1
|
Li X, Hu L, Xu F, Yu W, Wu Y, Deng J, Wei Z, Shi G, Zhang M. Ultrasensitive and selective vancomycin detection using aptamer-modified multi-doped laser-induced graphene extended-gate field-effect transistor. Talanta 2025; 295:128312. [PMID: 40373583 DOI: 10.1016/j.talanta.2025.128312] [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: 03/19/2025] [Revised: 05/04/2025] [Accepted: 05/11/2025] [Indexed: 05/17/2025]
Abstract
Vancomycin (Van), a widely utilized glycopeptide antibiotic in clinical settings, necessitates precise bloodstream concentration monitoring due to its narrow therapeutic window, ensuring drug efficacy while preventing adverse effects. In this work, we have engineered an extended-gate field-effect transistor (EG-FET) sensor tailored for vancomycin detection. This novel sensor configuration comprises a detachable multi-doped graphene EG electrode sensing element paired with a commercial field-effect transistor (FET). The EG electrode design integrates a sophisticated multistage doping process, incorporating MnO2 and Au nanoparticles into laser-induced graphene (LIG), thereby augmenting both functional and electrical characteristics of LIG. To achieve specific recognition, a vancomycin aptamer is immobilized onto the electrode surface, enabling selective binding with vancomycin and translating this interaction into a measurable electrical signal. This collaborative mechanism empowers the EG-FET sensor to exhibit exceptional sensitivity and selectivity towards vancomycin. Notably, the sensor demonstrates a wide linear response ranging from 1 nM to 100 μM, spanning the entire therapeutic window of vancomycin (6-35 μM), boasting an impressive detection limit of 0.187 nM. We have innovated a portable wireless sensing system, coupled with a Janus membrane for expedited plasma separation, consolidating a portable platform dedicated to vancomycin sensing. Furthermore, we have realized the detection of vancomycin concentration in patient's blood using this sensor, and the results are reliable. This comprehensive study underscores the immense potential of multi-doped graphene EG-FET sensors in the realm of antibiotic detection, thereby contributing a pivotal tool towards the realization of precision medicine strategies.
Collapse
Affiliation(s)
- Xinjie Li
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Dongchuan Road 500, Shanghai, 200241, China
| | - Linping Hu
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Dongchuan Road 500, Shanghai, 200241, China
| | - Feng Xu
- Department of Pharmacy, 6th People's Hospital South Campus, Shanghai Jiao Tong University, Shanghai, 201499, China.
| | - Wenbang Yu
- Jinhua University of Vocational Technology, Jinhua, 321017, China.
| | - Yixuan Wu
- Xi'an Jiaotong-liverpool University, Soochow, 215123, China
| | - Junhongyu Deng
- Xi'an Jiaotong-liverpool University, Soochow, 215123, China
| | - Zihan Wei
- Shanghai Rongxiang Biotechnology Co., Ltd, China
| | - Guoyue Shi
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Dongchuan Road 500, Shanghai, 200241, China
| | - Min Zhang
- School of Chemistry and Molecular Engineering, Shanghai Key Laboratory for Urban Ecological Processes and Eco-Restoration, East China Normal University, Dongchuan Road 500, Shanghai, 200241, China.
| |
Collapse
|
2
|
Emmons N, Duman Z, Erdal MK, Hespanha J, Kippin TE, Plaxco KW. Feedback Control over Plasma Drug Concentrations Achieves Rapid and Accurate Control over Solid-Tissue Drug Concentrations. ACS Pharmacol Transl Sci 2025; 8:1416-1423. [PMID: 40370982 PMCID: PMC12070313 DOI: 10.1021/acsptsci.5c00142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 03/31/2025] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Electrochemical aptamer-based (EAB) sensors enable the continuous, real-time monitoring of drugs and biomarkers in situ in the blood, brain, and peripheral tissues of live subjects. The real-time concentration information produced by these sensors provides unique opportunities to perform closed-loop, feedback-controlled drug delivery, by which the plasma concentration of a drug can be held constant or made to follow a specific, time-varying profile. Motivated by the observation that the site of action of many drugs is the solid tissues and not the blood, here we experimentally confirm that maintaining constant plasma drug concentrations also produces constant concentrations in the interstitial fluid (ISF). Using an intravenous EAB sensor we performed feedback control over the concentration of doxorubicin, an anthracycline chemotherapeutic, in the plasma of live rats. Using a second sensor placed in the subcutaneous space, we find drug concentrations in the ISF rapidly (30-60 min) match and then accurately (RMS deviation of 8 to 21%) remain at the feedback-controlled plasma concentration, validating the use of feedback-controlled plasma drug concentrations to control drug concentrations in the solid tissues that are the site of drug action. We expanded to pairs of sensors in the ISF, the outputs of the individual sensors track one another with good precision (R 2 = 0.95-0.99), confirming that the performance of in vivo EAB sensors matches that of prior, in vitro validation studies. These observations suggest EAB sensors could prove a powerful new approach to the high-precision personalization of drug dosing.
Collapse
Affiliation(s)
- Nicole
A. Emmons
- Department
of Psychological and Brain Sciences, University
of California, Santa Barbara 93106, United States
- Neuroscience
Research Institute, University of California, Santa Barbara 93106, United States
- Institute
for Collaborate Biotechnologies, University
of California, Santa Barbara 93106, United States
| | - Zeki Duman
- Department
of Electrical and Computer Engineering, University of California, Santa
Barbara 93106, United States
| | - Murat Kaan Erdal
- Department
of Electrical and Computer Engineering, University of California, Santa
Barbara 93106, United States
| | - João Hespanha
- Department
of Electrical and Computer Engineering, University of California, Santa
Barbara 93106, United States
| | - Tod E. Kippin
- Department
of Psychological and Brain Sciences, University
of California, Santa Barbara 93106, United States
- Neuroscience
Research Institute, University of California, Santa Barbara 93106, United States
- Institute
for Collaborate Biotechnologies, University
of California, Santa Barbara 93106, United States
| | - Kevin W. Plaxco
- Department
of Chemistry and Biochemistry, University
of California, Santa Barbara 93106, United States
- Department
of Bioengineering, University of California, Santa Barbara 93106, United States
- Institute
for Collaborate Biotechnologies, University
of California, Santa Barbara 93106, United States
| |
Collapse
|
3
|
Chen S, Fan S, Qiao Z, Wu Z, Lin B, Li Z, Riegler MA, Wong MYH, Opheim A, Korostynska O, Nielsen KM, Glott T, Martinsen ACT, Telle-Hansen VH, Lim CT. Transforming Healthcare: Intelligent Wearable Sensors Empowered by Smart Materials and Artificial Intelligence. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2500412. [PMID: 40167502 DOI: 10.1002/adma.202500412] [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: 01/07/2025] [Revised: 03/14/2025] [Indexed: 04/02/2025]
Abstract
Intelligent wearable sensors, empowered by machine learning and innovative smart materials, enable rapid, accurate disease diagnosis, personalized therapy, and continuous health monitoring without disrupting daily life. This integration facilitates a shift from traditional, hospital-centered healthcare to a more decentralized, patient-centric model, where wearable sensors can collect real-time physiological data, provide deep analysis of these data streams, and generate actionable insights for point-of-care precise diagnostics and personalized therapy. Despite rapid advancements in smart materials, machine learning, and wearable sensing technologies, there is a lack of comprehensive reviews that systematically examine the intersection of these fields. This review addresses this gap, providing a critical analysis of wearable sensing technologies empowered by smart advanced materials and artificial Intelligence. The state-of-the-art smart materials-including self-healing, metamaterials, and responsive materials-that enhance sensor functionality are first examined. Advanced machine learning methodologies integrated into wearable devices are discussed, and their role in biomedical applications is highlighted. The combined impact of wearable sensors, empowered by smart materials and machine learning, and their applications in intelligent diagnostics and therapeutics are also examined. Finally, existing challenges, including technical and compliance issues, information security concerns, and regulatory considerations are addressed, and future directions for advancing intelligent healthcare are proposed.
Collapse
Affiliation(s)
- Shuwen Chen
- Institute of Medical Equipment Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Shicheng Fan
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Zheng Qiao
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Zixiong Wu
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Baobao Lin
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Zhijie Li
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Michael A Riegler
- Simula Metropolitan Center for Digital Engineering, Oslo, 0167, Norway
| | | | - Arve Opheim
- Sunnaas Rehabilitation Hospital, Bjoernemyr, 1453, Norway
- Institute of Neuroscience and Physiology, Unit for Rehabilitation Medicine, University of Gothenburg, Gothenburg, 413 45, Sweden
| | - Olga Korostynska
- Department of Mechanical, Electronic and Chemical Engineering (MEK), Faculty of Technology, Art, and Design, TKD, Oslo Metropolitan University, OsloMet, Oslo, 0166, Norway
| | - Kaare Magne Nielsen
- Department of Life Science and Health, Faculty of Health Sciences, Oslo Metropolitan University, OsloMet, Oslo, 0130, Norway
- Intelligent Health, Faculty of Health Sciences and Faculty of Technology, Art and Design, Oslo Metropolitan University, OsloMet, Oslo, 0130, Norway
| | - Thomas Glott
- Sunnaas Rehabilitation Hospital, Bjoernemyr, 1453, Norway
| | - Anne Catrine T Martinsen
- Sunnaas Rehabilitation Hospital, Bjoernemyr, 1453, Norway
- Department of Rehabilitation Science and Health Technology, Faculty of Health Sciences, Oslo Metropolitan University, OsloMet, Oslo, 0130, Norway
| | - Vibeke H Telle-Hansen
- Intelligent Health, Faculty of Health Sciences and Faculty of Technology, Art and Design, Oslo Metropolitan University, OsloMet, Oslo, 0130, Norway
- Department of Nursing and Health Promotion, Faculty of Health Sciences, Oslo Metropolitan University, OsloMet, Oslo, 0130, Norway
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore, 119276, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore, 117411, Singapore
- Institute for Functional Intelligent Materials, National University of Singapore, Singapore, 117544, Singapore
- SIA-NUS Digital Aviation Corp Lab, National University of Singapore, Singapore, 117602, Singapore
- Institute for Digital Molecular Analytics and Science, Nanyang Technological University, Singapore, 636921, Singapore
| |
Collapse
|
4
|
Mayol B, Qubbaj IZ, Nava-Granados J, Vasquez K, Keene ST, Sempionatto JR. Aptamer and Oligonucleotide-Based Biosensors for Health Applications. BIOSENSORS 2025; 15:277. [PMID: 40422016 DOI: 10.3390/bios15050277] [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: 04/01/2025] [Revised: 04/22/2025] [Accepted: 04/23/2025] [Indexed: 05/28/2025]
Abstract
Aptamers have emerged as powerful molecular recognition elements for biosensing applications, offering high specificity, stability, and adaptability. This review explores key considerations in designing aptamer-based sensors (aptasensors), with a focus on biomarker selection, aptamer design, and detection and immobilization strategies. However, challenges such as biofluid stability and reversibility must be addressed to improve biosensor performance. In this study, the potential of aptamer-based platforms in diagnostics is explored, emphasizing their advantages and future applications. Looking ahead, advances in multifunctional aptamers, integration with nanomaterials, and computational optimization are highlighted as promising directions for enhancing their effectiveness in biosensing.
Collapse
Affiliation(s)
- Beatriz Mayol
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| | - I Zeina Qubbaj
- Department of Materials Science and NanoEngineering, Rice University, Houston, TX 77005, USA
| | - Julieta Nava-Granados
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| | - Katherine Vasquez
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| | - Scott T Keene
- Department of Materials Science and NanoEngineering, Rice University, Houston, TX 77005, USA
| | - Juliane R Sempionatto
- Department of Electrical and Computer Engineering, Rice University, Houston, TX 77005, USA
| |
Collapse
|
5
|
Huang J, Gu S, Zhou X, Liu Y, Zhang Z. Aptamer single-molecule dispersion on single-atom anchoring sites for high-selectivity in vivo detection. Chem Sci 2025:d5sc01852h. [PMID: 40321190 PMCID: PMC12044545 DOI: 10.1039/d5sc01852h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Accepted: 04/20/2025] [Indexed: 05/08/2025] Open
Abstract
Traditional aptasensors struggle to distinguish molecules with highly similar chemical structures due to the inherent flexibility of aptamers, which form 'nano-bushes' causing non-specific adsorption and reducing sensor specificity. To address this, we propose a novel strategy of anchoring aptamers at the single-molecule level onto atomic anchoring sites. We have designed a gold single-atom/titanium dioxide (Au SA/TiO2) photoelectrode to immobilize a dopamine (DA)-selective aptamer, enabling the fabrication of a photoelectrochemical single-molecule aptamer sensor (PEC-sm-aptasensor). This sensor can selectively detect DA in vivo in different brain regions of living mice. This advancement has revolutionized our understanding of DA variation in the prefrontal cortex of Parkinson's disease (PD) mice. In contrast to previous beliefs, we have discovered a new neurotransmitter dynamic pattern: while the total concentration of neurotransmitters decreases, the concentration of DA remains constant, thus not affecting cognitive levels. This finding is crucial for a more targeted understanding of PD and opens avenues for more effective treatments and diagnostic methods.
