and quantification of pollutants in water is key to regulating their presence in the environment. Biosensors offer several advantages, such as minimal sample preparation, short measurement times, high specificity and sensibility and low detection limits. The purpose of this review is to explore the different types of optical biosensors, focusing on their biological elements and their principle of operation, as well as recent applications in the detection of pollutants in water. According to our literature review, 33% of the publications used fluorescence-based biosensors, followed by surface plasmon resonance (SPR) with 28%. So far, SPR biosensors have achieved the best results in terms of detection limits. Although less common (22%), interferometers and resonators (4%) are also highly promising due to the low detection limits that can be reached using these techniques. In terms of biological recognition elements, 43% of the published works focused on antibodies due to their high affinity and stability, although they could be replaced with molecularly imprinted polymers. This review offers a unique compilation of the most recent work in the specific area of optical biosensing for water monitoring, focusing on both the biological element and the transducer used, as well as the type of target contaminant. Recent technological advances are discussed.
Collapse
Affiliation(s)
- Marcela Herrera-Domínguez
- Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey 64849, Mexico
| | - Gesuri Morales-Luna
- Departamento de Física y Matemáticas, Universidad Iberoamericana, Prolongación Paseo de la Reforma 880, Mexico City 01219, Mexico
| | - Jürgen Mahlknecht
- Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey 64849, Mexico
| | - Quan Cheng
- Department of Chemistry, University of California, Riverside, CA 92521, USA
| | - Iris Aguilar-Hernández
- Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey 64849, Mexico
- Correspondence: (I.A.-H.); (N.O.-S.)
| | - Nancy Ornelas-Soto
- Tecnológico de Monterrey, Escuela de Ingeniería y Ciencias, Ave. Eugenio Garza Sada 2501, Monterrey 64849, Mexico
- Correspondence: (I.A.-H.); (N.O.-S.)
| |
Collapse
|
8
|
Dreymann N, Möller A, Menger MM. Label-Free Determination of the Kinetic Parameters of Protein-Aptamer Interaction by Surface Plasmon Resonance. Methods Mol Biol 2023; 2570:141-153. [PMID: 36156780 DOI: 10.1007/978-1-0716-2695-5_11] [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: 01/25/2023]
Abstract
Due to their high specificity and affinity to target molecules, aptamers can be used as powerful tools in diagnostics, therapeutics, and environmental or food analytics. For the use in various applications, the detailed characterization of their binding behavior is an important step after selection to determine the interaction strength between the aptamer and its target and to find the best kinetics depending on the field of application. The surface plasmon resonance (SPR) spectroscopy is a powerful technology to investigate important parameters in molecular interaction, for example, kinetics, affinity, and specificity. The most-used system is the Biacore™ SPR system which comprises an optical biosensor for label-free monitoring of binding events in real time based on SPR. This biophysical phenomenon describes the changes in refractive index on a sensor surface which can be used to measure binding events and to determine kinetic constants. In this chapter, a detailed protocol for the determination of kinetic constants for protein-aptamer interaction is provided. An 82-nt long ssDNA aptamer which are targeted against human urokinase is used as a model system for determination of binding and dissociation constants using Biacore™ SPR technology. A detailed note section provides useful tips and pitfalls at the end of this chapter.
Collapse
Affiliation(s)
- Nico Dreymann
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalysis and Bioprocesses (IZI-BB), Potsdam, Germany.,Institute for Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Anja Möller
- Fraunhofer Institute for Cell Therapy and Immunology, Branch Bioanalysis and Bioprocesses (IZI-BB), Potsdam, Germany
| | - Marcus M Menger
- Branch Bioanalysis and Bioprocesses, Fraunhofer Institute for Cell Therapy and Immunology, Potsdam, Germany.
| |
Collapse
|
9
|
Klingel L, Siebert N, Troschke-Meurer S, Zumpe M, Ehlert K, Huber S, Loibner H, Mutschlechner O, Lode HN. Immune Response and Outcome of High-Risk Neuroblastoma Patients Immunized with Anti-Idiotypic Antibody Ganglidiomab: Results from Compassionate-Use Treatments. Cancers (Basel) 2022; 14:cancers14235802. [PMID: 36497290 PMCID: PMC9735439 DOI: 10.3390/cancers14235802] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/20/2022] [Accepted: 11/23/2022] [Indexed: 11/29/2022] Open
Abstract
(1) Background: High-risk neuroblastoma (HR-NB) is associated with a poor prognosis despite a multimodal high-intensity treatment regimen, including immunotherapy with anti-GD2 monoclonal antibodies (mAb). Here, we investigated the effects of an anti-idiotypic vaccine based on the mAb ganglidiomab that structurally mimics GD2. (2) Methods: Patients with HR-NB treated with anti-GD2 mAb dinutuximab beta and who achieved complete remission after frontline or salvage therapy were offered the vaccine (0.5 mg ganglidiomab adsorbed to Alhydrogel®). Side effects (CTCAE v4.03) and immune responses were determined on each visit. We also evaluated the time to relapse or progression until the last follow-up. (3) Results: Seven HR-NB patients (five frontlines, two relapsed) received 6-22 subcutaneous injections every two weeks. Six of the seven patients showed an immune response. The non-responding patient had a haploidentical stem cell transplantation as part of the previous treatment. No fever, pain, neuropathy, or toxicities ≥ grade 3 occurred during or post-treatment. All immunized patients did not experience relapses or progressions of their neuroblastoma. (4) Conclusions: This is the first-in-man use of the ganglidiomab vaccine, which was well-tolerated, and all patients not pre-treated by haploidentical transplantation developed vaccine-specific immune responses. These findings provide an important basis for the design of prospective clinical trials.
Collapse
Affiliation(s)
- Leah Klingel
- Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Nikolai Siebert
- Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Sascha Troschke-Meurer
- Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Maxi Zumpe
- Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Karoline Ehlert
- Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Stefanie Huber
- Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
| | - Hans Loibner
- Anyxis Immuno-Oncology GmbH, 1230 Vienna, Austria
| | | | - Holger N. Lode
- Pediatric Hematology and Oncology, University Medicine Greifswald, 17475 Greifswald, Germany
- Anyxis Immuno-Oncology GmbH, 1230 Vienna, Austria
- Correspondence: ; Tel.: +49-3834-86-6300
| |
Collapse
|
10
|
Fryer T, Rogers JD, Mellor C, Kohler TN, Minter R, Hollfelder F. Gigavalent Display of Proteins on Monodisperse Polyacrylamide Hydrogels as a Versatile Modular Platform for Functional Assays and Protein Engineering. ACS CENTRAL SCIENCE 2022; 8:1182-1195. [PMID: 36032770 PMCID: PMC9413441 DOI: 10.1021/acscentsci.2c00576] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Indexed: 06/15/2023]
Abstract
The assembly of robust, modular biological components into complex functional systems is central to synthetic biology. Here, we apply modular "plug and play" design principles to a solid-phase protein display system that facilitates protein purification and functional assays. Specifically, we capture proteins on polyacrylamide hydrogel display beads (PHD beads) made in microfluidic droplet generators. These monodisperse PHD beads are decorated with predefined amounts of anchors, methacrylate-PEG-benzylguanine (BG) and methacrylate-PEG-chloroalkane (CA), that react covalently with SNAP-/Halo-tag fusion proteins, respectively, in a specific, orthogonal, and stable fashion. Anchors, and thus proteins, are distributed throughout the entire bead volume, allowing attachment of ∼109 protein molecules per bead (⌀ 20 μm) -a higher density than achievable with commercial surface-modified beads. We showcase a diverse array of protein modules that enable the secondary capture of proteins, either noncovalently (IgG and SUMO-tag) or covalently (SpyCatcher, SpyTag, SnpCatcher, and SnpTag), in mono- and multivalent display formats. Solid-phase protein binding and enzymatic assays are carried out, and incorporating the photocleavable protein PhoCl enables the controlled release of modules via visible-light irradiation for functional assays in solution. We utilize photocleavage for valency engineering of an anti-TRAIL-R1 scFv, enhancing its apoptosis-inducing potency ∼50-fold through pentamerization.
Collapse
Affiliation(s)
- Thomas Fryer
- Department
of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, United Kingdom
- Antibody
Discovery and Protein Engineering, R&D, AstraZeneca, Milstein
Building, Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Joel David Rogers
- Department
of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, United Kingdom
- Antibody
Discovery and Protein Engineering, R&D, AstraZeneca, Milstein
Building, Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Christopher Mellor
- Department
of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - Timo N. Kohler
- Department
of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| | - Ralph Minter
- Antibody
Discovery and Protein Engineering, R&D, AstraZeneca, Milstein
Building, Granta Park, Cambridge CB21 6GH, United Kingdom
| | - Florian Hollfelder
- Department
of Biochemistry, University of Cambridge, 80 Tennis Court Road, Cambridge CB2 1GA, United Kingdom
| |
Collapse
|
11
|
Applications of Surface Plasmon Resonance and Biolayer Interferometry for Virus–Ligand Binding. Viruses 2022; 14:v14040717. [PMID: 35458446 PMCID: PMC9027846 DOI: 10.3390/v14040717] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 03/20/2022] [Accepted: 03/26/2022] [Indexed: 02/01/2023] Open
Abstract
Surface plasmon resonance and biolayer interferometry are two common real-time and label-free assays that quantify binding events by providing kinetic parameters. There is increased interest in using these techniques to characterize whole virus-ligand interactions, as the methods allow for more accurate characterization than that of a viral subunit-ligand interaction. This review aims to summarize and evaluate the uses of these technologies specifically in virus–ligand and virus-like particle–ligand binding cases to guide the field towards studies that apply these robust methods for whole virus-based studies.
Collapse
|
12
|
Oguntoye IO, Simone BK, Padmanabha S, Hartfield GZ, Amrollahi P, Hu TY, Ollanik AJ, Escarra MD. Silicon Nanodisk Huygens Metasurfaces for Portable and Low-Cost Refractive Index and Biomarker Sensing. ACS APPLIED NANO MATERIALS 2022; 5:3983-3991. [PMID: 35372799 PMCID: PMC8961735 DOI: 10.1021/acsanm.1c04443] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Accepted: 03/07/2022] [Indexed: 05/10/2023]
Abstract
Biomarker detection and bulk refractive index sensing are important across multiple industries ranging from early medical diagnosis to chemical process quality control. The bulky size, high cost, and complex architecture of existing refractive index and biomarker sensing technologies limit their use to highly skilled environments like hospitals, large food processing plants, and research labs. Here, we demonstrate a compact and inexpensive refractive index sensor based on resonant dielectric photonic nanoantenna arrays or metasurfaces. These dielectric resonances support Mie dipole and asymmetric resonances that shift with changes in their external environment. A single-wavelength transmission measurement in a portable (<250 in.3), low-cost (<$4000) sensor shows sensitivity to 1.9 × 10-6 change in the fluid refractive index without the use of a spectrometer or other complex optics. Our sensor assembly allows for measurements using multiple metasurfaces with identical resonances or varying resonance types for enhanced diagnostics on the same chip. Furthermore, a 10 kDa culture filtrate peptide CFP-10, a marker for human tuberculosis, is detected with our sensor with 10 pM resolution. This system has the potential to enable facile, fast, and highly sensitive measurements with adequate limits of detection for personalized biomedical diagnoses.
Collapse
Affiliation(s)
- Isaac O. Oguntoye
- Department
of Physics and Engineering Physics, Tulane
University, New Orleans, Louisiana 70118, United States
| | - Brittany K. Simone
- Department
of Physics and Engineering Physics, Tulane
University, New Orleans, Louisiana 70118, United States
| | - Siddharth Padmanabha
- Department
of Physics and Engineering Physics, Tulane
University, New Orleans, Louisiana 70118, United States
| | - George Z. Hartfield
- Department
of Physics and Engineering Physics, Tulane
University, New Orleans, Louisiana 70118, United States
| | - Pouya Amrollahi
- Center
of Cellular and Molecular Diagnostics, Tulane
University, New Orleans, Louisiana 70112, United States
| | - Tony Y. Hu
- Center
of Cellular and Molecular Diagnostics, Tulane
University, New Orleans, Louisiana 70112, United States
| | - Adam J. Ollanik
- Department
of Physics and Engineering Physics, Tulane
University, New Orleans, Louisiana 70118, United States
- Department
of Physics, University of Colorado Boulder, Boulder, Colorado 80309, United States
| | - Matthew D. Escarra
- Department
of Physics and Engineering Physics, Tulane
University, New Orleans, Louisiana 70118, United States
| |
Collapse
|
13
|
Mori S, Nomura K, Fujikawa K, Osawa T, Shionyu M, Yoda T, Shirai T, Tsuda S, Yoshizawa-Kumagaye K, Masuda S, Nishio H, Yoshiya T, Suzuki S, Muramoto M, Nishiyama KI, Shimamoto K. Intermolecular Interactions between a Membrane Protein and a Glycolipid Essential for Membrane Protein Integration. ACS Chem Biol 2022; 17:609-618. [PMID: 35239308 DOI: 10.1021/acschembio.1c00882] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Inducing newly synthesized proteins to appropriate locations is an indispensable biological function in every organism. Integration of proteins into biomembranes in Escherichia coli is mediated by proteinaceous factors, such as Sec translocons and an insertase YidC. Additionally, a glycolipid named MPIase (membrane protein integrase), composed of a long sugar chain and pyrophospholipid, was proven essential for membrane protein integration. We reported that a synthesized minimal unit of MPIase possessing only one trisaccharide, mini-MPIase-3, involves an essential structure for the integration activity. Here, to elucidate integration mechanisms using MPIase, we analyzed intermolecular interactions of MPIase or its synthetic analogs with a model substrate, the Pf3 coat protein, using physicochemical methods. Surface plasmon resonance (SPR) analyses revealed the importance of a pyrophosphate for affinity to the Pf3 coat protein. Compared with mini-MPIase-3, natural MPIase showed faster association and dissociation due to its long sugar chain despite the slight difference in affinity. To focus on more detailed MPIase substructures, we performed docking simulations and saturation transfer difference-nuclear magnetic resonance. These experiments yielded that the 6-O-acetyl group on glucosamine and the phosphate of MPIase play important roles leading to interactions with the Pf3 coat protein. The high affinity of MPIase to the hydrophobic region and the basic amino acid residues of the protein was suggested by docking simulations and proven experimentally by SPR using protein mutants devoid of target regions. These results demonstrated the direct interactions of MPIase with a substrate protein and revealed detailed mechanisms of membrane protein integration.
Collapse
Affiliation(s)
- Shoko Mori
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| | - Kaoru Nomura
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Kohki Fujikawa
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Tsukiho Osawa
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
| | - Masafumi Shionyu
- Department of Frontier Bioscience, Faculty of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura-cho, Nagahama, Shiga 526-0829, Japan
| | - Takao Yoda
- Department of Frontier Bioscience, Faculty of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura-cho, Nagahama, Shiga 526-0829, Japan
| | - Tsuyoshi Shirai
- Department of Frontier Bioscience, Faculty of Bioscience, Nagahama Institute of Bio-Science and Technology, 1266 Tamura-cho, Nagahama, Shiga 526-0829, Japan
| | - Shugo Tsuda
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Kumiko Yoshizawa-Kumagaye
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Shun Masuda
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Hideki Nishio
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Taku Yoshiya
- Peptide Institute, Inc., 7-2-9 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | - Sonomi Suzuki
- Department of Biological Chemistry and Food Sciences, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Maki Muramoto
- Department of Biological Chemistry and Food Sciences, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Ken-ichi Nishiyama
- Department of Biological Chemistry and Food Sciences, Faculty of Agriculture, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Keiko Shimamoto
- Bioorganic Research Institute, Suntory Foundation for Life Sciences, 8-1-1 Seikadai, Seika-cho, Soraku-gun, Kyoto 619-0284, Japan
- Department of Chemistry, Graduate School of Science, Osaka University, 1-1 Machikaneyama, Toyonaka, Osaka 560-0043, Japan
| |
Collapse
|
14
|
Emert-Sedlak LA, Moukha-Chafiq O, Shi H, Du S, Alvarado JJ, Pathak V, Tanner SG, Hunter RN, Nebane M, Chen L, Ilina TV, Ishima R, Zhang S, Kuzmichev YV, Wonderlich ER, Schader SM, Augelli-Szafran CE, Ptak RG, Smithgall TE. Inhibitors of HIV-1 Nef-Mediated Activation of the Myeloid Src-Family Kinase Hck Block HIV-1 Replication in Macrophages and Disrupt MHC-I Downregulation. ACS Infect Dis 2022; 8:91-105. [PMID: 34985256 PMCID: PMC9274903 DOI: 10.1021/acsinfecdis.1c00288] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
HIV-1 Nef is an attractive target for antiretroviral drug discovery because of its role in promoting HIV-1 infectivity, replication, and host immune system avoidance. Here, we applied a screening strategy in which recombinant HIV-1 Nef protein was coupled to activation of the Src-family tyrosine kinase Hck, which enhances the HIV-1 life cycle in macrophages. Nef stimulates recombinant Hck activity in vitro, providing a robust assay for chemical library screening. High-throughput screening of more than 730 000 compounds using the Nef·Hck assay identified six unique hit compounds that bound directly to recombinant Nef by surface plasmon resonance (SPR) in vitro and inhibited HIV-1 replication in primary macrophages in the 0.04 to 5 μM range without cytotoxicity. Eighty-four analogs were synthesized around an isothiazolone scaffold from this series, many of which bound to recombinant Nef and inhibited HIV-1 infectivity in the low to submicromolar range. Compounds in this series restored MHC-I to the surface of HIV-infected primary cells and disrupted a recombinant protein complex of Nef with the C-terminal tail of MHC-I and the μ1 subunit of the AP-1 endocytic trafficking protein. Nef inhibitors in this class have the potential to block HIV-1 replication in myeloid cells and trigger recognition of HIV-infected cells by the adaptive immune system in vivo.
Collapse
Affiliation(s)
- Lori A. Emert-Sedlak
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219
| | - Omar Moukha-Chafiq
- Department of Chemistry, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205
| | - Haibin Shi
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219
| | - Shoucheng Du
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219
| | - John J. Alvarado
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219
| | - Vibha Pathak
- Department of Chemistry, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205
| | - Samuel G. Tanner
- Department of Chemistry, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205
| | - Robert N. Hunter
- Department of Chemistry, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205
| | - Miranda Nebane
- Department of High-throughput Screening, Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205
| | - Li Chen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219
| | - Tatiana V. Ilina
- Department of Structural Biology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, Pittsburgh, PA 15260
| | - Rieko Ishima
- Department of Structural Biology, University of Pittsburgh School of Medicine, 3501 Fifth Avenue, Pittsburgh, PA 15260
| | - Sixue Zhang
- Department of High-throughput Screening, Drug Discovery Division, Southern Research Institute, 2000 Ninth Avenue South, Birmingham, AL 35205
| | - Yury V. Kuzmichev
- Department of Infectious Disease Research, Drug Development Division, Southern Research Institute, 431 Aviation Way, Frederick, MD 21701
| | - Elizabeth R. Wonderlich
- Department of Infectious Disease Research, Drug Development Division, Southern Research Institute, 431 Aviation Way, Frederick, MD 21701
| | - Susan M. Schader
- Department of Infectious Disease Research, Drug Development Division, Southern Research Institute, 431 Aviation Way, Frederick, MD 21701
| | | | - Roger G. Ptak
- Department of Infectious Disease Research, Drug Development Division, Southern Research Institute, 431 Aviation Way, Frederick, MD 21701
| | - Thomas E. Smithgall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, 450 Technology Drive, Pittsburgh, PA 15219
| |
Collapse
|
15
|
Lopez-Muñoz GA, Mughal S, Ramón-Azcón J. Sensors and Biosensors in Organs-on-a-Chip Platforms. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:55-80. [DOI: 10.1007/978-3-031-04039-9_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
16
|
Fujikawa K, Mori S, Nishiyama KI, Shimamoto K. A bacterial glycolipid essential for membrane protein integration. Adv Carbohydr Chem Biochem 2022; 81:95-129. [DOI: 10.1016/bs.accb.2022.09.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
17
|
Abstract
Plants are increasingly viewed as suitable expression hosts for the production of recombinant proteins, especially when oxidative folding and/or posttranslational modification is essential for protein stability and functionality. In contrast to traditional platforms such as yeast and mammalian cells, where the product is secreted into the culture medium, recombinant proteins expressed in plants are usually retained within the cells so additional effort is required during extraction and purification. Various extraction processes are used to release soluble proteins from plant tissues, followed by clarification to remove fibers and particulates before the target protein is purified. Fermentation media generally contain few proteins, making it easier to recover a secreted product, whereas the green juice extracted from plants usually contains a large number of host proteins that interfere with target isolation and purification. In this chapter, we describe the use of heat precipitation to remove a large portion of the host cell proteins, thus improving the efficiency of subsequent purification steps and the quality of the purified recombinant protein.
Collapse
Affiliation(s)
- Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
| |
Collapse
|
18
|
Spiegel H, Schillberg S, Nölke G. Production of Recombinant Proteins by Agrobacterium-Mediated Transient Expression. Methods Mol Biol 2022; 2480:89-102. [PMID: 35616859 DOI: 10.1007/978-1-0716-2241-4_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
The agroinfiltration of plant tissue is a robust method that allows the rapid and transient expression of recombinant proteins. Using wild-type plants as biomass, agroinfiltration exploits the ability of plants to synthesize even complex multimeric proteins that require oxidative folding and/or post-translational modifications, while avoiding the expensive and time-consuming creation of stably transformed plant lines. Here we describe a generic method for the transient expression of recombinant proteins in Nicotiana benthamiana at the small to medium laboratory scale, including appropriate binary vectors, the design and cloning of expression constructs, the transformation, selection, and cultivation of recombinant Agrobacterium tumefaciens, the infiltration of plants using a syringe or vacuum device, and finally the extraction of recombinant proteins from plant tissues.
Collapse
Affiliation(s)
- Holger Spiegel
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany.
| | - Stefan Schillberg
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
- Department of Phytopathology, Justus Liebig University Giessen, Giessen, Germany
| | - Greta Nölke
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Aachen, Germany
| |
Collapse
|
19
|
Armaghan F, Hajihassan Z. Engineering a variant of IL-17RA with high binding affinity to IL-17A for optimized immunotherapy. BIOTECHNOLOGY REPORTS (AMSTERDAM, NETHERLANDS) 2021; 32:e00682. [PMID: 34765462 PMCID: PMC8572878 DOI: 10.1016/j.btre.2021.e00682] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 09/25/2021] [Accepted: 10/01/2021] [Indexed: 11/08/2022]
Abstract
Immunotherapy is one of the most recently used treatments for numerous cancer types and also some autoimmune and inflammatory diseases. One of the valuable targets for immunotherapy is Interleukin-17A (IL-17A) or its receptor (IL-17RA) because overexpression of IL-17A as a pro-inflammatory cytokine is associated with several inflammatory, autoimmune and cancer diseases. In this study, the extracellular domain of IL-17RA involved in binding to IL-17A was mutated by using R software to achieve a variant with increased binding affinity to IL-17A. The ∆∆G value of -30.89 kcal/mol was calculated for the best variant (385) with point mutations of R265N, N91T, and W31K using the FoldX module. Also, the KD for its interaction with IL-17A was calculated 0.06 nM by surface plasmon resonance (SPR) technique. Our results indicated that variant 385 could bind to IL-17A with higher binding affinity than wild-type one, so, it can be a good therapeutic candidate for blocking IL-17A.
Collapse
Affiliation(s)
- Fatemeh Armaghan
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Zahra Hajihassan
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| |
Collapse
|
20
|
A-to-I mRNA Editing in a Ferric Siderophore Receptor Improves Competition for Iron in Xanthomonas oryzae pv. oryzicola. Microbiol Spectr 2021; 9:e0157121. [PMID: 34704802 PMCID: PMC8549721 DOI: 10.1128/spectrum.01571-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Iron is an essential element for the growth and survival of pathogenic bacteria; however, it is not fully understood how bacteria sense and respond to iron deficiency or excess. In this study, we show that xfeA in Xanthomonas oryzae pv. oryzicola senses extracytoplasmic iron and changes the hydrogen bonding network of ligand channel domains by adenosine-to-inosine (A-to-I) RNA editing. The frequency of A-to-I RNA editing during iron-deficient conditions increased by 76.87%, which facilitated the passage of iron through the XfeA outer membrane channel. When bacteria were subjected to high iron concentrations, the percentage of A-to-I editing in xfeA decreased, which reduced iron transport via XfeA. Furthermore, A-to-I RNA editing increased expression of multiple genes in the chemotaxis pathway, including methyl-accepting chemotaxis proteins (MCPs) that sense concentrations of exogenous ferrienterobactin (Fe-Ent) at the cytoplasmic membrane. A-to-I RNA editing helps X. oryzae pv. oryzicola move toward an iron-rich environment and supports our contention that editing in xfeA facilitates entry of a ferric siderophore. Overall, our results reveal a new signaling mechanism that bacteria use to adjust to iron concentrations. IMPORTANCE Adenosine-to-inosine (A-to-I) RNA editing, which is catalyzed by the adenosine deaminase RNA-specific family of enzymes, is a frequent posttranscriptional modification in metazoans. Research on A-to-I editing in bacteria is limited, and the importance of this editing is underestimated. In this study, we show that bacteria may use A-to-I editing as an alternative strategy to promote uptake of metabolic iron, and this form of editing can quickly and precisely modify RNA and subsequent protein sequences similar to an "on/off" switch. Thus, bacteria have the capacity to use a rapid switch-like mechanism to facilitate iron uptake and improve their competitiveness.
Collapse
|
21
|
Xiong H, Luo F, Zhou P, Yi J. Development of a reporter gene method to measure the bioactivity of anti-CD38 × CD3 bispecific antibody. Antib Ther 2021; 4:212-221. [PMID: 34676357 PMCID: PMC8524643 DOI: 10.1093/abt/tbab022] [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: 08/10/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 01/04/2023] Open
Abstract
Background A T cell-redirecting bispecific antibody (bsAb) consisting of a tumor-binding unit and a T cell-binding unit is a large group of antibody-based biologics against death-causing cancer diseases. The anti-CD38 × anti-CD3 bsAb (Y150) is potential for treating multiple myeloma (MM). When developing a cell-based reporter gene bioassay to assess the activities of Y150, it was found that the expression of CD38 on the human T lymphocyte cells (Jurkat) caused the nonspecific activation, which interfered with the specific T cells activation of mediated by the Y150 and CD38(+) tumor cells. Methods Here, we first knocked-out the CD38 expression on Jurkat T cell line by CRISPR-Cas9 technology, then developed a stable monoclonal CD38(−) Jurkat T cell line with an NFAT-RE driving luciferase expressing system. Further based on the CD38(−) Jurkat cell, we developed a reporter gene method to assess the bioactivity of the anti-CD38 × anti-CD3 bsAb. Results Knocking out CD38 expression abolished the nonspecific self-activation of the Jurkat cells. The selected stable monoclonal CD38(−) Jurkat T cell line assured the robustness of the report genes assay for the anti-CD38 × anti-CD3 bsAb. The relative potencies of the Y150 measured by the developed reporter gene assay were correlated with those by the flow-cytometry-based cell cytotoxicity assay and by the ELISA-based binding assay. Conclusions The developed reporter gene assay was mechanism of action-reflective for the bioactivity of anti-CD38 × anti-CD3 antibody, and suitable for the quality control for the bsAb product.
Collapse
Affiliation(s)
- Hui Xiong
- Wuhan YZY Biopharma Co., Ltd, Biolake City C2-1, No. 666 Gaoxin Road, Wuhan, Hubei 430075, China
| | - Fengyan Luo
- Wuhan YZY Biopharma Co., Ltd, Biolake City C2-1, No. 666 Gaoxin Road, Wuhan, Hubei 430075, China
| | - Pengfei Zhou
- Wuhan YZY Biopharma Co., Ltd, Biolake City C2-1, No. 666 Gaoxin Road, Wuhan, Hubei 430075, China
| | - Jizu Yi
- Wuhan YZY Biopharma Co., Ltd, Biolake City C2-1, No. 666 Gaoxin Road, Wuhan, Hubei 430075, China
| |
Collapse
|
22
|
Bheemavarapu LP, Shah MI, Joseph J, Sivaprakasam M. IQVision: An Image-Based Evaluation Tool for Quantitative Lateral Flow Immunoassay Kits. BIOSENSORS-BASEL 2021; 11:bios11070211. [PMID: 34203515 PMCID: PMC8428085 DOI: 10.3390/bios11070211] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022]
Abstract
The development of quantitative lateral flow immunoassay test strips involves a lot of research from kit manufacturers’ standpoint. Kit providers need to evaluate multiple parameters, including the location of test regions, sample flow speed, required sample volumes, reaction stability time, etc. A practical visualization tool assisting manufacturers in this process is very much required for the design of more sensitive and reliable quantitative LFIA test strips. In this paper, we present an image-based quantitative evaluation tool determining the practical functionality of fluorescence-labelled LFIA test cartridges. Image processing-based algorithms developed and presented in this paper provide a practical analysis of sample flow rates, reaction stability times of samples under test, and detect any abnormalities in test strips. Evaluation of the algorithm is done with Glycated Hemoglobin (HbA1C) and Vitamin D test cartridges. Practical sample flow progress for HbA1C test cartridges is demonstrated. The reaction stability time of HbA1C test samples is measured to be 12 min, while that of Vitamin D test samples is 24 min. Experimental evaluation of the abnormality detection algorithm is carried out, and sample flow abnormalities are detected with 100% accuracy while membrane irregularities are detected with 96% accuracy.
Collapse
Affiliation(s)
- Lalitha Pratyusha Bheemavarapu
- Department of Electrical Engineering, Indian Institute of Technology Madras, Chennai 600036, India; (J.J.); (M.S.)
- Correspondence:
| | - Malay Ilesh Shah
- Healthcare Technology Innovation Centre (HTIC), Indian Institute of Technology Madras, Chennai 600036, India;
| | - Jayaraj Joseph
- Department of Electrical Engineering, Indian Institute of Technology Madras, Chennai 600036, India; (J.J.); (M.S.)
| | - Mohanasankar Sivaprakasam
- Department of Electrical Engineering, Indian Institute of Technology Madras, Chennai 600036, India; (J.J.); (M.S.)
- Healthcare Technology Innovation Centre (HTIC), Indian Institute of Technology Madras, Chennai 600036, India;
| |
Collapse
|
23
|
Ettabib MA, Marti A, Liu Z, Bowden BM, Zervas MN, Bartlett PN, Wilkinson JS. Waveguide Enhanced Raman Spectroscopy for Biosensing: A Review. ACS Sens 2021; 6:2025-2045. [PMID: 34114813 DOI: 10.1021/acssensors.1c00366] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Waveguide enhanced Raman spectroscopy (WERS) utilizes simple, robust, high-index contrast dielectric waveguides to generate a strong evanescent field, through which laser light interacts with analytes residing on the surface of the waveguide. It offers a powerful tool for the direct identification and reproducible quantification of biochemical species and an alternative to surface enhanced Raman spectroscopy (SERS) without reliance on fragile noble metal nanostructures. The advent of low-cost laser diodes, compact spectrometers, and recent progress in material engineering, nanofabrication techniques, and software modeling tools have made realizing portable and cheap WERS Raman systems with high sensitivity a realistic possibility. This review highlights the latest progress in WERS technology and summarizes recent demonstrations and applications. Following an introduction to the fundamentals of WERS, the theoretical framework that underpins the WERS principles is presented. The main WERS design considerations are then discussed, and a review of the available approaches for the modification of waveguide surfaces for the attachment of different biorecognition elements is provided. The review concludes by discussing and contrasting the performance of recent WERS implementations, thereby providing a future roadmap of WERS technology where the key opportunities and challenges are highlighted.
Collapse
Affiliation(s)
- Mohamed A. Ettabib
- Zepler Institute for Photonics and Nanoelectronics, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Almudena Marti
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Zhen Liu
- Zepler Institute for Photonics and Nanoelectronics, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Bethany M. Bowden
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Michalis N. Zervas
- Zepler Institute for Photonics and Nanoelectronics, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - Philip N. Bartlett
- School of Chemistry, University of Southampton, Southampton SO17 1BJ, United Kingdom
| | - James S. Wilkinson
- Zepler Institute for Photonics and Nanoelectronics, University of Southampton, Southampton SO17 1BJ, United Kingdom
| |
Collapse
|
24
|
Thao NT, Hoang TX, Phan TB, Kim JY, Ta HKT, Trinh KTL, Tran NHT. Metal-enhanced sensing platform for the highly sensitive detection of C-reactive protein antibody and rhodamine B with applications in cardiovascular diseases and food safety. Dalton Trans 2021; 50:6962-6974. [PMID: 33929466 DOI: 10.1039/d0dt04353b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The potential applications of metal-enhanced fluorescence (MEF) devices include biosensors for the detection of trace amounts in biosciences, biotechnology, and pathogens that are relevant to medical diagnostics and food control. In the present study, the silver (Ag) film thickness (56 nm) of an MEF system was calibrated to maximize the depth-to-width ratio (Γ) of the surface plasmon resonance (SPR) active metal from reflectance dip curves. Upon plasmon coupling with thermally evaporated Ag, we demonstrated a 2.21-fold enhancement compared to the pristine flat substrate with the coefficient of variation (CV) ≈0.22% and the limit of detection (LOD) 0.001 mg L-1 of the concentration of an Alexa Fluor 488-labeled anti-C-reactive protein antibody (CRP@Alexa fluor 488). The structure was developed to simplify the in situ generation of biosensors for the surface-enhanced Raman spectroscopy (SERS) to determine Rhodamine B (RhB) with a highly robust performance. The procedure presented a simple and rapid sample pretreatment for the determination of RhB with a limit of quantification of 10-10 M and a satisfactory linear response (0.98). The results showed the excellent performance of the surface plasmon coupled emission (SPCE), which opens up possibilities for the accurate detection of small-volume and low-concentration target analytes due to the improved sensitivity and signal-to-noise ratio (SNR).
Collapse
Affiliation(s)
- Nguyen Thanh Thao
- Faculty of Materials Science and Technology, University of Science, Ho Chi Minh City, Viet Nam.
| | | | | | | | | | | | | |
Collapse
|
25
|
Chung H, Boriskina SV. Inverse design of a single-frequency diffractive biosensor based on the reporter cleavage detection mechanism. OPTICS EXPRESS 2021; 29:10780-10799. [PMID: 33820205 DOI: 10.1364/oe.421656] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 03/11/2021] [Indexed: 06/12/2023]
Abstract
Vertically interrogated porous silicon (PSi) interferometric biosensors have shown high potential for sensing bio-molecules as they combine high detection sensitivity with simplicity of fabrication, functionalization, optical coupling, and interfacing with microfluidic systems. However, most interferometric sensor designs require either broadband or wavelength-tunable light sources as well as wide-angle detection schemes, increasing their complexity and cost for point-of-care biosensing applications. The limit of detection of such sensors is also constrained by the small size and low refractive index of biological molecules, making it hard to detect very low concentrations of pathogens. In this work, we use a large-scale computational "inverse design" technique to demonstrate a single-frequency, fixed-angle PSi-based biosensor, which exploits a recently developed high-contrast reporter cleavage detection (HCCD) technique. The HCCD sensors detect high-index reporter cleavage events instead of low-index target analyte capture events as typical for traditional label-free optical biosensors. We use the inverse design approach to discover an optimal configuration of a PSi biosensor that makes use of the extended achievable range of cleavage-induced PSi effective index variations and can be interrogated at a single frequency and at a fixed angle. The optimized design in the form of a one-dimensional PSi grating exhibits the change in the reflectance up to 55 % at the interrogation angle of 12∘ and wavelength of 600 nm, which is caused by cleavage of Au nanoparticle reporters initially occupying 2% of the sensor surface area. The maximum possible change in reflectance is predicted to be 222 % (for a two-dimensional freeform design not amenable to fabrication). This demonstration may pave the way for developing new or redesigned conventional interferometric and colorimetric point-of-care biosensor systems in combination with the cleavage-based detection schemes.
Collapse
|
26
|
Lee S, Song H, Ahn H, Kim S, Choi JR, Kim K. Fiber-Optic Localized Surface Plasmon Resonance Sensors Based on Nanomaterials. SENSORS 2021; 21:s21030819. [PMID: 33530416 PMCID: PMC7865415 DOI: 10.3390/s21030819] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 01/16/2021] [Accepted: 01/20/2021] [Indexed: 12/31/2022]
Abstract
Applying fiber-optics on surface plasmon resonance (SPR) sensors is aimed at practical usability over conventional SPR sensors. Recently, field localization techniques using nanostructures or nanoparticles have been investigated on optical fibers for further sensitivity enhancement and significant target selectivity. In this review article, we explored varied recent research approaches of fiber-optics based localized surface plasmon resonance (LSPR) sensors. The article contains interesting experimental results using fiber-optic LSPR sensors for three different application categories: (1) chemical reactions measurements, (2) physical properties measurements, and (3) biological events monitoring. In addition, novel techniques which can create synergy combined with fiber-optic LSPR sensors were introduced. The review article suggests fiber-optic LSPR sensors have lots of potential for measurements of varied targets with high sensitivity. Moreover, the previous results show that the sensitivity enhancements which can be applied with creative varied plasmonic nanomaterials make it possible to detect minute changes including quick chemical reactions and tiny molecular activities.
Collapse
Affiliation(s)
- Seunghun Lee
- Departments of Congo-Mechatronics Engineering, Pusan National University, Busan 46241, Korea; (S.L.); (H.S.); (H.A.); (S.K.)
| | - Hyerin Song
- Departments of Congo-Mechatronics Engineering, Pusan National University, Busan 46241, Korea; (S.L.); (H.S.); (H.A.); (S.K.)
| | - Heesang Ahn
- Departments of Congo-Mechatronics Engineering, Pusan National University, Busan 46241, Korea; (S.L.); (H.S.); (H.A.); (S.K.)
| | - Seungchul Kim
- Departments of Congo-Mechatronics Engineering, Pusan National University, Busan 46241, Korea; (S.L.); (H.S.); (H.A.); (S.K.)
- Department of Optics and Mechatronics Engineering, Pusan National University, Busan 46241, Korea
| | - Jong-ryul Choi
- Medical Device Development Center, Daegu-Gyeongbuk Medical Innovation Foundation (DGMIF), Daegu 41061, Korea
- Correspondence: (J.-r.C.); (K.K.)
| | - Kyujung Kim
- Departments of Congo-Mechatronics Engineering, Pusan National University, Busan 46241, Korea; (S.L.); (H.S.); (H.A.); (S.K.)
- Department of Optics and Mechatronics Engineering, Pusan National University, Busan 46241, Korea
- Correspondence: (J.-r.C.); (K.K.)
| |
Collapse
|
27
|
Nangare S, Patil P. Nanoarchitectured Bioconjugates and Bioreceptors Mediated Surface Plasmon Resonance Biosensor for In Vitro Diagnosis of Alzheimer’s Disease: Development and Future Prospects. Crit Rev Anal Chem 2021; 52:1139-1169. [DOI: 10.1080/10408347.2020.1864716] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Sopan Nangare
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| | - Pravin Patil
- Department of Pharmaceutical Chemistry, H. R. Patel Institute of Pharmaceutical Education and Research, Shirpur, India
| |
Collapse
|
28
|
de Oliveira Vitarelli M, Elias MC. Quantifying the Affinity of Trypanosoma cruzi RPA-1 to the Single-Stranded DNA Overhang of the Telomere Using Surface Plasmon Resonance. Methods Mol Biol 2021; 2281:217-228. [PMID: 33847961 DOI: 10.1007/978-1-0716-1290-3_13] [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] [Indexed: 06/12/2023]
Abstract
Surface plasmon resonance (SPR) biosensors provide real-time binding affinity measurements between a pair of biomolecules, characterizing its interaction dynamics. An example of Trypanosoma cruzi's RPA-1 and a single-stranded DNA telomere sequence is presented with detailed guidelines and fundamentals for SPR technology.
Collapse
Affiliation(s)
- Marcela de Oliveira Vitarelli
- Cell Cycle Laboratory, Butantan Institute, São Paulo, SP, Brazil
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil
| | - Maria Carolina Elias
- Cell Cycle Laboratory, Butantan Institute, São Paulo, SP, Brazil.
- Center of Toxins, Immune Response and Cell Signaling (CeTICS), Butantan Institute, São Paulo, SP, Brazil.
| |
Collapse
|
29
|
Hampitak P, Jowitt TA, Melendrez D, Fresquet M, Hamilton P, Iliut M, Nie K, Spencer B, Lennon R, Vijayaraghavan A. A Point-of-Care Immunosensor Based on a Quartz Crystal Microbalance with Graphene Biointerface for Antibody Assay. ACS Sens 2020; 5:3520-3532. [PMID: 33103441 PMCID: PMC7706117 DOI: 10.1021/acssensors.0c01641] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 10/13/2020] [Indexed: 12/31/2022]
Abstract
We present a sensitive and low-cost immunoassay, based on a customized open-source quartz crystal microbalance coupled with graphene biointerface sensors (G-QCM), to quantify antibodies in undiluted patient serum. We demonstrate its efficacy for a specific antibody against the phospholipase A2 receptor (anti-PLA2R), which is a biomarker in primary membranous nephropathy. A novel graphene-protein biointerface was constructed by adsorbing a low concentration of denatured bovine serum albumin (dBSA) on the reduced graphene oxide (rGO) sensor surface. The dBSA film prevents the denaturation of the protein receptor on the rGO surface and serves as the cross-linker for immobilization of the receptor for anti-PLA2R antibodies on the surface. The detection limit and selectivity of this G-QCM biosensor was compared with a commercial QCM system. The G-QCM immunoassay exhibited good specificity and high sensitivity toward the target, with an order of magnitude better detection limit (of 100 ng/mL) compared to the commercial system, at a fraction of the cost and with considerable time saving. The results obtained from patient sera compared favorably with those from enzyme-linked immunosorbent assay, validating the feasibility of use in clinical applications. The multifunctional dBSA-rGO platform provides a promising biofunctionalization method for universal immunoassay and biosensors. With the advantages of inexpensive, rapid, and sensitive detection, the G-QCM sensor and instrument form an effective autoimmune disease screening tool.
Collapse
Affiliation(s)
- Piramon Hampitak
- Department
of Materials and National Graphene Institute, Faculty of Science and
Engineering, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| | - Thomas A. Jowitt
- School
of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| | - Daniel Melendrez
- Department
of Materials and National Graphene Institute, Faculty of Science and
Engineering, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| | - Maryline Fresquet
- School
of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| | - Patrick Hamilton
- Wellcome
Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and
Regenerative Medicine, School of Biological Sciences, Faculty of Biology
Medicine and Health, The University of Manchester,
Manchester Academic Health Science Centre, Manchester M13 9PT, U.K.
- Manchester
Academic Health Science Centre (MAHSC), The University of Manchester, Manchester M13 9PL, U.K.
| | - Maria Iliut
- Department
of Materials and National Graphene Institute, Faculty of Science and
Engineering, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| | - Kaiwen Nie
- Department
of Materials and National Graphene Institute, Faculty of Science and
Engineering, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| | - Ben Spencer
- Department
of Materials and National Graphene Institute, Faculty of Science and
Engineering, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| | - Rachel Lennon
- Wellcome
Centre for Cell-Matrix Research, Division of Cell-Matrix Biology and
Regenerative Medicine, School of Biological Sciences, Faculty of Biology
Medicine and Health, The University of Manchester,
Manchester Academic Health Science Centre, Manchester M13 9PT, U.K.
- Department
of Paediatric Nephrology, Royal Manchester Children’s Hospital,
Manchester University Hospitals NHS Foundation Trust, Manchester Academic Health Science Centre, Manchester M13 9WL, U.K.
| | - Aravind Vijayaraghavan
- Department
of Materials and National Graphene Institute, Faculty of Science and
Engineering, The University of Manchester, Oxford Road, Manchester M13 9PL, U.K.
| |
Collapse
|
30
|
Adamopoulos C, Gharia A, Niknejad A, Stojanović V, Anwar M. Microfluidic Packaging Integration with Electronic-Photonic Biosensors Using 3D Printed Transfer Molding. BIOSENSORS 2020; 10:bios10110177. [PMID: 33202594 PMCID: PMC7698318 DOI: 10.3390/bios10110177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/10/2020] [Accepted: 11/12/2020] [Indexed: 01/18/2023]
Abstract
Multiplexed sensing in integrated silicon electronic-photonic platforms requires microfluidics with both high density micro-scale channels and meso-scale features to accommodate for optical, electrical, and fluidic coupling in small, millimeter-scale areas. Three-dimensional (3D) printed transfer molding offers a facile and rapid method to create both micro and meso-scale features in complex multilayer microfluidics in order to integrate with monolithic electronic-photonic system-on-chips with multiplexed rows of 5 μm radius micro-ring resonators (MRRs), allowing for simultaneous optical, electrical, and microfluidic coupling on chip. Here, we demonstrate this microfluidic packaging strategy on an integrated silicon photonic biosensor, setting the basis for highly multiplexed molecular sensing on-chip.
Collapse
Affiliation(s)
- Christos Adamopoulos
- Department of Electrical Engineering and Computer Science, University of California Berkeley, Berkeley, CA 94720, USA; (A.G.); (A.N.); (V.S.)
- Correspondence:
| | - Asmaysinh Gharia
- Department of Electrical Engineering and Computer Science, University of California Berkeley, Berkeley, CA 94720, USA; (A.G.); (A.N.); (V.S.)
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA 94158, USA;
| | - Ali Niknejad
- Department of Electrical Engineering and Computer Science, University of California Berkeley, Berkeley, CA 94720, USA; (A.G.); (A.N.); (V.S.)
| | - Vladimir Stojanović
- Department of Electrical Engineering and Computer Science, University of California Berkeley, Berkeley, CA 94720, USA; (A.G.); (A.N.); (V.S.)
| | - Mekhail Anwar
- Department of Radiation Oncology, University of California San Francisco, San Francisco, CA 94158, USA;
| |
Collapse
|
31
|
Mataji-Kojouri A, Ozen MO, Shahabadi M, Inci F, Demirci U. Entangled Nanoplasmonic Cavities for Estimating Thickness of Surface-Adsorbed Layers. ACS NANO 2020; 14:8518-8527. [PMID: 32639713 DOI: 10.1021/acsnano.0c02797] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Plasmonic sensors provide real-time and label-free detection of biotargets with unprecedented sensitivity and detection limit. However, they usually lack the ability to estimate the thickness of the target layer formed on top of the sensing surface. Here, we report a sensing modality based on reflection spectroscopy of a nanoplasmonic Fabry-Perot cavity array, which exhibits characteristics of both surface plasmon polaritons and localized plasmon resonances and outperforms its conventional counterparts by providing the thickness of the surface-adsorbed layers. Through numerical simulations, we demonstrate that the designed plasmonic surface resembles two entangled Fabry-Perot cavities excited from both ends. Performance of the device is evaluated by studying sensor response in the refractive index (RI) measurement of aqueous glycerol solutions and during formation of a surface-adsorbed layer consisting of protein (i.e., NeutrAvidin) molecules. By tracking the resonance wavelengths of the two modes of the nanoplasmonic surface, it is therefore possible to measure the thickness of a homogeneous adsorbed layer and RI of the background solution with precisions better than 4 nm and 0.0001 RI units. Using numerical simulations, we show that the thickness estimation algorithm can be extended for layers consisting of nanometric analytes adsorbed on an antibody-coated sensor surface. Furthermore, performance of the device has been evaluated to detect exosomes. By providing a thickness estimation for adsorbed layers and differentiating binding events from background RI variations, this device can potentially supersede conventional plasmonic sensors.
Collapse
Affiliation(s)
- Amideddin Mataji-Kojouri
- Photonics Research Laboratory, Center of Excellence on Applied Electromagnetic Systems, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran 1439957131, Iran
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, Stanford School of Medicine, Stanford University, Palo Alto, California 94304, United States
| | - Mehmet Ozgun Ozen
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, Stanford School of Medicine, Stanford University, Palo Alto, California 94304, United States
| | - Mahmoud Shahabadi
- Photonics Research Laboratory, Center of Excellence on Applied Electromagnetic Systems, School of Electrical and Computer Engineering, College of Engineering, University of Tehran, Tehran 1439957131, Iran
| | - Fatih Inci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, Stanford School of Medicine, Stanford University, Palo Alto, California 94304, United States
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, Stanford School of Medicine, Stanford University, Palo Alto, California 94304, United States
| |
Collapse
|
32
|
Recent Strategic Advances in CFTR Drug Discovery: An Overview. Int J Mol Sci 2020; 21:ijms21072407. [PMID: 32244346 PMCID: PMC7177952 DOI: 10.3390/ijms21072407] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 03/25/2020] [Accepted: 03/27/2020] [Indexed: 12/13/2022] Open
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR)-rescuing drugs have already transformed cystic fibrosis (CF) from a fatal disease to a treatable chronic condition. However, new-generation drugs able to bind CFTR with higher specificity/affinity and to exert stronger therapeutic benefits and fewer side effects are still awaited. Computational methods and biosensors have become indispensable tools in the process of drug discovery for many important human pathologies. Instead, they have been used only piecemeal in CF so far, calling for their appropriate integration with well-tried CF biochemical and cell-based models to speed up the discovery of new CFTR-rescuing drugs. This review will give an overview of the available structures and computational models of CFTR and of the biosensors, biochemical and cell-based assays already used in CF-oriented studies. It will also give the reader some insights about how to integrate these tools as to improve the efficiency of the drug discovery process targeted to CFTR.
Collapse
|
33
|
Soules KR, Dmitriev A, LaBrie SD, Dimond ZE, May BH, Johnson DK, Zhang Y, Battaile KP, Lovell S, Hefty PS. Structural and ligand binding analyses of the periplasmic sensor domain of RsbU in Chlamydia trachomatis support a role in TCA cycle regulation. Mol Microbiol 2020; 113:68-88. [PMID: 31637787 PMCID: PMC7007330 DOI: 10.1111/mmi.14401] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2019] [Indexed: 12/17/2022]
Abstract
Chlamydia trachomatis is an obligate intracellular bacteria that undergo dynamic morphologic and physiologic conversions upon gaining an access to a eukaryotic cell. These conversions likely require the detection of key environmental conditions and regulation of metabolic activity. Chlamydia encodes homologs to proteins in the Rsb phosphoregulatory partner-switching pathway, best described in Bacillus subtilis. ORF CT588 has a strong sequence similarity to RsbU cytoplasmic phosphatase domain but also contains a unique periplasmic sensor domain that is expected to control the phosphatase activity. A 1.7 Å crystal structure of the periplasmic domain of the RsbU protein from C. trachomatis (PDB 6MAB) displays close structural similarity to DctB from Vibrio and Sinorhizobium. DctB has been shown, both structurally and functionally, to specifically bind to the tricarboxylic acid (TCA) cycle intermediate succinate. Surface plasmon resonance and differential scanning fluorimetry of TCA intermediates and potential metabolites from a virtual screen of RsbU revealed that alpha-ketoglutarate, malate and oxaloacetate bound to the RsbU periplasmic domain. Substitutions in the putative binding site resulted in reduced binding capabilities. An RsbU null mutant showed severe growth defects which could be restored through genetic complementation. Chemical inhibition of ATP synthesis by oxidative phosphorylation phenocopied the growth defect observed in the RsbU null strain. Altogether, these data support a model with the Rsb system responding differentially to TCA cycle intermediates to regulate metabolism and key differentiation processes.
Collapse
Affiliation(s)
- Katelyn R Soules
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA
| | - Aidan Dmitriev
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA
| | - Scott D LaBrie
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA
| | - Zoë E Dimond
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA
| | - Benjamin H May
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA
| | - David K Johnson
- Computational Chemical Biology Core Facility, Del Shankel Structural Biology Center, University of Kansas, Lawrence, KS, 66047, USA
| | - Yang Zhang
- Computational Medicine & Bioinformatics, University of Michigan, Ann Arbor, MI, 48109, USA
| | - Kevin P Battaile
- IMCA-CAT, Hauptman-Woodward Medical Research Institute, Argonne, IL, 60439, USA
| | - Scott Lovell
- Protein Structure Laboratory, Del Shankel Structural Biology Center, University of Kansas, Lawrence, KS, 66047, USA
| | - P Scott Hefty
- Department of Molecular Biosciences, University of Kansas, Lawrence, KS, 66045, USA
| |
Collapse
|
34
|
Protein binding kinetics quantification via coupled plasmonic-photonic resonance nanosensors in generic microplate reader. Biosens Bioelectron 2019; 142:111494. [DOI: 10.1016/j.bios.2019.111494] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 06/21/2019] [Accepted: 07/04/2019] [Indexed: 02/06/2023]
|
35
|
Aviñó A, Eritja R, Ciudad CJ, Noé V. Parallel Clamps and Polypurine Hairpins (PPRH) for Gene Silencing and Triplex‐Affinity Capture: Design, Synthesis, and Use. ACTA ACUST UNITED AC 2019; 77:e78. [DOI: 10.1002/cpnc.78] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
| | | | - Carlos J. Ciudad
- School of Pharmacy and IN2UBUniversity of Barcelona Barcelona Spain
| | - Verónica Noé
- School of Pharmacy and IN2UBUniversity of Barcelona Barcelona Spain
| |
Collapse
|
36
|
Pan Y, Chai X, Gao Q, Zhou L, Zhang S, Li L, Luan S. Dynamic Interactions of Plant CNGC Subunits and Calmodulins Drive Oscillatory Ca 2+ Channel Activities. Dev Cell 2019; 48:710-725.e5. [PMID: 30713075 DOI: 10.1016/j.devcel.2018.12.025] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Revised: 10/03/2018] [Accepted: 12/28/2018] [Indexed: 12/20/2022]
Abstract
Calcium is a universal signal in all eukaryotes, but the mechanism for encoding calcium signatures remains largely unknown. Calcium oscillations control pollen tube growth and fertilization in flowering plants, serving as a model for dissecting the molecular machines that mediate calcium fluctuations. We report that pollen-tube-specific cyclic nucleotide-gated channels (CNGC18, CNGC8, and CNGC7) together with calmodulin 2 (CaM2) constitute a molecular switch that either opens or closes the calcium channel depending on cellular calcium levels. Under low calcium, calcium-free calmodulin 2 (Apo-CaM2) interacts with CNGC18-CNGC8 complex, leading to activation of the influx channel and consequently increasing cytosolic calcium levels. Calcium-bound CaM2 dissociates from CNGC18/8 heterotetramer, closing the channel and initiating a downturn of cellular calcium levels. We further reconstituted the calcium oscillator in HEK293 cells, supporting the model that Ca2+-CaM-dependent regulation of CNGC channel activity provides an auto-regulatory feedback mechanism for calcium oscillations during pollen tube growth.
Collapse
Affiliation(s)
- Yajun Pan
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Xuyang Chai
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Qifei Gao
- Department of Plant and Microbial Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Liming Zhou
- Department of Plant and Microbial Biology, University of California at Berkeley, Berkeley, CA 94720, USA
| | - Sisi Zhang
- College of Life Sciences, Capital Normal University, Beijing 100048, China
| | - Legong Li
- College of Life Sciences, Capital Normal University, Beijing 100048, China.
| | - Sheng Luan
- Department of Plant and Microbial Biology, University of California at Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
37
|
Christodouleas DC, Kaur B, Chorti P. From Point-of-Care Testing to eHealth Diagnostic Devices (eDiagnostics). ACS CENTRAL SCIENCE 2018; 4:1600-1616. [PMID: 30648144 PMCID: PMC6311959 DOI: 10.1021/acscentsci.8b00625] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Indexed: 05/09/2023]
Abstract
Point-of-care devices were originally designed to allow medical testing at or near the point of care by health-care professionals. Some point-of-care devices allow medical self-testing at home but cannot fully cover the growing diagnostic needs of eHealth systems that are under development in many countries. A number of easy-to-use, network-connected diagnostic devices for self-testing are needed to allow remote monitoring of patients' health. This Outlook highlights the essential characteristics of diagnostic devices for eHealth settings and indicates point-of-care technologies that may lead to the development of new devices. It also describes the most representative examples of simple-to-use, point-of-care devices that have been used for analysis of untreated biological samples.
Collapse
Affiliation(s)
| | - Balwinder Kaur
- Department of Chemistry, University
of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| | - Parthena Chorti
- Department of Chemistry, University
of Massachusetts Lowell, Lowell, Massachusetts 01854, United States
| |
Collapse
|
38
|
Immobilized Enzyme Reactors: an Overview of Applications in Drug Discovery from 2008 to 2018. Chromatographia 2018. [DOI: 10.1007/s10337-018-3663-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
39
|
Voss OH, Lee HN, Tian L, Krzewski K, Coligan JE. Liposome Preparation for the Analysis of Lipid-Receptor Interaction and Efferocytosis. ACTA ACUST UNITED AC 2018; 120:14.44.1-14.44.21. [PMID: 29512142 DOI: 10.1002/cpim.43] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Efficient phagocytosis of apoptotic cells (efferocytosis) is essential for immune homeostasis. Phospholipids exposed on the surface of apoptotic cells, such as phosphatidylserine, supply important "eat-me" signals. Liposomes are lipid bilayer vesicles that can be generated from one or several types of phospholipids of interest. Thus, these vesicles offer versatility, flexibility, and, importantly, a three-dimensional structure for studying the interaction between lipids and their receptors as well as the lipid-receptor interaction-mediated signaling events controlling efferocytosis by cells like professional phagocytes. Here, we describe methods to prepare liposomes, perform liposome-based lipid-receptor binding assays, use liposomes to block efferocytosis, and utilize liposome-coated beads as apoptotic cell surrogates for phagocytosis. © 2018 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Oliver H Voss
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - Ha-Na Lee
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - Linjie Tian
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - Konrad Krzewski
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| | - John E Coligan
- Receptor Cell Biology Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland
| |
Collapse
|
40
|
Adamski CJ, Palzkill T. BLIP-II Employs Differential Hotspot Residues To Bind Structurally Similar Staphylococcus aureus PBP2a and Class A β-Lactamases. Biochemistry 2017; 56:1075-1084. [PMID: 28182405 DOI: 10.1021/acs.biochem.6b00978] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The interaction of β-lactamase inhibitory protein II (BLIP-II) with β-lactamases serves as a model system to investigate the principles underlying protein-protein interactions. Previous studies have focused on identifying the determinants of binding affinity and specificity between BLIP-II and class A β-lactamases. However, interactions between BLIP-II and other bacterial proteins have yet to be explored. Here, we provide evidence that BLIP-II binds penicillin binding protein 2a (PBP2a) from methicillin-resistant Staphylococcus aureus (MRSA) with a KD in the low micromolar range. In comparison to the binding constants for the potent interaction between BLIP-II and TEM-1 β-lactamase (KD = 0.5 pM), the on-rate for BLIP-II binding PBP2a is 44 000 times slower and the off-rate is 170 times faster. Therefore, a slow association rate is a limiting factor for the potency of the interaction between BLIP-II and PBP2a. Results from alanine scanning mutagenesis of the predicted interface residues of BLIP-II indicate that charged residues on the periphery of the BLIP-II interface play a critical role for binding PBP2a, in contrast to previous findings that aromatic residues at the center of the BLIP-II interface are critical for the interaction with β-lactamases. Interestingly, many of the alanine mutants at the BLIP-II interface increase kon for binding PBP2a, consistent with the association rate being a limiting factor for affinity. In summary, the results of the study reveal that BLIP-II binds PBP2a, although weakly compared to binding of β-lactamases, and provides insights into the different binding strategies used for these targets.
Collapse
Affiliation(s)
- Carolyn J Adamski
- Department of Biochemistry and Molecular Biology, ‡Department of Pharmacology, Baylor College of Medicine , Houston, Texas 77030, United States
| | - Timothy Palzkill
- Department of Biochemistry and Molecular Biology, ‡Department of Pharmacology, Baylor College of Medicine , Houston, Texas 77030, United States
| |
Collapse
|
41
|
Abstract
The use of optical biosensors for studying macromolecular interactions is gaining increasing popularity. In one study, 1514 papers that involved the application of biosensor data were identified for the year 2009 alone (Rich and Myszka, J Mol Recognit 24:892-914, 2011), the sheer volume and variety of which present a daunting task for the burgeoning biosensor user to accumulate and decipher. This chapter is designed to provide the reader with the tools necessary to prepare, design, and efficiently execute a kinetic experiment on Biacore. It is written to guide the Biacore user through basic theory, system maintenance, and assay setup while also offering some practical tips that we find useful for Biacore-based studies. Many kinetic-based screening assays require rigorous sample preparation and purification prior to analysis. To highlight these procedures, this protocol describes the kinetic characterization of single chain Fv (scFv) antibody fragments from crude bacterial lysates using an antibody affinity capture approach. Even though we specifically describe the capture of HA-tagged scFv antibody fragments to an anti-HA tag monoclonal antibody-immobilized surface prior to kinetic analysis, the same methodologies are universally applicable and can be used for practically any affinity pair and most Biacore systems.
Collapse
Affiliation(s)
- Paul Leonard
- Biomedical Diagnostics Institute, Dublin City University, Dublin 9, Ireland
- School of Biotechnology, Dublin City University, Dublin 9, Ireland
- National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland
| | - Stephen Hearty
- Biomedical Diagnostics Institute, Dublin City University, Dublin 9, Ireland
- School of Biotechnology, Dublin City University, Dublin 9, Ireland
- National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland
| | - Hui Ma
- Biomedical Diagnostics Institute, Dublin City University, Dublin 9, Ireland
| | - Richard O'Kennedy
- Biomedical Diagnostics Institute, Dublin City University, Dublin 9, Ireland.
- School of Biotechnology, Dublin City University, Dublin 9, Ireland.
- National Centre for Sensor Research, Dublin City University, Dublin 9, Ireland.
| |
Collapse
|
42
|
Arif A, Jia J, Halawani D, Fox PL. Experimental approaches for investigation of aminoacyl tRNA synthetase phosphorylation. Methods 2016; 113:72-82. [PMID: 27729295 DOI: 10.1016/j.ymeth.2016.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 10/04/2016] [Accepted: 10/06/2016] [Indexed: 02/04/2023] Open
Abstract
Phosphorylation of many aminoacyl tRNA synthetases (AARSs) has been recognized for decades, but the contribution of post-translational modification to their primary role in tRNA charging and decryption of genetic code remains unclear. In contrast, phosphorylation is essential for performance of diverse noncanonical functions of AARSs unrelated to protein synthesis. Phosphorylation of glutamyl-prolyl tRNA synthetase (EPRS) has been investigated extensively in our laboratory for more than a decade, and has served as an archetype for studies of other AARSs. EPRS is a constituent of the IFN-γ-activated inhibitor of translation (GAIT) complex that directs transcript-selective translational control in myeloid cells. Stimulus-dependent phosphorylation of EPRS is essential for its release from the parental multi-aminoacyl tRNA synthetase complex (MSC), for binding to other GAIT complex proteins, and for regulating the binding to target mRNAs. Importantly, phosphorylation is the common driving force for the context- and stimulus-dependent release, and non-canonical activity, of other AARSs residing in the MSC, for example, lysyl tRNA synthetase (KARS). Here, we describe the concepts and experimental methodologies we have used to investigate the influence of phosphorylation on the structure and function of EPRS. We suggest that application of these approaches will help to identify new functional phosphorylation event(s) in other AARSs and elucidate their possible roles in noncanonical activities.
Collapse
Affiliation(s)
- Abul Arif
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Jie Jia
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Dalia Halawani
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Paul L Fox
- Department of Cellular and Molecular Medicine, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
43
|
Hintersteiner B, Lingg N, Zhang P, Woen S, Hoi KM, Stranner S, Wiederkum S, Mutschlechner O, Schuster M, Loibner H, Jungbauer A. Charge heterogeneity: Basic antibody charge variants with increased binding to Fc receptors. MAbs 2016; 8:1548-1560. [PMID: 27559765 PMCID: PMC5098448 DOI: 10.1080/19420862.2016.1225642] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We identified active isoforms of the chimeric anti-GD2 antibody, ch14.18, a recombinant antibody produced in Chinese hamster ovary cells, which is already used in clinical trials.1,2,3 We separated the antibody by high resolution ion-exchange chromatography with linear pH gradient elution into acidic, main and basic charge variants on a preparative scale yielding enough material for an in-depth study of the sources and the effects of microheterogeneity. The binding affinity of the charge variants toward the antigen and various cell surface receptors was studied by Biacore. Effector functions were evaluated using cellular assays for antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity. Basic charge variants showed increased binding to cell surface receptor FcγRIIIa, which plays a major role in regulating effector functions. Furthermore, increased binding of the basic fractions to the neonatal receptor was observed. As this receptor mediates the prolonged half-life of IgG in human serum, this data may well hint at an increased serum half-life of these basic variants compared to their more acidic counterparts. Different glycoform patterns, C-terminal lysine clipping and N-terminal pyroglutamate formation were identified as the main structural sources for the observed isoform pattern. Potential differences in structural stability between individual charge variant fractions by nano differential scanning calorimetry could not been detected. Our in-vitro data suggests that the connection between microheterogeneity and the biological activity of recombinant antibody therapeutics deserves more attention than commonly accepted.
Collapse
Affiliation(s)
- Beate Hintersteiner
- a Department of Biotechnology , University of Natural Resources and Life Sciences, Vienna , Vienna , Austria
| | - Nico Lingg
- a Department of Biotechnology , University of Natural Resources and Life Sciences, Vienna , Vienna , Austria
| | - Peiqing Zhang
- b Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR) , Centros , Singapore
| | - Susanto Woen
- b Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR) , Centros , Singapore
| | - Kong Meng Hoi
- b Bioprocessing Technology Institute, Agency for Science, Technology and Research (A*STAR) , Centros , Singapore
| | - Stefan Stranner
- c Apeiron Biologics AG, Campus-Vienna-Biocenter , Vienna , Austria
| | | | | | - Manfred Schuster
- c Apeiron Biologics AG, Campus-Vienna-Biocenter , Vienna , Austria
| | - Hans Loibner
- c Apeiron Biologics AG, Campus-Vienna-Biocenter , Vienna , Austria
| | - Alois Jungbauer
- a Department of Biotechnology , University of Natural Resources and Life Sciences, Vienna , Vienna , Austria
| |
Collapse
|
44
|
Grover P, Shi H, Baumgartner M, Camacho CJ, Smithgall TE. Fluorescence Polarization Screening Assays for Small Molecule Allosteric Modulators of ABL Kinase Function. PLoS One 2015. [PMID: 26222440 PMCID: PMC4519180 DOI: 10.1371/journal.pone.0133590] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The ABL protein-tyrosine kinase regulates intracellular signaling pathways controlling diverse cellular processes and contributes to several forms of cancer. The kinase activity of ABL is repressed by intramolecular interactions involving its regulatory Ncap, SH3 and SH2 domains. Small molecules that allosterically regulate ABL kinase activity through its non-catalytic domains may represent selective probes of ABL function. Here we report a screening assay for chemical modulators of ABL kinase activity that target the regulatory interaction of the SH3 domain with the SH2-kinase linker. This fluorescence polarization (FP) assay is based on a purified recombinant ABL protein consisting of the N-cap, SH3 and SH2 domains plus the SH2-kinase linker (N32L protein) and a short fluorescein-labeled probe peptide that binds to the SH3 domain. In assay development experiments, we found that the probe peptide binds to the recombinant ABL N32L protein in vitro, producing a robust FP signal that can be competed with an excess of unlabeled peptide. The FP signal is not observed with control N32L proteins bearing either an inactivating mutation in the SH3 domain or enhanced SH3:linker interaction. A pilot screen of 1200 FDA-approved drugs identified four compounds that specifically reduced the FP signal by at least three standard deviations from the untreated controls. Secondary assays showed that one of these hit compounds, the antithrombotic drug dipyridamole, enhances ABL kinase activity in vitro to a greater extent than the previously described ABL agonist, DPH. Docking studies predicted that this compound binds to a pocket formed at the interface of the SH3 domain and the linker, suggesting that it activates ABL by disrupting this regulatory interaction. These results show that screening assays based on the non-catalytic domains of ABL can identify allosteric small molecule regulators of kinase function, providing a new approach to selective drug discovery for this important kinase system.
Collapse
Affiliation(s)
- Prerna Grover
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Haibin Shi
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Matthew Baumgartner
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Carlos J. Camacho
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
| | - Thomas E. Smithgall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
45
|
Abstract
Tissue inhibitor of metalloproteinases-3 (TIMP-3) belongs to a family of proteins that regulate the activity of matrix metalloproteinases (MMPs), which can process various bioactive molecules such as cell surface receptors, chemokines, and cytokines. Glycosaminoglycans (GAGs) interact with a number of proteins, thereby playing an essential role in the regulation of many physiological/patho-physiological processes. Both GAGs and TIMP/MMPs play a major role in many cell biological processes, including cell proliferation, migration, differentiation, angiogenesis, apoptosis, and host defense. In this report, a heparin biosensor was used to map the interaction between TIMP-3 and heparin and other GAGs by surface plasmon resonance spectroscopy. These studies show that TIMP-3 is a heparin-binding protein with an affinity of ~59 nM. Competition surface plasmon resonance analysis indicates that the interaction between TIMP-3 and heparin is chain-length dependent, and N-sulfo and 6-O-sulfo groups (rather than the 2-O-sulfo groups) in heparin are important in the interaction of heparin with TIMP-3. Other GAGs (including chondroitin sulfate (CS) type E (CS-E)and CS type B (CS-B)demonstrated strong binding to TIMP-3, while heparan sulfate (HS), CS type A (CSA), CS type C (CSC), and CS type D (CSD) displayed only weak binding affinity.
Collapse
|
46
|
Arghir I, Delport F, Spasic D, Lammertyn J. Smart design of fiber optic surfaces for improved plasmonic biosensing. N Biotechnol 2015; 32:473-84. [PMID: 25858811 DOI: 10.1016/j.nbt.2015.03.012] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2014] [Revised: 03/04/2015] [Accepted: 03/22/2015] [Indexed: 12/19/2022]
Abstract
Although the phenomenon of surface plasmon resonance (SPR) is known for more than a century now, traditional prism-based SPR platforms have hardly escaped the research laboratories despite being recognized for the sensitive and specific performance. Significant efforts have been made over the last years to overcome their existing limitations by coupling the SPR phenomenon to the fiber optic (FO) technology. While this platform has been promoted as cost-effective and simpler alternative capable of handling label-free bioassays, quantification and real-time monitoring of biomolecular interactions, examples of its applicability in sensing and biosensing remain to date very limited. The FO-SPR system is still in development and requires further advancements for reaching the stability and sensitivity of the benchmark SPR systems. Among existing strategies for device improvement, those based on modifying the FO tips using nanomaterials are mostly studied. These small-scale objects provide a wide range of possibilities for alternating the architecture of the FO sensitive zone, enabling also unique effects such as localized SPR (LSPR). This mini-review summarizes the latest innovations in the fabrication procedures which use nanoparticles or other nanomaterials, aiming at FO-SPR technology performance improvements, as well as addition of new device features and functionalities.
Collapse
Affiliation(s)
- Iulia Arghir
- KU Leuven, Department of Biosystems, MeBioS-Biosensor Group, Willem de Croylaan 42, 3000 Leuven, Belgium
| | - Filip Delport
- KU Leuven, Department of Biosystems, MeBioS-Biosensor Group, Willem de Croylaan 42, 3000 Leuven, Belgium
| | - Dragana Spasic
- KU Leuven, Department of Biosystems, MeBioS-Biosensor Group, Willem de Croylaan 42, 3000 Leuven, Belgium
| | - Jeroen Lammertyn
- KU Leuven, Department of Biosystems, MeBioS-Biosensor Group, Willem de Croylaan 42, 3000 Leuven, Belgium.
| |
Collapse
|
47
|
Davidoff SN, Ditto NT, Brooks AE, Eckman J, Brooks BD. Surface Plasmon Resonance for Therapeutic Antibody Characterization. ACTA ACUST UNITED AC 2015. [DOI: 10.1007/978-1-4939-2617-6_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
48
|
Comparing the measured affinity of 111In-labeled ligands for cellular receptors by monitoring gamma, beta, or X-ray radiation with three different LigandTracer® devices. J Radioanal Nucl Chem 2014. [DOI: 10.1007/s10967-014-3875-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
49
|
Chu R, Reczek D, Brondyk W. Capture-stabilize approach for membrane protein SPR assays. Sci Rep 2014; 4:7360. [PMID: 25484112 PMCID: PMC5154539 DOI: 10.1038/srep07360] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 11/19/2014] [Indexed: 02/01/2023] Open
Abstract
Measuring the binding kinetics of antibodies to intact membrane proteins by surface plasmon resonance has been challenging largely because of the inherent difficulties in capturing membrane proteins on chip surfaces while retaining their native conformation. Here we describe a method in which His-tagged CXCR5, a GPCR, was purified and captured on a Biacore chip surface via the affinity tag. The captured receptor protein was then stabilized on the chip surface by limited cross-linking. The resulting chip surface retained ligand binding activity and was used for monoclonal antibody kinetics assays by a standard Biacore kinetics assay method with a simple low pH regeneration step. We demonstrate the advantages of this whole receptor assay when compared to available peptide-based binding assays. We further extended the application of the capture-stabilize approach to virus-like particles and demonstrated its utility analyzing antibodies against CD52, a GPI-anchored protein, in its native membrane environment. The results are the first demonstration of chemically stabilized chip surfaces for membrane protein SPR assays.
Collapse
Affiliation(s)
- Ruiyin Chu
- Genzyme Corporation, 49 New York Avenue, Framingham, MA 01701, USA
| | - David Reczek
- Genzyme Corporation, 49 New York Avenue, Framingham, MA 01701, USA
| | - William Brondyk
- Genzyme Corporation, 49 New York Avenue, Framingham, MA 01701, USA
| |
Collapse
|
50
|
Neutralization of Virus Infectivity by Antibodies: Old Problems in New Perspectives. ACTA ACUST UNITED AC 2014; 2014. [PMID: 27099867 DOI: 10.1155/2014/157895] [Citation(s) in RCA: 160] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Neutralizing antibodies (NAbs) can be both sufficient and necessary for protection against viral infections, although they sometimes act in concert with cellular immunity. Successful vaccines against viruses induce NAbs but vaccine candidates against some major viral pathogens, including HIV-1, have failed to induce potent and effective such responses. Theories of how antibodies neutralize virus infectivity have been formulated and experimentally tested since the 1930s; and controversies about the mechanistic and quantitative bases for neutralization have continually arisen. Soluble versions of native oligomeric viral proteins that mimic the functional targets of neutralizing antibodies now allow the measurement of the relevant affinities of NAbs. Thereby the neutralizing occupancies on virions can be estimated and related to the potency of the NAbs. Furthermore, the kinetics and stoichiometry of NAb binding can be compared with neutralizing efficacy. Recently, the fundamental discovery that the intracellular factor TRIM21 determines the degree of neutralization of adenovirus has provided new mechanistic and quantitative insights. Since TRIM21 resides in the cytoplasm, it would not affect the neutralization of enveloped viruses, but its range of activity against naked viruses will be important to uncover. These developments bring together the old problems of virus neutralization-mechanism, stoichiometry, kinetics, and efficacy-from surprising new angles.
Collapse
|