Genetically Encoded Biosensor

A special issue of Biosensors (ISSN 2079-6374). This special issue belongs to the section "Biosensor and Bioelectronic Devices".

Deadline for manuscript submissions: closed (1 September 2021) | Viewed by 18958

Special Issue Editor

Molecular Physiology & Biophysics, Chemistry, and Biomedical Engineering, University of Virginia, Charlottesville, VA, USA
Interests: fluorescent and bioluminescent biosensors; protein engineering and cell physiology
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The past few decades have witnessed extraordinary advances in genetically encoded biosensors that have revolutionized the way biology can be studied. Genetically encoded biosensors have found a wide array of applications, such as exploration of live-cell biochemical and signaling networks, understanding of brain activities, and imaging of metabolism and disease markers. While there are various types of biosensors available, genetically encoded biosensors offer unique advantages over other types of biosensors in terms of their genetic encodability. Genetically encoded biosensors are thus ideal tools for live-cell and live-organism applications.

Genetically encoded biosensors are usually based on proteins or nucleic acids. They are designed and engineered to change signals (e.g., fluorescence or bioluminescence colors or intensities) in response to external stimuli or physiological changes, including pH fluctuations, metal ion homeostasis, cell signaling, membrane potential differences, phosphorylation, ubiquitination, redox reactions, and apoptosis.

The aim of this Special Issue is to highlight high-quality results (including original research articles and reviews) in the field of genetically encoded biosensors. Articles on hybrid biosensors composed of genetically encoded elements may also be submitted to this Special Issue. Articles that focus on less-common detection modalities (e.g., SPR, NMR, MRI, and PET) or that propose new ideas and new directions are particularly welcome.

Dr. Huiwang Ai
Guest Editor

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. Biosensors is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • biosensors
  • genetically encoded sensors
  • live-cell imaging
  • hybrid biosensors
  • biomarker detection
  • cell signaling
  • drug screening

Published Papers (5 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Other

15 pages, 3026 KiB  
Article
Circularly Permuted Far-Red Fluorescent Proteins
by Tianchen Wu, Yu Pang and Hui-wang Ai
Biosensors 2021, 11(11), 438; https://doi.org/10.3390/bios11110438 - 03 Nov 2021
Cited by 8 | Viewed by 2775
Abstract
The color palette of genetically encoded fluorescent protein indicators (GEFPIs) has expanded rapidly in recent years. GEFPIs with excitation and emission within the “optical window” above 600 nm are expected to be superior in many aspects, such as enhanced tissue penetration, reduced autofluorescence [...] Read more.
The color palette of genetically encoded fluorescent protein indicators (GEFPIs) has expanded rapidly in recent years. GEFPIs with excitation and emission within the “optical window” above 600 nm are expected to be superior in many aspects, such as enhanced tissue penetration, reduced autofluorescence and scattering, and lower phototoxicity. Circular permutation of fluorescent proteins (FPs) is often the first step in the process of developing single-FP-based GEFPIs. This study explored the tolerance of two far-red FPs, mMaroon1 and mCarmine, towards circular permutation. Several initial constructs were built according to previously reported circularly permuted topologies for other FP analogs. Mutagenesis was then performed on these constructs and screened for fluorescent variants. As a result, five circularly permuted far-red FPs (cpFrFPs) with excitation and emission maxima longer than 600 nm were identified. Some displayed appreciable brightness and efficient chromophore maturation. These cpFrFPs variants could be intriguing starting points to further engineer far-red GEFPIs for in vivo tissue imaging. Full article
(This article belongs to the Special Issue Genetically Encoded Biosensor)
Show Figures

Figure 1

12 pages, 5893 KiB  
Article
Versatile Cell and Animal Models for Advanced Investigation of Lead Poisoning
by De-Ming Yang and Yu-Fen Chang
Biosensors 2021, 11(10), 371; https://doi.org/10.3390/bios11100371 - 04 Oct 2021
Cited by 3 | Viewed by 2322
Abstract
The heavy metal, lead (Pb) can irreversibly damage the human nervous system. To help understand Pb-induced damage, we applied a genetically encoded Förster resonance energy transfer (FRET)-based Pb biosensor Met-lead 1.44 M1 to two living systems to monitor the concentration of Pb: induced [...] Read more.
The heavy metal, lead (Pb) can irreversibly damage the human nervous system. To help understand Pb-induced damage, we applied a genetically encoded Förster resonance energy transfer (FRET)-based Pb biosensor Met-lead 1.44 M1 to two living systems to monitor the concentration of Pb: induced pluripotent stem cell (iPSC)-derived cardiomyocytes as a semi-tissue platform and Drosophila melanogaster fruit flies as an in vivo animal model. Different FRET imaging modalities were used to obtain FRET signals, which represented the presence of Pb in the tested samples in different spatial dimensions. Using iPSC-derived cardiomyocytes, the relationship between beating activity (20–24 beats per minute, bpm) determined from the fluctuation of fluorescent signals and the concentrations of Pb represented by the FRET emission ratio values of Met-lead 1.44 M1 was revealed from simultaneous measurements. Pb (50 μM) affected the beating activity of cardiomyocytes, whereas two drugs that stop the entry of Pb differentially affected this beating activity: verapamil (2 μM) did not reverse the cessation of beating, whereas 2-APB (50 μM) partially restored this activity (16 bpm). The results clearly demonstrate the potential of this biosensor system as an anti-Pb drug screening application. In the Drosophila model, Pb was detected within the adult brain or larval central nervous system (Cha-gal4 > UAS-Met-lead 1.44 M1) using fast epifluorescence and high-resolution two-photon 3D FRET ratio image systems. The tissue-specific expression of Pb biosensors provides an excellent opportunity to explore the possible Pb-specific populations within living organisms. We believe that this integrated Pb biosensor system can be applied to the prevention of Pb poisoning and advanced research on Pb neurotoxicology. Full article
(This article belongs to the Special Issue Genetically Encoded Biosensor)
Show Figures

Figure 1

12 pages, 1811 KiB  
Article
A Co-Culture-Based Multiparametric Imaging Technique to Dissect Local H2O2 Signals with Targeted HyPer7
by Melike Secilmis, Hamza Yusuf Altun, Johannes Pilic, Yusuf Ceyhun Erdogan, Zeynep Cokluk, Busra Nur Ata, Gulsah Sevimli, Asal Ghaffari Zaki, Esra Nur Yigit, Gürkan Öztürk, Roland Malli and Emrah Eroglu
Biosensors 2021, 11(9), 338; https://doi.org/10.3390/bios11090338 - 14 Sep 2021
Cited by 6 | Viewed by 4685
Abstract
Multispectral live-cell imaging is an informative approach that permits detecting biological processes simultaneously in the spatial and temporal domain by exploiting spectrally distinct biosensors. However, the combination of fluorescent biosensors with distinct spectral properties such as different sensitivities, and dynamic ranges can undermine [...] Read more.
Multispectral live-cell imaging is an informative approach that permits detecting biological processes simultaneously in the spatial and temporal domain by exploiting spectrally distinct biosensors. However, the combination of fluorescent biosensors with distinct spectral properties such as different sensitivities, and dynamic ranges can undermine accurate co-imaging of the same analyte in different subcellular locales. We advanced a single-color multiparametric imaging method, which allows simultaneous detection of hydrogen peroxide (H2O2) in multiple cell locales (nucleus, cytosol, mitochondria) using the H2O2 biosensor HyPer7. Co-culturing of endothelial cells stably expressing differentially targeted HyPer7 biosensors paved the way for co-imaging compartmentalized H2O2 signals simultaneously in neighboring cells in a single experimental setup. We termed this approach COMPARE IT, which is an acronym for co-culture-based multiparametric imaging technique. Employing this approach, we detected lower H2O2 levels in mitochondria of endothelial cells compared to the cell nucleus and cytosol under basal conditions. Upon administering exogenous H2O2, the cytosolic and nuclear-targeted probes displayed similarly slow and moderate HyPer7 responses, whereas the mitochondria-targeted HyPer7 signal plateaued faster and reached higher amplitudes. Our results indicate striking differences in mitochondrial H2O2 accumulation of endothelial cells. Here, we present the method’s potential as a practicable and informative multiparametric live-cell imaging technique. Full article
(This article belongs to the Special Issue Genetically Encoded Biosensor)
Show Figures

Figure 1

12 pages, 2586 KiB  
Article
Influence of the luxR Regulatory Gene Dosage and Expression Level on the Sensitivity of the Whole-Cell Biosensor to Acyl-Homoserine Lactone
by Sergey Bazhenov, Uliana Novoyatlova, Ekaterina Scheglova, Vadim Fomin, Svetlana Khrulnova, Olga Melkina, Vladimir Chistyakov and Ilya Manukhov
Biosensors 2021, 11(6), 166; https://doi.org/10.3390/bios11060166 - 23 May 2021
Cited by 10 | Viewed by 3161
Abstract
Aliivibrio fischeri LuxR and Aliivibrio logei LuxR1 and LuxR2 regulatory proteins are quorum sensing transcriptional (QS) activators, inducing promoters of luxICDABEG genes in the presence of an autoinducer (3-oxo-hexanoyl-l-homoserine lactone). In the Aliivibrio cells, luxR genes are regulated by HNS, CRP, LitR, etc. [...] Read more.
Aliivibrio fischeri LuxR and Aliivibrio logei LuxR1 and LuxR2 regulatory proteins are quorum sensing transcriptional (QS) activators, inducing promoters of luxICDABEG genes in the presence of an autoinducer (3-oxo-hexanoyl-l-homoserine lactone). In the Aliivibrio cells, luxR genes are regulated by HNS, CRP, LitR, etc. Here we investigated the role of the luxR expression level in LuxI/R QS system functionality and improved the whole-cell biosensor for autoinducer detection. Escherichia coli-based bacterial lux-biosensors were used, in which Photorhabdus luminescensluxCDABE genes were controlled by LuxR-dependent promoters and luxR, luxR1, or luxR2 regulatory genes. We varied either the dosage of the regulatory gene in the cells using additional plasmids, or the level of the regulatory gene expression using the lactose operon promoter. It was shown that an increase in expression level, as well as dosage of the regulatory gene in biosensor cells, leads to an increase in sensitivity (the threshold concentration of AI is reduced by one order of magnitude) and to a two to threefold reduction in response time. The best parameters were obtained for a biosensor with an increased dosage of luxRA. fischeri (sensitivity to 3-oxo-hexanoyl-l-homoserine lactone reached 30–100 pM). Full article
(This article belongs to the Special Issue Genetically Encoded Biosensor)
Show Figures

Figure 1

Other

Jump to: Research

14 pages, 794 KiB  
Perspective
Genetically Encoded Fluorescent Indicators for Imaging Brain Chemistry
by Xiaoke Bi, Connor Beck and Yiyang Gong
Biosensors 2021, 11(4), 116; https://doi.org/10.3390/bios11040116 - 11 Apr 2021
Cited by 14 | Viewed by 5065
Abstract
Genetically encoded fluorescent indicators, combined with optical imaging, enable the detection of physiologically or behaviorally relevant neural activity with high spatiotemporal resolution. Recent developments in protein engineering and screening strategies have improved the dynamic range, kinetics, and spectral properties of genetically encoded fluorescence [...] Read more.
Genetically encoded fluorescent indicators, combined with optical imaging, enable the detection of physiologically or behaviorally relevant neural activity with high spatiotemporal resolution. Recent developments in protein engineering and screening strategies have improved the dynamic range, kinetics, and spectral properties of genetically encoded fluorescence indicators of brain chemistry. Such indicators have detected neurotransmitter and calcium dynamics with high signal-to-noise ratio at multiple temporal and spatial scales in vitro and in vivo. This review summarizes the current trends in these genetically encoded fluorescent indicators of neurotransmitters and calcium, focusing on their key metrics and in vivo applications. Full article
(This article belongs to the Special Issue Genetically Encoded Biosensor)
Show Figures

Figure 1

Back to TopTop