Collapse
Affiliation(s)
- Jing Huang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University Dongchuan Road 500 Shanghai 200241 China
| | - Shiting Gu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University Dongchuan Road 500 Shanghai 200241 China
| | - Xue Zhou
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University Dongchuan Road 500 Shanghai 200241 China
| | - Yibin Liu
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University Dongchuan Road 500 Shanghai 200241 China
| | - Zhonghai Zhang
- Shanghai Key Laboratory of Green Chemistry and Chemical Processes, School of Chemistry and Molecular Engineering, East China Normal University Dongchuan Road 500 Shanghai 200241 China
| |
Collapse
|
6
|
Abouali H, Keyvani F, Hosseini SA, Srikant S, Poudineh M. Continuous High-Throughput Plasma Separation for Blood Biomarker Sensing Using a Hydrodynamic Microfluidic Device. Adv Healthc Mater 2025; 14:e2404193. [PMID: 39972640 PMCID: PMC11973946 DOI: 10.1002/adhm.202404193] [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: 10/25/2024] [Revised: 02/05/2025] [Indexed: 02/21/2025]
Abstract
Continuous, cost-effective, high-throughput with admissible yield and purity of blood plasma separation is widely needed for biomarker detection in the clinic. The existing gold standard technique (centrifugation) and microfluidic technologies fall short of meeting these criteria. In this study, a microfluidic device design is demonstrated based on passive hydrodynamic principles to achieve admissible yield and purity plasma samples. Through computational and experimental assessments, it is shown that side channels with varying lengths are required to improve the plasma extraction rate. The optimized side channels in this device design use the formed cell-free layer regions in the expanded areas to extract plasma consistently and efficiently. These Hydrodynamic Continuous, High-Throughput Plasma Separator (HCHPS) microfluidic devices achieve a purity in the range of 47% to 64% with whole blood and maintaining a yield of 10% to 18%, with half hemolysis compared to gold standard centrifugation. These devices also separate the plasma from diluted blood with a purity in the range of 62% to 97% with a similar yield range. Additionally, whole human blood spiked with lactate was processed through the HCHPS device, and the separated plasma is collected and analyzed using two biosensing approaches, a bead-based fluorescence, and an electrochemical aptamer biosensing, confirming the quality of plasma for downstream biomarker detection.
Collapse
Affiliation(s)
- Hesam Abouali
- Department of Electrical and Computer EngineeringUniversity of WaterlooWaterlooONN2L 3G1Canada
| | - Fatemeh Keyvani
- Department of Electrical and Computer EngineeringUniversity of WaterlooWaterlooONN2L 3G1Canada
| | - Seied Ali Hosseini
- Department of Electrical and Computer EngineeringUniversity of WaterlooWaterlooONN2L 3G1Canada
| | - Sanjana Srikant
- Department of Electrical and Computer EngineeringUniversity of WaterlooWaterlooONN2L 3G1Canada
| | - Mahla Poudineh
- Department of Electrical and Computer EngineeringUniversity of WaterlooWaterlooONN2L 3G1Canada
| |
Collapse
|
7
|
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
|
8
|
Saunders J, Thompson IAP, Soh HT. Generalizable Molecular Switch Designs for In Vivo Continuous Biosensing. Acc Chem Res 2025; 58:703-713. [PMID: 39954262 PMCID: PMC11883736 DOI: 10.1021/acs.accounts.4c00721] [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: 11/05/2024] [Revised: 02/03/2025] [Accepted: 02/05/2025] [Indexed: 02/17/2025]
Abstract
Continuous biosensors have the potential to transform medicine, enabling healthcare to be more preventative and personalized as compared to the current standard of reactive diagnostics. Realizing this transformative potential requires biosensors that can function continuously in vivo without sample preparation and deliver molecular specificity, sensitivity, and high temporal resolution. Molecular switches stand out as a promising solution for creating such sensors for the continuous detection of many different types of molecules. Molecular switches are target-binding receptors designed such that binding causes a conformational change in the switch's structure. This structure switching induces a measurable signal change via reporters incorporated into the molecular switch, enabling highly specific, label-free sensing. However, there remains an outstanding need for generalizable switch designs that can be adapted for the detection of a wide range of molecular targets. In this Account, we chronicle the work our lab has done to develop generalizable molecular switch designs that allow more rapid development of high-performance biosensors across a broad range of biomarkers. Pioneering efforts toward molecular switch-based biosensing have employed aptamers─nucleic acid-based receptors with sequence-specific target affinity. However, most of these early demonstrations relied upon aptamers with intrinsic structure-switching capabilities. To accelerate aptamer switch design for more targets, we have applied rational design and knowledge of an aptamer's structure to engineer switching functionality into pre-existing aptamers. Our designs contained several structural parameters that enabled us to easily tune the sensitivity and binding kinetics of the resulting switches. Using such rationally designed aptamer switches, we demonstrated continuous optical detection of cortisol and dopamine at physiologically relevant concentrations in complex media. In an effort to move beyond aptamers with well-characterized structural properties, we developed a high-throughput screening method that allowed us to simultaneously screen millions of candidates derived from a single aptamer to find sensitive switches without any prior structural knowledge of the parent aptamer. In subsequent work, we reasoned that we could enhance our ability to design a broader range of biosensors by leveraging other classes of receptors besides aptamers. Antibodies offer excellent affinity and specificity for a wide range of targets, but lack the capacity for intrinsic structure switching. We therefore developed a set of strategies to augment antibodies with the capacity to act as molecular switches with a diverse range of target-binding properties. We combined both the high binding affinity of an antibody with the structure-switching capabilities of an aptamer to develop a chimeric switch with 100-fold enhanced sensitivity for a protein target and improved function in interferent-rich samples. In a second design, we developed a competitive immunoassay-inspired scheme to engineer switching behavior into an antibody for minutes-scale temporal resolution with nanomolar sensitivity. We used this competitive antibody-switch to demonstrate the first continuous detection of cortisol directly in whole blood. Together, these advances in molecular switch development have expanded our capability to rapidly engineer new continuous biosensors, thereby increasing opportunities to track health via a wide range of biomarkers to deliver more personalized and preventative medicine.
Collapse
Affiliation(s)
- Jason Saunders
- Department
of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - Ian A. P. Thompson
- Department
of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - Hyongsok Tom Soh
- Department
of Electrical Engineering, Stanford University, Stanford, California 94305, United States
- Department
of Radiology, Stanford University, Stanford, California 94305, United States
- Department
of Bioengineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
9
|
Scida K, Ornelas-Gatdula E, DePasquale M, Carr GV, Arroyo-Currás N. Therapeutic Drug Distribution across the Mouse Brain Is Heterogeneous as Revealed by In Vivo, Spatially Resolved Aptamer-Based Sensing. ACS Pharmacol Transl Sci 2025; 8:435-445. [PMID: 39990862 PMCID: PMC11843511 DOI: 10.1021/acsptsci.4c00579] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/28/2024] [Accepted: 12/26/2024] [Indexed: 02/25/2025]
Abstract
Drug discovery for central nervous system (CNS) targets is a high stakes process with estimated success rates below ten percent. Dose scaling, penetration through the blood-brain-barrier (BBB), and potency are among the various challenges involved in developing drugs for CNS targets. The standard approach to evaluate some of these parameters is dosing lead therapeutic compounds via intravenous delivery and assessing their brain levels via tissue homogenization and ex vivo quantification. Although a cost and time effective approach, brain homogenization lacks pharmacokinetic spatial resolution and normalizes drug levels to the entire brain volume. The brain, however, is known to have regional differences in cellular composition, transporters, BBB permeability, and drug-metabolizing enzymes, factors that could significantly affect pharmacological assessments during drug discovery. In this study we employ electrochemical aptamer-based sensors, a technology that allows in situ, real-time molecular monitoring in live animals, to reveal significant differences in the pharmacokinetics of drug uptake and accumulation in the brain of mice. Using vancomycin in the context of penetrating brain injury (PBI), our results highlight that potency may be significantly affected by PBI location. Additionally, more accurate dose scaling and delivery for deep brain wounds could be achieved by adjusting route of administration based on real-time-measured pharmacokinetic profiles, for example by changing delivery from intravenous to intracerebroventricular dosing. We emphasize the issue of establishing accurate pharmacological parameters during preclinical drug discovery efforts and underline the value of aptamer-based sensors for precise estimations of drug pharmacokinetics, transport across the BBB, and effective dose delivery during preclinical trials.
Collapse
Affiliation(s)
- Karen Scida
- Lieber
Institute for Brain Development, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21205, United States
| | - Elysse Ornelas-Gatdula
- 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
| | - Michael DePasquale
- Lieber
Institute for Brain Development, Johns Hopkins
University School of Medicine, Baltimore, Maryland 21205, United States
| | - Gregory V. Carr
- Lieber
Institute for Brain Development, 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
| | - 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
| |
Collapse
|
10
|
Bakestani RM, Wu Y, Glahn-Martínez B, Kippin TE, Plaxco KW, Kolkman RW. Carboxylate-Terminated Electrode Surfaces Improve the Performance of Electrochemical Aptamer-Based Sensors. ACS APPLIED MATERIALS & INTERFACES 2025; 17:8706-8714. [PMID: 39841926 PMCID: PMC11803614 DOI: 10.1021/acsami.4c21790] [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: 12/11/2024] [Revised: 01/13/2025] [Accepted: 01/14/2025] [Indexed: 01/24/2025]
Abstract
Electrochemical aptamer-based (EAB) sensors are a molecular measurement platform that enables the continuous, real-time measurement of a wide range of drugs and biomarkers in situ in the living body. EAB sensors are fabricated by depositing a thiol-modified, target-binding aptamer on the surface of a gold electrode, followed by backfilling with an alkanethiol to form a self-assembled monolayer. And while the majority of previously described EAB sensors have employed hydroxyl-terminated monolayers, a handful of studies have shown that altering the monolayer headgroup can strongly affect sensor performance. Here, using 4 different EAB sensors, we show that the mixed monolayers composed of mixtures of 6-carbon hydroxyl-terminated thiols and varying amounts of either 6- or 8-carbon, carboxylate-terminated thiols lead to improved EAB sensor performance. Specifically, the use of such mixed monolayers enhances the signal gain (the relative change in the signal seen upon target addition) for all tested sensors, often by several fold, both in buffer and whole blood at room temperature or physiological temperatures. Moreover, these improvements in gain are achieved without significant changes in the aptamer affinity or the stability of the resulting sensors. In addition to proving a ready means of improving EAB sensor performance, these results suggest that exploration of the chemistry of the electrode surface employed in such sensors could prove to be a fruitful means of advancing this unique in vivo sensing technology.
Collapse
Affiliation(s)
- Rose Mery Bakestani
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Yuyang Wu
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Bettina Glahn-Martínez
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa Barbara, California 93106, United States
- Department
of Analytical Chemistry, Faculty of Chemistry, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Tod E. Kippin
- Department
of Psychological and Brain Sciences, University
of California Santa Barbara, 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
- Biological
Engineering Graduate Program, University
of California Santa Barbara, Santa
Barbara, California 93106, United States
| | - Ruben W. Kolkman
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
11
|
Tan JH, Fraser AG. Quantifying metabolites using structure-switching aptamers coupled to DNA sequencing. Nat Biotechnol 2025:10.1038/s41587-025-02554-7. [PMID: 39905266 DOI: 10.1038/s41587-025-02554-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 01/06/2025] [Indexed: 02/06/2025]
Abstract
Here we report a method, smol-seq (small-molecule sequencing), using structure-switching aptamers (SSAs) and DNA sequencing to quantify metabolites. In smol-seq, each SSA detects a single target molecule and releases a unique DNA barcode on target binding. Sequencing the released barcodes can, thus, read out metabolite levels. We show that SSAs are highly specific and can be multiplexed to detect multiple targets in parallel, bringing the power of DNA sequencing to metabolomics.
Collapse
Affiliation(s)
- June H Tan
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada
| | - Andrew G Fraser
- The Donnelly Centre, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
12
|
Liu ZJ, Liang YQ, Li JY, Wu B, Huang C, Liu YW, Zhang CZ, Yang Y, Cai NQ, Chen JY, Lin XH. Engineered Aptamer-Derived Fluorescent Aptasensor: the Label-Free, Single-Step, Rapid Detection of Vancomycin in Clinical Samples. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2407799. [PMID: 39676392 DOI: 10.1002/smll.202407799] [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: 09/01/2024] [Revised: 11/27/2024] [Indexed: 12/17/2024]
Abstract
Currently, the reported vancomycin (VCM) aptamers, including the 3- (Kd = 9.13 × 10-6 m) and 4-truncated variants (Kd = 45.5 × 10-6 m), are engineered via stem truncation of the VCM parent aptamer, which inevitably compromises their affinities, thus affecting their clinical application within the VCM therapeutic window of 6.9-13.8 × 10-6 m. Herein, the binding pocket of the VCM parent aptamer is elucidated for the first time and we implemented the Post-SELEX modification strategy involving truncation and mutagenesis to refined the VCM parent aptamer. This yielded a VCM aptamer (ABC20-11) with an intramolecular G-triplex, an enhanced thioflavin T (ThT) fluorescence intensity, and an improved affinity (Kd = 0.591 × 10-6 m) and specificity (one-methyl level) for VCM. Utilizing a portable fluorescence detector specifically designed for rapidly detecting VCM concentration and leveraging the competitive binding between VCM and ThT to ABC20-11, a label-free fluorescent aptasensor is developed. This aptasensor exhibits exceptional analytical performances across various clinical samples (serum, cerebrospinal fluid, and joint fluid), with corresponding linear ranges of 0.5-50, 0.5-40, and 0.5-50 × 10-6 m and detection limits at 0.11, 0.12, and 0.16 × 10-6 m, respectively. Consequently, the proposed VCM aptasensor displays considerable clinical value and potential for use in rapid VCM detection.
