Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Anal Chem ; 93(29): 10251-10260, 2021 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-34264067

RESUMO

The implementation of accurate and sensitive molecular detection for the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is paramount to effectively control the ongoing coronavirus disease 2019 (COVID-19) pandemic. In this regard, we herein propose the specific and highly sensitive SARS-CoV-2 detection based on nanoyeast single-chain-variable fragment (scFv) and ultrasensitive plasmonic nanobox-integrated nanomixing microassay. Importantly, this designed platform showcases the utility of nanoyeast-scFvs as specific capture reagents targeting the receptor-binding domain (RBD) of the virus and as monoclonal antibody alternatives suitable for cost-effective mass production and frequent testing. By capitalizing on single-particle active nanoboxes as plasmonic nanostructures for surface-enhanced Raman scattering (SERS), the microassay utilizes highly sensitive Raman signals to indicate virus infection. The developed microassay further integrated nanomixing for accelerating molecular collisions. Through the synergistic working of nanoyeast-scFv, plasmonic nanoboxes, and nanomixing, the highly specific and sensitive SARS-CoV-2 detection is achieved as low as 17 virus/µL without any molecular amplification. We successfully demonstrate SARS-CoV-2 detection in saliva samples of simulated patients at clinically relevant viral loads, suggesting the possibility of this platform for accurate and noninvasive patient screening.


Assuntos
COVID-19 , Anticorpos de Cadeia Única , Humanos , SARS-CoV-2 , Saliva , Análise Espectral Raman
2.
J Am Chem Soc ; 141(20): 8128-8135, 2019 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-31074995

RESUMO

Allosteric protein switches are key controllers of information and energy processing in living organisms and are desirable engineered control tools in synthetic systems. Here we present a generally applicable strategy for construction of allosteric signaling systems with inputs and outputs of choice. We demonstrate conversion of constitutively active enzymes into peptide-operated synthetic allosteric ON switches by insertion of a calmodulin domain into rationally selected sites. Switches based on EGFP, glucose dehydrogenase, NanoLuciferase, and dehydrofolate reductase required minimal optimization and demonstrated a dynamic response ranging from 1.8-fold in the former case to over 200-fold in the latter case. The peptidic nature of the calmodulin ligand enables incorporation of such synthetic switch modules into higher order sensory architectures. Here, a ligand-mediated increase in proximity of the allosteric switch and the engineered activator peptide modulates biosensor's activity. Created biosensors were used to measure concentrations of clinically relevant drugs and biomarkers in plasma, saliva, and urine with accuracy comparable to that of the currently used clinical diagnostic assays. The approach presented is generalizable as it allows rapid construction of efficient protein switches that convert binding of a broad range of analytes into a biochemical activity of choice enabling construction of artificial signaling and metabolic circuits of potentially unlimited complexity.


Assuntos
Técnicas Biossensoriais/métodos , Glucose Desidrogenase/química , Proteínas Recombinantes de Fusão/química , Albumina Sérica Humana/urina , alfa-Amilases/análise , Acinetobacter calcoaceticus/enzimologia , Sequência de Aminoácidos , Proteínas de Bactérias/química , Proteínas de Bactérias/genética , Biomarcadores/sangue , Biomarcadores/urina , Calmodulina/química , Calmodulina/genética , Calmodulina/metabolismo , Proteínas de Ligação a Calmodulina/metabolismo , Ciclosporina/análise , Diabetes Mellitus/urina , Glucose Desidrogenase/genética , Humanos , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/genética , Saliva/química , Tacrolimo/análise , Proteínas de Ligação a Tacrolimo/química , Proteínas de Ligação a Tacrolimo/genética
3.
Biochim Biophys Acta Bioenerg ; 1858(12): 945-954, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28882760

RESUMO

To better understand metalloproteins with Mn-clusters, we have designed artificial four-helix bundles to have one, two, or three dinuclear metal centers able to bind Mn(II). Circular dichroism measurements showed that the Mn-proteins have substantial α-helix content, and analysis of electron paramagnetic resonance spectra is consistent with the designed number of bound Mn-clusters. The Mn-proteins were shown to catalyze the conversion of hydrogen peroxide into molecular oxygen. The loss of hydrogen peroxide was dependent upon the concentration of protein with bound Mn, with the proteins containing multiple Mn-clusters showing greater activity. Using an oxygen sensor, the oxygen concentration was found to increase with a rate up to 0.4µM/min, which was dependent upon the concentrations of hydrogen peroxide and the Mn-protein. In addition, the Mn-proteins were shown to serve as electron donors to bacterial reaction centers using optical spectroscopy. Similar binding of the Mn-proteins to reaction centers was observed with an average dissociation constant of 2.3µM. The Mn-proteins with three metal centers were more effective at this electron transfer reaction than the Mn-proteins with one or two metal centers. Thus, multiple Mn-clusters can be incorporated into four-helix bundles with the capability of performing catalysis and electron transfer to a natural protein.


