1
|
Chen Y, Fu KX, Cotton R, Ou Z, Kwak JW, Chien JC, Kesler V, Nyein HYY, Eisenstein M, Tom Soh H. A biochemical sensor with continuous extended stability in vivo. Nat Biomed Eng 2025:10.1038/s41551-025-01389-6. [PMID: 40410556 DOI: 10.1038/s41551-025-01389-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/03/2025] [Indexed: 05/25/2025]
Abstract
The development of biosensors that can detect specific analytes continuously, in vivo, in real time has proven difficult due to biofouling, probe degradation and signal drift that often occur in vivo. By drawing inspiration from intestinal mucosa that can protect host cell receptors in the presence of the gut microbiome, we develop a synthetic biosensor that can continuously detect specific target molecules in vivo. The biomimetic multicomponent sensor features the hierarchical nano-bio interface design with three-dimensional bicontinuous nanoporous structure, polymer coating and aptamer switches, balancing small-molecule sensing and surface protection in complex biological environments. Our system is stable for at least 1 month in undiluted serum in vitro or 1 week implanted within the blood vessels of free-moving rats, retaining over 50% baseline signal and reproducible calibration curves. We demonstrate that the implanted system can intravenously track pharmacokinetics in real time even after 4 days of continuous exposure to flowing blood within rat femoral vein. In this way, our work provides a generalizable design foundation for biosensors that can continuously operate in vivo for extended durations.
Collapse
Affiliation(s)
- Yihang Chen
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Kaiyu X Fu
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, IN, USA
| | - Renee Cotton
- Department of Comparative Medicine, Stanford University, Stanford, CA, USA
| | - Zihao Ou
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Jean Won Kwak
- Department of Radiology, Stanford University, Stanford, CA, USA
| | - Jun-Chau Chien
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Vladimir Kesler
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Hnin Yin Yin Nyein
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Michael Eisenstein
- Department of Radiology, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - H Tom Soh
- Department of Radiology, Stanford University, Stanford, CA, USA.
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA.
- Department of Bioengineering, Stanford University, Stanford, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
2
|
Fu B, Yuan Z, Fang G, Wang WJ, Xiong Z, Chen YC. Dielectric Nanocavity Enhanced Fluorescence Emission for Ultrasensitive Wavelength-Multiplexed Detection. NANO LETTERS 2025. [PMID: 40393953 DOI: 10.1021/acs.nanolett.5c01087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2025]
Abstract
This study demonstrates a novel biosensing platform utilizing a dielectric nanocavity to enhance fluorescence emission for the ultrasensitive detection of biomolecules. By coupling a silver (Ag) nanocube with a distributed Bragg reflector (DBR) mirror, we achieved a substantial fluorescence enhancement reaching a maximum enhancement factor of up to 855-fold and having quasi-single molecule sensitivity. The platform was successfully applied for multiplexed detection of four different miRNA biomarkers, showcasing its ability to detect multiple targets simultaneously with high sensitivity. The simplicity, rapid speed, and small detection volume (down to 0.5 μL) of this system make it suitable for high-throughput and large-area nanocavity imaging. Our findings offer a promising solution for ultrasensitive, multiplexed biosensing with potential applications in disease diagnosis, personalized medicine, and digital molecular diagnostics.
Collapse
Affiliation(s)
- Bowen Fu
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798 Singapore
- Institute for Digital Molecular Analytics and Science (IDMxS), Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921 Singapore
| | - Zhiyi Yuan
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798 Singapore
| | - Guocheng Fang
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798 Singapore
| | - Wen-Jie Wang
- Key Lab of Advanced Transducers and Intelligent Control System of Ministry of Education, Taiyuan University of Technology, Taiyuan 030024, P. R. China
| | - Zhongshu Xiong
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798 Singapore
| | - Yu-Cheng Chen
- School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798 Singapore
- Institute for Digital Molecular Analytics and Science (IDMxS), Nanyang Technological University, 59 Nanyang Drive, Singapore, 636921 Singapore
| |
Collapse
|
3
|
Yang Q, Pedreira-Rincón J, Balerdi-Sarasola L, Baptista-Pires L, Muñoz J, Camprubí-Ferrer D, Idili A, Parolo C. An aptamer-based electrochemical sensor for the quantification of the malaria biomarker lactate dehydrogenase. Biosens Bioelectron 2025; 274:117152. [PMID: 39884100 DOI: 10.1016/j.bios.2025.117152] [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: 10/12/2024] [Revised: 12/23/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025]
Abstract
Malaria is one of the most deadly infectious diseases, causing the death of hundreds of thousands of patients each year. Global efforts to combat malaria necessitate the implementation of novel rapid diagnostic tests deployable at the point of care. Here, we present the development of an electrochemical aptamer-based (EAB) sensor for the quantification of the malaria biomarker Plasmodium falciparum lactate dehydrogenase (PfLDH). Specifically, we demonstrate how, through careful optical and electrochemical characterization, we re-engineered the aptamer sequence to maximize its binding-induced conformational change, the core principle of EAB sensors. We then demonstrated that our biosensor can quantify clinically relevant concentrations of PfLDH in blood in a single step and within a few minutes. Overall, we believe these results demonstrate the promising potential of EAB sensors for the diagnosis and prognosis of malaria at the point of care.
Collapse
Affiliation(s)
- Qiuyue Yang
- Barcelona Institute for Global Health (ISGlobal), Barcelona, 08036, Spain.
| | - Julia Pedreira-Rincón
- Barcelona Institute for Global Health (ISGlobal), Barcelona, 08036, Spain; Facultat de Medicina i Ciències de la Salut, Universitatde Barcelona (UB), Barcelona, 08036, Spain
| | - Leire Balerdi-Sarasola
- Barcelona Institute for Global Health (ISGlobal), Barcelona, 08036, Spain; International Health Department, Hospital Clínic de Barcelona,Barcelona, 08036, Spain
| | - Luis Baptista-Pires
- I3N, Physics Department, University of Aveiro, Aveiro, 3810-193, Portugal; Graphenicalab: Parc Científic Tecnocampus, Mataró, 08302, Spain
| | - Jose Muñoz
- Barcelona Institute for Global Health (ISGlobal), Barcelona, 08036, Spain; Facultat de Medicina i Ciències de la Salut, Universitatde Barcelona (UB), Barcelona, 08036, Spain; International Health Department, Hospital Clínic de Barcelona,Barcelona, 08036, Spain
| | - Daniel Camprubí-Ferrer
- Barcelona Institute for Global Health (ISGlobal), Barcelona, 08036, Spain; International Health Department, Hospital Clínic de Barcelona,Barcelona, 08036, Spain
| | - Andrea Idili
- Department of Chemical Sciences and Technologies, University of Rome Tor Vergata, Via della Ricerca Scientifica, Rome 00133, Italy
| | - Claudio Parolo
- Barcelona Institute for Global Health (ISGlobal), Barcelona, 08036, Spain; INTERFIBIO Research Group, Departament d'Enginyeria Química, Universitat Rovira i Virgili, Tarragona, 43007, Spain.
| |
Collapse
|
4
|
Khan A, Anwar M, Rehman AU, Shokouhimehr M, Reis NM, Kalhoro KA, Zhang C, Liu Z. Biorecognition-based electrochemical sensors for highly sensitive C-reactive protein detection: A review. Int J Biol Macromol 2025; 304:140829. [PMID: 39938854 DOI: 10.1016/j.ijbiomac.2025.140829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 01/23/2025] [Accepted: 02/07/2025] [Indexed: 02/14/2025]
Abstract
Highly sensitive C-reactive protein (hsCRP) is a widely recognized biomarker for inflammation and cardiovascular diseases and plays a critical role in early diagnosis, risk assessment, and treatment monitoring. The development of sensitive and selective techniques for hsCRP detection is of paramount importance for clinical diagnostics. Electrochemical sensors have emerged as promising alternatives to traditional methods, offering rapid, cost-effective, and portable solutions for hsCRP analysis. This review comprehensively discusses advancements in biorecognition-based electrochemical sensors for hsCRP detection, focusing on label- and label-free approaches. This review highlights the sensor principles, designs, and performance, and emphasizes their advantages as well as limitations in various target applications. Recent studies have shown the potential of both label- and label-free-based sensors to achieve low detection limits and wide linear ranges comparable to traditional methods. In addition, we discuss the mechanisms, challenges, and future directions of biorecognition-based electrochemical sensors for hsCRP detection. This innovation can potentially revolutionize the diagnosis and treatment of cardiovascular and inflammatory diseases by enhancing the detection sensitivity and specificity. Ultimately, these advancements aim to improve patient outcomes by enabling earlier diagnosis, cost-effectiveness, and more precise monitoring, contributing to more effective management of cardiovascular health globally.
Collapse
Affiliation(s)
- Adil Khan
- School of Electronic Information, Central South University, Changsha 410083, China; School of Physics, Central South University, Changsha 410083, China
| | - Muhammad Anwar
- School of Electronic Information, Central South University, Changsha 410083, China; School of Physics, Central South University, Changsha 410083, China
| | - Atiq Ur Rehman
- School of Physics, Central South University, Changsha 410083, China
| | - Mohammadreza Shokouhimehr
- Department of Materials Science and Engineering, Research Institute of Advanced Materials, Seoul National University, Seoul 08826, Republic of Korea; Institute of Nanosensor Technology, Hanyang University, Ansan 15588, Republic of Korea
| | - Nuno M Reis
- Department of Chemical Engineering and Centre for Bioengineering & Biomedical Technologies (CBio), University of Bath, Claverton Down, Bath BA2 7AY, UK
| | - Kashif Ali Kalhoro
- School of Electronic Information, Central South University, Changsha 410083, China; School of Physics, Central South University, Changsha 410083, China; Sukkur IBA University, Sukkur 65200, Pakistan
| | - Chi Zhang
- School of Electronic Information, Central South University, Changsha 410083, China; School of Physics, Central South University, Changsha 410083, China
| | - Zhengchun Liu
- School of Electronic Information, Central South University, Changsha 410083, China; School of Physics, Central South University, Changsha 410083, China.
| |
Collapse
|
5
|
Luo S, Wu Q, Wang L, Qu H, Zheng L. Direct detection of doxorubicin in whole blood using a hydrogel-protected electrochemical aptamer-based biosensor. Talanta 2025; 285:127289. [PMID: 39613489 DOI: 10.1016/j.talanta.2024.127289] [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: 07/25/2024] [Revised: 11/22/2024] [Accepted: 11/24/2024] [Indexed: 12/01/2024]
Abstract
Electrochemical aptamer-based biosensors (EABs) have been developed for multiple important biomarkers for their convenient and real-time features. However, the application of EABs in complex biological fluids has been limited by the rapid loss of sensitivity and selectivity due to inactivation and biofouling of aptamer probes and electrodes. To address this issue, we report the preparation of a simple hydrogel-protected aptamer-based biosensor (HP-EAB) for direct detection of Doxorubicin (DOX) in whole blood. The aptamer provides excellent selectivity for the electrochemical sensor, allowing the prepared sensor to accurately detect DOX in a 50-fold diluted whole blood sample. The agarose hydrogel coating on the electrode surface allows the passage of small molecules while hindering the adsorption of biomolecules from the whole blood matrix to the electrode surface. The experimental results show that the prepared HP-EAB has high stability compared with the unprotected EAB, and the HP-EAB maintains excellent detection performance after 7 days of storage. The hydrogel coating can effectively reduce the non-specific response to the whole blood matrix and prolong the life-time of the sensor. When used to detect DOX in rabbit whole blood, the HP-EAB exhibited excellent detection performance with a detection limit of 25.9 nM (S/N = 3) and a detection range of 0.1 μM-50 μM. The developed HP-EAB provides an excellent platform for the rapid and accurate determination of important analytes in complex biological fluids.
Collapse
Affiliation(s)
- Songjia Luo
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Qingliu Wu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Lu Wang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China; Engineering Research Center of Bioprocess, Ministry of Education, Hefei University of Technology, Hefei, 230009, China.
| | - Hao Qu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China; Anhui Provincial International Science and Technology Cooperation Base for Major Metabolic Diseases and Nutritional Interventions, Hefei, 230009, China.
| | - Lei Zheng
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China; Intelligent Interconnected Systems Laboratory of Anhui Province, Hefei University of Technology, Hefei, 230009, China
| |
Collapse
|
6
|
Gouveia G, Saateh A, Swietlikowska A, Scarpellini C, Tsang E, Altug H, Merkx M, Dillen A, Leirs K, Spasic D, Lammertyn J, Gothelf KV, Bonedeau E, Porzberg N, Smeets RL, Koenen HJPM, Prins MWJ, de Jonge MI. Continuous Biosensing to Monitor Acute Systemic Inflammation, a Diagnostic Need for Therapeutic Guidance. ACS Sens 2025; 10:4-14. [PMID: 39692622 PMCID: PMC11773571 DOI: 10.1021/acssensors.4c02569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/14/2024] [Accepted: 12/03/2024] [Indexed: 12/19/2024]
Abstract
Continuous monitoring of acute inflammation can become a very important next step for guiding therapeutic interventions in severely ill patients. This Perspective discusses the current medical need for patients with acute inflammatory diseases and the potential of continuous biosensing technologies. First, we discuss biomarkers that could help to monitor the state of a patient with acute systemic inflammation based on theoretical studies and empirical data. Then, based on the state of the art, we describe sensing strategies that could be applied for the continuous monitoring of acute inflammatory biomarkers, followed by challenges that must be overcome. Nanoswitch-based continuous biosensors enable suitable measurement frequencies but still lack sensitivity, while regeneration risks lower sensor reliability. Developments are still needed in bioreceptors and molecular architectures, regeneration techniques, combined with suitable sampling and sample pretreatment methods, for bringing continuous biosensing of inflammation closer to reality. Furthermore, collaborations between healthcare professionals and scientists, regulatory bodies, and biosensor engineers are needed for a successful translation of sensing technologies from the laboratory to clinical practice.
