1
|
Mata Calidonio J, Maddox AI, Hamad-Schifferli K. A novel immunoassay technique using principal component analysis for enhanced detection of emerging viral variants. LAB ON A CHIP 2024; 24:3985-3995. [PMID: 39046406 DOI: 10.1039/d4lc00505h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/25/2024]
Abstract
Rapid diagnostics are critical infectious disease tools that are designed to detect a known biomarker using antibodies specific to that biomarker. However, a way to detect unknown disease variants has not yet been achieved in a paper test format. We describe here a route to make an adaptable paper immunoassay that can detect an unknown biomarker, demonstrating it on SARS-CoV-2 variants. The immunoassay repurposes cross reactive antibodies raised against the alpha variant. Gold nanoparticles of two different colors conjugated to two different antibodies create a colorimetric signal, and machine learning of the resulting colorimetric pattern is used to train the assay to discriminate between variants of alpha and Omicron BA.5. By using principal component analysis, the colorimetric test patterns can pick up and discriminate an unknown variant that it has not encountered before, Omicron BA.1. The test has an accuracy of 100% and a potential calculated discriminatory power of 900. We show that it can be used adaptively and that it can be used to pick up emerging variants without the need to raise new antibodies.
Collapse
Affiliation(s)
| | - Arianna I Maddox
- Department of Biology, University of Massachusetts Boston, Boston, MA, USA
| | - Kimberly Hamad-Schifferli
- Department of Engineering, University of Massachusetts Boston, Boston, MA, USA.
- School for the Environment, University of Massachusetts Boston, Boston, MA, USA
| |
Collapse
|
2
|
Breausche F, Somerlot A, Walder J, Osei K, Okyem S, Driskell JD. Immobilization of Thiol-Modified Horseradish Peroxidase on Gold Nanoparticles Enhances Enzyme Stability and Prevents Proteolytic Digestion. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:13957-13967. [PMID: 38919992 PMCID: PMC11238584 DOI: 10.1021/acs.langmuir.4c01180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 05/20/2024] [Accepted: 06/18/2024] [Indexed: 06/27/2024]
Abstract
The specificity and efficiency of enzyme-mediated reactions have the potential to positively impact many biotechnologies; however, many enzymes are easily degraded. Immobilization on a solid support has recently been explored to improve enzyme stability. This study aims to gain insights and facilitate enzyme adsorption onto gold nanoparticles (AuNPs) to form a stable bioconjugate through the installation of thiol functional groups that alter the protein chemistry. In specific, the model enzyme, horseradish peroxidase (HRP), is thiolated via Traut's reagent to increase the robustness and enzymatic activity of the bioconjugate. This study compares HRP and its thiolated analog (THRP) to deduce the impact of thiolation and AuNP-immobilization on the enzyme activity and stability. HRP, THRP, and their corresponding bioconjugates, HRP-AuNP and THRP-AuNP, were analyzed via UV-vis spectrophotometry, circular dichroism, zeta potential, and enzyme-substrate kinetics assays. Our data show a 5-fold greater adsorption for THRP on the AuNP, in comparison to HRP, that translated to a 5-fold increase in the THRP-AuNP bioconjugate activity. The thiolated and immobilized HRP exhibited a substantial improvement in stability at elevated temperatures (50 °C) and storage times (1 month) relative to the native enzyme in solution. Moreover, HRP, THRP, and their bioconjugates were incubated with trypsin to assess the susceptibility to proteolytic digestion. Our results demonstrate that THRP-AuNP bioconjugates maintain full enzymatic activity after 18 h of incubation with trypsin, whereas free HRP, free THRP, and HRP-AuNP conjugates are rendered inactive by trypsin treatment. These results highlight the potential for protein modification and immobilization to substantially extend enzyme shelf life, resist protease digestion, and enhance biological function to realize enzyme-enabled biotechnologies.
Collapse
Affiliation(s)
- Faith
E. Breausche
- Department
of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Annelise Somerlot
- Department
of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Jason Walder
- Department
of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Kwame Osei
- Department
of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Samuel Okyem
- Department
of Chemistry, University of Illinois at
Urbana−Champaign, Urbana, Illinois 61801, United States
| | - Jeremy D. Driskell
- Department
of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| |
Collapse
|
3
|
Goetz M, Thotathil N, Zhao Z, Mitragotri S. Vaccine adjuvants for infectious disease in the clinic. Bioeng Transl Med 2024; 9:e10663. [PMID: 39036089 PMCID: PMC11256182 DOI: 10.1002/btm2.10663] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/29/2024] [Accepted: 03/10/2024] [Indexed: 07/23/2024] Open
Abstract
Adjuvants, materials added to vaccines to enhance the resulting immune response, are important components of vaccination that are many times overlooked. While vaccines always include an antigen to tell the body what to vaccinate to, of equal importance the adjuvant provides the how, a significant factor in producing a complete response. The adjuvant space has been slow to develop with the first use of an adjuvant in a licensed vaccine occurring in the 1930s, and remaining the only adjuvant in licensed vaccines for the next 80 years. However, with vaccination at the forefront of protection against new and complex pathogens, it is important to consider all components when designing an effective vaccine. Here we summarize the adjuvant space in licensed vaccines as well as the novel adjuvant space in clinical trials with a specific focus on the materials utilized and their resulting impact on the immune response. We discuss five major categories of adjuvant materials: aluminum salts, nanoparticles, viral vectors, TLR agonists, and emulsions. For each category, we delve into the current clinical trials space, the impact of these materials on vaccination, as well as some of the ways in which they could be improved. Adjuvants present an exciting opportunity to improve vaccine responses and stability, this review will help inform about the current progress of this space. Translational impact statement In the aftermath of the COVID-19 pandemic, vaccines for infectious diseases have come into the spotlight. While antigens have always been an important focus of vaccine design, the adjuvant is a significant tool for enhancing the immune response to the vaccine that has been largely underdeveloped. This article provides a broad review of the history of adjuvants and, the current vaccine adjuvant space, and the progress seen in adjuvants in clinical trials. There is specific emphasis on the material landscape for adjuvants and their resulting mechanism of action. Looking ahead, while the novel vaccine adjuvant space features exciting new technologies and materials, there is still a need for more to meet the protective needs of new and complex pathogens.
Collapse
Affiliation(s)
- Morgan Goetz
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| | - Naaz Thotathil
- University of Massachusetts AmherstAmherstMassachusettsUSA
| | - Zongmin Zhao
- Department of Pharmaceutical SciencesCollege of Pharmacy, University of Illinois ChicagoChicagoIllinoisUSA
| | - Samir Mitragotri
- John A Paulson School of Engineering & Applied SciencesHarvard UniversityAllstonMassachusettsUSA
- Wyss Institute of Biologically Inspired EngineeringBostonMassachusettsUSA
| |
Collapse
|
4
|
Echeverri D, Calucho E, Marrugo-Ramírez J, Álvarez-Diduk R, Orozco J, Merkoçi A. Capacitive immunosensing at gold nanoparticle-decorated reduced graphene oxide electrodes fabricated by one-step laser nanostructuration. Biosens Bioelectron 2024; 252:116142. [PMID: 38401281 DOI: 10.1016/j.bios.2024.116142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/01/2024] [Accepted: 02/18/2024] [Indexed: 02/26/2024]
Abstract
Nanostructured electrochemical biosensors have ushered in a new era of diagnostic precision, offering enhanced sensitivity and specificity for clinical biomarker detection. Among them, capacitive biosensing enables ultrasensitive label-free detection of multiple molecular targets. However, the complexity and cost associated with conventional fabrication methods of nanostructured platforms hinder the widespread adoption of these devices. This study introduces a capacitive biosensor that leverages laser-engraved reduced graphene oxide (rGO) electrodes decorated with gold nanoparticles (AuNPs). The fabrication involves laser-scribed GO-Au3+ films, yielding rGO-AuNP electrodes, seamlessly transferred onto a PET substrate via a press-stamping methodology. These electrodes have a remarkable affinity for biomolecular recognition after being functionalized with specific bioreceptors. For example, initial studies with human IgG antibodies confirm the detection capabilities of the biosensor using electrochemical capacitance spectroscopy. Furthermore, the biosensor can quantify CA-19-9 glycoprotein, a clinical cancer biomarker. The biosensor exhibits a dynamic range from 0 to 300 U mL-1, with a limit of detection of 8.9 U mL-1. Rigorous testing with known concentrations of a pretreated CA-19-9 antigen from human fluids confirmed their accuracy and reliability in detecting the glycoprotein. This study signifies notable progress in capacitive biosensing for clinical biomarkers, potentially leading to more accessible and cost-effective point-of-care solutions.