Collapse
Affiliation(s)
- Zhou-Jie Liu
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Yu-Qi Liang
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Jia-Yi Li
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Bing Wu
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Chen Huang
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Yi-Wei Liu
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Chen-Zhi Zhang
- Department of Pharmacy, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Ye Yang
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Nai-Qing Cai
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Jin-Yuan Chen
- The First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
| | - Xin-Hua Lin
- Department of Pharmaceutical Analysis, Higher Educational Key Laboratory for Nano Biomedical Technology of Fujian Province, Faculty of Pharmacy, Fujian Medical University, Fuzhou, 350122, China
| |
Collapse
|
13
|
Emmons N, Duman Z, Erdal M, Kippin T, Hespanha J, Plaxco K. Feedback control over plasma drug concentrations achieves rapid and accurate control over solid-tissue drug concentrations. RESEARCH SQUARE 2025:rs.3.rs-5868915. [PMID: 39975897 PMCID: PMC11838736 DOI: 10.21203/rs.3.rs-5868915/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Electrochemical aptamer-based (EAB) sensors enable the continuous, real-time monitoring of drugs and biomarkers in situ in the blood, brain, and peripheral tissues of live subjects. The real-time concentration information produced by these sensors provides unique opportunities to perform closed-loop, feedback-controlled drug delivery, by which the plasma concentration of a drug can be held constant or made to follow a specific, time-varying profile. Motivated by the observation that the site of action of many drugs is the solid tissues and not the blood, here we experimentally confirm that maintaining constant plasma drug concentrations also produces constant concentrations in the interstitial fluid (ISF). Using an intravenous EAB sensor we performed feedback control over the concentration of doxorubicin, an anthracycline chemotherapeutic, in the plasma of live rats. Using a second sensor placed in the subcutaneous space, we find drug concentrations in the ISF rapidly (30-60 min) match and then accurately (RMS deviation of 8-21%) remain at the feedback-controlled plasma concentration, validating the use of feedback-controlled plasma drug concentrations to control drug concentrations in the solid tissues that are the site of drug action. We expanded to pairs of sensors in the ISF, the outputs of the individual sensors track one another with good precision (R 2 = 0.95-0.99), confirming that the performance of in vivo EAB sensors matches that of prior, in vitro validation studies. These observations suggest EAB sensors could prove a powerful new approach to the high-precision personalization of drug dosing.
Collapse
|
14
|
Bibani M, Casian M, Feier B, Bogdan D, Hosu-Stancioiu O, Ktari N, Kalfat R, Cristea C. Electrochemical aptasensor for the selective detection of vancomycin based on nanostructured "in-lab" printed electrodes. Mikrochim Acta 2025; 192:107. [PMID: 39863729 PMCID: PMC11762413 DOI: 10.1007/s00604-025-06952-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025]
Abstract
A label-free, flexible, and disposable aptasensor was designed for the rapid on-site detection of vancomycin (VAN) levels. The electrochemical sensor was based on lab-printed carbon electrodes (C-PE) enriched with cauliflower-shaped gold nanostructures (AuNSs), on which VAN-specific aptamers were immobilized as biorecognition elements and short-chain thiols as blocking agents. The AuNSs, characterized by scanning electron microscopy (SEM) and atomic force microscopy (AFM), enhanced the electrochemical properties of the platform and the aptamer immobilization active sites. The developed disposable aptasensor allowed label-free detection of VAN via electrochemical impedance spectroscopy (EIS) across a wide range of concentrations (50-1000 nM), with a limit of detection (LOD) of 1.721 nM. The aptasensor presented good selectivity against some commonly found interferences in human serum and milk and was successfully applied to the analysis of these samples.
Collapse
Affiliation(s)
- Malek Bibani
- Department of Analytical Chemistry, Faculty of Pharmacy, "Iuliu Hațieganu" University of Medicine and Pharmacy, 4 Pasteur Street, 400349, Cluj-Napoca, Romania
- Laboratoire Matériaux, Traitement Et Analyse, INRAP, BiotechPole Sidi-Thabet, 2020, Ariana, Tunisia
| | - Magdolna Casian
- Department of Analytical Chemistry, Faculty of Pharmacy, "Iuliu Hațieganu" University of Medicine and Pharmacy, 4 Pasteur Street, 400349, Cluj-Napoca, Romania
| | - Bogdan Feier
- Department of Analytical Chemistry, Faculty of Pharmacy, "Iuliu Hațieganu" University of Medicine and Pharmacy, 4 Pasteur Street, 400349, Cluj-Napoca, Romania.
| | - Diana Bogdan
- National Institute for Research and Development of Isotopic and Molecular Technologies, 67-103 Donat St., 400293, Cluj-Napoca, Romania
| | - Oana Hosu-Stancioiu
- Department of Analytical Chemistry, Faculty of Pharmacy, "Iuliu Hațieganu" University of Medicine and Pharmacy, 4 Pasteur Street, 400349, Cluj-Napoca, Romania
| | - Nadia Ktari
- Laboratoire Matériaux, Traitement Et Analyse, INRAP, BiotechPole Sidi-Thabet, 2020, Ariana, Tunisia
| | - Rafik Kalfat
- Laboratoire Matériaux, Traitement Et Analyse, INRAP, BiotechPole Sidi-Thabet, 2020, Ariana, Tunisia
| | - Cecilia Cristea
- Department of Analytical Chemistry, Faculty of Pharmacy, "Iuliu Hațieganu" University of Medicine and Pharmacy, 4 Pasteur Street, 400349, Cluj-Napoca, Romania
| |
Collapse
|
15
|
Behnam V, McManamen AM, Ballard HG, Aldana B, Tamimi M, Milosavić N, Stojanovic MN, Rubin MR, Sia SK. mPatch: A Wearable Hydrogel Microneedle Patch for In Vivo Optical Sensing of Calcium. Angew Chem Int Ed Engl 2025; 64:e202414871. [PMID: 39625999 DOI: 10.1002/anie.202414871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Accepted: 12/02/2024] [Indexed: 12/13/2024]
Abstract
This study presents an in vivo optical hydrogel microneedle platform that measures levels of analytes in interstitial fluid. The platform builds on a previously published technique for molding hydrogel microneedles by developing a composite hydrogel (i.e., PEGDA and polyacrylamide) that is sufficiently stiff to penetrate skin in the hydrated state and whose fluorescence changes dynamically-via a conjugated aptamer-depending on level of analyte. In a demonstration relevant to hypercalcemia, the hydrogel microneedle distinguished varying concentrations of calcium (within a range of 0 to 2 mM, which spans physiologically meaningful variations for hypoparathyroidism) within 10 minutes. In rats, a compact CMOS sensor measuring fluorescence from microneedles distinguished low hypercalcemic (1.7 mM) from high hypercalcemic (2.3 mM) ionized calcium levels as determined from reference blood measurements. Overall, this work demonstrates in vivo feasibility of a concept-which we call mPatch-for an optical hydrogel microneedle to measure small changes in levels of analytes in interstitial fluid, which does not rely on extraction of interstitial fluid out of the dermis.
Collapse
Affiliation(s)
- Vira Behnam
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Anika M McManamen
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Hannah G Ballard
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Bryan Aldana
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| | - Melissa Tamimi
- Institute of Comparative Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Nenad Milosavić
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Milan N Stojanovic
- Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Mishaela R Rubin
- Department of Medicine Endocrinology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Samuel K Sia
- Department of Biomedical Engineering, Columbia University, New York, NY 10027, USA
| |
Collapse
|
16
|
Drachuk I, Ramani N, Harbaugh S, Mirkin CA, Chávez JL. Implantable Fluorogenic DNA Biosensor for Stress Detection. ACS APPLIED MATERIALS & INTERFACES 2025; 17:130-139. [PMID: 39417681 DOI: 10.1021/acsami.4c08940] [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: 10/19/2024]
Abstract
Implantable sensors that can monitor analytes related to cognitive and physiological status have gained significant focus in recent years. We have developed an implantable biosensor to detect dehydroepiandrosterone sulfate (DHEA-S), a biomarker related to stress. The biosensor strategy was based on the principle of forced intercalation (FIT) aptamers designed to detect subtle intramolecular changes during aptamer-target binding events. By incorporating a steroid-specific fluorogenic aptamer into a hydrogel, the sensitivity and biostability of the FIT biosensor fiber were improved, which were essential for designing implantable sensors to monitor biomarker levels in the living body. The polyethylenimine-based hydrogel chosen for this study produced an optically transparent cross-linked network with optimal microstructure, physicochemical, and mechanical properties, making it suitable for optical biosensors. The in vitro studies showed that the biosensor fiber was successfully activated in human serum and skin analogue, providing a linear response to physiological concentrations of the steroid. We believe that this type of implantable platform can be effective in monitoring more complex biomarkers associated with physiological or psychological health.
Collapse
Affiliation(s)
- Irina Drachuk
- 711th Human Performance Wing, Human Effectiveness Directorate, AFRL, 2510 Fifth Street, Wright-Patterson AFB, Ohio 45433, United States
- UES, a BlueHalo Company, 4401 Dayton-Xenia Rd., Dayton, Ohio 45432, United States
| | - Namrata Ramani
- Department of Materials Science and Engineering and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Svetlana Harbaugh
- 711th Human Performance Wing, Human Effectiveness Directorate, AFRL, 2510 Fifth Street, Wright-Patterson AFB, Ohio 45433, United States
| | - Chad A Mirkin
- Department of Chemistry and International Institute for Nanotechnology, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - Jorge L Chávez
- 711th Human Performance Wing, Human Effectiveness Directorate, AFRL, 2510 Fifth Street, Wright-Patterson AFB, Ohio 45433, United States
| |
Collapse
|
17
|
Notley SR, Meade RD, Looney DP, Chapman CL, Potter AW, Fogarty A, Howlader T, Main LC, Friedl KE, Kenny GP. Physiological monitoring for occupational heat stress management: recent advancements and remaining challenges. Appl Physiol Nutr Metab 2025; 50:1-14. [PMID: 40063989 DOI: 10.1139/apnm-2024-0395] [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: 05/14/2025]
Abstract
Occupational heat stress poses a major threat to worker health and safety that is projected to worsen with global warming. To manage these adverse effects, most industries rely on administrative controls (stay times and work-to-rest allocations) that are designed to limit the rise in body core temperature in the "average" individual. However, due to the extensive inter- and intra-individual variation in thermoregulatory function, these administrative controls will result in some individuals having their work rate and productivity unnecessarily restricted (false positives), while others may be subject to rises in heat strain that compromise health (false negatives). Physiological monitoring has long been touted as a more effective approach for individualized protection from excessive heat stress. This has led to extensive interest in the use of wearable technology for heat stress management from both the scientific community and manufacturers of wearable devices, which has accelerated in the past decade. In this review, we evaluate the merits of the recent and emerging approaches to manage occupational heat strain with wearable physiological monitors. Against this background, we then describe the issues that we perceive to be unresolved regarding the use of wearable heat strain monitors and the research efforts needed to address those issues. Particular emphasis is directed to the efficacy of existing physiological indicators of heat strain, how to define upper limits for those indicators and the efforts required to rigorously validate emerging wearable heat strain monitoring devices.
Collapse
Affiliation(s)
- Sean R Notley
- Defence Science and Technology Group, Melbourne, Australia
| | - Robert D Meade
- Department of Epidemiology, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - David P Looney
- United States Army Research Institute of Environmental Medicine (USARIEM), Natick, MA, USA
| | - Christopher L Chapman
- United States Army Research Institute of Environmental Medicine (USARIEM), Natick, MA, USA
- Oak Ridge Institute for Science and Education, Oak Ridge, TN, USA
| | - Adam W Potter
- United States Army Research Institute of Environmental Medicine (USARIEM), Natick, MA, USA
| | - Alison Fogarty
- Defence Science and Technology Group, Melbourne, Australia
| | - Tabassum Howlader
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| | - Luana C Main
- Deakin University, Institute for Physical Activity and Nutrition, School of Exercise & Nutrition Sciences, Geelong, Victoria, Australia
| | - Karl E Friedl
- United States Army Research Institute of Environmental Medicine (USARIEM), Natick, MA, USA
| | - Glen P Kenny
- Human and Environmental Physiology Research Unit, School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
18
|
Fetter LC, McDonough MH, Kippin TE, Plaxco KW. Effects of Physiological-Scale Variation in Cations, pH, and Temperature on the Calibration of Electrochemical Aptamer-Based Sensors. ACS Sens 2024; 9:6675-6684. [PMID: 39570094 PMCID: PMC11855119 DOI: 10.1021/acssensors.4c02274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Electrochemical aptamer-based (EAB) sensors are the first technology supporting high-frequency, real-time, in vivo molecular measurements that is independent of the chemical reactivity of its targets, rendering it easily generalizable. As is true for all biosensors, however, EAB sensor performance is affected by the measurement environment, potentially reducing accuracy when this environment deviates from the conditions under which the sensor was calibrated. Here, we address this question by measuring the extent to which physiological-scale environmental fluctuations reduce the accuracy of a representative set of EAB sensors and explore the means of correcting these effects. To do so, we first calibrated sensors against vancomycin, phenylalanine, and tryptophan under conditions that match the average ionic strength, cation composition, pH, and temperature of healthy human plasma. We then assessed their accuracy in samples for which the ionic composition, pH, and temperature were at the lower and upper ends of their physiological ranges. Doing so, we find that physiologically relevant fluctuations in ionic strength, cation composition, and pH do not significantly harm EAB sensor accuracy. Specifically, all 3 of our test-bed sensors achieve clinically significant mean relative accuracy (i.e., better than 20%) over the clinically or physiologically relevant concentration ranges of their target molecules. In contrast, physiologically plausible variations away from the temperature used for calibration induce more substantial errors. With knowledge of the temperature in hand, however, these errors are easily ameliorated. It thus appears that physiologically induced changes in the sensing environment are likely not a major impediment to clinical application of this in vivo molecular monitoring technology.
Collapse
Affiliation(s)
- Lisa C Fetter
- Interdepartmental Program in Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Matthew H McDonough
- Department of Statistics and Applied Probability, University of California Santa Barbara, Santa Barbara, California 93106, United States
- Department of Bioengineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Tod E Kippin
- Interdepartmental Program in Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
- Department of Bioengineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
- Department of Psychological and Brain Sciences, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Kevin W Plaxco
- Interdepartmental Program in Biomolecular Science and Engineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
- Department of Bioengineering, University of California Santa Barbara, Santa Barbara, California 93106, United States
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
19
|
Santa C, Park S, Gejt A, Clark HA, Hengerer B, Sergelen K. Real-time monitoring of vancomycin using a split-aptamer surface plasmon resonance biosensor. Analyst 2024; 150:131-141. [PMID: 39584594 DOI: 10.1039/d4an01226g] [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: 11/26/2024]
Abstract
Real-time monitoring of therapeutic drugs is crucial for treatment management and pharmacokinetic studies. We present the optimization and affinity tuning of split-aptamer sandwich assay for real-time monitoring of the narrow therapeutic window drug vancomycin, using surface plasmon resonance (SPR). To achieve reversible, label-free sensing of small molecules by SPR, we adapted a vancomycin binding aptamer in a sandwich assay format through the split-aptamer approach. By evaluating multiple split sites within the secondary structure of the original aptamer, we identified position 27 (P27) as optimal for preserving target affinity, ensuring reversibility, and maximizing sensitivity. The assay demonstrated robust performance under physiologically relevant ranges of pH and divalent cations, and the specific ternary complex formation of the aptamer split segments with the analyte was confirmed by circular dichroism spectroscopy. Subsequently, we engineered a series of split-aptamer pairs with increasing complementarity in the stem regions, improving both the affinity and limit of detection up to 10-fold, as compared to the primary P27 pair. The kinetics of the engineered split-aptamer pairs were evaluated, revealing fast association and dissociation rates, confirming improved affinity and detection limits across variants. Most importantly, the reversibility of the assay, essential for real-time monitoring, was maintained in all pairs. Finally, the assay was further validated in complex biological matrices, including the cerebrospinal fluid from dogs and diluted plasma from rats, demonstrating functionality in biological environments and stability exceeding 9 hours. Our study paves the way for applications of split-aptamers in real-time monitoring of small molecules, with potential implications for in vivo therapeutic drug monitoring and pharmacokinetic studies.
Collapse
Affiliation(s)
| | | | - Artur Gejt
- BioMed X Institute, Heidelberg, Germany.
- Faculty of Biotechnology, Mannheim University of Applied Sciences, Germany
| | - Heather A Clark
- School of Biological and Health Systems Engineering, Arizona State University, USA
| | - Bastian Hengerer
- Boehringer Ingelheim Pharma GmbH & Co. KG, CNS Research, Germany
| | | |
Collapse
|
20
|
Nguyen MD, Osborne MT, Prevot GT, Churcher ZR, Johnson PE, Simine L, Dauphin-Ducharme P. Truncations and in silico docking to enhance the analytical response of aptamer-based biosensors. Biosens Bioelectron 2024; 265:116680. [PMID: 39213817 DOI: 10.1016/j.bios.2024.116680] [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] [Accepted: 08/14/2024] [Indexed: 09/04/2024]
Abstract
Aptamers are short oligonucleotides capable of binding specifically to various targets (i.e., small molecules, proteins, and whole cells) which have been introduced in biosensors such as in the electrochemical aptamer-based (E-AB) sensing platform. E-AB sensors are comprised of a redox-reporter-modified aptamer attached to an electrode that undergoes, upon target addition, a binding-induced change in electron transfer rates. To date, E-AB sensors have faced a limitation in the translatability of aptamers into the sensing platform presumably because sequences obtained from Systematic Evolution of Ligands by Exponential Enrichment (SELEX) are typically long (>80 nucleotides) and that obtaining structural information remains time and resource consuming. In response, we explore the utility of aptamer base truncations and in silico docking to improve their translatability into E-AB sensors. Here, we first apply this to the glucose aptamer, which we characterize in solution using NMR methods to guide design and translate truncated variants in E-AB biosensors. We further investigated the applicability of the truncation and computational approaches to four other aptamer systems (vancomycin, cocaine, methotrexate and theophylline) from which we derived functional E-AB sensors. We foresee that our strategy will increase the success rate of translating aptamers into sensing platforms to afford low-cost measurements of molecules directly in undiluted complex matrices.
Collapse
Affiliation(s)
- Minh-Dat Nguyen
- Département de chimie, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada
| | - Meghan T Osborne
- Department of Chemistry, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Guy Terence Prevot
- Département de chimie, Université de Sherbrooke, Sherbrooke, Québec, J1K 2R1, Canada
| | - Zachary R Churcher
- Department of Chemistry, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Philip E Johnson
- Department of Chemistry, York University, Toronto, Ontario, M3J 1P3, Canada
| | - Lena Simine
- Department of Chemistry, McGill University, Montreal, Quebec, H3A 0B8, Canada
| | | |
Collapse
|
21
|
Grzedowski AJ, Jun D, Mahey A, Zhou GC, Fernandez R, Bizzotto D. Engineering DNA Nanocube SAM Scaffolds for FRET-Based Biosensing: Interfacial Characterization and Sensor Demonstration. J Am Chem Soc 2024; 146:31560-31573. [PMID: 39527762 DOI: 10.1021/jacs.4c09240] [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: 11/16/2024]
Abstract
Decorating a gold surface with molecular-level control over the positioning of DNA probes was demonstrated using a self-assembled monolayer (SAM) of wireframe DNA nanocube structures. The DNA nanocubes were specifically adsorbed and oriented using thiol-modified DNA on one face of the cube. The DNA nanocube SAM had a uniform coverage over the gold single crystal bead electrode with a separation of 20-30 nm measured by AFM. The face of the nanocube furthest from the gold surface was designed to hybridize with two different sequences of a 50 base single-stranded DNA probe that was modified with a fluorophore. The first 20 bases were hybridized with the DNA nanocube. One of a pair of FRET fluorophores was used for each probe strand. The dimensions of the nanocube controlled the relative spacing between these fluorophores. When the DNA probes were single-stranded, a FRET signal was observed. FRET decreased to background levels when a complementary DNA target was hybridized to either probe, resulting in a turn-off sensor with little cross-talk between the individual hybridization events. Hybridization isotherms for one target gave KA = 170 pM and a detection limit <50 pM. In addition, the DNA nanocube SAM was configured to be used as a turn-on NeutrAvidin sensor using biotinylated DNA targets hybridized to each probe resulting in an increase in FRET. We show that the wireframe DNA nanocube can be an effective scaffold for preparing biosensors with controlled separation between surface-bound probes facilitating precise sensor surface design and enabling a wide range of sensing modalities with more than one signal available for correlative confirmation of the target binding.
Collapse
Affiliation(s)
- Adrian Jan Grzedowski
- AMPEL and Department of Chemistry,University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1M1,Canada
| | - Daniel Jun
- Deparment of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 2A1,Canada
| | - Amita Mahey
- Deparment of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 2A1,Canada
| | - Geyang Chris Zhou
- AMPEL and Department of Chemistry,University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1M1,Canada
| | - Rachel Fernandez
- Deparment of Microbiology and Immunology, University of British Columbia, 2350 Health Sciences Mall, Vancouver, BC V6T 2A1,Canada
| | - Dan Bizzotto
- AMPEL and Department of Chemistry,University of British Columbia, 2036 Main Mall, Vancouver, BC V6T 1M1,Canada
| |
Collapse
|
22
|
Silver K, Smith A, Colling HV, Tenorio N, Rowland TJ, Bonham AJ. Electrochemical Aptamer-Based Biosensor for Detecting Pap31, a Biomarker for Carrion's Disease. SENSORS (BASEL, SWITZERLAND) 2024; 24:7295. [PMID: 39599072 PMCID: PMC11598354 DOI: 10.3390/s24227295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/08/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024]
Abstract
Carrion's disease, caused by infection with the bacterium Bartonella bacilliformis (B. bacilliformis), is effectively treated with antibiotics, but reaches fatality rates of ~90% if untreated. Current diagnostic methods are limited, insufficiently sensitive, or require laboratory technology unavailable in endemic areas. Electrochemical aptamer-based (E-AB) biosensors provide a potential solution for this unmet need, as these biosensors are portable, sensitive, and can rapidly report the detection of small molecule targets. Here, we developed an E-AB biosensor to detect Pap31, a biomarker of Carrion's disease and an outer membrane protein in B. bacilliformis. We identified an aptamer with Pap31-specific binding affinity using a magnetic pull-down assay with magnetic bead-bound Pap31 and an aptamer library followed by electrophoretic mobility shift assays. We incorporated the Pap31-binding aptamer into a DNA oligonucleotide that changes conformation upon binding Pap31. The resultant Pap31 E-AB biosensor produced a rapid, significant, and target-specific electrical current readout in the buffer, demonstrating an apparent KD of 0.95 nM with a limit of detection of 0.1 nM, and no significant signal change when challenged with off-target proteins. This proof-of-concept Pap31 biosensor design is a first step toward the development of more rapid, sensitive, and portable diagnostic tools for detecting Carrion's disease.
Collapse
Affiliation(s)
- Keaton Silver
- Department of Chemistry & Biochemistry, Metropolitan State University of Denver, Denver, CO 80204, USA; (K.S.); (H.V.C.); (N.T.)
| | - Andrew Smith
- Department of Biology, University of Kansas Medical Center, Kansas City, KS 66061, USA;
| | - Haley V. Colling
- Department of Chemistry & Biochemistry, Metropolitan State University of Denver, Denver, CO 80204, USA; (K.S.); (H.V.C.); (N.T.)
| | - Nico Tenorio
- Department of Chemistry & Biochemistry, Metropolitan State University of Denver, Denver, CO 80204, USA; (K.S.); (H.V.C.); (N.T.)
| | | | - Andrew J. Bonham
- Department of Chemistry & Biochemistry, Metropolitan State University of Denver, Denver, CO 80204, USA; (K.S.); (H.V.C.); (N.T.)
| |
Collapse
|
23
|
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
|
24
|
Stangherlin S, Ding Y, Liu J. Dissociation Constant (K d) Measurement for Small-Molecule Binding Aptamers: Homogeneous Assay Methods and Critical Evaluations. SMALL METHODS 2024:e2401572. [PMID: 39511863 DOI: 10.1002/smtd.202401572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/27/2024] [Indexed: 11/15/2024]
Abstract
Since 1990, numerous aptamers have been isolated and discovered for use in various analytical, biomedical, and environmental applications. This trend continues to date. A critical step in the characterization of aptamer binding is to measure its binding affinity toward both target and non-target molecules. Dissociation constant (Kd) is the most commonly used value in characterizing aptamer binding. In this article, homogenous assays are reviewed for aptamers that can bind small-molecule targets. The reviewed methods include label-free methods, such as isothermal titration calorimetry, intrinsic fluorescence of target molecules, DNA staining dyes, and nuclease digestion assays, and labeled methods, such as the strand displacement reaction. Some methods are not recommended, such as those based on the aggregation of gold nanoparticles and the desorption of fluorophore-labeled DNA from nanomaterials. The difference between the measured apparent Kd and the true Kd of aptamer binding is stressed. In addition, avoiding the titration regime and paying attention to the time required to reach equilibrium are discussed. Finally, it is important to include mutated non-binding sequences as controls.
Collapse
Affiliation(s)
- Stefen Stangherlin
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Yuzhe Ding
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| | - Juewen Liu
- Department of Chemistry, Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, ON, N2L 3G1, Canada
| |
Collapse
|
25
|
Keyvani F, GhavamiNejad P, Saleh MA, Soltani M, Zhao Y, Sadeghzadeh S, Shakeri A, Chelle P, Zheng H, Rahman FA, Mahshid S, Quadrilatero J, Rao PPN, Edginton A, Poudineh M. Integrated Electrochemical Aptamer Biosensing and Colorimetric pH Monitoring via Hydrogel Microneedle Assays for Assessing Antibiotic Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2309027. [PMID: 39250329 PMCID: PMC11538706 DOI: 10.1002/advs.202309027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 06/27/2024] [Indexed: 09/11/2024]
Abstract
Current methods for therapeutic drug monitoring (TDM) have a long turnaround time as they involve collecting patients' blood samples followed by transferring the samples to medical laboratories where sample processing and analysis are performed. To enable real-time and minimally invasive TDM, a microneedle (MN) biosensor to monitor the levels of two important antibiotics, vancomycin (VAN) and gentamicin (GEN) is developed. The MN biosensor is composed of a hydrogel MN (HMN), and an aptamer-functionalized flexible (Flex) electrode, named HMN-Flex. The HMN extracts dermal interstitial fluid (ISF) and transfers it to the Flex electrode where sensing of the target antibiotics happens. The HMN-Flex performance is validated ex vivo using skin models as well as in vivo in live rat animal models. Data is leveraged from the HMN-Flex system to construct pharmacokinetic profiles for VAN and GEN and compare these profiles with conventional blood-based measurements. Additionally, to track pH and monitor patient's response during antibiotic treatment, an HMN is developed that employs a colorimetric method to detect changes in the pH, named HMN-pH assay, whose performance has been validated both in vitro and in vivo. Further, multiplexed antibiotic and pH detection is achieved by simultaneously employing the HMN-pH and HMN-Flex on live animals.
Collapse
Affiliation(s)
- Fatemeh Keyvani
- Department of Electrical and Computer EngineeringFaculty of EngineeringUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Peyman GhavamiNejad
- Department of Electrical and Computer EngineeringFaculty of EngineeringUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Mahmoud Ayman Saleh
- Department of BioengineeringMcGill University815 Sherbrooke St. WMontrealQuebecH3A 0C3Canada
| | - Mohammad Soltani
- Department of Electrical and Computer EngineeringFaculty of EngineeringUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Yusheng Zhao
- School of PharmacyUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Sadegh Sadeghzadeh
- Department of Electrical and Computer EngineeringFaculty of EngineeringUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Arash Shakeri
- School of PharmacyUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Pierre Chelle
- School of PharmacyUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Hanjia Zheng
- Department of Electrical and Computer EngineeringFaculty of EngineeringUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Fasih A. Rahman
- Department of Kinesiology and Health SciencesUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Sarah Mahshid
- Department of BioengineeringMcGill University815 Sherbrooke St. WMontrealQuebecH3A 0C3Canada
| | - Joe Quadrilatero
- Department of Kinesiology and Health SciencesUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Praveen P. N. Rao
- School of PharmacyUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Andrea Edginton
- School of PharmacyUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| | - Mahla Poudineh
- Department of Electrical and Computer EngineeringFaculty of EngineeringUniversity of WaterlooWaterlooOntarioN2L 3G1Canada
| |
Collapse
|
26
|
Kaiyum YA, Hoi Pui Chao E, Dhar L, Shoara AA, Nguyen MD, Mackereth CD, Dauphin-Ducharme P, Johnson PE. Ligand-Induced Folding in a Dopamine-Binding DNA Aptamer. Chembiochem 2024:e202400493. [PMID: 39370408 DOI: 10.1002/cbic.202400493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 08/30/2024] [Accepted: 10/04/2024] [Indexed: 10/08/2024]
Abstract
Aptamers are often employed as molecular recognition elements in the development of different types of biosensors. Many of these biosensors take advantage of the aptamer having a ligand-induced structure-formation binding mechanism. However, this binding mechanism is poorly understood. Here we use isothermal titration calorimetry, circular dichroism spectroscopy and NMR spectroscopy to study the binding and ligand-induced structural change exhibited by a dopamine-binding DNA aptamer. We analysed a series of aptamers where we shorten the terminal stem that contains the 5' and 3' termini of the aptamer sequence. All aptamers bind dopamine in an enthalpically driven process coupled with an unfavorable entropy. A general trend of the aptamer having a weaker binding affinity is observed as the terminal stem is shortened. For all aptamers studied, numerous signals appear in the imino region of the 1H NMR spectrum indicating that new structure forms with ligand binding. However, it is only when this region of structure formation in the aptamer is brought close to the sensor surface that we obtain a functional electrochemical aptamer-based biosensor.
Collapse
Affiliation(s)
- Yunus A Kaiyum
- Department of Chemistry, York University, 4700 Keele St., Toronto, Ontario, M3 J 1P3, Canada
| | - Emily Hoi Pui Chao
- Department of Chemistry, York University, 4700 Keele St., Toronto, Ontario, M3 J 1P3, Canada
| | - Lakshmi Dhar
- Department of Chemistry, York University, 4700 Keele St., Toronto, Ontario, M3 J 1P3, Canada
| | - Aron A Shoara
- Department of Chemistry, York University, 4700 Keele St., Toronto, Ontario, M3 J 1P3, Canada
| | - Minh-Dat Nguyen
- Département de chimie, Université de Sherbrooke, Sherbrooke, Québec, J1 K 2R1, Canada
| | - Cameron D Mackereth
- University of Bordeaux, Inserm U1212, CNRS UMR 5320, ARNA, Bordeaux, FR-33000, France
| | | | - Philip E Johnson
- Department of Chemistry, York University, 4700 Keele St., Toronto, Ontario, M3 J 1P3, Canada
| |
Collapse
|
27
|
Gerson J, Erdal MK, Dauphin-Ducharme P, Idili A, Hespanha JP, Plaxco KW, Kippin TE. A high-precision view of intercompartmental drug transport via simultaneous, seconds-resolved, in situ measurements in the vein and brain. Br J Pharmacol 2024; 181:3869-3885. [PMID: 38877797 PMCID: PMC11890181 DOI: 10.1111/bph.16471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 04/06/2024] [Accepted: 04/19/2024] [Indexed: 06/16/2024] Open
Abstract
BACKGROUND AND PURPOSE The ability to measure specific molecules at multiple sites within the body simultaneously, and with a time resolution of seconds, could greatly advance our understanding of drug transport and elimination. EXPERIMENTAL APPROACH As a proof-of-principle demonstration, here we describe the use of electrochemical aptamer-based (EAB) sensors to measure transport of the antibiotic vancomycin from the plasma (measured in the jugular vein) to the cerebrospinal fluid (measured in the lateral ventricle) of live rats with temporal resolution of a few seconds. KEY RESULTS In our first efforts, we made measurements solely in the ventricle. Doing so we find that, although the collection of hundreds of concentration values over a single drug lifetime enables high-precision estimates of the parameters describing intracranial transport, due to a mathematical equivalence, the data produce two divergent descriptions of the drug's plasma pharmacokinetics that fit the in-brain observations equally well. The simultaneous collection of intravenous measurements, however, resolves this ambiguity, enabling high-precision (typically of ±5 to ±20% at 95% confidence levels) estimates of the key pharmacokinetic parameters describing transport from the blood to the cerebrospinal fluid in individual animals. CONCLUSIONS AND IMPLICATIONS The availability of simultaneous, high-density 'in-vein' (plasma) and 'in-brain' (cerebrospinal fluid) measurements provides unique opportunities to explore the assumptions almost universally employed in earlier compartmental models of drug transport, allowing the quantitative assessment of, for example, the pharmacokinetic effects of physiological processes such as the bulk transport of the drug out of the CNS via the dural venous sinuses.
Collapse
Affiliation(s)
- Julian Gerson
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, California, USA
- Neuroscience Research Institute, University of California, Santa Barbara, Santa Barbara, California, USA
| | - Murat Kaan Erdal
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, Santa Barbara, California, USA
| | - Philippe Dauphin-Ducharme
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, California, USA
| | - Andrea Idili
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, California, USA
- Department of Chemical Science and Technologies, University of Rome Tor Vergata, Rome, Italy
| | - Joao P. Hespanha
- Department of Electrical and Computer Engineering, University of California, Santa Barbara, Santa Barbara, California, USA
| | - Kevin W. Plaxco
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, California, USA
- Center for Bioengineering, 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
| | - Tod E. Kippin
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, California, USA
- Neuroscience Research Institute, 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
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, Santa Barbara, California, USA
| |
Collapse
|
28
|
Lubken R, Lin YT, Haenen SRR, Bergkamp MH, Yan J, Nommensen PA, Prins MWJ. Continuous Biosensor Based on Particle Motion: How Does the Concentration Measurement Precision Depend on Time Scale? ACS Sens 2024; 9:4924-4933. [PMID: 39166946 PMCID: PMC11443519 DOI: 10.1021/acssensors.4c01586] [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: 06/27/2024] [Revised: 07/26/2024] [Accepted: 07/30/2024] [Indexed: 08/23/2024]
Abstract
Continuous biosensors measure concentration-time profiles of biomolecular substances in order to allow for comparisons of measurement data over long periods of time. To make meaningful comparisons of time-dependent data, it is essential to understand how measurement imprecision depends on the time interval between two evaluation points, as the applicable imprecision determines the significance of measured concentration differences. Here, we define a set of measurement imprecisions that relate to different sources of variation and different time scales, ranging from minutes to weeks, and study these using statistical analyses of measurement data. The methodology is exemplified for Biosensing by Particle Motion (BPM), a continuous, affinity-based sensing technology with single-particle and single-molecule resolution. The studied BPM sensor measures specific small molecules (glycoalkaloids) in an industrial food matrix (potato fruit juice). Measurements were performed over several months at two different locations, on nearly 50 sensor cartridges with in total more than 1000 fluid injections. Statistical analyses of the measured signals and concentrations show that the relative residuals are normally distributed, allowing extraction and comparisons of the proposed imprecision parameters. The results indicate that sensor noise is the most important source of variation followed by sample pretreatment. Variations caused by fluidic transport, changes of the sensor during use (drift), and variations due to different sensor cartridges and cartridge replacements appear to be small. The imprecision due to sensor noise is recorded over few-minute time scales and is attributed to stochastic fluctuations of the single-molecule measurement principle, false-positive signals in the signal processing, and nonspecific interactions. The developed methodology elucidates both time-dependent and time-independent factors in the measurement imprecision, providing essential knowledge for interpreting concentration-time profiles as well as for further development of continuous biosensing technologies.
Collapse
Affiliation(s)
| | - Yu-Ting Lin
- Helia
Biomonitoring, Eindhoven 5612 AR, The Netherlands
| | | | | | - Junhong Yan
- Helia
Biomonitoring, Eindhoven 5612 AR, The Netherlands
| | | | - Menno W. J. Prins
- Helia
Biomonitoring, Eindhoven 5612 AR, The Netherlands
- Department
of Biomedical Engineering, Eindhoven University
of Technology, Eindhoven 5612 AZ, The Netherlands
- Department
of Applied Physics, Eindhoven University
of Technology, Eindhoven 5612 AZ, The Netherlands
- Institute
for Complex Molecular Systems (ICMS), Eindhoven
University of Technology, Eindhoven 5612 AZ, The Netherlands
| |
Collapse
|
29
|
Yang Y, Gao X, Widdicombe B, Zhang X, Zielinski JL, Cheng T, Gunatilaka A, Leung KK, Plaxco KW, Rajasekharan Unnithan R, Stewart AG. Dual-Purpose Aptamer-Based Sensors for Real-Time, Multiplexable Monitoring of Metabolites in Cell Culture Media. ACS NANO 2024; 18. [PMID: 39255458 PMCID: PMC11441400 DOI: 10.1021/acsnano.4c06813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/12/2024]
Abstract
The availability of high-frequency, real-time measurements of the concentrations of specific metabolites in cell culture systems will enable a deeper understanding of cellular metabolism and facilitate the application of good laboratory practice standards in cell culture protocols. However, currently available approaches to this end either are constrained to single-time-point and single-parameter measurements or are limited in the range of detectable analytes. Electrochemical aptamer-based (EAB) biosensors have demonstrated utility in real-time monitoring of analytes in vivo in blood and tissues. Here, we characterize a pH-sensing capability of EAB sensors that is independent of the specific target analyte of the aptamer sequence. We applied this dual-purpose EAB to the continuous measurement of pH and phenylalanine in several in vitro cell culture settings. The miniature EAB sensor that we developed exhibits rapid response times, good stability, high repeatability, and biologically relevant sensitivity. We also developed and characterized a leak-free reference electrode that mitigates the potential cytotoxic effects of silver ions released from conventional reference electrodes. Using the resulting dual-purpose sensor, we performed hourly measurements of pH and phenylalanine concentrations in the medium superfusing cultured epithelial tumor cell lines (A549, MDA-MB-23) and a human fibroblast cell line (MRC-5) for periods of up to 72 h. Our scalable technology may be multiplexed for high-throughput monitoring of pH and multiple analytes in support of the broad metabolic qualification of microphysiological systems.
Collapse
Affiliation(s)
- Yiling Yang
- Department
of Electrical and Electronic Engineering, University of Melbourne, Melbourne, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, Melbourne, Victoria 3010, Australia
| | - Xumei Gao
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, Melbourne, Victoria 3010, Australia
| | - Bryce Widdicombe
- Department
of Electrical and Electronic Engineering, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Xiaodan Zhang
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
| | - Jana Lorraine Zielinski
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
| | - Tianhong Cheng
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
| | - Avanka Gunatilaka
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
| | - Kaylyn K. Leung
- Department
of Chemistry and Biochemistry, University
of California Santa Barbara, 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
| | - Ranjith Rajasekharan Unnithan
- Department
of Electrical and Electronic Engineering, University of Melbourne, Melbourne, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, Melbourne, Victoria 3010, Australia
| | - Alastair G. Stewart
- Department
of Biochemistry and Pharmacology, University
of Melbourne, Melbourne, Victoria 3010, Australia
- ARC
Centre for Personalised Therapeutics Technologies, Melbourne, Victoria 3010, Australia
| |
Collapse
|
30
|
Probst D, Batchu K, Younce JR, Sode K. Levodopa: From Biological Significance to Continuous Monitoring. ACS Sens 2024; 9:3828-3839. [PMID: 39047295 PMCID: PMC11348912 DOI: 10.1021/acssensors.4c00602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 06/21/2024] [Accepted: 07/03/2024] [Indexed: 07/27/2024]
Abstract
A continuous levodopa sensor can improve the quality of life for patients suffering with Parkinson's disease by enhancing levodopa titration and treatment effectiveness; however, its development is currently hindered by the absence of a specific levodopa molecular recognition element and limited insights into how real-time monitoring might affect clinical outcomes. This gap in research contributes to clinician uncertainty regarding the practical value of continuous levodopa monitoring data. This paper examines the current state of levodopa sensing and the inherent limitations in today's methods. Further, these challenges are described, including aspects such as interference from the metabolic pathway and adjunct medications, temporal resolution, and clinical questions, with a specific focus on a comprehensive selection of molecules, such as adjunct medications and structural isomers, as an interferent panel designed to assess and validate future levodopa sensors. We review insights and lessons from previously reported levodopa sensors and present a comparative analysis of potential molecular recognition elements, discussing their advantages and drawbacks.
Collapse
Affiliation(s)
- David Probst
- Joint
Department of Biomedical Engineering, The
University of North Carolina at Chapel Hill and North Carolina State
University, Chapel Hill, North Carolina 27599, United States
| | - Kartheek Batchu
- Joint
Department of Biomedical Engineering, The
University of North Carolina at Chapel Hill and North Carolina State
University, Chapel Hill, North Carolina 27599, United States
| | - John Robert Younce
- Department
of Neurology, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Koji Sode
- Joint
Department of Biomedical Engineering, The
University of North Carolina at Chapel Hill and North Carolina State
University, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
31
|
Ye C, Lukas H, Wang M, Lee Y, Gao W. Nucleic acid-based wearable and implantable electrochemical sensors. Chem Soc Rev 2024; 53:7960-7982. [PMID: 38985007 PMCID: PMC11308452 DOI: 10.1039/d4cs00001c] [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: 07/11/2024]
Abstract
The rapid advancements in nucleic acid-based electrochemical sensors for implantable and wearable applications have marked a significant leap forward in the domain of personal healthcare over the last decade. This technology promises to revolutionize personalized healthcare by facilitating the early diagnosis of diseases, monitoring of disease progression, and tailoring of individual treatment plans. This review navigates through the latest developments in this field, focusing on the strategies for nucleic acid sensing that enable real-time and continuous biomarker analysis directly in various biofluids, such as blood, interstitial fluid, sweat, and saliva. The review delves into various nucleic acid sensing strategies, emphasizing the innovative designs of biorecognition elements and signal transduction mechanisms that enable implantable and wearable applications. Special perspective is given to enhance nucleic acid-based sensor selectivity and sensitivity, which are crucial for the accurate detection of low-level biomarkers. The integration of such sensors into implantable and wearable platforms, including microneedle arrays and flexible electronic systems, actualizes their use in on-body devices for health monitoring. We also tackle the technical challenges encountered in the development of these sensors, such as ensuring long-term stability, managing the complexity of biofluid dynamics, and fulfilling the need for real-time, continuous, and reagentless detection. In conclusion, the review highlights the importance of these sensors in the future of medical engineering, offering insights into design considerations and future research directions to overcome existing limitations and fully realize the potential of nucleic acid-based electrochemical sensors for healthcare applications.
Collapse
Affiliation(s)
- Cui Ye
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA, USA.
| | - Heather Lukas
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA, USA.
| | - Minqiang Wang
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA, USA.
| | - Yerim Lee
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA, USA.
| | - Wei Gao
- Andrew and Peggy Cherng Department of Medical Engineering, Division of Engineering and Applied Science, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
32
|
Liu T, Simine L. DeltaGzip: Computing Biopolymer-Ligand Binding Affinity via Kolmogorov Complexity and Lossless Compression. J Chem Inf Model 2024; 64:5617-5623. [PMID: 38980667 DOI: 10.1021/acs.jcim.4c00461] [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: 07/10/2024]
Abstract
The design of biosequences for biosensing and therapeutics is a challenging multistep search and optimization task. In principle, computational modeling may speed up the design process by virtual screening of sequences based on their binding affinities to target molecules. However, in practice, existing machine-learned models trained to predict binding affinities lack the flexibility with respect to reaction conditions, and molecular dynamics simulations that can incorporate reaction conditions suffer from high computational costs. Here, we describe a computational approach called DeltaGzip that evaluates the free energy of binding in biopolymer-ligand complexes from ultrashort equilibrium molecular dynamics simulations. The entropy of binding is evaluated using the Kolmogorov complexity definition of entropy and approximated using a lossless compression algorithm, Gzip. We benchmark the method on a well-studied data set of protein-ligand complexes comparing the predictions of DeltaGzip to the free energies of binding obtained using Jarzynski equality and experimental measurements.
Collapse
Affiliation(s)
- Tao Liu
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| | - Lena Simine
- Department of Chemistry, McGill University, Montreal, Quebec H3A 0B8, Canada
| |
Collapse
|
33
|
Verrinder E, Gerson J, Leung K, Kippin TE, Plaxco KW. Dual-Frequency, Ratiometric Approaches to EAB Sensor Interrogation Support the Calibration-Free Measurement of Specific Molecules In Vivo. ACS Sens 2024; 9:3205-3211. [PMID: 38775190 DOI: 10.1021/acssensors.4c00516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/29/2024]
Abstract
Electrochemical aptamer-based (EAB) sensors represent the first molecular measurement technology that is both (1) independent of the chemical reactivity of the target, and thus generalizable to many targets and (2) able to function in an accurate, drift-corrected manner in situ in the living body. Signaling in EAB sensors is generated when an electrode-bound aptamer binds to its target ligand, altering the rate of electron transfer from an attached redox reporter and producing an easily detectable change in peak current when the sensor is interrogated using square wave voltammetry. Due to differences in the microscopic surface area of the interrogating electrodes, the baseline peak currents obtained from EAB sensors, however, can be highly variable. To overcome this, we have historically performed single-point calibration using measurements performed in a single sample of known target concentration. Here, however, we explore approaches to EAB sensor operation that negate the need to perform even single-point calibration of individual sensors. These are a ratiometric approach employing the ratio of the peak currents observed at two distinct square wave frequencies, and a kinetic differential measurement approach that employs the difference between peak currents seen at the two frequencies. Using in vivo measurements of vancomycin and phenylalanine as our test bed, we compared the output of these methods with that of the same sensor when single-point calibration was employed. Doing so we find that both methods support accurately drift-corrected measurements in vivo in live rats, even when employing rather crudely handmade devices. By removing the need to calibrate each individual sensor in a sample of known target concentration, these interrogation methods should significantly simplify the use of EAB sensors for in vivo applications.
Collapse
Affiliation(s)
- Elsi Verrinder
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| | - Julian Gerson
- Department of Chemistry and Biochemistry, University of California, Santa Barbara, Santa Barbara, California 93106, United States
- Department of Psychological and Brain Sciences, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| | - Kaylyn Leung
- Department of Chemistry and Biochemistry, 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, 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
- Biological Engineering Graduate Program, University of California, Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
34
|
Chung J, Billante A, Flatebo C, Leung KK, Gerson J, Emmons N, Kippin TE, Sepunaru L, Plaxco KW. Effects of storage conditions on the performance of an electrochemical aptamer-based sensor. SENSORS & DIAGNOSTICS 2024; 3:1044-1050. [PMID: 38882472 PMCID: PMC11170682 DOI: 10.1039/d4sd00066h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 04/11/2024] [Indexed: 06/18/2024]
Abstract
The electrochemical aptamer-based (EAB) sensor platform is the only molecular monitoring approach yet reported that is (1) real time and effectively continuous, (2) selective enough to deploy in situ in the living body, and (3) independent of the chemical or enzymatic reactivity of its target, rendering it adaptable to a wide range of analytes. These attributes suggest the EAB platform will prove to be an important tool in both biomedical research and clinical practice. To advance this possibility, here we have explored the stability of EAB sensors upon storage, using retention of the target recognizing aptamer, the sensor's signal gain, and the affinity of the aptamer as our performance metrics. Doing so we find that low-temperature (-20 °C) storage is sufficient to preserve sensor functionality for at least six months without the need for exogenous preservatives.
Collapse
Affiliation(s)
- Julia Chung
- Interdepartmental Program in Biomolecular Science and Engineering, University of California Santa Barbara Santa Barbara California 93106 USA
| | - Adriana Billante
- Department of Chemistry and Biochemistry, University of California Santa Barbara Santa Barbara California 93106 USA
| | - Charlotte Flatebo
- Institute for Collaborative Biotechnologies, University of California Santa Barbara Santa Barbara California 93106 USA
| | - Kaylyn K Leung
- Department of Chemistry and Biochemistry, University of California Santa Barbara Santa Barbara California 93106 USA
- Center for Bioengineering, University of California Santa Barbara Santa Barbara California 93106 USA
| | - Julian Gerson
- Center for Bioengineering, University of California Santa Barbara Santa Barbara California 93106 USA
- Department of Psychological and Brain Sciences, University of California Santa Barbara Santa Barbara California 93106 USA
| | - Nicole Emmons
- Center for Bioengineering, University of California Santa Barbara Santa Barbara California 93106 USA
- Department of Psychological and Brain Sciences, University of California Santa Barbara Santa Barbara California 93106 USA
| | - Tod E Kippin
- Interdepartmental Program in Biomolecular Science and Engineering, University of California Santa Barbara Santa Barbara California 93106 USA
- Department of Psychological and Brain Sciences, University of California Santa Barbara Santa Barbara California 93106 USA
| | - Lior Sepunaru
- Department of Chemistry and Biochemistry, University of California Santa Barbara Santa Barbara California 93106 USA
| | - Kevin W Plaxco
- Interdepartmental Program in Biomolecular Science and Engineering, University of California Santa Barbara Santa Barbara California 93106 USA
- Department of Chemistry and Biochemistry, University of California Santa Barbara Santa Barbara California 93106 USA
- Institute for Collaborative Biotechnologies, University of California Santa Barbara Santa Barbara California 93106 USA
- Center for Bioengineering, University of California Santa Barbara Santa Barbara California 93106 USA
| |
Collapse
|
35
|
Taguchi Y, Toma K, Iitani K, Arakawa T, Iwasaki Y, Mitsubayashi K. In Vitro Performance of a Long-Range Surface Plasmon Hydrogel Aptasensor for Continuous and Real-Time Vancomycin Measurement in Human Serum. ACS APPLIED MATERIALS & INTERFACES 2024; 16:28162-28171. [PMID: 38767334 DOI: 10.1021/acsami.4c03805] [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: 05/22/2024]
Abstract
This study investigated the suitability of surface modification for a long-range surface plasmon (LRSP) aptasensor using two different hydrogels, aiming at real-time monitoring of vancomycin (VCM) in undiluted serum and blood. Three different layer structures were formed on a gold surface of LRSP sensor chip using poly[2-methacryloyloxyethyl phosphorylcholine (MPC)-co-N-methacryloyl-(L)-tyrosinemethylester (MAT)] (PMM) and poly[MPC-co-2-ethylhexyl methacrylate (EHMA)-co-MAT] (PMEM). The peptide aptamer for VCM was immobilized in PMM and PMEM via MAT. Among four differently prepared sensor chips, the LRSP hydrogel aptasensor with PMM, referred to as the PMM hydrogel, exhibited the highest sensor output and superior antifouling properties. Following the optimization of the PMM hydrogel preparation conditions, the shelf life of the PMM hydrogel was determined to exceed 2 weeks, and the same sensor chip could be used for 102 days without significant performance deterioration. The PMM hydrogel was then applied for VCM measurement in undiluted serum in vitro, where it demonstrated a limit of detection of 0.098 μM and a dynamic range of 0.18-100 μM, covering the therapeutic range. Additionally, the PMM hydrogel enabled the continuous measurement of various VCM concentrations in serum without rinsing and showed a concentration-dependent output in undiluted blood. These findings underscore the potential of the PMM hydrogel for real-time and direct monitoring of VCM in body fluids.
Collapse
Affiliation(s)
- Yui Taguchi
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Koji Toma
- College of Engineering, Shibaura Institute of Technology, 3-7-5 Toyosu, Koto-ku, Tokyo 135-8548, Japan
| | - Kenta Iitani
- Department of Biomedical Devices and Instrumentation, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Takahiro Arakawa
- Department of Electric and Electronic Engineering, Tokyo University of Technology, 1404-1 Katakura, Hachioji City, Tokyo 192-0982, Japan
| | - Yasuhiko Iwasaki
- Faculty of Chemistry, Materials and Bioengineering, Kansai University, 3-3-35 Yamate-cho, Suita-shi, Osaka 564-8680, Japan
| | - Kohji Mitsubayashi
- Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
- Department of Biomedical Devices and Instrumentation, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| |
Collapse
|
36
|
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
|
37
|
Leung KK, Gerson J, Emmons N, Heemstra JM, Kippin TE, Plaxco KW. The Use of Xenonucleic Acids Significantly Reduces the In Vivo Drift of Electrochemical Aptamer-Based Sensors. Angew Chem Int Ed Engl 2024; 63:e202316678. [PMID: 38500260 PMCID: PMC11821280 DOI: 10.1002/anie.202316678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 02/09/2024] [Accepted: 03/17/2024] [Indexed: 03/20/2024]
Abstract
Electrochemical aptamer-based sensors support the high-frequency, real-time monitoring of molecules-of-interest in vivo. Achieving this requires methods for correcting the sensor drift seen during in vivo placements. While this correction ensures EAB sensor measurements remain accurate, as drift progresses it reduces the signal-to-noise ratio and precision. Here, we show that enzymatic cleavage of the sensor's target-recognizing DNA aptamer is a major source of this signal loss. To demonstrate this, we deployed a tobramycin-detecting EAB sensor analog fabricated with the DNase-resistant "xenonucleic acid" 2'O-methyl-RNA in a live rat. In contrast to the sensor employing the equivalent DNA aptamer, the 2'O-methyl-RNA aptamer sensor lost very little signal and had improved signal-to-noise. We further characterized the EAB sensor drift using unstructured DNA or 2'O-methyl-RNA oligonucleotides. While the two devices drift similarly in vitro in whole blood, the in vivo drift of the 2'O-methyl-RNA-employing device is less compared to the DNA-employing device. Studies of the electron transfer kinetics suggested that the greater drift of the latter sensor arises due to enzymatic DNA degradation. These findings, coupled with advances in the selection of aptamers employing XNA, suggest a means of improving EAB sensor stability when they are used to perform molecular monitoring in the living body.
Collapse
Affiliation(s)
- Kaylyn K. Leung
- Department of Chemistry and Biochemistry, 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
- Center for Bioengineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Nicole Emmons
- Department of Psychological and Brain Sciences, University of California, Santa Barbara
- Center for Bioengineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Jennifer M. Heemstra
- Department of Chemistry, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Tod E. Kippin
- Department of Psychological and Brain Sciences, University of California, Santa Barbara
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara
| | - Kevin W. Plaxco
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, CA 93106, USA
- Center for Bioengineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| |
Collapse
|
38
|
Abeykoon SW, White RJ. Single Voltammetric Sweep Calibration-Free Interrogation of Electrochemical Aptamer-Based Sensors Employing Continuous Square Wave Voltammetry. Anal Chem 2024; 96:6958-6967. [PMID: 38662230 PMCID: PMC12014223 DOI: 10.1021/acs.analchem.3c05920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Continuous square wave voltammetry (cSWV) is a technique that enables the continuous collection of current data (at 100 kHz) to maximize the information content obtainable from a single voltammetric sweep. This data collection procedure results in the generation of multiple voltammograms corresponding to different effective square wave frequencies. The application of cSWV brings significant benefits to electrochemical aptamer-based (E-AB) sensors. The E-AB sensor platform permits continuous real-time monitoring of small biological molecules. Traditionally, E-AB sensors report only on changes in analyte concentration rather than absolute quantification in matrices when basal concentrations are not known a priori. This is because they exhibit a voltammetric peak current even in the absence of a target. However, using a dual-frequency approach, calibration-free sensing can be performed effectively, eliminating the sensor-to-sensor variation by taking ratiometric current responses obtained at two different frequencies from two different voltammetric sweeps. In employing our approach, cSWV provides a great advantage over the conventionally used square wave voltammetry since the required voltammograms are collected with a single sweep, which improves the temporal resolution of the measurement when considering the current at multiple frequencies for improved accuracy and reduced surface interrogation. Moreover, we show here that using cSWV provides significantly improved concentration predictions. E-AB sensors sensitive to ATP and tobramycin were interrogated across a wide range of concentrations. With this approach, cSWV allowed us to estimate the target concentration, retaining up to an ±5% error of the expected concentration when tested in buffer and complex media.
Collapse
Affiliation(s)
- Sanduni W. Abeykoon
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221-0172, United States
| | - Ryan J. White
- Department of Chemistry, University of Cincinnati, Cincinnati, Ohio 45221-0172, United States
| |
Collapse
|
39
|
Wu Y, Shi J, Kippin TE, Plaxco KW. Codeposition Enhances the Performance of Electrochemical Aptamer-Based Sensors. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2024; 40:8703-8710. [PMID: 38616608 PMCID: PMC11821552 DOI: 10.1021/acs.langmuir.4c00585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/16/2024]
Abstract
Electrochemical aptamer-based (EAB) sensors, a minimally invasive means of performing high-frequency, real-time measurement of drugs and biomarkers in situ in the body, have traditionally been fabricated by depositing their target-recognizing aptamer onto an interrogating gold electrode using a "sequential" two-step method involving deposition of the thiol-modified oligonucleotide (typically for 1 h) followed by incubation in mercaptohexanol solution (typically overnight) to complete the formation of a stable, self-assembled monolayer. Here we use EAB sensors targeting vancomycin, tryptophan, and phenylalanine to show that "codeposition", a less commonly employed EAB fabrication method in which the thiol-modified aptamer and the mercaptohexanol diluent are deposited on the electrode simultaneously and for as little as 1 h, improves the signal gain (relative change in signal upon the addition of high concentrations of the target) of the vancomycin and tryptophan sensors without significantly reducing their stability. In contrast, the gain of the phenylalanine sensor is effectively identical irrespective of the fabrication approach employed. This sensor, however, appears to employ binding-induced displacement of the redox reporter rather than binding-induced folding as its signal transduction mechanism, suggesting in turn a mechanism for the improvement observed for the other two sensors. Codeposition thus not only provides a more convenient means of fabricating EAB sensors but also can improve their performance.
Collapse
Affiliation(s)
- Yuyang Wu
- Department of Chemistry and Biochemistry, University of California Santa Barbara, Santa Barbara, California 93106, United States
| | - Jinyuan Shi
- Department of Chemistry and Biochemistry, 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, Santa Barbara, California 93106, United States
- Department of Molecular Cellular and Developmental Biology, University of California, Santa Barbara, California 93106, United States
- Neuroscience Research Institute, University of California, Santa Barbara, California 93106, United States
- Biological Engineering Graduate Program, University of California Santa Barbara, 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
- Biological Engineering Graduate Program, University of California Santa Barbara, Santa Barbara, California 93106, United States
| |
Collapse
|
40
|
Campuzano S, Barderas R, Moreno-Casbas MT, Almeida Á, Pingarrón JM. Pursuing precision in medicine and nutrition: the rise of electrochemical biosensing at the molecular level. Anal Bioanal Chem 2024; 416:2151-2172. [PMID: 37420009 PMCID: PMC10951035 DOI: 10.1007/s00216-023-04805-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/10/2023] [Accepted: 06/13/2023] [Indexed: 07/09/2023]
Abstract
In the era that we seek personalization in material things, it is becoming increasingly clear that the individualized management of medicine and nutrition plays a key role in life expectancy and quality of life, allowing participation to some extent in our welfare and the use of societal resources in a rationale and equitable way. The implementation of precision medicine and nutrition are highly complex challenges which depend on the development of new technologies able to meet important requirements in terms of cost, simplicity, and versatility, and to determine both individually and simultaneously, almost in real time and with the required sensitivity and reliability, molecular markers of different omics levels in biofluids extracted, secreted (either naturally or stimulated), or circulating in the body. Relying on representative and pioneering examples, this review article critically discusses recent advances driving the position of electrochemical bioplatforms as one of the winning horses for the implementation of suitable tools for advanced diagnostics, therapy, and precision nutrition. In addition to a critical overview of the state of the art, including groundbreaking applications and challenges ahead, the article concludes with a personal vision of the imminent roadmap.
Collapse
Affiliation(s)
- Susana Campuzano
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain.
| | - Rodrigo Barderas
- UFIEC, Instituto de Salud Carlos III, Majadahonda, 28220, Madrid, Spain
| | - Maria Teresa Moreno-Casbas
- Nursing and Healthcare Research Unit (Investén-isciii), Instituto de Salud Carlos III, Madrid, Spain
- Biomedical Research Center Network for Frailty and Healthy Ageing (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Ángeles Almeida
- Instituto de Biología Funcional y Genómica, CSIC, Universidad de Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca, Hospital Universitario de Salamanca, CSIC, Universidad de Salamanca, Salamanca, Spain
| | - José M Pingarrón
- Departamento de Química Analítica, Facultad de CC. Químicas, Universidad Complutense de Madrid, 28040, Madrid, Spain
| |
Collapse
|
41
|
Kong D, Thompson IAP, Maganzini N, Eisenstein M, Soh HT. Aptamer-Antibody Chimera Sensors for Sensitive, Rapid, and Reversible Molecular Detection in Complex Samples. ACS Sens 2024; 9:1168-1177. [PMID: 38407035 DOI: 10.1021/acssensors.3c01638] [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: 02/27/2024]
Abstract
The development of receptors suitable for the continuous detection of analytes in complex, interferent-rich samples remains challenging. Antibodies are highly sensitive but difficult to engineer in order to introduce signaling functionality, while aptamer switches are easy to construct but often yield only a modest target sensitivity. We present here a programmable antibody and DNA aptamer switch (PANDAS), which combines the desirable properties of both receptors by using a nucleic acid tether to link an analyte-specific antibody to an internal strand-displacement (ISD)-based aptamer switch that recognizes the same target through different epitopes. The antibody increases PANDAS analyte binding due to its high affinity, and the effective concentration between the two receptors further enhances two-epitope binding and fluorescent aptamer signaling. We developed a PANDAS sensor for the clotting protein thrombin and show that a tuned design achieves a greater than 300-fold enhanced sensitivity compared to that of using an aptamer alone. This design also exhibits reversible binding, enabling repeated measurements with a temporal resolution of ∼10 min, and retains excellent sensitivity even in interferent-rich samples. With future development, this PANDAS approach could enable the adaptation of existing protein-binding aptamers with modest affinity to sensors that deliver excellent sensitivity and minute-scale resolution in minimally prepared biological specimens.
Collapse
Affiliation(s)
- Dehui Kong
- Department of Radiology, Stanford University, Stanford, California 94305, United States
| | - Ian A P Thompson
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - Nicolo Maganzini
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - Michael Eisenstein
- Department of Radiology, Stanford University, Stanford, California 94305, United States
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| | - Hyongsok Tom Soh
- Department of Radiology, Stanford University, Stanford, California 94305, United States
- Department of Electrical Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
42
|
Liang WS, Beaulieu-Jones B, Smalley S, Snyder M, Goetz LH, Schork NJ. Emerging therapeutic drug monitoring technologies: considerations and opportunities in precision medicine. Front Pharmacol 2024; 15:1348112. [PMID: 38545548 PMCID: PMC10965556 DOI: 10.3389/fphar.2024.1348112] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Accepted: 02/27/2024] [Indexed: 11/11/2024] Open
Abstract
In recent years, the development of sensor and wearable technologies have led to their increased adoption in clinical and health monitoring settings. One area that is in early, but promising, stages of development is the use of biosensors for therapeutic drug monitoring (TDM). Traditionally, TDM could only be performed in certified laboratories and was used in specific scenarios to optimize drug dosage based on measurement of plasma/blood drug concentrations. Although TDM has been typically pursued in settings involving medications that are challenging to manage, the basic approach is useful for characterizing drug activity. TDM is based on the idea that there is likely a clear relationship between plasma/blood drug concentration (or concentration in other matrices) and clinical efficacy. However, these relationships may vary across individuals and may be affected by genetic factors, comorbidities, lifestyle, and diet. TDM technologies will be valuable for enabling precision medicine strategies to determine the clinical efficacy of drugs in individuals, as well as optimizing personalized dosing, especially since therapeutic windows may vary inter-individually. In this mini-review, we discuss emerging TDM technologies and their applications, and factors that influence TDM including drug interactions, polypharmacy, and supplement use. We also discuss how using TDM within single subject (N-of-1) and aggregated N-of-1 clinical trial designs provides opportunities to better capture drug response and activity at the individual level. Individualized TDM solutions have the potential to help optimize treatment selection and dosing regimens so that the right drug and right dose may be matched to the right person and in the right context.
Collapse
Affiliation(s)
- Winnie S. Liang
- Net/Bio Inc, Los Angeles, CA, United States
- Translational Genomics Research Institute (TGen), Phoenix, AZ, United States
| | - Brett Beaulieu-Jones
- Net/Bio Inc, Los Angeles, CA, United States
- University of Chicago, Chicago, IL, United States
| | | | - Michael Snyder
- Net/Bio Inc, Los Angeles, CA, United States
- Stanford University, Stanford, CA, United States
| | | | - Nicholas J. Schork
- Net/Bio Inc, Los Angeles, CA, United States
- Translational Genomics Research Institute (TGen), Phoenix, AZ, United States
| |
Collapse
|
43
|
Wang X, Kong F, Liu Y, Lv S, Zhang K, Sun S, Liu J, Wang M, Cai X, Jin H, Yan S, Luo J. 17β-estradiol biosensors based on different bioreceptors and their applications. Front Bioeng Biotechnol 2024; 12:1347625. [PMID: 38357703 PMCID: PMC10864596 DOI: 10.3389/fbioe.2024.1347625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 01/16/2024] [Indexed: 02/16/2024] Open
Abstract
17β-Estradiol (E2) is a critical sex steroid hormone, which has significant effects on the endocrine systems of both humans and animals. E2 is also believed to play neurotrophic and neuroprotective roles in the brain. Biosensors present a powerful tool to detect E2 because of their small, efficient, and flexible design. Furthermore, Biosensors can quickly and accurately obtain detection results with only a small sampling amount, which greatly meets the detection of the environment, food safety, medicine safety, and human body. This review focuses on previous studies of biosensors for detecting E2 and divides them into non-biometric sensors, enzyme biosensors, antibody biosensors, and aptamer biosensors according to different bioreceptors. The advantages, disadvantages, and design points of various bioreceptors for E2 detection are analyzed and summarized. Additionally, applications of different bioreceptors of E2 detection are presented and highlight the field of environmental monitoring, food and medicine safety, and disease detection in recent years. Finally, the development of E2 detection by biosensor is prospected.
Collapse
Affiliation(s)
- Xinyi Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Fanli Kong
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Yaoyao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Shiya Lv
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Kui Zhang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Shutong Sun
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Juntao Liu
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Mixia Wang
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Xinxia Cai
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| | - Hongyan Jin
- Obstetrics and Gynecology Department, Peking University First Hospital, Beijing, China
| | - Shi Yan
- Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, China
| | - Jinping Luo
- State Key Laboratory of Transducer Technology, Aerospace Information Research Institute, Chinese Academy of Sciences, Beijing, China
- School of Electronic, Electrical and Communication Engineering, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
Watkins Z, McHenry A, Heikenfeld J. Wearing the Lab: Advances and Challenges in Skin-Interfaced Systems for Continuous Biochemical Sensing. ADVANCES IN BIOCHEMICAL ENGINEERING/BIOTECHNOLOGY 2024; 187:223-282. [PMID: 38273210 DOI: 10.1007/10_2023_238] [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
Continuous, on-demand, and, most importantly, contextual data regarding individual biomarker concentrations exemplify the holy grail for personalized health and performance monitoring. This is well-illustrated for continuous glucose monitoring, which has drastically improved outcomes and quality of life for diabetic patients over the past 2 decades. Recent advances in wearable biosensing technologies (biorecognition elements, transduction mechanisms, materials, and integration schemes) have begun to make monitoring of other clinically relevant analytes a reality via minimally invasive skin-interfaced devices. However, several challenges concerning sensitivity, specificity, calibration, sensor longevity, and overall device lifetime must be addressed before these systems can be made commercially viable. In this chapter, a logical framework for developing a wearable skin-interfaced device for a desired application is proposed with careful consideration of the feasibility of monitoring certain analytes in sweat and interstitial fluid and the current development of the tools available to do so. Specifically, we focus on recent advancements in the engineering of biorecognition elements, the development of more robust signal transduction mechanisms, and novel integration schemes that allow for continuous quantitative analysis. Furthermore, we highlight the most compelling and promising prospects in the field of wearable biosensing and the challenges that remain in translating these technologies into useful products for disease management and for optimizing human performance.
Collapse
Affiliation(s)
- Zach Watkins
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA.
| | - Adam McHenry
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| | - Jason Heikenfeld
- Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| |
Collapse
|
47
|
Hariri AA, Cartwright AP, Dory C, Gidi Y, Yee S, Thompson IAP, Fu KX, Yang K, Wu D, Maganzini N, Feagin T, Young BE, Afshar BH, Eisenstein M, Digonnet MJF, Vuckovic J, Soh HT. Modular Aptamer Switches for the Continuous Optical Detection of Small-Molecule Analytes in Complex Media. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2304410. [PMID: 37975267 DOI: 10.1002/adma.202304410] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 11/10/2023] [Indexed: 11/19/2023]
Abstract
Aptamers are a promising class of affinity reagents because signal transduction mechanisms can be built into the reagent, so that they can directly produce a physically measurable output signal upon target binding. However, endowing the signal transduction functionality into an aptamer remains a trial-and-error process that can compromise its affinity or specificity and typically requires knowledge of the ligand binding domain or its structure. In this work, a design architecture that can convert an existing aptamer into a "reversible aptamer switch" whose kinetic and thermodynamic properties can be tuned without a priori knowledge of the ligand binding domain or its structure is described. Finally, by combining these aptamer switches with evanescent-field-based optical detection hardware that minimizes sample autofluorescence, this study demonstrates the first optical biosensor system that can continuously measure multiple biomarkers (dopamine and cortisol) in complex samples (artificial cerebrospinal fluid and undiluted plasma) with second and subsecond-scale time responses at physiologically relevant concentration ranges.
Collapse
Affiliation(s)
- Amani A Hariri
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Alyssa P Cartwright
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Constantin Dory
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Yasser Gidi
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Steven Yee
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Ian A P Thompson
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Kaiyu X Fu
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Kiyoul Yang
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Diana Wu
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
| | - Nicolò Maganzini
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Trevor Feagin
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Brian E Young
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
| | - Behrad Habib Afshar
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | | | - Michel J F Digonnet
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Jelena Vuckovic
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - H Tom Soh
- Department of Radiology, Stanford University, Stanford, CA, 94305, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA, 94305, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
48
|
Fang Z, Zhang H, Guo J, Guo J. Overview of therapeutic drug monitoring and clinical practice. Talanta 2024; 266:124996. [PMID: 37562225 DOI: 10.1016/j.talanta.2023.124996] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/29/2023] [Accepted: 07/25/2023] [Indexed: 08/12/2023]
Abstract
With the rapid development of clinical pharmacy in China, therapeutic drug monitoring (TDM) has become an essential tool for guiding rational clinical drug use and is widely concerned. TDM is a tool that combines pharmacokinetic and pharmacodynamic knowledge to optimize personalized drug therapy, which can improve treatment outcomes, reduce drug-drug toxicity, and avoid the risk of developing drug resistance. To effectively implement TDM, accurate and sophisticated analytical methods are required. By researching the literature published in recent years, we summarize the types of commonly monitored drugs, therapeutic windows, and clinical assays and track the trends and hot spots of therapeutic drug monitoring. The purpose is to provide guidelines for clinical blood drug concentration monitoring, to implement individualized drug delivery programs better, to ensure the rational use of drugs for patients, and to provide a reference for the group to carry out related topics in the future.
Collapse
Affiliation(s)
- Zijun Fang
- University of Southwest Petroleum University, College of Mechanical and Electrical Engineering, Chengdu, China
| | - He Zhang
- University of Southwest Petroleum University, College of Mechanical and Electrical Engineering, Chengdu, China
| | - Jiuchuan Guo
- University of Electronic Science and Technology of China, Chengdu, China.
| | - Jinhong Guo
- School of Sensing Science and Engineering, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
49
|
Carou-Senra P, Rodríguez-Pombo L, Monteagudo-Vilavedra E, Awad A, Alvarez-Lorenzo C, Basit AW, Goyanes A, Couce ML. 3D Printing of Dietary Products for the Management of Inborn Errors of Intermediary Metabolism in Pediatric Populations. Nutrients 2023; 16:61. [PMID: 38201891 PMCID: PMC10780524 DOI: 10.3390/nu16010061] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 12/21/2023] [Accepted: 12/22/2023] [Indexed: 01/12/2024] Open
Abstract
The incidence of Inborn Error of Intermediary Metabolism (IEiM) diseases may be low, yet collectively, they impact approximately 6-10% of the global population, primarily affecting children. Precise treatment doses and strict adherence to prescribed diet and pharmacological treatment regimens are imperative to avert metabolic disturbances in patients. However, the existing dietary and pharmacological products suffer from poor palatability, posing challenges to patient adherence. Furthermore, frequent dose adjustments contingent on age and drug blood levels further complicate treatment. Semi-solid extrusion (SSE) 3D printing technology is currently under assessment as a pioneering method for crafting customized chewable dosage forms, surmounting the primary limitations prevalent in present therapies. This method offers a spectrum of advantages, including the flexibility to tailor patient-specific doses, excipients, and organoleptic properties. These elements are pivotal in ensuring the treatment's efficacy, safety, and adherence. This comprehensive review presents the current landscape of available dietary products, diagnostic methods, therapeutic monitoring, and the latest advancements in SSE technology. It highlights the rationale underpinning their adoption while addressing regulatory aspects imperative for their seamless integration into clinical practice.
Collapse
Affiliation(s)
- Paola Carou-Senra
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Materials Institute (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (P.C.-S.); (L.R.-P.); (C.A.-L.)
| | - Lucía Rodríguez-Pombo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Materials Institute (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (P.C.-S.); (L.R.-P.); (C.A.-L.)
| | - Einés Monteagudo-Vilavedra
- Servicio de Neonatología, Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Congénitas, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Universidad de Santiago de Compostela, RICORS, CIBERER, MetabERN, 15706 Santiago de Compostela, Spain;
| | - Atheer Awad
- Department of Clinical, Pharmaceutical and Biological Sciences, University of Hertfordshire, College Lane, Hatfield AL10 9AB, UK;
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Materials Institute (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (P.C.-S.); (L.R.-P.); (C.A.-L.)
| | - Abdul W. Basit
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK;
- FABRX Ltd., Henwood House, Henwood, Ashford, Kent TN24 8DH, UK
- FABRX Artificial Intelligence, 27543 O Saviñao, Spain
| | - Alvaro Goyanes
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Materials Institute (iMATUS) and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; (P.C.-S.); (L.R.-P.); (C.A.-L.)
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK;
- FABRX Ltd., Henwood House, Henwood, Ashford, Kent TN24 8DH, UK
- FABRX Artificial Intelligence, 27543 O Saviñao, Spain
| | - María L. Couce
- Servicio de Neonatología, Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Congénitas, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Universidad de Santiago de Compostela, RICORS, CIBERER, MetabERN, 15706 Santiago de Compostela, Spain;
| |
Collapse
|
50
|
Seibold JM, Abeykoon SW, Ross AE, White RJ. Development of an Electrochemical, Aptamer-Based Sensor for Dynamic Detection of Neuropeptide Y. ACS Sens 2023; 8:4504-4511. [PMID: 38033269 PMCID: PMC11214579 DOI: 10.1021/acssensors.3c00855] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
The ability to monitor dynamic changes in neuropeptide Y (NPY) levels in complex environments can have an impact on many fields, including neuroscience and immunology. Here, we describe the development of an electrochemical, aptamer-based (E-AB) sensor for the dynamic (reversible) measurement of physiologically relevant (nanomolar) concentrations of neuropeptide Y. The E-AB sensors are fabricated using a previously described 80 nucleotide aptamer1 reported to specifically bind NPY with a binding affinity Kd = 0.3 ± 0.2 uM. We investigated two redox tag placement locations on the aptamer sequence (terminal vs internal) and various sensor fabrication and interrogation parameters to tune the performance of the resulting sensor. The best-performing sensor architecture displayed a physiologically relevant dynamic range (nM) and low limit of detection and is selective among competitors and similar molecules. The development of this sensor accomplishes two breakthroughs: first, the development of a nonmicrofluidic aptamer-based electrochemical sensor that can detect NPY on a physiologically relevant (seconds to minutes) time scale and across a relevant concentration range; second, the expansion of the range of molecules for which an electrochemical, aptamer-based sensor can be used.
Collapse
Affiliation(s)
- Jordan M. Seibold
- University of Cincinnati Department of Chemistry 312 College Dr. 404 Crosley Tower Cincinnati, OH 45221-0172
| | - Sanduni W. Abeykoon
- University of Cincinnati Department of Chemistry 312 College Dr. 404 Crosley Tower Cincinnati, OH 45221-0172
| | - Ashley E. Ross
- University of Cincinnati Department of Chemistry 312 College Dr. 404 Crosley Tower Cincinnati, OH 45221-0172
| | - Ryan J. White
- University of Cincinnati Department of Chemistry 312 College Dr. 404 Crosley Tower Cincinnati, OH 45221-0172
- Department of Electrical and Computer Engineering
| |
Collapse
|