Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
JACS Au ; 4(2): 760-770, 2024 Feb 26.
Article in English | MEDLINE | ID: mdl-38425914

ABSTRACT

We reported over 20 years ago MNS-4.1, the first DNA aptamer with a micromolar affinity for cocaine. MNS-4.1 is based on a structural motif that is very common in any random pool of oligonucleotides, and it is actually a nonspecific hydrophobic receptor with wide cross-reactivity with alkaloids and steroids. Despite such weaknesses preventing broad applications, this aptamer became widely used in proof-of-concept demonstrations of new formats of biosensors. We now report a series of progressively improved DNA aptamers recognizing cocaine, with the final optimized receptors having low nanomolar affinity and over a thousand-fold selectivity over the initial cross-reactants. In the process of optimization, we tested different methods to eliminate cross-reactivities and improve affinity, eventually achieving properties that are comparable to those of the reported monoclonal antibody candidates for the therapy of overdose. Multiple aptamers that we now report share structural motifs with the previously reported receptor for serotonin. Further mutagenesis studies revealed a palindromic, highly adaptable, broadly cross-reactive hydrophobic motif that could be rebuilt through mutagenesis, expansion of linker regions, and selections into receptors with exceptional affinities and varying specificities.

2.
ACS Nano ; 18(8): 6286-6297, 2024 Feb 27.
Article in English | MEDLINE | ID: mdl-38355286

ABSTRACT

Single-molecule proteomics based on nanopore technology has made significant advances in recent years. However, to achieve nanopore sensing with single amino acid resolution, several bottlenecks must be tackled: controlling nanopore sizes with nanoscale precision and slowing molecular translocation events. Herein, we address these challenges by integrating amino acid-specific DNA aptamers into interface nanopores with dynamically tunable pore sizes. A phenylalanine aptamer was used as a proof-of-concept: aptamer recognition of phenylalanine moieties led to the retention of specific peptides, slowing translocation speeds. Importantly, while phenylalanine aptamers were isolated against the free amino acid, the aptamers were determined to recognize the combination of the benzyl or phenyl and the carbonyl group in the peptide backbone, enabling binding to specific phenylalanine-containing peptides. We decoupled specific binding between aptamers and phenylalanine-containing peptides from nonspecific interactions (e.g., electrostatics and hydrophobic interactions) using optical waveguide lightmode spectroscopy. Aptamer-modified interface nanopores differentiated peptides containing phenylalanine vs. control peptides with structurally similar amino acids (i.e., tyrosine and tryptophan). When the duration of aptamer-target interactions inside the nanopore were prolonged by lowering the applied voltage, discrete ionic current levels with repetitive motifs were observed. Such reoccurring signatures in the measured signal suggest that the proposed method has the possibility to resolve amino acid-specific aptamer recognition, a step toward single-molecule proteomics.


Subject(s)
Aptamers, Nucleotide , Nanopores , Amino Acids , Peptides , Aptamers, Nucleotide/chemistry , Phenylalanine
3.
Nat Nanotechnol ; 19(5): 660-667, 2024 May.
Article in English | MEDLINE | ID: mdl-38233588

ABSTRACT

Small molecules such as neurotransmitters are critical for biochemical functions in living systems. While conventional ultraviolet-visible spectroscopy and mass spectrometry lack portability and are unsuitable for time-resolved measurements in situ, techniques such as amperometry and traditional field-effect detection require a large ensemble of molecules to reach detectable signal levels. Here we demonstrate the potential of carbon-nanotube-based single-molecule field-effect transistors (smFETs), which can detect the charge on a single molecule, as a new platform for recognizing and assaying small molecules. smFETs are formed by the covalent attachment of a probe molecule, in our case a DNA aptamer, to a carbon nanotube. Conformation changes on binding are manifest as discrete changes in the nanotube electrical conductance. By monitoring the kinetics of conformational changes in a binding aptamer, we show that smFETs can detect and quantify serotonin at the single-molecule level, providing unique insights into the dynamics of the aptamer-ligand system. In particular, we show the involvement of G-quadruplex formation and the disruption of the native hairpin structure in the conformational changes of the serotonin-aptamer complex. The smFET is a label-free approach to analysing molecular interactions at the single-molecule level with high temporal resolution, providing additional insights into complex biological processes.