Assuntos
Manganês/química , Metaloproteínas/química , Oxigênio/química , Conformação Proteica em alfa-Hélice , Sítios de Ligação , Dicroísmo Circular , Espectroscopia de Ressonância de Spin Eletrônica , Peróxido de Hidrogênio/química , Peróxido de Hidrogênio/metabolismo , Metaloproteínas/síntese química , Metaloproteínas/metabolismo , Modelos Moleculares , Ligação Proteica
4.
Biochim Biophys Acta ; 1857(5): 539-547, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26392146

RESUMO

A compelling target for the design of electron transfer proteins with novel cofactors is to create a model for the oxygen-evolving complex, a Mn4Ca cluster, of photosystem II. A mononuclear Mn cofactor can be added to the bacterial reaction center, but the addition of multiple metal centers is constrained by the native protein architecture. Alternatively, metal centers can be incorporated into artificial proteins. Designs for the addition of dinuclear metal centers to four-helix bundles resulted in three artificial proteins with ligands for one, two, or three dinuclear metal centers able to bind Mn. The three-dimensional structure determined by X-ray crystallography of one of the Mn-proteins confirmed the design features and revealed details concerning coordination of the Mn center. Electron transfer between these artificial Mn-proteins and bacterial reaction centers was investigated using optical spectroscopy. After formation of a light-induced, charge-separated state, the experiments showed that the Mn-proteins can donate an electron to the oxidized bacteriochlorophyll dimer of modified reaction centers, with the Mn-proteins having additional metal centers being more effective at this electron transfer reaction. Modeling of the structure of the Mn-protein docked to the reaction center showed that the artificial protein likely binds on the periplasmic surface similarly to cytochrome c2, the natural secondary donor. Combining reaction centers with exogenous artificial proteins provides the opportunity to create ligands and investigate the influence of inhomogeneous protein environments on multinuclear redox-active metal centers. This article is part of a Special Issue entitled Biodesign for Bioenergetics--the design and engineering of electronic transfer cofactors, proteins and protein networks, edited by Ronald L. Koder and J.L. Ross Anderson.


Assuntos
Proteínas de Bactérias/química , Manganês/metabolismo , Metaloproteínas/química , Engenharia de Proteínas/métodos , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Sítios de Ligação/genética , Coenzimas/química , Coenzimas/genética , Coenzimas/metabolismo , Humanos , Manganês/química , Metaloproteínas/genética , Metaloproteínas/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Secundária de Proteína
5.
Bioconjug Chem ; 25(2): 424-32, 2014 Feb 19.
Artigo em Inglês | MEDLINE | ID: mdl-24410055

RESUMO

BH3 peptides are key mediators of apoptosis and have served as the lead structures for the development of anticancer therapeutics. Previously, we reported the application of a simple cysteine-based side chain cross-linking chemistry to NoxaBH3 peptides that led to the generation of the cross-linked NoxaBH3 peptides with increased cell permeability and higher inhibitory activity against Mcl-1 ( Muppidi, A., Doi, K., Edwardraja, S., Drake, E. J., Gulick, A. M., Wang, H.-G., Lin, Q. ( 2012 ) J. Am. Chem. Soc. 134 , 14734 ). To deliver cross-linked NoxaBH3 peptides selectively into cancer cells for enhanced efficacy and reduced systemic toxicity, here we report the conjugation of the NoxaBH3 peptides with the extracellular ubiquitin, a recently identified endogenous ligand for CXCR4, a chemokine receptor overexpressed in cancer cells. The resulting ubiquitin-NoxaBH3 peptide conjugates showed increased inhibitory activity against Mcl-1 and selective killing of the CXCR4-expressing cancer cells. The successful delivery of the NoxaBH3 peptides by ubiquitin into cancer cells suggests that the ubiquitin/CXCR4 axis may serve as a general route for the targeted delivery of anticancer agents.