Collapse
Affiliation(s)
- Guilherme Gouveia
- Department
of Laboratory Medicine, Laboratory of Medical Immunology, Radboud
Community for Infectious Diseases, Radboud
University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Abtin Saateh
- Institute
of Bioengineering, École Polytechnique
Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Anna Swietlikowska
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven 5600MB, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, Eindhoven 5600MB, The Netherlands
| | - Claudia Scarpellini
- Department
of Biosystems - Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Emily Tsang
- Department
of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus 8000 C, Denmark
| | - Hatice Altug
- Institute
of Bioengineering, École Polytechnique
Fédérale de Lausanne (EPFL), Lausanne 1015, Switzerland
| | - Maarten Merkx
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven 5600MB, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, Eindhoven 5600MB, The Netherlands
| | - Annelies Dillen
- Department
of Biosystems - Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Karen Leirs
- Department
of Biosystems - Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Dragana Spasic
- Department
of Biosystems - Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Jeroen Lammertyn
- Department
of Biosystems - Biosensors Group, KU Leuven, Willem de Croylaan 42, 3001 Leuven, Belgium
| | - Kurt V. Gothelf
- Department
of Chemistry and Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus 8000 C, Denmark
| | - Estelle Bonedeau
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Nicola Porzberg
- Department
of Chemical Biology, Max Planck Institute
for Medical Research, Jahnstrasse 29, 69120 Heidelberg, Germany
| | - Ruben L. Smeets
- Department
of Laboratory Medicine, Laboratory of Medical Immunology, Radboud
Community for Infectious Diseases, Radboud
University Medical Center, Nijmegen 6500 HB, The Netherlands
- Department
of Laboratory Medicine, Radboudumc Laboratory for Diagnostics, Radboud University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Hans J. P. M. Koenen
- Department
of Laboratory Medicine, Laboratory of Medical Immunology, Radboud
Community for Infectious Diseases, Radboud
University Medical Center, Nijmegen 6500 HB, The Netherlands
| | - Menno W. J. Prins
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, Eindhoven 5600MB, The Netherlands
- Department
of Biomedical Engineering, Eindhoven University
of Technology, Eindhoven 5600MB, The Netherlands
- Department
of Applied Physics, Eindhoven University
of Technology, Eindhoven 5600MB, The Netherlands
- Helia Biomonitoring, De Lismortel 31, 5612 AR Eindhoven, The Netherlands
| | - Marien I. de Jonge
- Department
of Laboratory Medicine, Laboratory of Medical Immunology, Radboud
Community for Infectious Diseases, Radboud
University Medical Center, Nijmegen 6500 HB, The Netherlands
| |
Collapse
|
7
|
Duan H, Peng S, He S, Tang S, Goda K, Wang CH, Li M. Wearable Electrochemical Biosensors for Advanced Healthcare Monitoring. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411433. [PMID: 39588557 PMCID: PMC11727287 DOI: 10.1002/advs.202411433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 11/13/2024] [Indexed: 11/27/2024]
Abstract
Recent advancements in wearable electrochemical biosensors have opened new avenues for on-body and continuous detection of biomarkers, enabling personalized, real-time, and preventive healthcare. While glucose monitoring has set a precedent for wearable biosensors, the field is rapidly expanding to include a wider range of analytes crucial for disease diagnosis, treatment, and management. In this review, recent key innovations are examined in the design and manufacturing underpinning these biosensing platforms including biorecognition elements, signal transduction methods, electrode and substrate materials, and fabrication techniques. The applications of these biosensors are then highlighted in detecting a variety of biochemical markers, such as small molecules, hormones, drugs, and macromolecules, in biofluids including interstitial fluid, sweat, wound exudate, saliva, and tears. Additionally, the review also covers recent advances in wearable electrochemical biosensing platforms, such as multi-sensory integration, closed-loop control, and power supply. Furthermore, the challenges associated with critical issues are discussed, such as biocompatibility, biofouling, and sensor degradation, and the opportunities in materials science, nanotechnology, and artificial intelligence to overcome these limitations.
Collapse
Affiliation(s)
- Haowei Duan
- School of Mechanical and Manufacturing EngineeringThe University of New South WalesSydneyNSW2052Australia
| | - Shuhua Peng
- School of Mechanical and Manufacturing EngineeringThe University of New South WalesSydneyNSW2052Australia
| | - Shuai He
- School of Mechanical and Manufacturing EngineeringThe University of New South WalesSydneyNSW2052Australia
| | - Shi‐Yang Tang
- School of Mechanical and Manufacturing EngineeringThe University of New South WalesSydneyNSW2052Australia
| | - Keisuke Goda
- Department of ChemistryThe University of TokyoTokyo113‐0033Japan
- Department of BioengineeringUniversity of CaliforniaLos AngelesCalifornia90095USA
- Institute of Technological SciencesWuhan UniversityHubei430072China
| | - Chun H. Wang
- School of Mechanical and Manufacturing EngineeringThe University of New South WalesSydneyNSW2052Australia
| | - Ming Li
- School of Mechanical and Manufacturing EngineeringThe University of New South WalesSydneyNSW2052Australia
| |
Collapse
|
8
|
Hao X, Song W, Wang Y, Qin J, Jiang Z. Recent Advancements in Electrochemical Sensors Based on MOFs and Their Derivatives. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2408624. [PMID: 39676419 DOI: 10.1002/smll.202408624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 11/19/2024] [Indexed: 12/17/2024]
Abstract
Metal-organic frameworks (MOFs) are composed of metal nodes and organic linkers that can self-assemble into an infinite network. The high porosity and large surface area of MOFs facilitate the effective enrichment and mass transfer of analytes, which can enhance the signal response and improve the sensitivity of electrochemical sensors. Additionally, MOFs and their derivatives possess the properties of unsaturated metal sites and tunable structures, collectively demonstrating their potential for electrochemical sensing. This paper summarizes the preparation methods, structural properties, and applications of MOFs and their derivatives in electrochemical sensing, emphasizing sensors' selectivity and sensitivity from the perspectives of direct and indirect detection. Additionally, it also explores future directions and prospects for MOFs in electrochemical sensing, with the aim of overcoming current limitations through innovative approaches.
Collapse
Affiliation(s)
- Xi Hao
- School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Weihua Song
- Xuanwu Hospital Capital Medical University, Beijing, 100037, China
| | - Yinghui Wang
- The Second Affiliated Hospital of Luohe Medical College, Luohe, Henan, 462005, China
| | - Jieling Qin
- School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Zhenqi Jiang
- School of Medical Technology, School of Chemistry and Chemical Engineering, Beijing Institute of Technology, Beijing, 100081, China
| |
Collapse
|
9
|
Zargartalebi H, Mirzaie S, GhavamiNejad A, Ahmed SU, Esmaeili F, Geraili A, Flynn CD, Chang D, Das J, Abdrabou A, Sargent EH, Kelley SO. Active-reset protein sensors enable continuous in vivo monitoring of inflammation. Science 2024; 386:1146-1153. [PMID: 39636994 DOI: 10.1126/science.adn2600] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 06/11/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024]
Abstract
Continuous measurement of proteins in vivo is important for real-time disease management and prevention. Implantable sensors for monitoring small molecules such as glucose have been available for more than a decade. However, analysis of proteins remains an unmet need because the lower physiological levels require that sensors have high affinities, which are linked to long complexation half-lives (t1/2 ~20 hours) and slow equilibration when concentrations decrease. We report active-reset sensors by use of high-frequency oscillations to accelerate dissociation, which enables regeneration of the unbound form of the sensor within 1 minute. When implemented within implanted devices, these sensors allow for real-time, in vivo monitoring of proteins within interstitial fluid. Active-reset protein sensors track biomarker levels on a physiological timescale for inflammation monitoring in living animals.
Collapse
Affiliation(s)
- H Zargartalebi
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - S Mirzaie
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - A GhavamiNejad
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - S U Ahmed
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
- Robert H. Laurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
| | - F Esmaeili
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
| | - A Geraili
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
| | - C D Flynn
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - D Chang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
| | - J Das
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
| | - A Abdrabou
- Robert H. Laurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA
| | - E H Sargent
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
- Department of Electrical and Computer Engineering, University of Toronto, Toronto, ON, Canada
- Department of Electrical and Computer Engineering, Northwestern University, Evanston, IL, USA
| | - S O Kelley
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
- Department of Chemistry, Weinberg College of Arts and Sciences, Northwestern University, Evanston, IL, USA
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, Canada
- Robert H. Laurie Comprehensive Cancer Center, Northwestern University, Chicago, IL, USA
- Chan Zuckerberg Biohub Chicago, Chicago, IL, USA
| |
Collapse
|
10
|
Wang S. Continuous monitoring with a shake. Science 2024; 386:1093-1094. [PMID: 39637003 DOI: 10.1126/science.adt8928] [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: 12/07/2024]
Abstract
Oscillating signals rapidly reset a sensor for real-time protein detection in the body.
Collapse
Affiliation(s)
- Sihong Wang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, IL, USA
- Nanoscience and Technology Division and Center for Molecular Engineering, Argonne National Laboratory, Lemont, IL, USA
| |
Collapse
|
11
|
Feng L, Gao RY, Chen ZM, Qin SN, Cao YJ, Salminen K, Sun JJ, Wu SH. Cold-hot Janus electrochemical aptamer-based sensor for calibration-free determination of biomolecules. Biosens Bioelectron 2024; 264:116642. [PMID: 39126905 DOI: 10.1016/j.bios.2024.116642] [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: 04/12/2024] [Revised: 07/16/2024] [Accepted: 08/06/2024] [Indexed: 08/12/2024]
Abstract
Real-time, high-frequency measurements of pharmaceuticals, metabolites, exogenous antigens, and other biomolecules in biological samples can provide critical information for health management and clinical diagnosis. Electrochemical aptamer-based (EAB) sensor is a promising analytical technique capable of achieving these goals. However, the issues of insufficient sensitivity, frequent calibration and lack of adapted portable electrochemical device limit its practical application in immediate detection. In response we have fabricated an on-chip-integrated, cold-hot Janus EAB (J-EAB) sensor based on the thermoelectric coolers (TECs). Attributed to the Peltier effect, the enhanced/suppressed current response can be generated simultaneously on cold/hot sides of the J-EAB sensor. The ratio of the current responses on the cold and hot sides was used as the detection signal, enabling rapid on-site, calibration-free determination of small molecules (procaine) as well as macromolecules (SARS-CoV-2 spike protein) in single step, with detection limits of 1 μM and 10 nM, respectively. We have further demonstrated that the J-EAB sensor is effective in improving the ease and usability of the actual detection process, and is expected to provide a universal, low-cost, fast and easy potential analytical tool for other clinically important biomarkers, drugs or pharmaceutical small molecules.
Collapse
Affiliation(s)
- Lei Feng
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Run-Yu Gao
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Zhi-Min Chen
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Sai-Nan Qin
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Yi-Jie Cao
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Kalle Salminen
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, 350108, China
| | - Jian-Jun Sun
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, 350108, China.
| | - Shao-Hua Wu
- Ministry of Education Key Laboratory for Analytical Science of Food Safety and Biology, Fujian Provincial Key Laboratory of Analysis and Detection Technology for Food Safety, College of Chemistry, Fuzhou University, Fuzhou, 350108, China.
| |
Collapse
|
12
|
Mack J, Murray R, Lynch K, Arroyo-Currás N. 3D-printed electrochemical cells for multi-point aptamer-based drug measurements. SENSORS & DIAGNOSTICS 2024; 3:1533-1541. [PMID: 39157417 PMCID: PMC11325214 DOI: 10.1039/d4sd00192c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 08/03/2024] [Indexed: 08/20/2024]
Abstract
Electrochemical aptamer-based (E-AB) sensors achieve detection and quantitation of biomedically relevant targets such as small molecule drugs and protein biomarkers in biological samples. E-ABs are usually fabricated on commercially available macroelectrodes which, although functional for rapid sensor prototyping, can be costly and are not compatible with the microliter sample volumes typically available in biorepositories for clinical validation studies. Seeking to develop a multi-point sensing platform for sensor validation in sample volumes characteristic of clinical studies, we report a protocol for in-house assembly of 3D-printed E-ABs. We employed a commercially available 3D stereolithographic printer (FormLabs, $5k USD) for electrochemical cell fabrication and directly embedded electrodes within the 3D-printed cell structure. This approach offers a reproducible and reusable electrode fabrication process resulting in four independent and simultaneous measurements for statistically weighted results. We demonstrate compatibility with aptamer sequences binding antibiotics and antineoplastic agents. We also demonstrate a proof-of-concept validation of serum vancomycin measurements using clinical samples. Our results demonstrate that 3D-printing can be used in conjunction with E-ABs for accessible, rapid, and statistically meaningful validation of E-AB sensors in biological matrices.
Collapse
Affiliation(s)
- John Mack
- Biochemistry, Cellular and Molecular Biology Program, Johns Hopkins University School of Medicine 316 Hunterian Building, 725 North Wolfe Street Baltimore MD 21205 USA +1 443 287 4798
| | - Raygan Murray
- Biochemistry, Cellular and Molecular Biology Program, Johns Hopkins University School of Medicine 316 Hunterian Building, 725 North Wolfe Street Baltimore MD 21205 USA +1 443 287 4798
| | - Kenedi Lynch
- Amgen Scholars Program, Krieger School of Arts and Sciences, Johns Hopkins University Baltimore MD 21218 USA
| | - Netzahualcóyotl Arroyo-Currás
- Biochemistry, Cellular and Molecular Biology Program, Johns Hopkins University School of Medicine 316 Hunterian Building, 725 North Wolfe Street Baltimore MD 21205 USA +1 443 287 4798
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine Baltimore MD 21205 USA
| |
Collapse
|
13
|
Jensen IM, Clark V, Kirby HL, Arroyo-Currás N, Jenkins DM. Tuning N-heterocyclic carbene wingtips to form electrochemically stable adlayers on metals. MATERIALS ADVANCES 2024; 5:7052-7060. [PMID: 39156595 PMCID: PMC11325317 DOI: 10.1039/d4ma00648h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
Self-assembled monolayers (SAMs) are employed in electrochemical biosensors to passivate and functionalize electrode surfaces. These monolayers prevent the occurrence of undesired electrochemical reactions and act as scaffolds for coupling bioaffinity reagents. Thiols are the most common adlayer used for this application; however, the thiol-gold bond is susceptible to competitive displacement by naturally occurring solvated thiols in biological fluids, as well as to desorption under continuous voltage interrogation. To overcome these issues, N-heterocyclic carbene (NHC) monolayers have been proposed as an alternative for electrochemical biosensor applications due to the strong carbon-gold bond. To maximize the effectiveness of NHCs for SAMs, a thorough understanding of both the steric effects of wingtip substituents and NHC precursor type to the passivation of electrode surfaces is required. In this study, five different NHC wingtips as well as two kinds of NHC precursors were evaluated. The best performing NHC adlayers can be cycled continuously for four days (over 30 000 voltammetric cycles) without appreciably desorbing from the electrode surface. Benchmark thiol monolayers, in contrast, rapidly desorb after only twelve hours. Investigations also show NHC adlayer formation on other biosensor-relevant electrodes such as platinum and palladium.
Collapse
Affiliation(s)
- Isabel M Jensen
- Department of Chemistry University of Tennessee Knoxville Knoxville TN 37996 USA
| | - Vincent Clark
- Chemistry-Biology Interface Program Johns Hopkins University Baltimore MD 21218 USA
| | - Harper L Kirby
- Department of Chemistry University of Tennessee Knoxville Knoxville TN 37996 USA
| | - Netzahualcóyotl Arroyo-Currás
- Chemistry-Biology Interface Program Johns Hopkins University Baltimore MD 21218 USA
- Department of Pharmacology and Molecular Sciences Johns Hopkins University School of Medicine Baltimore MD 21205 USA
| | - David M Jenkins
- Department of Chemistry University of Tennessee Knoxville Knoxville TN 37996 USA
| |
Collapse
|
14
|
Gupta V, Pham A, Dick JE. Planar Disk μ-Aptasensors by Monolayer Assembly in a Dissolving Microdroplet. Anal Chem 2024. [PMID: 39152900 DOI: 10.1021/acs.analchem.4c01043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/19/2024]
Abstract
Electrochemical aptamer-based sensors provide a highly modular platform for real-time monitoring of small molecules. Their ability to selectively recognize target molecules in complex environments like biological fluids makes them an attractive technology for the analysis of micro- and nanoscale systems. The signal-to-noise of the measurement depends on the electroactive surface (i.e., how many aptamers one can place), which has previously precluded miniaturization of aptamer-based sensors to planar disk ultramicroelectrodes (r ∼ 5-10 μm). Here, we employ a concentration enrichment strategy based on the active dissolution of an aqueous, aptamer-containing microdroplet on an ultramicroelectrode submerged in an organic continuous phase (1,2-dichloroethane). We show consistent voltammetric signal increase as a function of droplet lifetime, indicating the successful immobalization of the thiol-terminated aminoglycoside aptamers to the electrode surface. We observe a diagnostic methylene blue peak and 10-fold increase in current magnitude as compared to bare microelectrodes. We report robust sensor behavior with a linear dynamic range extending from milli- to micromolar concentrations of kanamycin in buffer. This research offers a successful method for optimized electrochemical aptamer-based sensor fabrication and miniaturization on ultramicroelectrodes without the need for electrode surface area enhancement.
Collapse
Affiliation(s)
- Vanshika Gupta
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - AnhThu Pham
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
| | - Jeffrey E Dick
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Elmore Family School of Electrical and Computer Engineering, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
15
|
Younas R, Jubeen F, Bano N, Andreescu S, Zhang H, Hayat A. Covalent organic frameworks (COFs) as carrier for improved drug delivery and biosensing applications. Biotechnol Bioeng 2024; 121:2017-2049. [PMID: 38665008 DOI: 10.1002/bit.28718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 06/13/2024]
Abstract
Porous organic frameworks (POFs) represent a significant subclass of nanoporous materials in the field of materials science, offering exceptional characteristics for advanced applications. Covalent organic frameworks (COFs), as a novel and intriguing type of porous material, have garnered considerable attention due to their unique design capabilities, diverse nature, and wide-ranging applications. The unique structural features of COFs, such as high surface area, tuneable pore size, and chemical stability, render them highly attractive for various applications, including targeted and controlled drug release, as well as improving the sensitivity and selectivity of electrochemical biosensors. Therefore, it is crucial to comprehend the methods employed in creating COFs with specific properties that can be effectively utilized in biomedical applications. To address this indispensable fact, this review paper commences with a concise summary of the different methods and classifications utilized in synthesizing COFs. Second, it highlights the recent advancements in COFs for drug delivery, including drug carriers as well as the classification of drug delivery systems and biosensing, encompassing drugs, biomacromolecules, small biomolecules and the detection of biomarkers. While exploring the potential of COFs in the biomedical field, it is important to acknowledge the limitations that researchers may encounter, which could impact the practicality of their applications. Third, this paper concludes with a thought-provoking discussion that thoroughly addresses the challenges and opportunities associated with leveraging COFs for biomedical applications. This review paper aims to contribute to the scientific community's understanding of the immense potential of COFs in improving drug delivery systems and enhancing the performance of biosensors in biomedical applications.
Collapse
Affiliation(s)
- Rida Younas
- State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, Shandong, China
- Department of Chemistry, Govt College Women University, Faisalabad, Pakistan
| | - Farhat Jubeen
- Department of Chemistry, Govt College Women University, Faisalabad, Pakistan
| | - Nargis Bano
- Department of Physics and Astronomy College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Silvana Andreescu
- Department of Chemistry and Biomolecular Science, Clarkson University, Potsdam, New York, USA
| | - Hongxia Zhang
- State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, Shandong, China
| | - Akhtar Hayat
- State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, Shandong, China
- Interdisciplinary Research Centre in Biomedical Materials, COMSATS University Islamabad, Lahore, Punjab, Pakistan
| |
Collapse
|
16
|
Whitehouse WL, Lo LHY, Kinghorn AB, Shiu SCC, Tanner JA. Structure-Switching Electrochemical Aptasensor for Rapid, Reagentless, and Single-Step Nanomolar Detection of C-Reactive Protein. ACS APPLIED BIO MATERIALS 2024; 7:3721-3730. [PMID: 38485932 DOI: 10.1021/acsabm.4c00061] [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: 06/18/2024]
Abstract
C-reactive protein (CRP) is an acute-phase reactant and sensitive indicator for sepsis and other life-threatening pathologies, including systemic inflammatory response syndrome. Currently, clinical turn-around times for established CRP detection methods take between 30 min to hours or even days from centralized laboratories. Here, we report the development of an electrochemical biosensor using redox probe-tagged DNA aptamers, functionalized onto inexpensive, commercially available screen-printed electrodes. Binding-induced conformational switching of the CRP-targeting aptamer induces a specific and selective signal-ON event, which enables single-step and reagentless detection of CRP in as little as 1 min. The aptasensor limit of detection spans approximately 20-60 nM in 50% human serum with dynamic response windows spanning 1-200 or 1-500 nM (R = 0.97/R = 0.98 respectively). The sensor is stable for at least 1 week and can be reused numerous times, as judged from repeated real-time dosing and dose-response assays. By decoupling binding events from the signal induction mechanism, structure-switching electrochemical aptamer-based sensors provide considerable advantages over their adsorption-based counterparts. Our work expands on the retinue of such sensors reported in the literature and is the first instance of structure-switching electrochemical aptamer-based sensors (SS-EABs) for reagentless, voltammetric CRP detection. We hope this study inspires further investigations into the suitability of SS-EABs for diagnostics, which will aid translational R&D toward fully realized devices aimed at point-of-care applications or for broader use by the public.
Collapse
Affiliation(s)
- William L Whitehouse
- Advanced Biomedical Instrumentation Center, Hong Kong Science Park, Shatin, New Territories, Hong Kong, China
| | - Louisa H Y Lo
- Advanced Biomedical Instrumentation Center, Hong Kong Science Park, Shatin, New Territories, Hong Kong, China
| | - Andrew B Kinghorn
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Simon C C Shiu
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Julian A Tanner
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
17
|
Ahmadian-Alam L, Andrade A, Song E. Electrochemical detection of glutamate and histamine using redox-labeled stimuli-responsive polymer as a synthetic target receptor. ACS APPLIED POLYMER MATERIALS 2024; 6:5630-5641. [PMID: 39444408 PMCID: PMC11498899 DOI: 10.1021/acsapm.4c00121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2024]
Abstract
Glutamate (Glu) and histamine (His) are two major neurotransmitters that play many critical roles in brain physiological functions and neurological disorders. Therefore, specific and sensitive monitoring of Glu and His is essential in the diagnosis and treatment of various mental health and neurodegenerative disorders. Both being non-electroactive species, direct electrochemical detection of Glu and His has been challenging. Herein, we report a stimuli-responsive polymer-based biosensor for the electrochemical detection of Glu and His. The polymer-based target receptors consist of a linear chain stimuli-responsive templated polymer hybrid that is labeled with an osmium-based redox-active reporter molecules to elicit conformation-dependent electrochemical responses. The polymers are then attached to a gold electrode to implement an electrochemical sensor. The cyclic voltammetry (CV) and square-wave voltammetry (SWV) results confirmed the polymers' conformational changes due to the specific target (i.e., Glu and His) recognition and the corresponding electrochemical detection capabilities. The voltammetry results indicate that this biosensor can be used as a 'signal-on' and 'signal-off' sensors for the detection of Glu and His concentrations, respectively. The developed biosensor also showed excellent regeneration capability by fully recovering the initial current signal after rinsing with deionized water. To further validate the polymer's utility as a target bioreceptor, the surface plasmon resonance (SPR) technique was used to characterize the binding affinity between the designed polymers and the target chemical. The SPR results exhibited the equilibrium dissociation constants (KD) of 2.40 μM and 1.54 μM for the polymer-Glu and polymer-His interactions, respectively. The results obtained this work strongly suggest that the proposed sensing technology could potentially be used as a platform for monitoring non-electroactive neurochemicals from animal models.
Collapse
Affiliation(s)
- Leila Ahmadian-Alam
- Department of Electrical & Computer Engineering, University of New Hampshire, Durham, NH 03824, United States
| | - Arturo Andrade
- Department of Neuroscience, Brown University, Providence, RI 02912, United States
- Robert J. & Nancy D. Carney Institute for Brain Science, Brown University, Providence, RI 02912, United States
| | - Edward Song
- Department of Electrical & Computer Engineering, University of New Hampshire, Durham, NH 03824, United States
| |
Collapse
|
18
|
Ritz AJ, Stuehr OM, Comer DN, Lazenby RA. Controlling Gold Morphology Using Electrodeposition for the Preparation of Electrochemical Aptamer-Based Sensors. ACS APPLIED BIO MATERIALS 2024; 7:1925-1935. [PMID: 38369768 DOI: 10.1021/acsabm.3c01254] [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: 02/20/2024]
Abstract
Nanostructuring of gold surfaces to enhance electroactive surface area has proven to significantly enhance the performance of electrochemical aptamer-based (E-AB) sensors, particularly for electrodes on the microscale. Unlike for sensors fabricated on polished gold surfaces, predicting the behavior of E-AB sensors on surfaces with varied gold morphologies becomes more intricate due to the effects of surface roughness and the shapes and sizes of surface features on supporting a self-assembled monolayer. In this study, we explored the impact of gold morphology characteristics on sensor performance, evaluating parameters such as signal change in response to the addition of the target analyte, aptamer probe packing density, and continuous sensing ability. Our findings reveal that surface area enhancement can either enhance or diminish sensor performance for gold nanostructured E-AB sensors, contingent upon the surface morphology. In particular, our results indicate that the aptamer packing density and target analyte signal change results are heavily dependent on gold nanostructure size and features. Sensing surfaces with larger nanoparticle diameters, which were prepared using electrodeposition at a constant potential, had a reduced aptamer packing density and exhibited diminished sensor performance. However, the equivalent packing density of polished electrodes did not yield the equivalent signal change. Other surfaces that were prepared using pulsed waveform electrodeposition achieved optimal signal change with a deposition time, tdep, of 120 s, and increased deposition time with enhanced electroactive surface area resulted in minimized signal changes and more rapid sensor degradation. By investigating sensing surfaces with varied morphologies, we have demonstrated that enhancing the electroactive surface does not always enhance the signal change of the sensor, and aptamer packing density alone does not dictate observed signal change trends. We anticipate that understanding how electrodeposition techniques enhance or diminish sensor performance will pave the way for further exploration of nanostructure-aptamer relationships, contributing to the future development of optimized, miniaturized electrochemical aptamer-based sensors for continuous, in vivo sensing.
Collapse
Affiliation(s)
- Amanda J Ritz
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Olivia M Stuehr
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Danté N Comer
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| | - Robert A Lazenby
- Department of Chemistry & Biochemistry, Florida State University, Tallahassee, Florida 32306, United States
| |
Collapse
|
19
|
de Bakker M, Kimenai DM, Mills NL. Challenges and opportunities for biomarker discovery to predict imminent myocardial infarction. NATURE CARDIOVASCULAR RESEARCH 2024; 3:102-103. [PMID: 39196200 DOI: 10.1038/s44161-024-00424-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Affiliation(s)
- Marie de Bakker
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Dorien M Kimenai
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK
| | - Nicholas L Mills
- BHF Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, UK.
- Usher Institute, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
20
|
Zhao P, Song S, He Z, Dai G, Liu D, Shen J, Asakawa T, Zheng M, Lu H. Development of a novel cholesterol tag-based system for trans-membrane transport of protein drugs. Biosci Trends 2024; 17:503-507. [PMID: 38072446 DOI: 10.5582/bst.2023.01285] [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: 02/02/2024]
Abstract
The main technological difficulties of developing an intracellular (transmembrane) transport system for protein drugs lie in two points: i) overcoming the barriers in the cellular membrane, and ii) loading enough protein drugs, and particularly high-dose proteins, into particles. To address these two technological problems, we recently developed a novel cholesterol tag (C-Tag)-based transmembrane transport system. This pilot study found that the C-Tag dramatically improved the cellular uptake of Fab (902-fold, vs. Fab alone) into living cells, indicating that it successfully achieved transmembrane transport. Moreover, C-Tag-mediated membrane transport was verified using micron-scale large unilamellar vesicles (LUVs, approximately 1.5 μm)-based particles. The C-Tagged Fab was able to permeate the liposomal bilayer and it greatly enhanced (a 10.1-fold increase vs. Fab alone) internalization of proteins into the LUV-based particles, indicating that the C-Tag loaded enough proteins into particles for use of high-dose proteins. Accordingly, we established a novel C-Tag-based transport system that has overcome the known technological difficulties of protein transmembrane delivery, and this might be a useful technology for drug development in the future.
Collapse
Affiliation(s)
- Pengfei Zhao
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- College of Physics and Optoelectronic Engineering, Shenzhen Key Laboratory of Photonics and Biophotonics, Key Laboratory of Optoelectronic Devices and Systems of the Ministry of Education and Guangdong Province, Shenzhen University, Shenzhen, China
| | - Shuo Song
- Shenzhen Samii Medical Center, Shenzhen, Guangdong, China
| | - Zhuojun He
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Guiqin Dai
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Deliang Liu
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Jiayin Shen
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Tetsuya Asakawa
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| | - Mingbin Zheng
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- Key Laboratory for Nanomedicine, Guangdong Medical University, Dongguan, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- Shenzhen Samii Medical Center, Shenzhen, Guangdong, China
| | - Hongzhou Lu
- Institute of Neurology, National Clinical Research Center for Infectious Diseases, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
- National Clinical Research Center for Infectious Disease, Shenzhen Clinical Medical Research Center for Tuberculosis, Institute for Hepatology, the Third People's Hospital of Shenzhen, Shenzhen, Guangdong, China
| |
Collapse
|
21
|
Liu Y, Mack JO, Shojaee M, Shaver A, George A, Clarke W, Patel N, Arroyo-Currás N. Analytical Validation of Aptamer-Based Serum Vancomycin Monitoring Relative to Automated Immunoassays. ACS Sens 2024; 9:228-235. [PMID: 38110361 PMCID: PMC10826698 DOI: 10.1021/acssensors.3c01868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/27/2023] [Accepted: 12/05/2023] [Indexed: 12/20/2023]
Abstract
The practice of monitoring therapeutic drug concentrations in patient biofluids can significantly improve clinical outcomes while simultaneously minimizing adverse side effects. A model example of this practice is vancomycin dosing in intensive care units. If dosed correctly, vancomycin can effectively treat methicillin-resistant streptococcus aureus (MRSA) infections. However, it can also induce nephrotoxicity or fail to kill the bacteria if dosed too high or too low, respectively. Although undeniably important to achieve effectiveness, therapeutic drug monitoring remains inconvenient in practice due primarily to the lengthy process of sample collection, transport to a centralized facility, and analysis using costly instrumentation. Adding to this workflow is the possibility of backlogs at centralized clinical laboratories, which is not uncommon and may result in additional delays between biofluid sampling and concentration measurement, which can negatively affect clinical outcomes. Here, we explore the possibility of using point-of-care electrochemical aptamer-based (E-AB) sensors to minimize the time delay between biofluid sampling and drug measurement. Specifically, we conducted a clinical agreement study comparing the measurement outcomes of E-AB sensors to the benchmark automated competitive immunoassays for vancomycin monitoring in serum. Our results demonstrate that E-ABs are selective for free vancomycin─the active form of the drug, over total vancomycin. In contrast, competitive immunoassays measure total vancomycin, including both protein-bound and free drug. Accounting for these differences in a pilot study consisting of 85 clinical samples, we demonstrate that the E-AB vancomycin measurement achieved a 95% positive correlation rate with the benchmark immunoassays. Therefore, we conclude that E-AB sensors could provide clinically useful stratification of patient samples at trough sampling to guide effective vancomycin dose recommendations.
Collapse
Affiliation(s)
- Yu Liu
- ZiO
Health Ltd., The Tower,
St George Wharf, London SW82BW, U.K.
| | - John O. Mack
- Biochemistry,
Cellular and Molecular Biology Program, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Maryam Shojaee
- ZiO
Health Ltd., The Tower,
St George Wharf, London SW82BW, U.K.
| | - Alexander Shaver
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Ankitha George
- ZiO
Health Ltd., The Tower,
St George Wharf, London SW82BW, U.K.
| | - William Clarke
- Department
of Pathology, Johns Hopkins University School
of Medicine, Baltimore, Maryland 21205, United States
| | - Neel Patel
- ZiO
Health Ltd., The Tower,
St George Wharf, London SW82BW, U.K.
| | - Netzahualcóyotl Arroyo-Currás
- Biochemistry,
Cellular and Molecular Biology Program, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| |
Collapse
|
22
|
Chamorro A, Rossetti M, Bagheri N, Porchetta A. Rationally Designed DNA-Based Scaffolds and Switching Probes for Protein Sensing. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2024; 187:71-106. [PMID: 38273204 DOI: 10.1007/10_2023_235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2024]
Abstract
The detection of a protein analyte and use of this type of information for disease diagnosis and physiological monitoring requires methods with high sensitivity and specificity that have to be also easy to use, rapid and, ideally, single step. In the last 10 years, a number of DNA-based sensing methods and sensors have been developed in order to achieve quantitative readout of protein biomarkers. Inspired by the speed, specificity, and versatility of naturally occurring chemosensors based on structure-switching biomolecules, significant efforts have been done to reproduce these mechanisms into the fabrication of artificial biosensors for protein detection. As an alternative, in scaffold DNA biosensors, different recognition elements (e.g., peptides, proteins, small molecules, and antibodies) can be conjugated to the DNA scaffold with high accuracy and precision in order to specifically interact with the target protein with high affinity and specificity. They have several advantages and potential, especially because the transduction signal can be drastically enhanced. Our aim here is to provide an overview of the best examples of structure switching-based and scaffold DNA sensors, as well as to introduce the reader to the rational design of innovative sensing mechanisms and strategies based on programmable functional DNA systems for protein detection.
Collapse
Affiliation(s)
| | - Marianna Rossetti
- Department of Chemistry, University of Rome Tor Vergata, Rome, Italy
| | - Neda Bagheri
- Department of Chemistry, University of Rome Tor Vergata, Rome, Italy
| | | |
Collapse
|
23
|
Gurukandure A, Somasundaram S, Kurian ASN, Khuda N, Easley CJ. Building a Nucleic Acid Nanostructure with DNA-Epitope Conjugates for a Versatile Approach to Electrochemical Protein Detection. Anal Chem 2023; 95:18122-18129. [PMID: 38032341 PMCID: PMC10720615 DOI: 10.1021/acs.analchem.3c03512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Revised: 11/10/2023] [Accepted: 11/13/2023] [Indexed: 12/01/2023]
Abstract
The recent surge of effort in nucleic-acid-based electrochemical (EC) sensors has been fruitful, yet there remains a need for more generalizable EC platforms for sensing multiple classes of clinically relevant targets. We recently reported a nucleic acid nanostructure for simple, economical, and more generalizable EC readout of a range of analytes, including small molecules, peptides, proteins, and antibodies. The nanostructure is built through on-electrode enzymatic ligation of three oligonucleotides for attachment, binding, and signaling. However, the generalizable detection of larger proteins remains a challenge. Here, we adapted the sensor to quantify larger proteins in a more generic manner through conjugating the protein's minimized antibody-binding epitope to the central DNA strand. This concept was verified using creatine kinase (CK-MM), a biomarker of muscle damage and several disorders for which rapid clinical sensing is important. DNA-epitope conjugates permitted a competitive immunoassay for the CK protein at the electrode via square-wave voltammetry (SWV). Sensing through a signal-off mechanism, the anti-CK antibody limit of detection (LOD) was 5 nM with a response time as low as 3 min. Antibody displacement by native protein analytes gave a signal-on response with the CK sensing range from the LOD of 14 nM up to 100 nM, overlapping with the normal (nonelevated) human clinical range (3-37 nM), and the sensor was validated in 98% human serum. While a need for improved DNA-epitope conjugate purification was identified, overall, this approach allows the quantification of a generic protein- or peptide-binding antibody and should facilitate future quantitative EC readouts of clinically relevant proteins that were previously inaccessible to EC techniques.
Collapse
Affiliation(s)
- Asanka Gurukandure
- Department of Chemistry and
Biochemistry, Auburn University, Auburn, Alabama 36849, United States
| | - Subramaniam Somasundaram
- Department of Chemistry and
Biochemistry, Auburn University, Auburn, Alabama 36849, United States
| | - Amanda S. N. Kurian
- Department of Chemistry and
Biochemistry, Auburn University, Auburn, Alabama 36849, United States
| | - Niamat Khuda
- Department of Chemistry and
Biochemistry, Auburn University, Auburn, Alabama 36849, United States
| | - Christopher J. Easley
- Department of Chemistry and
Biochemistry, Auburn University, Auburn, Alabama 36849, United States
| |
Collapse
|
24
|
Wang Z, Chang D, Sargent EH, Kelley SO. Apta FastZ: An Algorithm for the Rapid Identification of Aptamers with Defined Binding Affinities. Anal Chem 2023; 95:17438-17443. [PMID: 37991715 DOI: 10.1021/acs.analchem.3c02881] [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: 11/23/2023]
Abstract
Real-time biomolecular monitoring requires biosensors based on affinity reagents, such as aptamers, with moderate to low affinities for the best binding dynamics and signal gain. We recently reported Pro-SELEX, an approach that utilizes parallelized SELEX and high-content bioinformatics for the selection of aptamers with predefined binding affinities. The Pro-SELEX pipeline relies on an algorithm, termed AptaZ, that can predict the binding affinities of selected aptamers. The original AptaZ algorithm is computationally complex and slows the overall throughput of Pro-SELEX. Here, we present Apta FastZ, a rapid equivalent of AptaZ. The Apta FastZ algorithm considers the spare nature of the sequences from SELEX and is coded to avoid unnecessary comparison between sequences. As a result, Apta FastZ achieved a 10 to 40-fold faster computing speed compared to the original AptaZ algorithm while maintaining identical outcomes, allowing the bioinformatics to be completed within 1-10 h for large-scale data sets. We further validated the affinity of myeloperoxidase aptamers predicted by Apta FastZ by experiments and observed a high level of linear correlation between predicted scores and measured affinities. Taken together, the implementation of Apta FastZ could greatly accelerate the current Pro-SELEX workflow, allowing customized aptamers to be discovered within 3 days using preselected DNA libraries.
Collapse
Affiliation(s)
- Zongjie Wang
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
| | - Dingran Chang
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
| | - Edward H Sargent
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Electrical and Computer Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering, University of Toronto, Toronto M5S 3G4, Canada
| | - Shana O Kelley
- Department of Biomedical Engineering, McCormick School of Engineering, Northwestern University, Evanston, Illinois 60208, United States
- Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto M5S 3M2, Canada
- Department of Chemistry, Weinberg College of Arts & Sciences, Northwestern University, Evanston, Illinois 60208, United States
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Simpson Querrey Institute, Northwestern University, Chicago, Illinois 60611, United States
- Chan Zuckerberg Biohub Chicago, Chicago, Illinois 60607, United States
| |
Collapse
|
25
|
Myres GJ, Kitt JP, Harris JM. Raman Scattering Reveals Ion-Dependent G-Quadruplex Formation in the 15-mer Thrombin-Binding Aptamer upon Association with α-Thrombin. Anal Chem 2023; 95:16160-16168. [PMID: 37870982 DOI: 10.1021/acs.analchem.3c02751] [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: 10/25/2023]
Abstract
The discovery of DNA aptamers that bind biomolecular targets has enabled significant innovations in biosensing. Aptamers form secondary structures that exhibit selective high-affinity interactions with their binding partners. The binding of its target by an aptamer is often accompanied by conformational changes, and sensing by aptamers often relies on these changes to provide readout signals from extrinsic labels to detect target association. Many biosensing applications involve aptamers immobilized to surfaces, but methods to characterize conformations of immobilized aptamers and their in situ response have been lacking. To address this challenge, we have developed a structurally informative Raman spectroscopy method to determine conformations of the 15-mer thrombin-binding aptamer (TBA) immobilized on porous silica surfaces. The TBA is of interest because its binding of α-thrombin depends on the aptamer forming an antiparallel G-quadruplex, which is thought to drive signal changes that allow thrombin-binding to be detected. However, specific metal cations also stabilize the G-quadruplex conformation of the aptamer, even in the absence of its protein target. To develop a deeper understanding of the conformational response of the TBA, we utilize Raman spectroscopy to quantify the effects of the metal cations, K+ (stabilizing) and Li+ (nonstabilizing), on G-quadruplex versus unfolded populations of the TBA. In K+ or Li+ solutions, we then detect the association of α-thrombin with the immobilized aptamer, which can be observed in Raman scattering from the bound protein. The results show that the association of α-thrombin in K+ solutions produces no detectable change in aptamer conformation, which is found in the G-quadruplex form both before and after binding its target. In Li+ solutions, however, where the TBA is unfolded prior to α-thrombin association, protein binding occurs with the formation of a G-quadruplex by the aptamer.
Collapse
Affiliation(s)
- Grant J Myres
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| | - Jay P Kitt
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| | - Joel M Harris
- Department of Chemistry, University of Utah, 315 South 1400 East, Salt Lake City, Utah 84112-0850, United States
| |
Collapse
|
26
|
Wu G, Zhang ET, Qiang Y, Esmonde C, Chen X, Wei Z, Song Y, Zhang X, Schneider MJ, Li H, Sun H, Weng Z, Santaniello S, He J, Lai RY, Li Y, Bruchas MR, Zhang Y. Long-Term In Vivo Molecular Monitoring Using Aptamer-Graphene Microtransistors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.18.562080. [PMID: 37905115 PMCID: PMC10614860 DOI: 10.1101/2023.10.18.562080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2023]
Abstract
Long-term, real-time molecular monitoring in complex biological environments is critical for our ability to understand, prevent, diagnose, and manage human diseases. Aptamer-based electrochemical biosensors possess the promise due to their generalizability and a high degree of selectivity. Nevertheless, the operation of existing aptamer-based biosensors in vivo is limited to a few hours. Here, we report a first-generation long-term in vivo molecular monitoring platform, named aptamer-graphene microtransistors (AGMs). The AGM incorporates a layer of pyrene-(polyethylene glycol)5-alcohol and DNase inhibitor-doped polyacrylamide hydrogel coating to reduce biofouling and aptamer degradation. As a demonstration of function and generalizability, the AGM achieves the detection of biomolecules such as dopamine and serotonin in undiluted whole blood at 37 °C for 11 days. Furthermore, the AGM successfully captures optically evoked dopamine release in vivo in mice for over one week and demonstrates the capability to monitor behaviorally-induced endogenous dopamine release even after eight days of implantation in freely moving mice. The results reported in this work establish the potential for chronic aptamer-based molecular monitoring platforms, and thus serve as a new benchmark for molecular monitoring using aptamer-based technology.
Collapse
Affiliation(s)
- Guangfu Wu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Eric T. Zhang
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
| | - Yingqi Qiang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Colin Esmonde
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
| | - Xingchi Chen
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
| | - Zichao Wei
- Department of Chemistry, University of Connecticut, Storrs, CT 06269, USA
| | - Yang Song
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Xincheng Zhang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Michael J. Schneider
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Huijie Li
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - He Sun
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Zhengyan Weng
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| | - Sabato Santaniello
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
| | - Jie He
- Department of Chemistry, University of Connecticut, Storrs, CT 06269, USA
| | - Rebecca Y. Lai
- Department of Chemistry, University of Nebraska-Lincoln, Lincoln, NE 68588, USA
| | - Yan Li
- Department of Chemical and Biomedical Engineering, FAMU-FSU College of Engineering, Florida State University, Tallahassee, FL 32306, USA
| | - Michael R. Bruchas
- Department of Bioengineering, University of Washington, Seattle, WA 98195, USA
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98195, USA
- Center for Neurobiology of Addiction, Pain, and Emotion, University of Washington, Seattle, WA 98195, USA
- Department of Pharmacology, University of Washington, Seattle, WA 98195, USA
| | - Yi Zhang
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT 06269, USA
- Institute of Materials Science, University of Connecticut, Storrs, CT 06269, USA
| |
Collapse
|
27
|
Bergkamp MH, Cajigas S, van IJzendoorn LJ, Prins MWJ. Real-time continuous monitoring of dynamic concentration profiles studied with biosensing by particle motion. LAB ON A CHIP 2023; 23:4600-4609. [PMID: 37772830 DOI: 10.1039/d3lc00410d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/30/2023]
Abstract
Real-time monitoring-and-control of biological systems requires lab-on-a-chip sensors that are able to accurately measure concentration-time profiles with a well-defined time delay and accuracy using only small amounts of sampled fluid. Here, we study real-time continuous monitoring of dynamic concentration profiles in a microfluidic measurement chamber. Step functions and sinusoidal oscillations of concentrations were generated using two pumps and a herringbone mixer. Concentrations in the bulk of the measurement chamber were quantified using a solution with a dye and light absorbance measurements. Concentrations near the surface were measured using a reversible cortisol sensor based on particle motion. The experiments show how the total time delay of the real-time sensor has contributions from advection, diffusion, reaction kinetics at the surface and signal processing. The total time delay of the studied real-time cortisol sensor was ∼90 seconds for measuring 63% of the concentration change. Monitoring of sinusoidal cortisol concentration-time profiles showed that the sensor has a low-pass frequency response with a cutoff frequency of ∼4 mHz and a lag time of ∼60 seconds. The described experimental methodology paves the way for the development of monitoring-and-control in lab-on-a-chip systems and for further engineering of the analytical characteristics of real-time continuous biosensors.
Collapse
Affiliation(s)
- Max H Bergkamp
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AE Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AE Eindhoven, The Netherlands
| | | | - Leo J van IJzendoorn
- Department of Applied Physics and Science Education, Eindhoven University of Technology, 5612 AE Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AE Eindhoven, The Netherlands
| | - Menno W J Prins
- Department of Biomedical Engineering, Eindhoven University of Technology, 5612 AE Eindhoven, The Netherlands.
- Department of Applied Physics and Science Education, Eindhoven University of Technology, 5612 AE Eindhoven, The Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, 5612 AE Eindhoven, The Netherlands
- Helia Biomonitoring, 5612 AR Eindhoven, The Netherlands
| |
Collapse
|
28
|
Tsai YC, Weng WY, Yeh YT, Chien JC. Dual-Aptamer Drift Canceling Techniques to Improve Long-Term Stability of Real-Time Structure-Switching Aptasensors. ACS Sens 2023; 8:3380-3388. [PMID: 37671977 DOI: 10.1021/acssensors.3c00509] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
This paper presents a dual-aptamer scheme to mitigate signal drifts caused by structure-switching aptamers during long-term monitoring. Electrochemical aptamer-based (E-AB) biosensors have recently shown great potential for continuous in vivo monitoring. However, the accuracy of detection is often limited by signaling drifts. Traditional approaches rely on kinetic differential measurements (KDM) coupled with square-wave voltammetry to eliminate these drifts. Yet, we have discovered that KDM does not apply universally to all aptamers, as their responses at different SWV frequencies heavily rely on their structure-switching characteristics and the electron transfer (ET) kinetics of the redox reporters. In light of this, we propose a "dual-aptamer" scheme that utilizes two aptamers, each responding differently to the same target molecule to cancel out drift. These paired aptamers are identified through (1) screening from an existing pool of aptamers and (2) engineering the signaling behavior of the redox reporters. We demonstrate the differential signaling of the aptamer pair in the presence of ampicillin and ATP molecules and show that the pair exhibits similar drifts in undiluted goat serum. By implementing drift cancelation, sensor drift is reduced by a factor of 370. Additionally, the differential signaling enables an increased recording throughput by leveraging differential readout electronics. The authors believe that the proposed technique holds significant benefits for long-term in vivo monitoring.
Collapse
Affiliation(s)
- Ya-Chen Tsai
- Department of Electrical Engineering, National Taiwan University, No. 1, Section 4, Roosevelt Rd, Da'an District, Taipei City 10617, Taiwan
| | - Wei-Yang Weng
- Graduate Institute of Electronics Engineering, National Taiwan University, No. 1, Section 4, Roosevelt Rd, Da'an District, Taipei City 10617, Taiwan
| | - Yu-Tung Yeh
- Department of Electrical Engineering, National Taiwan University, No. 1, Section 4, Roosevelt Rd, Da'an District, Taipei City 10617, Taiwan
| | - Jun-Chau Chien
- Department of Electrical Engineering, National Taiwan University, No. 1, Section 4, Roosevelt Rd, Da'an District, Taipei City 10617, Taiwan
- Graduate Institute of Electronics Engineering, National Taiwan University, No. 1, Section 4, Roosevelt Rd, Da'an District, Taipei City 10617, Taiwan
| |
Collapse
|
29
|
Kim M, Panagiotakopoulou M, Chen C, Ruiz SB, Ganesh K, Tammela T, Heller DA. Micro-engineering and nano-engineering approaches to investigate tumour ecosystems. Nat Rev Cancer 2023; 23:581-599. [PMID: 37353679 PMCID: PMC10528361 DOI: 10.1038/s41568-023-00593-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/25/2023] [Indexed: 06/25/2023]
Abstract
The interactions among tumour cells, the tumour microenvironment (TME) and non-tumour tissues are of interest to many cancer researchers. Micro-engineering approaches and nanotechnologies are under extensive exploration for modelling these interactions and measuring them in situ and in vivo to investigate therapeutic vulnerabilities in cancer and extend a systemic view of tumour ecosystems. Here we highlight the greatest opportunities for improving the understanding of tumour ecosystems using microfluidic devices, bioprinting or organ-on-a-chip approaches. We also discuss the potential of nanosensors that can transmit information from within the TME or elsewhere in the body to address scientific and clinical questions about changes in chemical gradients, enzymatic activities, metabolic and immune profiles of the TME and circulating analytes. This Review aims to connect the cancer biology and engineering communities, presenting biomedical technologies that may expand the methodologies of the former, while inspiring the latter to develop approaches for interrogating cancer ecosystems.
Collapse
Affiliation(s)
- Mijin Kim
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
| | | | - Chen Chen
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Tri-Institutional PhD Program in Chemical Biology, Sloan Kettering Institute, New York, NY, USA
| | - Stephen B Ruiz
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Karuna Ganesh
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
| | - Tuomas Tammela
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA
- Cancer Biology and Genetics Program, Sloan Kettering Institute, New York, NY, USA
| | - Daniel A Heller
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA.
- Graduate School of Medical Sciences, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
30
|
Roehrich B, Leung KK, Gerson J, Kippin TE, Plaxco KW, Sepunaru L. Calibration-Free, Seconds-Resolved In Vivo Molecular Measurements using Fourier-Transform Impedance Spectroscopy Interrogation of Electrochemical Aptamer Sensors. ACS Sens 2023; 8:3051-3059. [PMID: 37584531 PMCID: PMC10463274 DOI: 10.1021/acssensors.3c00632] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 08/02/2023] [Indexed: 08/17/2023]
Abstract
Electrochemical aptamer-based (EAB) sensors are capable of measuring the concentrations of specific molecules in vivo, in real time, and with a few-second time resolution. For their signal transduction mechanism, these sensors utilize a binding-induced conformational change in their target-recognizing, redox-reporter-modified aptamer to alter the rate of electron transfer between the reporter and the supporting electrode. While a variety of voltammetric techniques have been used to monitor this change in kinetics, they suffer from various drawbacks, including time resolution limited to several seconds and sensor-to-sensor variation that requires calibration to remove. Here, however, we show that the use of fast Fourier transform electrochemical impedance spectroscopy (FFT-EIS) to interrogate EAB sensors leads to improved (here better than 2 s) time resolution and calibration-free operation, even when such sensors are deployed in vivo. To showcase these benefits, we demonstrate the approach's ability to perform real-time molecular measurements in the veins of living rats.
Collapse
Affiliation(s)
- Brian Roehrich
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Kaylyn K. Leung
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa Barbara, California 93106, United States
- Center
for Bioengineering, University of California
Santa Barbara, Santa Barbara, California 93106, United States
| | - Julian Gerson
- Department
of Psychological and Brain Sciences, University
of California, Santa Barbara, California 93106, United States
- Center
for Bioengineering, University of California
Santa Barbara, Santa Barbara, California 93106, United States
| | - Tod E. Kippin
- Department
of Psychological and Brain Sciences, University
of California, Santa Barbara, California 93106, United States
- Department
of Molecular Cellular and Developmental Biology, University of California, Santa
Barbara, California 93106,United States
| | - Kevin W. Plaxco
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa Barbara, California 93106, United States
- Center
for Bioengineering, University of California
Santa Barbara, Santa Barbara, California 93106, United States
| | - Lior Sepunaru
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
31
|
Kurian ASN, Gurukandure A, Dovgan I, Kolodych S, Easley CJ. Thermofluorimetric Analysis (TFA) using Probes with Flexible Spacers: Application to Direct Antibody Sensing and to Antibody-Oligonucleotide (AbO) Conjugate Valency Monitoring. Anal Chem 2023; 95:11680-11686. [PMID: 37490525 PMCID: PMC10421636 DOI: 10.1021/acs.analchem.3c01590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2023]
Abstract
Antibodies have long been recognized as clinically relevant biomarkers of disease. The onset of a disease often stimulates antibody production in low quantities, making it crucial to develop sensitive, specific, and easy-to-use antibody assay platforms. Antibodies are also extensively used as probes in bioassays, and there is a need for simpler methods to evaluate specialized probes, such as antibody-oligonucleotide (AbO) conjugates. Previously, we demonstrated that thermofluorimetric analysis (TFA) of analyte-driven DNA assembly can be leveraged to detect protein biomarkers using AbO probes. A key advantage of this technique is its ability to circumvent autofluorescence arising from biological samples, which otherwise hampers homogeneous assays. The analysis of differential DNA melt curves (dF/dT) successfully distinguishes the signal from the background and interferences. Expanding the applicability of TFA further, herein we demonstrate a unique proximity based TFA assay for antibody quantification that is functional in 90% human plasma. We show that the conformational flexibility of the DNA-based proximity probes is critically important for optimal performance in these assays. To promote stable, proximity-induced hybridization of the short DNA strands, substitution of poly(ethylene glycol) (PEG) spacers in place of ssDNA segments led to improved conformational flexibility and sensor performance. Finally, by applying these flexible spacers to study AbO conjugates directly, we validate this modified TFA approach as a novel tool to elucidate the probe valency, clearly distinguishing between monovalent and multivalent AbOs and reducing the reagent amounts by 12-fold.
Collapse
Affiliation(s)
- Amanda S. N. Kurian
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL, 36849
| | - Asanka Gurukandure
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL, 36849
| | | | | | | |
Collapse
|
32
|
Flatebo C, Conkright WR, Beckner ME, Batchelor RH, Kippin TE, Heikenfeld J, Plaxco KW. Efforts toward the continuous monitoring of molecular markers of performance. J Sci Med Sport 2023; 26 Suppl 1:S46-S53. [PMID: 36841706 DOI: 10.1016/j.jsams.2023.01.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 01/04/2023] [Accepted: 01/28/2023] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Technologies supporting the continuous, real-time measurement of blood oxygen saturation and plasma glucose levels have improved our ability to monitor performance status. Our ability to monitor other molecular markers of performance, however, including the hormones known to indicate overtraining and general health, has lagged. That is, although a number of other molecular markers of performance status have been identified, we have struggled to develop viable technologies supporting their real-time monitoring in the body. Here we review biosensor approaches that may support such measurements, as well as the molecules potentially of greatest interest to monitor. DESIGN Narrative literature review. METHOD Literature review. RESULTS Significant effort has been made to harness the specificity, affinity, and generalizability of biomolecular recognition in a platform technology supporting continuous in vivo molecular measurements. Most biosensor approaches, however, are either not generalizable to most targets, or fail when challenged in the complex environments found in vivo. Electrochemical aptamer-based sensors, in contrast, are the first technology to simultaneously achieve both of these critical attributes. In an effort to illustrate the potential of this platform technology, we both critically review the literature describing it and briefly survey some of the molecular performance markers we believe will prove advantageous to monitor using it. CONCLUSIONS Electrochemical aptamer-based sensors may be the first truly generalizable technology for monitoring specific molecules in situ in the body and how adaptation of the platform to subcutaneous microneedles will enable the real-time monitoring of performance markers via a wearable, minimally invasive device.
Collapse
Affiliation(s)
- Charlotte Flatebo
- Institute for Collaborative Biotechnologies, University of California Santa Barbara, USA
| | | | | | | | - Tod E Kippin
- Neuroscience Research Institute, Department of Psychological and Brain Sciences, University of California Santa Barbara, USA
| | - Jason Heikenfeld
- Biomedical, Electrical, and Chemical Engineering, Director Novel Devices Laboratory, University of Cincinnati, USA
| | - Kevin W Plaxco
- Institute for Collaborative Biotechnologies, University of California Santa Barbara, USA; Department of Chemistry and Biochemistry, Biological Engineering Graduate Program, University of California Santa Barbara, USA.
| |
Collapse
|
33
|
Young T, Clark V, Arroyo-Currás N, Heikenfeld J. Perspective-The Feasibility of Continuous Protein Monitoring in Interstitial Fluid. ECS SENSORS PLUS 2023; 2:027001. [PMID: 37128505 PMCID: PMC10140668 DOI: 10.1149/2754-2726/accd7e] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/09/2023] [Indexed: 05/03/2023]
Abstract
Real-time continuous monitoring of proteins in-vivo holds great potential for personalized medical applications. Unfortunately, a prominent knowledge gap exists in the fundamental biology regarding protein transfer and correlation between interstitial fluid and blood. Additionally, technological sensing will require affinity-based platforms that cannot be robustly protected in-vivo and will therefore be challenged in sensitivity, longevity, and fouling over multi-day to week timelines. Here we use electrochemical aptamer sensors as a model system to discuss further research necessary to achieve continuous protein sensing.
Collapse
Affiliation(s)
- Thomas Young
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, United States of America
| | - Vincent Clark
- Chemistry-Biology Interface Program, Zanvyl Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, MD 21218, United States of America
| | - Netzahualcóyotl Arroyo-Currás
- Chemistry-Biology Interface Program, Zanvyl Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, MD 21218, United States of America
| | - Jason Heikenfeld
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH 45221, United States of America
- Department of Electrical & Computer Engineering, University of Cincinnati, Cincinnati, OH 45221, United States of America
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States of America
| |
Collapse
|
34
|
Shetty SS, Moosa B, Zhang L, Alshankiti B, Baslyman W, Mani V, Khashab NM, Salama KN. Polyoxometalate-cyclodextrin supramolecular entities for real-time in situ monitoring of dopamine released from neuroblastoma cells. Biosens Bioelectron 2023; 229:115240. [PMID: 36963326 DOI: 10.1016/j.bios.2023.115240] [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: 12/06/2022] [Revised: 02/27/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023]
Abstract
Optimized and sensitive biomarker detection has recently been shown to have a critical impact on quality of diagnosis and medical care options. In this research study, polyoxometalate-γ-cyclodextrin metal-organic framework (POM-γCD MOF) was utilized as an electrocatalyst to fabricate highly selective sensors to detect in-situ released dopamine. The POM-γCD MOF produced multiple modes of signals for dopamine including electrochemical, colorimetric, and smartphone read-outs. Real-time quantitative monitoring of SH-SY5Y neuroblastoma cellular dopamine production was successfully demonstrated under various stimuli at different time intervals. The POM-CD MOF sensor and linear regression model were used to develop a smartphone read-out platform, which converts dopamine visual signals to digital signals within a few seconds. Ultimately, POM-γCD MOFs can play a significant role in the diagnosis and treatment of various diseases that involve dopamine as a significant biomarker.
Collapse
Affiliation(s)
- Saptami Suresh Shetty
- Sensors Lab, Advanced Membranes and Porous Materials Center, Computer, Electrical and Mathematical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Basem Moosa
- Smart Hybrid Materials Research Group (SHMs), Advanced Membranes and Porous Materials Center (AMPMC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Li Zhang
- Sensors Lab, Advanced Membranes and Porous Materials Center, Computer, Electrical and Mathematical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia
| | - Buthainah Alshankiti
- Smart Hybrid Materials Research Group (SHMs), Advanced Membranes and Porous Materials Center (AMPMC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Walaa Baslyman
- Smart Hybrid Materials Research Group (SHMs), Advanced Membranes and Porous Materials Center (AMPMC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia
| | - Veerappan Mani
- Sensors Lab, Advanced Membranes and Porous Materials Center, Computer, Electrical and Mathematical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| | - Niveen M Khashab
- Smart Hybrid Materials Research Group (SHMs), Advanced Membranes and Porous Materials Center (AMPMC), King Abdullah University of Science and Technology (KAUST), Thuwal, 23955, Saudi Arabia.
| | - Khaled Nabil Salama
- Sensors Lab, Advanced Membranes and Porous Materials Center, Computer, Electrical and Mathematical Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, 23955-6900, Saudi Arabia.
| |
Collapse
|
35
|
Parolo C, Idili A, Heikenfeld J, Plaxco KW. Conformational-switch biosensors as novel tools to support continuous, real-time molecular monitoring in lab-on-a-chip devices. LAB ON A CHIP 2023; 23:1339-1348. [PMID: 36655710 PMCID: PMC10799767 DOI: 10.1039/d2lc00716a] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Recent years have seen continued expansion of the functionality of lab on a chip (LOC) devices. Indeed LOCs now provide scientists and developers with useful and versatile platforms across a myriad of chemical and biological applications. The field still fails, however, to integrate an often important element of bench-top analytics: real-time molecular measurements that can be used to "guide" a chemical response. Here we describe the analytical techniques that could provide LOCs with such real-time molecular monitoring capabilities. It appears to us that, among the approaches that are general (i.e., that are independent of the reactive or optical properties of their targets), sensing strategies relying on binding-induced conformational change of bioreceptors are most likely to succeed in such applications.
Collapse
Affiliation(s)
- Claudio Parolo
- Barcelona Institute for Global Health, Hospital Clínic Universitat de Barcelona, 08036, Barcelona, Spain
| | - Andrea Idili
- Department of Chemical Science and Technologies, University of Rome, Tor Vergata, 00133 Rome, Italy
| | - Jason Heikenfeld
- Novel Devices Laboratory, University of Cincinnati, Cincinnati, Ohio, USA
| | - Kevin W Plaxco
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, California, USA.
- Interdepartmental Program in Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, California, USA
| |
Collapse
|
36
|
Khuda N, Somasundaram S, Urgunde AB, Easley CJ. Ionic Strength and Hybridization Position near Gold Electrodes Can Significantly Improve Kinetics in DNA-Based Electrochemical Sensors. ACS APPLIED MATERIALS & INTERFACES 2023; 15:5019-5027. [PMID: 36661270 PMCID: PMC10370289 DOI: 10.1021/acsami.2c22741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
A variety of electrochemical (EC) biosensors play critical roles in disease diagnostics. More recently, DNA-based EC sensors have been established as promising for detecting a wide range of analyte classes. Since most of these sensors rely on the high specificity of DNA hybridization for analyte binding or structural control, it is crucial to understand the kinetics of hybridization at the electrode surface. In this work, we have used methylene blue-labeled DNA strands to monitor the kinetics of DNA hybridization at the electrode surface with square-wave voltammetry. By varying the position of the double-stranded DNA segment relative to the electrode surface as well as the bulk solution's ionic strength (0.125-1.00 M), we observed significant interferences with DNA hybridization closer to the surface, with more substantial interference at lower ionic strength. As a demonstration of the effect, toehold-mediated strand displacement reactions were slowed and diminished close to the surface, while strategic placement of the DNA binding site improved reaction rates and yields. This work manifests that both the salt concentration and DNA hybridization site relative to the electrode are important factors to consider when designing DNA-based EC sensors that measure hybridization directly at the electrode surface.
Collapse
Affiliation(s)
- Niamat Khuda
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL 36849, USA
| | | | - Ajay B. Urgunde
- Department of Chemistry and Biochemistry, Auburn University, Auburn, AL 36849, USA
| | | |
Collapse
|
37
|
Dong T, Matos Pires NM, Yang Z, Jiang Z. Advances in Electrochemical Biosensors Based on Nanomaterials for Protein Biomarker Detection in Saliva. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2205429. [PMID: 36585368 PMCID: PMC9951322 DOI: 10.1002/advs.202205429] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 11/20/2022] [Indexed: 06/02/2023]
Abstract
The focus on precise medicine enhances the need for timely diagnosis and frequent monitoring of chronic diseases. Moreover, the recent pandemic of severe acute respiratory syndrome coronavirus 2 poses a great demand for rapid detection and surveillance of viral infections. The detection of protein biomarkers and antigens in the saliva allows rapid identification of diseases or disease changes in scenarios where and when the test response at the point of care is mandated. While traditional methods of protein testing fail to provide the desired fast results, electrochemical biosensors based on nanomaterials hold perfect characteristics for the detection of biomarkers in point-of-care settings. The recent advances in electrochemical sensors for salivary protein detection are critically reviewed in this work, with emphasis on the role of nanomaterials to boost the biosensor analytical performance and increase the reliability of the test in human saliva samples. Furthermore, this work identifies the critical factors for further modernization of the nanomaterial-based electrochemical sensors, envisaging the development and implementation of next-generation sample-in-answer-out systems.
Collapse
Affiliation(s)
- Tao Dong
- Department of Microsystems‐ IMSFaculty of TechnologyNatural Sciences and Maritime SciencesUniversity of South‐Eastern Norway‐USNP.O. Box 235Kongsberg3603Norway
| | - Nuno Miguel Matos Pires
- Chongqing Key Laboratory of Micro‐Nano Systems and Intelligent TransductionCollaborative Innovation Center on Micro‐Nano Transduction and Intelligent Eco‐Internet of ThingsChongqing Key Laboratory of Colleges and Universities on Micro‐Nano Systems Technology and Smart TransducingNational Research Base of Intelligent Manufacturing ServiceChongqing Technology and Business UniversityNan'an DistrictChongqing400067China
| | - Zhaochu Yang
- Chongqing Key Laboratory of Micro‐Nano Systems and Intelligent TransductionCollaborative Innovation Center on Micro‐Nano Transduction and Intelligent Eco‐Internet of ThingsChongqing Key Laboratory of Colleges and Universities on Micro‐Nano Systems Technology and Smart TransducingNational Research Base of Intelligent Manufacturing ServiceChongqing Technology and Business UniversityNan'an DistrictChongqing400067China
| | - Zhuangde Jiang
- Chongqing Key Laboratory of Micro‐Nano Systems and Intelligent TransductionCollaborative Innovation Center on Micro‐Nano Transduction and Intelligent Eco‐Internet of ThingsChongqing Key Laboratory of Colleges and Universities on Micro‐Nano Systems Technology and Smart TransducingNational Research Base of Intelligent Manufacturing ServiceChongqing Technology and Business UniversityNan'an DistrictChongqing400067China
- State Key Laboratory for Manufacturing Systems EngineeringInternational Joint Laboratory for Micro/Nano Manufacturing and Measurement TechnologyXi'an Jiaotong UniversityXi'an710049China
| |
Collapse
|
38
|
Dey S, Dolci M, Zijlstra P. Single-Molecule Optical Biosensing: Recent Advances and Future Challenges. ACS PHYSICAL CHEMISTRY AU 2023; 3:143-156. [PMID: 36968450 PMCID: PMC10037498 DOI: 10.1021/acsphyschemau.2c00061] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 12/22/2022] [Accepted: 12/22/2022] [Indexed: 01/13/2023]
Abstract
In recent years, the sensitivity and specificity of optical sensors has improved tremendously due to improvements in biochemical functionalization protocols and optical detection systems. As a result, single-molecule sensitivity has been reported in a range of biosensing assay formats. In this Perspective, we summarize optical sensors that achieve single-molecule sensitivity in direct label-free assays, sandwich assays, and competitive assays. We describe the advantages and disadvantages of single-molecule assays and summarize future challenges in the field including their optical miniaturization and integration, multimodal sensing capabilities, accessible time scales, and compatibility with real-life matrices such as biological fluids. We conclude by highlighting the possible application areas of optical single-molecule sensors that include not only healthcare but also the monitoring of the environment and industrial processes.
Collapse
Affiliation(s)
- Swayandipta Dey
- Eindhoven University of Technology, Department of Applied Physics, Eindhoven 5600 MB, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven, 5600 MB, The Netherlands
- Eindhoven Hendrik Casimir Institute, Eindhoven, 5600 MB, The Netherlands
| | - Mathias Dolci
- Eindhoven University of Technology, Department of Applied Physics, Eindhoven 5600 MB, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven, 5600 MB, The Netherlands
- Eindhoven Hendrik Casimir Institute, Eindhoven, 5600 MB, The Netherlands
| | - Peter Zijlstra
- Eindhoven University of Technology, Department of Applied Physics, Eindhoven 5600 MB, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven, 5600 MB, The Netherlands
- Eindhoven Hendrik Casimir Institute, Eindhoven, 5600 MB, The Netherlands
| |
Collapse
|
39
|
Punetha A, Kotiya D. Advancements in Oncoproteomics Technologies: Treading toward Translation into Clinical Practice. Proteomes 2023; 11:2. [PMID: 36648960 PMCID: PMC9844371 DOI: 10.3390/proteomes11010002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 01/03/2023] [Accepted: 01/04/2023] [Indexed: 01/12/2023] Open
Abstract
Proteomics continues to forge significant strides in the discovery of essential biological processes, uncovering valuable information on the identity, global protein abundance, protein modifications, proteoform levels, and signal transduction pathways. Cancer is a complicated and heterogeneous disease, and the onset and progression involve multiple dysregulated proteoforms and their downstream signaling pathways. These are modulated by various factors such as molecular, genetic, tissue, cellular, ethnic/racial, socioeconomic status, environmental, and demographic differences that vary with time. The knowledge of cancer has improved the treatment and clinical management; however, the survival rates have not increased significantly, and cancer remains a major cause of mortality. Oncoproteomics studies help to develop and validate proteomics technologies for routine application in clinical laboratories for (1) diagnostic and prognostic categorization of cancer, (2) real-time monitoring of treatment, (3) assessing drug efficacy and toxicity, (4) therapeutic modulations based on the changes with prognosis and drug resistance, and (5) personalized medication. Investigation of tumor-specific proteomic profiles in conjunction with healthy controls provides crucial information in mechanistic studies on tumorigenesis, metastasis, and drug resistance. This review provides an overview of proteomics technologies that assist the discovery of novel drug targets, biomarkers for early detection, surveillance, prognosis, drug monitoring, and tailoring therapy to the cancer patient. The information gained from such technologies has drastically improved cancer research. We further provide exemplars from recent oncoproteomics applications in the discovery of biomarkers in various cancers, drug discovery, and clinical treatment. Overall, the future of oncoproteomics holds enormous potential for translating technologies from the bench to the bedside.
Collapse
Affiliation(s)
- Ankita Punetha
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Rutgers University, 225 Warren St., Newark, NJ 07103, USA
| | - Deepak Kotiya
- Department of Pharmacology and Nutritional Sciences, University of Kentucky, 900 South Limestone St., Lexington, KY 40536, USA
| |
Collapse
|
40
|
Abdrabou A, Duong BTV, Chen K, Atwal RS, Labib M, Lin S, Angers S, Kelley SO. nuPRISM: Microfluidic Genome-Wide Phenotypic Screening Platform for Cellular Nuclei. ACS CENTRAL SCIENCE 2022; 8:1618-1626. [PMID: 36589880 PMCID: PMC9801500 DOI: 10.1021/acscentsci.2c00836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Indexed: 06/17/2023]
Abstract
Genome-wide loss-of-function screens are critical tools to identify novel genetic regulators of intracellular proteins. However, studying the changes in the organelle-specific expression profile of intracellular proteins can be challenging due to protein localization differences across the whole cell, hindering context-dependent protein expression and activity analyses. Here, we describe nuPRISM, a microfluidics chip specifically designed for large-scale isolated nuclei sorting. The new device enables rapid genome-wide loss-of-function phenotypic CRISPR-Cas9 screens directed at intranuclear targets. We deployed this technology to identify novel genetic regulators of β-catenin nuclear accumulation, a phenotypic hallmark of APC-mutated colorectal cancer. nuPRISM expands our ability to capture aberrant nuclear morphological and functional traits associated with distinctive signal transduction and subcellular localization-driven functional processes with substantial resolution and high throughput.
Collapse
Affiliation(s)
- Abdalla
M. Abdrabou
- Department
of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Bill T. V. Duong
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Kangfu Chen
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| | - Randy Singh Atwal
- Department
of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
| | - Mahmoud Labib
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60611, United States
| | - Sichun Lin
- Terrence
Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Stephane Angers
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
- Department
of Biochemistry, Faculty of Medicine, University
of Toronto, Toronto, Ontario M5S 1A8, Canada
- Terrence
Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Toronto, Ontario M5S 3E1, Canada
| | - Shana O. Kelley
- Department
of Biochemistry and Molecular Genetics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, United States
- Department
of Chemistry, Northwestern University, Evanston, Illinois 60611, United States
- Department
of Biomedical Engineering, Northwestern
University, Evanston, Illinois 60611, United States
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario M5S 3M2, Canada
| |
Collapse
|
41
|
Shaver A, Mahlum J, Scida K, Johnston ML, Aller Pellitero M, Wu Y, Carr GV, Arroyo-Currás N. Optimization of Vancomycin Aptamer Sequence Length Increases the Sensitivity of Electrochemical, Aptamer-Based Sensors In Vivo. ACS Sens 2022; 7:3895-3905. [PMID: 36417705 PMCID: PMC9791989 DOI: 10.1021/acssensors.2c01910] [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] [Received: 08/31/2022] [Accepted: 11/14/2022] [Indexed: 11/24/2022]
Abstract
The measurement of serum vancomycin levels at the clinic is critical to optimizing dosing given the narrow therapeutic window of this antibiotic. Current approaches to quantitate serum vancomycin levels are based on immunoassays, which are multistep methods requiring extensive processing of patient samples. As an alternative, vancomycin-binding electrochemical, aptamer-based sensors (E-ABs) were developed to simplify the workflow of vancomycin monitoring. E-ABs enable the instantaneous measurement of serum vancomycin concentrations without the need for sample dilution or other processing steps. However, the originally reported vancomycin-binding E-ABs had a dissociation constant of 45 μM, which is approximately 1 order of magnitude higher than the recommended trough concentrations of vancomycin measured in patients. This limited sensitivity hinders the ability of E-ABs to accurately support vancomycin monitoring. To overcome this problem, here we sought to optimize the length of the vancomycin-binding aptamer sequence to enable a broader dynamic range in the E-AB platform. Our results demonstrate, via isothermal calorimetry and E-AB calibrations in undiluted serum, that superior affinity and near-equal sensor gain in vitro can be achieved using a one-base-pair-longer aptamer than the truncated sequence originally reported. We validate the impact of the improved binding affinity in vivo by monitoring vancomycin levels in the brain cortex of live mice following intravenous administration. While the original sequence fails to resolve vancomycin concentrations from baseline noise (SNR = 1.03), our newly reported sequence provides an SNR of 1.62 at the same dose.
Collapse
Affiliation(s)
- Alexander Shaver
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - J.D. Mahlum
- Chemistry-Biology
Interface Program, Zanvyl Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Karen Scida
- Lieber
Institute for Brain Development, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21205, United States
| | - Melanie L. Johnston
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Biochemistry,
Cellular and Molecular Biology, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21287, United States
| | - Miguel Aller Pellitero
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Yao Wu
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Gregory V. Carr
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Lieber
Institute for Brain Development, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21205, United States
| | - Netzahualcóyotl Arroyo-Currás
- Department
of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Chemistry-Biology
Interface Program, Zanvyl Krieger School of Arts & Sciences, Johns Hopkins University, Baltimore, Maryland 21218, United States
- Biochemistry,
Cellular and Molecular Biology, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21287, United States
| |
Collapse
|
42
|
Yang J, Gan X, Song X, Yuan R, Xiang Y. Apamer-based sensitive and label-free electrochemical detection of neutrophil gelatinase-associated lipocalin via recycling amplification cascades. Anal Chim Acta 2022; 1233:340515. [DOI: 10.1016/j.aca.2022.340515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/26/2022] [Accepted: 10/10/2022] [Indexed: 11/15/2022]
|
43
|
Downs AM, Plaxco KW. Real-Time, In Vivo Molecular Monitoring Using Electrochemical Aptamer Based Sensors: Opportunities and Challenges. ACS Sens 2022; 7:2823-2832. [PMID: 36205360 PMCID: PMC9840907 DOI: 10.1021/acssensors.2c01428] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The continuous, real-time measurement of specific molecules in situ in the body would greatly improve our ability to understand, diagnose, and treat disease. The vast majority of continuous molecular sensing technologies, however, either (1) rely on the chemical or enzymatic reactivity of their targets, sharply limiting their scope, or (2) have never been shown (and likely will never be shown) to operate in the complex environments found in vivo. Against this background, here we review electrochemical aptamer-based (EAB) sensors, an electrochemical approach to real-time molecular monitoring that has now seen 15 years of academic development. The strengths of the EAB platform are significant: to date it is the only molecular measurement technology that (1) functions independently of the chemical reactivity of its targets, and is thus general, and (2) supports in vivo measurements. Specifically, using EAB sensors we, and others, have already reported the real-time, seconds-resolved measurements of multiple, unrelated drugs and metabolites in situ in the veins and tissues of live animals. Against these strengths, we detail the platform's remaining weaknesses, which include still limited measurement duration (hours, rather than the more desirable days) and the difficulty in obtaining sufficiently high performance aptamers against new targets, before then detailing promising approaches overcoming these hurdles. Finally, we close by exploring the opportunities we believe this potentially revolutionary technology (as well as a few, possibly competing, technologies) will create for both researchers and clinicians.
Collapse
Affiliation(s)
- Alex M. Downs
- Sandia National Laboratories, Albuquerque, NM 87106, USA
| | - Kevin W. Plaxco
- Center for Bioengineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA,Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA,Corresponding author:
| |
Collapse
|
44
|
Chamorro-Garcia A, Parolo C, Ortega G, Idili A, Green J, Ricci F, Plaxco KW. The sequestration mechanism as a generalizable approach to improve the sensitivity of biosensors and bioassays. Chem Sci 2022; 13:12219-12228. [PMID: 36349092 PMCID: PMC9601244 DOI: 10.1039/d2sc03901j] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/16/2022] [Indexed: 07/25/2023] Open
Abstract
Biosensors and bioassays, both of which employ proteins and nucleic acids to detect specific molecular targets, have seen significant applications in both biomedical research and clinical practice. This success is largely due to the extraordinary versatility, affinity, and specificity of biomolecular recognition. Nevertheless, these receptors suffer from an inherent limitation: single, saturable binding sites exhibit a hyperbolic relationship (the "Langmuir isotherm") between target concentration and receptor occupancy, which in turn limits the sensitivity of these technologies to small variations in target concentration. To overcome this and generate more responsive biosensors and bioassays, here we have used the sequestration mechanism to improve the steepness of the input/output curves of several bioanalytical methods. As our test bed for this we employed sensors and assays against neutrophil gelatinase-associated lipocalin (NGAL), a kidney biomarker for which enhanced sensitivity will improve the monitoring of kidney injury. Specifically, by introducing sequestration we have improved the responsiveness of an electrochemical aptamer based (EAB) biosensor, and two bioassays, a paper-based "dipstick" assay and an enzyme-linked immunosorbent assay (ELISA). Doing so we have narrowed the dynamic range of these sensors and assays several-fold, thus enhancing their ability to measure small changes in target concentration. Given that introducing sequestration requires only the addition of the appropriate concentration of a high-affinity "depletant," the mechanism appears simple and easily adaptable to tuning the binding properties of the receptors employed in a wide range of biosensors and bioassays.
Collapse
Affiliation(s)
- Alejandro Chamorro-Garcia
- Department of Chemistry and Biochemistry University of California Santa Barbara (UCSB) Santa Barbara CA 93106 USA
- Dipartimento di Scienze e Tecnologie Chimiche, University of Rome, Tor Vergata, Via della Ricerca Scientifica 00133 Rome Italy
| | - Claudio Parolo
- ISGlobal-Barcelona Institute for Global Health Carrer del Rosselló 132 08036 Barcelona Spain
| | - Gabriel Ortega
- Ikerbasque, Basque Foundation for Science 48013 Bilbao Spain
- Precision Medicine and Metabolism Laboratory, CIC BioGUNE, Basque Research and Technology Alliance, Parque Tecnológico de Bizkaia 48160 Derio Spain
| | - Andrea Idili
- Dipartimento di Scienze e Tecnologie Chimiche, University of Rome, Tor Vergata, Via della Ricerca Scientifica 00133 Rome Italy
| | - Joshua Green
- Department of Chemistry and Biochemistry University of California Santa Barbara (UCSB) Santa Barbara CA 93106 USA
| | - Francesco Ricci
- Dipartimento di Scienze e Tecnologie Chimiche, University of Rome, Tor Vergata, Via della Ricerca Scientifica 00133 Rome Italy
| | - Kevin W Plaxco
- Department of Chemistry and Biochemistry University of California Santa Barbara (UCSB) Santa Barbara CA 93106 USA
| |
Collapse
|
45
|
Buskermolen AD, Lin YT, van Smeden L, van Haaften RB, Yan J, Sergelen K, de Jong AM, Prins MWJ. Continuous biomarker monitoring with single molecule resolution by measuring free particle motion. Nat Commun 2022; 13:6052. [PMID: 36229441 PMCID: PMC9561105 DOI: 10.1038/s41467-022-33487-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022] Open
Abstract
There is a need for sensing technologies that can continuously monitor concentration levels of critical biomolecules in applications such as patient care, fundamental biological research, biotechnology and food industry, as well as the environment. However, it is fundamentally difficult to develop measurement technologies that are not only sensitive and specific, but also allow monitoring over a broad concentration range and over long timespans. Here we describe a continuous biomolecular sensing methodology based on the free diffusion of biofunctionalized particles hovering over a sensor surface. The method records digital events due to single-molecule interactions and enables biomarker monitoring at picomolar to micromolar concentrations without consuming any reagents. We demonstrate the affinity-based sensing methodology for DNA-based sandwich and competition assays, and for an antibody-based cortisol assay. Additionally, the sensor can be dried, facilitating storage over weeks while maintaining its sensitivity. We foresee that this will enable the development of continuous monitoring sensors for applications in fundamental research, for studies on organs on a chip, for the monitoring of patients in critical care, and for the monitoring of industrial processes and bioreactors as well as ecological systems.
Collapse
Affiliation(s)
- Alissa D. Buskermolen
- grid.6852.90000 0004 0398 8763Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands ,grid.6852.90000 0004 0398 8763Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Yu-Ting Lin
- grid.6852.90000 0004 0398 8763Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands ,grid.6852.90000 0004 0398 8763Department of Applied Physics, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Laura van Smeden
- grid.6852.90000 0004 0398 8763Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands ,grid.6852.90000 0004 0398 8763Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Rik B. van Haaften
- grid.6852.90000 0004 0398 8763Department of Applied Physics, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Junhong Yan
- Helia Biomonitoring, Eindhoven, the Netherlands
| | - Khulan Sergelen
- grid.6852.90000 0004 0398 8763Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands ,grid.6852.90000 0004 0398 8763Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Arthur M. de Jong
- grid.6852.90000 0004 0398 8763Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands ,grid.6852.90000 0004 0398 8763Department of Applied Physics, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Menno W. J. Prins
- grid.6852.90000 0004 0398 8763Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands ,grid.6852.90000 0004 0398 8763Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven, the Netherlands ,grid.6852.90000 0004 0398 8763Department of Applied Physics, Eindhoven University of Technology, Eindhoven, the Netherlands ,Helia Biomonitoring, Eindhoven, the Netherlands
| |
Collapse
|
46
|
Electrochemical Microneedles: Innovative Instruments in Health Care. BIOSENSORS 2022; 12:bios12100801. [PMID: 36290938 PMCID: PMC9599258 DOI: 10.3390/bios12100801] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 09/21/2022] [Accepted: 09/24/2022] [Indexed: 11/16/2022]
Abstract
As a significant part of drug therapy, the mode of drug transport has attracted worldwide attention. Efficient drug delivery methods not only markedly improve the drug absorption rate, but also reduce the risk of infection. Recently, microneedles have combined the advantages of subcutaneous injection administration and transdermal patch administration, which is not only painless, but also has high drug absorption efficiency. In addition, microneedle-based electrochemical sensors have unique capabilities for continuous health state monitoring, playing a crucial role in the real-time monitoring of various patient physiological indicators. Therefore, they are commonly applied in both laboratories and hospitals. There are a variety of reports regarding electrochemical microneedles; however, the comprehensive introduction of new electrochemical microneedles is still rare. Herein, significant work on electrochemical microneedles over the past two years is summarized, and the main challenges faced by electrochemical microneedles and future development directions are proposed.
Collapse
|
47
|
Chozinski T, Ferguson BS, Fisher W, Ge S, Gong Q, Kang H, McDermott J, Scott A, Shi W, Trausch JJ, Verch T, Vukovich M, Wang J, Wu JE, Yang Q. Development of an Aptamer-Based Electrochemical Microfluidic Device for Viral Vaccine Quantitation. Anal Chem 2022; 94:6146-6155. [PMID: 35410467 DOI: 10.1021/acs.analchem.1c05093] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Global deployment of vaccines poses significant challenges in the distribution and use of the accompanying immunoassays, one of the standard methods for quality control of vaccines, particularly when establishing assays in countries worldwide to support testing/release upon importation. This work describes our effort toward developing an integrated, portable device to carry out affinity assays for viral particles quantification in viral vaccines by incorporating (i) aptamers, (ii) microfluidic devices, and (iii) electrochemical detection. We generated and characterized more than eight aptamers against multiple membrane proteins of cytomegalovirus (CMV), which we used as a model system and designed and fabricated electrochemical microfluidic devices to measure CMV concentrations in a candidate vaccine under development. The aptamer-based assays provided a half maximal effective concentration, EC50, of 12 U/mL, comparable to that of an ELISA using a pair of antibodies (EC50 60 U/mL). The device measured relative CMV concentrations accurately (within ±10% bias) and precisely (11%, percent relative standard deviation). This work represents the critical first steps toward developing simple, affordable, and robust affinity assays for global deployment without the need for sensitive equipment and extensive analyst training.
Collapse
Affiliation(s)
- Tyler Chozinski
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - B Scott Ferguson
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - William Fisher
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - Shencheng Ge
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Qiang Gong
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - Hui Kang
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - John McDermott
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - Alexander Scott
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - Wentao Shi
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - Jeremiah J Trausch
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Thorsten Verch
- Merck & Co., Inc., 770 Sumneytown Pike, West Point, Pennsylvania 19486, United States
| | - Matthew Vukovich
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - Jinpeng Wang
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - J Emma Wu
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| | - Qin Yang
- Aptitude Medical Systems, 125 Cremona Drive, Suite 100, Goleta, California 93117, United States
| |
Collapse
|
48
|
Mamun AA, Zhao F. In-Plane Si Microneedles: Fabrication, Characterization, Modeling and Applications. MICROMACHINES 2022; 13:657. [PMID: 35630124 PMCID: PMC9146885 DOI: 10.3390/mi13050657] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Revised: 04/16/2022] [Accepted: 04/17/2022] [Indexed: 01/26/2023]
Abstract
Microneedles are getting more and more attention in research and commercialization since their advancement in the 1990s due to the advantages over traditional hypodermic needles such as minimum invasiveness, low material and fabrication cost, and precise needle geometry control, etc. The design and fabrication of microneedles depend on various factors such as the type of materials used, fabrication planes and techniques, needle structures, etc. In the past years, in-plane and out-of-plane microneedle technologies made by silicon (Si), polymer, metal, and other materials have been developed for numerous biomedical applications including drug delivery, sample collections, medical diagnostics, and bio-sensing. Among these microneedle technologies, in-plane Si microneedles excel by the inherent properties of Si such as mechanical strength, wear resistance, biocompatibility, and structural advantages of in-plane configuration such as a wide range of length, readiness of integration with other supporting components, and complementary metal-oxide-semiconductor (CMOS) compatible fabrication. This article aims to provide a review of in-plane Si microneedles with a focus on fabrication techniques, theoretical and numerical analysis, experimental characterization of structural and fluidic behaviors, major applications, potential challenges, and future prospects.
Collapse
Affiliation(s)
| | - Feng Zhao
- Micro/Nanoelectronics and Energy Laboratory, School of Engineering and Computer Science, Washington State University, Vancouver, WA 98686, USA;
| |
Collapse
|
49
|
Downs AM, Gerson J, Leung KK, Honeywell KM, Kippin T, Plaxco KW. Improved calibration of electrochemical aptamer-based sensors. Sci Rep 2022; 12:5535. [PMID: 35365672 PMCID: PMC8976050 DOI: 10.1038/s41598-022-09070-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 02/24/2022] [Indexed: 12/22/2022] Open
Abstract
Electrochemical aptamer-based (EAB) sensors support the real-time, high frequency measurement of pharmaceuticals and metabolites in-situ in the living body, rendering them a potentially powerful technology for both research and clinical applications. Here we explore quantification using EAB sensors, examining the impact of media selection and temperature on measurement performance. Using freshly-collected, undiluted whole blood at body temperature as both our calibration and measurement conditions, we demonstrate accuracy of better than ± 10% for the measurement of our test bed drug, vancomycin. Comparing titrations collected at room and body temperature, we find that matching the temperature of calibration curve collection to the temperature used during measurements improves quantification by reducing differences in sensor gain and binding curve midpoint. We likewise find that, because blood age impacts the sensor response, calibrating in freshly collected blood can improve quantification. Finally, we demonstrate the use of non-blood proxy media to achieve calibration without the need to collect fresh whole blood.
Collapse
Affiliation(s)
- Alex M Downs
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, 93106, USA.,Center for Bioengineering, University of California Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Julian Gerson
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Kaylyn K Leung
- Center for Bioengineering, University of California Santa Barbara, Santa Barbara, CA, 93106, USA.,Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Kevin M Honeywell
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Tod Kippin
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, CA, 93106, USA.,The Neuroscience Research Institute and Department of Molecular Cellular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, 93106, USA
| | - Kevin W Plaxco
- Department of Mechanical Engineering, University of California Santa Barbara, Santa Barbara, CA, 93106, USA. .,Center for Bioengineering, University of California Santa Barbara, Santa Barbara, CA, 93106, USA. .,Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA, 93106, USA.
| |
Collapse
|
50
|
Xiao Z, Huang Q, Yang Y, Liu M, Chen Q, Huang J, Xiang Y, Long X, Zhao T, Wang X, Zhu X, Tu S, Ai K. Emerging early diagnostic methods for acute kidney injury. Theranostics 2022; 12:2963-2986. [PMID: 35401836 PMCID: PMC8965497 DOI: 10.7150/thno.71064] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Many factors such as trauma and COVID-19 cause acute kidney injury (AKI). Late AKI have a very high incidence and mortality rate. Early diagnosis of AKI provides a critical therapeutic time window for AKI treatment to prevent progression to chronic renal failure. However, the current clinical detection based on creatinine and urine output isn't effective in diagnosing early AKI. In recent years, the early diagnosis of AKI has made great progress with the advancement of information technology, nanotechnology, and biomedicine. These emerging methods are mainly divided into two aspects: First, predicting AKI through models construct by machine learning; Second, early diagnosis of AKI through detection of newly-discovered early biomarkers. Currently, these methods have shown great potential and become an attractive tool for the early diagnosis of AKI. Therefore, it is very important to discuss and summarize these methods for the early diagnosis of AKI. In this review, we first systematically summarize the application of machine learning in AKI prediction algorithms and specific scenarios. In addition, we introduce the key role of early biomarkers in the progress of AKI, and then comprehensively summarize the application of emerging detection technologies for early AKI. Finally, we discuss current challenges and prospects of machine learning and biomarker detection. The review is expected to provide new insights for early diagnosis of AKI, and provided important inspiration for the design of early diagnosis of other major diseases.
Collapse
Affiliation(s)
- Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China, 410078
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China, 410008
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China, 410008
| | - Yuqi Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China, 410008
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China, 410008
| | - Min Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China, 410008
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, P.R. China, 410008
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China, 410078
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Jia Huang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China, 410078
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yuting Xiang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China, 410078
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xingyu Long
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China, 410078
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Tianjiao Zhao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China, 410078
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiaoyuan Wang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China, 410078
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Xiaoyu Zhu
- Hunan Key Laboratory of Oral Health Research, Hunan 3D Printing Engineering Research Center of Oral Care, Hunan Clinical Research Center of Oral Major Diseases and Oral Health, Xiangya Stomatological Hospital, and Xiangya School of Stomatology, Central South University, Hunan, 410008, Changsha, China
| | - Shiqi Tu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China, 410078
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, P.R. China, 410078
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| |
Collapse
|