Subject(s)
Aptamers, Nucleotide , Nanotubes, Carbon , Serotonin , Transistors, Electronic , Aptamers, Nucleotide/chemistry , Nanotubes, Carbon/chemistry , Kinetics , Ligands , Serotonin/chemistry , Serotonin/metabolism , Biosensing Techniques/methods , Biosensing Techniques/instrumentation
4.
Faraday Discuss ; 250(0): 43-59, 2024 Mar 11.
Article in English | MEDLINE | ID: mdl-37970875

ABSTRACT

Aptamers that undergo large conformational rearrangements at the surface of electrolyte-gated field-effect transistor (EG-FETs)-based biosensors can overcome the Debye length limitation in physiological high ionic strength environments. For the sensitive detection of small molecules, carbon nanotubes (CNTs) that approach the dimensions of analytes of interest are promising channel materials for EG-FETs. However, functionalization of CNTs with bioreceptors using frequently reported surface modification strategies (e.g., π-π stacking), requires highly pristine CNTs deposited through methods that are incompatible with low-cost fabrication methods and flexible substrates. In this work, we explore alternative non-covalent surface chemistry to functionalize CNTs with aptamers. We harnessed the adhesive properties of poly-D-lysine (PDL), to coat the surface of CNTs and then grafted histamine-specific DNA aptamers electrostatically in close proximity to the CNT semiconducting channel. The layer-by-layer assembly was monitored by complementary techniques such as X-ray photoelectron spectroscopy, optical waveguide lightmode spectroscopy, and fluorescence microscopy. Surface characterization confirmed histamine aptamer integration into PDL-coated CNTs and revealed ∼5-fold higher aptamer surface coverage when using CNT networks with high surface areas. Specific aptamers assembled on EG-CNTFETs enabled histamine detection in undiluted high ionic strength solutions in the concentration range of 10 nM to 100 µM. Sequence specificity was demonstrated via parallel measurements with control EG-CNTFETs functionalized with scrambled DNA. Histamine aptamer-modified EG-CNTFETs showed high selectivity vs. histidine, the closest structural analog and precursor to histamine. Taken together, these results implied that target-specific aptamer conformational changes on CNTs facilitate signal transduction, which was corroborated by circular dichroism spectroscopy. Our work suggests that layer-by-layer polymer chemistry enables integration of structure-switching aptamers into flexible EG-CNTFETs for small-molecule biosensing.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Nanotubes, Carbon , Polylysine , Transistors, Electronic , Histamine , Nanotubes, Carbon/chemistry , Polymers/chemistry , Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods
5.
Science ; 380(6648): 942-948, 2023 06 02.
Article in English | MEDLINE | ID: mdl-37262137

ABSTRACT

Aptameric receptors are important biosensor components, yet our ability to identify them depends on the target structures. We analyzed the contributions of individual functional groups on small molecules to binding within 27 target-aptamer pairs, identifying potential hindrances to receptor isolation-for example, negative cooperativity between sterically hindered functional groups. To increase the probability of aptamer isolation for important targets, such as leucine and voriconazole, for which multiple previous selection attempts failed, we designed tailored strategies focused on overcoming individual structural barriers to successful selections. This approach enables us to move beyond standardized protocols into functional group-guided searches, relying on sequences common to receptors for targets and their analogs to serve as anchors in regions of vast oligonucleotide spaces wherein useful reagents are likely to be found.


Subject(s)
Antifungal Agents , Aptamers, Nucleotide , Biosensing Techniques , Leucine , SELEX Aptamer Technique , Voriconazole , Aptamers, Nucleotide/chemistry , SELEX Aptamer Technique/methods , Leucine/blood , Voriconazole/analysis , Antifungal Agents/analysis
6.
Bioconjug Chem ; 34(1): 124-132, 2023 01 18.
Article in English | MEDLINE | ID: mdl-36044602

ABSTRACT

Electrochemical aptamer-based (EAB) sensors utilize the binding-induced conformational change of an electrode-attached, redox-reporter-modified aptamer to transduce target recognition into an easily measurable electrochemical output. Because this signal transduction mechanism is single-step and rapidly reversible, EAB sensors support high-frequency, real-time molecular measurements, and because it recapitulates the reagentless, conformation-linked signaling seen in vivo among naturally occurring receptors, EAB sensors are selective enough to work in the complex, time-varying environments found in the living body. The fabrication of EAB sensors, however, requires that their target-recognizing aptamer be modified such that (1) it undergoes the necessary binding-induced conformational change and (2) that the thermodynamics of this "conformational switch" are tuned to ensure that they reflect an acceptable trade-off between affinity and signal gain. That is, even if an "as-selected" aptamer achieves useful affinity and specificity, it may fail when adapted to the EAB platform because it lacks the binding-induced conformational change required to support EAB signaling. In this paper we reveal the spectroscopy-guided approaches we use to modify aptamers such that they support the necessary binding-induced conformational change. Specifically, using newly reported aptamers, we demonstrate the systematic design of EAB sensors achieving clinically and physiologically relevant specificity, limits of detection, and dynamic range against the targets methotrexate and tryptophan.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Aptamers, Nucleotide/chemistry , Biosensing Techniques/methods , Oxidation-Reduction , Electrodes , Spectrum Analysis , Electrochemical Techniques/methods
7.
ACS Sens ; 8(1): 150-157, 2023 01 27.
Article in English | MEDLINE | ID: mdl-36534756

ABSTRACT

Dose-limiting toxicity and significant patient-to-patient pharmacokinetic variability often render it difficult to achieve the safe and effective dosing of drugs. This is further compounded by the slow, cumbersome nature of the analytical methods used to monitor patient-specific pharmacokinetics, which inevitably rely on blood draws followed by post-facto laboratory analysis. Motivated by the pressing need for improved "therapeutic drug monitoring", we are developing electrochemical aptamer-based (EAB) sensors, a minimally invasive biosensor architecture that can provide real-time, seconds-resolved measurements of drug levels in situ in the living body. A key advantage of EAB sensors is that they are generalizable to the detection of a wide range of therapeutic agents because they are independent of the chemical or enzymatic reactivity of their targets. Three of the four therapeutic drug classes that have, to date, been shown measurable using in vivo EAB sensors, however, bind to nucleic acids as part of their mode of action, leaving open questions regarding the extent to which the approach can be generalized to therapeutics that do not. Here, we demonstrate real-time, in vivo measurements of plasma methotrexate, an antimetabolite (a mode of action not reliant on DNA binding) chemotherapeutic, following human-relevant dosing in a live rat animal model. By providing hundreds of drug concentration values, the resulting seconds-resolved measurements succeed in defining key pharmacokinetic parameters, including the drug's elimination rate, peak plasma concentration, and exposure (area under the curve), with unprecedented 5 to 10% precision. With this level of precision, we easily identify significant (>2-fold) differences in drug exposure occurring between even healthy rats given the same mass-adjusted methotrexate dose. By providing a real-time, seconds-resolved window into methotrexate pharmacokinetics, such measurements can be used to precisely "individualize" the dosing of this significantly toxic yet vitally important chemotherapeutic.


Subject(s)
Aptamers, Nucleotide , Biosensing Techniques , Nucleic Acids , Humans , Rats , Animals , Methotrexate , Biosensing Techniques/methods , Drug Monitoring/methods
8.
Sci Adv ; 8(1): eabk0967, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34985954

ABSTRACT

Wearable technologies for personalized monitoring require sensors that track biomarkers often present at low levels. Cortisol­a key stress biomarker­is present in sweat at low nanomolar concentrations. Previous wearable sensing systems are limited to analytes in the micromolar-millimolar ranges. To overcome this and other limitations, we developed a flexible field-effect transistor (FET) biosensor array that exploits a previously unreported cortisol aptamer coupled to nanometer-thin-film In2O3 FETs. Cortisol levels were determined via molecular recognition by aptamers where binding was transduced to electrical signals on FETs. The physiological relevance of cortisol as a stress biomarker was demonstrated by tracking salivary cortisol levels in participants in a Trier Social Stress Test and establishing correlations between cortisol in diurnal saliva and sweat samples. These correlations motivated the development and on-body validation of an aptamer-FET array­based smartwatch equipped with a custom, multichannel, self-referencing, and autonomous source measurement unit enabling seamless, real-time cortisol sweat sensing.

9.
Environ Int ; 158: 107000, 2022 01.
Article in English | MEDLINE | ID: mdl-34991260

ABSTRACT

Per- and polyfluoroalkyl substances (PFAS) are widely detected environmental contaminants, and there is a great need for development of sensor technologies for rapid and continuous monitoring of PFAS. In this study, we have developed fluorescence based aptasensor that can possibly monitor perfluorooctanoic acid (PFOA) in water with limit of detection (LOD) of 0.17 µM. This is first to report the successful isolation of PFAS binding ssDNA aptamers. The obtained aptamer selectively binds PFOA with dissociation constant (KD) of 5.5 µM. Specific aptamer binding sites to PFOA were identified and the length of the fluorinated carbons was a key binding factor rather than the functional group. The aptamer binding to structurally similar PFAS compounds (i.e., perfluorocarboxylic acids and perfluorosulfonic acids with 4-8 carbon chains) was also investigated; the aptamer KD values were 6.5 and 3.3 µM for perfluoroheptanoic acid and perfluorohexanesulfonic acid, respectively, while other analogs did not bind to the aptamer. The presence of major inorganic ions and dissolved organic matter had negligible influences on the aptamer performance (<14% at a 10 mM concentration), and the aptamer performance was also robust in real wastewater effluent conditions, with a KD of 7.4 µM for PFOA. Fluorescence-based aptasensor developed in this study is adequate in monitoring PFOA levels in water contaminated with the accident spills and heavy usage of fire-fighting foams near the industrial sites and military bases. More importantly, the study opens up new capability of aptasensors to efficiently monitor the trace amount of various PFAS compounds and other fluorinated alternatives in natural and engineered water environments.


Subject(s)
Alkanesulfonic Acids , Fluorocarbons , Water Pollutants, Chemical , Caprylates , Dissolved Organic Matter , Fluorescence , Fluorocarbons/analysis , Water , Water Pollutants, Chemical/analysis
10.
J Hazard Mater ; 427: 127939, 2022 04 05.
Article in English | MEDLINE | ID: mdl-34893377

ABSTRACT

Fipronil is a broad-spectrum insecticide widely used in agriculture and residential areas; its indiscriminate use leads to environmental pollution and poses health hazards. Early detection of fipronil is critical to prevent the deleterious effects. However, current insecticide analysis methods such as HPLC, LC/MS, and GC/MS are incompetent; they are costly, immobile, time-consuming, laborious, and need skilled technicians. Hence, a sensitive, specific, and cheap biosensor are essential to containing the contamination. Here, we designed two novel biosensors-the first design relied on fluorescent labeling/quenching, while the second sensor focused on label-free detection using Thioflavin T displacement. Altogether, we identified four candidate aptamers, predicted secondary structures, and performed 3D molecular modeling to predict the binding pocket of fipronil in FiPA6B aptamer. Furthermore, the aptameric sensors showed high sensitivity to fipronil of sub-ppb level LOD, attributed to stringent experimental design. The biosensors displayed high specificity against other phenylpyrazole insecticides and demonstrated robust sensitivity for fipronil in real samples like cabbage and cucumber. Notably, to the best of our knowledge, this is the first demonstration of noncanonical G4-quadruplex-like aptamer binding to fipronil, verified using CD spectroscopy. Such aptasensors possess considerable potential for real-time measurements of hazardous insecticides as point-of-care technology.


Subject(s)
Biosensing Techniques , Insecticides , DNA , Pyrazoles
11.
Sci Transl Med ; 13(585)2021 03 17.
Article in English | MEDLINE | ID: mdl-33731432

ABSTRACT

Tracking antimalarial drug use and efficacy is essential for monitoring the current spread of antimalarial drug resistance. However, available methods for determining tablet quality and patient drug use are often inaccessible, requiring well-equipped laboratories capable of performing liquid chromatography-mass spectrometry (LC-MS). Here, we report the development of aptamer-based fluorescent sensors for the rapid, specific detection of the antimalarial compounds piperaquine and mefloquine-two slow-clearing partner drugs in current first-line artemisinin-based combination therapies (ACTs). Highly selective DNA aptamers were identified that bind piperaquine and mefloquine with dissociation constants (K d's) measured in the low nanomolar range via two independent methods. The aptamers were isolated from a library of single-stranded DNA molecules using a capture-systematic evolution of ligands by exponential enrichment (SELEX) technique and then adapted into structure-switching aptamer fluorescent sensors. Sensor performance was optimized for the detection of drug from human serum and crushed tablets, resulting in two sensing platforms. The patient sample platform was validated against an LC-MS standard drug detection method in samples from healthy volunteers and patients with malaria. This assay provides a rapid and inexpensive method for tracking antimalarial drug use and quality for the containment and study of parasite resistance, a major priority for malaria elimination campaigns. This sensor platform allows for flexibility of sample matrix and can be easily adapted to detect other small-molecule drugs.


Subject(s)
Antimalarials , Aptamers, Nucleotide , Malaria , Quinolines , Antimalarials/therapeutic use , Aptamers, Nucleotide/therapeutic use , Humans , Malaria/diagnosis , Malaria/drug therapy , Mefloquine/therapeutic use , Quinolines/therapeutic use
12.
ACS Appl Mater Interfaces ; 13(8): 9425-9435, 2021 Mar 03.
Article in English | MEDLINE | ID: mdl-33410656

ABSTRACT

Oligonucleotide receptors (aptamers), which change conformation upon target recognition, enable electronic biosensing under high ionic-strength conditions when coupled to field-effect transistors (FETs). Because highly negatively charged aptamer backbones are influenced by ion content and concentration, biosensor performance and target sensitivities were evaluated under application conditions. For a recently identified dopamine aptamer, physiological concentrations of Mg2+ and Ca2+ in artificial cerebrospinal fluid produced marked potentiation of dopamine FET-sensor responses. By comparison, divalent cation-associated signal amplification was not observed for FET sensors functionalized with a recently identified serotonin aptamer or a previously reported dopamine aptamer. Circular dichroism spectroscopy revealed Mg2+- and Ca2+-induced changes in target-associated secondary structure for the new dopamine aptamer, but not the serotonin aptamer nor the old dopamine aptamer. Thioflavin T displacement corroborated the Mg2+ dependence of the new dopamine aptamer for target detection. These findings imply allosteric binding interactions between divalent cations and dopamine for the new dopamine aptamer. Developing and testing sensors in ionic environments that reflect intended applications are best practices for identifying aptamer candidates with favorable attributes and elucidating sensing mechanisms.


Subject(s)
Aptamers, Nucleotide/chemistry , Calcium/chemistry , Dopamine/analysis , Magnesium/chemistry , Benzothiazoles/chemistry , Biosensing Techniques/instrumentation , Biosensing Techniques/methods , Dopamine/chemistry , Electrochemical Techniques/instrumentation , Electrochemical Techniques/methods , G-Quadruplexes/drug effects , Serotonin/analysis , Serotonin/chemistry , Transistors, Electronic
13.
iScience ; 21: 328-340, 2019 Nov 22.
Article in English | MEDLINE | ID: mdl-31698247

ABSTRACT

Digital health promises a paradigm shift for medicine where biomarkers in individuals are continuously monitored to improve diagnosis and treatment of disease. To that end, a technology for minimally invasive quantification of endogenous analytes in bodily fluids will be required. Here, we describe a strategy for designing and fabricating hydrogel microfilaments that can penetrate the skin while allowing for optical fluorescence sensing. The polyacrylamide formulation was selected to provide high elastic modulus in the dehydrated state and optical transparency in the hydrated state. The microfilaments can be covalently tethered to a fluorescent aptamer to enable functional sensing. The microfilament array can penetrate the skin with low pain and without breaking, contact the dermal interstitial fluid, and be easily removed from the skin. In the future, hydrogel microfilaments could be integrated with a wearable fluorometer to serve as a platform for continuous, minimally invasive monitoring of intradermal biomarkers.

14.
ACS Sens ; 4(12): 3308-3317, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31631652

ABSTRACT

Determination of the amino acid phenylalanine is important for lifelong disease management in patients with phenylketonuria, a genetic disorder in which phenylalanine accumulates and persists at levels that alter brain development and cause permanent neurological damage and cognitive dysfunction. Recent approaches for treating phenylketonuria focus on injectable medications that efficiently break down phenylalanine but sometimes result in detrimentally low phenylalanine levels. We have identified new DNA aptamers for phenylalanine in two formats, initially as fluorescent sensors and then, incorporated with field-effect transistors (FETs). Aptamer-FET sensors detected phenylalanine over a wide range of concentrations (fM to mM). para-Chlorophenylalanine, which inhibits the enzyme that converts phenylalanine to tyrosine, was used to induce hyperphenylalaninemia during brain development in mice. Aptamer-FET sensors were specific for phenylalanine versus para-chlorophenylalanine and differentiated changes in mouse serum phenylalanine at levels expected in patients. Aptamer-FETs can be used to investigate models of hyperphenylalanemia in the presence of structurally related enzyme inhibitors, as well as naturally occurring amino acids. Nucleic acid-based receptors that discriminate phenylalanine analogs, some that differ by a single substituent, indicate a refined ability to identify aptamers with binding pockets tailored for high affinity and specificity. Aptamers of this type integrated into FETs enable rapid, electronic, label-free phenylalanine sensing.


Subject(s)
Aptamers, Nucleotide/chemistry , DNA/chemistry , Phenylalanine/blood , Transistors, Electronic , Animals , Electrochemical Techniques/instrumentation , Electrochemical Techniques/methods , Fenclonine , Mice , Phenylalanine/chemistry , Phenylketonurias/blood , Phenylketonurias/chemically induced
15.
ACS Sens ; 4(10): 2832-2837, 2019 10 25.
Article in English | MEDLINE | ID: mdl-31556293

ABSTRACT

The electrochemical aptamer-based (E-AB) sensing platform appears to be a convenient (rapid, single-step, and calibration-free) and modular approach to measure concentrations of specific molecules (irrespective of their chemical reactivity) directly in blood and even in situ in the living body. Given these attributes, the platform may thus provide significant opportunities to render therapeutic drug monitoring (the clinical practice in which dosing is adjusted in response to plasma drug measurements) as frequent and convenient as the measurement of blood sugar has become for diabetics. The ability to measure arbitrary molecules in the body in real time could even enable closed-loop feedback control over plasma drug levels in a manner analogous to the recently commercialized controlled blood sugar systems. As initial exploration of this, we describe here the selection of an aptamer against vancomycin, a narrow therapeutic window antibiotic for which therapeutic monitoring is a critical part of the standard of care, and its adaptation into an electrochemical aptamer-based (E-AB) sensor. Using this sensor, we then demonstrate: (i) rapid (seconds) and convenient (single-step and calibration-free) measurement of plasma vancomycin in finger-prick-scale samples of whole blood, (ii) high-precision measurement of subject-specific vancomycin pharmacokinetics (in a rat animal model), and (iii) high-precision, closed-loop feedback control over plasma levels of the drug (in a rat animal model). The ability to not only track (with continuous-glucose-monitor-like measurement frequency and convenience) but also actively control plasma drug levels provides an unprecedented route toward improving therapeutic drug monitoring and, more generally, the personalized, high-precision delivery of pharmacological interventions.


Subject(s)
Anti-Bacterial Agents/blood , Aptamers, Nucleotide/chemistry , Drug Monitoring/methods , Electrochemical Techniques/methods , Vancomycin/blood , Animals , Anti-Bacterial Agents/chemistry , Cattle , Male , Rats, Sprague-Dawley , Vancomycin/chemistry
16.
ACS Appl Mater Interfaces ; 11(12): 11262-11269, 2019 Mar 27.
Article in English | MEDLINE | ID: mdl-30848118

ABSTRACT

Physical isolation of molecular computing elements holds the potential for increasing system complexity by enabling the reuse of standardized components and by protecting the components from environmental degradation. However, once elements have been compartmentalized, methods for communicating into these compartments are needed. We report the compartmentalization of steroid-responsive DNA aptamers within giant unilamellar vesicles (GUVs) that are permeable to steroid inputs. Monodisperse GUVs are loaded with aptamers using a microfluidic platform. We demonstrate the target-specific activation of individual aptamers within the GUVs and then load two noninterfering aptamers into the same GUV and demonstrate specific responses to all possible combinations of the two input steroids. Crucially, GUVs prevent the degradation of DNA components by nucleases, providing a potential mechanism for deploying nucleic acid components in vivo. Importantly, our compartments also prevent nonspecific cross-talk between complementary strands, thereby providing a method for parallel execution of cross-reacting molecular logic components. Thus, we provide a mechanism for spatially organizing molecular computing elements, which will increase system modularity by allowing standardized components to be reused.


Subject(s)
Aptamers, Nucleotide/metabolism , Unilamellar Liposomes/chemistry , Aptamers, Nucleotide/chemistry , Base Pairing , Deoxyribonucleases/metabolism , Fluorometry , Microfluidics , Microscopy, Confocal , Unilamellar Liposomes/metabolism
17.
Science ; 362(6412): 319-324, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30190311

ABSTRACT

Detection of analytes by means of field-effect transistors bearing ligand-specific receptors is fundamentally limited by the shielding created by the electrical double layer (the "Debye length" limitation). We detected small molecules under physiological high-ionic strength conditions by modifying printed ultrathin metal-oxide field-effect transistor arrays with deoxyribonucleotide aptamers selected to bind their targets adaptively. Target-induced conformational changes of negatively charged aptamer phosphodiester backbones in close proximity to semiconductor channels gated conductance in physiological buffers, resulting in highly sensitive detection. Sensing of charged and electroneutral targets (serotonin, dopamine, glucose, and sphingosine-1-phosphate) was enabled by specifically isolated aptameric stem-loop receptors.


Subject(s)
Aptamers, Nucleotide/chemistry , Biosensing Techniques , Dopamine/analysis , Glucose/analysis , Lysophospholipids/analysis , Serotonin/analysis , Sphingosine/analogs & derivatives , Sphingosine/analysis , Transistors, Electronic
18.
Nucleic Acids Res ; 46(8): e43, 2018 05 04.
Article in English | MEDLINE | ID: mdl-29361056

ABSTRACT

Aptamer-based sensors offer a powerful tool for molecular detection, but the practical implementation of these biosensors is hindered by costly and laborious sequence engineering and chemical modification procedures. We report a simple strategy for directly isolating signal-reporting aptamers in vitro through systematic evolution of ligands by exponential enrichment (SELEX) that transduce binding events into a detectable change of absorbance via target-induced displacement of a small-molecule dye. We first demonstrate that diethylthiatricarbocyanine (Cy7) can stack into DNA three-way junctions (TWJs) in a sequence-independent fashion, greatly altering the dye's absorbance spectrum. We then design a TWJ-containing structured library and isolate an aptamer against 3,4-methylenedioxypyrovalerone (MDPV), a synthetic cathinone that is an emerging drug of abuse. This aptamer intrinsically binds Cy7 within its TWJ domain, but MDPV efficiently displaces the dye, resulting in a change in absorbance within seconds. This assay is label-free, and detects nanomolar concentrations of MDPV. It also recognizes other synthetic cathinones, offering the potential to detect newly-emerging designer drugs, but does not detect structurally-similar non-cathinone compounds or common cutting agents. Moreover, we demonstrate that the Cy7-displacement colorimetric assay is more sensitive than a conventional strand-displacement fluorescence assay. We believe our strategy offers an effective generalized approach for the development of sensitive dye-displacement colorimetric assays for other small-molecule targets.


Subject(s)
Aptamers, Nucleotide/isolation & purification , SELEX Aptamer Technique/methods , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/genetics , Base Sequence , Benzodioxoles/analysis , Biosensing Techniques/methods , Carbocyanines/chemistry , Colorimetry/methods , Coloring Agents/chemistry , DNA/chemistry , DNA/genetics , Molecular Structure , Pyrrolidines/analysis , Synthetic Cathinone
19.
J Electrochem Soc ; 164(5): B3122-B3129, 2017.
Article in English | MEDLINE | ID: mdl-29170564

ABSTRACT

Systematic evolution of ligands by exponential enrichment (SELEX) offers a powerful method to isolate affinity oligonucleotides known as aptamers, which can then be used in a wide range of applications from drug delivery to biosensing. However, conventional SELEX methods rely on labor intensive and time consuming benchtop operations. A simplified microfluidic approach is presented which allows integration of the affinity selection and amplification stages of SELEX for the isolation of target-binding oligonucleotides by combining bead-based biochemical reactions with free solution electrokinetic oligonucleotide transfer. Free solution electrokinetics allows coupling of affinity selection and amplification for closed loop oligonucleotide enrichment without the need for offline processes, flow handling components or gel components, while bead based selection and amplification allow efficient manipulation of reagents and reaction products thereby realizing on-chip loop closure and integration of the entire SELEX process. Thus the approach is capable of multi-round enrichment of oligonucleotides using simple transfer processes while maintaining a high level of device integration, as demonstrated by the isolation of an aptamer pool against a protein target (IgA) with significantly higher binding affinity than the starting library in approximately 4 hours of processing time.

20.
ACS Chem Biol ; 12(12): 3103-3112, 2017 12 15.
Article in English | MEDLINE | ID: mdl-29083858

ABSTRACT

Artificial receptors for hydrophobic molecules usually have moderate affinities and limited selectivities. We describe three new classes of high affinity hydrophobic receptors for nonaromatic steroids based on deoxyribonucleotides, obtained through five high stringency selections coupled with tailored counter-selections. The isolation of multiple classes of high affinity steroid receptors demonstrates the surprising breadth of moderately sized hydrophobic binding motifs (<40 nucleotides) available to natural nucleic acids. Studies of interactions with analogs indicate that two classes, four-way junctions and 4XGN motifs, comprise receptors with shapes that prevent binding of specific steroid conjugates used in counter-selections. Furthermore, they strongly prefer nonhydroxylated steroid cores, which is typical for hydrophobic receptors. The third new class accommodates hydroxyl groups in high-affinity, high-selectivity binding pockets, thus reversing the preferences of the first two classes. The high-affinity binding of aptamers to targets efficiently inhibits double-helix formation in the presence of the complementary oligonucleotides. The high affinity of some of these receptors and tailored elimination of binding through counter-selections ensures that these new aptamers will enable clinical chemistry applications.


Subject(s)
Dehydroepiandrosterone Sulfate/chemistry , Desoxycorticosterone/analogs & derivatives , Nucleic Acids/chemistry , Receptors, Steroid/chemistry , Receptors, Steroid/metabolism , Steroids/chemistry , Desoxycorticosterone/chemistry , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL
...