Assuntos
Proteína de Sequência 1 de Leucemia de Células Mieloides/antagonistas & inibidores , Neoplasias/patologia , Fragmentos de Peptídeos/administração & dosagem , Proteínas Proto-Oncogênicas/administração & dosagem , Ubiquitina/química , Sequência de Aminoácidos , Linhagem Celular Tumoral , Polarização de Fluorescência , Humanos , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Proteínas Proto-Oncogênicas/química
6.
Nat Nanotechnol ; 18(10): 1222-1229, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37291255

RESUMO

Accurate and early detection of biomarkers provides the molecular evidence for disease management, allowing prompt actions and timely treatments to save lives. Multivalent biomolecular interactions between the probe and biomarker as well as controlled probe orientation on material surfaces are keys for highly sensitive detection. Here we report the bioengineering of programmable and multifunctional nanoprobes, which can provide rapid, specific and highly sensitive detection of emerging diseases in a range of widely used diagnostic systems. These nanoprobes composed of nanosized cell wall fragments, termed as synthetic bionanofragments (SynBioNFs), are generated by the fragmentation of genetically programmed yeast cells. SynBioNFs display multiple copies of biomolecules for high-affinity target binding and molecular handles for the precisely orientated attachment on surfaces used in diagnostic platforms. SynBioNFs are demonstrated for the capture and detection of SARS-CoV-2 virions using multiple diagnostic platforms, including surface-enhanced Raman scattering, fluorescence, electrochemical and colorimetric-based lateral flow systems with sensitivity comparable with the gold-standard reverse-transcription quantitative polymerase chain reaction.


Assuntos
SARS-CoV-2 , Saccharomyces cerevisiae , Saccharomyces cerevisiae/genética , Indicadores e Reagentes , SARS-CoV-2/genética
7.
J Am Chem Soc ; 134(36): 14734-7, 2012 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-22920569

RESUMO

Direct chemical modifications provide a simple and effective means to "translate" bioactive helical peptides into potential therapeutics targeting intracellular protein-protein interactions. We previously showed that distance-matching bisaryl cross-linkers can reinforce peptide helices containing two cysteines at the i and i+7 positions and confer cell permeability to the cross-linked peptides. Here we report the first crystal structure of a biphenyl-cross-linked Noxa peptide in complex with its target Mcl-1 at 2.0 Å resolution. Guided by this structure, we remodeled the surface of this cross-linked peptide through side-chain substitution and N-methylation and obtained a pair of cross-linked peptides with substantially increased helicity, cell permeability, proteolytic stability, and cell-killing activity in Mcl-1-overexpressing U937 cells.


Assuntos
Permeabilidade da Membrana Celular/efeitos dos fármacos , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Humanos , Modelos Moleculares , Estrutura Molecular , Proteína de Sequência 1 de Leucemia de Células Mieloides , Peptídeos/química , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células U937
8.
J Mol Biol ; 434(17): 167678, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-35709893

RESUMO

Biological information processing networks rely on allosteric protein switches that dynamically interconvert biological signals. Construction of their artificial analogues is a central goal of synthetic biology and bioengineering. Receptor domain insertion is one of the leading methods for constructing chimeric protein switches. Here we present an in vitro expression-based platform for the analysis of chimeric protein libraries for which traditional cell survival or cytometric high throughput assays are not applicable. We utilise this platform to screen a focused library of chimeras between PQQ-glucose dehydrogenase and calmodulin. Using this approach, we identified 50 chimeras (approximately 23% of the library) that were activated by calmodulin-binding peptides. We analysed performance parameters of the active chimeras and demonstrated that their dynamic range and response times are anticorrelated, pointing to the existence of an inherent thermodynamic trade-off. We show that the structure of the ligand peptide affects both the response and activation kinetics of the biosensors suggesting that the structure of a ligand:receptor complex can influence the chimera's activation pathway. In order to understand the extent of structural changes in the reporter protein induced by the receptor domains, we have analysed one of the chimeric molecules by CD spectroscopy and hydrogen-deuterium exchange mass spectrometry. We concluded that subtle ligand-induced changes in the receptor domain propagated into the GDH domain and affected residues important for substrate and cofactor binding. Finally, we used one of the identified chimeras to construct a two-component rapamycin biosensor and demonstrated that core switch optimisation translated into improved biosensor performance.