Collapse
Affiliation(s)
- Danilo Echeverri
- Nanobioelectronics & Biosensors Group, Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and the Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193, Barcelona, Spain; Max Planck Tandem Group in Nanobioengineering, Institute of Chemistry, Faculty of Natural and Exact Sciences, University of Antioquia, Complejo Ruta N, Calle 67 N° 52-20, 050010, Medellín, Colombia
| | - Enric Calucho
- Nanobioelectronics & Biosensors Group, Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and the Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Bellaterra, 08193, Barcelona, Spain
| | - Jose Marrugo-Ramírez
- Nanobioelectronics & Biosensors Group, Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and the Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Bellaterra, 08193, Barcelona, Spain
| | - Ruslán Álvarez-Diduk
- Nanobioelectronics & Biosensors Group, Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and the Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193, Barcelona, Spain.
| | - Jahir Orozco
- Max Planck Tandem Group in Nanobioengineering, Institute of Chemistry, Faculty of Natural and Exact Sciences, University of Antioquia, Complejo Ruta N, Calle 67 N° 52-20, 050010, Medellín, Colombia.
| | - Arben Merkoçi
- Nanobioelectronics & Biosensors Group, Institut Català de Nanociència i Nanotecnologia (ICN2), CSIC and the Barcelona Institute of Science and Technology (BIST), Campus UAB, Bellaterra, 08193, Barcelona, Spain; ICREA Institució Catalana de Recerca i Estudis Avançats, Passeig de Lluís Companys, 23, 08010, Barcelona, Spain.
| |
Collapse
|
5
|
Steegmüller T, Kratky T, Gollwitzer L, Schwaminger SP, Berensmeier S. Development of a New Affinity Gold Polymer Membrane with Immobilized Protein A. MEMBRANES 2024; 14:31. [PMID: 38392658 PMCID: PMC10890041 DOI: 10.3390/membranes14020031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/08/2024] [Accepted: 01/19/2024] [Indexed: 02/24/2024]
Abstract
New and highly selective stationary phases for affinity membrane chromatography have the potential to significantly enhance the efficiency and specificity of therapeutic protein purification by reduced mass transfer limitations. This work developed and compared different immobilization strategies for recombinant Protein A ligands to a gold-sputtered polymer membrane for antibody separation in terms of functionalization and immobilization success, protein load, and stability. Successful, functionalization was validated via X-ray photoelectron spectroscopy (XPS). Here, a recombinant Protein A ligand was coupled by N-hydroxysuccinimide (NHS)/N-(3-dimethylaminopropyl)-N'-ethylcarbodiimide (EDC) chemistry to carboxy-functionalized, gold-sputtered membranes. We achieved a binding capacity of up to 104 ± 17 mg of the protein ligand per gram of the gold-sputtered membrane. The developed membranes were able to successfully capture and release the monoclonal antibody (mAb) Trastuzumab, as well as antibodies from fresh frozen human blood plasma in both static and dynamic setups. Therefore, they demonstrated successful functionalization and immobilization strategies. The antibody load was tested using bicinchoninic acid (BCA), ultraviolet-visible spectroscopy (UV-vis) measurements, and sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE). The outcome is a fully functional affinity membrane that can be implemented in a variety of different antibody purification processes, eliminating the need for creating individualized strategies for modifying the surface to suit different substrates or conditions.
Collapse
Affiliation(s)
- Tobias Steegmüller
- Chair of Bioseparation Engineering, TUM School of Engineering and Design, Technical University of Munich, Boltzmannstraße 15, 85748 Garching, Germany
| | - Tim Kratky
- Associate Professorship Physical Chemistry with Focus on Catalysis, TUM School of Natural Sciences, Technical University of Munich, Lichtenbergstraße 4, 85748 Garching, Germany
| | - Lena Gollwitzer
- Chair of Bioseparation Engineering, TUM School of Engineering and Design, Technical University of Munich, Boltzmannstraße 15, 85748 Garching, Germany
| | - Sebastian Patrick Schwaminger
- Chair of Bioseparation Engineering, TUM School of Engineering and Design, Technical University of Munich, Boltzmannstraße 15, 85748 Garching, Germany
- Division of Medicinal Chemistry, Otto-Loewi Research Center, Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
- BioTechMed-Graz, Mozartgasse 12, 8010 Graz, Austria
| | - Sonja Berensmeier
- Chair of Bioseparation Engineering, TUM School of Engineering and Design, Technical University of Munich, Boltzmannstraße 15, 85748 Garching, Germany
- Munich Institute of Integrated Materials, Energy and Process Engineering, Technical University of Munich, Lichtenbergstraße 4a, 85748 Garching, Germany
| |
Collapse
|
6
|
Ebbah E, Amissah A, Kim JH, Driskell JD. SERS-based immunoassay on a plasmonic syringe filter for improved sampling and labeling efficiency of biomarkers. Analyst 2023; 149:221-230. [PMID: 38018888 DOI: 10.1039/d3an01899g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2023]
Abstract
Rapid, sensitive, and quantitative detection of biomarkers is needed for early diagnosis of disease and surveillance of infectious outbreaks. Here, we exploit a plasmonic syringe filter and surface-enhanced Raman spectroscopy (SERS) in the development of a rapid detection system, using human IgG as a model diagnostic biomarker. The novel assay design facilitates multiple passages of the sample and labeling solution through the detection zone enabling us to investigate and maximize sampling efficiency to the capture substrate. The vertical flow immunoassay process in this study involves the utilization of filter paper embedded with gold nanoparticles (AuNPs) to form a plasmonic substrate. Capture antibody (anti-human IgG) is then immobilized onto the prepared plasmonic paper and inserted into a vertical flow device (syringe filter holder). Sample solution is passed through the filter paper and the target antigen (human IgG) is selectively captured by the immobilized antibody to form an antibody-antigen complex. Next, functionalized AuNPs as extrinsic Raman labels (ERLs) are passed through the filter paper to label the captured biomarker molecules forming a layered structure. This sandwiched geometry enhances plasmonic coupling and SERS signal to provide highly sensitive detection of biomolecules. Systematic studies to investigate the impact of multiple infuse/withdraw cycles of the sample and labeling solutions reveal that antigen and ERL binding are maximized with 10 and 20 cycles, respectively. The optimized assay achieves a detection limit of ∼0.2 ng mL-1 for human IgG with a total assay time of less than 5 minutes, meeting the demands for rapid point of care diagnostics. Additionally, the optimized platform was implemented in the quantitative analysis of the SARS-CoV-2 nucleocapsid protein, the typical target in commercial, FDA-approved rapid antigen tests for COVID-19.
