[go: up one dir, main page]

Next Issue
Volume 12, November
Previous Issue
Volume 12, September
 
 

Biosensors, Volume 12, Issue 10 (October 2022) – 141 articles

Cover Story (view full-size image): Cardiovascular disease is the number one cause of death worldwide. Heart attacks account for 85% of this high mortality. Unfortunately, a significant number of heart attack survivors die within the first year because the waiting times for a diagnosis are critical for these patients. The clinical diagnostic procedure is not yet suitable for an early diagnosis. This paper presents a field effect transistor (FET) biosensor for the rapid, sensitive quantification of heart attack biomarker detection. The developed bio-FET combines the use of an excellent semiconducting channel with a biocompatible nanosheet material, which provides a bridge between the solid-state device and the bio-recognition element. This new bio-FET demonstrated the sensitive determination of heart attack biomarkers within a few minutes using only a few drops of a sample. View this paper
  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Section
Select all
Export citation of selected articles as:
13 pages, 1619 KiB  
Article
Efficient Entrapment of Alpha-Synuclein Biotinylated Antibody in KCC-1-NH-CS2 and Application for the Sensitive Diagnosis of Parkinson’s Using Recognition of Biomarker: An Innovative Electrochemical Label-Free Immunosensor for the Biomedical Analysis of Neurodegenerative Diseases
by Hossein Navay Baghban, Mohammad Hasanzadeh, Yuqian Liu and Farzad Seidi
Biosensors 2022, 12(10), 911; https://doi.org/10.3390/bios12100911 - 21 Oct 2022
Cited by 5 | Viewed by 2257
Abstract
The early detection of Parkinson’s disease (PD) is a critical issue in terms of efficiency. Alpha-synuclein (α-Syn) is a biomarker in PD checks. Alpha-synuclein (α-syn) is the major constituent of Lewy bodies and a pathogenic hallmark of all synucleinopathies, including PDs, dementia with [...] Read more.
The early detection of Parkinson’s disease (PD) is a critical issue in terms of efficiency. Alpha-synuclein (α-Syn) is a biomarker in PD checks. Alpha-synuclein (α-syn) is the major constituent of Lewy bodies and a pathogenic hallmark of all synucleinopathies, including PDs, dementia with Lewy bodies, and multiple system atrophy. In this study, KCC-1-NH-CS2 was conjugated with biotinylated Ab and entrapped in P(β-CD) polymer cavities. Using this approach, a novel electrochemical label-free immunosensor was designed for the quantification of α-syn in real human samples. For this purpose, the glassy carbon electrode electropolymerized with P(β-CD) biopolymer provided an excellent matrix for entrapping of KCC-1-NH-CS2 loaded with the biotinylated antibody of α-syn. Using the chronoamperometric technique, the proposed immunosensor shows a suitable range of 0.02 to 64 ng/mL for the determination of α-syn. Additionally, a low limit of quantification of the engineered biosensor was obtained at 0.02 ng/mL. The developed immunosensor’s adequate stability, sensitivity, and selectivity, together with its ease of manufacture, make it a promising diagnostic technique for further research. This study also will pave the way for further applications of the synergetic effect of β-CD and KCC-1-NH-CS2 for biomedical analysis in the near future. Full article
(This article belongs to the Special Issue Advances in Amplification Methods for Biosensors)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) CV characterization of step-by-step fabrication of immunosensor, the supporting electrolyte was 0.05 M solution [Fe (CN)<sub>6</sub>]<sup>3−/4−</sup>/KCl with a potential range from −1 to 1, scan rate = 0.1 V/s. (<b>B</b>) Comparison of peak currents recorded in the process of immunosensor fabrication.</p>
Full article ">Figure 2
<p>ChA curves of the GCE-P(β-CD)-KCC-1-NH-CS<sub>2</sub> in different concentrations of α-Syn (0.02, 2, 4, 8, 16, 64 ng/mL). The supporting electrolyte is 0.01 M ferricyanide/ferrocyanide. The applied potential was 0.3 V vs. Ag/AgCl, and the record time was set to 140 s. Inset: related calibration curve (n = 4, SD = 2.44).</p>
Full article ">Figure 3
<p>(<b>A</b>) CV of GCE-P(β-CD) at scan rates ranging from 0.002 to 1 V/s. (<b>B</b>) Dependence of the E<sub>pa</sub> vs. Ln<span class="html-italic">v</span>. (<b>C</b>) Dependence of LnI<sub>p</sub> vs. Ln<span class="html-italic">v</span>. (<b>D</b>) Dependence of Ip to the square root of sweep rates.</p>
Full article ">Figure 4
<p>(<b>A</b>) DPV of GCE-β-CD–KCC-1-NH-CS<sub>2</sub>-Ab-BSA in the presence of 16 ng/mL α-syn in three repetitions. The supporting electrolyte was 0.05 M solution [Fe (CN)<sub>6</sub>]<sup>3−/4−</sup>/KCl; the potential range was from -1 to 1. (<b>B</b>) Comparison of 1st, 2nd, and 3rd peak currents (SD = 2.13, n = 4).</p>
Full article ">Scheme 1
<p>The fabrication procedure of the electrochemical immunosensor for the recognition of α-syn biomarker.</p>
Full article ">
78 pages, 16259 KiB  
Review
Graphene and Its Derivatives: Synthesis and Application in the Electrochemical Detection of Analytes in Sweat
by Anoop Singh, Aamir Ahmed, Asha Sharma and Sandeep Arya
Biosensors 2022, 12(10), 910; https://doi.org/10.3390/bios12100910 - 21 Oct 2022
Cited by 29 | Viewed by 6739
Abstract
Wearable sensors and invasive devices have been studied extensively in recent years as the demand for real-time human healthcare applications and seamless human–machine interaction has risen exponentially. An explosion in sensor research throughout the globe has been ignited by the unique features such [...] Read more.
Wearable sensors and invasive devices have been studied extensively in recent years as the demand for real-time human healthcare applications and seamless human–machine interaction has risen exponentially. An explosion in sensor research throughout the globe has been ignited by the unique features such as thermal, electrical, and mechanical properties of graphene. This includes wearable sensors and implants, which can detect a wide range of data, including body temperature, pulse oxygenation, blood pressure, glucose, and the other analytes present in sweat. Graphene-based sensors for real-time human health monitoring are also being developed. This review is a comprehensive discussion about the properties of graphene, routes to its synthesis, derivatives of graphene, etc. Moreover, the basic features of a biosensor along with the chemistry of sweat are also discussed in detail. The review mainly focusses on the graphene and its derivative-based wearable sensors for the detection of analytes in sweat. Graphene-based sensors for health monitoring will be examined and explained in this study as an overview of the most current innovations in sensor designs, sensing processes, technological advancements, sensor system components, and potential hurdles. The future holds great opportunities for the development of efficient and advanced graphene-based sensors for the detection of analytes in sweat. Full article
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Structure of graphene. Reproduced from [<a href="#B35-biosensors-12-00910" class="html-bibr">35</a>] under common creative 3.0. license. (Copyright 2020, IntechOpen, London, UK).</p>
Full article ">Figure 2
<p>Synthesis approach for graphene.</p>
Full article ">Figure 3
<p>Graphite oxidation route schemes.</p>
Full article ">Figure 4
<p>A schematic diagram of the DC arc discharge apparatus. (Reproduced from [<a href="#B70-biosensors-12-00910" class="html-bibr">70</a>] with permission. Copyright 2012, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 5
<p>The FT-IR of solvents and schematic illustration of the exfoliation mechanism of graphene in the binary solvent system. (<b>a</b>) The development of bonding (hydrogen) between DMF and NBA molecules. (<b>b</b>) The exfoliation of graphite into few-layer graphene. (<b>c</b>) Graphite powder and graphene–DMF/3NBA dispersions. (Reproduced from [<a href="#B76-biosensors-12-00910" class="html-bibr">76</a>] with permission. Copyright 2015, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 6
<p>Schematic diagram of the two-step epitaxial graphene growth model. Reproduced from [<a href="#B118-biosensors-12-00910" class="html-bibr">118</a>] under common creative.</p>
Full article ">Figure 7
<p>The schematic representation summarizing the chemical reactions performed for functionalization. (Reproduced from [<a href="#B132-biosensors-12-00910" class="html-bibr">132</a>] with permission. Copyright 2015, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 8
<p>Structure of various graphene derivatives: (<b>a</b>) graphene and reduced graphene oxide (reproduced from [<a href="#B133-biosensors-12-00910" class="html-bibr">133</a>] under common creative 4.0), (<b>b</b>) graphene nanoribbons (reproduced from [<a href="#B134-biosensors-12-00910" class="html-bibr">134</a>] under common creative), (<b>c</b>) graphene nanowalls (reproduced from [<a href="#B135-biosensors-12-00910" class="html-bibr">135</a>] under common creative 4.0), and (<b>d</b>) graphene quantum dots (reproduced from [<a href="#B136-biosensors-12-00910" class="html-bibr">136</a>] with permission. Copyright 2012, American Chemical Society, Washington, WA, USA).</p>
Full article ">Figure 9
<p>Representations of defects in graphene-like materials using pictorial models. (<b>a</b>) defects in the hexagonal sp<sup>2</sup> hybridised carbon lattice, (<b>b</b>) Stone–Thrower–Wales defects, (<b>c</b>) replacement of carbon with another element inside the hexagonal lattice, (<b>d</b>) carbon defects, such as vacancies, edges, adatoms, interstitials, carbon chains, etc., that are not sp<sup>2</sup> hybridised, and (<b>e</b>) consequence of considerable deformation of graphene, folding-induced defects. (Reproduced from [<a href="#B199-biosensors-12-00910" class="html-bibr">199</a>] with permission. Copyright 2010, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 10
<p>The Poisson’s ratio of graphene nanoribbons with different widths. (<b>a</b>) Armchair nanoribbons and (<b>b</b>) zigzag nanoribbons. A graphene nanoribbon with 10:890 chirality under uniaxial tensile loading: (<b>c</b>) zero strain, (<b>d</b>) 10% strain, (<b>e</b>) 12% strain, and (<b>f</b>) 16% strain. (Reproduced from [<a href="#B207-biosensors-12-00910" class="html-bibr">207</a>] with permission. Copyright 2014, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 11
<p>The electrical characteristics of a graphene/hexagonal boron nitride bilayer. (<b>a</b>) layer of graphene/h−BN bilayer before stress, (<b>b</b>) graphene/h−BN bilayer structural model after full relaxation, (<b>c</b>) the interlayer binding energy curve for each super monomer, (<b>d</b>) strain maps for graphene and hexagonal boron nitride, and (<b>e</b>) lattice mismatch. (Reproduced from [<a href="#B246-biosensors-12-00910" class="html-bibr">246</a>] with permission. Copyright 2017, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 12
<p>Hybrid graphene–PCM thermal characteristics: (<b>a</b>) temperature-dependent enhancement factor of the thermal conductivity of graphene—paraffin composites with various graphene loadings. For comparison, the outcomes for pristine paraffin are also shown: (<b>b</b>) temperature-dependent specific heat of the composites and the reference pure paraffin. (Reproduced from [<a href="#B262-biosensors-12-00910" class="html-bibr">262</a>] with permission. Copyright 2013, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 13
<p>Graphene materials’ promising application areas.</p>
Full article ">Figure 14
<p>Skin-patchable sensors have a number of desirable characteristics.</p>
Full article ">Figure 15
<p>Wearable biosensor classification.</p>
Full article ">Figure 16
<p>Over the last decade, examples of integrated wearable sweat sensors have been created. (Reproduced from [<a href="#B404-biosensors-12-00910" class="html-bibr">404</a>] with permission. Copyright 2021, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 17
<p>Overview of the sweat-monitoring wearable multi-electrode system: (<b>a</b>) medical application; (<b>b</b>) the custom made and adaptable electrochemical multi-sensing system; (<b>c</b>) the flexible electrochemical multi-electrode platform; (<b>d</b>) bending test; (<b>e</b>) example of interface with read-out electronics. (Reproduced from [<a href="#B405-biosensors-12-00910" class="html-bibr">405</a>] under common creative License. Copyright 1969, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 18
<p>(<b>a</b>) Human participants were tested on the wearable platform using this experimental arrangement. (<b>b</b>) Thirty minutes after sweating began, the potassium and sodium levels in a volunteer’s sweat were measured using electrolyte analysis. (<b>c</b>) Analysis of the sweat of a volunteer after 30 min of indoor cycling to compare in situ and ex situ measures of potassium and sodium. (Reproduced from [<a href="#B405-biosensors-12-00910" class="html-bibr">405</a>] under common creative License. Copyright 1969, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 19
<p>Graphene-modified electrode manufacturing techniques are shown schematically in this diagram. (Reproduced from [<a href="#B440-biosensors-12-00910" class="html-bibr">440</a>] with permission. Copyright 2018, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 20
<p>(<b>a</b>) Molecular patterns of different D-glucose isomers. (<b>b</b>) The whole process of oxidising glucose. Slow hydrogen abstraction and oxidation transform D-glucose into D-glucono-δ-lactone, which is then rapidly hydrolysed into D-gluconic acid. (Reproduced from [<a href="#B452-biosensors-12-00910" class="html-bibr">452</a>] with permission. Copyright 2018, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 21
<p>(<b>a</b>) The mechanism of oxidation of glucose. (<b>b</b>) The effect of reactive species in the fast conversion of glucose into glucono-δ-lactone. (Reproduced from [<a href="#B458-biosensors-12-00910" class="html-bibr">458</a>] under common creative License 4.0).</p>
Full article ">Figure 22
<p>Results of electrochemical characterizations. (<b>a</b>) different electrochemical in vitro characterisation test configurations, (<b>b</b>) CV conducted in K<sub>3</sub>[Fe(CN)<sub>6</sub>] solution to compare the redox properties of pristine LIG with PtNPs and acetic acid-treated LIG with PtNPs at a scan rate of 50 mV/s, (<b>c</b>) cyclic voltammogram’s cathodic peak current, (<b>d</b>) urrent response of glucose and interferences such as AP, AA, NaCl, and UA, (<b>e</b>) current responses of different LIG−based electrode samples, (<b>f</b>) The current response of the various glucose concentration from ultra-low glucose levels. (<b>g</b>) Linear regression functional curve. (<b>h</b>) Current response under 1 μM glucose injection. (<b>i</b>) Daily variation of the sensitivity of the as-produced acetic acid-treated LIG/PtNPs/GOx electrode. (Reproduced from [<a href="#B471-biosensors-12-00910" class="html-bibr">471</a>] with permission. Copyright 2020, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 23
<p>Preparation of a three-dimensional microfluidic electrochemical biosensor based on paper. (<b>a</b>) preparation of a three-dimensional microfluidic electrochemical biosensor based on paper, (<b>b</b>) 8.2 mm hydrophilic zone, (<b>c</b>,<b>d</b>) carbon as counter and working electrode, and (<b>e</b>) reference electrode Ag/AgCl). (Reproduced from [<a href="#B472-biosensors-12-00910" class="html-bibr">472</a>] with permission. Copyright 2019, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 24
<p>Schematics of the developed wearable glucose biosensor based on sweat. Photos taken with an optical camera (<b>a</b>,<b>b</b>) of the developed biosensor. Diagram of the whole biosensor (<b>c</b>), with an expanded view (<b>d</b>). (Reproduced from [<a href="#B473-biosensors-12-00910" class="html-bibr">473</a>] with permission. Copyright 2018, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 25
<p>Diagram illustrating direct electron transport in (<b>a</b>) LOx, (<b>b</b>) LDH, (<b>c</b>) cross-linker attached LOx (<b>d</b>) cross-linker attached LDH; and for mediated electron transmission in (<b>e</b>) LDH and (<b>f</b>) LOx. (Reproduced from [<a href="#B555-biosensors-12-00910" class="html-bibr">555</a>] with permission. Copyright 2018, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 26
<p>(<b>a</b>) Lactate sweat sensor optical visualisation. (<b>b</b>) The electrochemical affinity-based lactate detection technology using graphene oxide across a membrane is shown schematically. (Reproduced from [<a href="#B556-biosensors-12-00910" class="html-bibr">556</a>] with permission. Copyright 2020, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 27
<p>Scheme of the biosensing platform. (Reproduced from [<a href="#B558-biosensors-12-00910" class="html-bibr">558</a>] with permission. Copyright 2015, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 28
<p>The creation of the lactate oxidase-based biosensor and the solution synthesis of the chevron−shaped GNR. (Reproduced from [<a href="#B559-biosensors-12-00910" class="html-bibr">559</a>] with permission. Copyright 2022, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 29
<p>Cyclic voltammograms of Lactate in 0.1 M phosphate buffer with 1 mM HMF. (Reproduced from [<a href="#B559-biosensors-12-00910" class="html-bibr">559</a>] with permission. Copyright 2022, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 30
<p>Illustrations showing the structure of the sweat glands, the release of biomarkers, and a wearable biosensor for detection of uric acid in sweat. (Reproduced from [<a href="#B563-biosensors-12-00910" class="html-bibr">563</a>] with permission. Copyright 2021, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 31
<p>Working mechanism of graphene-based UA biosensor. (Reproduced from [<a href="#B564-biosensors-12-00910" class="html-bibr">564</a>] under common creative.).</p>
Full article ">Figure 32
<p>(<b>a</b>) Schematic representation of the LIG-based sensor platform’s manufacturing process. (<b>b</b>) An illustration of the sensor platform mounted to the wrist and the results of such sensing. (Reproduced from [<a href="#B576-biosensors-12-00910" class="html-bibr">576</a>] under common creative.).</p>
Full article ">Figure 33
<p>(<b>a</b>–<b>e</b>) The K<sup>+</sup>-GCE, K<sup>+</sup>-GPE, Na<sup>+</sup>-GPE, Cl<sup>−</sup>GPE, and H<sup>+</sup>-GPE calibration curves and open-circuit potential responses. (<b>f</b>) Interference testing for Na<sup>+</sup>-GPE. (Reproduced from [<a href="#B577-biosensors-12-00910" class="html-bibr">577</a>] with permission. Copyright 2019, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 34
<p>(<b>a</b>) An image of the detection instrument. (<b>b</b>) Results of in-sweat pH and target ion concentration real-time monitoring. (<b>c</b>) Sweat analysis findings in comparison to real-time test samples. (Reproduced from [<a href="#B577-biosensors-12-00910" class="html-bibr">577</a>] with permission. Copyright 2019, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 35
<p>Diagram illustrating the many stages associated with producing modified SPEs. (Reproduced from [<a href="#B578-biosensors-12-00910" class="html-bibr">578</a>] with permission. Copyright 2021, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">Figure 36
<p>(<b>A</b>) Cyclic voltammetry was performed using 100 μL of 100 mM NaCl in the absence (dashed line) and presence (solid line) of oxygen, using Bare-SPE (black) and CB-modified SPE, with a scan rate of 25 mV/s. (<b>B</b>) Different NaCl concentrations were used to achieve electrochemical impedance spectroscopy using and CB-SPE. (Reproduced from [<a href="#B578-biosensors-12-00910" class="html-bibr">578</a>] with permission. Copyright 2021, Elsevier, Amsterdam, The Netherlands).</p>
Full article ">
12 pages, 3748 KiB  
Article
Glucose Incorporated Graphite Matrix for Electroanalysis of Trimethoprim
by Rakesh R. Sawkar, Mahesh M. Shanbhag, Suresh M. Tuwar, Ravindra S. Veerapur and Nagaraj P. Shetti
Biosensors 2022, 12(10), 909; https://doi.org/10.3390/bios12100909 - 21 Oct 2022
Cited by 5 | Viewed by 1981
Abstract
The antibiotic drug trimethoprim (TMP) is used to treat bacterial infections in humans and animals, and frequently TMP is used along with sulfonamides. However, a large portion of TMP is excreted in its active state, which poses a severe problem to humans and [...] Read more.
The antibiotic drug trimethoprim (TMP) is used to treat bacterial infections in humans and animals, and frequently TMP is used along with sulfonamides. However, a large portion of TMP is excreted in its active state, which poses a severe problem to humans and the environment. A sensitive, rapid, cost-effective analytical tool is required to monitor the TMP concentration in biological and environmental samples. Hence, this study proposed an analytical methodology to analyze TMP in clinical, biological and environmental samples. The investigations were carried out using a glucose-modified carbon paste electrode (G-CPE) employing voltammetric techniques. Electrochemical behavior was examined with 0.5 mM TMP solution at optimum pH 3.4 (Phosphate Buffer Solution, I = 0.2 M). The influence of scan rate on the electro-oxidation of TMP was studied within the range of 0.05 to 0.55 V/s. The effect of pH and scan rate variations revealed proton transfer during oxidation. Moreover, diffusion phenomena governed the irreversibility of the electrode reaction. A probable and suitable electrode interaction and reaction mechanism was proposed for the electrochemical oxidation of TMP. Further, the TMP was quantitatively estimated with the differential pulse voltammetry (DPV) technique in the concentration range from 9.0 × 10−7 to 1.0 × 10−4 M. The tablet, spiked water and urine analysis demonstrated that the selected method and developed electrode were rapid, simple, sensitive, and cost-effective. Full article
(This article belongs to the Special Issue Electrochemical (Bio-) Sensors in Biological Applications)
Show Figures