Assuntos
Regulação Alostérica , Calmodulina , Glucose Desidrogenase , Biblioteca de Peptídeos , Proteínas Recombinantes de Fusão , Calmodulina/química , Calmodulina/genética , Glucose Desidrogenase/química , Glucose Desidrogenase/genética , Ligantes , Ligação Proteica , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética , Termodinâmica
9.
Nat Commun ; 13(1): 789, 2022 02 10.
Artigo em Inglês | MEDLINE | ID: mdl-35145068

RESUMO

Allostery enables proteins to interconvert different biochemical signals and form complex metabolic and signaling networks. We hypothesize that circular permutation of proteins increases the probability of functional coupling of new N- and C- termini with the protein's active center through increased local structural disorder. To test this we construct a synthetically allosteric version of circular permutated NanoLuc luciferase that can be activated through ligand-induced intramolecular non-covalent cyclisation. This switch module is tolerant of the structure of binding domains and their ligands, and can be used to create biosensors of proteins and small molecules. The developed biosensors covers a range of emission wavelengths and displays sensitivity as low as 50pM and dynamic range as high as 16-fold and could quantify their cognate ligand in human fluids. We apply hydrogen exchange kinetic mass spectroscopy to analyze time resolved structural changes in the developed biosensors and observe ligand-mediated folding of newly created termini.


Assuntos
Regulação Alostérica , Luciferases/genética , Luciferases/metabolismo , Engenharia Metabólica , Regulação Alostérica/genética , Regulação da Expressão Gênica , Humanos , Ligantes , Luciferases/química , Modelos Moleculares
10.
J Am Chem Soc ; 133(39): 15316-9, 2011 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-21899368

RESUMO

Bioorthogonal reactions suitable for functionalization of genetically or metabolically encoded alkynes, for example, copper-catalyzed azide-alkyne cycloaddition reaction ("click chemistry"), have provided chemical tools to study biomolecular dynamics and function in living systems. Despite its prominence in organic synthesis, copper-free Sonogashira cross-coupling reaction suitable for biological applications has not been reported. In this work, we report the discovery of a robust aminopyrimidine-palladium(II) complex for copper-free Sonogashira cross-coupling that enables selective functionalization of a homopropargylglycine (HPG)-encoded ubiquitin protein in aqueous medium. A wide range of aromatic groups including fluorophores and fluorinated aromatic compounds can be readily introduced into the HPG-containing ubiquitin under mild conditions with good to excellent yields. The suitability of this reaction for functionalization of HPG-encoded ubiquitin in Escherichia coli was also demonstrated. The high efficiency of this new catalytic system should greatly enhance the utility of Sonogashira cross-coupling in bioorthogonal chemistry.


Assuntos
Alcinos/metabolismo , Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Escherichia coli/citologia , Glicina/análogos & derivados , Ubiquitina/química , Ubiquitina/metabolismo , Água/química , Glicina/metabolismo , Modelos Moleculares , Paládio/química , Conformação Proteica , Solubilidade
11.
Bioelectrochemistry ; 138: 107735, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33482577

RESUMO

The Implication (IMPLY) and Inhibition (INHIB) Boolean logic gates were realized using switchable chimeric pyrroloquinoline quinone-dependent glucose dehydrogenase (PQQ-GDH-Clamp) containing a fused affinity clamp unit recognizing a signal-peptide. The second component of the logic gate was the wild-type PQQ-glucose dehydrogenase working cooperatively with the PQQ-GDH-Clamp enzyme. The IMPLY and INHIB gates were realized using the same enzyme composition activated with differently defined input signals, thus representing reconfigurable logic systems. The logic gates were first tested while operating in a solution with optical analysis of the output signals. Then, the enzymes were immobilized on a buckypaper electrode for electrochemical transduction of the output signals. The switchable modified electrodes mimicking the IMPLY or INHIB logic gates were integrated with an oxygen-reducing electrode modified with bilirubin oxidase to operate as a biofuel cell activated/inhibited by various input signal combinations processed either by IMPLY or INHIB logic gates. The switchable biofuel cell was used as a self-powered device triggering molecule release function controlled by the logically processed molecule signals.


