1
|
Mudugamuwa A, Roshan U, Hettiarachchi S, Cha H, Musharaf H, Kang X, Trinh QT, Xia HM, Nguyen N, Zhang J. Periodic Flows in Microfluidics. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2404685. [PMID: 39246195 PMCID: PMC11636114 DOI: 10.1002/smll.202404685] [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: 06/08/2024] [Revised: 08/24/2024] [Indexed: 09/10/2024]
Abstract
Microfluidics, the science and technology of manipulating fluids in microscale channels, offers numerous advantages, such as low energy consumption, compact device size, precise control, fast reaction, and enhanced portability. These benefits have led to applications in biomedical assays, disease diagnostics, drug discovery, neuroscience, and so on. Fluid flow within microfluidic channels is typically in the laminar flow region, which is characterized by low Reynolds numbers but brings the challenge of efficient mixing of fluids. Periodic flows are time-dependent fluid flows, featuring repetitive patterns that can significantly improve fluid mixing and extend the effective length of microchannels for submicron and nanoparticle manipulation. Besides, periodic flow is crucial in organ-on-a-chip (OoC) for accurately modeling physiological processes, advancing disease understanding, drug development, and personalized medicine. Various techniques for generating periodic flows have been reported, including syringe pumps, peristalsis, and actuation based on electric, magnetic, acoustic, mechanical, pneumatic, and fluidic forces, yet comprehensive reviews on this topic remain limited. This paper aims to provide a comprehensive review of periodic flows in microfluidics, from fundamental mechanisms to generation techniques and applications. The challenges and future perspectives are also discussed to exploit the potential of periodic flows in microfluidics.
Collapse
Affiliation(s)
- Amith Mudugamuwa
- Queensland Micro and Nanotechnology CentreGriffith UniversityBrisbaneQLD4111Australia
| | - Uditha Roshan
- Queensland Micro and Nanotechnology CentreGriffith UniversityBrisbaneQLD4111Australia
| | - Samith Hettiarachchi
- Queensland Micro and Nanotechnology CentreGriffith UniversityBrisbaneQLD4111Australia
| | - Haotian Cha
- Queensland Micro and Nanotechnology CentreGriffith UniversityBrisbaneQLD4111Australia
| | - Hafiz Musharaf
- Queensland Micro and Nanotechnology CentreGriffith UniversityBrisbaneQLD4111Australia
| | - Xiaoyue Kang
- Queensland Micro and Nanotechnology CentreGriffith UniversityBrisbaneQLD4111Australia
| | - Quang Thang Trinh
- Queensland Micro and Nanotechnology CentreGriffith UniversityBrisbaneQLD4111Australia
| | - Huan Ming Xia
- School of Mechanical EngineeringNanjing University of Science and TechnologyNanjing210094P. R. China
| | - Nam‐Trung Nguyen
- Queensland Micro and Nanotechnology CentreGriffith UniversityBrisbaneQLD4111Australia
| | - Jun Zhang
- Queensland Micro and Nanotechnology CentreGriffith UniversityBrisbaneQLD4111Australia
- School of Engineering and Built EnvironmentGriffith UniversityBrisbaneQLD4111Australia
| |
Collapse
|
2
|
Li R, Fan H, Chen Y, Yin S, Liu GL, Li Y, Huang L. MXene-Graphene Oxide Heterostructured Films for Enhanced Metasurface Plasmonic Biosensing in Continuous Glucose Monitoring. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2410376. [PMID: 39569760 DOI: 10.1002/advs.202410376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 10/22/2024] [Indexed: 11/22/2024]
Abstract
Non-invasive biosensors have attracted attention for their potential to obtain continuous, real-time physiological information through measurements of biochemical markers, such as one of the most important-glucose, in biological fluids. Although some optical sensing materials are used in non-invasive devices for continuous glucose monitoring (CGM), surface or localized plasmon sensing material are seldom applied in CGM owing to modest sensitivity and bulk sensing apparatus. Herein, a metasurface (MGMSPR) biosensor based on the metasurface plasmon resonance chip modified with heterostructured Ti3C2 MXene-Graphene oxide (MG) is reported, which potentially enables ultra-sensitive glucose detection. The sensor consists of a dual-channel microfluidic device integrated with silver mirror enhanced MGMSPR chips. Not only does it promote the entry of glucose oxidase (GOD) into the internal pores and enhance the stable fixation of GOD in the membrane, but also the integration of MG material provides a high specific surface area and unique electronic properties, thereby significantly enhancing the sensitivity of the MGMSPR sensor. The detection limit of MGMSPR biosensor is 106.8 µM. This pioneering approach opens new avenues for monitoring physiological parameters and process analytical technology on an optical platform, providing continuous health monitoring and production process control through optical sensors.
Collapse
Affiliation(s)
- Rui Li
- College of Life Science and Technology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1037 LuoYu Road, Wuhan, 430070, P. R. China
| | - Hongli Fan
- College of Life Science and Technology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1037 LuoYu Road, Wuhan, 430070, P. R. China
| | - Youqian Chen
- College of Life Science and Technology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1037 LuoYu Road, Wuhan, 430070, P. R. China
| | - Shaoping Yin
- School of Pharmacy, Jiangsu Provincial Engineering Research Center of Traditional Chinese Medicine External Medication Development and Application, Nanjing University of Chinese Medicine, Nanjing, 210023, P. R. China
| | - Gang L Liu
- College of Life Science and Technology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1037 LuoYu Road, Wuhan, 430070, P. R. China
| | - Yanan Li
- Biosensor R&D Department, Liangzhun (Wuhan) Life Technology Co., Ltd., Wuhan, 430070, P. R. China
| | - Liping Huang
- College of Life Science and Technology, State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, 1037 LuoYu Road, Wuhan, 430070, P. R. China
- School of Food Science and Pharmaceutical Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
- Biosensor R&D Department, Liangzhun (Wuhan) Life Technology Co., Ltd., Wuhan, 430070, P. R. China
| |
Collapse
|
3
|
Zhang S, Staples AE. Microfluidic-based systems for the management of diabetes. Drug Deliv Transl Res 2024; 14:2989-3008. [PMID: 38509342 PMCID: PMC11445324 DOI: 10.1007/s13346-024-01569-y] [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] [Accepted: 03/04/2024] [Indexed: 03/22/2024]
Abstract
Diabetes currently affects approximately 500 million people worldwide and is one of the most common causes of mortality in the United States. To diagnose and monitor diabetes, finger-prick blood glucose testing has long been used as the clinical gold standard. For diabetes treatment, insulin is typically delivered subcutaneously through cannula-based syringes, pens, or pumps in almost all type 1 diabetic (T1D) patients and some type 2 diabetic (T2D) patients. These painful, invasive approaches can cause non-adherence to glucose testing and insulin therapy. To address these problems, researchers have developed miniaturized blood glucose testing devices as well as microfluidic platforms for non-invasive glucose testing through other body fluids. In addition, glycated hemoglobin (HbA1c), insulin levels, and cellular biomechanics-related metrics have also been considered for microfluidic-based diabetes diagnosis. For the treatment of diabetes, insulin has been delivered transdermally through microdevices, mostly through microneedle array-based, minimally invasive injections. Researchers have also developed microfluidic platforms for oral, intraperitoneal, and inhalation-based delivery of insulin. For T2D patients, metformin, glucagon-like peptide 1 (GLP-1), and GLP-1 receptor agonists have also been delivered using microfluidic technologies. Thus far, clinical studies have been widely performed on microfluidic-based diabetes monitoring, especially glucose sensing, yet technologies for the delivery of insulin and other drugs to diabetic patients with microfluidics are still mostly in the preclinical stage. This article provides a concise review of the role of microfluidic devices in the diagnosis and monitoring of diabetes, as well as the delivery of pharmaceuticals to treat diabetes using microfluidic technologies in the recent literature.
Collapse
Affiliation(s)
- Shuyu Zhang
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Blacksburg, VA, 24061, USA.
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, 24061, USA.
| | - Anne E Staples
- Virginia Tech-Wake Forest School of Biomedical Engineering and Sciences, Blacksburg, VA, 24061, USA
- Department of Biomedical Engineering and Mechanics, Virginia Tech, Blacksburg, VA, 24061, USA
| |
Collapse
|
4
|
Zhang Y, Zheng XT, Zhang X, Pan J, Thean AVY. Hybrid Integration of Wearable Devices for Physiological Monitoring. Chem Rev 2024; 124:10386-10434. [PMID: 39189683 DOI: 10.1021/acs.chemrev.3c00471] [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: 08/28/2024]
Abstract
Wearable devices can provide timely, user-friendly, non- or minimally invasive, and continuous monitoring of human health. Recently, multidisciplinary scientific communities have made significant progress regarding fully integrated wearable devices such as sweat wearable sensors, saliva sensors, and wound sensors. However, the translation of these wearables into markets has been slow due to several reasons associated with the poor system-level performance of integrated wearables. The wearability consideration for wearable devices compromises many properties of the wearables. Besides, the limited power capacity of wearables hinders continuous monitoring for extended duration. Furthermore, peak-power operations for intensive computations can quickly create thermal issues in the compact form factor that interfere with wearability and sensor operations. Moreover, wearable devices are constantly subjected to environmental, mechanical, chemical, and electrical interferences and variables that can invalidate the collected data. This generates the need for sophisticated data analytics to contextually identify, include, and exclude data points per multisensor fusion to enable accurate data interpretation. This review synthesizes the challenges surrounding the wearable device integration from three aspects in terms of hardware, energy, and data, focuses on a discussion about hybrid integration of wearable devices, and seeks to provide comprehensive guidance for designing fully functional and stable wearable devices.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Xin Ting Zheng
- Institute of Materials Research and Engineering (IMRE), Agency for Science Technology and Research (A*STAR), 2 Fusionopolis Way, Innovis #08-03, Singapore 138634, Singapore
| | - Xiangyu Zhang
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Jieming Pan
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117576, Singapore
| | - Aaron Voon-Yew Thean
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117576, Singapore
| |
Collapse
|
5
|
Gonçalves Pereira J, Fernandes J, Mendes T, Gonzalez FA, Fernandes SM. Artificial Intelligence to Close the Gap between Pharmacokinetic/Pharmacodynamic Targets and Clinical Outcomes in Critically Ill Patients: A Narrative Review on Beta Lactams. Antibiotics (Basel) 2024; 13:853. [PMID: 39335027 PMCID: PMC11428226 DOI: 10.3390/antibiotics13090853] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Antimicrobial dosing can be a complex challenge. Although a solid rationale exists for a link between antibiotic exposure and outcome, conflicting data suggest a poor correlation between pharmacokinetic/pharmacodynamic targets and infection control. Different reasons may lead to this discrepancy: poor tissue penetration by β-lactams due to inflammation and inadequate tissue perfusion; different bacterial response to antibiotics and biofilms; heterogeneity of the host's immune response and drug metabolism; bacterial tolerance and acquisition of resistance during therapy. Consequently, either a fixed dose of antibiotics or a fixed target concentration may be doomed to fail. The role of biomarkers in understanding and monitoring host response to infection is also incompletely defined. Nowadays, with the ever-growing stream of data collected in hospitals, utilizing the most efficient analytical tools may lead to better personalization of therapy. The rise of artificial intelligence and machine learning has allowed large amounts of data to be rapidly accessed and analyzed. These unsupervised learning models can apprehend the data structure and identify homogeneous subgroups, facilitating the individualization of medical interventions. This review aims to discuss the challenges of β-lactam dosing, focusing on its pharmacodynamics and the new challenges and opportunities arising from integrating machine learning algorithms to personalize patient treatment.
Collapse
Affiliation(s)
- João Gonçalves Pereira
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
- Serviço de Medicina Intensiva, Hospital Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal
| | - Joana Fernandes
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Serviço de Medicina Intensiva, Centro Hospitalar de Trás-os-Montes e Alto Douro, 5000-508 Vila Real, Portugal
| | - Tânia Mendes
- Serviço de Medicina Interna, Hospital Vila Franca de Xira, 2600-009 Vila Franca de Xira, Portugal
| | - Filipe André Gonzalez
- Serviço de Medicina Intensiva, Hospital Garcia De Orta, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| | - Susana M Fernandes
- Grupo de Investigação e Desenvolvimento em Infeção e Sépsis, Serviço de Medicina Intensiva, Hospital Santa Maria, Clínica Universitária de Medicina Intensiva, Faculdade de Medicina, Universidade de Lisboa, 1649-004 Lisbon, Portugal
| |
Collapse
|
6
|
d'Astous ÉV, Dauphin-Ducharme P. Whole blood multiplex measurements using electrochemical aptamer-based biosensors. Chem Commun (Camb) 2024; 60:6419-6422. [PMID: 38828657 DOI: 10.1039/d4cc01452a] [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: 06/05/2024]
Abstract
Simultaneous measurements of various molecules ("multiplex") using electrochemical biosensors typically require multiple electrode implementations, which for neonates, hemophiliacs, etc. is problematic. Here, we introduce the oxazine ATTO 700 into electrochemical aptamer-based biosensors to achieve "true" multiplex, continuous and real-time measurements of two different molecules in undiluted whole blood using a single electrode.
Collapse
Affiliation(s)
- Élodie V d'Astous
- Université de Sherbrooke, Département de Chimie, 2500 boul. de l'Université, Sherbrooke, J1K 2R1, Canada.
| | - Philippe Dauphin-Ducharme
- Université de Sherbrooke, Département de Chimie, 2500 boul. de l'Université, Sherbrooke, J1K 2R1, Canada.
| |
Collapse
|
7
|
Oharazawa A, Maimaituxun G, Watanabe K, Nishiyasu T, Fujii N. Metabolome analyses of skin dialysate: Insights into skin interstitial fluid biomarkers. J Dermatol Sci 2024; 114:141-147. [PMID: 38740531 DOI: 10.1016/j.jdermsci.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 03/27/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024]
Abstract
BACKGROUND Metabolites in biofluids can serve as biomarkers for diagnosing diseases and monitoring body conditions. Among the available biofluids, interstitial fluid (ISF) in the skin has garnered considerable attention owing to its advantages, which include inability to clot, easy access to the skin, and possibility of incorporating wearable devices. However, the scientific understanding of skin ISF composition is limited. OBJECTIVE In this study, we aimed to compare metabolites between skin dialysate containing metabolites from the skin ISF and venous blood (plasma) samples, both collected under resting states. METHODS We collected forearm skin dialysate using intradermal microdialysis alongside venous blood (plasma) samples from 12 healthy young adults. We analyzed these samples using capillary electrophoresis-fourier transform mass spectrometry-based metabolomics (CE-FTMS). RESULTS Significant positive correlations were observed in 39 metabolites between the skin dialysate and plasma, including creatine (a mitochondrial disease biomarker), 1-methyladenosine (an early detection of cancer biomarker), and trimethylamine N-oxide (a posterior predictor of heart failure biomarker). Based on the Human Metabolome Technologies database, we identified 12 metabolites unique to forearm skin dialysate including nucleic acids, benzoate acids, fatty acids, amino acids, ascorbic acid, 3-methoxy-4-hydroxyphenylethyleneglycol (an Alzheimer's disease biomarker), and cysteic acid (an acute myocardial infarction biomarker). CONCLUSION We show that some venous blood biomarkers may be predicted from skin dialysate or skin ISF, and that these fluids may serve as diagnostic and monitoring tools for health and clinical conditions.
Collapse
Affiliation(s)
| | - Gulinu Maimaituxun
- Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan
| | - Koichi Watanabe
- Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan
| | - Takeshi Nishiyasu
- Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan; Advanced Research Initiative for Human High Performance (ARIHHP), Japan
| | - Naoto Fujii
- Institute of Health and Sport Sciences, University of Tsukuba, Tsukuba, Japan; Advanced Research Initiative for Human High Performance (ARIHHP), Japan.
| |
Collapse
|
8
|
Wang X, Zhang Z, Li P, Xu J, Zheng Y, Sun W, Xie M, Wang J, Pan X, Lei X, Wang J, Chen J, Chen Y, Wang SJ, Lei T. Ultrastable N-Type Semiconducting Fiber Organic Electrochemical Transistors for Highly Sensitive Biosensors. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2400287. [PMID: 38433667 DOI: 10.1002/adma.202400287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/27/2024] [Indexed: 03/05/2024]
Abstract
Organic electrochemical transistors (OECTs) have attracted increasing attention due to their merits of high transconductance, low operating voltage, and good biocompatibility, ideal for biosensors. However, further advances in their practical applications face challenges of low n-type performance and poor stability. Here, it is demonstrated that wet-spinning the commercially available n-type conjugated polymer poly(benzimidazobenzophenanthroline) (BBL) into highly aligned and crystalline fibers enhances both OECT performance and stability. Although BBL is only soluble in high-boiling-point strong acids, it can be wet-spun into high-quality fibers with adjustable diameters. The BBL fiber OECTs exhibit a record-high area-normalized transconductance (gm,A) of 2.40 µS µm-2 and over 10 times higher figure-of-merit (µC*) than its thin-film counterparts. More importantly, these fiber OECTs exhibit remarkable stability with no noticeable performance attenuation after 1500 cycles over 4 h operation, outperforming all previously reported n-type OECTs. The superior performance and stability can be attributed to shorter π-π stacking distance and ordered molecular arrangement in the fibers, endowing the BBL fiber OECT-based biosensors with outstanding sensitivity while keeping a miniaturized form factor. This work demonstrates that, beyond new material development, developing new fabrication technology is also crucial for addressing the performance and stability issues in n-type OECTs.
Collapse
Affiliation(s)
- Xiu Wang
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Zhi Zhang
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Peiyun Li
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Jingcao Xu
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Yuting Zheng
- College of Engineering, Peking University, Beijing, 100871, P. R. China
| | - Wenxi Sun
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Mingyue Xie
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Juanrong Wang
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Xiran Pan
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Xun Lei
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Jingyi Wang
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Jupeng Chen
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Yiheng Chen
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| | - Shu-Jen Wang
- Department of Physics, Hong Kong Baptist University, Hong Kong, SAR, P. R. China
| | - Ting Lei
- Key Laboratory of Polymer Chemistry and Physics of Ministry of Education, School of Materials Science and Engineering, Peking University, Beijing, 100871, P. R. China
| |
Collapse
|
9
|
Xie Y, He J, He W, Iftikhar T, Zhang C, Su L, Zhang X. Enhanced Interstitial Fluid Extraction and Rapid Analysis via Vacuum Tube-Integrated Microneedle Array Device. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308716. [PMID: 38502884 DOI: 10.1002/advs.202308716] [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: 11/14/2023] [Revised: 02/06/2024] [Indexed: 03/21/2024]
Abstract
Advancing the development of point-of-care testing (POCT) sensors that utilize interstitial fluid (ISF) presents considerable obstacles in terms of rapid sampling and analysis. Herein, an innovative strategy is introduced that involves the use of a 3D-printed, hollow microneedle array patch (MAP), in tandem with a vacuum tube (VT) connected through a hose, to improve ISF extraction efficiency and facilitate expedited analysis. The employment of negative pressure by the VT allows the MAP device to effectively gather ≈18 µL of ISF from the dermis of a live rabbit ear within a concise period of 5 min. This methodology enables the immediate and minimally invasive measurement of glucose levels within the body, employing personal healthcare meters for quantification. The fusion of the VT and MAP technologies provides for their effortless integration into a comprehensive and mobile system for ISF analysis, accomplished by preloading the hose with custom sensing papers designed to detect specific analytes. Moreover, the design and functionality of this integrated VT-MAP system are intuitively user-friendly, eliminating the requirement for specialized medical expertise. This feature enhances its potential to make a significant impact on the field of decentralized personal healthcare.
Collapse
Affiliation(s)
- Yuanting Xie
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
- Shenzhen Key Laboratory of Nano-Biosensing Technology, Marshall Laboratory of Biomedical Engineering, International Health Science Innovation Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China
| | - Jinhua He
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Wenqing He
- Guangdong Laboratory of Artificial Intelligence and Digital Economy (SZ), Shenzhen University, Shenzhen, 518060, China
| | - Tayyaba Iftikhar
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Chuangjie Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Lei Su
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
- Shenzhen Key Laboratory of Nano-Biosensing Technology, Marshall Laboratory of Biomedical Engineering, International Health Science Innovation Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China
| | - Xueji Zhang
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
- Shenzhen Key Laboratory of Nano-Biosensing Technology, Marshall Laboratory of Biomedical Engineering, International Health Science Innovation Center, Shenzhen University Medical School, Shenzhen University, Shenzhen, 518060, China
| |
Collapse
|
10
|
Lu B, Lunn J, Nightingale AM, Niu X. Highly sensitive absorbance measurement using droplet microfluidics integrated with an oil extraction and long pathlength detection flow cell. Front Chem 2024; 12:1394388. [PMID: 38803381 PMCID: PMC11129082 DOI: 10.3389/fchem.2024.1394388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 04/16/2024] [Indexed: 05/29/2024] Open
Abstract
In droplet microfluidics, UV-Vis absorption spectroscopy along with colorimetric assays have been widely used for chemical and biochemical analysis. However, the sensitivity of the measurement can be limited by the short optical pathlength. Here we report a novel design to enhance the sensitivity by removing oil and converting the droplets into a single-phase aqueous flow, which can be measured within a U-shape channel with long optical pathlength. The flow cells were fabricated via 3D printing. The calibration results have demonstrated complete oil removal and effective optical pathlengths similar to the designed channel lengths (from 5 to 20 mm). The flow cell was further employed in a droplet microfluidic-based phosphate sensing system. The measured phosphate levels displayed excellent consistency with data obtained from traditional UV spectroscopy analysis. This flow cell design overcomes the limitations of short optical pathlengths in droplet microfluidics and has the potential to be used for in situ and continuous monitoring.
Collapse
Affiliation(s)
| | | | | | - Xize Niu
- Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
11
|
Zhao X, Kolbinger FR, Distler M, Weitz J, Makarov D, Bachmann M, Baraban L. Portable droplet-based real-time monitoring of pancreatic α-amylase in postoperative patients. Biosens Bioelectron 2024; 251:116034. [PMID: 38359666 DOI: 10.1016/j.bios.2024.116034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/21/2023] [Accepted: 01/11/2024] [Indexed: 02/17/2024]
Abstract
Postoperative complications after pancreatic surgery are frequent and can be life-threatening. Current clinical diagnostic strategies involve time-consuming quantification of α-amylase activity in abdominal drain fluid, which is performed on the first and third postoperative day. The lack of real-time monitoring may delay adjustment of medical treatment upon complications and worsen prognosis for patients. We report a bedside portable droplet-based millifluidic device enabling real-time sensing of drain α-amylase activity for postoperative monitoring of patients undergoing pancreatic surgery. Here, a tiny amount of drain liquid of patient samples is continuously collected and co-encapsulated with a starch reagent in nanoliter-sized droplets to track the fluorescence intensity released upon reaction with α-amylase. Comparing the α-amylase levels of 32 patients, 97 % of the results of the droplet-based millifluidic system matched the clinical data. Our method reduces the α-amylase assay duration to approximately 3 min with the limit of detection 7 nmol/s·L, enabling amylase activity monitoring at the bedside in clinical real-time. The presented droplet-based platform can be extended for analysis of different body fluids, diseases, and towards a broader range of biomarkers, including lipase, bilirubin, lactate, inflammation, or liquid biopsy markers, paving the way towards new standards in postoperative patient monitoring.
Collapse
Affiliation(s)
- Xinne Zhao
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V, 01328, Dresden, Germany.
| | - Fiona R Kolbinger
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav. Carus, TUD Dresden University of Technology, Germany; Else Kröner Fresenius Center for Digital Health (EKFZ), TUD Dresden University of Technology, Germany.
| | - Marius Distler
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav. Carus, TUD Dresden University of Technology, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, University Hospital and Faculty of Medicine Carl Gustav. Carus, TUD Dresden University of Technology, Germany; Else Kröner Fresenius Center for Digital Health (EKFZ), TUD Dresden University of Technology, Germany
| | - Denys Makarov
- Institute of Ion Beam Physics and Materials Research, Helmholtz-Zentrum Dresden-Rossendorf e. V, 01328, Dresden, Germany.
| | - Michael Bachmann
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V, 01328, Dresden, Germany.
| | - Larysa Baraban
- Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf e. V, 01328, Dresden, Germany; Else Kröner Fresenius Center for Digital Health (EKFZ), TUD Dresden University of Technology, Germany.
| |
Collapse
|
12
|
Zhou J, Dong J, Hou H, Huang L, Li J. High-throughput microfluidic systems accelerated by artificial intelligence for biomedical applications. LAB ON A CHIP 2024; 24:1307-1326. [PMID: 38247405 DOI: 10.1039/d3lc01012k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
High-throughput microfluidic systems are widely used in biomedical fields for tasks like disease detection, drug testing, and material discovery. Despite the great advances in automation and throughput, the large amounts of data generated by the high-throughput microfluidic systems generally outpace the abilities of manual analysis. Recently, the convergence of microfluidic systems and artificial intelligence (AI) has been promising in solving the issue by significantly accelerating the process of data analysis as well as improving the capability of intelligent decision. This review offers a comprehensive introduction on AI methods and outlines the current advances of high-throughput microfluidic systems accelerated by AI, covering biomedical detection, drug screening, and automated system control and design. Furthermore, the challenges and opportunities in this field are critically discussed as well.
Collapse
Affiliation(s)
- Jianhua Zhou
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Jianpei Dong
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Hongwei Hou
- Beijing Life Science Academy, Beijing 102209, China
| | - Lu Huang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China.
- Key Laboratory of Sensing Technology and Biomedical Instruments of Guangdong Province, School of Biomedical Engineering, Sun Yat-sen University, Shenzhen 518107, China
| | - Jinghong Li
- Department of Chemistry, Center for BioAnalytical Chemistry, Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Tsinghua University, Beijing 100084, China.
- New Cornerstone Science Laboratory, Shenzhen 518054, China
- Beijing Life Science Academy, Beijing 102209, China
- Center for BioAnalytical Chemistry, Hefei National Laboratory of Physical Science at Microscale, University of Science and Technology of China, Hefei 230026, China
| |
Collapse
|
13
|
Saha T, Mukherjee S, Dickey MD, Velev OD. Harvesting and manipulating sweat and interstitial fluid in microfluidic devices. LAB ON A CHIP 2024; 24:1244-1265. [PMID: 38197332 DOI: 10.1039/d3lc00874f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2024]
Abstract
Microfluidic devices began to be used to facilitate sweat and interstitial fluid (ISF) sensing in the mid-2010s. Since then, numerous prototypes involving microfluidics have been developed in different form factors for sensing biomarkers found in these fluids under in vitro, ex vivo, and in vivo (on-body) settings. These devices transport and manipulate biofluids using microfluidic channels composed of silicone, polymer, paper, or fiber. Fluid flow transport and sample management can be achieved by controlling the flow rate, surface morphology of the channel, and rate of fluid evaporation. Although many devices have been developed for estimating sweat rate, electrolyte, and metabolite levels, only a handful have been able to proceed beyond laboratory testing and reach the stage of clinical trials and commercialization. To further this technology, this review reports on the utilization of microfluidics towards sweat and ISF management and transport. The review is distinguished from other recent reviews by focusing on microfluidic principles of sweat and ISF generation, transport, extraction, and management. Challenges and prospects are highlighted, with a discussion on how to transition such prototypes towards personalized healthcare monitoring systems.
Collapse
Affiliation(s)
- Tamoghna Saha
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA.
| | - Sneha Mukherjee
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA.
| | - Michael D Dickey
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA.
| | - Orlin D Velev
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, NC 27695, USA.
| |
Collapse
|
14
|
Pan D, Hu J, Wang B, Xia X, Cheng Y, Wang C, Lu Y. Biomimetic Wearable Sensors: Emerging Combination of Intelligence and Electronics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2303264. [PMID: 38044298 PMCID: PMC10837381 DOI: 10.1002/advs.202303264] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 10/03/2023] [Indexed: 12/05/2023]
Abstract
Owing to the advancement of interdisciplinary concepts, for example, wearable electronics, bioelectronics, and intelligent sensing, during the microelectronics industrial revolution, nowadays, extensively mature wearable sensing devices have become new favorites in the noninvasive human healthcare industry. The combination of wearable sensing devices with bionics is driving frontier developments in various fields, such as personalized medical monitoring and flexible electronics, due to the superior biocompatibilities and diverse sensing mechanisms. It is noticed that the integration of desired functions into wearable device materials can be realized by grafting biomimetic intelligence. Therefore, herein, the mechanism by which biomimetic materials satisfy and further enhance system functionality is reviewed. Next, wearable artificial sensory systems that integrate biomimetic sensing into portable sensing devices are introduced, which have received significant attention from the industry owing to their novel sensing approaches and portabilities. To address the limitations encountered by important signal and data units in biomimetic wearable sensing systems, two paths forward are identified and current challenges and opportunities are presented in this field. In summary, this review provides a further comprehensive understanding of the development of biomimetic wearable sensing devices from both breadth and depth perspectives, offering valuable guidance for future research and application expansion of these devices.
Collapse
Affiliation(s)
- Donglei Pan
- College of Light Industry and Food EngineeringGuangxi UniversityNanningGuangxi530004China
- Key Laboratory of Industrial BiocatalysisMinistry of EducationDepartment of Chemical EngineeringTsinghua UniversityBeijing100084China
| | - Jiawang Hu
- Key Laboratory of Industrial BiocatalysisMinistry of EducationDepartment of Chemical EngineeringTsinghua UniversityBeijing100084China
| | - Bin Wang
- Key Laboratory of Industrial BiocatalysisMinistry of EducationDepartment of Chemical EngineeringTsinghua UniversityBeijing100084China
| | - Xuanjie Xia
- Key Laboratory of Industrial BiocatalysisMinistry of EducationDepartment of Chemical EngineeringTsinghua UniversityBeijing100084China
| | - Yifan Cheng
- Key Laboratory of Industrial BiocatalysisMinistry of EducationDepartment of Chemical EngineeringTsinghua UniversityBeijing100084China
| | - Cheng‐Hua Wang
- College of Light Industry and Food EngineeringGuangxi UniversityNanningGuangxi530004China
| | - Yuan Lu
- Key Laboratory of Industrial BiocatalysisMinistry of EducationDepartment of Chemical EngineeringTsinghua UniversityBeijing100084China
| |
Collapse
|
15
|
Lou C, Yang H, Hou Y, Huang H, Qiu J, Wang C, Sang Y, Liu H, Han L. Microfluidic Platforms for Real-Time In Situ Monitoring of Biomarkers for Cellular Processes. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307051. [PMID: 37844125 DOI: 10.1002/adma.202307051] [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: 07/17/2023] [Revised: 09/05/2023] [Indexed: 10/18/2023]
Abstract
Cellular processes are mechanisms carried out at the cellular level that are aimed at guaranteeing the stability of the organism they comprise. The investigation of cellular processes is key to understanding cell fate, understanding pathogenic mechanisms, and developing new therapeutic technologies. Microfluidic platforms are thought to be the most powerful tools among all methodologies for investigating cellular processes because they can integrate almost all types of the existing intracellular and extracellular biomarker-sensing methods and observation approaches for cell behavior, combined with precisely controlled cell culture, manipulation, stimulation, and analysis. Most importantly, microfluidic platforms can realize real-time in situ detection of secreted proteins, exosomes, and other biomarkers produced during cell physiological processes, thereby providing the possibility to draw the whole picture for a cellular process. Owing to their advantages of high throughput, low sample consumption, and precise cell control, microfluidic platforms with real-time in situ monitoring characteristics are widely being used in cell analysis, disease diagnosis, pharmaceutical research, and biological production. This review focuses on the basic concepts, recent progress, and application prospects of microfluidic platforms for real-time in situ monitoring of biomarkers in cellular processes.
Collapse
Affiliation(s)
- Chengming Lou
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Hongru Yang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Ying Hou
- Institute for Advanced Interdisciplinary Research (IAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Haina Huang
- Institute for Advanced Interdisciplinary Research (IAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Jichuan Qiu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Chunhua Wang
- Institute for Advanced Interdisciplinary Research (IAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Yuanhua Sang
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
| | - Hong Liu
- State Key Laboratory of Crystal Materials, Shandong University, Jinan, 250100, P. R. China
- Institute for Advanced Interdisciplinary Research (IAIR), University of Jinan, Jinan, 250022, P. R. China
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, Shandong, 266000, P. R. China
| |
Collapse
|
16
|
Friedel M, Thompson IAP, Kasting G, Polsky R, Cunningham D, Soh HT, Heikenfeld J. Opportunities and challenges in the diagnostic utility of dermal interstitial fluid. Nat Biomed Eng 2023; 7:1541-1555. [PMID: 36658344 DOI: 10.1038/s41551-022-00998-9] [Citation(s) in RCA: 73] [Impact Index Per Article: 36.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 12/06/2022] [Indexed: 01/21/2023]
Abstract
The volume of interstitial fluid (ISF) in the human body is three times that of blood. Yet, collecting diagnostically useful ISF is more challenging than collecting blood because the extraction of dermal ISF disrupts the delicate balance of pressure between ISF, blood and lymph, and because the triggered local inflammation further skews the concentrations of many analytes in the extracted fluid. In this Perspective, we overview the most meaningful differences in the make-up of ISF and blood, and discuss why ISF cannot be viewed generally as a diagnostically useful proxy for blood. We also argue that continuous sensing of small-molecule analytes in dermal ISF via rapid assays compatible with nanolitre sample volumes or via miniaturized sensors inserted into the dermis can offer clinically advantageous utility, particularly for the monitoring of therapeutic drugs and of the status of the immune system.
Collapse
Affiliation(s)
- Mark Friedel
- Novel Device Laboratory, Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA
| | - Ian A P Thompson
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Gerald Kasting
- The James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, Cincinnati, OH, USA
| | - Ronen Polsky
- Nano and Micro Sensors, Sandia National Laboratories, Albuquerque, NM, USA
| | - David Cunningham
- Department of Chemistry and Physics, Southeast Missouri State University, Cape Girardeau, MO, USA
| | - Hyongsok Tom Soh
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA.
- Department of Radiology, Stanford University, Stanford, CA, USA.
| | - Jason Heikenfeld
- Novel Device Laboratory, Department of Biomedical Engineering, University of Cincinnati, Cincinnati, OH, USA.
| |
Collapse
|
17
|
Lathia R, Nagpal S, Modak CD, Mishra S, Sharma D, Reddy BS, Nukala P, Bhat R, Sen P. Tunable encapsulation of sessile droplets with solid and liquid shells. Nat Commun 2023; 14:6445. [PMID: 37833273 PMCID: PMC10575970 DOI: 10.1038/s41467-023-41977-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 09/22/2023] [Indexed: 10/15/2023] Open
Abstract
Droplet encapsulations using liquid or solid shells are of significant interest in microreactors, drug delivery, crystallization, and cell growth applications. Despite progress in droplet-related technologies, tuning micron-scale shell thickness over a large range of droplet sizes is still a major challenge. In this work, we report capillary force assisted cloaking using hydrophobic colloidal particles and liquid-infused surfaces. The technique produces uniform solid and liquid shell encapsulations over a broad range (5-200 μm shell thickness for droplet volume spanning over four orders of magnitude). Tunable liquid encapsulation is shown to reduce the evaporation rate of droplets by up to 200 times with a wide tunability in lifetime (1.5 h to 12 days). Further, we propose using the technique for single crystals and cell/spheroid culture platforms. Stimuli-responsive solid shells show hermetic encapsulation with tunable strength and dissolution time. Moreover, scalability, and versatility of the technique is demonstrated for on-chip applications.
Collapse
Affiliation(s)
- Rutvik Lathia
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Satchit Nagpal
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Chandantaru Dey Modak
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Satyarthi Mishra
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Deepak Sharma
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Bheema Sankar Reddy
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Pavan Nukala
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Ramray Bhat
- Department of Developmental Biology and Genetics, Indian Institute of Science, Bangalore, 560012, India
- Department of BioSystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India
| | - Prosenjit Sen
- Centre for Nano Science and Engineering, Indian Institute of Science, Bangalore, 560012, India.
- Department of BioSystems Science and Engineering, Indian Institute of Science, Bangalore, 560012, India.
| |
Collapse
|
18
|
Hu X, Abbasi R, Wachsmann-Hogiu S. Microfluidics on lensless, semiconductor optical image sensors: challenges and opportunities for democratization of biosensing at the micro-and nano-scale. NANOPHOTONICS (BERLIN, GERMANY) 2023; 12:3977-4008. [PMID: 39635640 PMCID: PMC11501743 DOI: 10.1515/nanoph-2023-0301] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 09/29/2023] [Indexed: 12/07/2024]
Abstract
Optical image sensors are 2D arrays of pixels that integrate semiconductor photodiodes and field effect transistors for efficient photon conversion and processing of generated electrons. With technological advancements and subsequent democratization of these sensors, opportunities for integration with microfluidics devices are currently explored. 2D pixel arrays of such optical image sensors can reach dimensions larger than one centimeter with a sub-micrometer pixel size, for high spatial resolution lensless imaging with large field of view, a feat that cannot be achieved with lens-based optical microscopy. Moreover, with advancements in fabrication processes, the field of microfluidics has evolved to develop microfluidic devices with an overall size below one centimeter and individual components of sub-micrometer size, such that they can now be implemented onto optical image sensors. The convergence of these fields is discussed in this article, where we review fundamental principles, opportunities, challenges, and outlook for integration, with focus on contact-mode imaging configuration. Most recent developments and applications of microfluidic lensless contact-based imaging to the field of biosensors, in particular those related to the potential for point of need applications, are also discussed.
Collapse
Affiliation(s)
- Xinyue Hu
- Department of Bioengineering, McGill University, Montreal, QC H3A 0C3, Canada
| | - Reza Abbasi
- Department of Bioengineering, McGill University, Montreal, QC H3A 0C3, Canada
| | | |
Collapse
|
19
|
Yao Y, Huang W, Chen J, Liu X, Bai L, Chen W, Cheng Y, Ping J, Marks TJ, Facchetti A. Flexible and Stretchable Organic Electrochemical Transistors for Physiological Sensing Devices. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209906. [PMID: 36808773 DOI: 10.1002/adma.202209906] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 01/31/2023] [Indexed: 06/18/2023]
Abstract
Flexible and stretchable bioelectronics provides a biocompatible interface between electronics and biological systems and has received tremendous attention for in situ monitoring of various biological systems. Considerable progress in organic electronics has made organic semiconductors, as well as other organic electronic materials, ideal candidates for developing wearable, implantable, and biocompatible electronic circuits due to their potential mechanical compliance and biocompatibility. Organic electrochemical transistors (OECTs), as an emerging class of organic electronic building blocks, exhibit significant advantages in biological sensing due to the ionic nature at the basis of the switching behavior, low driving voltage (<1 V), and high transconductance (in millisiemens range). During the past few years, significant progress in constructing flexible/stretchable OECTs (FSOECTs) for both biochemical and bioelectrical sensors has been reported. In this regard, to summarize major research accomplishments in this emerging field, this review first discusses structure and critical features of FSOECTs, including working principles, materials, and architectural engineering. Next, a wide spectrum of relevant physiological sensing applications, where FSOECTs are the key components, are summarized. Last, major challenges and opportunities for further advancing FSOECT physiological sensors are discussed.
Collapse
Affiliation(s)
- Yao Yao
- School of Biosystems Engineering and Food Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, P. R. China
- Innovation Platform of Micro/Nano Technology for Biosensing, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311200, P. R. China
- Department of Chemistry and the Materials Research Center, Northwestern University, Sheridan Road, Evanston, IL, 60208, USA
| | - Wei Huang
- Department of Chemistry and the Materials Research Center, Northwestern University, Sheridan Road, Evanston, IL, 60208, USA
- School of Automation Engineering, University of Electronic Science and Technology of China (UESTC), Chengdu, Sichuan, 611731, P. R. China
| | - Jianhua Chen
- Department of Chemistry and the Materials Research Center, Northwestern University, Sheridan Road, Evanston, IL, 60208, USA
| | - Xiaoxue Liu
- School of Biosystems Engineering and Food Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, P. R. China
- Innovation Platform of Micro/Nano Technology for Biosensing, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311200, P. R. China
| | - Libing Bai
- School of Automation Engineering, University of Electronic Science and Technology of China (UESTC), Chengdu, Sichuan, 611731, P. R. China
| | - Wei Chen
- School of Biosystems Engineering and Food Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, P. R. China
| | - Yuhua Cheng
- School of Automation Engineering, University of Electronic Science and Technology of China (UESTC), Chengdu, Sichuan, 611731, P. R. China
| | - Jianfeng Ping
- School of Biosystems Engineering and Food Science, Zhejiang University, 866 Yuhangtang Road, Hangzhou, 310058, P. R. China
- Innovation Platform of Micro/Nano Technology for Biosensing, ZJU-Hangzhou Global Scientific and Technological Innovation Center, Hangzhou, 311200, P. R. China
| | - Tobin J Marks
- Department of Chemistry and the Materials Research Center, Northwestern University, Sheridan Road, Evanston, IL, 60208, USA
| | - Antonio Facchetti
- Department of Chemistry and the Materials Research Center, Northwestern University, Sheridan Road, Evanston, IL, 60208, USA
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, Norrköping, 60174, Sweden
| |
Collapse
|
20
|
Yang J, Luo R, Yang L, Wang X, Huang Y. Microneedle-Integrated Sensors for Extraction of Skin Interstitial Fluid and Metabolic Analysis. Int J Mol Sci 2023; 24:9882. [PMID: 37373027 DOI: 10.3390/ijms24129882] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/02/2023] [Accepted: 06/06/2023] [Indexed: 06/29/2023] Open
Abstract
Skin interstitial fluid (ISF) has emerged as a fungible biofluid sample for blood serum and plasma for disease diagnosis and therapy. The sampling of skin ISF is highly desirable considering its easy accessibility, no damage to blood vessels, and reduced risk of infection. Particularly, skin ISF can be sampled using microneedle (MN)-based platforms in the skin tissues, which exhibit multiple advantages including minimal invasion of the skin tissues, less pain, ease of carrying, capacity for continuous monitoring, etc. In this review, we focus on the current development of microneedle-integrated transdermal sensors for collecting ISF and detecting specific disease biomarkers. Firstly, we discussed and classified microneedles according to their structural design, including solid MNs, hollow MNs, porous MNs, and coated MNs. Subsequently, we elaborate on the construction of MN-integrated sensors for metabolic analysis with highlights on the electrochemical, fluorescent, chemical chromogenic, immunodiagnostic, and molecular diagnostic MN-integrated sensors. Finally, we discuss the current challenges and future direction for developing MN-based platforms for ISF extraction and sensing applications.
Collapse
Affiliation(s)
- Jie Yang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Ruiyu Luo
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| | - Lei Yang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Xiaocheng Wang
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health), Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Yong Huang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
21
|
Fernandes J, Karra N, Swindle EJ, Morgan H. Droplet fluidics for time-dependent analysis of barrier permeability in an epithelial barrier on chip system. RSC Adv 2023; 13:14494-14500. [PMID: 37179995 PMCID: PMC10173818 DOI: 10.1039/d3ra00470h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
A droplet generator has been developed that interfaces with a barrier-on-chip platform for temporal analyte compartmentalisation and analysis. Droplets are generated every 20 minutes in 8 separate parallel microchannels, with an average droplet volume of 9.47 ± 0.6 μL, allowing simultaneous analysis of 8 different experiments. The device was tested using an epithelial barrier model by monitoring the diffusion of a fluorescent high molecular weight dextran molecule. The epithelial barrier was perturbed using detergent leading to a peak at 3-4 hours, correlating with simulations. For the untreated (control) a constant, very low level of dextran diffusion was observed. The epithelial cell barrier properties were also continuously measured using electrical impedance spectroscopy to extract an equivalent trans epithelial resistance.
Collapse
Affiliation(s)
- Joao Fernandes
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton UK
| | - Nikita Karra
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton UK
| | - Emily J Swindle
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton UK
- Institute for Life Sciences, University of Southampton UK
- NIHR Southampton Biomedical Research Centre, University Hospital Southampton UK
| | - Hywel Morgan
- Electronics and Computer Science, Faculty of Physical Sciences and Engineering, University of Southampton UK
- Institute for Life Sciences, University of Southampton UK
| |
Collapse
|
22
|
Mintz Hemed N, Leal-Ortiz S, Zhao ET, Melosh NA. On-Demand, Reversible, Ultrasensitive Polymer Membrane Based on Molecular Imprinting Polymer. ACS NANO 2023; 17:5632-5643. [PMID: 36913954 PMCID: PMC10062346 DOI: 10.1021/acsnano.2c11618] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 03/08/2023] [Indexed: 06/18/2023]
Abstract
The development of in vivo, longitudinal, real-time monitoring devices is an essential step toward continuous, precision health monitoring. Molecularly imprinted polymers (MIPs) are popular sensor capture agents that are more robust than antibodies and have been used for sensors, drug delivery, affinity separations, assays, and solid-phase extraction. However, MIP sensors are typically limited to one-time use due to their high binding affinity (>107 M-1) and slow-release kinetics (<10-4 μM/sec). To overcome this challenge, current research has focused on stimuli-responsive MIPs (SR-MIPs), which undergo a conformational change induced by external stimuli to reverse molecular binding, requiring additional chemicals or outside stimuli. Here, we demonstrate fully reversible MIP sensors based on electrostatic repulsion. Once the target analyte is bound within a thin film MIP on an electrode, a small electrical potential successfully releases the bound molecules, enabling repeated, accurate measurements. We demonstrate an electrostatically refreshed dopamine sensor with a 760 pM limit of detection, linear response profile, and accuracy even after 30 sensing-release cycles. These sensors could repeatedly detect <1 nM dopamine released from PC-12 cells in vitro, demonstrating they can longitudinally measure low concentrations in complex biological environments without clogging. Our work provides a simple and effective strategy for enhancing the use of MIPs-based biosensors for all charged molecules in continuous, real-time health monitoring and other sensing applications.
Collapse
Affiliation(s)
- Nofar Mintz Hemed
- Department
of Materials Science and Engineering, Stanford
University, Stanford, California 94305, United States
| | - Sergio Leal-Ortiz
- Department
of Psychiatry and Behavioral Sciences, Stanford
University, Stanford, California 94304, United States
| | - Eric T. Zhao
- Department
of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Nicholas A. Melosh
- Department
of Materials Science and Engineering, Stanford
University, Stanford, California 94305, United States
| |
Collapse
|
23
|
Luo Y, Abidian MR, Ahn JH, Akinwande D, Andrews AM, Antonietti M, Bao Z, Berggren M, Berkey CA, Bettinger CJ, Chen J, Chen P, Cheng W, Cheng X, Choi SJ, Chortos A, Dagdeviren C, Dauskardt RH, Di CA, Dickey MD, Duan X, Facchetti A, Fan Z, Fang Y, Feng J, Feng X, Gao H, Gao W, Gong X, Guo CF, Guo X, Hartel MC, He Z, Ho JS, Hu Y, Huang Q, Huang Y, Huo F, Hussain MM, Javey A, Jeong U, Jiang C, Jiang X, Kang J, Karnaushenko D, Khademhosseini A, Kim DH, Kim ID, Kireev D, Kong L, Lee C, Lee NE, Lee PS, Lee TW, Li F, Li J, Liang C, Lim CT, Lin Y, Lipomi DJ, Liu J, Liu K, Liu N, Liu R, Liu Y, Liu Y, Liu Z, Liu Z, Loh XJ, Lu N, Lv Z, Magdassi S, Malliaras GG, Matsuhisa N, Nathan A, Niu S, Pan J, Pang C, Pei Q, Peng H, Qi D, Ren H, Rogers JA, Rowe A, Schmidt OG, Sekitani T, Seo DG, Shen G, Sheng X, Shi Q, Someya T, Song Y, Stavrinidou E, Su M, Sun X, Takei K, Tao XM, Tee BCK, Thean AVY, Trung TQ, Wan C, Wang H, Wang J, Wang M, Wang S, Wang T, Wang ZL, Weiss PS, Wen H, Xu S, Xu T, Yan H, Yan X, Yang H, Yang L, Yang S, Yin L, Yu C, Yu G, Yu J, Yu SH, Yu X, Zamburg E, Zhang H, Zhang X, Zhang X, Zhang X, Zhang Y, Zhang Y, Zhao S, Zhao X, Zheng Y, Zheng YQ, Zheng Z, Zhou T, Zhu B, Zhu M, Zhu R, Zhu Y, Zhu Y, Zou G, Chen X. Technology Roadmap for Flexible Sensors. ACS NANO 2023; 17:5211-5295. [PMID: 36892156 PMCID: PMC11223676 DOI: 10.1021/acsnano.2c12606] [Citation(s) in RCA: 249] [Impact Index Per Article: 124.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Humans rely increasingly on sensors to address grand challenges and to improve quality of life in the era of digitalization and big data. For ubiquitous sensing, flexible sensors are developed to overcome the limitations of conventional rigid counterparts. Despite rapid advancement in bench-side research over the last decade, the market adoption of flexible sensors remains limited. To ease and to expedite their deployment, here, we identify bottlenecks hindering the maturation of flexible sensors and propose promising solutions. We first analyze challenges in achieving satisfactory sensing performance for real-world applications and then summarize issues in compatible sensor-biology interfaces, followed by brief discussions on powering and connecting sensor networks. Issues en route to commercialization and for sustainable growth of the sector are also analyzed, highlighting environmental concerns and emphasizing nontechnical issues such as business, regulatory, and ethical considerations. Additionally, we look at future intelligent flexible sensors. In proposing a comprehensive roadmap, we hope to steer research efforts towards common goals and to guide coordinated development strategies from disparate communities. Through such collaborative efforts, scientific breakthroughs can be made sooner and capitalized for the betterment of humanity.
Collapse
Affiliation(s)
- Yifei Luo
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
- Innovative Centre for Flexible Devices (iFLEX), School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Mohammad Reza Abidian
- Department of Biomedical Engineering, University of Houston, Houston, Texas 77024, United States
| | - Jong-Hyun Ahn
- School of Electrical and Electronic Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Deji Akinwande
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Microelectronics Research Center, The University of Texas at Austin, Austin, Texas 78758, United States
| | - Anne M Andrews
- Department of Chemistry and Biochemistry, California NanoSystems Institute, and Department of Psychiatry and Biobehavioral Sciences, Semel Institute for Neuroscience and Human Behavior, and Hatos Center for Neuropharmacology, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Markus Antonietti
- Colloid Chemistry Department, Max Planck Institute of Colloids and Interfaces, 14476 Potsdam, Germany
| | - Zhenan Bao
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Magnus Berggren
- Laboratory of Organic Electronics, Department of Science and Technology, Campus Norrköping, Linköping University, 83 Linköping, Sweden
- Wallenberg Initiative Materials Science for Sustainability (WISE) and Wallenberg Wood Science Center (WWSC), SE-100 44 Stockholm, Sweden
| | - Christopher A Berkey
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94301, United States
| | - Christopher John Bettinger
- Department of Biomedical Engineering and Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania 15213, United States
| | - Jun Chen
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Peng Chen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Wenlong Cheng
- Nanobionics Group, Department of Chemical and Biological Engineering, Monash University, Clayton, Australia, 3800
- Monash Institute of Medical Engineering, Monash University, Clayton, Australia3800
| | - Xu Cheng
- Applied Mechanics Laboratory, Department of Engineering Mechanics, Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, PR China
| | - Seon-Jin Choi
- Division of Materials of Science and Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul 04763, Republic of Korea
| | - Alex Chortos
- School of Mechanical Engineering, Purdue University, West Lafayette, Indiana 47906, United States
| | - Canan Dagdeviren
- Media Lab, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Reinhold H Dauskardt
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94301, United States
| | - Chong-An Di
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - Michael D Dickey
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Xiangfeng Duan
- Department of Chemistry and Biochemistry, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Antonio Facchetti
- Department of Chemistry and the Materials Research Center, Northwestern University, Evanston, Illinois 60208, United States
| | - Zhiyong Fan
- Department of Electronic and Computer Engineering and Department of Chemical and Biological Engineering, The Hong Kong University of Science and Technology, Clear Water Bay, Kowloon, Hong Kong SAR, China
| | - Yin Fang
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Jianyou Feng
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and Laboratory of Advanced Materials, Fudan University, Shanghai 200438, PR China
| | - Xue Feng
- Laboratory of Flexible Electronics Technology, Department of Engineering Mechanics, Tsinghua University, Beijing 100084, China
| | - Huajian Gao
- School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), 1 Fusionopolis Way, #16-16 Connexis, Singapore 138632, Republic of Singapore
| | - Wei Gao
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, California, 91125, United States
| | - Xiwen Gong
- Department of Chemical Engineering, Department of Materials Science and Engineering, Department of Electrical Engineering and Computer Science, Applied Physics Program, and Macromolecular Science and Engineering Program, University of Michigan, Ann Arbor, Michigan, 48109 United States
| | - Chuan Fei Guo
- Department of Materials Science and Engineering, Southern University of Science and Technology, Shenzhen 518055, China
| | - Xiaojun Guo
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication, School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Martin C Hartel
- Department of Bioengineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Zihan He
- Beijing National Laboratory for Molecular Sciences, CAS Key Laboratory of Organic Solids, Institute of Chemistry, Chinese Academy of Sciences, Beijing 100190, China
| | - John S Ho
- Institute for Health Innovation and Technology, National University of Singapore, Singapore 117599, Singapore
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117583, Singapore
- The N.1 Institute for Health, National University of Singapore, Singapore 117456, Singapore
| | - Youfan Hu
- School of Electronics and Center for Carbon-Based Electronics, Peking University, Beijing 100871, China
| | - Qiyao Huang
- School of Fashion and Textiles, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Yu Huang
- Department of Materials Science and Engineering, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Fengwei Huo
- Key Laboratory of Flexible Electronics (KLOFE) and Institute of Advanced Materials (IAM), Nanjing Tech University (NanjingTech), 30 South Puzhu Road, Nanjing 211816, PR China
| | - Muhammad M Hussain
- mmh Labs, Elmore Family School of Electrical and Computer Engineering, Purdue University, West Lafayette, Indiana 47906, United States
| | - Ali Javey
- Electrical Engineering and Computer Sciences, University of California, Berkeley, California 94720, United States
- Materials Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States
| | - Unyong Jeong
- Department of Materials Science and Engineering, Pohang University of Science and Engineering (POSTECH), Pohang, Gyeong-buk 37673, Korea
| | - Chen Jiang
- Department of Electronic Engineering, Tsinghua University, Beijing 100084, China
| | - Xingyu Jiang
- Department of Biomedical Engineering, Southern University of Science and Technology, No 1088, Xueyuan Road, Xili, Nanshan District, Shenzhen, Guangdong 518055, PR China
| | - Jiheong Kang
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
| | - Daniil Karnaushenko
- Research Center for Materials, Architectures and Integration of Nanomembranes (MAIN), Chemnitz University of Technology, Chemnitz 09126, Germany
| | | | - Dae-Hyeong Kim
- Center for Nanoparticle Research, Institute for Basic Science (IBS), School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Il-Doo Kim
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon 34141, Republic of Korea
| | - Dmitry Kireev
- Department of Electrical and Computer Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- Microelectronics Research Center, The University of Texas at Austin, Austin, Texas 78758, United States
| | - Lingxuan Kong
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
| | - Chengkuo Lee
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117583, Singapore
- Center for Intelligent Sensors and MEMS (CISM), National University of Singapore, Singapore 117608, Singapore
- National University of Singapore Suzhou Research Institute (NUSRI), Suzhou Industrial Park, Suzhou 215123, China
- NUS Graduate School-Integrative Sciences and Engineering Programme (ISEP), National University of Singapore, Singapore 119077, Singapore
| | - Nae-Eung Lee
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, Kyunggi-do 16419, Republic of Korea
| | - Pooi See Lee
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Singapore-HUJ Alliance for Research and Enterprise (SHARE), Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Tae-Woo Lee
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
- School of Chemical and Biological Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Institute of Engineering Research, Research Institute of Advanced Materials, Seoul National University, Soft Foundry, Seoul 08826, Republic of Korea
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Fengyu Li
- College of Chemistry and Materials Science, Jinan University, Guangzhou, Guangdong 510632, China
| | - Jinxing Li
- Department of Biomedical Engineering, Department of Electrical and Computer Engineering, Neuroscience Program, BioMolecular Science Program, and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48823, United States
| | - Cuiyuan Liang
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Chwee Teck Lim
- Department of Biomedical Engineering, National University of Singapore, Singapore 117583, Singapore
- Mechanobiology Institute, National University of Singapore, Singapore 117411, Singapore
- Institute for Health Innovation and Technology, National University of Singapore, Singapore 119276, Singapore
| | - Yuanjing Lin
- School of Microelectronics, Southern University of Science and Technology, Shenzhen 518055, China
| | - Darren J Lipomi
- Department of Nano and Chemical Engineering, University of California, San Diego, La Jolla, California 92093-0448, United States
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts, 02134, United States
| | - Kai Liu
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Nan Liu
- Beijing Key Laboratory of Energy Conversion and Storage Materials, College of Chemistry, Beijing Normal University, Beijing 100875, PR China
| | - Ren Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts, 02134, United States
| | - Yuxin Liu
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
- Department of Biomedical Engineering, N.1 Institute for Health, Institute for Health Innovation and Technology (iHealthtech), National University of Singapore, Singapore 119077, Singapore
| | - Yuxuan Liu
- Department of Mechanical and Aerospace Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Zhiyuan Liu
- Neural Engineering Centre, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China 518055
| | - Zhuangjian Liu
- Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), 1 Fusionopolis Way, #16-16 Connexis, Singapore 138632, Republic of Singapore
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
| | - Nanshu Lu
- Department of Aerospace Engineering and Engineering Mechanics, Department of Electrical and Computer Engineering, Department of Mechanical Engineering, Department of Biomedical Engineering, Texas Materials Institute, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Zhisheng Lv
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
| | - Shlomo Magdassi
- Institute of Chemistry and the Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem 9190401, Israel
| | - George G Malliaras
- Electrical Engineering Division, Department of Engineering, University of Cambridge CB3 0FA, Cambridge United Kingdom
| | - Naoji Matsuhisa
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo 153-8505, Japan
| | - Arokia Nathan
- Darwin College, University of Cambridge, Cambridge CB3 9EU, United Kingdom
| | - Simiao Niu
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, United States
| | - Jieming Pan
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117583, Singapore
| | - Changhyun Pang
- School of Chemical Engineering and Samsung Advanced Institute for Health Science and Technology, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Qibing Pei
- Department of Materials Science and Engineering, Department of Mechanical and Aerospace Engineering, California NanoSystems Institute, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Huisheng Peng
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and Laboratory of Advanced Materials, Fudan University, Shanghai 200438, PR China
| | - Dianpeng Qi
- School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, Heilongjiang 150001, China
| | - Huaying Ren
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, California, 90095, United States
| | - John A Rogers
- Querrey Simpson Institute for Bioelectronics, Northwestern University, Evanston, Illinois 60208, United States
- Department of Materials Science and Engineering, Department of Mechanical Engineering, Department of Biomedical Engineering, Departments of Electrical and Computer Engineering and Chemistry, and Department of Neurological Surgery, Northwestern University, Evanston, Illinois 60208, United States
| | - Aaron Rowe
- Becton, Dickinson and Company, 1268 N. Lakeview Avenue, Anaheim, California 92807, United States
- Ready, Set, Food! 15821 Ventura Blvd #450, Encino, California 91436, United States
| | - Oliver G Schmidt
- Research Center for Materials, Architectures and Integration of Nanomembranes (MAIN), Chemnitz University of Technology, Chemnitz 09126, Germany
- Material Systems for Nanoelectronics, Chemnitz University of Technology, Chemnitz 09107, Germany
- Nanophysics, Faculty of Physics, TU Dresden, Dresden 01062, Germany
| | - Tsuyoshi Sekitani
- The Institute of Scientific and Industrial Research (SANKEN), Osaka University, Osaka, Japan 5670047
| | - Dae-Gyo Seo
- Department of Materials Science and Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Guozhen Shen
- School of Integrated Circuits and Electronics, Beijing Institute of Technology, Beijing 100081, China
| | - Xing Sheng
- Department of Electronic Engineering, Beijing National Research Center for Information Science and Technology, Institute for Precision Medicine, Center for Flexible Electronics Technology, and IDG/McGovern Institute for Brain Research, Tsinghua University, Beijing, 100084, China
| | - Qiongfeng Shi
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117583, Singapore
- Center for Intelligent Sensors and MEMS (CISM), National University of Singapore, Singapore 117608, Singapore
- National University of Singapore Suzhou Research Institute (NUSRI), Suzhou Industrial Park, Suzhou 215123, China
| | - Takao Someya
- Department of Electrical Engineering and Information Systems, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | - Yanlin Song
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing, Beijing 100190, China
| | - Eleni Stavrinidou
- Laboratory of Organic Electronics, Department of Science and Technology, Linköping University, SE-601 74 Norrkoping, Sweden
| | - Meng Su
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing, Beijing 100190, China
| | - Xuemei Sun
- State Key Laboratory of Molecular Engineering of Polymers, Department of Macromolecular Science, and Laboratory of Advanced Materials, Fudan University, Shanghai 200438, PR China
| | - Kuniharu Takei
- Department of Physics and Electronics, Osaka Metropolitan University, Sakai, Osaka 599-8531, Japan
| | - Xiao-Ming Tao
- Research Institute for Intelligent Wearable Systems, School of Fashion and Textiles, Hong Kong Polytechnic University, Hong Kong, China
| | - Benjamin C K Tee
- Materials Science and Engineering, National University of Singapore, Singapore 117575, Singapore
- iHealthtech, National University of Singapore, Singapore 119276, Singapore
| | - Aaron Voon-Yew Thean
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117583, Singapore
- Singapore Hybrid-Integrated Next-Generation μ-Electronics Centre (SHINE), Singapore 117583, Singapore
| | - Tran Quang Trung
- School of Advanced Materials Science and Engineering, Sungkyunkwan University, Suwon, Kyunggi-do 16419, Republic of Korea
| | - Changjin Wan
- School of Electronic Science and Engineering, Nanjing University, Nanjing 210023, China
| | - Huiliang Wang
- Department of Biomedical Engineering, University of Texas at Austin, Austin, Texas 78712, United States
| | - Joseph Wang
- Department of Nanoengineering, University of California, San Diego, California 92093, United States
| | - Ming Wang
- Frontier Institute of Chip and System, State Key Laboratory of Integrated Chip and Systems, Zhangjiang Fudan International Innovation Center, Fudan University, Shanghai, 200433, China
- the Shanghai Qi Zhi Institute, 41th Floor, AI Tower, No.701 Yunjin Road, Xuhui District, Shanghai 200232, China
| | - Sihong Wang
- Pritzker School of Molecular Engineering, The University of Chicago, Chicago, Illinois, 60637, United States
| | - Ting Wang
- State Key Laboratory of Organic Electronics and Information Displays and Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Nanjing University of Posts and Telecommunications, 9 Wenyuan Road, Nanjing 210023, China
| | - Zhong Lin Wang
- Beijing Institute of Nanoenergy and Nanosystems, Chinese Academy of Sciences, Beijing 100083, China
- Georgia Institute of Technology, Atlanta, Georgia 30332-0245, United States
| | - Paul S Weiss
- California NanoSystems Institute, Department of Chemistry and Biochemistry, Department of Bioengineering, and Department of Materials Science and Engineering, University of California, Los Angeles, Los Angeles, California 90095, United States
| | - Hanqi Wen
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore 637457, Singapore
- Institute of Flexible Electronics Technology of THU, Jiaxing, Zhejiang, China 314000
| | - Sheng Xu
- Department of Nanoengineering, Department of Electrical and Computer Engineering, Materials Science and Engineering Program, and Department of Bioengineering, University of California San Diego, La Jolla, California, 92093, United States
| | - Tailin Xu
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong, 518060, PR China
| | - Hongping Yan
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
| | - Xuzhou Yan
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Hui Yang
- Tianjin Key Laboratory of Molecular Optoelectronic Sciences, Department of Chemistry, School of Science, Tianjin University, Tianjin, China, 300072
| | - Le Yang
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
- Department of Materials Science and Engineering, National University of Singapore (NUS), 9 Engineering Drive 1, #03-09 EA, Singapore 117575, Singapore
| | - Shuaijian Yang
- School of Biomedical Sciences, Faculty of Biological Sciences, University of Leeds, Leeds, LS2 9JT, United Kingdom
| | - Lan Yin
- School of Materials Science and Engineering, The Key Laboratory of Advanced Materials of Ministry of Education, State Key Laboratory of New Ceramics and Fine Processing, and Center for Flexible Electronics Technology, Tsinghua University, Beijing, 100084, China
| | - Cunjiang Yu
- Department of Engineering Science and Mechanics, Department of Biomedical Engineering, Department of Material Science and Engineering, Materials Research Institute, Pennsylvania State University, University Park, Pennsylvania, 16802, United States
| | - Guihua Yu
- Materials Science and Engineering Program and Walker Department of Mechanical Engineering, The University of Texas at Austin, Austin, Texas, 78712, United States
| | - Jing Yu
- School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Shu-Hong Yu
- Department of Chemistry, Institute of Biomimetic Materials and Chemistry, Hefei National Research Center for Physical Science at the Microscale, University of Science and Technology of China, Hefei 230026, China
| | - Xinge Yu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | - Evgeny Zamburg
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117583, Singapore
- Singapore Hybrid-Integrated Next-Generation μ-Electronics Centre (SHINE), Singapore 117583, Singapore
| | - Haixia Zhang
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication; Beijing Advanced Innovation Center for Integrated Circuits, School of Integrated Circuits, Peking University, Beijing 100871, China
| | - Xiangyu Zhang
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117583, Singapore
- Singapore Hybrid-Integrated Next-Generation μ-Electronics Centre (SHINE), Singapore 117583, Singapore
| | - Xiaosheng Zhang
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China
| | - Xueji Zhang
- School of Biomedical Engineering, Health Science Center, Shenzhen University, Shenzhen, Guangdong 518060, PR China
| | - Yihui Zhang
- Applied Mechanics Laboratory, Department of Engineering Mechanics; Laboratory of Flexible Electronics Technology, Tsinghua University, Beijing 100084, PR China
| | - Yu Zhang
- Department of Electrical and Computer Engineering, National University of Singapore, Singapore 117583, Singapore
- Singapore Hybrid-Integrated Next-Generation μ-Electronics Centre (SHINE), Singapore 117583, Singapore
| | - Siyuan Zhao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, Massachusetts, 02134, United States
| | - Xuanhe Zhao
- Department of Mechanical Engineering, Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts, 02139, United States
| | - Yuanjin Zheng
- Center for Integrated Circuits and Systems, School of Electrical and Electronic Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore 639798, Singapore
| | - Yu-Qing Zheng
- National Key Laboratory of Science and Technology on Micro/Nano Fabrication; School of Integrated Circuits, Peking University, Beijing 100871, China
| | - Zijian Zheng
- Department of Applied Biology and Chemical Technology, Faculty of Science, Research Institute for Intelligent Wearable Systems, Research Institute for Smart Energy, The Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong SAR, China
| | - Tao Zhou
- Center for Neural Engineering, Department of Engineering Science and Mechanics, The Huck Institutes of the Life Sciences, Materials Research Institute, The Pennsylvania State University, University Park, Pennsylvania 16802, United States
| | - Bowen Zhu
- Key Laboratory of 3D Micro/Nano Fabrication and Characterization of Zhejiang Province, School of Engineering, Westlake University, Hangzhou 310024, China
| | - Ming Zhu
- Institute for Digital Molecular Analytics and Science (IDMxS), Nanyang Technological University, 59 Nanyang Drive, Singapore 636921, Singapore
| | - Rong Zhu
- Department of Precision Instrument, Tsinghua University, Beijing 100084, China
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, 90064, United States
| | - Yong Zhu
- Department of Mechanical and Aerospace Engineering, Department of Materials Science and Engineering, and Department of Biomedical Engineering, North Carolina State University, Raleigh, North Carolina 27695, United States
| | - Guijin Zou
- Institute of High Performance Computing (IHPC), Agency for Science, Technology and Research (A*STAR), 1 Fusionopolis Way, #16-16 Connexis, Singapore 138632, Republic of Singapore
| | - Xiaodong Chen
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), 2 Fusionopolis Way, #08-03 Innovis, Singapore 138634, Republic of Singapore
- Innovative Center for Flexible Devices (iFLEX), Max Planck-NTU Joint Laboratory for Artificial Senses, School of Materials Science and Engineering, Nanyang Technological University, Singapore 639798, Singapore
| |
Collapse
|
24
|
Kashaninejad N, Nguyen NT. Microfluidic solutions for biofluids handling in on-skin wearable systems. LAB ON A CHIP 2023; 23:913-937. [PMID: 36628970 DOI: 10.1039/d2lc00993e] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
On-skin wearable systems for biofluid sampling and biomarker sensing can revolutionize the current practices in healthcare monitoring and personalized medicine. However, there is still a long path toward complete market adoption and acceptance of this fascinating technology. Accordingly, microfluidic science and technology can provide excellent solutions for bridging the gap between basic research and clinical research. The research gap has led to the emerging field of epidermal microfluidics. Moreover, recent advances in the fabrication of highly flexible and stretchable microfluidic systems have revived the concept of micro elastofluidics, which can provide viable solutions for on-skin wearable biofluid handling. In this context, this review highlights the current state-of-the-art platforms in this field and discusses the potential technologies that can be used for on-skin wearable devices. Toward this aim, we first compare various microfluidic platforms that could be used for on-skin wearable devices. These platforms include semiconductor-based, polymer-based, liquid metal-based, paper-based, and textile-based microfluidics. Next, we discuss how these platforms can enhance the stretchability of on-skin wearable biosensors at the device level. Next, potential microfluidic solutions for collecting, transporting, and controlling the biofluids are discussed. The application of finger-powered micropumps as a viable solution for precise and on-demand biofluid pumping is highlighted. Finally, we present the future directions of this field by emphasizing the applications of droplet-based microfluidics, stretchable continuous-flow micro elastofluidics, stretchable superhydrophobic surfaces, liquid beads as a form of digital micro elastofluidics, and topological liquid diodes that received less attention but have enormous potential to be integrated into on-skin wearable devices.
Collapse
Affiliation(s)
- Navid Kashaninejad
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia.
| | - Nam-Trung Nguyen
- Queensland Micro- and Nanotechnology Centre, Griffith University, 170 Kessels Road, Nathan, QLD 4111, Australia.
| |
Collapse
|
25
|
Deng Z, Guo L, Chen X, Wu W. Smart Wearable Systems for Health Monitoring. SENSORS (BASEL, SWITZERLAND) 2023; 23:s23052479. [PMID: 36904682 PMCID: PMC10007426 DOI: 10.3390/s23052479] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 06/12/2023]
Abstract
Smart wearable systems for health monitoring are highly desired in personal wisdom medicine and telemedicine. These systems make the detecting, monitoring, and recording of biosignals portable, long-term, and comfortable. The development and optimization of wearable health-monitoring systems have focused on advanced materials and system integration, and the number of high-performance wearable systems has been gradually increasing in recent years. However, there are still many challenges in these fields, such as balancing the trade-off between flexibility/stretchability, sensing performance, and the robustness of systems. For this reason, more evolution is required to promote the development of wearable health-monitoring systems. In this regard, this review summarizes some representative achievements and recent progress of wearable systems for health monitoring. Meanwhile, a strategy overview is presented about selecting materials, integrating systems, and monitoring biosignals. The next generation of wearable systems for accurate, portable, continuous, and long-term health monitoring will offer more opportunities for disease diagnosis and treatment.
Collapse
Affiliation(s)
- Zhiyong Deng
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou 730000, China
- Nuclear Power Institute of China, Huayang, Shuangliu District, Chengdu 610213, China
| | - Lihao Guo
- School of Advanced Materials and Nanotechnology, Interdisciplinary Research Center of Smart Sensors, Xidian University, Xi’an 710126, China
| | - Ximeng Chen
- School of Nuclear Science and Technology, Lanzhou University, Lanzhou 730000, China
| | - Weiwei Wu
- School of Advanced Materials and Nanotechnology, Interdisciplinary Research Center of Smart Sensors, Xidian University, Xi’an 710126, China
| |
Collapse
|
26
|
Krämer J, Grimm LM, Zhong C, Hirtz M, Biedermann F. A supramolecular cucurbit[8]uril-based rotaxane chemosensor for the optical tryptophan detection in human serum and urine. Nat Commun 2023; 14:518. [PMID: 36720875 PMCID: PMC9889744 DOI: 10.1038/s41467-023-36057-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 01/13/2023] [Indexed: 02/02/2023] Open
Abstract
Sensing small biomolecules in biofluids remains challenging for many optical chemosensors based on supramolecular host-guest interactions due to adverse interplays with salts, proteins, and other biofluid components. Instead of following the established strategy of developing alternative synthetic binders with improved affinities and selectivity, we report a molecular engineering approach that addresses this biofluid challenge. Here we introduce a cucurbit[8]uril-based rotaxane chemosensor feasible for sensing the health-relevant biomarker tryptophan at physiologically relevant concentrations, even in protein- and lipid-containing human blood serum and urine. Moreover, this chemosensor enables emission-based high-throughput screening in a microwell plate format and can be used for label-free enzymatic reaction monitoring and chirality sensing. Printed sensor chips with surface-immobilized rotaxane-microarrays are used for fluorescence microscopy imaging of tryptophan. Our system overcomes the limitations of current supramolecular host-guest chemosensors and will foster future applications of supramolecular sensors for molecular diagnostics.
Collapse
Affiliation(s)
- Joana Krämer
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Laura M Grimm
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Chunting Zhong
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
- Karlsruhe Nano Micro Facility (KNMFi), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany
| | - Michael Hirtz
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany.
- Karlsruhe Nano Micro Facility (KNMFi), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany.
| | - Frank Biedermann
- Institute of Nanotechnology (INT), Karlsruhe Institute of Technology (KIT), Hermann-von-Helmholtz Platz 1, 76344, Eggenstein-Leopoldshafen, Germany.
| |
Collapse
|
27
|
Fu J, Wang H, Na R, Jisaihan A, Wang Z, Ohno Y. Recent advancements in digital health management using multi-modal signal monitoring. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2023; 20:5194-5222. [PMID: 36896542 DOI: 10.3934/mbe.2023241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Healthcare is the method of keeping or enhancing physical and mental well-being with its aid of illness and injury prevention, diagnosis, and treatment. The majority of conventional healthcare practices involve manual management and upkeep of client demographic information, case histories, diagnoses, medications, invoicing, and drug stock upkeep, which can result in human errors that have an impact on clients. By linking all the essential parameter monitoring equipment through a network with a decision-support system, digital health management based on Internet of Things (IoT) eliminates human errors and aids the doctor in making more accurate and timely diagnoses. The term "Internet of Medical Things" (IoMT) refers to medical devices that have the ability to communicate data over a network without requiring human-to-human or human-to-computer interaction. Meanwhile, more effective monitoring gadgets have been made due to the technology advancements, and these devices can typically record a few physiological signals simultaneously, including the electrocardiogram (ECG) signal, the electroglottography (EGG) signal, the electroencephalogram (EEG) signal, and the electrooculogram (EOG) signal. Yet, there has not been much research on the connection between digital health management and multi-modal signal monitoring. To bridge the gap, this article reviews the latest advancements in digital health management using multi-modal signal monitoring. Specifically, three digital health processes, namely, lower-limb data collection, statistical analysis of lower-limb data, and lower-limb rehabilitation via digital health management, are covered in this article, with the aim to fully review the current application of digital health technology in lower-limb symptom recovery.
Collapse
Affiliation(s)
- Jiayu Fu
- Department of Mathematical Health Science, Graduate School of Medicine, Osaka University, Osaka 5650871, Japan
| | - Haiyan Wang
- Ma'anshan University, maanshan 243000, China
| | - Risu Na
- Department of Mathematical Health Science, Graduate School of Medicine, Osaka University, Osaka 5650871, Japan
- Shanghai Jian Qiao University, Shanghai 201315, China
| | - A Jisaihan
- Department of Mathematical Health Science, Graduate School of Medicine, Osaka University, Osaka 5650871, Japan
| | - Zhixiong Wang
- Department of Mathematical Health Science, Graduate School of Medicine, Osaka University, Osaka 5650871, Japan
- Ma'anshan University, maanshan 243000, China
| | - Yuko Ohno
- Department of Mathematical Health Science, Graduate School of Medicine, Osaka University, Osaka 5650871, Japan
| |
Collapse
|
28
|
Lapizco-Encinas BH, Zhang YV. Microfluidic systems in clinical diagnosis. Electrophoresis 2023; 44:217-245. [PMID: 35977346 DOI: 10.1002/elps.202200150] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 02/01/2023]
Abstract
The use of microfluidic devices is highly attractive in the field of biomedical and clinical assessments, as their portability and fast response time have become crucial in providing opportune therapeutic treatments to patients. The applications of microfluidics in clinical diagnosis and point-of-care devices are continuously growing. The present review article discusses three main fields where miniaturized devices are successfully employed in clinical applications. The quantification of ions, sugars, and small metabolites is examined considering the analysis of bodily fluids samples and the quantification of this type of analytes employing real-time wearable devices. The discussion covers the level of maturity that the devices have reached as well as cost-effectiveness. The analysis of proteins with clinical relevance is presented and organized by the function of the proteins. The last section covers devices that can perform single-cell metabolomic and proteomic assessments. Each section discusses several strategically selected recent reports on microfluidic devices successfully employed for clinical assessments, to provide the reader with a wide overview of the plethora of novel systems and microdevices developed in the last 5 years. In each section, the novel aspects and main contributions of each reviewed report are highlighted. Finally, the conclusions and future outlook section present a summary and speculate on the future direction of the field of miniaturized devices for clinical applications.
Collapse
Affiliation(s)
- Blanca H Lapizco-Encinas
- Microscale Bioseparations Laboratory and Biomedical Engineering Department, Rochester Institute of Technology, Rochester, New York, USA
| | - Yan Victoria Zhang
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
29
|
Yang H, Hu Y, Yin X, Huang J, Qiao C, Hu Z, He C, Huo D, Hou C. A disposable and sensitive non-enzymatic glucose sensor based on a 3D-Mn-doped NiO nanoflower-modified flexible electrode. Analyst 2023; 148:153-162. [DOI: 10.1039/d2an01495e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Herein, Mn-doped NiO nano-enzyme composites with high catalytic performance and excellent conductivity were grown on 3D CFC via hydrothermal and calcination methods to construct an efficient flexible glucose sensor.
Collapse
Affiliation(s)
- Huisi Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Yian Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Xinxue Yin
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Jiaqing Huang
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Cailin Qiao
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Zhikun Hu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Congjuan He
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
| | - Danqun Huo
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
- Chongqing Key Laboratory of Bio-perception & Intelligent Information Processing, School of Microelectronics and Communication Engineering, Chongqing University, Chongqing, 400044, PR China
| | - Changjun Hou
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China
- Liquor Making Biology Technology and Application of Key Laboratory of Sichuan Province, College of Bioengineering, Sichuan University of Science and Engineering, Zigong, 643000, PR China
| |
Collapse
|
30
|
Bran A, Tanase NO, Balan C. Interface Dynamics and the Influence of Gravity on Droplet Generation in a Y-microchannel. MICROMACHINES 2022; 13:1941. [PMID: 36363964 PMCID: PMC9696363 DOI: 10.3390/mi13111941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 11/02/2022] [Accepted: 11/07/2022] [Indexed: 06/16/2023]
Abstract
The present experimental investigation is focused on the influence of gravity upon water-droplet formation in a Y-microchannel filled with oil. The flows are in the Stokes regime, with very small capillary numbers and Ohnesorge numbers less than one. The study was performed in a square-cross-section channel, with a = 1.0 mm as the characteristic dimension and a flow rate ratio κ in a range between 0.55 and 1.8. The interface dynamics in the vicinity of breakup and the transitory plug flow regime after the detachment of the droplet were analysed. The dependence of droplet length L was correlated with the channel position against the gravity and κ parameters. The results of the work prove that, for κ=1, the droplet length L is independent of channel orientation.
Collapse
|
31
|
Coles L, Oluwasanya PW, Karam N, Proctor CM. Fluidic enabled bioelectronic implants: opportunities and challenges. J Mater Chem B 2022; 10:7122-7131. [PMID: 35959561 PMCID: PMC9518646 DOI: 10.1039/d2tb00942k] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 07/26/2022] [Indexed: 11/21/2022]
Abstract
Bioelectronic implants are increasingly facilitating novel strategies for clinical diagnosis and treatment. The integration of fluidic technologies into such implants enables new complementary routes for sensing and therapy alongside electrical interaction. Indeed, these two technologies, electrical and fluidic, can work synergistically in a bioelectronics implant towards the fabrication of a complete therapeutic platform. In this perspective article, the leading applications of fluidic enabled bioelectronic implants are highlighted and methods of operation and material choices are discussed. Furthermore, a forward-looking perspective is offered on emerging opportunities as well as critical materials and technological challenges.
Collapse
Affiliation(s)
- Lawrence Coles
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK.
| | - Pelumi W Oluwasanya
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK.
| | - Nuzli Karam
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK.
| | - Christopher M Proctor
- Electrical Engineering Division, Department of Engineering, University of Cambridge, Cambridge, UK.
| |
Collapse
|
32
|
Chavez‐Pineda OG, Rodriguez‐Moncayo R, Cedillo‐Alcantar DF, Guevara‐Pantoja PE, Amador‐Hernandez JU, Garcia‐Cordero JL. Microfluidic systems for the analysis of blood‐derived molecular biomarkers. Electrophoresis 2022; 43:1667-1700. [DOI: 10.1002/elps.202200067] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 06/18/2022] [Accepted: 06/22/2022] [Indexed: 12/19/2022]
Affiliation(s)
- Oriana G. Chavez‐Pineda
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Roberto Rodriguez‐Moncayo
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Diana F. Cedillo‐Alcantar
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Pablo E. Guevara‐Pantoja
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Josue U. Amador‐Hernandez
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
| | - Jose L. Garcia‐Cordero
- Laboratory of Microtechnologies Applied to Biomedicine (LMAB) Centro de Investigación y de Estudios Avanzados (Cinvestav) Monterrey Nuevo León Mexico
- Roche Institute for Translational Bioengineering (ITB) Roche Pharma Research and Early Development, Roche Innovation Center Basel Basel Switzerland
| |
Collapse
|
33
|
Chandra S, Hu T. From Prevention to Therapy: A Roadmap of Nanotechnologies to Stay Ahead of Future Pandemics. ACS NANO 2022; 16:9985-9993. [PMID: 35793456 PMCID: PMC9330760 DOI: 10.1021/acsnano.2c04148] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Several recent viral outbreaks, culminating in the COVID-19 pandemic, have illustrated the need for comprehensive improvement in the detection, control, and treatment of emerging viruses that exhibit the potential to cause epidemics. Nanotechnology approaches have the potential to make major contributions in all these areas. This perspective is intended to outline how nanotechnology can be employed to improve upon respiratory disease detection and containment measures, and therapeutics, with a particular emphasis on applications that can address key areas, including home diagnostics, contact tracing, and the evaluation of durability of vaccine protection over time and against future variants. Nanotechnology offers potent tools to address these needs, but further research is required to validate these applications to address needs of future epidemics.
Collapse
Affiliation(s)
- Sutapa Chandra
- Center
for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
- Department
of Biochemistry and Molecular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| | - Tony Hu
- Center
for Cellular and Molecular Diagnostics, Tulane University School of Medicine, New Orleans, Louisiana 70112, United States
- Department
of Biochemistry and Molecular Biology, Tulane
University School of Medicine, New Orleans, Louisiana 70112, United States
| |
Collapse
|
34
|
Teixidor J, Novello S, Ortiz D, Menin L, Lashuel HA, Bertsch A, Renaud P. On-Demand Nanoliter Sampling Probe for the Collection of Brain Fluid. Anal Chem 2022; 94:10415-10426. [PMID: 35786947 DOI: 10.1021/acs.analchem.2c01577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Continuous fluidic sampling systems allow collection of brain biomarkers in vivo. Here, we propose a new sequential and intermittent sampling paradigm using droplets, called Droplet on Demand (DoD). It is implemented in a microfabricated neural probe and alternates phases of analyte removal from the tissue and phases of equilibration of the concentration in the tissue. It allows sampling droplets loaded with molecules from the brain extracellular fluid punctually, without the long transient equilibration periods typical of continuous methods. It uses an accurately defined fluidic sequence with controlled timings, volumes, and flow rates, and correct operation is verified by the embedded electrodes and a flow sensor. As a proof of concept, we demonstrated the application of this novel approach in vitro and in vivo, to collect glucose in the brain of mice, with a temporal resolution of 1-2 min and without transient regime. Absolute quantification of the glucose level in the samples was performed by direct infusion nanoelectrospray ionization Fourier transform mass spectrometry (nanoESI-FTMS). By adjusting the diffusion time and the perfusion volume of DoD, the fraction of molecules recovered in the samples can be tuned to mirror the tissue concentration at accurate points in time. Moreover, this makes quantification of biomarkers in the brain possible within acute experiments of only 20-120 min. DoD provides a complementary tool to continuous microdialysis and push-pull sampling probes. Thus, the advances allowed by DoD will benefit quantitative molecular studies in the brain, i.e., for molecules involved in volume transmission or for protein aggregates that form in neurodegenerative diseases over long periods.
Collapse
Affiliation(s)
- Joan Teixidor
- Microsystems Laboratory 4 (STI-IEM-LMIS4), École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Salvatore Novello
- Laboratory of Molecular and Chemical Biology of Neurodegeneration (SV-BMI-LMNN), EPFL, 1015 Lausanne, Switzerland
| | - Daniel Ortiz
- Mass Spectrometry and Elemental Analysis Platform (SB-ISIC-MSEAP), EPFL, 1015 Lausanne, Switzerland
| | - Laure Menin
- Mass Spectrometry and Elemental Analysis Platform (SB-ISIC-MSEAP), EPFL, 1015 Lausanne, Switzerland
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration (SV-BMI-LMNN), EPFL, 1015 Lausanne, Switzerland
| | - Arnaud Bertsch
- Microsystems Laboratory 4 (STI-IEM-LMIS4), École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Philippe Renaud
- Microsystems Laboratory 4 (STI-IEM-LMIS4), École Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| |
Collapse
|
35
|
Li S, Zhang R, Zhang G, Shuai L, Chang W, Hu X, Zou M, Zhou X, An B, Qian D, Liu Z. Microfluidic manipulation by spiral hollow-fibre actuators. Nat Commun 2022; 13:1331. [PMID: 35288561 PMCID: PMC8921237 DOI: 10.1038/s41467-022-29088-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 02/16/2022] [Indexed: 12/31/2022] Open
Abstract
A microfluidic manipulation system that can sense a liquid and control its flow is highly desirable. However, conventional sensors and motors have difficulty fitting the limited space in microfluidic devices; moreover, fast sensing and actuation are required because of the fast liquid flow in the hollow fibre. In this study, fast torsional and tensile actuators were developed using hollow fibres employing spiral nonlinear stress, which can sense the fluid temperature and sort the fluid into the desired vessels. The fluid-driven actuation exhibited a highly increased response speed (27 times as fast as that of air-driven actuation) and increased power density (90 times that of an air-driven solid fibre actuator). A 0.5 K fluid temperature fluctuation produced a 20° rotation of the hollow fibre. These high performances originated from increments in both heat transfer and the average bias angle, which was understood through theoretical analysis. This work provides a new design strategy for intelligent microfluidics and inspiration for soft robots and smart devices for biological, optical, or magnetic applications.
Collapse
Affiliation(s)
- Sitong Li
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry and College of Pharmacy, Key Laboratory of Functional Polymer Materials, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Rui Zhang
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Guanghao Zhang
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry and College of Pharmacy, Key Laboratory of Functional Polymer Materials, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Luyizheng Shuai
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry and College of Pharmacy, Key Laboratory of Functional Polymer Materials, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Wang Chang
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry and College of Pharmacy, Key Laboratory of Functional Polymer Materials, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Xiaoyu Hu
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry and College of Pharmacy, Key Laboratory of Functional Polymer Materials, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Min Zou
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry and College of Pharmacy, Key Laboratory of Functional Polymer Materials, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Xiang Zhou
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry and College of Pharmacy, Key Laboratory of Functional Polymer Materials, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China
| | - Baigang An
- School of Chemical Engineering, University of Science and Technology Liaoning, Anshan, 114051, China
| | - Dong Qian
- Department of Mechanical Engineering, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Zunfeng Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Chemistry and College of Pharmacy, Key Laboratory of Functional Polymer Materials, Frontiers Science Center for New Organic Matter, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
36
|
Elvira KS, Gielen F, Tsai SSH, Nightingale AM. Materials and methods for droplet microfluidic device fabrication. LAB ON A CHIP 2022; 22:859-875. [PMID: 35170611 PMCID: PMC9074766 DOI: 10.1039/d1lc00836f] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 01/21/2022] [Indexed: 05/19/2023]
Abstract
Since the first reports two decades ago, droplet-based systems have emerged as a compelling tool for microbiological and (bio)chemical science, with droplet flow providing multiple advantages over standard single-phase microfluidics such as removal of Taylor dispersion, enhanced mixing, isolation of droplet contents from surfaces, and the ability to contain and address individual cells or biomolecules. Typically, a droplet microfluidic device is designed to produce droplets with well-defined sizes and compositions that flow through the device without interacting with channel walls. Successful droplet flow is fundamentally dependent on the microfluidic device - not only its geometry but moreover how the channel surfaces interact with the fluids. Here we summarise the materials and fabrication techniques required to make microfluidic devices that deliver controlled uniform droplet flow, looking not just at physical fabrication methods, but moreover how to select and modify surfaces to yield the required surface/fluid interactions. We describe the various materials, surface modification techniques, and channel geometry approaches that can be used, and give examples of the decision process when determining which material or method to use by describing the design process for five different devices with applications ranging from field-deployable chemical analysers to water-in-water droplet creation. Finally we consider how droplet microfluidic device fabrication is changing and will change in the future, and what challenges remain to be addressed in the field.
Collapse
Affiliation(s)
- Katherine S Elvira
- Department of Chemistry, Faculty of Science, University of Victoria, BC, Canada
| | - Fabrice Gielen
- Living Systems Institute, College of Engineering, Physics and Mathematics, University of Exeter, Exeter, EX4 4QD, UK
| | - Scott S H Tsai
- Department of Mechanical and Industrial Engineering, Ryerson University, ON, Canada
- Institute for Biomedical Engineering, Science, and Technology (iBEST)-a partnership between Ryerson University and St. Michael's Hospital, ON, Canada
- Keenan Research Centre for Biomedical Science, St. Michael's Hospital, ON, Canada
| | - Adrian M Nightingale
- Mechanical Engineering, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, UK
- Centre of Excellence for Continuous Digital Chemical Engineering Science, Faculty of Engineering and Physical Sciences, University of Southampton, Southampton, SO17 1BJ, UK.
| |
Collapse
|
37
|
Jeerapan I, Moonla C, Thavarungkul P, Kanatharana P. Lab on a body for biomedical electrochemical sensing applications: The next generation of microfluidic devices. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 187:249-279. [PMID: 35094777 DOI: 10.1016/bs.pmbts.2021.07.025] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
This chapter highlights applications of microfluidic devices toward on-body biosensors. The emerging application of microfluidics to on-body bioanalysis is a new strategy to establish systems for the continuous, real-time, and on-site determination of informative markers present in biofluids, such as sweat, interstitial fluid, blood, saliva, and tear. Electrochemical sensors are attractive to integrate with such microfluidics due to the possibility to be miniaturized. Moreover, on-body microfluidics coupled with bioelectronics enable smart integration with modern information and communication technology. This chapter discusses requirements and several challenges when developing on-body microfluidics such as difficulties in manipulating small sample volumes while maintaining mechanical flexibility, power-consumption efficiency, and simplicity of total automated systems. We describe key components, e.g., microchannels, microvalves, and electrochemical detectors, used in microfluidics. We also introduce representatives of advanced lab-on-a-body microfluidics combined with electrochemical sensors for biomedical applications. The chapter ends with a discussion of the potential trends of research in this field and opportunities. On-body microfluidics as modern total analysis devices will continue to bring several fascinating opportunities to the field of biomedical and translational research applications.
Collapse
Affiliation(s)
- Itthipon Jeerapan
- Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand; Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, Thailand.
| | - Chochanon Moonla
- School of Chemistry, Institute of Science, Suranaree University of Technology, Nakhon Ratchasima, Thailand
| | - Panote Thavarungkul
- Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand; Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| | - Proespichaya Kanatharana
- Division of Physical Science, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, Thailand; Center of Excellence for Trace Analysis and Biosensor, Prince of Songkla University, Hat Yai, Songkhla, Thailand
| |
Collapse
|
38
|
Lubken RM, de Jong AM, Prins MWJ. Real-Time Monitoring of Biomolecules: Dynamic Response Limits of Affinity-Based Sensors. ACS Sens 2022; 7:286-295. [PMID: 34978190 PMCID: PMC8805115 DOI: 10.1021/acssensors.1c02307] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
![]()
Sensors for monitoring
biomolecular dynamics in biological systems
and biotechnological processes in real time, need to accurately and
precisely reconstruct concentration–time profiles. This requirement
becomes challenging when transport processes and biochemical kinetics
are important, as is typically the case for biomarkers at low concentrations.
Here, we present a comprehensive methodology to study the concentration–time
profiles generated by affinity-based sensors that continuously interact
with a biological system of interest. Simulations are performed for
sensors with diffusion-based sampling (e.g., a sensor
patch on the skin) and advection-based sampling (e.g., a sensor connected to a catheter). The simulations clarify how
transport processes and molecular binding kinetics result in concentration
gradients and time delays in the sensor system. Using these simulations,
measured and true concentration–time profiles of insulin were
compared as a function of sensor design parameters. The results lead
to guidelines on how biomolecular monitoring sensors can be designed
for optimal bioanalytical performance in terms of concentration and
time properties.
Collapse
Affiliation(s)
- Rafiq M. Lubken
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven 5612 AZ, the Netherlands
- Institute for Complex Molecular Systems (ICMS), Eindhoven University of Technology, Eindhoven 5612 AZ, the Netherlands
| | - Arthur M. de Jong
- 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
| | - Menno W. J. Prins
- 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
- Helia Biomonitoring, Eindhoven 5612 AZ, the Netherlands
| |
Collapse
|
39
|
Liu J, Fang X, Zhang Z, Liu Z, Liu J, Sun K, Yuan Z, Yu J, Chiu DT, Wu C. Long-Term In Vivo Glucose Monitoring by Polymer-Dot Transducer in an Injectable Hydrogel Implant. Anal Chem 2022; 94:2195-2203. [PMID: 35034435 DOI: 10.1021/acs.analchem.1c04730] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Optical sensors have attracted a great deal of interest for glucose detection. However, their practical applications for continuous glucose monitoring are still constrained by operational reliability in subcutaneous tissues. Here, we show an implantable hydrogel platform embedded with luminescent polymer dots (Pdots) for sensitive and long-term glucose monitoring. We use Pdot transducer in a polyacrylamide hydrogel matrix to construct an implantable platform. The hydrogel-Pdot transducer showed bright luminescence with ratiometric response to glucose changes. The in vitro and in vivo sensitivities of the hydrogel implant were enhanced by varying the enzyme concentration and injection volume. After implantation, the hydrogel with Pdot transducer remained at the implanted site without migration for 1 month and can be removed from the subcutaneous tissue for further analysis. Our results indicate that the hydrogel-Pdot platform maintains the intrinsic sensing property with excellent stability during 1 month implantation, while fibrous capsule formation on the implant in some cases needs to be solved for long-term continuous glucose monitoring.
Collapse
Affiliation(s)
- Jing Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China.,Faculty of Health Science, University of Macau, Taipa, Macau SAR 999078, China
| | - Xiaofeng Fang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Zhe Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Zhihe Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Jie Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Kai Sun
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| | - Zhen Yuan
- Faculty of Health Science, University of Macau, Taipa, Macau SAR 999078, China
| | - Jiangbo Yu
- Department of Chemistry and Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Daniel T Chiu
- Department of Chemistry and Bioengineering, University of Washington, Seattle, Washington 98195, United States
| | - Changfeng Wu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen 518055, Guangdong, China
| |
Collapse
|
40
|
Zhang S, Zeng J, Wang C, Feng L, Song Z, Zhao W, Wang Q, Liu C. The Application of Wearable Glucose Sensors in Point-of-Care Testing. Front Bioeng Biotechnol 2021; 9:774210. [PMID: 34957071 PMCID: PMC8692794 DOI: 10.3389/fbioe.2021.774210] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/18/2021] [Indexed: 12/18/2022] Open
Abstract
Diabetes and its complications have become a worldwide concern that influences human health negatively and even leads to death. The real-time and convenient glucose detection in biofluids is urgently needed. Traditional glucose testing is detecting glucose in blood and is invasive, which cannot be continuous and results in discomfort for the users. Consequently, wearable glucose sensors toward continuous point-of-care glucose testing in biofluids have attracted great attention, and the trend of glucose testing is from invasive to non-invasive. In this review, the wearable point-of-care glucose sensors for the detection of different biofluids including blood, sweat, saliva, tears, and interstitial fluid are discussed, and the future trend of development is prospected.
Collapse
Affiliation(s)
- Sheng Zhang
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Ningbo Research Institute, Zhejiang University, Hangzhou, China
| | - Junyan Zeng
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Ningbo Research Institute, Zhejiang University, Hangzhou, China
| | - Chunge Wang
- School of Mechanical and Energy Engineering, Ningbo Tech University, Ningbo, China
| | - Luying Feng
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Ningbo Research Institute, Zhejiang University, Hangzhou, China
| | - Zening Song
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Ningbo Research Institute, Zhejiang University, Hangzhou, China
| | - Wenjie Zhao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Ningbo Research Institute, Zhejiang University, Hangzhou, China
| | - Qianqian Wang
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Ningbo Research Institute, Zhejiang University, Hangzhou, China
- College of Chemical and Biological Engineering, Zhejiang University, Hangzhou, China
| | - Chen Liu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Ningbo Research Institute, Zhejiang University, Hangzhou, China
| |
Collapse
|
41
|
Pan Y, Shan D, Ding LL, Yang XD, Xu K, Huang H, Wang JF, Ren HQ. Developing a generally applicable electrochemical sensor for detecting macrolides in water with thiophene-based molecularly imprinted polymers. WATER RESEARCH 2021; 205:117670. [PMID: 34583204 DOI: 10.1016/j.watres.2021.117670] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/30/2021] [Accepted: 09/11/2021] [Indexed: 05/05/2023]
Abstract
Our screening data revealed the threat macrolide antibiotics, especially azithromycin (AZN), posed to human health with its increasing occurrence in water environment. The electrochemical sensor based on molecularly imprinted polymer (MIP) is a promising platform that caters for the next generation of intelligent wastewater treatment plants (WWTPs) by virtue of its wide tolerance to water from all sources and in-situ monitoring. However, low initiation potentials of cross-linking monomers contributed by the electron-rich circumstance allowed them to usurp sites designed for functional monomers when electrically stimulated, leading to an unsatisfactory binding capacity. Another uncertainty is that multiple reaction sites of cross-linking monomers granted them complex polymerization routes and made it difficult to ensure the consistency of preparation. Serval monomers had been investigated with electrochemical tools and the performance of sensors constructed with these monomers were compared in this study. Based on the results, we proposed a protocol in which a novel functional monomer possessing a stronger electron-donating group, phenyl, was adopted to compete for the dominance in electropolymerization. Beyond that, the cross-linking monomer was modified with electron-withdrawing groups to raise its initiation potential. A monothiophene with a moderate initiation potential was also recruited as the linker to address the steric hindrance. In this way, polymerization proceeded in a specific order. It is worth mentioning that the Marangoni flow is an ideal tool to deal with the Coffee-ring deposition while drop-casting. The resulting sensor showed good performance with a limitation of detection (LOD) of 0.120 μM for AZN and a satisfactory selectivity, and the design can be applied to constructing sensors for a variety of macrolide antibiotics.
Collapse
Affiliation(s)
- Yao Pan
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Dan Shan
- School of Environmental and Biological Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Li-Li Ding
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Xu-Dong Yang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Ke Xu
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Hui Huang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Jin-Feng Wang
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China
| | - Hong-Qiang Ren
- State Key Laboratory of Pollution Control and Resource Reuse, School of the Environment, Nanjing University, Nanjing 210023, China.
| |
Collapse
|
42
|
Wang R, Zhou Y, Ghanbari Ghalehjoughi N, Mawaldi Y, Wang X. Ion-Induced Phase Transfer of Cationic Dyes for Fluorescence-Based Electrolyte Sensing in Droplet Microfluidics. Anal Chem 2021; 93:13694-13702. [PMID: 34590485 DOI: 10.1021/acs.analchem.1c03394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Fluorescence-based sensing in droplet microfluidics requires small sample volumes, allows for high-throughput assays, and does not suffer from photobleaching as each flowing sensor is only scanned one time. In this paper, we report a selective and sensitive fluorescence-based ion-sensing methodology in droplet microfluidics using a T-junction PDMS chip. The oil stream is doped with sensor ingredients including an ionophore, a cation exchanger, and a permanently cationic fluorophore as the optical reporter. Electrolyte cations from the aqueous sample are extracted into oil segments and displace the cationic dyes into aqueous droplets. Laser-induced fluorescence of the two immiscible phases is collected alternately, which is in clear contrast to most other ion-selective optode configurations such as nanoparticle suspensions that rely on mixed optical signals of two phases. The cation exchanger, tetrakis[3,5-bis(trifluoromethyl)phenyl]borate, is found to dramatically enhance the dye emission in the nonpolar sensing oil by preventing ion-pairing interactions and aggregations of the dye molecules, providing new insights into the mechanism of cationic dye-based ion sensors. The high dye brightness allows us to use low concentrations of sensing chemicals (e.g., 10 μM) in the oil and attain high sensitivity for detection of ions in an equal volume of sample. Using valinomycin as the ionophore and methylene blue as the dye, K+ is detected with a response time of ∼11 s, a logarithmic linear range of 10-5 to 10-2 M, a 20-fold total fluorescence response, >1000-fold selectivity against other electrolyte cations, and negligible cross-sensitivity toward the sample pH. The K+ concentration in untreated and undiluted whole blood and sweat samples is successfully determined by this microfluidic sensing method without optical interference from the droplet sample to the sensing oil. Detection of other ionic analytes can be achieved using the corresponding ionophores.
Collapse
Affiliation(s)
- Renjie Wang
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Yang Zhou
- School of Chemical Engineering and Technology, Key Laboratory of Advanced Materials of Tropical Island Resources of Ministry of Education, Hainan University, Haikou, Hainan 570228, China
| | | | - Yazan Mawaldi
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| | - Xuewei Wang
- Department of Chemistry, Virginia Commonwealth University, Richmond, Virginia 23284, United States
| |
Collapse
|
43
|
Quantitative electrolysis of droplet contents in microfluidic channels. Concept and experimental validation. Electrochim Acta 2021. [DOI: 10.1016/j.electacta.2021.139017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
44
|
Leroy A, Teixidor J, Bertsch A, Renaud P. In-flow electrochemical detection of chemicals in droplets with pyrolysed photoresist electrodes: application as a module for quantification of microsampled dopamine. LAB ON A CHIP 2021; 21:3328-3337. [PMID: 34250532 DOI: 10.1039/d1lc00116g] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
The electrochemical quantification of analytes in droplets of PBS separated by a fluorinated phase was investigated. PDMS-fused silica chips with pyrolysed photoresist electrodes were prepared using a simple fabrication technique and used to analyze droplets in flow. Potentiostatic chronoamperometry provided current readouts consistent with mass transport and the concentration inside the droplets. This paper highlights measurements of dopamine in droplets in T-junction microfluidic chips at unprecedently low concentrations, with a limit of detection of 207 nM and a linear range of 0.21-20 μM, giving results similar to continuous flow electrochemistry and allowing the analysis in the striatal extracellular range (<1 μM). The system was applied to the quick and reliable on-line detection of dopamine concentration steps in droplets collected with a microsampling probe in vitro, demonstrating the usefulness of the electrochemical device as a quantification module for microsampled chemicals in droplets.
Collapse
Affiliation(s)
- Albert Leroy
- EPFL-STI-IMT-LMIS4, École Polytechnique Fédérale de Lausanne, Station 17, CH-1015 Lausanne, Switzerland.
| | | | | | | |
Collapse
|
45
|
Phan LMT, Vo TAT, Hoang TX, Selvam SP, Pham HL, Kim JY, Cho S. Trending Technology of Glucose Monitoring during COVID-19 Pandemic: Challenges in Personalized Healthcare. ADVANCED MATERIALS TECHNOLOGIES 2021; 6:2100020. [PMID: 34179343 PMCID: PMC8212092 DOI: 10.1002/admt.202100020] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 02/18/2021] [Indexed: 05/11/2023]
Abstract
The COVID-19 pandemic has continued to spread rapidly, and patients with diabetes are at risk of experiencing rapid progression and poor prognosis for appropriate treatment. Continuous glucose monitoring (CGM), which includes accurately tracking fluctuations in glucose levels without raising the risk of coronavirus exposure, becomes an important strategy for the self-management of diabetes during this pandemic, efficiently contributing to the diabetes care and the fight against COVID-19. Despite being less accurate than direct blood glucose monitoring, wearable noninvasive systems can encourage patient adherence by guaranteeing reliable results through high correlation between blood glucose levels and glucose concentrations in various other biofluids. This review highlights the trending technologies of glucose sensors during the ongoing COVID-19 pandemic (2019-2020) that have been developed to make a significant contribution to effective management of diabetes and prevention of coronavirus spread, from off-body systems to wearable on-body CGM devices, including nanostructure and sensor performance in various biofluids. The advantages and disadvantages of various human biofluids for use in glucose sensors are also discussed. Furthermore, the challenges faced by wearable CGM sensors with respect to personalized healthcare during and after the pandemic are deliberated to emphasize the potential future directions of CGM devices for diabetes management.
Collapse
Affiliation(s)
- Le Minh Tu Phan
- Department of Electronic EngineeringGachon UniversitySeongnam‐siGyeonggi‐do13120Republic of Korea
- School of Medicine and PharmacyThe University of DanangDanang550000Vietnam
| | - Thuy Anh Thu Vo
- Department of Life ScienceGachon UniversitySeongnam‐siGyeonggi‐do461‐701Republic of Korea
| | - Thi Xoan Hoang
- Department of Life ScienceGachon UniversitySeongnam‐siGyeonggi‐do461‐701Republic of Korea
| | - Sathish Panneer Selvam
- Department of Electronic EngineeringGachon UniversitySeongnam‐siGyeonggi‐do13120Republic of Korea
| | - Hoang Lan Pham
- Department of Life ScienceGachon UniversitySeongnam‐siGyeonggi‐do461‐701Republic of Korea
| | - Jae Young Kim
- Department of Life ScienceGachon UniversitySeongnam‐siGyeonggi‐do461‐701Republic of Korea
| | - Sungbo Cho
- Department of Electronic EngineeringGachon UniversitySeongnam‐siGyeonggi‐do13120Republic of Korea
- Department of Health Sciences and TechnologyGAIHSTGachon UniversityIncheon21999Republic of Korea
| |
Collapse
|
46
|
Macdonald A, Hawkes LA, Corrigan DK. Recent advances in biomedical, biosensor and clinical measurement devices for use in humans and the potential application of these technologies for the study of physiology and disease in wild animals. Philos Trans R Soc Lond B Biol Sci 2021; 376:20200228. [PMID: 34176326 PMCID: PMC8237170 DOI: 10.1098/rstb.2020.0228] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/21/2021] [Indexed: 12/30/2022] Open
Abstract
The goal of achieving enhanced diagnosis and continuous monitoring of human health has led to a vibrant, dynamic and well-funded field of research in medical sensing and biosensor technologies. The field has many sub-disciplines which focus on different aspects of sensor science; engaging engineers, chemists, biochemists and clinicians, often in interdisciplinary teams. The trends which dominate include the efforts to develop effective point of care tests and implantable/wearable technologies for early diagnosis and continuous monitoring. This review will outline the current state of the art in a number of relevant fields, including device engineering, chemistry, nanoscience and biomolecular detection, and suggest how these advances might be employed to develop effective systems for measuring physiology, detecting infection and monitoring biomarker status in wild animals. Special consideration is also given to the emerging threat of antimicrobial resistance and in the light of the current SARS-CoV-2 outbreak, zoonotic infections. Both of these areas involve significant crossover between animal and human health and are therefore well placed to seed technological developments with applicability to both human and animal health and, more generally, the reviewed technologies have significant potential to find use in the measurement of physiology in wild animals. This article is part of the theme issue 'Measuring physiology in free-living animals (Part II)'.
Collapse
Affiliation(s)
- Alexander Macdonald
- Department of Biomedical Engineering, Wolfson Centre, University of Strathclyde, 106 Rottenrow, Glasgow G1 1XQ, UK
| | - Lucy A. Hawkes
- Department of Biosciences, University of Exeter, Hatherly Laboratories, Prince of Wales Road, Exeter EX4 4PS, UK
| | - Damion K. Corrigan
- Department of Biomedical Engineering, Wolfson Centre, University of Strathclyde, 106 Rottenrow, Glasgow G1 1XQ, UK
| |
Collapse
|
47
|
Mou L, Xia Y, Jiang X. Epidermal Sensor for Potentiometric Analysis of Metabolite and Electrolyte. Anal Chem 2021; 93:11525-11531. [PMID: 34378909 DOI: 10.1021/acs.analchem.1c01940] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Wearable epidermal sensors that can provide noninvasive and continuous analysis of metabolites and electrolytes in sweat have great significance for healthcare monitoring. This study reports an epidermal sensor that can wirelessly, noninvasively, and potentiometrically analyze metabolites and electrolytes. Potentiometry-based ion-selective electrodes (ISE) are most widely used for detecting electrolytes, such as Na+ and K+. We develop an enzyme-based glucose ISE for potentiometric analysis of sweat glucose. The glucose ISE sensor is obtained by modifying a glucose oxidase layer (GOD) on an H+ ISE sensor. GOD catalyzes glucose to generate H+. The generated H+ passes through the H+ selective membrane to change the potential of the electrode. We have fully examined the limit of detection, detecting range, and stability of our epidermal sensor. Meanwhile, using this epidermal sensor, we can easily analyze the relationship between blood glucose and sweat glucose. The concentration curve of sweat glucose can represent blood glucose concentration, significantly contributing to sports and chronic disease monitoring.
Collapse
Affiliation(s)
- Lei Mou
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou, Guangdong 510150, P. R. China.,Department of Biomedical Engineering, Southern University of Science and Technology, No 1088, Xueyuan Road, Xili, Nanshan District, Shenzhen, Guangdong 518055, P. R. China
| | - Yong Xia
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou, Guangdong 510150, P. R. China
| | - Xingyu Jiang
- Department of Clinical Laboratory, Third Affiliated Hospital of Guangzhou Medical University, No. 63 Duobao Road, Liwan District, Guangzhou, Guangdong 510150, P. R. China.,Department of Biomedical Engineering, Southern University of Science and Technology, No 1088, Xueyuan Road, Xili, Nanshan District, Shenzhen, Guangdong 518055, P. R. China
| |
Collapse
|
48
|
Tharmatt A, Malhotra D, Sharma H, Bedi N. Pharmaceutical Perspective in Wearable Drug Delivery Systems. Assay Drug Dev Technol 2021; 19:386-401. [PMID: 34339259 DOI: 10.1089/adt.2021.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Humans have been dealing with health problems for millions of years. Normal health services need well-trained personnel and high-cost diagnostic tests, which forces patients to go to hospitals if medical treatment is required. To address this, prototype testing has been carried out into the wearable drug delivery health care perspectives. Researchers have devised a wide variety of formulations for the treatment of various diseases at home by performing real-time monitoring of different routes of drug administration such as ocular, transdermal, intraoral, intracochlear, and several more. A comprehensive review of the different types of wearable drug delivery systems with respect to their manufacturing, mechanism of action and specifications has been done. In the pharmaceutical context, these devices are technologically well-equipped interfaces for diverse physicochemical signals. Above mentioned information with a broader perspective has also been discussed in this article. Several wearable drug delivery systems have been introduced in the market in recent years. But a lot of testing needs to be conducted to address the numerous obstacles before the wearable devices are successfully launched in the market.
Collapse
Affiliation(s)
- Abhay Tharmatt
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Danish Malhotra
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Hamayal Sharma
- Department of Pharmacy, Birla Institute of Technology and Science (BITS) Pilani, Pilani, India
| | - Neena Bedi
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| |
Collapse
|
49
|
Monitoring of Lactate in Interstitial Fluid, Saliva and Sweat by Electrochemical Biosensor: The Uncertainties of Biological Interpretation. CHEMOSENSORS 2021. [DOI: 10.3390/chemosensors9080195] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Lactate electrochemical biosensors were fabricated using Pediococcus sp lactate oxidase (E.C. 1.1.3.2), an external polyurethane membrane laminate diffusion barrier and an internal ionomeric polymer barrier (sulphonated polyether ether sulphone polyether sulphone, SPEES PES). In a needle embodiment, a Pt wire working electrode was retained within stainless steel tubing serving as pseudoreference. The construct gave linearity to at least 25 mM lactate with 0.17 nA/mM lactate sensitivity. A low permeability inner membrane was also unexpectedly able to increase linearity. Responses were oxygen dependent at pO2 < 70 mmHg, irrespective of the inclusion of an external diffusion barrier membrane. Subcutaneous tissue was monitored in Sprague Dawley rats, and saliva and sweat during exercise in human subjects. The tissue sensors registered no response to intravenous Na lactate, indicating a blood-tissue lactate barrier. Salivary lactate allowed tracking of blood lactate during exercise, but lactate levels were substantially lower than those in blood (0–3.5 mM vs. 1.6–12.1 mM), with variable degrees of lactate partitioning from blood, evident both between subjects and at different exercise time points. Sweat lactate during exercise measured up to 23 mM but showed highly inconsistent change as exercise progressed. We conclude that neither tissue interstitial fluid nor sweat are usable as surrogates for blood lactate, and that major reappraisal of lactate sensor use is indicated for any extravascular monitoring strategy for lactate.
Collapse
|
50
|
Shi N, Mohibullah M, Easley CJ. Active Flow Control and Dynamic Analysis in Droplet Microfluidics. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2021; 14:133-153. [PMID: 33979546 PMCID: PMC8956363 DOI: 10.1146/annurev-anchem-122120-042627] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Droplet-based microfluidics has emerged as an important subfield within the microfluidic and general analytical communities. Indeed, several unique applications such as digital assay readout and single-cell sequencing now have commercial systems based on droplet microfluidics. Yet there remains room for this research area to grow. To date, most analytical readouts are optical in nature, relatively few studies have integrated sample preparation, and passive means for droplet formation and manipulation have dominated the field. Analytical scientists continue to expand capabilities by developing droplet-compatible method adaptations, for example, by interfacing to mass spectrometers or automating droplet sampling for temporally resolved analysis. In this review, we highlight recently developed fluidic control techniques and unique integrations of analytical methodology with droplet microfluidics-focusing on automation and the connections to analog/digital domains-and we conclude by offering a perspective on current challenges and future applications.
Collapse
Affiliation(s)
- Nan Shi
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849, USA;
| | - Md Mohibullah
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849, USA;
| | - Christopher J Easley
- Department of Chemistry and Biochemistry, Auburn University, Auburn, Alabama 36849, USA;
| |
Collapse
|