Figure 1

Figure 1
<p>AFM image of (<b>A</b>) CPE and (<b>B</b>) G-CPE, (<b>C</b>) and (<b>D</b>) SEM images of G-CPE, and (<b>E</b>) EDS image of G-CPE.</p>
Full article ">Figure 2
<p>(<b>A</b>) Cyclic voltammograms of (<b>a</b>) blank CPE, (<b>b</b>) blank G-CPE for buffer solution, (<b>c</b>) nascent CPE, and (<b>d</b>) G-CPE for 0.5 mM TMP solution, (<b>B</b>) Peak current responses at fabricated electrodes.</p>
Full article ">Figure 3
<p>(<b>A</b>) Cyclic voltammograms obtained for 0.5 mM TMP at different pH; (<b>B</b>) Plot of Ip vs. pH; (<b>C</b>) Plot of Ep vs. pH.</p>
Full article ">Figure 4
<p>(<b>A</b>) Differential pulse voltammograms obtained for various TMP concentrations from 0.9 µM–100 µM (1–13); (<b>B</b>) Dependence of Ip vs. concentration.</p>
Full article ">Scheme 1
<p>Probable interaction of glucose and trimethoprim.</p>
Full article ">Scheme 2
<p>Probable electrooxidation mechanism of TMP at G-CPE.</p>
Full article ">
14 pages, 3570 KiB  
Article
A Novel Activated Biochar-Based Immunosensor for Rapid Detection of E. coli O157:H7
by Abdus Sobhan, Fei Jia, Lisa Cooney Kelso, Sonatan Kumar Biswas, Kasiviswanathan Muthukumarappan, Changyong Cao, Lin Wei and Yanbin Li
Biosensors 2022, 12(10), 908; https://doi.org/10.3390/bios12100908 - 21 Oct 2022
Cited by 9 | Viewed by 2506
Abstract
E. coli O157:H7, one of the major foodborne pathogens, can cause a significant threat to the safety of foods. The aim of this research is to develop an activated biochar-based immunosensor that can rapidly detect E. coli O157:H7 cells without incubation in pure [...] Read more.
E. coli O157:H7, one of the major foodborne pathogens, can cause a significant threat to the safety of foods. The aim of this research is to develop an activated biochar-based immunosensor that can rapidly detect E. coli O157:H7 cells without incubation in pure culture. Biochar was developed from corn stalks using proprietary reactors and then activated using steam-activation treatment. The developed activated biochar presented an enhanced surface area of 830.78 m2/g. To develop the biosensor, the gold electrode of the sensor was first coated with activated biochar and then functionalized with streptavidin as a linker and further immobilized with biotin-labeled anti-E. coli polyclonal antibodies (pAbs). The optimum concentration of activated biochar for sensor development was determined to be 20 mg/mL. Binding of anti-E. coli pAbs with E. coli O157:H7 resulted in a significant increase in impedance amplitude from 3.5 to 8.5 kΩ when compared to an only activated biochar-coated electrode. The developed immunosensor was able to detect E. coli O157:H7 cells with a limit of detection of 4 log CFU/mL without incubation. Successful binding of E. coli O157:H7 onto an activated biochar-based immunosensor was observed on the microelectrode surface in scanning electron microscopy (SEM) images. Full article
(This article belongs to the Section Biosensor and Bioelectronic Devices)
Show Figures

Figure 1

Figure 1
<p>Formulation of corn stalk biochar through proprietary reactors and activation followed by steam activation treatment.</p>
Full article ">Figure 2
<p>Scheme of whole systems for electrochemical workstation and impedance analyzer, including the fabricated electrode.</p>
Full article ">Figure 3
<p>Stepwise developmental process of the activated biochar-based immunosensor to rapidly detect <span class="html-italic">E. coli</span> cells.</p>
Full article ">Figure 4
<p>Physical and electrical properties analyzed for activated biochar compared to biochar sample. (<b>A</b>) Raman spectra (IR); (<b>B</b>) BET analysis and current measurement; (<b>C</b>) optical images of biochar after pyrolysis and biochar after steam activation.</p>
Full article ">Figure 5
<p>(<b>A</b>) Resistance difference (∆R) of activated biochar-based immunosensor reacted with PBS as control and <span class="html-italic">E. coli</span> O157:H7; (<b>B</b>) spectra of impedance amplitude; (<b>C</b>) spectra of phase angle after stepwise-immobilization of activated biochar, streptavidin as linker, anti-<span class="html-italic">E. coli</span> antibodies on the immunosensor, and their response to <span class="html-italic">E. coli</span> O157:H7; (<b>D</b>) an equivalent circuit based on electrochemical system of the immunosensor.</p>
Full article ">Figure 6
<p>The specificity of custom-prepared anti-<span class="html-italic">E. coli</span> pAbs with selected bacterial strains determined by (<b>A</b>) dot blot assay; and (<b>B</b>) activated biochar-based immunosensor. PBS indicates phosphate-buffered saline as control.</p>
Full article ">Figure 7
<p>Resistance difference (∆R) of activated biochar-based immunosensor reacted with the selected concentration of <span class="html-italic">E. coli</span> O157:H7 from 10<sup>1</sup> to 10<sup>8</sup> CFU/mL. Different letters (a–c) denote a significant statistical difference among the concentrations at <span class="html-italic">p</span> &lt; 0.05. PBS stands for phosphate-buffered saline as control.</p>
Full article ">Figure 8
<p>Scanning electron microscopic images of the developed immunosensor. (<b>A</b>) Activated biochar-based electrode with 2 mm scale bar; (<b>B</b>) activated biochar with 10 µm scale bar; (<b>C</b>) streptavidin-linker-modified electrode; (<b>D</b>) anti-<span class="html-italic">E. coli</span> antibody-immobilized biosensor reacted to PBS; (<b>E</b>) antibody-immobilized electrode with captured <span class="html-italic">E. coli</span> cells with 5 µm scale bar; (<b>F</b>) captured <span class="html-italic">E. coli</span> with 10 µm scale bar.</p>
Full article ">
14 pages, 2171 KiB  
Article
Trace Determination of Grouper Nervous Necrosis Virus in Contaminated Larvae and Pond Water Samples Using Label-Free Fiber Optic Nanoplasmonic Biosensor
by Yuan-Yu Chen, Chih-Lu Wu, Chia-Wei Hsu, Chih-Hui Wang, Chung-Rui Su, Chun-Jen Huang, Hau-Ren Chen, Lai-Kwan Chau and Shau-Chun Wang
Biosensors 2022, 12(10), 907; https://doi.org/10.3390/bios12100907 - 21 Oct 2022
Cited by 2 | Viewed by 1958
Abstract
We developed a fast (<20 min), label-free fiber optic particle plasmon resonance (FOPPR) immunosensing method to detect nervous necrosis virus (NNV), which often infects high-value economic aquatic species, such as grouper. Using spiked NNV particles in a phosphate buffer as samples, the standard [...] Read more.
We developed a fast (<20 min), label-free fiber optic particle plasmon resonance (FOPPR) immunosensing method to detect nervous necrosis virus (NNV), which often infects high-value economic aquatic species, such as grouper. Using spiked NNV particles in a phosphate buffer as samples, the standard calibration curve obtained was linear (R2 = 0.99) and the limit of detection (LOD) achieved was 2.75 × 104 TCID50/mL, which is superior to that obtained using enzyme-linked immunosorbent assay (ELISA). By using an enhancement method called fiber optic nanogold-linked immunosorbent assay (FONLISA), the LOD can be further improved to <1 TCID50/mL, which is comparable to that found by the conventional qPCR method. Employing the larvae homogenate samples of NNV-infected grouper, the results obtained by the FOPPR biosensor agree with those obtained by the quantitative polymerase chain reaction (qPCR) method. We also examined pond water samples from an infected container in an indoor aquaculture facility. The lowest detectable level of NNV coat protein was found to be 0.17 μg/mL, which is one order lower than the LOD reported by ELISA. Therefore, we demonstrated the potential of the FOPPR biosensor as an outbreak surveillance tool, which is able to give warning indication even when the trend of larvae death toll increment is still not clear. Full article
(This article belongs to the Special Issue Waveguide Biosensors)
Show Figures

Figure 1

Figure 1
<p>The schematic illustration of FOPPR biosensor system used in this study: A. light-emitting diode (LED); B. sensing chip; C. sample inlet port; D. sample outlet port; E. sensing fiber with one unclad segment; F. photodiode; G. circuit board with LED driver and photoreceiver amplification circuit; H. data acquisition card; I. personal computer. In the upper right, the left imbedded photograph shows the upper and lower slides of a sensor chip, while the right photograph shows the sensing fiber with one unclad segment.</p>
Full article ">Figure 2
<p>(<b>a</b>) The sensorgram obtained by sequentially injecting NNV standard solutions at concentration of (1) 1.0 × 10<sup>3</sup>, (2) 2.5 × 10<sup>3</sup>, (3) 5.0 × 10<sup>3</sup>, (4) 7.5 × 10<sup>3</sup>, (5) 1.0 × 10<sup>4</sup>, (6) 2.5 × 10<sup>4</sup>, (7) 5.0 × 10<sup>4</sup>, (8) 7.5 × 10<sup>4</sup>, (9) 1.0 × 10<sup>5</sup>, (10) 2.5 × 10<sup>5</sup>, and (11) 5.0 × 10<sup>5</sup> TCID<sub>50</sub>/mL. (<b>b</b>) The standard calibration curve of the normalized response (ΔI/I<sub>0</sub>) versus the logarithm value of NNV concentration (log[C]). Each point is the mean of 3 repeated measurements.</p>
Full article ">Figure 3
<p>The sensorgram obtained by sequentially injecting diluted filtrate samples of NNV-infected larvae homogenate with dilution factor of (1) 320, (2) 160, (3) 80, and (4) 40.</p>
Full article ">Figure 4
<p>(<b>a</b>) The sensorgram obtained by sequentially injecting NNV standard solutions at concentrations of (1) 1.0 × 10<sup>0</sup>, (2) 1.0 × 10<sup>1</sup>, (3) 1.0 × 10<sup>2</sup>, (4) 1.0 × 10<sup>3</sup>, and (5) 1.0 × 10<sup>4</sup> TCID<sub>50</sub>/mL in the presence of AuNP@Ab<sup>D</sup> conjugate. (<b>b</b>) The standard calibration curve of the normalized response (ΔI/I<sub>0</sub>) versus the logarithm value of NNV concentration (log[C]).</p>
Full article ">Figure 5
<p>(<b>a</b>) The sensorgram obtained by sequentially injecting NNV coat protein standard solutions at concentrations of (1) 1.4 × 10<sup>−7</sup>, (2) 2.8 × 10<sup>−7</sup>, (3) 5.6 × 10<sup>−7</sup>, (4) 1.25 × 10<sup>−6</sup>, (5) 2.5 × 10<sup>−6</sup>, (6) 5.0 × 10<sup>−6</sup>, (7) 1.0 × 10<sup>−5</sup>, and (8) 2.0 × 10<sup>−5</sup> to g/mL. (<b>b</b>) The corresponding standard calibration curve of the normalized response (ΔI/I<sub>0</sub>) versus the logarithm value of NNV coat protein concentration (log[C]), over the range of 1.4 × 10<sup>−7</sup> to 2.5 × 10<sup>−6</sup> g/mL.</p>
Full article ">Figure 6
<p>The detected concentration of NNV coat proteins in an infected pond using FOPPR biosensor (curve A) from the eighth day to the nineteenth day during a larvae cultivation process; the death toll of grouper larvae (curve B) in the same time span.</p>
Full article ">Scheme 1
<p>Schematic of the steps involved in the fabrication of sensing fibers and the corresponding biosensing strategies for (<b>a</b>) direct method and (<b>b</b>) FONLISA method.</p>
Full article ">
14 pages, 36650 KiB  
Communication
An Innovative Simple Electrochemical Levofloxacin Sensor Assembled from Carbon Paste Enhanced with Nano-Sized Fumed Silica
by Amany M. Fekry
Biosensors 2022, 12(10), 906; https://doi.org/10.3390/bios12100906 - 21 Oct 2022
Cited by 13 | Viewed by 2184
Abstract
A new electrochemical sensor for the detection of levofloxacin (LV) was efficiently realized. The aim was to develop a new, cheap, and simple sensor for the detection of LV, which is used in various infections due to its pharmacological importance. It consists of [...] Read more.
A new electrochemical sensor for the detection of levofloxacin (LV) was efficiently realized. The aim was to develop a new, cheap, and simple sensor for the detection of LV, which is used in various infections due to its pharmacological importance. It consists of carbon paste (CP) enhanced with nano-sized fumed silica (NFS). NFS has a very low bulk density and a large surface area. The carbon paste-enhanced NFS electrode (NFS/CPE) showed great electrocatalytic activity in the oxidation of 1.0 mM LV in Britton–Robinson buffer (BR) at pH values ranging from 3.0 to 8.0. Cyclic voltammetry (CV) and differential pulse voltammetry (DPV) were used; the peak current value (Ip) of the NFS/CPE sensor was 2.7 times that of the bare electrode, ensuring its high electrocatalytic activity. Electrochemical impedance spectroscopy (EIS) was performed at a peak potential (Ep) of +1066 mV, yielding a resistance of 10 kΩ for the designed NFS/CPE sensor compared to 2461 kΩ for the bare electrode, indicating the high conductivity of the modified sensor and verifying the data observed using the CV technique. Surface descriptions were determined by scanning electron microscopy (SEM) and energy dispersive X-ray analysis (EDX). The variation in the concentration of LV (2.0 to 1000 µM) was considered in BR buffer (pH = 5.0) at a scan rate (SR) of 10 mV/s by the NFS/CPE. The detection and quantification limits were 0.09 µM and 0.30 µM, respectively. To evaluate the application of LV in real samples, this procedure was established on Quinostarmax 500 mg tablets and human plasma samples. Reasonable results were obtained for the detection of LV. Full article
Show Figures

Figure 1

Figure 1
<p>SEM images of (<b>A</b>) CPE and (<b>B</b>) NFS/CPE electrodes; (<b>C</b>) TEM image (size distribution histogram as inset); and (<b>D</b>) EDX spectra of nano-sized fumed silica improved carbon paste electrode.</p>
Full article ">Figure 2
<p>(<b>A</b>) CVs (SR = 50 mV/s) and Nyquist plots (at peak potential) of 1 mM LV using BR buffer (pH 5.0) at (<b>B</b>) CPE and (<b>C</b>) NFS/CPE. <b>Inset C:</b> fitting model.</p>
Full article ">Figure 3
<p>CVs of LV at dissimilar pH values by NFS/CPE, at a scan rate of 50 mV/s. <b>Inset:</b> (<b>A</b>) Deviation of anodic peak current (I<sub>pa</sub>), and (<b>B</b>) potential (E<sub>pa</sub>), with pH at NFS/CPE.</p>
Full article ">Figure 4
<p>(<b>A</b>) CVs of 1.0 mM LV in BR (pH 5.0) by means of various SRs (10.0–400 mV s<sup>−1</sup>). Inset: The I<sub>pa</sub> against the square root of SR at NFS/CPE. (<b>B</b>) The logarithm of the I<sub>pa</sub> and the logarithm of the SR at NFS/CPE. (<b>C</b>) The E<sub>pa</sub> against the logarithm of the SR at NFS/CPE. (<b>D</b>) The I<sub>pa</sub> against the square root of SR at CPE.</p>
Full article ">Figure 4 Cont.
<p>(<b>A</b>) CVs of 1.0 mM LV in BR (pH 5.0) by means of various SRs (10.0–400 mV s<sup>−1</sup>). Inset: The I<sub>pa</sub> against the square root of SR at NFS/CPE. (<b>B</b>) The logarithm of the I<sub>pa</sub> and the logarithm of the SR at NFS/CPE. (<b>C</b>) The E<sub>pa</sub> against the logarithm of the SR at NFS/CPE. (<b>D</b>) The I<sub>pa</sub> against the square root of SR at CPE.</p>
Full article ">Figure 5
<p>Outcome of the accumulation of time on I<sub>pa</sub> for 1.0 mM of LV in BR (pH 5.0) at NFS/CPE. Inset: the corresponding data as CVs.</p>
Full article ">Figure 6
<p>Calibration curve of LV using NFS/CPE.</p>
Full article ">Figure 7
<p>Instantaneous determination of 6 × 10<sup>−4</sup> M LV and ACOP.</p>
Full article ">Scheme 1
<p>Recommended oxidation reaction mechanism of LV.</p>
Full article ">
22 pages, 2405 KiB  
Review
Bacteriophage-Based Biosensors: A Platform for Detection of Foodborne Bacterial Pathogens from Food and Environment
by Rashad R. Al-Hindi, Addisu D. Teklemariam, Mona G. Alharbi, Ibrahim Alotibi, Sheren A. Azhari, Ishtiaq Qadri, Turki Alamri, Steve Harakeh, Bruce M. Applegate and Arun K. Bhunia
Biosensors 2022, 12(10), 905; https://doi.org/10.3390/bios12100905 - 21 Oct 2022
Cited by 29 | Viewed by 6055
Abstract
Foodborne microorganisms are an important cause of human illness worldwide. Two-thirds of human foodborne diseases are caused by bacterial pathogens throughout the globe, especially in developing nations. Despite enormous developments in conventional foodborne pathogen detection methods, progress is limited by the assay complexity [...] Read more.
Foodborne microorganisms are an important cause of human illness worldwide. Two-thirds of human foodborne diseases are caused by bacterial pathogens throughout the globe, especially in developing nations. Despite enormous developments in conventional foodborne pathogen detection methods, progress is limited by the assay complexity and a prolonged time-to-result. The specificity and sensitivity of assays for live pathogen detection may also depend on the nature of the samples being analyzed and the immunological or molecular reagents used. Bacteriophage-based biosensors offer several benefits, including specificity to their host organism, the detection of only live pathogens, and resistance to extreme environmental factors such as organic solvents, high temperatures, and a wide pH range. Phage-based biosensors are receiving increasing attention owing to their high degree of accuracy, specificity, and reduced assay times. These characteristics, coupled with their abundant supply, make phages a novel bio-recognition molecule in assay development, including biosensors for the detection of foodborne bacterial pathogens to ensure food safety. This review provides comprehensive information about the different types of phage-based biosensor platforms, such as magnetoelastic sensors, quartz crystal microbalance, and electrochemical and surface plasmon resonance for the detection of several foodborne bacterial pathogens from various representative food matrices and environmental samples. Full article
(This article belongs to the Special Issue Microbial Biosensors for Environmental Monitoring)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram showing the structure of major phage families: (<b>A</b>) <span class="html-italic">Myoviridae</span> (e.g., T4); (<b>B</b>) filamentous <span class="html-italic">Inoviridae</span> (e.g., M13); (<b>C</b>) long and noncontractile <span class="html-italic">Siphoviridae</span> (e.g., λ phage); (<b>D</b>) <span class="html-italic">Leviviridae</span>; and (<b>E</b>) short <span class="html-italic">Podoviridae</span> (e.g., T7).</p>
Full article ">Figure 2
<p>Schematic diagram showing the various intracellular cytoplasmic factors (biomarkers) released following phage infection. Abbr.: AK, adenosine kinase; ATP, adenosine triphosphate.</p>
Full article ">Figure 3
<p>Strategies used for immobilization of phages on biosensor platforms.</p>
Full article ">Figure 4
<p>Schematic diagram illustrating the working principle of a surface plasmon resonance sensor (SPR) using bacteriophages.</p>
Full article ">Figure 5
<p>Schematic illustration of phage-based impedimetric biosensor, showing steps involved in phage immobilization and target bacteria detection: (<b>A</b>) surface modification of gold electrode using chemical linker (e.g., cysteamine); (<b>B</b>) cross-linker/enhancement using 1,4 dithiocyanate (PDICT); (<b>C</b>) immobilization of phages and treatment with ethanolamine to block nonspecific binding; (<b>D</b>) capture of target bacteria; and (<b>E</b>) impedance measurement (detection of target pathogen).</p>
Full article ">Figure 6
<p>Schematic diagram elucidating the detection principle of the magnetoelastic phage sensor platform.</p>
Full article ">
13 pages, 4305 KiB  
Article
A Paper-Based Electrochemical Sensor Based on PtNP/COFTFPB−DHzDS@rGO for Sensitive Detection of Furazolidone
by Rongfang Chen, Xia Peng, Yonghai Song and Yan Du
Biosensors 2022, 12(10), 904; https://doi.org/10.3390/bios12100904 - 21 Oct 2022
Cited by 11 | Viewed by 2021
Abstract
Herein, a paper-based electrochemical sensor based on PtNP/COFTFPB−DHzDS@rGO was developed for the sensitive detection of furazolidone. A cluster-like covalent organic framework (COFTFPB−DHzDS) was successfully grown on the surface of amino-functional reduced graphene oxide (rGO-NH2) to avoid serious [...] Read more.
Herein, a paper-based electrochemical sensor based on PtNP/COFTFPB−DHzDS@rGO was developed for the sensitive detection of furazolidone. A cluster-like covalent organic framework (COFTFPB−DHzDS) was successfully grown on the surface of amino-functional reduced graphene oxide (rGO-NH2) to avoid serious self-aggregation, which was further loaded with platinum nanoparticles (PtNPs) with high catalytic activity as nanozyme to obtain PtNP/COFTFPB−DHzDS@rGO nanocomposites. The morphology of PtNP/COFTFPB−DHzDS@rGO nanocomposites was characterized, and the results showed that the smooth rGO surface became extremely rough after the modification of COFTFPB−DHzDS. Meanwhile, ultra-small PtNPs with sizes of around 1 nm were precisely anchored on COFTFPB−DHzDS to maintain their excellent catalytic activity. The conventional electrodes were used to detect furazolidone and showed a detection limit as low as 5 nM and a linear range from 15 nM to 110 μM. In contrast, the detection limit for the paper-based electrode was 0.23 μM, and the linear range was 0.69–110 μM. The results showed that the paper-based electrode can be used to detect furazolidone. This sensor is a potential candidate for the detection of furazolidone residue in human serum and fish samples. Full article
(This article belongs to the Special Issue Paper-Based Biosensors)
Show Figures

Figure 1

Figure 1
<p>SEM images of (<b>a</b>) rGO, (<b>b</b>) COF<sub>TFPB−DHzDS</sub>@rGO, and (<b>c</b>) PtNP/COF<sub>TFPB−DHzDS</sub>@rGO. TEM images of (<b>d</b>) rGO, (<b>e</b>) COF<sub>TFPB−DHzDS</sub>@rGO, and (<b>f</b>) PtNP/COF<sub>TFPB−DHzDS</sub>@rGO.</p>
Full article ">Figure 2
<p>(<b>a</b>) FTIR spectra of rGO, rGO-NH<sub>2</sub>, and COF<sub>TFPB−DHzDS</sub>@rGO; (<b>b</b>) XPS spectrum of PtNP/COF<sub>TFPB−DHzDS</sub>@rGO; (<b>c</b>–<b>f</b>) fine XPS spectra of Pt 4f, C 1s, N 1s, and O 1s, respectively.</p>
Full article ">Figure 3
<p>(<b>a</b>) CVs curves and (<b>b</b>) EIS diagram (inset is equivalent circuit diagram) of bare GCE (curve a), COF<sub>TFPB−DHzDS</sub>/GCE (curve b), and PtNP/COF<sub>TFPB−DHzDS</sub>@rGO/GCE (curve c) in 0.1 M KCl solution containing 5 mM [Fe (CN)<sub>6</sub>]<sup>3−/4−</sup>; (<b>c</b>) CVs of PtNP/COF<sub>TFPB−DHzDS</sub>@rGO/GCE at different scan rates (25 mV s<sup>−1</sup> to 225 mV s<sup>−1</sup>) in 0.1 M N<sub>2</sub>-statured phosphate-buffered solution (pH = 7.0) with 10 μM furazolidone; (<b>d</b>) the corresponding fitting curves between current density and scan rates; (<b>e</b>) the plots of peak current density versus different concentrations of TFPB, (<b>f</b>) different concentrations of potassium chloroplatinate, (<b>g</b>) different pH levels, and (<b>h</b>) different concentrations of PtNP/COF<sub>TFPB−DHzDS</sub>@rGO.</p>
Full article ">Figure 4
<p>(<b>a</b>) DPV diagram of GCE (curves a and b), COF<sub>TFPB−DHzDS</sub>/GCE (curves c and d), and PtNP/COF<sub>TFPB−DHzDS</sub>@rGO/GCE (curves e and f) before (<b>a</b>,<b>c</b>,<b>e</b>) and after (<b>b</b>,<b>d</b>,<b>f</b>) the addition of furazolidone in N<sub>2</sub>-saturated 0.2 M phosphate-buffered solution (pH = 7); (<b>b</b>) DPV of PtNP/COF<sub>TFPB−DHzDS</sub>@rGO/GCE in N<sub>2</sub>-saturated 0.2 M phosphate-buffered solution (pH = 7) in the presence of furazolidone with different concentration; (<b>c</b>) the linear curve of the corresponding peak current density and concentration; (<b>d</b>) histogram of PtNP/COF<sub>TFPB−DHzDS</sub>@rGO/GCE for different interfering substances (glucose, sodium carbonate, thiourea, NaCl, UA, DA, mannose); (<b>e</b>) current responses of six different PtNP/COF<sub>TFPB−DHzDS</sub>@rGO/GCE to 100 μM furazolidone; (<b>f</b>) current responses of PtNP/COF<sub>TFPB−DHzDS</sub>@rGO/GCE to 100 μM furazolidone after different days.</p>
Full article ">Figure 5
<p>(<b>a</b>) CVs curves and (<b>b</b>) EIS diagram of ePAD (curve a) and PtNP/COF<sub>TFPB−DHzDS</sub>@rGO/ePAD (curve b) in 0.1 M KCl solution containing 5 mM [Fe (CN)<sub>6</sub>]<sup>3−/4−</sup>; (<b>c</b>) DPV of furazolidone with different concentrations detected by PtNP/COF <sub>TFPB−DHzDS</sub>@rGO/ePAD; (<b>d</b>) linear curves corresponding to peak current density and concentration of PtNP/COF<sub>TFPB−DHzDs</sub>@rGO/ePAD.</p>
Full article ">Scheme 1
<p>Schematic illustration of the synthesis of PtNP/COF<sub>TFPB−DHzDS</sub>@rGO.</p>
Full article ">Scheme 2
<p>Schematic illustration of the preparation of paper-based electrodes.</p>
Full article ">
10 pages, 2428 KiB  
Article
Probing Subcellular Iron Availability with Genetically Encoded Nitric Oxide Biosensors
by Gulsah Sevimli, Amy E. Alston, Felix Funk, Beat Flühmann, Roland Malli, Wolfgang F. Graier and Emrah Eroglu
Biosensors 2022, 12(10), 903; https://doi.org/10.3390/bios12100903 - 21 Oct 2022
Cited by 3 | Viewed by 3775
Abstract
Cellular iron supply is required for various biochemical processes. Measuring bioavailable iron in cells aids in obtaining a better understanding of its biochemical activities but is technically challenging. Existing techniques have several constraints that make precise localization difficult, and the lack of a [...] Read more.
Cellular iron supply is required for various biochemical processes. Measuring bioavailable iron in cells aids in obtaining a better understanding of its biochemical activities but is technically challenging. Existing techniques have several constraints that make precise localization difficult, and the lack of a functional readout makes it unclear whether the tested labile iron is available for metalloproteins. Here, we use geNOps; a ferrous iron-dependent genetically encoded fluorescent nitric oxide (NO) biosensor, to measure available iron in cellular locales. We exploited the nitrosylation-dependent fluorescence quenching of geNOps as a direct readout for cellular iron absorption, distribution, and availability. Our findings show that, in addition to ferrous iron salts, the complex of iron (III) with N,N’-bis (2-hydroxybenzyl)ethylenediamine-N,N’-diacetic acid (HBED) can activate the iron (II)-dependent NO probe within intact cells. Cell treatment for only 20 min with iron sucrose was also sufficient to activate the biosensor in the cytosol and mitochondria significantly; however, ferric carboxymaltose failed to functionalize the probe, even after 2 h of cell treatment. Our findings show that the geNOps approach detects available iron (II) in cultured cells and can be applied to assay functional iron (II) at the (sub)cellular level. Full article
(This article belongs to the Special Issue Genetically Encoded Biosensors for Biomedical Applications)
Show Figures

Figure 1

Figure 1
<p>NO biosensor reports labile iron (II) in a concentration-dependent manner. (<b>A</b>) The cartoon shows iron (II) dependency of the NO biosensor geNOps. If iron salts are provided to cells, only ferrous iron can enter the cell cytosol through DMT1 channels but not ferric iron. Non-heme iron consisting of geNOps can sense NO in an iron (II)-concentration-dependent manner. (<b>B</b>) Average curves show real-time NO signals in response to 10 μM NOC-7 in HEK293T cells expressing O-geNOps. Cells have been treated with equimolar concentrations of FeSO<sub>4</sub> and Vitamin C, respectively, for 20 min before imaging experiments: 500 μM (black curve, <span class="html-italic">n</span> = 3/60), 300 μM (dark grey curve, <span class="html-italic">n</span> = 3/54), 100 μM (green curve, <span class="html-italic">n</span> = 3/56), 30 μM (blue curve, <span class="html-italic">n</span> = 3/42), and control no treatment (light grey curve, <span class="html-italic">n</span> = 3/73). (<b>C</b>) Bars represent the statistical analysis of iron (II) dependent geNOps responses shown in panel (<b>B</b>). All values are given as ±SD, and Dunnet’s multiple comparison test was applied. ** 0 vs. 30 um, Mean Difference (Diff): −5.525, q = 5.182, 95% Confidential Interval (CI) of Diff: −8.607 to −2.443; *** 0 vs. 100 um; Mean Diff: −9.870; q = 9.257, 95% CI of Diff: −12.95 to −6.79; *** 0 vs. 300 um; Mean Diff: −11.400; q = 10.69, 95% CI of Diff: −14.48 to −8.32; *** 0 vs. 500 um; Mean Diff: −13.800; q = 12.94, 95% CI of Diff: −16.88 to −10.71.</p>
Full article ">Figure 2
<p>Evaluation of the ferric iron chelator HBED and its effectiveness in different cultured cells to activate geNOps. (<b>A</b>) Representative wide-field images show immortalized endothelial cells (Ea.hy926), (<b>B</b>) human embryonic kidney cells (HEK293T), and (<b>C</b>) cervical cancer cells (HeLa) expressing cytosolic O-geNOps. <span class="html-italic">Lower Panels:</span> Average curves show NO responses of cells in response to 10 μM NOC-7 that have been treated with equimolar concentrations of HBED and Vitamin C, 1 mM, respectively, for 20 min before imaging experiments. The left panel shows Ea.hy926 cells (control grey curve, <span class="html-italic">n</span> = 3/60; HBED-treated, red curve, <span class="html-italic">n</span> = 3/74), middle panel HEK293T cells (control grey curve, <span class="html-italic">n</span> = 3/54; HBED-treated, red curve, <span class="html-italic">n</span> = 3/66) and the right panel shows HeLa cells (control grey curve, <span class="html-italic">n</span> = 3/29; HBED-treated, red curve, <span class="html-italic">n</span> = 3/18). All values are given as mean ± SD, and Student’s <span class="html-italic">t</span>-test was applied to compare maximum amplitudes between HBED-treated cells and control cells. (<b>A</b>) *** <span class="html-italic">p</span> = 0.0003, (<b>B</b>) ** <span class="html-italic">p</span> = 0.0081, (<b>C</b>) ** <span class="html-italic">p</span> = 0.0014.</p>
Full article ">Figure 3
<p>Evaluation of the effectiveness of IS and FCM in cultured Eahy.926 cells to activate geNOps. (<b>A</b>) The upper panel shows representative real-time traces of intracellular NO signals in response to 10 μM NOC-7 reflecting the labile iron pool in Ea.hy926 cells upon treatment with IS (red curve) or FCM (orange curve) vs. control cells (grey curves). Cells were pretreated with 500 μg iron/mL with the respective drug for 20 min before the imaging experiment. The lower panel shows statistical analysis. Control (grey curve, <span class="html-italic">n</span> = 3/46), FCM (orange curve, <span class="html-italic">n</span> = 3/47), and IS (red curve, <span class="html-italic">n</span> = 3/55). Panel (<b>B</b>) shows the same experimental setup as in panel (<b>A</b>), with the difference that cell pretreatment with IS and FCM was extended to 2 h. Control (grey curve, <span class="html-italic">n</span> = 3/29), FCM (orange curve, <span class="html-italic">n</span> = 3/28), and IS (red curve, <span class="html-italic">n</span> = 3/32). All values are given as mean ± SD, and Dunnet’s multiple comparison test was applied. (<b>A</b>) *** <span class="html-italic">p</span> = 0.0002, (<b>B</b>) *** <span class="html-italic">p</span> = 0.0001.</p>
Full article ">Figure 4
<p>Assessing iron (II) levels in subcellular locales upon IS treatment. (<b>A</b>) Representative wide-field images show HeLa cells stably expressing O-geNOps-NES (orange) transiently transfected with mitochondria-targeted C-geNOps (Mito-C-geNOp, cyan). (<b>B</b>) Real-time traces of intracellular NO signals in response to 10 μM NOC-7 in the cell cytosol (red curve and bar, co-imaged with O-geNOps, <span class="html-italic">n</span> = 3/18) or mitochondria (black curve and bar, co-imaged with C-geNOps, <span class="html-italic">n</span> = 3/18). All values are given as mean ± SD, and Student’s <span class="html-italic">t</span>-test was applied to compare maximum amplitudes between mitochondria and cytosolic NO signals.</p>
Full article ">
12 pages, 2796 KiB  
Article
Glycated Hemoglobin Electrochemical Immunosensor Based on Screen-Printed Electrode
by Yuliang Zhao, Hongyu Zhang, Yang Li, Xiaoai Wang, Liang Zhao, Jianghong Xu, Zhikun Zhan, Guanglie Zhang and Wen Jung Li
Biosensors 2022, 12(10), 902; https://doi.org/10.3390/bios12100902 - 21 Oct 2022
Cited by 2 | Viewed by 2895
Abstract
An electrochemical HbA1c sensor with high sensitivity and good specificity is proposed based on the electrochemical immune principle. The reproducibility and conductivity of the electrode are improved by depositing gold nanoparticles (AuNPs) on the surface of the screen-printed electrode (SPE). The HbA1c antibodies [...] Read more.
An electrochemical HbA1c sensor with high sensitivity and good specificity is proposed based on the electrochemical immune principle. The reproducibility and conductivity of the electrode are improved by depositing gold nanoparticles (AuNPs) on the surface of the screen-printed electrode (SPE). The HbA1c antibodies are immobilized on the surface of the modified electrode by adsorption to capture the HbA1c in the sample. The hindering effect of HbA1c on the electrode transfer reaction was exploited as the HbA1c detection mechanism. The electrode’s properties were characterized by electrochemical impedance spectroscopy (EIS), and the measurement properties of the electrode were analyzed using differential pulse voltammetry (DPV) and cyclic voltammetry (CV). The experimental results show that the peak current signal of the electrochemical immunosensor produced a linear response to HbA1c in the concentration range of 20–200 μg/mL, a linear relationship coefficient of 0.9812, a detection limit of 15.5 µg/mL, and a sensitivity of 0.0938 µA/µg·mL−1. The sensor delivered satisfactory repeatability, stability, and anti-interference performance. Due to its small size, high sensitivity, and wide linear detection range, it is expected to play a significant role in managing diabetes at home. Full article
(This article belongs to the Special Issue Biosensors for Bioanalytical and Healthcare Applications)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Dimensions of the SPE. (<b>B</b>) Electrodeposition of SPE in the mixture of 1 mM HAuCl<sub>4</sub> and 10 mM K<sub>2</sub>SO<sub>4</sub>. (<b>C</b>) Anti-HbA1c incubation and capture HbA1c. (<b>D</b>) AuNPs/anti-HbA1c/HbA1c electrode for K<sub>3</sub> [Fe(CN)<sub>6</sub>] testing.</p>
Full article ">Figure 2
<p>Schematic diagram of the proposed electrochemical HbA1c immunosensor.</p>
Full article ">Figure 3
<p>(<b>A</b>) CV response of SPEs before and after AuNPs modification. (<b>B</b>) DPV response of bare SPE, AuNPs SPE, AuNPs/anti-HbA1c SPE and AuNPs/anti-HbA1c/HbA1c SPE. (<b>C</b>) Complex impedance characteristics of electrodes in different states. (<b>D</b>) Charge-transfer resistance of electrodes in different states.</p>
Full article ">Figure 4
<p>(<b>A</b>) SEM image of the SPE (2000 times). (<b>B</b>) SEM image of the SPE after AuNPs modification (2000 times). (<b>C</b>) SEM image of the SPE (8000 times). (<b>D</b>) SEM image of the SPE after AuNPs modification (8000 times). (<b>E</b>) EDX spectrum of the bare electrode. (<b>F</b>) EDX spectrum of the AuNPs-modified electrode.</p>
Full article ">Figure 5
<p>Measurement curves and linear fitting diagram of the modified working electrode for different concentrations of HbA1c. (<b>A</b>) DPV measurement curves of the modified working electrode for different concentrations of HbA1c. (<b>B</b>) Linear fitting diagram of DPV peak current curves. (<b>C</b>) CV measurement curves of the modified working electrode for different concentrations of HbA1c. (<b>D</b>) Linear fitting diagram of CV current curves.</p>
Full article ">Figure 6
<p>(<b>A</b>) Repeatability measurement: current of the immune electrodes to HbA1c of 10 μg/mL, 40 μg/mL, 100 μg/mL, and 150 μg/mL (insert: peak current comparison). (<b>B</b>) Specificity measurement: current of the immune electrodes to HbA1c, BSA, GLU, FRU, and VC. (<b>C</b>) Long-term measurement: 0 h, 24 h, 72 h. (<b>D</b>) Repeated experiments at 72 h.</p>
Full article ">
33 pages, 3655 KiB  
Systematic Review
Image Quality Improvement Techniques and Assessment Adequacy in Clinical Optoacoustic Imaging: A Systematic Review
by Ioannis Dimaridis, Patmaa Sridharan, Vasilis Ntziachristos, Angelos Karlas and Leontios Hadjileontiadis
Biosensors 2022, 12(10), 901; https://doi.org/10.3390/bios12100901 - 20 Oct 2022
Cited by 6 | Viewed by 3108
Abstract
Optoacoustic imaging relies on the detection of optically induced acoustic waves to offer new possibilities in morphological and functional imaging. As the modality matures towards clinical application, research efforts aim to address multifactorial limitations that negatively impact the resulting image quality. In an [...] Read more.
Optoacoustic imaging relies on the detection of optically induced acoustic waves to offer new possibilities in morphological and functional imaging. As the modality matures towards clinical application, research efforts aim to address multifactorial limitations that negatively impact the resulting image quality. In an endeavor to obtain a clear view on the limitations and their effects, as well as the status of this progressive refinement process, we conduct an extensive search for optoacoustic image quality improvement approaches that have been evaluated with humans in vivo, thus focusing on clinically relevant outcomes. We query six databases (PubMed, Scopus, Web of Science, IEEE Xplore, ACM Digital Library, and Google Scholar) for articles published from 1 January 2010 to 31 October 2021, and identify 45 relevant research works through a systematic screening process. We review the identified approaches, describing their primary objectives, targeted limitations, and key technical implementation details. Moreover, considering comprehensive and objective quality assessment as an essential prerequisite for the adoption of such approaches in clinical practice, we subject 36 of the 45 papers to a further in-depth analysis of the reported quality evaluation procedures, and elicit a set of criteria with the intent to capture key evaluation aspects. Through a comparative criteria-wise rating process, we seek research efforts that exhibit excellence in quality assessment of their proposed methods, and discuss features that distinguish them from works with similar objectives. Additionally, informed by the rating results, we highlight areas with improvement potential, and extract recommendations for designing quality assessment pipelines capable of providing rich evidence. Full article
(This article belongs to the Special Issue Optical Imaging and Biophotonic Sensors (OIBS))
Show Figures

Figure 1

Figure 1
<p>Optoacoustic principle and imaging configurations. <b>Above</b>, the optoacoustic effect, in which light absorbed by chromophores in tissue results in emission of ultrasound. <b>Below</b>, categorization of optoacoustic imaging configurations into microscopic, mesoscopic, and macroscopic, with the corresponding tissue penetration depth limits and resolution orders of magnitude, as reported in relevant reviews [<a href="#B8-biosensors-12-00901" class="html-bibr">8</a>,<a href="#B9-biosensors-12-00901" class="html-bibr">9</a>,<a href="#B23-biosensors-12-00901" class="html-bibr">23</a>].</p>
Full article ">Figure 2
<p>Examples of optoacoustic images. Top: Microscopic images of the lipid core of a human carotid atheroma, adapted from [<a href="#B12-biosensors-12-00901" class="html-bibr">12</a>] with permission from Elsevier. (<b>a</b>) OAM image of embedded red blood cells. (<b>b</b>) Brightfield microscopy image. (<b>c</b>) Two-photon excitation microscopy fluorescence image. Middle: (<b>d</b>) RSOM image of the skin microvasculature on the dorsal aspect of the forearm, adapted from [<a href="#B24-biosensors-12-00901" class="html-bibr">24</a>], CC BY 4.0 (<a href="http://creativecommons.org/licences/by/4.0/" target="_blank">http://creativecommons.org/licences/by/4.0/</a>, accessed on 26 July 2022). Horizontal lines demarcate the epidermis (EP) and dermis (DR). The color scale represents the size of the imaged microvessels, with red representing large vessels, orange middle-sized vessels, and green small vessels. Bottom: Ultrasound and MSOT images of the carotid artery of a patient with carotid atherosclerosis, adapted from [<a href="#B17-biosensors-12-00901" class="html-bibr">17</a>] with permission from Elsevier. The arterial lumen is demarcated in red, and the plaque area with a yellow dashed line. (<b>e</b>) Ultrasound image. (<b>f</b>) MSOT image at 800 nm. (<b>g</b>) Magnification of the arterial cross-section in the ultrasound image. (<b>h</b>) Magnification of the same cross-section in the spectrally unmixed MSOT image in (<b>f</b>), showing the total hemoglobin (THb) signal. (<b>i</b>) Same magnification showing the lipids signal.</p>
Full article ">Figure 3
<p>Image domain manifestations of limitations in hardware and algorithms. The upper thick line represents the skin boundary. The circular shapes represent optoacoustic absorbers in tissue. (<b>a</b>) Original, unaffected image. (<b>b</b>) Background noise due to limited illumination intensity. (<b>c</b>) Deformation resulting from acoustic mismatch. (<b>d</b>) Loss of high-frequency information (blurring) due to limited bandwidth acquisition. (<b>e</b>) Incomplete structure rendering due to limited-view acquisition. (<b>f</b>) Negative values (represented with blue) near dominant absorbers. (<b>g</b>) Aliasing artifacts. (<b>h</b>) Smearing due to inadequate modelling of transducer detector dimensions.</p>
Full article ">Figure 4
<p>Image domain manifestations of limitations in tissue and in the acquisition process. The upper thick line represents the skin boundary. The shapes below represent optoacoustic absorbers in tissue. (<b>a</b>) Original, unaffected image. (<b>b</b>) Reduced contrast at depth due to optical attenuation. (<b>c</b>) Reduced resolution of deeper structures due to acoustic attenuation. (<b>d</b>) Reflection artifacts due to acoustic heterogeneity. (<b>e</b>) Clutter originating from optical absorbers outside the imaging plane. (<b>f</b>) Directional blurring due to motion.</p>
Full article ">Figure 5
<p>An overview of the literature search and screening procedure. The flow diagram was created according to PRISMA 2020 guidelines for systematic reviews [<a href="#B56-biosensors-12-00901" class="html-bibr">56</a>].</p>
Full article ">Figure 6
<p>Illustrative overviews of the rating template and procedure for a single study. The relationships between the individual ratings, experiment category-specific subtotals, and total and normalized total ratings are described. <b>Above</b>, the overview for all subgroups except for S<sub>MOT</sub>. <b>Below</b>, the overview for the S<sub>MOT</sub> subgroup.</p>
Full article ">Figure 7
<p>Plots of the normalized total ratings of all studies included in the subgroup analysis. On the <b>left</b>, the ratings, sorted in increasing order, are displayed as points. On the <b>right</b>, the corresponding box plot with the same points are overlaid. Points that correspond to ratings greater than the upper quartile selection threshold (<math display="inline"><semantics> <mrow> <msub> <mi>Q</mi> <mn>3</mn> </msub> <mo>=</mo> <mn>1.1</mn> </mrow> </semantics></math>) are colored red.</p>
Full article ">Figure 8
<p>Visual overview of the individual ratings for the human-involving experiments covering all criteria, subgroups, and studies. In case of criteria not applicable to certain subgroups, the corresponding space is left blank. The rating scale is as follows: 0: absent, 1: lacking, 2: adequate, 3: ample, 4: thorough. The points corresponding to different subgroups have been assigned unique colors to facilitate their visual identification.</p>
Full article ">
14 pages, 3431 KiB  
Article
A Closed-Loop Approach to Fight Coronavirus: Early Detection and Subsequent Treatment
by Guoguang Rong, Yuqiao Zheng, Xi Yang, Kangjian Bao, Fen Xia, Huihui Ren, Sumin Bian, Lan Li, Bowen Zhu and Mohamad Sawan
Biosensors 2022, 12(10), 900; https://doi.org/10.3390/bios12100900 - 20 Oct 2022
Cited by 3 | Viewed by 2462
Abstract
The recent COVID-19 pandemic has caused tremendous damage to the social economy and people’s health. Some major issues fighting COVID-19 include early and accurate diagnosis and the shortage of ventilator machines for critical patients. In this manuscript, we describe a novel solution to [...] Read more.
The recent COVID-19 pandemic has caused tremendous damage to the social economy and people’s health. Some major issues fighting COVID-19 include early and accurate diagnosis and the shortage of ventilator machines for critical patients. In this manuscript, we describe a novel solution to deal with COVID-19: portable biosensing and wearable photoacoustic imaging for early and accurate diagnosis of infection and magnetic neuromodulation or minimally invasive electrical stimulation to replace traditional ventilation. The solution is a closed-loop system in that the three modules are integrated together and form a loop to cover all-phase strategies for fighting COVID-19. The proposed technique can guarantee ubiquitous and onsite detection, and an electrical hypoglossal stimulator can be more effective in helping severe patients and reducing complications caused by ventilators. Full article
Show Figures

Figure 1

Figure 1
<p>Proposed closed-loop solution for COVID-19, including three main parts: portable biosensor, wearable photoacoustic imaging, and breath detection and magnetic as well as electrical stimulation.</p>
Full article ">Figure 2
<p>Biosensor and its portable detection system. Two different porous silicon layers construct the resonant microcavity, with an optical refractive index and thickness of n<sub>1</sub>, t<sub>1,</sub> and n<sub>2</sub>, t<sub>2</sub>. There are two 4-periods structures sandwiching a defect layer with thickness N times t<sub>1</sub>. N can be any number except 1. For details, see reference [<a href="#B11-biosensors-12-00900" class="html-bibr">11</a>]. Due to the nanoporous structures of porous silicon, a conformally deposited metal thin film is also nanoporous, which allows LSPR excitation by incident light.</p>
Full article ">Figure 3
<p>Flexible electronic device for photoacoustic sensing.</p>
Full article ">Figure 4
<p>Breathing monitor and subsequent electrical neurostimulation interface.</p>
Full article ">Figure 5
<p>The biosensor’s response for detecting SARS-CoV-2 N protein (10 ng/μL). Above: characteristic valley of the reflection spectrum used for tracking the spectral shift is circled in red. Below: real-time shift of the characteristic valley as the N protein of SARS-CoV-2 is binding with the specific antibody.</p>
Full article ">Figure 6
<p>Simulation results of photoacoustic imaging for lung tissues: (<b>a</b>) Optical absorption at 30 ns, (<b>b</b>) Optical fluence and temperature change from 0–60 ns at points (5, 6).</p>
Full article ">Figure 7
<p>Results of the force sensor for respiration monitoring, (<b>a</b>,<b>b</b>) Working principle of the PPy-based force sensor, (<b>c</b>) Current-time curves of the sensor with different applied forces. (<b>d</b>) The magnified view of the current-time curves at applied force ranges from 0.05 kPa to 0.5 kPa.</p>
Full article ">Figure 8
<p>(<b>a</b>) SEM image of micro-pyramidal PPy-based PDMS surface. The inset illustrates a detailed SEM image of the individual structure. (<b>b</b>) Optical image of the device.</p>
Full article ">Figure 9
<p>COMSOL-based simulation results of the magnetic field coupling link show the power transfer efficiency related to the load resistance.</p>
Full article ">
13 pages, 3417 KiB  
Article
A Novel Paper-Based Electrochemical Biosensor Based on N,O-Rich Covalent Organic Frameworks for Carbaryl Detection
by Yawen Xiao, Na Wu, Li Wang and Lili Chen
Biosensors 2022, 12(10), 899; https://doi.org/10.3390/bios12100899 - 20 Oct 2022
Cited by 10 | Viewed by 2260
Abstract
A new N,O-rich covalent organic framework (COFDHNDA-BTH) was synthesized by an amine-aldehyde condensation reaction between 2,6-dialdehyde-1,5-dihydroxynaphthalene (DHNDA) and 1,3,5-phenyltriformylhydrazine (BTH) for carbaryl detection. The free NH, OH, and C=O groups of COFDHNDA-BTH not only covalently couples with acetylcholinesterase (AChE) into [...] Read more.
A new N,O-rich covalent organic framework (COFDHNDA-BTH) was synthesized by an amine-aldehyde condensation reaction between 2,6-dialdehyde-1,5-dihydroxynaphthalene (DHNDA) and 1,3,5-phenyltriformylhydrazine (BTH) for carbaryl detection. The free NH, OH, and C=O groups of COFDHNDA-BTH not only covalently couples with acetylcholinesterase (AChE) into the pores of COFDHNDA-BTH, but also greatly improves the catalytic activity of AChE in the constrained environment of COFDHNDA-BTH’s pore. Under the catalysis of AChE, the acetylthiocholine (ATCl) was decomposed into positively charged thiocholine (TCl), which was captured on the COFDHNDA-BTH modified electrode. The positive charges of TCl can attract anionic probe [Fe(CN)6]3−/4− on the COFDHNDA-BTH-modified electrode to show a good oxidation peak at 0.25 V (versus a saturated calomel electrode). The carbaryl detection can inhibit the activity of AChE, resulting in the decrease in the oxidation peak. Therefore, a turn-off electrochemical carbaryl biosensor based on a flexible carbon paper electrode loaded with COFDHNDA-BTH and AChE was constructed using the oxidation peak of an anionic probe [Fe(CN)6]3−/4− as the detection signal. The detection limit was 0.16 μM (S/N = 3), and the linear range was 0.48~35.0 μM. The sensor has good selectivity, repeatability, and stability, and has a good application prospect in pesticide detection. Full article
(This article belongs to the Special Issue Paper-Based Biosensors)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) SEM image of COF<sub>DHNDA-BTH</sub>. (<b>b</b>) TEM image of COF<sub>DHNDA-BTH</sub>. (<b>c</b>) FTIR spectra of DHNDA, BTH, and COF<sub>DHNDA-BTH</sub>. (<b>d</b>) Experimental and refined XRD pattern as well as the difference.</p>
Full article ">Figure 2
<p>(<b>a</b>) Low-resolution and (<b>b</b>) high-resolution SEM image of commercial carbon paper.</p>
Full article ">Figure 3
<p>(<b>a</b>) CVs and (<b>b</b>) EIS of GCE (curve a), COF<sub>DHNDA-BTH</sub>/GCE (curve b), and AChE/COF<sub>DHNDA-BTH</sub>/GCE (curve c) in 0.1 M KCl with 5.0 mM [Fe(CN)<sub>6</sub>]<sup>3</sup><sup>−/4−</sup>.</p>
Full article ">Figure 4
<p>(<b>a</b>) Effect of COF<sub>DHNDA-BTH</sub> dosage on AChE/COF<sub>DHNDA-BTH</sub>/GCE on peak current density of 5.0 mM [Fe(CN)<sub>6</sub>]<sup>3−/4−</sup>. (<b>b</b>) Plot of inhibition rate versus incubation time. (<b>c</b>) Plot of peak current density versus AChE concentration. (<b>d</b>) Relationship between concentration of ATCl and peak current density in 0.1 M KCl with 5.0 mM [Fe(CN)<sub>6</sub>]<sup>3−/4−</sup>.</p>
Full article ">Figure 5
<p>(<b>a</b>) CVs and (<b>c</b>) EIS (inset is its equivalent circuit) of AChE/COF<sub>DHNDA-BTH</sub>/GCE in 0.1 M KCl + 5.0 mM [Fe(CN)6]<sup>3−/4−</sup> with different concentrations of carbaryl. (<b>b</b>,<b>d</b>) Corresponding linear relationship curves.</p>
Full article ">Figure 6
<p>(<b>a</b>) CVs of AChE/COF<sub>DHNDA-BTH</sub>/portable paper-based electrode in 0.1 M KCl + 5.0 mM [Fe(CN)6]<sup>3−/4−</sup> with carbaryl different concentrations. (<b>b</b>) Corresponding linear relationship curve.</p>
Full article ">Figure 7
<p>(<b>a</b>) Selectivity test of electrochemical carbaryl biosensor. (<b>b</b>) Current response histogram of electrochemical carbaryl biosensor measured for 30 consecutive days of carbaryl. (<b>c</b>) Current response histogram of six AChE/COF<sub>DHNDA-BTH</sub>/GCE to detect carbaryl.</p>
Full article ">Scheme 1
<p>Schematic diagram of preparation process of COF<sub>DHNDA-BTH</sub> and detection principle of electrochemical sensor based on AChE/COF<sub>DHNDA-BTH</sub>/GCE.</p>
Full article ">Scheme 2
<p>Preparation process of paper-based electrode and the picture of obtained paper-based electrode.</p>
Full article ">
25 pages, 2733 KiB  
Review
Advances in Biosensing Technologies for Diagnosis of COVID-19
by Sulaiman Alsalameh, Khalid Alnajjar, Tariq Makhzoum, Noor Al Eman, Ismail Shakir, Tanveer Ahmad Mir, Khaled Alkattan, Raja Chinnappan and Ahmed Yaqinuddin
Biosensors 2022, 12(10), 898; https://doi.org/10.3390/bios12100898 - 20 Oct 2022
Cited by 14 | Viewed by 3773
Abstract
The COVID-19 pandemic has severely impacted normal human life worldwide. Due to its rapid community spread and high mortality statistics, the development of prompt diagnostic tests for a massive number of samples is essential. Currently used traditional methods are often expensive, time-consuming, laboratory-based, [...] Read more.
The COVID-19 pandemic has severely impacted normal human life worldwide. Due to its rapid community spread and high mortality statistics, the development of prompt diagnostic tests for a massive number of samples is essential. Currently used traditional methods are often expensive, time-consuming, laboratory-based, and unable to handle a large number of specimens in resource-limited settings. Because of its high contagiousness, efficient identification of SARS-CoV-2 carriers is crucial. As the advantages of adopting biosensors for efficient diagnosis of COVID-19 increase, this narrative review summarizes the recent advances and the respective reasons to consider applying biosensors. Biosensors are the most sensitive, specific, rapid, user-friendly tools having the potential to deliver point-of-care diagnostics beyond traditional standards. This review provides a brief introduction to conventional methods used for COVID-19 diagnosis and summarizes their advantages and disadvantages. It also discusses the pathogenesis of COVID-19, potential diagnostic biomarkers, and rapid diagnosis using biosensor technology. The current advancements in biosensing technologies, from academic research to commercial achievements, have been emphasized in recent publications. We covered a wide range of topics, including biomarker detection, viral genomes, viral proteins, immune responses to infection, and other potential proinflammatory biomolecules. Major challenges and prospects for future application in point-of-care settings are also highlighted. Full article
Show Figures

Figure 1

Figure 1
<p>SARS-CoV-2 diagnosis by nucleic acid amplification method, qRT-PCR. (<b>1</b>,<b>2</b>). Nasopharyngeal swabs were taken from the patients. (<b>3</b>). RNA extracted/purified by RNA purification kit. (<b>4</b>). Extracted RNA is reverse transcribed to complementary DNA and amplification using the target-specific primers. (<b>5</b>). Quantitative real-time PCR (qRT-PCR) to identify the positive samples. Conducted to identify positive samples in real-time from the fluorescence of the dsDNA intercalating dyes such as SYBR green.</p>
Full article ">Figure 2
<p>Schematic diagram and operational principle of biosensors. The sample is introduced to the biosensor; the bioreceptor interacts with the target molecule. The transducer coupled with the receptor transfer the respective signal to the amplifier, then the amplified signal is converted into readable signals.</p>
Full article ">Figure 3
<p>Schematic representation of various genomic parts and their organization in the SARS-CoV-2 viral particle.</p>
Full article ">Figure 4
<p>Schematic representation of loop-mediated isothermal amplification of SARS-CoV-2 RNA amplification.</p>
Full article ">Figure 5
<p>(<b>a</b>) Pictorial representation of lateral flow immunoassay working principle. (<b>b</b>) The assay setup in the commercial test pad and (<b>c</b>) Interpretation of assay results positive/negative and the stability of the testing pad.</p>
Full article ">
12 pages, 2809 KiB  
Article
A High-Performance Self-Supporting Electrochemical Biosensor to Detect Aflatoxin B1
by Yunfei Zhang, Tingting Lin, Yi Shen and Hongying Li
Biosensors 2022, 12(10), 897; https://doi.org/10.3390/bios12100897 - 20 Oct 2022
Cited by 9 | Viewed by 2473
Abstract
High-performance electrochemical biosensors for the rapid detection of aflatoxin B1 (AFB1) are urgently required in the food industry. Herein, a multi-scaled electrochemical biosensor was fabricated by assembling carboxylated polystyrene nanospheres, an aptamer and horseradish peroxidase into a free-standing carbon nanofiber/carbon felt [...] Read more.
High-performance electrochemical biosensors for the rapid detection of aflatoxin B1 (AFB1) are urgently required in the food industry. Herein, a multi-scaled electrochemical biosensor was fabricated by assembling carboxylated polystyrene nanospheres, an aptamer and horseradish peroxidase into a free-standing carbon nanofiber/carbon felt support. The resulting electrochemical biosensor possessed an exceptional performance, owing to the unique structures as well as the synergistic effects of the components. The 3D porous carbon nanofiber/carbon felt support served as an ideal substrate, owing to the excellent conductivity and facile diffusion of the reactants. The integration of carboxylated polystyrene nanospheres with horseradish peroxidase was employed as a signal amplification probe to enhance the electrochemical responses via catalyzing the decomposition of hydrogen peroxide. With the aid of the aptamer, the prepared sensors could quantitatively detect AFB1 in wine and soy sauce samples via differential pulse voltammetry. The recovery rates of AFB1 in the samples were between 87.53% and 106.71%. The limit of detection of the biosensors was 0.016 pg mL−1. The electrochemical biosensors also had excellent sensitivity, reproducibility, specificity and stability. The synthetic strategy reported in this work could pave a new route to fabricate high-performance electrochemical biosensors for the detection of mycotoxins. Full article
Show Figures

Figure 1

Figure 1
<p>Digital photos of the hydrophilicity of the CF materials before and after thermal treatment. (<b>a</b>) Activated CF; (<b>b</b>) pristine CF. (<b>c</b>) CVs of the CF (black line) and activated CF (red line) were recorded in 1 mM [Fe(CN)<sub>6</sub>]<sup>3</sup><sup>−</sup><sup>/4</sup><sup>−</sup> with a scan rate of 10 mV s<sup>−</sup><sup>1</sup>.</p>
Full article ">Figure 2
<p>FESEM micrographs of Ni-Cu oxalate-decorated CF (<b>a</b>,<b>b</b>) and SSE (<b>c</b>,<b>d</b>) at different scales.</p>
Full article ">Figure 3
<p>(<b>a</b>) CVs of the SSE (black line) and Au/SSE (red line) electrodes recorded in 0.1 M NaOH with a scan rate of 10 mV s<sup>−</sup><sup>1</sup>. (<b>b</b>) CVs of the as-prepared electrodes recorded in 1 mM [Fe(CN)<sub>6</sub>]<sup>3−</sup> with a scan rate of 10 mV s<sup>−</sup><sup>1</sup>.</p>
Full article ">Figure 4
<p>(<b>a</b>) I<sub>p</sub> response of the biosensor to different concentrations of AFB<sub>1</sub> in a large concentration range of 0~5000 pg mL<sup>−</sup><sup>1</sup>. DPV responses of the biosensor recorded in 10 mM H<sub>2</sub>O<sub>2</sub> with varying concentrations of AFB<sub>1</sub> (<b>b</b>) and corresponding calibration curves of the biosensor for the detection of AFB<sub>1</sub> (<b>c</b>). Error bars were obtained from three independent measurements. (<b>d</b>) I-t curve of the biosensor recorded in varying H<sub>2</sub>O<sub>2</sub> concentrations.</p>
Full article ">Figure 5
<p>(<b>a</b>) Reproducibility of six electrode samples (S1–S6). (<b>b</b>) Stability of an as-prepared electrode. Error bars shown were obtained from three independent measurements.</p>
Full article ">Figure 6
<p>(<b>a</b>) Recovery rates of the developed biosensors for the detection of AFB<sub>1</sub> and the mixture of AFB<sub>1</sub> and OTA. Error bars were obtained from three independent measurements. (<b>b</b>) Selectivity test of the biosensor toward H<sub>2</sub>O<sub>2</sub> by interference with Na<sup>+</sup>, K<sup>+</sup>, Mg<sup>2+</sup>, AA, CA, starch and G.</p>
Full article ">Scheme 1
<p>Stepwise illustration of fabrication procedure of the electrochemical biosensor based on the self-supported electrode.</p>
Full article ">
11 pages, 1426 KiB  
Article
A Lateral Flow Device for Point-of-Care Detection of Doxorubicin
by Tania Pomili, Francesca Gatto and Pier Paolo Pompa
Biosensors 2022, 12(10), 896; https://doi.org/10.3390/bios12100896 - 19 Oct 2022
Cited by 4 | Viewed by 2076
Abstract
A simple, rapid, and sensitive point-of-care (POC) device for the on-site detection of doxorubicin was developed. The proposed method relies on the naked-eye detection of the intrinsic fluorescence of the drug in a lateral flow device (LFD) configuration, exploiting the biological recognition of [...] Read more.
A simple, rapid, and sensitive point-of-care (POC) device for the on-site detection of doxorubicin was developed. The proposed method relies on the naked-eye detection of the intrinsic fluorescence of the drug in a lateral flow device (LFD) configuration, exploiting the biological recognition of DNA probes and avoiding the use of expensive antibodies and sophisticated instrumentations. The POC assay does not require any pre-treatment or purification step and provides an immediate visual readout, achieving a limit of detection as low as ca. 1 ng doxorubicin, outperforming several laboratory-based instrumental techniques. The POC method was proven useful for the detection of trace amounts of the drug both in the case of water solutions (to simulate surface contaminations) and in urine samples, opening promising perspectives for routine monitoring of doxorubicin, with potential benefit to healthcare workers and personalized chemotherapies. Full article
(This article belongs to the Special Issue Biosensing for Point-of-Care Diagnostics)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the LFD for the assessment of doxorubicin contamination on surfaces and in urine. The device is composed of a running pad made of positively charged nylon and an absorbent pad, both laminated on the backing card and partially overlapped (2 mm). The test zone is realized by dropping 0.5 µL of the DNA solution onto the nylon pad. The test samples are deposited on the lateral side of the running pad and, flowing through the membrane, they reach the recognition element on the test zone. When doxorubicin molecules are present in the test sample, they intercalate with the immobilized DNA probes, giving an intense fluorescence, detectable by the naked eye, when excited by a UV lamp. In the case of non-contaminated samples, no intercalation occurs and, hence, no fluorescence is visualized.</p>
Full article ">Figure 2
<p>(<b>A</b>) Representative photograph of the proposed devices tested with decreasing doxorubicin amounts, ranging from 160 to 0 ng, as reported in the upper part of the image (160, 80, 40, 16, 8, 4, 1.5, 0 ng, roughly corresponding to 10, 5, 2.5, 1, 0.5, 0.25, 0.1 µM); (<b>B</b>) analysis of the optical response of the device as a function of doxorubicin amounts (ΔG values were obtained subtracting the G coordinate of the sample from that of the blank (in RGB coordinates system)). G values were recorded in 9 different experiments.</p>
Full article ">Figure 3
<p>Representative photograph of the LFD devices tested with real urine sample spiked with increasing doxorubicin amounts, as reported in the upper part of the image (240, 160, 80, 40, 16, 0 ng, roughly corresponding to 15, 10, 5, 2.5, 1, 0 µM).</p>
Full article ">
11 pages, 4584 KiB  
Article
Effect of the Combination of Gold Nanoparticles and Polyelectrolyte Layers on SERS Measurements
by Antonello Nucera, Rossella Grillo, Carmen Rizzuto, Riccardo Cristoforo Barberi, Marco Castriota, Thomas Bürgi, Roberto Caputo and Giovanna Palermo
Biosensors 2022, 12(10), 895; https://doi.org/10.3390/bios12100895 - 19 Oct 2022
Cited by 4 | Viewed by 2059
Abstract
In this study, polyelectrolyte (PE) layers are deposited on substrates made by glass covered with an array of gold nanoparticles (GNPs). In particular, the samples studied have 0 PE layers (GGPE0), 3 PE layers (GGPE3), 11 PE layers (GGPE [...] Read more.
In this study, polyelectrolyte (PE) layers are deposited on substrates made by glass covered with an array of gold nanoparticles (GNPs). In particular, the samples studied have 0 PE layers (GGPE0), 3 PE layers (GGPE3), 11 PE layers (GGPE11), and 21 PE layers (GGPE21). All samples have been studied by micro-Raman spectroscopy. An acetic acid solution (10% v/v) has been used as a standard solution in order to investigate the SERS effect induced by different numbers of PE layers in each sample. The Surface Enhancement Raman Spectroscopy (SERS) effect correlating to the number of PE layers deposited on the samples has been shown. This effect is explained in terms of an increase in the interaction between the photon of the laser source and the plasmonic band of the GNPs due to a change of the permittivity of the surrounding medium around the GNPs. The trends of the ratios of the intensities of the Raman bands of the acetic acid solution (acetic acid and water molecules) on the band at 1098 cm1 ascribed to the substrates increase, and the number of PE layers increases. Full article
(This article belongs to the Special Issue High-Efficiency Surface-Enhanced Raman Scattering Biosensing)
Show Figures

Figure 1

Figure 1
<p>Schematic view of the four samples: GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>0</mn> </msub> </semantics></math> (glass substrate/GNPs), GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>3</mn> </msub> </semantics></math> (glass substrate/GNPs/3 layers of PE material), GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>11</mn> </msub> </semantics></math> (glass substrate/GNPs/11 layers of PE material), and GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>21</mn> </msub> </semantics></math> (glass substrate/GNPs/21 layer of PE material).</p>
Full article ">Figure 2
<p>Localized surface plasmon band (LSPR) of the GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>0</mn> </msub> </semantics></math>, GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>3</mn> </msub> </semantics></math>, GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>11</mn> </msub> </semantics></math>, and GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>21</mn> </msub> </semantics></math> sample.</p>
Full article ">Figure 3
<p>RepresentativeRaman spectra collected on the 10% <span class="html-italic">v</span>/<span class="html-italic">v</span> acetic acid solution (<b>a</b>) in a cuvette and (<b>b</b>) on the top surfaces of the glass substrate.</p>
Full article ">Figure 4
<p>Representative Raman spectra of 10% <span class="html-italic">v</span>/<span class="html-italic">v</span> acetic acid solution in the ranges between 200 and 1300 cm<math display="inline"><semantics> <msup> <mrow/> <mrow> <mo>−</mo> <mn>1</mn> </mrow> </msup> </semantics></math> and between 2800 and 3800 cm<math display="inline"><semantics> <msup> <mrow/> <mrow> <mo>−</mo> <mn>1</mn> </mrow> </msup> </semantics></math> on (<b>a</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>0</mn> </msub> </semantics></math> (glass substrate/GNPs), (<b>b</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>3</mn> </msub> </semantics></math> (glass substrate/GNPs/3 layers of PE material), (<b>c</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>11</mn> </msub> </semantics></math> (glass substrate/GNPs/11 layers of PE material), and (<b>d</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>21</mn> </msub> </semantics></math> (glass substrate/GNPs/21 layer of PE material). The modes of the substrates are in red, while the modes of the solutions are in black. The peaks have been identified by comparisons with other spectra in the literature.</p>
Full article ">Figure 5
<p>Fitting procedure in ranges between 800 and 1250 cm<math display="inline"><semantics> <msup> <mrow/> <mrow> <mo>−</mo> <mn>1</mn> </mrow> </msup> </semantics></math> (on the left) and between 2800 and 3800 cm<math display="inline"><semantics> <msup> <mrow/> <mrow> <mo>−</mo> <mn>1</mn> </mrow> </msup> </semantics></math> (on the right) of the Raman bands of 10% <span class="html-italic">v</span>/<span class="html-italic">v</span> acetic acid solution deposited on (<b>a</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>0</mn> </msub> </semantics></math> (glass substrate/GNPs), (<b>b</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>3</mn> </msub> </semantics></math> (glass substrate/GNPs/3 layers of PE material), (<b>c</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>11</mn> </msub> </semantics></math> (glass substrate/GNPs/11 layers of PE material), and (<b>d</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>21</mn> </msub> </semantics></math> (glass substrate/GNPs/21 layer of PE material). The grey line is the experimental spectra, the green lines are single fitting curves, and the red line is the total fitting curve.</p>
Full article ">Figure 6
<p>Trends, as a function of the number of the PE layers, of the ratios of intensities bands that fall at frequencies indicated as subscripts in I<math display="inline"><semantics> <msub> <mrow/> <mn>890</mn> </msub> </semantics></math>/I<math display="inline"><semantics> <msub> <mrow/> <mn>1098</mn> </msub> </semantics></math> (<b>a</b>), I<math display="inline"><semantics> <msub> <mrow/> <mn>2950</mn> </msub> </semantics></math>/I<math display="inline"><semantics> <msub> <mrow/> <mn>1098</mn> </msub> </semantics></math> (<b>b</b>), and (I<math display="inline"><semantics> <msub> <mrow/> <mn>3186</mn> </msub> </semantics></math> + I<math display="inline"><semantics> <msub> <mrow/> <mn>3307</mn> </msub> </semantics></math> + I<math display="inline"><semantics> <msub> <mrow/> <mn>3458</mn> </msub> </semantics></math> + I<math display="inline"><semantics> <msub> <mrow/> <mn>3593</mn> </msub> </semantics></math>)/I<math display="inline"><semantics> <msub> <mrow/> <mn>1098</mn> </msub> </semantics></math> (<b>c</b>) and that have been calculated by the fitting procedure shown above (see <a href="#biosensors-12-00895-f004" class="html-fig">Figure 4</a>).</p>
Full article ">Figure 7
<p>Finite element method (FEM) simulations reporting the local enhancement near-field around (<b>a</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>0</mn> </msub> </semantics></math>, (<b>b</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>3</mn> </msub> </semantics></math>, (<b>c</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>11</mn> </msub> </semantics></math>, and (<b>d</b>) GGPE<math display="inline"><semantics> <msub> <mrow/> <mn>21</mn> </msub> </semantics></math>.</p>
Full article ">
22 pages, 4735 KiB  
Review
Emerging Biosensing Technologies towards Early Sepsis Diagnosis and Management
by Andrea Bonini, Angela Gilda Carota, Noemi Poma, Federico Maria Vivaldi, Denise Biagini, Daria Bottai, Alessio Lenzi, Arianna Tavanti, Fabio Di Francesco and Tommaso Lomonaco
Biosensors 2022, 12(10), 894; https://doi.org/10.3390/bios12100894 - 18 Oct 2022
Cited by 8 | Viewed by 3270
Abstract
Sepsis is defined as a systemic inflammatory dysfunction strictly associated with infectious diseases, which represents an important health issue whose incidence is continuously increasing worldwide. Nowadays, sepsis is considered as one of the main causes of death that mainly affects critically ill patients [...] Read more.
Sepsis is defined as a systemic inflammatory dysfunction strictly associated with infectious diseases, which represents an important health issue whose incidence is continuously increasing worldwide. Nowadays, sepsis is considered as one of the main causes of death that mainly affects critically ill patients in clinical settings, with a higher prevalence in low-income countries. Currently, sepsis management still represents an important challenge, since the use of traditional techniques for the diagnosis does not provide a rapid response, which is crucial for an effective infection management. Biosensing systems represent a valid alternative due to their characteristics such as low cost, portability, low response time, ease of use and suitability for point of care/need applications. This review provides an overview of the infectious agents associated with the development of sepsis and the host biomarkers suitable for diagnosis and prognosis. Special focus is given to the new emerging biosensing technologies using electrochemical and optical transduction techniques for sepsis diagnosis and management. Full article
(This article belongs to the Special Issue Feature Review Papers for Biosensors)
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) comparison of the schematic flow between sepsis and normal host response to an infection; (<b>b</b>) geographical incidence of sepsis worldwide [<a href="#B9-biosensors-12-00894" class="html-bibr">9</a>]. Copyright © 1969, Elsevier.</p>
Full article ">Figure 2
<p>Traditional techniques to detect pathogens and biomarkers associated to sepsis: (<b>a</b>) Blood culture-based approach for microorganism identification; (<b>b</b>) DNA/RNA amplification-based technique (RT-PCR); (<b>c</b>) Matrix-Assisted Laser Desorption/Ionization Time-of-Flight Mass Spectrometry; (<b>d</b>) Immunoenzymatically serological assay for biomarkers detection.</p>
Full article ">Figure 3
<p>Examples of electrochemical biosensors approaches for sepsis diagnosis: (<b>a</b>) representation of all components of point of care biosensor for CRP detection, reprinted from [<a href="#B68-biosensors-12-00894" class="html-bibr">68</a>] Creative common CC BY 4.0; (<b>b</b>) assay schematic of CRISPR/cas12a based biosensor for <span class="html-italic">E. coli</span> and <span class="html-italic">S. aureus</span> detection, reprinted with permission of [<a href="#B81-biosensors-12-00894" class="html-bibr">81</a>], copyright 2021 Elsevier B.V.; (<b>c</b>) fabrication steps and electrode schematic for PCT and CRP detection, reprinted with permission [<a href="#B73-biosensors-12-00894" class="html-bibr">73</a>], copyright 2021 Wiley-VCH GmbH.</p>
Full article ">Figure 4
<p>Other examples of electrochemical biosensor approaches for sepsis diagnosis: (<b>a</b>) magnetic micromotors-based assay immunoassay for CRP detection, reprinted with permission [<a href="#B67-biosensors-12-00894" class="html-bibr">67</a>] Copyright 2020 Elsevier B.V; (<b>b</b>) assay schematic of point of care integrated sensor for cytokines detection, reprinted with permission of [<a href="#B76-biosensors-12-00894" class="html-bibr">76</a>] Copyright © 2018, American Chemical Society.</p>
Full article ">Figure 5
<p>Examples of optical biosensor approaches for sepsis diagnosis: (<b>a</b>) steps for fabrication of the fiber optic SPR biosensor fort the CRP detection, reprinted with permission of [<a href="#B89-biosensors-12-00894" class="html-bibr">89</a>] Creative common CC BY 4.0; (<b>b</b>) schematic sandwich assay for PCT detection, reprinted with permission of [<a href="#B90-biosensors-12-00894" class="html-bibr">90</a>] Copyright 2019 Elsevier B.V.; (<b>c</b>) schematic sensing principle for CRP detection, reprinted from [<a href="#B97-biosensors-12-00894" class="html-bibr">97</a>] Creative common CC BY 4.0; (<b>d</b>) schematic of the aptamer-gold nanoparticle-based assay for the detection of mouse IL-6, reprinted from [<a href="#B91-biosensors-12-00894" class="html-bibr">91</a>] Creative common CC BY 4.0; (<b>e</b>) schematic representation of the method for detect urease positive bacteria involved in sepsis, reprinted with permission of [<a href="#B93-biosensors-12-00894" class="html-bibr">93</a>], Copyright © 2019, American Chemical Society; (<b>f</b>) schematic showing the working principle of selective isolation and detection of bacteria from whole blood involved in sepsis, reprinted [<a href="#B95-biosensors-12-00894" class="html-bibr">95</a>], Creative common CC BY 4.0.</p>
Full article ">
15 pages, 2789 KiB  
Article
A Paper-Based Analytical Device Integrated with Smartphone: Fluorescent and Colorimetric Dual-Mode Detection of β-Glucosidase Activity
by Wei-Yi Zhang, Tao Tian, Li-Jing Peng, Hang-Yu Zhou, Hao Zhang, Hua Chen and Feng-Qing Yang
Biosensors 2022, 12(10), 893; https://doi.org/10.3390/bios12100893 - 18 Oct 2022
Cited by 15 | Viewed by 3012
Abstract
In this work, indoxyl-glucoside was used as the substrate to develop a cost-effective, paper-based analytical device for the fluorescent and colorimetric dual-mode detection of β-glucosidase activity through a smartphone. The β-glucosidase can hydrolyze the colorless substrate indoxyl-glucoside to release indoxyl, which will be [...] Read more.
In this work, indoxyl-glucoside was used as the substrate to develop a cost-effective, paper-based analytical device for the fluorescent and colorimetric dual-mode detection of β-glucosidase activity through a smartphone. The β-glucosidase can hydrolyze the colorless substrate indoxyl-glucoside to release indoxyl, which will be self-oxidized to generate green products in the presence of oxygen. Meanwhile, the green products emit bright blue-green fluorescence under ultraviolet–visible light irradiation at 365 nm. Fluorescent or colorimetric images were obtained by a smartphone, and the red-green-blue channels were analyzed by the Adobe Photoshop to quantify the β-glucosidase activity. Under the optimum conditions, the relative fluorescent and colorimetric signals have a good linear relationship with the activity of β-glucosidase, in the range of 0.01–1.00 U/mL and 0.25–5.00 U/mL, and the limits of detection are 0.005 U/mL and 0.0668 U/mL, respectively. The activities of β-glucosidase in a crude almond sample measured by the fluorescent and colorimetric methods were 23.62 ± 0.53 U/mL and 23.86 ± 0.25 U/mL, respectively. In addition, the spiked recoveries of normal human serum and crude almond samples were between 87.5% and 118.0%. In short, the paper-based device, combined with a smartphone, can provide a simple, environmentally friendly, and low-cost method for the fluorescent and colorimetric dual-mode detection of β-glucosidase activity. Full article
(This article belongs to the Special Issue Paper-Based Biosensors)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of fluorescent and colorimetric dual-mode detection of β-glucosidase, based on the paper-based device using indoxyl-glucoside as a substrate.</p>
Full article ">Figure 2
<p>Structure diagram of homemade camera obscura (<b>A</b>) and paper-based device (<b>B</b>).</p>
Full article ">Figure 3
<p>(<b>A</b>) Reaction mechanism of indoxyl-glucoside in the presence of β-glucosidase. (<b>B</b>) Fluorescent and colorimetric diagrams of paper-based device in the presence of different reagents: (<b>a</b>) pure filter paper, (<b>b</b>) indoxyl-glucoside, (<b>c</b>) β-glucosidase, (<b>d</b>) indoxyl-glucoside and β-glucosidase.</p>
Full article ">Figure 4
<p>Intensity of RGB channels in fluorescent images irradiated by ultraviolet light at 365 nm of the paper-based device with different activity of β-glucosidase.</p>
Full article ">Figure 5
<p>Effect of pH (<b>A</b>), temperature (<b>B</b>), indoxyl-glucoside concentration (<b>C</b>), and reaction time (<b>D</b>) on the fluorescent detection of β-glucosidase by the paper-based device, β-glucosidase = 1.0 U/mL. Inset: effect of temperature on the fluorescent detection of β-glucosidase by the paper-based device, β-glucosidase = 0.0 U/mL.</p>
Full article ">Figure 6
<p>The relative fluorescent value of the paper-based device changed with the activity of β-glucosidase. Inset: calibration plot in the linear range of 0.01–1.00 U/mL. Photographs: fluorescent images of paper-based device at different β-glucosidase activity.</p>
Full article ">Figure 7
<p>(<b>A</b>) Intensity of RGB channels in colorimetric images of the paper-based devices with different activity of β-glucosidase. (<b>B</b>) The relative colorimetric value of the paper-based device is changed with the activity of β-glucosidase. Inset: calibration plot in the linear range of 0.25–5.00 U/mL. Photographs: colorimetric images of paper-based device at different β-glucosidase activity.</p>
Full article ">Figure 8
<p>Selectivity and interference study of the paper-based device for β-glucosidase detection: (<b>A</b>) fluorescence, (<b>B</b>) colorimetry.</p>
Full article ">
30 pages, 5471 KiB  
Review
Recent Advancements in Nanobiosensors: Current Trends, Challenges, Applications, and Future Scope
by Madhusudan B. Kulkarni, Narasimha H. Ayachit and Tejraj M. Aminabhavi
Biosensors 2022, 12(10), 892; https://doi.org/10.3390/bios12100892 - 18 Oct 2022
Cited by 50 | Viewed by 12315
Abstract
In recent years, there has been immense advancement in the development of nanobiosensors as these are a fundamental need of the hour that act as a potential candidate integrated with point-of-care-testing for several applications, such as healthcare, the environment, energy harvesting, electronics, and [...] Read more.
In recent years, there has been immense advancement in the development of nanobiosensors as these are a fundamental need of the hour that act as a potential candidate integrated with point-of-care-testing for several applications, such as healthcare, the environment, energy harvesting, electronics, and the food industry. Nanomaterials have an important part in efficiently sensing bioreceptors such as cells, enzymes, and antibodies to develop biosensors with high selectivity, peculiarity, and sensibility. It is virtually impossible in science and technology to perform any application without nanomaterials. Nanomaterials are distinguished from fine particles used for numerous applications as a result of being unique in properties such as electrical, thermal, chemical, optical, mechanical, and physical. The combination of nanostructured materials and biosensors is generally known as nanobiosensor technology. These miniaturized nanobiosensors are revolutionizing the healthcare domain for sensing, monitoring, and diagnosing pathogens, viruses, and bacteria. However, the conventional approach is time-consuming, expensive, laborious, and requires sophisticated instruments with skilled operators. Further, automating and integrating is quite a challenging process. Thus, there is a considerable demand for the development of nanobiosensors that can be used along with the POCT module for testing real samples. Additionally, with the advent of nano/biotechnology and the impact on designing portable ultrasensitive devices, it can be stated that it is probably one of the most capable ways of overcoming the aforementioned problems concerning the cumulative requirement for the development of a rapid, economical, and highly sensible device for analyzing applications within biomedical diagnostics, energy harvesting, the environment, food and water, agriculture, and the pharmaceutical industry. Full article
(This article belongs to the Special Issue Application of Nanomaterials for Biosensors)
Show Figures

Figure 1

Figure 1
<p>Schematic of 0-D, 1-D, 2-D, and 3-D, nanostructured materials.</p>
Full article ">Figure 2
<p>Various modules of typical biosensors.</p>
Full article ">Figure 3
<p>Schematic representation of several techniques that are used for the synthesis of nanomaterials.</p>
Full article ">Figure 4
<p>Schematic of the top-down and bottom-up approaches of nanomaterials synthesis.</p>
Full article ">Figure 5
<p>Schematic representation of nanobiosensors, from analyte to application.</p>
Full article ">Figure 6
<p>Schematic of several applications of nanobiosensors.</p>
Full article ">Figure 7
<p>Shows the process of nanobiosensors used for various biomedical and diagnostics applications.</p>
Full article ">Figure 8
<p>(<b>A</b>) Structure diagram of an all-in-one system for sensing nucleic acid using LAMP [<a href="#B156-biosensors-12-00892" class="html-bibr">156</a>]. (<b>B</b>) The 3-D-printed microfluidic analytical devices are applied for plasma separation and the iterative fabrication process workflow [<a href="#B157-biosensors-12-00892" class="html-bibr">157</a>].</p>
Full article ">Figure 9
<p>Detailed SWOT analysis of nanomaterials and biosensors.</p>
Full article ">Figure 10
<p>Future scope of nanobiosensors.</p>
Full article ">
17 pages, 3746 KiB  
Article
Detection of SARS-CoV-2 Neutralizing Antibodies in Vaccinated Pregnant Women and Neonates by Using a Lateral Flow Immunoassay Coupled with a Spectrum-Based Reader
by Wei-Chun Chen, Yen-Pin Lin, Chao-Min Cheng, Ching-Fen Shen, Chang-Wei Li, Yu-Kuo Wang, Ting-Ying Shih, Chitsung Hong, Ting-Chang Chang and Ching-Ju Shen
Biosensors 2022, 12(10), 891; https://doi.org/10.3390/bios12100891 - 18 Oct 2022
Cited by 3 | Viewed by 2629
Abstract
The focus of this study was to investigate the detection of neutralizing antibodies (Nabs) in maternal serum and cord blood as the targeted samples by employing a lateral flow immunoassay combined with a spectrum reader (LFI-SR) and the correlation of Nab protection against [...] Read more.
The focus of this study was to investigate the detection of neutralizing antibodies (Nabs) in maternal serum and cord blood as the targeted samples by employing a lateral flow immunoassay combined with a spectrum reader (LFI-SR) and the correlation of Nab protection against different types of SARS-CoV-2. We enrolled 20 pregnant women who were vaccinated with the Moderna (mRNA-1273) vaccine during pregnancy and collected 40 samples during delivery. We used an LFI-SR for the level of spike protein receptor binding domain antibody (SRBD IgG) as Nabs and examined the correlation of the SRBD IgG concentration and Nab inhibition rates (NabIR) via enzyme-linked immunosorbent assays (ELISA). The LFI-SR had high confidence for the SRBD IgG level (p < 0.0001). Better NabIR were found in wild-type SARS-CoV-2 (WT) compared to Delta-type (DT) and Omicron-type (OT). Women with two-dose vaccinations demonstrated greater NabIR than those with a single dose. The cut-off value of the SRBD IgG level by the LFI-SR for NabIR to DT (≥30%; ≥70%) was 60.15 and 150.21 ng/mL for mothers (both p = 0.005), and 156.31 (p = 0.011) and 230.20 ng/mL (p = 0.006) for babies, respectively. An additional vaccine booster may be considered for those mothers with SRBD IgG levels < 60.15 ng/mL, and close protection should be given for those neonates with SRBD IgG levels < 150.21 ng/mL, since there is no available vaccine for them. Full article
(This article belongs to the Special Issue Optical Biosensors for Health, Food and Environment (Bio)markers)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) The colorimetric change mechanism aspect of the lateral flow immunoassay strip. (<b>B</b>) The colorimetric change in a lateral flow rapid test strip for detecting neutralizing antibodies. The control line is shown as a blue dotted line, the test line as a yellow dotted line. (<b>C</b>) The spectrum analyzer used in our study. The analyzer can load a lateral flow immunoassay and can be used in combination with a spectrum-based reader. (<b>D</b>) The reflectance spectra range of the control line and test line. (<b>E</b>) Standard curve of SRBD IgG by a lateral flow immunoassay/spectrum analyzer. SRBD, spike protein receptor binding domain; ** <span class="html-italic">p</span> &lt; 0.01; *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 2
<p>(<b>A</b>) Scatter plot showing the correlation between SRBD IgG by the lateral flow immunoassay/spectrum analyzer and ELISA for maternal serum. (<b>B</b>) Scatter plot showing the correlation between SRBD IgG by the lateral flow immunoassay/spectrum analyzer and ELISA in neonatal cord blood. The red spot (a) in <a href="#app1-biosensors-12-00891" class="html-app">Table S3</a> was case 17 and was censored in the correlation line due to it being an outlier. SRBD, spike protein receptor binding domain; ELISA, enzyme-linked immunosorbent assay. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 3
<p>(<b>A</b>) Scatter plot showing the correlation between SRBD IgG by the lateral flow immunoassay/spectrum analyzer and the neutralizing antibody inhibition rates against wild-type SARS-CoV-2 by ELISA in maternal serum. (<b>B</b>) Scatter plot showing the correlation between SRBD IgG by the lateral flow immunoassay/spectrum analyzer and the neutralizing antibody inhibition rates against wild-type SARS-CoV-2 by ELISA in neonatal cord blood. SRBD, spike protein receptor binding domain; ELISA, enzyme-linked immunosorbent assay. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 4
<p>(<b>A</b>) Scatter plot showing the correlation between SRBD IgG by the lateral flow immunoassay/spectrum analyzer and the neutralizing antibody inhibition rates against Delta-type SARS-CoV-2 by ELISA in maternal serum. (<b>B</b>) Scatter plot showing the correlation between SRBD IgG by the lateral flow immunoassay/spectrum analyzer and the neutralizing antibody inhibition rates against Delta-type SARS-CoV-2 by ELISA in neonatal cord blood. SRBD, spike protein receptor binding domain; ELISA, enzyme-linked immunosorbent assay. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure 5
<p>(<b>A</b>) Scatter plot showing the correlation between SRBD IgG by the lateral flow immunoassay/spectrum analyzer and the neutralizing antibody concentration against wild-type SARS-CoV-2 by ELISA in maternal serum. (<b>B</b>) Scatter plot showing the correlation between SRBD IgG by the lateral flow immunoassay/spectrum analyzer and the neutralizing antibody concentration against wild-type SARS-CoV-2 by ELISA in neonatal cord blood. SRBD, spike protein receptor binding domain; ELISA, enzyme-linked immunosorbent assay. *** <span class="html-italic">p</span> &lt; 0.001.</p>
Full article ">Figure A1
<p>(<b>A</b>) ROC curve of S1RBD in maternal serum by the immunoassay/spectrum analyzer for Nab inhibition rate ≧ 30% for Delta-type SARS-CoV-2. (<b>B</b>) ROC curve of S1RBD in neonatal cord blood by the immunoassay/spectrum analyzer for Nab inhibition rate ≧ 30% for Delta-type SARS-CoV-2. (<b>C</b>) ROC curve of S1RBD in maternal serum by the immunoassay/spectrum analyzer for Nab inhibition rate ≧ 70% for Delta-type SARS-CoV-2. (<b>D</b>) ROC curve of S1RBD in neonatal cord blood by the immunoassay/spectrum analyzer for Nab inhibition rate ≧ 70% for Delta-type SARS-CoV-2. ROC curve, receiver operating characteristic curve; SRBD, spike protein receptor binding domain; AUC, area under curve.</p>
Full article ">Figure A2
<p>(<b>A</b>) Scatter plot showing the correlation between SRBD IgG by the lateral flow immunoassay/spectrum analyzer and the neutralizing antibody inhibition rates against Omicron-type SARS-CoV-2 by ELISA in maternal serum. (<b>B</b>) Scatter plot showing the correlation between SRBD IgG by the lateral flow immunoassay/spectrum analyzer and the neutralizing antibody inhibition rates against Omicron-type SARS-CoV-2 by ELISA in neonatal cord blood. SRBD, spike protein receptor binding domain; ELISA, enzyme-linked immunosorbent assay.</p>
Full article ">
17 pages, 6675 KiB  
Review
SARS-CoV-2-on-Chip for Long COVID Management
by Jayesh Cherusseri, Claire Mary Savio, Mohammad Khalid, Vishal Chaudhary, Arshid Numan, Sreekanth J. Varma, Amrutha Menon and Ajeet Kaushik
Biosensors 2022, 12(10), 890; https://doi.org/10.3390/bios12100890 - 18 Oct 2022
Cited by 20 | Viewed by 3897
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a “wicked evil” in this century due to its extended progression and huge human mortalities. Although the diagnosis of SARS-CoV-2 viral infection is made simple and practical by employing reverse transcription polymerase chain reaction [...] Read more.
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has become a “wicked evil” in this century due to its extended progression and huge human mortalities. Although the diagnosis of SARS-CoV-2 viral infection is made simple and practical by employing reverse transcription polymerase chain reaction (RT-PCR) investigation, the process is costly, complex, time-consuming, and requires experts for testing and the constraints of a laboratory. Therefore, these challenges have raised the paradigm of on-site portable biosensors on a single chip, which reduces human resources and enables remote access to minimize the overwhelming burden on the existing global healthcare sector. This article reviews the recent advancements in biosensors for long coronavirus disease (COVID) management using a multitude of devices, such as point-of-care biosensors and lab-on-chip biosensors. Furthermore, it details the shift in the paradigm of SARS-CoV-2-on-chip biosensors from the laboratory to on-site detection with intelligent and economical operation, representing near-future diagnostic technologies for public health emergency management. Full article
Show Figures

Figure 1

Figure 1
<p>(<b>a</b>) Process of detection and treatment by existing diagnostic techniques and point-of-care tests. Reprinted with permission from Springer Nature [<a href="#B4-biosensors-12-00890" class="html-bibr">4</a>], Copyright (2020). (<b>b</b>) Various methods available to diagnose COVID-19. Reprinted with permission from Springer Nature [<a href="#B5-biosensors-12-00890" class="html-bibr">5</a>], Copyright (2020).</p>
Full article ">Figure 2
<p>Paper-based molecular diagnostic device. Reprinted with permission from Elsevier B.V. [<a href="#B10-biosensors-12-00890" class="html-bibr">10</a>], Copyright (2015).</p>
Full article ">Figure 3
<p>Schematic diagram depicts the advantages of lab-on-chip devices for COVID-19 testing. Reprinted with permission from Springer Nature [<a href="#B4-biosensors-12-00890" class="html-bibr">4</a>], Copyright (2020).</p>
Full article ">Figure 4
<p>Structure and binding of SARS-CoV-2 virus to human cells. Reproduced with permission from [<a href="#B20-biosensors-12-00890" class="html-bibr">20</a>], Creative Commons.</p>
Full article ">Figure 5
<p>Different types of RNA detection methods, such as (<b>a</b>) RT-PCR testing and (<b>b</b>) SARS-CoV-2 RT-LAMP testing. Reprinted with permission from Springer Nature [<a href="#B32-biosensors-12-00890" class="html-bibr">32</a>], Copyright (2021).</p>
Full article ">Figure 6
<p>Antigen detection associated with the SARS-CoV-2 virus. Reprinted with permission from Springer Nature [<a href="#B32-biosensors-12-00890" class="html-bibr">32</a>], Copyright (2021).</p>
Full article ">Figure 7
<p>Schematic diagram representing the “ASSURED” guidelines featuring point-of-care devices. Reproduced with permi ssion from [<a href="#B39-biosensors-12-00890" class="html-bibr">39</a>], Creative Commons.</p>
Full article ">Figure 8
<p>Schematic diagram of a point-of-care test. Reproduced with permission from [<a href="#B39-biosensors-12-00890" class="html-bibr">39</a>]. Reproduced with permission from [<a href="#B39-biosensors-12-00890" class="html-bibr">39</a>], Creative Commons.</p>
Full article ">Figure 9
<p>Overview of a rapid diagnostic serological test. Reprinted with permission from [<a href="#B45-biosensors-12-00890" class="html-bibr">45</a>] under a Creative Commons Attribution License.</p>
Full article ">Figure 10
<p>A schematic showing the concept of μ-PAD printing. Reprinted with permission from [<a href="#B53-biosensors-12-00890" class="html-bibr">53</a>] under the Creative Commons Attribution License CC-BY-NC-ND.</p>
Full article ">Figure 11
<p>(<b>A</b>) Various components of a COVID-19 ePAD; SARS-CoV-2 antibody detection principle (<b>B</b>); and detection procedure (<b>C</b>). Reprinted with permission from Elsevier B.V. [<a href="#B57-biosensors-12-00890" class="html-bibr">57</a>], Copyright (2020).</p>
Full article ">Figure 12
<p>Schematic diagram of the LFIA calorimetric test strips. Reprinted with permission from Elsevier B.V. [<a href="#B57-biosensors-12-00890" class="html-bibr">57</a>], Copyright (2020).</p>
Full article ">
30 pages, 4893 KiB  
Review
Progress in Data Acquisition of Wearable Sensors
by Zixuan Liu, Jingjing Kong, Menglong Qu, Guangxin Zhao and Cheng Zhang
Biosensors 2022, 12(10), 889; https://doi.org/10.3390/bios12100889 - 18 Oct 2022
Cited by 8 | Viewed by 4193
Abstract
Wearable sensors have demonstrated wide applications from medical treatment, health monitoring to real-time tracking, human-machine interface, smart home, and motion capture because of the capability of in situ and online monitoring. Data acquisition is extremely important for wearable sensors, including modules of probes, [...] Read more.
Wearable sensors have demonstrated wide applications from medical treatment, health monitoring to real-time tracking, human-machine interface, smart home, and motion capture because of the capability of in situ and online monitoring. Data acquisition is extremely important for wearable sensors, including modules of probes, signal conditioning, and analog-to-digital conversion. However, signal conditioning, analog-to-digital conversion, and data transmission have received less attention than probes, especially flexible sensing materials, in research on wearable sensors. Here, as a supplement, this paper systematically reviews the recent progress of characteristics, applications, and optimizations of transistor amplifiers and typical filters in signal conditioning, and mainstream analog-to-digital conversion strategies. Moreover, possible research directions on the data acquisition of wearable sensors are discussed at the end of the paper. Full article
(This article belongs to the Section Intelligent Biosensors and Bio-Signal Processing)
Show Figures

Figure 1

Figure 1
<p>Wearable sensor working processes of wearable sensors.</p>
Full article ">Figure 2
<p>Figure of contents, including amplifiers pursuing high linearity and low power consumption; filters suitable for signal and noise characteristics; high-performance ADCs with both noise rejection and energy efficiency.</p>
Full article ">Figure 3
<p>Transistor structures and optimized designs. (<b>a</b>) The structures of three transistors. Structure of classical insulated gate type FET (i); OTFT (ii) [<a href="#B34-biosensors-12-00889" class="html-bibr">34</a>]; reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License, Copyright 2004, The Authors, published by Elsevier; and OECT (iii) [<a href="#B35-biosensors-12-00889" class="html-bibr">35</a>]; reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License, Copyright 2013, The Authors, published by Springer Nature. (<b>b</b>) Sub-thermionic organic transistors. (i) False-color cross-section transmission electron microscopy image of the transistor. (ii) Comparison of intrinsic gain among different TFT technologies. (iii) An array of flexible devices that fit on your fingertips [<a href="#B36-biosensors-12-00889" class="html-bibr">36</a>]. Reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License, Copyright 2021, The Authors, published by Springer Nature. (<b>c</b>) Nanoscale flexible OECT (i) with high on/off current ratio up to 3 × 10<sup>8</sup> (ii) and sub-threshold swing down to 70 mV/decade (iii) [<a href="#B37-biosensors-12-00889" class="html-bibr">37</a>]. Reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License, Copyright 2022, The Authors, published by AAAS. (<b>d</b>) Vertical OECT-based inverter. (i) Schematic cross-section of the complementary inverter. (ii) The wiring diagram when the cofacial pair inverter amplifies the ECG voltage. (iii) Comparison of the recorded ECG signal between from the cofacial pair inverter and from medical electrodes using a benchtop digital multimeter [<a href="#B38-biosensors-12-00889" class="html-bibr">38</a>]. Reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License, Copyright 2021, The Authors, published by AAAS.</p>
Full article ">Figure 4
<p>CMRR enhancement approaches of amplifiers. (<b>a</b>) PMC technology to reduce OTFT mismatch. (i) The parallel transistors are electrically isolated prior to connection, and electrical connection is provided by interconnecting metal lines through the insulating layer. (ii) The PMC process which is based on the compensation method in ref [<a href="#B43-biosensors-12-00889" class="html-bibr">43</a>]. (iii) Output characteristics of two-sided OTFTs before and after PMC [<a href="#B58-biosensors-12-00889" class="html-bibr">58</a>]. Reproduced with permission, Copyright 2019, Springer Nature. (<b>b</b>) CMFF compensated bias current design. (i) First stage of fully differential amplifier. (ii) Second stage differential to single ended amplifier. (iii) Schematic diagram of the replica bias circuit used to set the common mode current of the input stage [<a href="#B44-biosensors-12-00889" class="html-bibr">44</a>]. Adapted under the terms of the CC-BY 4.0 Creative Commons Attribution License. Copyright 2020, The Authors, published by MDPI.</p>
Full article ">Figure 5
<p>Characteristics and applications of three different types of filters. (<b>a</b>) Filter frequencies of Butterworth filter (i), Elliptic filter (ii), Chebyshev I filter (iii), and Chebyshev II filter (iv). (<b>b</b>) Wearable devices with different orders of Butterworth filters. (i) Gait and dynamic balance sensors using second-order Butterworth filters [<a href="#B66-biosensors-12-00889" class="html-bibr">66</a>]. Reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License. Copyright 2019, The Authors, published by IEEE. (ii) Eye-tracking device with third-order Butterworth filter [<a href="#B67-biosensors-12-00889" class="html-bibr">67</a>]. Reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License. Copyright 2020, The Authors, published by AAAS. (iii) Physiological monitoring clothing for firefighters using a fourth-order Butterworth filter [<a href="#B68-biosensors-12-00889" class="html-bibr">68</a>]. Reproduced with permission. Copyright 2018, IEEE. (iv) In-ear PPG monitoring device with a fourth-order Butterworth filter. (v) Heart rate estimation during slow walking [<a href="#B69-biosensors-12-00889" class="html-bibr">69</a>]. Reproduced with permission. Copyright 2020, IEEE. (<b>c</b>) An ambulatory ECG monitoring system. (i) Front view of the electrocardiograph. ii. The response of Chebyshev filter at high pass filtering [<a href="#B70-biosensors-12-00889" class="html-bibr">70</a>]. Reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License. Copyright 2021, The Authors, published by MDPI. (<b>d</b>) Biopotential signal detection front-end with Elliptic filter. The block diagram (i), magnitude response of the proposed notch filter (ii), and low pass filter (iii) [<a href="#B71-biosensors-12-00889" class="html-bibr">71</a>]. Reproduced with permission, Copyright 2020, IEEE.</p>
Full article ">Figure 6
<p>Application and innovation of filters. (<b>a</b>) Insulated EMG sensing. (i) The myoelectric sensor is fixed on the human forearm through a cuff. Butterworth-filtered (ii) and Chebyshev-filtered (iii) signal at contracted muscle. (iv) Qualitative sketches of systolic EMG and typical motion artifacts in the lower range. (v) Difference between artifact and EMG signal losses [<a href="#B78-biosensors-12-00889" class="html-bibr">78</a>]. Reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License. Copyright 2019, The Authors, published by MDPI. (<b>b</b>) Optimal filter selection in PPG. (i) SQI for all types of filters. The excellent (ii) and unfit (iii) PPG signal processing comparison between Butterworth filter (blue) and Chebyshev filter (red) [<a href="#B79-biosensors-12-00889" class="html-bibr">79</a>]. Reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License. Copyright 2018, The Authors, published by Springer Nature. (<b>c</b>) The use of PPG for assessing hypertension. Difference between PAT and PTT (i) and the filters’ impact on PPG morphology (ii) [<a href="#B90-biosensors-12-00889" class="html-bibr">90</a>]. Adapted under the terms of the CC-BY 4.0 Creative Commons Attribution License. Copyright 2021, The Authors, published by Springer Nature. (<b>d</b>) EMG signal processing instrument (i) with adaptive filter. Comparison of EMG signal before (ii) and after (iii) processing and noisy EMG signal before (iv) and after processing (v) [<a href="#B85-biosensors-12-00889" class="html-bibr">85</a>]. Reproduced with permission, Copyright 2019, IEEE.</p>
Full article ">Figure 7
<p>Block diagrams of five types of ADCs. (i) Flash ADC [<a href="#B96-biosensors-12-00889" class="html-bibr">96</a>]. (ii) Pipelined ADC. (iii) Dual slope ADC. (iv) N-order SAR ADC [<a href="#B96-biosensors-12-00889" class="html-bibr">96</a>]. Reproduced with permission. Copyright 2016, IEEE (v) Sigma-delta ADC.</p>
Full article ">Figure 8
<p>ADC linearity enhancement and energy efficient design. (<b>a</b>) Capacitor mismatch solution with a SAR ADC structure which has 14-bit capacitor and resistor combination (i) that break the original INL (ii) for a smaller margin of error (iii). In addition, the improvement in SNDR (iv) and SFDR (v) it brings is considerable [<a href="#B110-biosensors-12-00889" class="html-bibr">110</a>]. Reproduced with permission, Copyright 2018, The Authors, published by IEEE. (<b>b</b>) On the basis of (<b>a</b>), the capacitor allocation is improved (ii), and the optimization strategy is added (iii). In addition, it shows more ideal SNDR (iv) and SFDR (v) [<a href="#B111-biosensors-12-00889" class="html-bibr">111</a>]. Reproduced under the terms of the CC-BY 4.0 Creative Commons Attribution License, Copyright 2018, The Authors, published by IEEE. (<b>c</b>) Special CS design. DAC array with 12-bit CS-SAR ADC, with sampling cycles from φ1 to φ4 (i) and quantization cycle φ5 (ii), achieved the power consumption; (iv) is much lower than that of Nyquist mode (iii) [<a href="#B116-biosensors-12-00889" class="html-bibr">116</a>]. Reproduced with permission, Copyright 2017, IEEE. (<b>d</b>) SAR ADC structure (i) with dynamic comparator, bypass-switching process (ii), and code recovery operation. Both bypass-switching technology (iii) and dynamic comparison technology (iv) can significantly improve energy efficiency [<a href="#B117-biosensors-12-00889" class="html-bibr">117</a>]. Reproduced with permission, Copyright 2018, IEEE.</p>
Full article ">
16 pages, 5058 KiB  
Article
Kinetics of Drug Molecule Interactions with a Newly Developed Nano-Gold-Modified Spike Protein Electrochemical Receptor Sensor
by Dingqiang Lu, Danyang Liu, Xinqian Wang, Yujiao Liu, Yixuan Liu, Ruijuan Ren and Guangchang Pang
Biosensors 2022, 12(10), 888; https://doi.org/10.3390/bios12100888 - 17 Oct 2022
Cited by 2 | Viewed by 2138
Abstract
In March 2020, the World Health Organization (WHO) declared COVID-19 a pandemic, and the spike protein has been reported to be an important drug target for anti-COVID-19 treatment. As such, in this study, we successfully developed a novel electrochemical receptor biosensor by immobilizing [...] Read more.
In March 2020, the World Health Organization (WHO) declared COVID-19 a pandemic, and the spike protein has been reported to be an important drug target for anti-COVID-19 treatment. As such, in this study, we successfully developed a novel electrochemical receptor biosensor by immobilizing the SARS-CoV-2 spike protein and using AuNPs-HRP as an electrochemical signal amplification system. Moreover, the time-current method was used to quantify seven antiviral drug compounds, such as arbidol and chloroquine diphosphate. The results show that the spike protein and the drugs are linearly correlated within a certain concentration range and that the detection sensitivity of the sensor is extremely high. In the low concentration range of linear response, the kinetics of receptor–ligand interactions are similar to that of an enzymatic reaction. Among the investigated drug molecules, bromhexine exhibits the smallest Ka value, and thus, is most sensitively detected by the sensor. Hydroxychloroquine exhibits the largest Ka value. Molecular docking simulations of the spike protein with six small-molecule drugs show that residues of this protein, such as Asp, Trp, Asn, and Gln, form hydrogen bonds with the -OH or -NH2 groups on the branched chains of small-molecule drugs. The electrochemical receptor biosensor can directly quantify the interaction between the spike protein and drugs such as abidor and hydroxychloroquine and perform kinetic studies with a limit of detection 3.3 × 1020 mol/L, which provides a new research method and idea for receptor–ligand interactions and pharmacodynamic evaluation. Full article
(This article belongs to the Special Issue Nanoengineering for Advanced Biosensors)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of the fabrication process of the spike protein receptor biosensor.</p>
Full article ">Figure 2
<p>(<b>a</b>) Cyclic voltammetry characterization of spike protein receptor sensor electrode assembly modifications. (<b>b</b>) the impact of thionine on electrochemical properties characterized by cyclic voltammetry; curve 1 is the cyclic voltammogram of Thi-Chit/GCE and curve 2 is the cyclic voltammogram of Chit/GCE. The peak current increases gradually when thi is present, indicating that thi facilitates the transfer of electrons between the electrode and the substrate. Cyclic voltammetry was performed in 1 × 10<sup>−</sup><sup>3</sup> mol/L K<sub>3</sub>Fe(CN)<sub>6</sub> (containing 0.20 mol/L KNO<sub>3</sub>) solution with a scanning potential range of −0.1 to 0.6 V and a scanning rate of 50 mV/s, using a three-electrode system; the prepared S protein receptor sensor was used as the working electrode, Ag/AgCl electrode as the reference electrode and platinum wire electrode as the counter electrode.</p>
Full article ">Figure 3
<p>SEM of sensor assembly process: (<b>a</b>) GCE; (<b>b</b>) Thi-Chit/GCE; (<b>c</b>) AuNPs-HRP/Thi-Chit/GCE; (<b>d</b>) spike protein/AuNPs-HRP/Thi-Chit/GCE; (<b>e</b>) BSA/spike protein/AuNPs-HRP/Thi-Chit/GCE.</p>
Full article ">Figure 4
<p>Rate of change of response current in the detection range (the concentration range of Shuanghuanglian oral liquid, arbidol, and lopinavir were 1 × 10<sup>−</sup><sup>20</sup> mol/L, ribavirin and chloroquine diphosphate were 1 × 10<sup>−</sup><sup>19</sup> mol/L, hydroxychloroquine was 1 × 10<sup>−</sup><sup>14</sup> mol/L, bromhexine was 1 × 10<sup>−</sup><sup>21</sup> mol/L, and KCl was 1 × 10<sup>−</sup><sup>15</sup> mol/L as control).</p>
Full article ">Figure 5
<p>Variation of the rate of change of the response current as a function of drug concentration: (<b>A</b>) Shuanghuanglian oral liquid 10<sup>−</sup><sup>20</sup>–10<sup>−</sup><sup>18</sup> mol/L; (<b>B</b>) abridol 10<sup>−</sup><sup>20</sup>–10<sup>−</sup><sup>18</sup> mol/L; (<b>C</b>) chloroquine diphosphate 10<sup>−</sup><sup>19</sup>–10<sup>−</sup><sup>17</sup> mol/L; (<b>D</b>) lopinavir 10<sup>−</sup><sup>19</sup>–10<sup>−</sup><sup>17</sup> mol/L; (<b>E</b>) ribavirin 10<sup>−</sup><sup>19</sup>–10<sup>−</sup><sup>17</sup> mol/L; (<b>F</b>) bromhexine 10<sup>−</sup><sup>21</sup>–10<sup>−</sup><sup>19</sup> mol/L; and (<b>G</b>) hydroxychloroquine 10<sup>−</sup><sup>14</sup>–10<sup>−</sup><sup>12</sup> mol/L; (<b>H</b>) KCl 10<sup>−</sup><sup>15</sup>–10<sup>−</sup><sup>13</sup> mol/L as control.</p>
Full article ">Figure 6
<p>Fitted response curves: (<b>A</b>) Shuanghuanglian oral liquid 10<sup>−</sup><sup>20</sup>–10<sup>−</sup><sup>18</sup> mol/L; (<b>B</b>) abridol 10<sup>−</sup><sup>20</sup>–10<sup>−</sup><sup>18</sup> mol/L; (<b>C</b>) chloroquine diphosphate 10<sup>−</sup><sup>19</sup>–10<sup>−</sup><sup>17</sup> mol/L; (<b>D</b>) lopinavir 10<sup>−</sup><sup>19</sup>–10<sup>−</sup><sup>17</sup> mol/L; (<b>E</b>) ribavirin 10<sup>−</sup><sup>19</sup>–10<sup>−</sup><sup>17</sup> mol/L; (<b>F</b>) bromhexine 10<sup>−</sup><sup>21</sup>–10<sup>−</sup><sup>19</sup> mol/L; and (<b>G</b>) hydroxychloroquine 10<sup>−</sup><sup>14</sup>–10<sup>−</sup><sup>12</sup> mol/L (<b>H</b>) KCl 10<sup>−</sup><sup>15</sup>–10<sup>−</sup><sup>13</sup> mol/L as control.</p>
Full article ">Figure 7
<p>Double reciprocal plots of the fitted response curves of (<b>A</b>) Shuanghuanglian oral liquid 10<sup>−</sup><sup>20</sup>–10<sup>−</sup><sup>18</sup> mol/L; (<b>B</b>) abridol 10<sup>−</sup><sup>20</sup>–10<sup>−</sup><sup>18</sup> mol/L; (<b>C</b>) chloroquine diphosphate 10<sup>−</sup><sup>19</sup>–10<sup>−</sup><sup>17</sup> mol/L; (<b>D</b>) lopinavir 10<sup>−</sup><sup>19</sup>–10<sup>−</sup><sup>17</sup> mol/L; (<b>E</b>) ribavirin 10<sup>−</sup><sup>19</sup>–10<sup>−</sup><sup>17</sup> mol/L; (<b>F</b>) bromhexine 10<sup>−</sup><sup>21</sup>–10<sup>−</sup><sup>19</sup> mol/L; and (<b>G</b>) hydroxychloroquine 10<sup>−</sup><sup>14</sup>–10<sup>−</sup><sup>12</sup> mol/L.</p>
Full article ">Figure 8
<p>3D schematic diagram of the molecular docking simulation of the spike protein with (<b>A</b>) lopinavir, (<b>B</b>) chloroquine diphosphate, (<b>C</b>) abridol, (<b>D</b>) hydroxychloroquine, (<b>E</b>) ribavirin, and (<b>F</b>) bromhexine.</p>
Full article ">
10 pages, 3023 KiB  
Article
A Sensitive Aptamer Fluorescence Anisotropy Sensor for Cd2+ Using Affinity-Enhanced Aptamers with Phosphorothioate Modification
by Hao Yu and Qiang Zhao
Biosensors 2022, 12(10), 887; https://doi.org/10.3390/bios12100887 - 17 Oct 2022
Cited by 8 | Viewed by 2519
Abstract
Rapid and sensitive detection of heavy metal cadmium ions (Cd2+) is of great significance to food safety and environmental monitoring, as Cd2+ contamination and exposure cause serious health risk. In this study we demonstrated an aptamer-based fluorescence anisotropy (FA) sensor [...] Read more.
Rapid and sensitive detection of heavy metal cadmium ions (Cd2+) is of great significance to food safety and environmental monitoring, as Cd2+ contamination and exposure cause serious health risk. In this study we demonstrated an aptamer-based fluorescence anisotropy (FA) sensor for Cd2+ with a single tetramethylrhodamine (TMR)-labeled 15-mer Cd2+ binding aptamer (CBA15), integrating the strengths of aptamers as affinity recognition elements for preparation, stability, and modification, and the advantages of FA for signaling in terms of sensitivity, simplicity, reproducibility, and high throughput. In this sensor, the Cd2+-binding-induced aptamer structure change provoked significant alteration of FA responses. To acquire better sensing performance, we further introduced single phosphorothioate (PS) modification of CBA15 at a specific phosphate backbone position, to enhance aptamer affinity by possible strong interaction between sulfur and Cd2+. The aptamer with PS modification at the third guanine (G) nucleotide (CBA15-G3S) had four times higher affinity than CBA15. Using as an aptamer probe CBA15-G3S with a TMR label at the 12th T, we achieved sensitive selective FA detection of Cd2+, with a detection limit of 6.1 nM Cd2+. This aptamer-based FA sensor works in a direct format for detection without need for labeling Cd2+, overcoming the limitations of traditional competitive immuno-FA assay using antibodies and fluorescently labeled Cd2+. This FA method enabled the detection of Cd2+ in real water samples, showing broad application potential. Full article
(This article belongs to the Special Issue Label-Free Biosensor)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Schematic diagram of fluorescence anisotropy sensor for Cd<sup>2+</sup> using TMR-labeled aptamers. (<b>B</b>) The predicted secondary structure of the aptamer CBA15, with single TMR labeled on the 12th thymine base.</p>
Full article ">Figure 2
<p>(<b>A</b>) FA responses of CBA15 with TMR labels at different sites (20 nM). (<b>B</b>) FA changes of TMR-labeled CBA15 caused by 500 nM Cd<sup>2+</sup>. Δr was obtained by subtracting the FA values of the blank sample from the FA values of 500 nM Cd<sup>2+</sup>.</p>
Full article ">Figure 3
<p>FA changes (Δr) of different TMR-labeled aptamers upon binding with various concentrations of Cd<sup>2+</sup>. Δr was obtained by subtracting the FA values of blank samples from the FA values of various concentrations of Cd<sup>2+</sup>.</p>
Full article ">Figure 4
<p>ITC analysis of aptamers (<b>A</b>) CBA15-G3S and (<b>B</b>) CBA15 with Cd<sup>2+</sup>. The top graph shows raw data for ITC titration, and the bottom graph displays the binding curve obtained by integrating the heats of each spike. The difference between PS modification and the phosphate (PO) group in the backbone of the aptamer is shown.</p>
Full article ">Figure 5
<p>(<b>A</b>) Effects of NaCl concentration on FA responses of blank sample solution and the solution containing 1000 nM Cd<sup>2+</sup> with 20 nM CBA15-G3S-T12-TMR. (<b>B</b>) The relationship between FA changes (Δr) caused by Cd<sup>2+</sup> and NaCl concentration.</p>
Full article ">Figure 6
<p>(<b>A</b>) FA detection of Cd<sup>2+</sup> using CBA15-G3S-T12-TMR. (<b>B</b>) Selectivity test of the FA sensor using CBA15-G3S-T12-TMR for Cd<sup>2+</sup> detection. A 20 nM aptamer probe was used, and the concentrations of tested metal ions were 1000 nM.</p>
Full article ">
13 pages, 3118 KiB  
Article
Colorimetric Detection of Urease-Producing Microbes Using an Ammonia-Responsive Flexible Film Sensor
by Yunsoo Chang, Tae-Eon Park, Seung-Woo Lee and Eun-Hee Lee
Biosensors 2022, 12(10), 886; https://doi.org/10.3390/bios12100886 - 17 Oct 2022
Cited by 5 | Viewed by 3271
Abstract
Urease-producing (ureolytic) microbes have given rise to environmental and public health concerns because they are thought to contribute to emissions of ammonia and to be a virulence factor for infections. Therefore, it is highly important to have the ability to detect such microbes. [...] Read more.
Urease-producing (ureolytic) microbes have given rise to environmental and public health concerns because they are thought to contribute to emissions of ammonia and to be a virulence factor for infections. Therefore, it is highly important to have the ability to detect such microbes. In this study, a poly(dimethylsiloxane) (PDMS)-based colorimetric film sensor was employed for the detection of urease-producing microbes. The sensor was able to detect the enzyme activity of commercially available urease, as the color and absorbance spectrum of the sensor was observed to change upon being exposed to the reaction catalyzed by urease. The ratio of the absorbance of the sensor at 640 nm to that at 460 nm (A640/A460) was linearly proportional to the amount of urease present. The performance of the sensor was validated by the results of a sensitivity and selectivity analysis towards thirteen different bacterial strains. Based on the development of blue color of the sensor, the tested bacteria were classified as strongly positive, moderately positive, weakly positive, or negative urease producers. The response of the sensor to ureolytic bacteria was verified using the urease inhibitor phenyl phosphorodiamidate (PPDA). Additionally, the sensor achieved the selective detection of ureolytic bacteria even in the presence of non-ureolytic bacteria. In addition, a used sensor could be reverted to its original state by being subjected to simple aeration, and in this way the same sensor could be used at least five times for the detection of bacterial urease activity. Full article
(This article belongs to the Section Biosensors and Healthcare)
Show Figures

Figure 1

Figure 1
<p>Schematic diagram of the colorimetric detection of microbial urease activity using the designed film sensor. (<b>a</b>) Composition of the colorimetric film sensor. (<b>b</b>) Reaction mechanism of the enzymatically catalyzed hydrolysis of urea. (<b>c</b>) Schematic of the experimental procedure used for the detection of ureolytic bacteria. (<b>d</b>) Reversible changes in the color of the sensor upon its exposure to ammonia gas and subsequent aeration.</p>
Full article ">Figure 2
<p>Detection of urease enzyme using the colorimetric film sensor. (<b>a</b>) Absorbance spectra and (<b>b</b>) A<sub>640</sub>/A<sub>460</sub> values of sensors exposed to various units of urease. The dashed line depicts the A<sub>640</sub>/A<sub>460</sub> value of the sensor not exposed to urease (0.00 U, negative control). Symbols and error bars indicate the mean and standard deviations of biological triplicates. Insets show photographs of the sensors exposed to the various units of urease.</p>
Full article ">Figure 3
<p>Colorimetric detection of ureolytic activity in <span class="html-italic">Klebsiella pneumoniae</span> (<span class="html-italic">K. pneumoniae</span>). (<b>a</b>) Absorbance spectra and (<b>b</b>) A<sub>640</sub>/A<sub>460</sub> values of the colorimetric film sensors exposed to <span class="html-italic">K. pneumoniae</span> for the indicated incubation times. Symbols and error bars indicate the mean and standard deviations of biological triplicates. Insets show photographs of film sensors at each incubation time.</p>
Full article ">Figure 4
<p>(<b>a</b>–<b>f</b>) Colorimetric detection of ureolytic bacteria using the designed film sensor. The A<sub>640</sub>/A<sub>460</sub> values of sensors exposed to indicated bacteria plotted against incubation duration. Insets show photographs of the sensors. The dashed lines each indicate the A<sub>640</sub>/A<sub>460</sub> value of the sensor in the absence of inoculum (negative control). Symbols and error bars represent the mean and standard deviations of biological triplicates.</p>
Full article ">Figure 5
<p>Verification of ureolytic activity in <span class="html-italic">Klebsiella pneumoniae</span> (<span class="html-italic">K. pneumoniae</span>) using a urease inhibitor, namely phenyl phosphorodiamidate (PPDA). The A<sub>640</sub>/A<sub>460</sub> values and photographs (in insets showing colors) of film sensors in the absence and presence of PPDA are presented.</p>
Full article ">Figure 6
<p>Colorimetric detection of urease activity of <span class="html-italic">Klebsiella pneumoniae</span> (<span class="html-italic">K. pneumoniae</span>) in the presence of non-ureolytic bacteria. The A<sub>640</sub>/A<sub>460</sub> values and photographs of film sensors each acquired after an incubation duration of 10 h. A: <span class="html-italic">Citrobacter koseri</span> (<span class="html-italic">C. koseri</span>) + <span class="html-italic">Escherichia coli</span> (<span class="html-italic">E. coli</span>) + <span class="html-italic">Klebsiella michiganensis</span> (<span class="html-italic">K. michiganensis</span>) + <span class="html-italic">Providencia alcalifaciens</span> (<span class="html-italic">P. alcalifaciens</span>) (non-ureolytic bacteria only), B: <span class="html-italic">K. pneumoniae</span> only, C: <span class="html-italic">K. pneumoniae</span> + <span class="html-italic">C. koseri</span>, D: <span class="html-italic">K. pneumoniae</span> + <span class="html-italic">E. coli</span>, E: <span class="html-italic">K. pneumoniae</span> + <span class="html-italic">K. michiganensis</span>, F: <span class="html-italic">K. pneumoniae</span> + <span class="html-italic">P. alcalifaciens</span>, and G: <span class="html-italic">K. pneumoniae</span> + <span class="html-italic">C. koseri</span> + <span class="html-italic">E. coli</span> + <span class="html-italic">K. michiganensis</span> + <span class="html-italic">P. alcalifaciens</span>. Different lowercases indicate a statistical significance between the means of sample sets at a significance level (<span class="html-italic">p</span>-value) of 0.05.</p>
Full article ">Figure 7
<p>Reusability of the colorimetric film sensor. The A<sub>640</sub>/A<sub>460</sub> values and photographs (showing colors) of a sensor reused five times are presented.</p>
Full article ">
12 pages, 1971 KiB  
Article
Graphene-Binding Peptide in Fusion with SARS-CoV-2 Antigen for Electrochemical Immunosensor Construction
by Beatriz A. Braz, Manuel Hospinal-Santiani, Gustavo Martins, Cristian S. Pinto, Aldo J. G. Zarbin, Breno C. B. Beirão, Vanete Thomaz-Soccol, Márcio F. Bergamini, Luiz H. Marcolino-Junior and Carlos R. Soccol
Biosensors 2022, 12(10), 885; https://doi.org/10.3390/bios12100885 - 17 Oct 2022
Cited by 17 | Viewed by 2530
Abstract
The development of immunosensors to detect antibodies or antigens has stood out in the face of traditional methods for diagnosing emerging diseases such as the one caused by the SARS-CoV-2 virus. The present study reports the construction of a simplified electrochemical immunosensor using [...] Read more.
The development of immunosensors to detect antibodies or antigens has stood out in the face of traditional methods for diagnosing emerging diseases such as the one caused by the SARS-CoV-2 virus. The present study reports the construction of a simplified electrochemical immunosensor using a graphene-binding peptide applied as a recognition site to detect SARS-CoV-2 antibodies. A screen-printed electrode was used for sensor preparation by adding a solution of peptide and reduced graphene oxide (rGO). The peptide-rGO suspension was characterized by scanning electron microscopy (SEM), Raman spectroscopy, and Fourier transform infrared spectroscopy (FT-IR). The electrochemical characterization (electrochemical impedance spectroscopy—EIS, cyclic voltammetry—CV and differential pulse voltammetry—DPV) was performed on the modified electrode. The immunosensor response is based on the decrease in the faradaic signal of an electrochemical probe resulting from immunocomplex formation. Using the best set of experimental conditions, the analytic curve obtained showed a good linear regression (r2 = 0.913) and a limit of detection (LOD) of 0.77 μg mL−1 for antibody detection. The CV and EIS results proved the efficiency of device assembly. The high selectivity of the platform, which can be attributed to the peptide, was demonstrated by the decrease in the current percentage for samples with antibody against the SARS-CoV-2 S protein and the increase in the other antibodies tested. Additionally, the DPV measurements showed a clearly distinguishable response in assays against human serum samples, with sera with a response above 95% being considered negative, whereas responses below this value were considered positive. The diagnostic platform developed with specific peptides is promising and has the potential for application in the diagnosis of other infections that lead to high antibody titers. Full article
(This article belongs to the Special Issue Recent Advances in the Screen-Printed Electrochemical (Bio)sensors)
Show Figures

Figure 1

Figure 1
<p>Schematic illustration of immunosensor step-by-step construction.</p>
Full article ">Figure 2
<p>(<b>a</b>) SEM image, (<b>b</b>) FTIR spectra and (<b>c</b>) Raman spectra of rGO (black line) and mixture peptide + rGO (red line).</p>
Full article ">Figure 3
<p>(<b>a</b>) DPVs obtained for each step of the immunosensor construction with peptide, rGO and BSA, with 1 mmol L<sup>−1</sup> K<sub>3</sub>[Fe(CN)<sub>6</sub>] in PBS 0.1 mol L<sup>−1</sup>. (<b>b</b>) Summarized data obtained from probe current peak to each step of building up the sensor (<span class="html-italic">n</span> = 3, ±SD).</p>
Full article ">Figure 4
<p>(<b>a</b>) Analytical curve obtained through probe current peak intensity vs. antibody concentration (<span class="html-italic">n</span> = 3, ±SD) on 0.1 mol L<sup>−1</sup> PBS, pH 7.4. (<b>b</b>) Summarized DPV assays for negative human serum spiked with different AbS concentrations (<span class="html-italic">n</span> = 3, ± SD).</p>
Full article ">Figure 5
<p>DPV data summarized for selectivity test against YFV antibodies, AbN SARS-CoV-2 and a mixture with AbS, YFV and AbN (<span class="html-italic">n</span> = 3, ± SD).</p>
Full article ">Figure 6
<p>Summarized DPV data for assays with negative and positive human serum (<span class="html-italic">n</span> = 3, ± SD).</p>
Full article ">
14 pages, 3712 KiB  
Article
Detecting the PEX Like Domain of Matrix Metalloproteinase-14 (MMP-14) with Therapeutic Conjugated CNTs
by D. Vieira, J. Barralet, E. J. Harvey and G. Merle
Biosensors 2022, 12(10), 884; https://doi.org/10.3390/bios12100884 - 17 Oct 2022
Cited by 1 | Viewed by 1993
Abstract
Matrix metalloproteinases (MMPs) are essential proteins acting directly in the breakdown of the extra cellular matrix and so in cancer invasion and metastasis. Given its impact on tumor angiogenesis, monitoring MMP-14 provides strategic insights on cancer severity and treatment. In this work, we [...] Read more.
Matrix metalloproteinases (MMPs) are essential proteins acting directly in the breakdown of the extra cellular matrix and so in cancer invasion and metastasis. Given its impact on tumor angiogenesis, monitoring MMP-14 provides strategic insights on cancer severity and treatment. In this work, we report a new approach to improve the electrochemical interaction of the MMP-14 with the electrode surface while preserving high specificity. This is based on the detection of the hemopexin (PEX) domain of MMP-14, which has a greater availability with a stable and low-cost commercial molecule, as a recognition element. This molecule, called NSC-405020, is specific of the PEX domain of MMP-14 within the binding pocket. Through the covalent grafting of the NSC-405020 molecule on carbon nanotubes (CNTs), we were able to detect and quantify MMP-14 using electrochemical impedance spectroscopy with a linear range of detection of 10 ng⋅mL−1 to 100 ng⋅mL−1, and LOD of 7.5 ng⋅mL−1. The specificity of the inhibitory small molecule was validated against the PEX domain of MMP-1. The inhibitor loaded CNTs system showed as a desirable candidate to become an alternative to the conventional recognition bioelements for the detection of MMP-14. Full article
(This article belongs to the Special Issue Biosensing and Diagnosis of Cancer)
Show Figures

Figure 1

Figure 1
<p>(<b>A</b>) Schematic representation of NSC405020 molecule grafted onto CNTs surface followed by (<b>B</b>) SEM images for pristine, acylated, and inhibitor loaded CNTs (low and high magnification).</p>
Full article ">Figure 2
<p>(<b>A</b>) FTIR spectrum (top) of pristine, acylated, and inhibitor loaded CNTs, respectively. The spectrum (down) of NSC 405020 molecule is also demonstrated. (<b>B</b>) CVs for GCE, pristine, acylated, and inhibitor loaded CNTs in PBS (pH 7.40) containing 10 mmol⋅mL<sup>−1</sup> of K<sub>4</sub>[Fe (CN)<sub>6</sub>]<sup>−</sup>, 10 mmol mL<sup>−1</sup> of K<sub>3</sub>[Fe (CN)<sub>6</sub>]<sup>−</sup> and 10 mmol.mL<sup>−1</sup> of NaCl, scan rate 10 mV.s<sup>−1</sup>.</p>
Full article ">Figure 3
<p>(<b>A</b>) Nyquist plots of inhibitor loaded CNTs before and after interaction with different concentrations of MPP-14 for 10 minutes in PBS (pH 7.40) containing 10 mmol.mL<sup>−1</sup> of K<sub>3</sub>[Fe (CN)<sub>6</sub>]<sup>−</sup> and 10 mmol.mL<sup>−1</sup> of NaCl. (<b>B</b>) Linear fit of EIS response for different concentrations of MMP-14, presenting LOD of 7.5 ng⋅mL<sup>−1</sup>. Significant difference between measurements was obtained at <span class="html-italic">p</span>-value &lt; 0.05 (n = 2). Protein concentration varying from 10 ng.mL<sup>−1</sup> to 100 ng.mL<sup>−1</sup>, applied potential of +0.20 V, from 50 kHz to 500 Hz, amplitude 50 mV.</p>
Full article ">Figure 4
<p>(<b>A</b>) The inhibitor/protein interaction between small molecule NSC 405020 and the binding pocket of MMP-14 and (<b>B</b>) Schematic of the MMP-14 detection through the binding mechanism of protein and inhibitory small molecule as the recognition element.</p>
Full article ">Figure 5
<p>EIS response for the inhibitor loaded CNT after individual interaction with 50 ng⋅mL<sup>−1</sup> of MMP-1 and MMP-14 in PBS (pH 7.40) containing 10 mmol⋅mL<sup>−1</sup> of K<sub>4</sub>[Fe (CN)<sub>6</sub>]<sup>−</sup> 10 mmol⋅mL<sup>−1</sup> of K<sub>3</sub>[Fe (CN)<sub>6</sub>]<sup>−</sup> and 10 mmol⋅mL<sup>−1</sup> of NaCl. The inhibitor loaded CNT showed specificity to MMP-14 (applied potential of +0.20 V, from 50 kHz to 500 Hz, amplitude 50 mV).</p>
Full article ">
11 pages, 1752 KiB  
Article
Applications of Immunomagnetic Reduction Technology as a Biosensor in Therapeutic Evaluation of Chinese Herbal Medicine in Tauopathy Alleviation of an AD Drosophila Model
by Ming-Tsan Su, Chen-Wen Lu, Wen-Jhen Wu, Yong-Sin Jheng, Shieh-Yueh Yang, Wu-Chang Chuang, Ming-Chung Lee and Chung-Hsin Wu
Biosensors 2022, 12(10), 883; https://doi.org/10.3390/bios12100883 - 17 Oct 2022
Cited by 2 | Viewed by 2323
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. The most convincing biomarkers in the blood for AD are currently β-amyloid (Aβ) and Tau protein because amyloid plaques and neurofibrillary tangles are pathological hallmarks in the brains of patients with AD. The [...] Read more.
Alzheimer’s disease (AD) is the most common form of dementia. The most convincing biomarkers in the blood for AD are currently β-amyloid (Aβ) and Tau protein because amyloid plaques and neurofibrillary tangles are pathological hallmarks in the brains of patients with AD. The development of assay technologies in diagnosing early-stage AD is very important. The study of human AD subjects is hindered by ethical and technical limitations. Thus, many studies have therefore turned to AD animal models, such as Drosophila melanogaster, to explore AD pathology. However, AD biomarkers such as Aβ and p-Tau protein in Drosophilamelanogaster occur at extremely low levels and are difficult to detect precisely. In this study, we applied the immunomagnetic reduction (IMR) technology of nanoparticles for the detection of p-Tau expressions in hTauR406W flies, an AD Drosophila model. Furthermore, we used IMR technology as a biosensor in the therapeutic evaluation of Chinese herbal medicines in hTauR406W flies with Tau-induced toxicity. To uncover the pathogenic pathway and identify therapeutic interventions of Chinese herbal medicines in Tau-induced toxicity, we modeled tauopathy in the notum of hTauR406W flies. Our IMR data showed that the selected Chinese herbal medicines can significantly reduce p-Tau expressions in hTauR406W flies. Using evidence of notal bristle quantification and Western blotting analysis, we confirmed the validity of the IMR data. Thus, we suggest that IMR can serve as a new tool for measuring tauopathy and therapeutic evaluation of Chinese herbal medicine in an AD Drosophila model. Full article
(This article belongs to the Special Issue Biosensors for Earlier Diagnosis of Alzheimer’s Disease)
Show Figures

Figure 1

Figure 1
<p>Schematic illustrating the more favorable therapeutic effects of Chinese herbal medicine treatment, which alleviates Tau-induced toxicity in hTau<sup>R406W</sup> <span class="html-italic">Drosophila</span> flies.</p>
Full article ">Figure 2
<p>IMR detecting p-Tau 181 and standard curve: (<b>A</b>) A scheme of the physical mechanism of IMR assay. IMR is a method of assaying target molecules through measuring reductions in the mixed-frequency magnetic susceptibility of magnetic reagents resulting from the association between magnetic nanoparticles and target molecules. A magnetic nanoparticle coated with p-Tau 181 bioprobes is presented in the diagram. (<b>B</b>) Standard curve of p-Tau181 measured by IMR technique. The IMR value increased with the p-Tau 181 concentration.</p>
Full article ">Figure 3
<p>IMR can be a biosensor in therapeutic evaluation of Chinese herbal medicine for tauopathy alleviation in AD hTau<sup>R406W</sup> flies: (<b>A</b>) Relationship between the p-Tau 181 concentrations and the number of hTau<sup>R406W</sup> flies (sampled number = 50, 100, 200) obtained through IMR assay. (<b>B</b>) The diagram Quantified comparison of p-Tau 181 concentrations and IMR values in hTau<sup>R406W</sup> flies (sampled number = 100) under sham and Chinese herbal medicine (CHM) treatment (three replicate experiments for each group). Values are mean ± SEM (** <span class="html-italic">p</span> &lt; 0.01, Kruskal–Wallis non-parametric test for multiple comparisons and followed by the Mann–Whitney non-parametric test for comparisons of two independent samples).</p>
Full article ">Figure 4
<p>Chinese herbal medicine treatment showed favorable therapeutic effects for tauopathy alleviation by notal bristle quantification: (<b>A</b>) Numbers of notal bristle (as indicated by arrows) within the red frames of the hTau<sup>R406W</sup> flies with sham and Chinese herbal medicine (CHM) treatments were notably lower than in their WT counterparts; hTau<sup>R406W</sup> flies under CHM treatments demonstrated higher notal bristle numbers than the hTau<sup>R406W</sup> flies under sham treatment. (<b>B</b>) Quantified comparison of the number of notal bristles among hTau<sup>R406W</sup> flies under sham and CHM treatments, and their WT counterparts (sampled number of flies = 30 for each group, three replicate experiments for each group). Values are mean ± SEM (** <span class="html-italic">p</span> &lt; 0.01, two-way ANOVA followed by Mann–Whitney non-parametric test for comparisons).</p>
Full article ">Figure 5
<p>Chinese herbal medicine treatment showed favorable therapeutic effects for tauopathy alleviation by Western blotting: (<b>A</b>) Example of Western blotting analysis of p-Tau 181 and total Tau expressions in hTau<sup>R406W</sup> flies and their WT counterparts under sham and Chinese herbal medicine (CHM) treatments. (<b>B</b>) Quantified comparison of the ratio of p-Tau 181 to total Tau expressions among hTau<sup>R406W</sup> flies under sham and CHM treatment (three replicate experiments for each group). Values are mean ± SEM (** <span class="html-italic">p</span> &lt; 0.01, Kruskal–Wallis non-parametric test for multiple comparisons and followed by the Mann–Whitney non-parametric test for comparisons of two independent samples). kDa, kilodaltons.</p>
Full article ">
25 pages, 3671 KiB  
Review
Semiconductor Multimaterial Optical Fibers for Biomedical Applications
by Lingyu Shen, Chuanxin Teng, Zhuo Wang, Hongyi Bai, Santosh Kumar and Rui Min
Biosensors 2022, 12(10), 882; https://doi.org/10.3390/bios12100882 - 17 Oct 2022
Cited by 9 | Viewed by 5017
Abstract
Integrated sensors and transmitters of a wide variety of human physiological indicators have recently emerged in the form of multimaterial optical fibers. The methods utilized in the manufacture of optical fibers facilitate the use of a wide range of functional elements in microscale [...] Read more.
Integrated sensors and transmitters of a wide variety of human physiological indicators have recently emerged in the form of multimaterial optical fibers. The methods utilized in the manufacture of optical fibers facilitate the use of a wide range of functional elements in microscale optical fibers with an extensive variety of structures. This article presents an overview and review of semiconductor multimaterial optical fibers, their fabrication and postprocessing techniques, different geometries, and integration in devices that can be further utilized in biomedical applications. Semiconductor optical fiber sensors and fiber lasers for body temperature regulation, in vivo detection, volatile organic compound detection, and medical surgery will be discussed. Full article
(This article belongs to the Special Issue New Progress in Optical Fiber-Based Biosensors)
Show Figures

Figure 1

Figure 1
<p>A summary of this review about biomedical semiconductor optical fibers.</p>
Full article ">Figure 2
<p>Material fabrication techniques. (Reprinted with permission from ref. [<a href="#B51-biosensors-12-00882" class="html-bibr">51</a>]. Copyright 2021 Ursula J. Gibson et al.): (<b>a</b>) CZ growth, (<b>b</b>) FZ growth, (<b>c</b>) LHPG, and (<b>d</b>) LPE.</p>
Full article ">Figure 3
<p>Fiber fabrication techniques. (Reprinted with permission from ref. [<a href="#B93-biosensors-12-00882" class="html-bibr">93</a>]. Copyright 2021 Hei Chit Leo Tsui et al.): (<b>a</b>) thermal drawing and (<b>b</b>) HPCVD.</p>
Full article ">Figure 4
<p>Postprocessing techniques: (<b>a</b>) RPP, (<b>b</b>) Laser treatment (Reprinted with permission from ref. [<a href="#B93-biosensors-12-00882" class="html-bibr">93</a>]. Copyright 2021 Hei Chit Leo Tsui et al.), and (<b>c</b>) Interfacial modifier.</p>
Full article ">Figure 5
<p>Semiconductor optical fibers of different geometries: (<b>a</b>) semiconductor core fiber (Reprinted with permission from ref. [<a href="#B119-biosensors-12-00882" class="html-bibr">119</a>]. Copyright 2010 Optical Society of America), (<b>b</b>) metal-semiconductor-insulator fiber, (<b>c</b>) MOFs (Reprinted with permission from ref. [<a href="#B120-biosensors-12-00882" class="html-bibr">120</a>]. Copyright 2003 Optical Society of America), and (<b>d</b>) PBG fiber (Reprinted with permission from ref. [<a href="#B121-biosensors-12-00882" class="html-bibr">121</a>]. Copyright 2006 Optical Society of America).</p>
Full article ">Figure 6
<p>Common semiconductor implantable devices. (Reprinted with permission from ref. [<a href="#B168-biosensors-12-00882" class="html-bibr">168</a>]. Copyright 2019 PM Kumar et al.).</p>
Full article ">Figure 7
<p>OE fiber-integrated devices for in vivo temperature sensing (Reprinted with permission from ref. [<a href="#B180-biosensors-12-00882" class="html-bibr">180</a>]. Copyright 2022 He Ding et al.): (<b>a</b>) a mouse implanted with a fiber sensor for temperature detection; (<b>b</b>) sagittal CT slice reconstruction; and (<b>c</b>) dynamic temperature data acquired by the fiber sensor in the mouse brain.</p>
Full article ">Figure 8
<p>Semiconductor gas sensor applications. (Reprinted with permission from ref. [<a href="#B194-biosensors-12-00882" class="html-bibr">194</a>]. Copyright 2020 MV Nikolic et al.): (<b>a</b>) NO gas sensor for environmental protection; (<b>b</b>) metal-oxide nanocolumns sensor for fire detection; (<b>c</b>) smelling nano aerial sensor for gas source localization and mapping; (<b>d</b>) self-powered sensor wristband for health and fitness applications; (<b>e</b>) ammonia emissions measured using GasFinder open-path lasers; (<b>f</b>) ethanol vapor detection gas sensor for medical systems; and (<b>g</b>) multiple hazard gas detector for air quality monitoring.</p>
Full article ">Figure 9
<p>PBG fiber laser (Reprinted with permission from ref. [<a href="#B121-biosensors-12-00882" class="html-bibr">121</a>]. Copyright 2006 Optical Society of America): (<b>a</b>) Fiber structure; (<b>b</b>) Fluorescence spectrum shown in red, and measured reflection band gap shown in black; (<b>c</b>) Dependence of the laser energy on the pump energy.</p>
Full article ">
Previous Issue
Next Issue
Back to TopTop