Assuntos
Eletroquímica/métodos , Lógica , Fontes de Energia Bioelétrica , Eletrodos , Glucose Desidrogenase/metabolismo
12.
ACS Sens ; 6(10): 3596-3603, 2021 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-34637274

RESUMO

Enzymatic polypeptide proteolysis is a widespread and powerful biological control mechanism. Over the last few years, substantial progress has been made in creating artificial proteolytic systems where an input of choice modulates the protease activity and thereby the activity of its substrates. However, all proteolytic systems developed so far have relied on the direct proteolytic cleavage of their effectors. Here, we propose a new concept where protease biosensors with a tunable input uncage a signaling peptide, which can then transmit a signal to an allosteric protein reporter. We demonstrate that both the cage and the regulatory domain of the reporter can be constructed from the same peptide-binding domain, such as calmodulin. To demonstrate this concept, we constructed a proteolytic rapamycin biosensor and demonstrated its quantitative actuation on fluorescent, luminescent, and electrochemical reporters. Using the latter, we constructed sensitive bioelectrodes that detect the messenger peptide release and quantitatively convert the recognition event into electric current. We discuss the application of such systems for the construction of in vitro sensory arrays and in vivo signaling circuits.


Assuntos
Técnicas Biossensoriais , Calmodulina , Calmodulina/metabolismo , Peptídeo Hidrolases , Proteólise , Transdução de Sinais
13.
ACS Biomater Sci Eng ; 7(12): 5850-5860, 2021 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-34738789

RESUMO

Bioengineered yeast bio-nanomaterials termed nanoyeasts displaying antibody single-chain variable fragments (scFvs) against diagnostic targets are a promising alternative to monoclonal antibodies (mAbs). A potential limitation for translating nanoyeasts into diagnostic tools is batch-to-batch variability. Herein, we demonstrate a systematic approach for cost-efficient production of highly specific nanoyeasts that enabled accurate dengue virus (DENV) detection by immunoassay (2.5% CV). Yeasts bioengineered to surface express DENV-specific scFvs (up to 66% of the total cell population) were fragmented into nanoyeast fractions trialing sonication, bead beating, and high-pressure disruption methods. Nanoyeast fractions from sonication had optimal target binding, uniform particle size (±89 nm), were stable, and retained diagnostic activity for 7 days at 37 °C compared to traditional mAbs that lost activity after 1 day at 37 °C. We engineered a panel of nanoyeast scFvs targeting DENV nonstructural protein 1 (NS1): (i) specific for serotyping DENV 1-4 and (ii) cross-reactive anti-DENV scFvs that are suitable for "yes/no" diagnostic applications. We demonstrate highly specific nanoyeast scFvs for serotyping DENV. We show that nanoyeast scFvs specifically detect NS1 in simulated patient plasma with a limit of detection of 250 ng/mL, the concentration found in infected patients.


Assuntos
Vírus da Dengue , Dengue , Anticorpos de Cadeia Única , Anticorpos Antivirais , Materiais Biocompatíveis , Dengue/diagnóstico , Vírus da Dengue/genética , Humanos , Anticorpos de Cadeia Única/genética , Proteínas não Estruturais Virais
14.
Biotechnol Bioeng ; 106(3): 367-75, 2010 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-20178123

RESUMO

Typically, single chain Fv antibodies are unable to fold properly under a reducing cytoplasm because of the reduction of disulfide bonds. The inability to fold limits both the production of the functional scFvs and their targeting against antigens, which are generally executed in a reducing cytoplasm. In this study, the target scFv CDR was grafted with stable human consensus framework sequences, which enabled the generation of a foldable scFv in a reducing cytoplasm of Escherichia coli. Additionally, the structural features affecting the folding efficiency of the engineered scFv were identified by analyzing the predicted structure. An anti-c-Met scFv, which was a cytoplasmic non-foldable protein, was redesigned as the model system. This study confirmed that the engineered anti-c-Met scFv was folded into its native form in the cytoplasm of E. coli BL21(DE3) without a significant loss in the specific binding activity against c-Met antigen. The structures of the wild-type anti-c-Met scFv and the engineered scFv were predicted using homology modeling. A comparative analysis based on the sequence and structure showed that the hydrophobicity of 12 solvent exposed residues decreased, and two newly formed salt bridges might have improved the folding efficiency of the engineered scFv under the reducing condition.