Collapse
Affiliation(s)
- Eunice Ebbah
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| | - Anthony Amissah
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| | - Jun-Hyun Kim
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| | - Jeremy D Driskell
- Department of Chemistry, Illinois State University, Normal, IL 61790, USA.
| |
Collapse
|
7
|
Dégardin M, Liberelle B, Oliverio R, Baniahmad SF, Darviot C, Largillière I, Henry O, Durocher Y, Banquy X, Meunier M, De Crescenzo G. Coiled-Coil-Based Biofunctionalization of 100 nm Gold Nanoparticles with the Trastuzumab Antibody for the Detection of HER2-Positive Cancer Cells. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:12235-12247. [PMID: 37581531 DOI: 10.1021/acs.langmuir.3c01621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/16/2023]
Abstract
We compared different biofunctionalization strategies for immobilizing trastuzumab, an IgG targeting the HER2 biomarker, onto 100 nm spherical gold nanoparticles because of the E/K coiled-coil peptide heterodimer. First, Kcoil peptides were grafted onto the gold surface while their Ecoil partners were genetically encoded at the C-terminus of trastuzumab's Fc region, allowing for a strong and specific interaction between the antibodies and the nanoparticles. Gold nanoparticles with no Kcoil peptides on their surface were also produced to immobilize Ecoil-tagged trastuzumab antibodies via the specific adsorption of their negatively charged Ecoil tags on the positively charged gold surface. Finally, the nonspecific adsorption of wild-type trastuzumab on the gold surface was also assessed, with and without Kcoil peptides grafted on it beforehand. We developed a thorough workflow to systematically compare the immobilization strategies regarding the stability of nanoparticles, antibody coverage, and ability to specifically bind to HER2-positive breast cancer cells. All nanoparticles were highly monodisperse and retained their localized surface plasmon resonance properties after biofunctionalization. A significant increase in the amount of immobilized antibodies was observed with the two oriented coil-based strategies compared to nonspecific adsorption. Finally, all biofunctionalization strategies allowed for the detection of HER2-positive breast cancer cells, but among the investigated approaches, we recommend using the E/K coiled-coil-based strategy for gold nanoparticle biofunctionalization because it allows for the qualitative and quantitative detection of HER2-positive cells with a higher contrast compared to HER2-negative cells.
Collapse
Affiliation(s)
- Médéric Dégardin
- Department of Chemical Engineering, Polytechnique Montréal, H3T 1J4 Montréal, Québec, Canada
- Laser Processing and Plasmonics Laboratory (LP2L), Department of Engineering Physics, Polytechnique Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Benoit Liberelle
- Department of Chemical Engineering, Polytechnique Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Romane Oliverio
- Department of Chemical Engineering, Polytechnique Montréal, H3T 1J4 Montréal, Québec, Canada
- Faculty of Pharmacy, Axe Formulation et Analyse du Médicament (AFAM), Université de Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Seyed Farzad Baniahmad
- Human Health Therapeutics Research Centre, National Research Council of Canada, Building Montréal-Royalmount, H4P 2R2 Montréal, Québec, Canada
| | - Cécile Darviot
- Laser Processing and Plasmonics Laboratory (LP2L), Department of Engineering Physics, Polytechnique Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Isabelle Largillière
- Laser Processing and Plasmonics Laboratory (LP2L), Department of Engineering Physics, Polytechnique Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Olivier Henry
- Department of Chemical Engineering, Polytechnique Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Yves Durocher
- Human Health Therapeutics Research Centre, National Research Council of Canada, Building Montréal-Royalmount, H4P 2R2 Montréal, Québec, Canada
| | - Xavier Banquy
- Faculty of Pharmacy, Axe Formulation et Analyse du Médicament (AFAM), Université de Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Michel Meunier
- Laser Processing and Plasmonics Laboratory (LP2L), Department of Engineering Physics, Polytechnique Montréal, H3T 1J4 Montréal, Québec, Canada
| | - Gregory De Crescenzo
- Department of Chemical Engineering, Polytechnique Montréal, H3T 1J4 Montréal, Québec, Canada
| |
Collapse
|
8
|
Andrzejewska W, Peplińska B, Litowczenko J, Obstarczyk P, Olesiak-Bańska J, Jurga S, Lewandowski M. SARS-CoV-2 Virus-like Particles with Plasmonic Au Cores and S1-Spike Protein Coronas. ACS Synth Biol 2023; 12:2320-2328. [PMID: 37449651 PMCID: PMC10443039 DOI: 10.1021/acssynbio.3c00133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Indexed: 07/18/2023]
Abstract
The COVID-19 pandemic has stimulated the scientific world to intensify virus-related studies aimed at the development of quick and safe ways of detecting viruses in the human body, studying the virus-antibody and virus-cell interactions, and designing nanocarriers for targeted antiviral therapies. However, research on dangerous viruses can only be performed in certified laboratories that follow strict safety procedures. Thus, developing deactivated virus constructs or safe-to-use virus-like objects, which imitate real viruses and allow performing virus-related studies in any research laboratory, constitutes an important scientific challenge. Such species, called virus-like particles (VLPs), contain instead of capsids with viral DNA/RNA empty or synthetic cores with real virus proteins attached to them. We have developed a method for the preparation of VLPs imitating the virus responsible for the COVID-19 disease: the SARS-CoV-2. The particles have Au cores surrounded by "coronas" of S1 domains of the virus's spike protein. Importantly, they are safe to use and specifically interact with SARS-CoV-2 antibodies. Moreover, Au cores exhibit localized surface plasmon resonance (LSPR), which makes the synthesized VLPs suitable for biosensing applications. During the studies, the effect allowed us to visualize the interaction between the VLPs and the antibodies and identify the characteristic vibrational signals. What is more, additional functionalization of the particles with a fluorescent label revealed their potential in studying specific virus-related interactions. Notably, the universal character of the developed synthesis method makes it potentially applicable for fabricating VLPs imitating other life-threatening viruses.
Collapse
Affiliation(s)
- Weronika Andrzejewska
- NanoBioMedical
Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland
| | - Barbara Peplińska
- NanoBioMedical
Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland
| | - Jagoda Litowczenko
- NanoBioMedical
Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland
| | - Patryk Obstarczyk
- Institute
of Advanced Materials, Wroclaw University
of Science and Technology, Wybrzeże Wyspiańskiego 2, 50-370 Wrocław, Poland
| | - Joanna Olesiak-Bańska
- Institute
of Advanced Materials, Wroclaw University
of Science and Technology, Wybrzeże Wyspiańskiego 2, 50-370 Wrocław, Poland
| | - Stefan Jurga
- NanoBioMedical
Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland
| | - Mikołaj Lewandowski
- NanoBioMedical
Centre, Adam Mickiewicz University, Wszechnicy Piastowskiej 3, 61-614 Poznań, Poland
| |
Collapse
|
9
|
Ning S, Chang HC, Fan KC, Hsiao PY, Feng C, Shoemaker D, Chen RT. A point-of-care biosensor for rapid detection and differentiation of COVID-19 virus (SARS-CoV-2) and influenza virus using subwavelength grating micro-ring resonator. APPLIED PHYSICS REVIEWS 2023; 10:021410. [PMID: 37265478 PMCID: PMC10228026 DOI: 10.1063/5.0146079] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 05/05/2023] [Indexed: 06/03/2023]
Abstract
In the context of continued spread of coronavirus disease 2019 (COVID-19) caused by SARS-CoV-2 and the emergence of new variants, the demand for rapid, accurate, and frequent detection is increasing. Moreover, the new predominant strain, Omicron variant, manifests more similar clinical features to those of other common respiratory infections. The concurrent detection of multiple potential pathogens helps distinguish SARS-CoV-2 infection from other diseases with overlapping symptoms, which is significant for providing tailored treatment to patients and containing the outbreak. Here, we report a lab-on-a-chip biosensing platform for SARS-CoV-2 detection based on the subwavelength grating micro-ring resonator. The sensing surface is functionalized by specific antibody against SARS-CoV-2 spike protein, which could produce redshifts of resonant peaks by antigen-antibody combination, thus achieving quantitative detection. Additionally, the sensor chip is integrated with a microfluidic chip featuring an anti-backflow Y-shaped structure that enables the concurrent detection of two analytes. In this study, we realized the detection and differentiation of COVID-19 and influenza A H1N1. Experimental results indicate that the limit of detection of our device reaches 100 fg/ml (1.31 fM) within 15 min detecting time, and cross-reactivity tests manifest the specificity of the optical diagnostic assay. Furthermore, the integrated packaging and streamlined workflow facilitate its use for clinical applications. Thus, the biosensing platform presents a promising approach for attaining highly sensitive, selective, multiplexed, and quantitative point-of-care diagnosis and distinction between COVID-19 and influenza.
Collapse
Affiliation(s)
- Shupeng Ning
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas 78758, USA
| | - Hao-Chen Chang
- Omega Optics, Inc., 8500 Shoal Creek Blvd., Austin, Texas 78757, USA
| | - Kang-Chieh Fan
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas 78758, USA
| | - Po-Yu Hsiao
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas 78758, USA
| | - Chenghao Feng
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas 78758, USA
| | - Devan Shoemaker
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas 78758, USA
| | - Ray T. Chen
- Author to whom correspondence should be addressed:
| |
Collapse
|
10
|
Lee JH, Chapman DV, Saltzman WM. Nanoparticle Targeting with Antibodies in the Central Nervous System. BME FRONTIERS 2023; 4:0012. [PMID: 37849659 PMCID: PMC10085254 DOI: 10.34133/bmef.0012] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/19/2023] [Indexed: 10/19/2023] Open
Abstract
Treatments for disease in the central nervous system (CNS) are limited because of difficulties in agent penetration through the blood-brain barrier, achieving optimal dosing, and mitigating off-target effects. The prospect of precision medicine in CNS treatment suggests an opportunity for therapeutic nanotechnology, which offers tunability and adaptability to address specific diseases as well as targetability when combined with antibodies (Abs). Here, we review the strategies to attach Abs to nanoparticles (NPs), including conventional approaches of chemisorption and physisorption as well as attempts to combine irreversible Ab immobilization with controlled orientation. We also summarize trends that have been observed through studies of systemically delivered Ab-NP conjugates in animals. Finally, we discuss the future outlook for Ab-NPs to deliver therapeutics into the CNS.
Collapse
Affiliation(s)
| | | | - W. Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
| |
Collapse
|
11
|
Ogunlusi T, Driskell JD. Controlled Temporal Release of Serum Albumin Immobilized on Gold Nanoparticles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2023; 39:3720-3728. [PMID: 36857653 DOI: 10.1021/acs.langmuir.2c03429] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Proteins adsorbed to gold nanoparticles (AuNPs) form bioconjugates and are critical to many emerging technologies for drug delivery, diagnostics, therapies, and other biomedical applications. A thorough understanding of the interaction between the immobilized protein and AuNP is essential for the bioconjugate to perform as designed. Here, we explore a correlation between the number of solvent-accessible thiol groups on a protein and the protein desorption rate from the AuNP surface in the presence of a competing protein. The chemical modification of human serum albumin (HSA) was carried out to install additional free thiols using Traut's reagent and create a library of HSA analogues by tailoring the molar excess of the Traut's reagent. We pre-adsorbed HSA variants onto the AuNP surface, and the resulting bioconjugates were then exposed to IgG antibody, and protein exchange was monitored as a function of time. We found that the rate of HSA displacement from the AuNP correlated with the experimentally measured number of accessible free thiol groups. Additionally, bioconjugates were synthesized using thiolated analogues of bovine serum albumin (BSA) and suspended in serum as a model for a complex sample matrix. Similarly, desorption rates with serum proteins were modulated with solvent-accessible thiols on the immobilized protein. These results further highlight the key role of Au-S bonds in the formation of protein-AuNP conjugates and provide a pathway to systematically control the number of free thiols on a protein, enabling the controlled release of protein from the surface of AuNP.
Collapse
Affiliation(s)
- Tosin Ogunlusi
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Jeremy D Driskell
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| |
Collapse
|
12
|
Didier CM, Orrico JF, Cepeda Torres OS, Castro JM, Baksh A, Rajaraman S. Microfabricated polymer-metal biosensors for multifarious data collection from electrogenic cellular models. MICROSYSTEMS & NANOENGINEERING 2023; 9:22. [PMID: 36875634 PMCID: PMC9974480 DOI: 10.1038/s41378-023-00488-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 12/19/2022] [Accepted: 01/09/2023] [Indexed: 05/28/2023]
Abstract
Benchtop tissue cultures have become increasingly complex in recent years, as more on-a-chip biological technologies, such as microphysiological systems (MPS), are developed to incorporate cellular constructs that more accurately represent their respective biological systems. Such MPS have begun facilitating major breakthroughs in biological research and are poised to shape the field in the coming decades. These biological systems require integrated sensing modalities to procure complex, multiplexed datasets with unprecedented combinatorial biological detail. In this work, we expanded upon our polymer-metal biosensor approach by demonstrating a facile technology for compound biosensing that was characterized through custom modeling approaches. As reported herein, we developed a compound chip with 3D microelectrodes, 3D microfluidics, interdigitated electrodes (IDEs) and a microheater. The chip was subsequently tested using the electrical/electrochemical characterization of 3D microelectrodes with 1 kHz impedance and phase recordings and IDE-based high-frequency (~1 MHz frequencies) impedimetric analysis of differential localized temperature recordings, both of which were modeled through equivalent electrical circuits for process parameter extraction. Additionally, a simplified antibody-conjugation strategy was employed for a similar IDE-based analysis of the implications of a key analyte (l-glutamine) binding to the equivalent electrical circuit. Finally, acute microfluidic perfusion modeling was performed to demonstrate the ease of microfluidics integration into such a polymer-metal biosensor platform for potential complimentary localized chemical stimulation. Overall, our work demonstrates the design, development, and characterization of an accessibly designed polymer-metal compound biosensor for electrogenic cellular constructs to facilitate comprehensive MPS data collection.
Collapse
Affiliation(s)
- Charles M. Didier
- NanoScience Technology Center, University of Central Florida, 4353 Scorpius Street, Research I, Suite 231, FL 32816 Orlando, USA
- Burnett School of Biomedical Sciences, University of Central Florida, 6900 Lake Nona Blvd, FL 32827 Orlando, USA
| | - Julia F. Orrico
- NanoScience Technology Center, University of Central Florida, 4353 Scorpius Street, Research I, Suite 231, FL 32816 Orlando, USA
| | - Omar S. Cepeda Torres
- NanoScience Technology Center, University of Central Florida, 4353 Scorpius Street, Research I, Suite 231, FL 32816 Orlando, USA
- Department of Biomedical Engineering, Polytechnic University of Puerto Rico, 377, 00918, Ponce de Leon, San Juan, Puerto Rico
| | - Jorge Manrique Castro
- NanoScience Technology Center, University of Central Florida, 4353 Scorpius Street, Research I, Suite 231, FL 32816 Orlando, USA
- Department of Electrical and Computer Engineering, University of Central Florida, 4238 Scorpius Street, FL 32816 Orlando, USA
| | - Aliyah Baksh
- NanoScience Technology Center, University of Central Florida, 4353 Scorpius Street, Research I, Suite 231, FL 32816 Orlando, USA
| | - Swaminathan Rajaraman
- NanoScience Technology Center, University of Central Florida, 4353 Scorpius Street, Research I, Suite 231, FL 32816 Orlando, USA
- Burnett School of Biomedical Sciences, University of Central Florida, 6900 Lake Nona Blvd, FL 32827 Orlando, USA
- Department of Electrical and Computer Engineering, University of Central Florida, 4238 Scorpius Street, FL 32816 Orlando, USA
- Department of Materials Science and Engineering, University of Central Florida, 12760 Pegasus Drive, Engineering I, Suite 207, FL 32816 Orlando, USA
| |
Collapse
|
13
|
Sarkis M, Minassian G, Mitri N, Rahme K, Fracasso G, El Hage R, Ghanem E. D2B-Functionalized Gold Nanoparticles: Promising Vehicles for Targeted Drug Delivery to Prostate Cancer. ACS APPLIED BIO MATERIALS 2023; 6:819-827. [PMID: 36755401 PMCID: PMC9945086 DOI: 10.1021/acsabm.2c00975] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Despite the multitude of therapeutic agents available to treat prostate cancer (PC), there are still no effective and safe measures to treat the tumor. It remains a challenge to develop a simple approach to target PC with specific antibodies. In our study, D2B monoclonal antibodies against a prostate-specific membrane antigen (PSMA) were used. We investigated the functionalization of gold nanoparticles (AuNPs) with D2B to generate favorable physicochemical and biological properties that mediate specific binding to PC. For this purpose, AuNPs with a size of about 25 nm were synthesized in water using sodium citrate as a reducing and stabilizing agent and then coated with D2B. Major physicochemical properties of naked and D2B-coated AuNPs were investigated by ultraviolet-visible (UV-vis) spectroscopy, dynamic light scattering (DLS), and zeta potential measurements. The successful binding of D2B to AuNPs-citrate caused a 15 nm red shift in the UV-vis. This was assessed by DLS as an increase in zeta potential from ∼-45 to ∼-23 mV and in the size of AuNPs from ∼25 to ∼63 nm. Scanning electron microscopy confirmed the size shift of AuNPs, which was detected as an exterior organic layer of D2Bs surrounding each AuNP. Even at high exposure levels of the bioconjugates, PSMA-PC-3 cells exhibited minimal cytotoxicity. The specific and dose-dependent binding of AuNPs-D2B to PC-3-PSMA cells was validated by flow cytometry analysis. Our data provide effective drug delivery systems in PC theranostics.
Collapse
Affiliation(s)
- Monira Sarkis
- Department of Sciences, Notre Dame University-Louaize, 72 Zouk Mosbeh, Lebanon
| | - Georges Minassian
- Department of Sciences, Notre Dame University-Louaize, 72 Zouk Mosbeh, Lebanon
| | - Nadim Mitri
- Department of Sciences, Notre Dame University-Louaize, 72 Zouk Mosbeh, Lebanon
| | - Kamil Rahme
- Department of Sciences, Notre Dame University-Louaize, 72 Zouk Mosbeh, Lebanon.,School of Chemistry & AMBER Centre, University College Cork, T12 YN60 Cork, Ireland
| | - Giulio Fracasso
- Department of Medicine, University of Verona, I-37134 Verona, Italy
| | - Roland El Hage
- Laboratory of Physical Chemistry of Materials (LCPM), PR2N (EDST), Faculty of Sciences II, Lebanese University, Campus Fanar P.O. Box 90656, 1103 Beirut, Lebanon.,Polymers Composites and Hybrids (PCH), IMT Mines Ales, 30100 Ales, France
| | - Esther Ghanem
- Department of Sciences, Notre Dame University-Louaize, 72 Zouk Mosbeh, Lebanon.,biobank.cy-Center of Excellence in Biobanking and Biomedical Research, Molecular Medicine Research Center, University of Cyprus, 1678 Nicosia, Cyprus
| |
Collapse
|
14
|
Riley MB, Strandquist E, Weitzel CS, Driskell JD. Structure and activity of native and thiolated α-chymotrypsin adsorbed onto gold nanoparticles. Colloids Surf B Biointerfaces 2022; 220:112867. [PMID: 36182820 DOI: 10.1016/j.colsurfb.2022.112867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/16/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022]
Abstract
A detailed understanding of protein-nanoparticle interactions is critical to realize the full potential of bioconjugate-enabled technologies. Parameters that lead to conformational changes in protein structure upon adsorption must be identified and controlled to mitigate loss of biological function. We hypothesized that the installation of thiol functional groups on a protein will facilitate robust adsorption to gold nanoparticles (AuNPs) and prevent protein unfolding to achieve thermodynamic stability. Here we investigated the adsorption behavior of α-chymotrypsin (ChT) and a thiolated analog of α-chymotrypsin (T-ChT) with AuNPs. ChT, which does not present any free thiols, was modified with 2-iminothiolane (Traut's reagent) to synthesize T-ChT consisting of two free thiols. Protein adsorption to AuNPs was monitored with dynamic light scattering and UV-vis spectrophotometry, and fluorescence spectra were acquired to assess changes in protein structure induced by interaction with the AuNP. The biological function of ChT, T-ChT, and respective bioconjugates were compared using a colorimetric enzymatic assay. The thiolated analog exhibited a greater affinity for the AuNP than the unmodified ChT, as determined from adsorption isotherms. The ChT protein formed a soft protein corona in which the enzyme denatures with prolonged exposure to AuNPs and, subsequently, lost enzymatic function. Conversely, the T-ChT formed a robust hard corona on the AuNP and retained structure and function. These data support the hypothesis, provide further insight into protein-AuNP interactions, and identify a simple chemical approach to synthesize robust and functional conjugates.
Collapse
Affiliation(s)
- McKenzie B Riley
- Department of Chemistry, Illinois State University, Normal, IL 61790, United States
| | - Evan Strandquist
- Department of Chemistry, Illinois State University, Normal, IL 61790, United States
| | | | - Jeremy D Driskell
- Department of Chemistry, Illinois State University, Normal, IL 61790, United States.
| |
Collapse
|
15
|
Ligiero CBP, Fernandes TS, D'Amato DL, Gaspar FV, Duarte PS, Strauch MA, Fonseca JG, Meirelles LGR, Bento da Silva P, Azevedo RB, Aparecida de Souza Martins G, Archanjo BS, Buarque CD, Machado G, Percebom AM, Ronconi CM. Influence of particle size on the SARS-CoV-2 spike protein detection using IgG-capped gold nanoparticles and dynamic light scattering. MATERIALS TODAY. CHEMISTRY 2022; 25:100924. [PMID: 35475288 PMCID: PMC9023328 DOI: 10.1016/j.mtchem.2022.100924] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 03/27/2022] [Accepted: 03/29/2022] [Indexed: 05/03/2023]
Abstract
Due to the unprecedented and ongoing nature of the coronavirus outbreak, the development of rapid immunoassays to detect severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and its highly contagious variants is an important and challenging task. Here, we report the development of polyclonal antibody-functionalized spherical gold nanoparticle biosensors as well as the influence of the nanoparticle sizes on the immunoassay response to detect the SARS-CoV-2 spike protein by dynamic light scattering. By monitoring the increment in the hydrodynamic diameter (ΔDH) by dynamic light scattering measurements in the antigen-antibody interaction, SARS-CoV-2 S-protein can be detected in only 5 min. The larger the nanoparticles, the larger ΔDH in the presence of spike protein. From adsorption isotherm, the calculated binding constant (K D ) was 83 nM and the estimated limit of detection was 13 ng/mL (30 pM). The biosensor was stable up to 90 days at 4 °C. Therefore, the biosensor developed in this work could be potentially applied as a fast and sensible immunoassay to detect SARS-CoV-2 infection in patient samples.
Collapse
Affiliation(s)
- C B P Ligiero
- Departamento de Química Inorgânica, Universidade Federal Fluminense (UFF), Campus Do Valonguinho, Outeiro de São João Batista, S/n, 24020-141, Niterói, RJ, Brazil
| | - T S Fernandes
- Departamento de Química Inorgânica, Universidade Federal Fluminense (UFF), Campus Do Valonguinho, Outeiro de São João Batista, S/n, 24020-141, Niterói, RJ, Brazil
| | - D L D'Amato
- Departamento de Química Inorgânica, Universidade Federal Fluminense (UFF), Campus Do Valonguinho, Outeiro de São João Batista, S/n, 24020-141, Niterói, RJ, Brazil
| | - F V Gaspar
- Departamento de Química Inorgânica, Universidade Federal Fluminense (UFF), Campus Do Valonguinho, Outeiro de São João Batista, S/n, 24020-141, Niterói, RJ, Brazil
- Departamento de Química, Pontifícia Universidade Católica Do Rio de Janeiro (PUC-Rio), Rio de Janeiro, RJ, 22451-900, Brazil
| | - P S Duarte
- Departamento de Química, Pontifícia Universidade Católica Do Rio de Janeiro (PUC-Rio), Rio de Janeiro, RJ, 22451-900, Brazil
| | - M A Strauch
- Gerência de Desenvolvimento Tecnológico, Instituto Vital Brazil, Niterói, RJ, 24230-410, Brazil
| | - J G Fonseca
- Gerência de Desenvolvimento Tecnológico, Instituto Vital Brazil, Niterói, RJ, 24230-410, Brazil
| | - L G R Meirelles
- Fazenda Instituto Vital Brazil, Niterói, RJ, 24230-410, Brazil
| | - P Bento da Silva
- Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, 70910-900, Brazil
| | - R B Azevedo
- Departamento de Genética e Morfologia, Instituto de Ciências Biológicas, Universidade de Brasília, Brasília, 70910-900, Brazil
| | - G Aparecida de Souza Martins
- Programa de Pós-graduação Em Ciência e Tecnologia de Alimentos, Universidade Federal Do Tocantins, 77001-090, Brazil
| | - B S Archanjo
- Divisão de Metrologia de Materiais, Instituto Nacional de Metrologia, Qualidade e Tecnologia (Inmetro), Duque de Caxias, RJ, 25250-020, Brazil
| | - C D Buarque
- Departamento de Química, Pontifícia Universidade Católica Do Rio de Janeiro (PUC-Rio), Rio de Janeiro, RJ, 22451-900, Brazil
| | - G Machado
- Centro de Tecnologias Estratégicas Do Nordeste, Av. Prof. Luiz Freire 01, Recife, Pernambuco, 50740-540, Brazil
| | - A M Percebom
- Departamento de Química, Pontifícia Universidade Católica Do Rio de Janeiro (PUC-Rio), Rio de Janeiro, RJ, 22451-900, Brazil
| | - C M Ronconi
- Departamento de Química Inorgânica, Universidade Federal Fluminense (UFF), Campus Do Valonguinho, Outeiro de São João Batista, S/n, 24020-141, Niterói, RJ, Brazil
| |
Collapse
|
16
|
António M, Lima T, Vitorino R, Daniel-da-Silva AL. Label-free dynamic light scattering assay for C-reactive protein detection using magnetic nanoparticles. Anal Chim Acta 2022; 1222:340169. [DOI: 10.1016/j.aca.2022.340169] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 07/11/2022] [Accepted: 07/12/2022] [Indexed: 11/26/2022]
|
17
|
Cameron SJ, Sheng J, Hosseinian F, Willmore WG. Nanoparticle Effects on Stress Response Pathways and Nanoparticle-Protein Interactions. Int J Mol Sci 2022; 23:7962. [PMID: 35887304 PMCID: PMC9323783 DOI: 10.3390/ijms23147962] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) are increasingly used in a wide variety of applications and products; however, NPs may affect stress response pathways and interact with proteins in biological systems. This review article will provide an overview of the beneficial and detrimental effects of NPs on stress response pathways with a focus on NP-protein interactions. Depending upon the particular NP, experimental model system, and dose and exposure conditions, the introduction of NPs may have either positive or negative effects. Cellular processes such as the development of oxidative stress, the initiation of the inflammatory response, mitochondrial function, detoxification, and alterations to signaling pathways are all affected by the introduction of NPs. In terms of tissue-specific effects, the local microenvironment can have a profound effect on whether an NP is beneficial or harmful to cells. Interactions of NPs with metal-binding proteins (zinc, copper, iron and calcium) affect both their structure and function. This review will provide insights into the current knowledge of protein-based nanotoxicology and closely examines the targets of specific NPs.
Collapse
Affiliation(s)
- Shana J. Cameron
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - Jessica Sheng
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
| | - Farah Hosseinian
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - William G. Willmore
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
- Institute of Biochemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
18
|
Mendes JP, Coelho LCC, Jorge PAS, Pereira CM. Differential Refractometric Biosensor for Reliable Human IgG Detection: Proof of Concept. BIOSENSORS 2022; 12:515. [PMID: 35884318 PMCID: PMC9312733 DOI: 10.3390/bios12070515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/08/2022] [Accepted: 07/09/2022] [Indexed: 06/15/2023]
Abstract
A new sensing platform based on long-period fiber gratings (LPFGs) for direct, fast, and selective detection of human immunoglobulin G (IgG; Mw = 150 KDa) was developed and characterized. The transducer's high selectivity is based on the specific interaction of a molecularly imprinted polymer (MIPs) design for IgG detection. The sensing scheme is based on differential refractometric measurements, including a correction system based on a non-imprinted polymer (NIP)-coated LPFG, allowing reliable and more sensitive measurements, improving the rejection of false positives in around 30%. The molecular imprinted binding sites were performed on the surface of a LPFG with a sensitivity of about 130 nm/RIU and a FOM of 16 RIU-1. The low-cost and easy to build device was tested in a working range from 1 to 100 nmol/L, revealing a limit of detection (LOD) and a sensitivity of 0.25 nmol/L (0.037 µg/mL) and 0.057 nm.L/nmol, respectively. The sensor also successfully differentiates the target analyte from the other abundant elements that are present in the human blood plasma.
Collapse
Affiliation(s)
- João P. Mendes
- Centro de Investigação em Química UP (CIQUP)—Instituto de Ciências Moleculares (IMS), Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal; (J.P.M.); (C.M.P.)
- INESC TEC—Instituto de Engenharia de Sistemas e Computadores, Tecnologia e Ciência, Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal;
- Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal
| | - Luís C. C. Coelho
- INESC TEC—Instituto de Engenharia de Sistemas e Computadores, Tecnologia e Ciência, Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal;
- Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal
| | - Pedro A. S. Jorge
- INESC TEC—Instituto de Engenharia de Sistemas e Computadores, Tecnologia e Ciência, Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal;
- Departamento de Física e Astronomia, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal
| | - Carlos M. Pereira
- Centro de Investigação em Química UP (CIQUP)—Instituto de Ciências Moleculares (IMS), Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal; (J.P.M.); (C.M.P.)
- Departamento de Química e Bioquímica, Faculdade de Ciências da Universidade do Porto, Rua do Campo Alegre, 687, 4169-007 Porto, Portugal
| |
Collapse
|
19
|
Kim K, Son T, Hong JS, Kwak TJ, Jeong MH, Weissleder R, Im H. Physisorption of Affinity Ligands Facilitates Extracellular Vesicle Detection with Low Non-Specific Binding to Plasmonic Gold Substrates. ACS APPLIED MATERIALS & INTERFACES 2022; 14:10.1021/acsami.2c07317. [PMID: 35653580 PMCID: PMC9715849 DOI: 10.1021/acsami.2c07317] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Plasmonic biosensors are increasingly being used for the analysis of extracellular vesicles (EVs) originating from disease areas. However, the high non-specific binding of EVs to a gold-sensing surface has been a critical problem and hindered the true translational potential. Here, we report that direct antibody immobilization on the plasmonic gold surface via physisorption shows excellent capture of cancer-derived EVs with ultralow non-specific binding even at very high concentrations. Contrary to commonly used methods that involve thiol-based linker attachment and an EDC/sulfo-NHS reaction, we show a higher specific capture rate and >50-fold lower non-specific on citrate-capped plain and nanopatterned gold surfaces. The method provides a simple, fast, and reproducible means to functionalize plasmonic gold surfaces with antibodies for robust EV biosensing.
Collapse
Affiliation(s)
- Kihyeun Kim
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Taehwang Son
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Jae-Sang Hong
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Tae Joon Kwak
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Mi Ho Jeong
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
- Cancer Center, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Hyungsoon Im
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
20
|
Drozd M, Duszczyk A, Ivanova P, Pietrzak M. Interactions of proteins with metal-based nanoparticles from a point of view of analytical chemistry - Challenges and opportunities. Adv Colloid Interface Sci 2022; 304:102656. [PMID: 35367856 DOI: 10.1016/j.cis.2022.102656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/18/2022] [Accepted: 03/23/2022] [Indexed: 11/01/2022]
Abstract
Interactions of proteins with nanomaterials draw attention of many research groups interested in fundamental phenomena. However, alongside with valuable information regarding physicochemical aspects of such processes and their mechanisms, they more and more often prove to be useful from a point of view of bioanalytics. Deliberate use of processes based on adsorption of proteins on nanoparticles (or vice versa) allows for a development of new analytical methods and improvement of the existing ones. It also leads to obtaining of nanoparticles of desired properties and functionalities, which can be used as elements of analytical tools for various applications. Due to interactions with nanoparticles, proteins can also gain new functionalities or lose their interfering potential, which from perspective of bioanalytics seems to be very inviting and attractive. In the framework of this article we will discuss the bioanalytical potential of interactions of proteins with a chosen group of nanoparticles, and implementation of so driven processes for biosensing. Moreover, we will show both positive and negative (opportunities and challenges) aspects resulting from the presence of proteins in media/samples containing metal-based nanoparticles or their precursors.
Collapse
|
21
|
Wu SY, Wu FG, Chen X. Antibody-Incorporated Nanomedicines for Cancer Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2109210. [PMID: 35142395 DOI: 10.1002/adma.202109210] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2021] [Revised: 02/06/2022] [Indexed: 06/14/2023]
Abstract
Antibody-based cancer therapy, one of the most significant therapeutic strategies, has achieved considerable success and progress over the past decades. Nevertheless, obstacles including limited tumor penetration, short circulation half-lives, undesired immunogenicity, and off-target side effects remain to be overcome for the antibody-based cancer treatment. Owing to the rapid development of nanotechnology, antibody-containing nanomedicines that have been extensively explored to overcome these obstacles have already demonstrated enhanced anticancer efficacy and clinical translation potential. This review intends to offer an overview of the advancements of antibody-incorporated nanoparticulate systems in cancer treatment, together with the nontrivial challenges faced by these next-generation nanomedicines. Diverse strategies of antibody immobilization, formats of antibodies, types of cancer-associated antigens, and anticancer mechanisms of antibody-containing nanomedicines are provided and discussed in this review, with an emphasis on the latest applications. The current limitations and future research directions on antibody-containing nanomedicines are also discussed from different perspectives to provide new insights into the construction of anticancer nanomedicines.
Collapse
Affiliation(s)
- Shun-Yu Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Fu-Gen Wu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, 2 Sipailou Road, Nanjing, 210096, P. R. China
| | - Xiaoyuan Chen
- Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, 119077, Singapore
| |
Collapse
|
22
|
Development of Neuropeptide Y and Cell-Penetrating Peptide MAP Adsorbed onto Lipid Nanoparticle Surface. Molecules 2022; 27:molecules27092734. [PMID: 35566093 PMCID: PMC9101637 DOI: 10.3390/molecules27092734] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/20/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
Functionalization of nanoparticles surfaces have been widely used to improve diagnostic and therapeutic biological outcome. Several methods can be applied to modify nanoparticle surface; however, in this article we focus toward a simple and less time-consuming method. We applied an adsorption method on already formulated nanostructured lipid carriers (NLC) to functionalize these nanoparticles with three distinct peptides sequences. We selected a cell-penetrating peptide (CPP), a lysine modified model amphipathic peptide (Lys(N3)-MAP), CPP/drug complex, and the neuropeptide Y. The aim of this work is to evaluate the effect of several parameters such as peptide concentration, different types of NLC, different types of peptides, and incubation medium on the physicochemical proprieties of NLC and determine if adsorption occurs. The preliminary results from zeta potential analysis indicate some evidence that this method was successful in adsorbing three types of peptides onto NLC. Several non-covalent interactions appear to be involved in peptide adsorption with the possibility of three adsorption peptide hypothesis that may occur with NLC in solution. Moreover, and for the first time, in silico docking analysis demonstrated strong interaction between CPP MAP and NPY Y1 receptor with high score values when compared to standard antagonist and NPY.
Collapse
|
23
|
Iyisan B, Simon J, Avlasevich Y, Baluschev S, Mailaender V, Landfester K. Antibody-Functionalized Carnauba Wax Nanoparticles to Target Breast Cancer Cells. ACS APPLIED BIO MATERIALS 2022; 5:622-629. [PMID: 35014837 PMCID: PMC8864612 DOI: 10.1021/acsabm.1c01090] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
![]()
Development of safer
nanomedicines for drug delivery applications
requires immense efforts to improve clinical outcomes. Targeting a
specific cell, biocompatibility and biodegradability are vital properties
of a nanoparticle to fulfill the safety criteria in medical applications.
Herein, we fabricate antibody-functionalized carnauba wax nanoparticles
encapsulated a hydrophobic drug mimetic, which is potentially interesting
for clinical use due to the inert and nontoxic properties of natural
waxes. The nanoparticles are synthesized applying miniemulsion methods
by solidifying molten wax droplets and further evaporating the solvent
from the dispersion. The pH-selective adsorption of antibodies (IgG1,
immunoglobulin G1, and CD340, an antihuman HER2 antibody) onto the
nanoparticle surface is performed for practical and effective functionalization,
which assists to overcome the complexity in chemical modification
of carnauba wax. The adsorption behavior of the antibodies is studied
using isothermal titration calorimetry (ITC), which gives thermodynamic
parameters including the enthalpy, association constant, and stoichiometry
of the functionalization process. Both antibodies exhibit strong binding
at pH 2.7. The CD340-decorated wax nanoparticles show specific cell
interaction toward BT474 breast cancer cells and retain the targeting
function even after 6 months of storage period.
Collapse
Affiliation(s)
- Banu Iyisan
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.,Institute of Biomedical Engineering, Boğaziçi University, 34684 Çengelköy, Istanbul, Turkey
| | - Johanna Simon
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.,Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Yuri Avlasevich
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| | - Stanislav Baluschev
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.,Faculty of Physics, University of Sofia "Saint Kliment Ohridski", James Bourchier 5, 1164 Sofia, Bulgaria
| | - Volker Mailaender
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.,Dermatology Clinic, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany
| |
Collapse
|
24
|
Sotnikov DV, Byzova NA, Zherdev AV, Dzantiev BB. Retention of Activity by Antibodies Immobilized on Gold Nanoparticles of Different Sizes: Fluorometric Method of Determination and Comparative Evaluation. NANOMATERIALS 2021; 11:nano11113117. [PMID: 34835881 PMCID: PMC8625478 DOI: 10.3390/nano11113117] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 11/12/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022]
Abstract
Antibody–nanoparticle conjugates are widely used analytical reagents. An informative parameter reflecting the conjugates’ properties is the number of antibodies per nanoparticle that retain their antigen-binding ability. Estimation of this parameter is characterized by a lack of simple, reproducible methods. The proposed method is based on the registration of fluorescence of tryptophan residues contained in proteins and combines sequential measurements of first the immobilized antibody number and then the bound protein antigen number. Requirements for the measurement procedure have been determined to ensure reliable and accurate results. Using the developed technique, preparations of spherical gold nanoparticles obtained by the most common method of citrate reduction of gold salts (the Turkevich–Frens method) and varying in average diameter from 15 to 55 nm have been characterized. It was shown that the number of antibodies (immunoglobulins G) bound by one nanoparticle ranged from 30 to 194 during adsorptive unoriented monolayer immobilization. C-reactive protein was considered as the model antigen. The percentage of antibody valences that retained their antigen-binding properties in the conjugate increased from 17 to 34% with an increase in the diameter of gold nanoparticles. The proposed method and the results of the study provide tools to assess the capabilities of the preparations of gold nanoparticles and their conjugates as well as the expediency of seeking the best techniques for various practical purposes.
Collapse
|
25
|
Okyem S, Awotunde O, Ogunlusi T, Riley MB, Driskell JD. Probing the Mechanism of Antibody-Triggered Aggregation of Gold Nanoparticles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2021; 37:2993-3000. [PMID: 33621098 DOI: 10.1021/acs.langmuir.1c00100] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The unique physicochemical properties of gold nanoparticles (AuNPs) provide many opportunities to develop novel biomedical technologies. The surface chemistry of AuNPs can be engineered to perform a variety of functions, including targeted binding, cellular uptake, or stealthlike properties through the immobilization of biomolecules, such as proteins. It is well established that proteins can spontaneously adsorb onto AuNPs, to form a stable and functional bioconjugate; however, the protein-AuNP interaction may result in the formation of less desirable protein-AuNP aggregates. Therefore, it is imperative to investigate the protein-AuNP interaction and elucidate the mechanism by which protein triggers AuNP aggregation. Herein, we systematically investigated the interaction of immunoglobulin G (IgG) antibody with citrate-capped AuNPs as a function of solution pH. We found that the addition of antibody triggers the aggregation of AuNPs for pH < 7.5, whereas a monolayer of antibody adsorbs onto the AuNP to form a stable bioconjugate when the antibody is added to AuNPs at pH ≥ 7.5. Our data identifies electrostatic bridging between the antibody and the negatively charged AuNPs as the mechanism by which aggregation occurs and rules out protein unfolding and surface charge depletion as potential causes. Furthermore, we found that the electrostatic bridging of AuNPs is reversible within the first few hours of interaction, but the protein-AuNP interactions strengthen over 24 h, after which the protein-AuNP aggregate is irreversibly formed. From this data, we developed a straightforward approach to acrylate the basic residues on the antibody to prevent protein-induced aggregation of AuNP over a wide pH range. The results of this study provide additional insight into antibody-nanoparticle interactions and provide a pathway to control the interaction with the potential to enhance the conjugate function.
Collapse
Affiliation(s)
- Samuel Okyem
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Olatunde Awotunde
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Tosin Ogunlusi
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - McKenzie B Riley
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Jeremy D Driskell
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| |
Collapse
|
26
|
Frimpong R, Jang W, Kim JH, Driskell JD. Rapid vertical flow immunoassay on AuNP plasmonic paper for SERS-based point of need diagnostics. Talanta 2021; 223:121739. [DOI: 10.1016/j.talanta.2020.121739] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 09/22/2020] [Accepted: 10/04/2020] [Indexed: 01/09/2023]
|
27
|
Arslan FB, Ozturk Atar K, Calis S. Antibody-mediated drug delivery. Int J Pharm 2021; 596:120268. [PMID: 33486037 DOI: 10.1016/j.ijpharm.2021.120268] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2020] [Revised: 12/30/2020] [Accepted: 12/31/2020] [Indexed: 01/10/2023]
Abstract
Passive and active targeted nanoparticulate delivery systems show promise to compensate for lacking properties of conventional therapy such as side effects, insufficient efficiency and accumulation of the drug at target site, poor pharmacokinetic properties etc. For active targeting, physically or covalently conjugated ligands, including monoclonal antibodies and their fragments, are consistently used and researched for targeting delivery systems or drugs to their target site. Currently, there are several FDA approved actively targeted antibody-drug conjugates, whereas no active targeted delivery system is in clinical use at present. However, efforts to successfully formulate actively targeted delivery systems continue. The scope of this review will be the use of monoclonal antibodies and their fragments as targeting ligands. General information about targeted delivery and antibodies will be given at the first half of the review. As for the second half, fragmentation of antibodies and conjugation approaches will be explained. Monoclonal antibodies and their fragments as targeting ligands and approaches for conjugating these ligands to nanoparticulate delivery systems and drugs will be the main focus of this review, polyclonal antibodies will not be included.
Collapse
Affiliation(s)
- Fatma Betul Arslan
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Kivilcim Ozturk Atar
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Sema Calis
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey.
| |
Collapse
|
28
|
Awotunde O, Okyem S, Chikoti R, Driskell JD. Role of Free Thiol on Protein Adsorption to Gold Nanoparticles. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2020; 36:9241-9249. [PMID: 32686419 DOI: 10.1021/acs.langmuir.0c01550] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Protein-gold nanoparticle (AuNP) bioconjugates have many potential applications in nanomedicine. A thorough understanding of the interaction between the protein and the AuNP is critical to engineering these functional bioconjugates with desirable properties. In this work, we investigate the role of free thiols presented by the protein on the stability of the protein-AuNP conjugate. Human serum albumin (HSA) was modified with 2-iminothiolane (Traut's reagent) to introduce additional thiols onto the protein surface, and three variants of HSA were synthesized to present 1, 5, and 20 free thiols by controlling the molar excess of the chemical modifier. Protein exchange studies on AuNPs were conducted using these HSA species and an IgG antibody which exhibited 10 free thiols. Antibody-AuNP conjugates were synthesized, purified, and dispersed in solutions containing each of the HSA species. No protein exchange was detected with the HSA or modified HSA containing 5 thiols; however, 85% of the antibody was displaced on the AuNP surface by the extensively thiolated HSA presenting 20 free thiols. Furthermore, the impact of the protein adsorption sequence was probed in which each of the HSA species were preadsorbed onto the AuNP and dispersed in a solution of antibody. The antibody fully displaced the HSA with a single thiol from the AuNP within 3 h, required 24 h to completely displace the modified HSA containing 5 thiols, and was unable to displace the modified HSA containing 20 thiols. These results indicate that the number of Au-S interactions governs the binding interaction between the protein and the AuNP. This work provides further insight into the protein-AuNP binding mechanism and identifies important design principles for engineered proteins to optimize bioconjugates.
Collapse
Affiliation(s)
- Olatunde Awotunde
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Samuel Okyem
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Rishika Chikoti
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| | - Jeremy D Driskell
- Department of Chemistry, Illinois State University, Normal, Illinois 61790, United States
| |
Collapse
|
29
|
Pollok NE, Rabin C, Walgama CT, Smith L, Richards I, Crooks RM. Electrochemical Detection of NT-proBNP Using a Metalloimmunoassay on a Paper Electrode Platform. ACS Sens 2020; 5:853-860. [PMID: 32154707 DOI: 10.1021/acssensors.0c00167] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
In this paper, we demonstrate an electrochemical method for detection of the heart failure biomarker, N-terminal prohormone brain natriuretic peptide (NT-proBNP). The approach is based on a paper electrode assembly and a metalloimmunoassay; it is intended for eventual integration into a home-use sensor. Sensing of NT-proBNP relies on the formation of a sandwich immunoassay and electrochemical quantification of silver nanoparticle (AgNP) labels attached to the detection antibodies (Abs). There are four important outcomes reported in this article. First, compared to physisorption of the detection Abs on the AgNP labels, a 27-fold increase in signal is observed when a heterobifunctional cross-linker is used to facilitate this labeling. Second, the assay is selective in that it does not cross-react with other cardiac natriuretic peptides. Third, the assay forms in undiluted human serum (though the electrochemical analysis is carried out in buffer). Finally, and most important, the assay is able to detect NT-proBNP at concentrations between 0.58 and 2.33 nM. This performance approaches the critical NT-proBNP concentration threshold often used by physicians for risk stratification purposes: ∼0.116 nM.
Collapse
Affiliation(s)
- Nicole E. Pollok
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Charlie Rabin
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Charuksha T. Walgama
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Leilani Smith
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| | - Ian Richards
- Interactives Executive Excellence LLC, Austin, Texas 78733, United States
| | - Richard M. Crooks
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th Street, Stop A1590, Austin, Texas 78712-1224, United States
| |
Collapse
|
30
|
Pollok NE, Rabin C, Smith L, Crooks RM. Orientation-Controlled Bioconjugation of Antibodies to Silver Nanoparticles. Bioconjug Chem 2019; 30:3078-3086. [PMID: 31730333 PMCID: PMC6920564 DOI: 10.1021/acs.bioconjchem.9b00737] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Here we report on the use of heterobifunctional cross-linkers (HBCLs) to control the number, orientation, and activity of immunoglobulin G antibodies (Abs) conjugated to silver nanoparticles (AgNPs). A hydrazone conjugation method resulted in exclusive modification of the polysaccharide chains present on the fragment crystallizable region of the Abs, leaving the antigen-binding regions accessible. Two HBCLs, each having a hydrazide terminal group, were synthesized and tested for effectiveness. The two HBCLs differed in two respects, however: (1) either a thiol or a dithiolane group was used for attachment to the AgNP; and (2) the spacer arm was either a PEG chain or an alkyl chain. Both cross-linkers immobilized 5 ± 1 Abs on the surface of each 20-nm-diameter AgNP. Electrochemical results, obtained using a half-metalloimmunoassay, proved that Abs conjugated to AgNPs via either of the two HBCLs were 4 times more active than those conjugated by the more common physisorption technique. This finding confirmed that the HBCLs exerted orientational control over the Abs. We also demonstrated that the AgNP-HBCL-Ab conjugates were stable and active for at least 2 weeks. Finally, we found that the stability of the HBCLs themselves was related to the nature of their spacer arms. Specifically, the results showed that the HBCL having the alkyl chain is chemically stable for at least 90 days, making it the preferred cross-linker for bioassays.
Collapse
Affiliation(s)
| | | | - Leilani Smith
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th St., Stop A1590, Austin, TX, 78712-1224, U.S.A
| | - Richard M. Crooks
- Department of Chemistry, The University of Texas at Austin, 100 E. 24th St., Stop A1590, Austin, TX, 78712-1224, U.S.A
| |
Collapse
|