Assuntos
Proteínas Proto-Oncogênicas c-met/imunologia , Anticorpos de Cadeia Única/genética , Anticorpos de Cadeia Única/imunologia , Citoplasma/química , Citoplasma/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Humanos , Modelos Moleculares , Oxirredução , Ligação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/imunologia , Proteínas Recombinantes/metabolismo , Anticorpos de Cadeia Única/química , Anticorpos de Cadeia Única/metabolismo
15.
J Phys Chem Lett ; 11(14): 5549-5554, 2020 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-32602718

RESUMO

The construction of allosteric protein switches is a key goal of synthetic biology. Such switches can be compiled into signaling systems mimicking information and energy processing systems of living organisms. Here we demonstrate construction of a biocatalytic electrode functionalized with a recombinant chimeric protein between pyrroloquinoline quinone-dependent glucose dehydrogenase and calmodulin. This electrode could be activated by calmodulin-binding peptide and showed a high bioelectrocatalytic current (ca. 300 µA) due to efficient direct electron transfer. In order to expand the types of inputs that can be used to activate the developed electrode, we constructed a caged version of calmodulin-binding peptide that could be proteolytically uncaged using a protease of choice. Finally, the complexity of the switchable bioelectrochemical system was further increased by the use of almost any kind of molecule/biomolecule or electronic signal, unequivocally proving the orthogonality of the aforementioned system.


Assuntos
Calmodulina/metabolismo , Glucose Desidrogenase/metabolismo , Proteínas Recombinantes de Fusão/metabolismo , Regulação Alostérica , Cálcio/metabolismo , Calmodulina/química , Calmodulina/genética , Proteínas de Ligação a Calmodulina/metabolismo , Técnicas Eletroquímicas/instrumentação , Eletrodos , Enzimas Imobilizadas/química , Enzimas Imobilizadas/metabolismo , Glucose/química , Glucose Desidrogenase/química , Mutação , Oxirredução , Ligação Proteica , Proteínas Recombinantes de Fusão/química
16.
Chem Commun (Camb) ; 56(64): 9206-9209, 2020 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-32662462

RESUMO

We report a novel approach for magneto-controlled activation of an artificial electro-enzymatic cascade. The input signal triggers release of a caged ligand peptide, its proteolytic processing and activation of an artificial allosteric enzyme based on PQQ-dependent glucose dehydrogenase. The developed cascade was used to assemble a magneto-controlled biofuel cell.


Assuntos
Fontes de Energia Bioelétrica , Proteínas de Ligação a Calmodulina/química , Calmodulina/química , Glucose Desidrogenase/química , Nanopartículas de Magnetita/química , Alanina Transaminase/química , Regulação Alostérica , Aminoácido Oxirredutases/química , Técnicas Eletroquímicas/instrumentação , Técnicas Eletroquímicas/métodos , Eletrodos , Enzimas Imobilizadas/química , Glucose/química , Campos Magnéticos , Nanotubos de Carbono/química , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/química , Proteínas Recombinantes de Fusão/química
17.
ACS Synth Biol ; 9(6): 1306-1314, 2020 06 19.
Artigo em Inglês | MEDLINE | ID: mdl-32339455

RESUMO

The ability of proteins to interconvert unrelated biochemical inputs and outputs underlays most energy and information processing in biology. A common conversion mechanism involves a conformational change of a protein receptor in response to a ligand binding or a covalent modification, leading to allosteric activity modulation of the effector domain. Designing such systems rationally is a central goal of synthetic biology and protein engineering. A two-component sensory system based on the scaffolding of modules in the presence of an analyte is one of the most generalizable biosensor architectures. An inherent problem of such systems is dependence of the response on the absolute and relative concentrations of the components. Here we use the example of two-component sensory systems based on calmodulin-operated synthetic switches to analyze and address this issue. We constructed "caged" versions of the activating domain thereby creating a thermodynamic barrier for spontaneous activation of the system. We demonstrate that the caged biosensor architectures could operate at concentrations spanning 3 orders of magnitude and are applicable to electrochemical, luminescent, and fluorescent two-component biosensors. We analyzed the activation kinetics of the caged biosensors and determined that the core allosteric switch is likely to be the rate limiting component of the system. These findings provide guidance for predictable engineering of robust sensory systems with inputs and outputs of choice.


Assuntos
Técnicas Biossensoriais/métodos , Calmodulina/metabolismo , Regulação Alostérica/efeitos da radiação , Calmodulina/genética , Glucose 1-Desidrogenase/genética , Glucose 1-Desidrogenase/metabolismo , Cinética , Ligantes , Luz , Peptídeos/química , Peptídeos/metabolismo , Engenharia de Proteínas , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/isolamento & purificação , Sirolimo/química , Sirolimo/metabolismo
18.
J Microbiol Biotechnol ; 18(6): 1186-90, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18600066

RESUMO

Single-chain Fv (scFv) antibody against c-Met is expected to be employed in clinical treatment or imaging of cancer cells owing to the important biological roles of c-Met in the proliferation of malignancies. Here, we show that the productivity of scFv against c-Met in Escherichia coli is significantly influenced by the orientation of its variable domains. We generated anti-c-Met scFv antibodies with two different domain orders (i.e., VL-linker-VH and VH-linker-VL), expressed them in the cytoplasm of E. coli trx/ gor deleted mutant, and compared their specific activities as well as their productivities. Productivity of total and functional anti-c-Met scFv with VH/VL orientation was more than five times higher than that with VL/VH format. Coexpression of DsbC enhanced the yield of soluble amounts of anti-c-Met scFv protein for both constructs. The purified scFv antibodies of the two different formats exhibited almost the same antigen-binding activities. We also compared the productivities and specific activities of anti-c-Met diabodies with VH/VL or VL/VH formats and obtained similar results to the case of scFv antibodies.


Assuntos
Fragmentos de Imunoglobulinas/imunologia , Região Variável de Imunoglobulina/imunologia , Engenharia de Proteínas , Proteínas Proto-Oncogênicas c-met/imunologia , Anticorpos Biespecíficos/imunologia , Anticorpos Antineoplásicos/imunologia , Afinidade de Anticorpos , Clonagem Molecular , Escherichia coli/genética , Escherichia coli/imunologia , Humanos , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Leves de Imunoglobulina/imunologia , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Especificidade por Substrato
19.
ACS Synth Biol ; 6(12): 2241-2247, 2017 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-28937743

RESUMO

The molecular recognition of carbohydrates plays a fundamental role in many biological processes. However, the development of carbohydrate-binding reagents for biomedical research and use poses a challenge due to the generally poor affinity of proteins toward sugars in aqueous solution. Here, we describe the effective molecular recognition of pyranose monosaccharides (in particular, galactose and mannose) by a rationally designed protein receptor based on the human lipocalin scaffold (Anticalin). Complexation relies on reversible covalent cis-diol boronate diester formation with a genetically encoded l-boronophenylalanine (Bpa) residue which was incorporated as a non-natural amino acid at a sterically permissive position in the ligand pocket of the Anticalin, as confirmed by X-ray crystallography. Compared with the metal-ion and/or avidity-dependent oligovalent lectins that prevail in nature, our approach offers a novel and promising route to generate tight sugar-binding reagents both as research reagents and for biomedical applications.


Assuntos
Ácidos Borônicos/química , Galactose/química , Lipocalinas/química , Manose/química , Sítios de Ligação , Cristalografia por Raios X , Humanos , Lipocalinas/genética
20.
J Phys Chem B ; 119(43): 13825-33, 2015 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-26201933

RESUMO

The design of binding sites for divalent metals in artificial proteins is a productive platform for examining the characteristics of metal-ligand interactions. In this report, we investigate the spectroscopic properties of small peptides and four-helix bundles that bind Cu(II). Three small peptides, consisting of 15 amino acid residues, were designed to have two arms, each containing a metal-binding site comprised of different combinations of imidazole and carboxylate side chains. Two four-helix bundles each had a binding site for a central dinuclear metal cofactor, with one design incorporating additional potential metal ligands at two identical sites. The small peptides displayed pH-dependent, metal-induced changes in the circular dichroism spectra, consistent with large changes in the secondary structure upon metal binding, while the spectra of the four-helix bundles showed a predominant α-helix content but only small structural changes upon metal binding. Electron paramagnetic resonance spectra were measured at X-band revealing classic Cu(II) axial patterns with hyperfine coupling peaks for the small peptides and four-helix bundles exhibiting a range of values that were related to the specific chemical natures of the ligands. The variety of electronic structures allow us to define the distinctive environment of each metal-binding site in these artificial systems, including the designed additional binding sites in one of the four-helix bundles.


Assuntos
Cobre/química , Metaloproteínas/química , Sítios de Ligação , Ácidos Carboxílicos/química , Espectroscopia de Ressonância de Spin Eletrônica , Concentração de Íons de Hidrogênio , Imidazóis/química , Ligantes , Modelos Moleculares
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA