Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Analyst ; 145(8): 3148-3156, 2020 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-32191233

RESUMO

Continued interest in protein therapeutics has motivated the development of improved bioanalytical tools to support development programs. LC-MS offers specificity, sensitivity, and multiplexing capabilities without the need for target-specific reagents, making it a valuable alternative to ligand binding assays. Immunoaffinity purification (IP) and enzymatic digestion are critical, yet extensive and time-consuming components of the "gold standard" bottom-up approach to LC-MS-based protein quantitation. In the present work, commercially available technology, based on membrane-immobilized reagents in spin column and plate format, is applied to reduce IP and digestion times from hours to minutes. For a standard monoclonal antibody, the lower limit of quantitation was 0.1 ng µL-1 compared to 0.05 ng µL-1 for the standard method. A pharmacokinetics (PK) study dosing Herceptin in rat was analyzed by both the membrane and the standard method with a total sample processing time of 4 h and 20 h, respectively. The calculated concentrations at each time point agreed within 8% between both methods, and PK values including area under the curve (AUC), half-life (T1/2), mean residence time (MRT), clearance (CL), and volume of distribution (Vdss) agreed within 6% underscoring the utility of the membrane methodology for quantitative bioanalysis workflows.


Assuntos
Cromatografia de Afinidade/métodos , Enzimas Imobilizadas/química , Membranas Artificiais , Trastuzumab/análise , Sequência de Aminoácidos , Animais , Masculino , Estudo de Prova de Conceito , Proteólise , Ratos Sprague-Dawley , Proteína Estafilocócica A/química , Fatores de Tempo , Trastuzumab/química , Trastuzumab/isolamento & purificação , Trastuzumab/farmacocinética , Tripsina/química
2.
Methods Enzymol ; 624: 249-264, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31370932

RESUMO

Over the past decade, photoswitchable molecules have been emerging as attractive tools for investigating biological processes with spatiotemporal resolution in a minimally invasive fashion. Photoswitches built on light-sensitive proteins or domains have significantly advanced neuronal and cellular studies. To install photosensitivity to general proteins and to enable high specificity for modulation, photoswitchable click amino acids (PSCaas) based on azobenzene have been developed and recently genetically incorporated into proteins via the expansion of the genetic code. PSCaas allow targeting selected sites in a protein for high specificity and are generally applicable to various proteins. In addition, PSCaas contain a click functional group, which selectively reacts with an appropriately positioned cysteine forming a photocontrollable bridge on the protein in situ. The photocontrollable bridge enables reversible modulation of the secondary structure of the spanned region and thus the function of the protein. In this chapter we describe the design and genetic encoding of PSCaa. Protocols are presented for incorporating PSCaa into a model protein calmodulin to build the bridge followed by photocontrol of calmodulin's conformation and binding function.


Assuntos
Aminoácidos/genética , Compostos Azo/química , Optogenética/métodos , Proteínas/genética , Aminoácidos/química , Animais , Calmodulina/química , Calmodulina/genética , Escherichia coli/química , Escherichia coli/genética , Código Genético , Humanos , Isomerismo , Luz , Modelos Moleculares , Processos Fotoquímicos , Proteínas/química
3.
Biosens Bioelectron ; 118: 188-194, 2018 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-30077871

RESUMO

Neuronal nitric oxide synthase (nNOS) is an enzyme responsible for catalyzing the production of the crucial cellular signalling molecule, nitric oxide (NO), through its interaction with the PDZ domain of α-syntrophin protein. In this study, a novel light-driven photoswitchable peptide-based biosensor, modelled on the nNOS ß-finger, is used to detect and control its interaction with α-syntrophin. An azobenzene photoswitch incorporated into the peptide backbone allows reversible switching between a trans photostationary state devoid of secondary structure, and a cis photostationary state possessing a well-defined antiparallel ß-strand geometry, as revealed by molecular modelling. Electrochemical impedance spectroscopy (EIS) is used to successfully detect the interaction between the gold electrode bound peptide in its cis photostationary state and a wide range of concentrations of α-syntrophin protein, highlighting both the qualitative and quantitative properties of the sensor. Furthermore, EIS demonstrates that the probe in its random trans photostationary state does not bind to the target protein. The effectiveness of the biosensor is further endorsed by the high thermal stability of the photostationary state of the cis-isomer, and the ability to actively control biomolecular interactions using light. This approach allows detection and control of binding to yield a regenerable on-off biosensor.


Assuntos
Técnicas Biossensoriais/métodos , Proteínas/metabolismo , Peptídeos , Ligação Proteica , Estrutura Secundária de Proteína
4.
J Am Chem Soc ; 140(35): 11058-11066, 2018 09 05.
Artigo em Inglês | MEDLINE | ID: mdl-30132658

RESUMO

Acidic vesicles and organelles play fundamental roles in a broad range of cellular events such as endocytosis, lysosomal degradation, synaptic transmission, pathogen fate, and drug delivery. Fluorescent reporters will be invaluable for studying these complex and multifunctional systems with spatiotemporal resolution, yet common fluorescent proteins are generally nonfluorescent at acidic conditions due to the decrease of anionic chromophores upon protonation, but are fluorescent at physiological pH, creating interfering fluorescence from nonvesicle regions. Here we developed a novel acid-brightening fluorescent protein (abFP) that fluoresces strongly at acidic pH but is nonfluorescent at or above neutral pH, boasting a pH profile opposite to that of common fluorescent proteins. Through expansion of the genetic code, we incorporated a quinoline-containing amino acid Qui into the chromophore of EGFP to reverse the chromophore charge. Protonation of Qui rendered a cationic chromophore, which resulted in unique fluorescence increase only at acidic pH in vitro, in E. coli cells, and on the mammalian cell surface. We further demonstrated that abFP-tagged δ opioid receptors were fluorescently imaged in lysosome showing distinct features and without background fluorescence from other cellular regions, whereas EGFP-tagged receptors were invisible in lysosome. This Qui-rendered cationic chromophore strategy may be generally applied to other fluorescent proteins to generate a palette of colors for acidic imaging with minimal background, and these abFPs should facilitate the study of molecules in association with various acidic vesicles and organelles in different cells and model organisms.


Assuntos
Proteínas Luminescentes/química , Quinolinas/química , Aminoácidos/química , Aminoácidos/genética , Portadores de Fármacos/química , Sistemas de Liberação de Medicamentos , Fluorescência , Código Genético , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Proteínas Luminescentes/genética , Modelos Moleculares , Estrutura Molecular , Organelas/química , Organelas/genética
5.
Nat Chem Biol ; 13(8): 842-844, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28604697

RESUMO

Access to phosphoproteins with stoichiometric and site-specific phosphorylation status is key to understanding the role of protein phosphorylation. Here we report an efficient method to generate pure, active phosphotyrosine-containing proteins by genetically encoding a stable phosphotyrosine analog that is convertible to native phosphotyrosine. We demonstrate its general compatibility with proteins of various sizes, phosphotyrosine sites and functions, and reveal a possible role of tyrosine phosphorylation in negative regulation of ubiquitination.


Assuntos
Código Genético/genética , Fosfotirosina/genética , Fosfotirosina/metabolismo , Proteínas/genética , Proteínas/metabolismo , Animais , Fosforilação , Proteínas/química , Tirosina/metabolismo
6.
Elife ; 62017 05 23.
Artigo em Inglês | MEDLINE | ID: mdl-28534738

RESUMO

Engineering light-sensitivity into proteins has wide ranging applications in molecular studies and neuroscience. Commonly used tethered photoswitchable ligands, however, require solvent-accessible protein labeling, face structural constrains, and are bulky. Here, we designed a set of optocontrollable NMDA receptors by directly incorporating single photoswitchable amino acids (PSAAs) providing genetic encodability, reversibility, and site tolerance. We identified several positions within the multi-domain receptor endowing robust photomodulation. PSAA photoisomerization at the GluN1 clamshell hinge is sufficient to control glycine sensitivity and activation efficacy. Strikingly, in the pore domain, flipping of a M3 residue within a conserved transmembrane cavity impacts both gating and permeation properties. Our study demonstrates the first detection of molecular rearrangements in real-time due to the reversible light-switching of single amino acid side-chains, adding a dynamic dimension to protein site-directed mutagenesis. This novel approach to interrogate neuronal protein function has general applicability in the fast expanding field of optopharmacology.


Assuntos
Luz , Receptores de Glutamato/metabolismo , Receptores de Glutamato/efeitos da radiação , Animais , Isomerismo , Camundongos , Ratos , Receptores de Glutamato/química , Receptores de Glutamato/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/efeitos da radiação
7.
J Am Chem Soc ; 138(45): 14832-14835, 2016 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-27797495

RESUMO

Chemical reactivity is essential for functional modification of biomolecules with small molecules and the development of covalent drugs. The reactivity between a chemical functional group of a small molecule and that of a large biomolecule cannot be reliably predicted from the reactivity of the corresponding functional groups separately installed on two small molecules, because the proximity effect on reactivity resulting from the binding of the small molecule to the biomolecule is challenging to achieve by mixing two small molecules. Here we present a new strategy to determine the chemical reactivity of two functional groups in the context of close proximity afforded by proteins. The functional groups to be tested were separately installed at the interface of two interacting proteins in the format of amino acid side chains via the expansion of the genetic code. Reaction of the two functional groups resulted in covalent cross-linking of interacting proteins, readily detectable by gel electrophoresis. Using this strategy, we evolved new synthetases to genetically encode Nε-fluoroacetyllysine (FAcK), an isosteric fluorine analogue of acetyllysine. We demonstrated that fluoroacetamide installed on FAcK, previously thought inert to biological functional groups, actually reacted with the thiol group of cysteine when in proximity. This strategy should be valuable for accurately evaluating chemical reactivity of small molecules toward large biomolecules, which will help avoid undesired side reactions of drugs and expand the repertoire of functional groups to covalently target biomolecules.


Assuntos
Ligases/química , Acetilação , Aminoácidos/química , Ligases/metabolismo , Lisina/química , Lisina/genética , Lisina/metabolismo , Methanosarcina/química , Conformação Molecular
8.
Nat Commun ; 7: 11964, 2016 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-27321135

RESUMO

Fluorescence labelling of an intracellular biomolecule in native living cells is a powerful strategy to achieve in-depth understanding of the biomolecule's roles and functions. Besides being nontoxic and specific, desirable labelling probes should be highly cell permeable without nonspecific interactions with other cellular components to warrant high signal-to-noise ratio. While it is critical, rational design for such probes is tricky. Here we report the first predictive model for cell permeable background-free probe development through optimized lipophilicity, water solubility and charged van der Waals surface area. The model was developed by utilizing high-throughput screening in combination with cheminformatics. We demonstrate its reliability by developing CO-1 and AzG-1, a cyclooctyne- and azide-containing BODIPY probe, respectively, which specifically label intracellular target organelles and engineered proteins with minimum background. The results provide an efficient strategy for development of background-free probes, referred to as 'tame' probes, and novel tools for live cell intracellular imaging.


Assuntos
Azidas/química , Compostos de Boro/química , Ciclo-Octanos/química , Corantes Fluorescentes/síntese química , Imagem Molecular/métodos , Coloração e Rotulagem/métodos , Animais , Células CHO , Linhagem Celular Tumoral , Nucléolo Celular/metabolismo , Nucléolo Celular/ultraestrutura , Cricetulus , Desenho de Fármacos , Corantes Fluorescentes/metabolismo , Expressão Gênica , Complexo de Golgi/metabolismo , Complexo de Golgi/ultraestrutura , Ensaios de Triagem em Larga Escala , Humanos , Lisossomos/metabolismo , Lisossomos/ultraestrutura , Mitocôndrias/metabolismo , Mitocôndrias/ultraestrutura , Osteoblastos/metabolismo , Osteoblastos/ultraestrutura , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Razão Sinal-Ruído
9.
Chem Commun (Camb) ; 52(29): 5140-3, 2016 Apr 14.
Artigo em Inglês | MEDLINE | ID: mdl-26996321

RESUMO

Although small molecule covalent inhibitors have been widely explored, macromolecular covalent inhibitors are more difficult to design and implement. Here we present a strategy to enable a peptide to bind to its target protein covalently via proximity-enabled bioreactivity, improving its activity of inhibiting the p53-Mdm4 interaction by 10-fold.


Assuntos
Proteínas Nucleares/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Mapas de Interação de Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Sequência de Aminoácidos , Proteínas de Ciclo Celular , Descoberta de Drogas , Humanos , Modelos Moleculares , Proteínas Nucleares/antagonistas & inibidores , Mapeamento de Interação de Proteínas , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteína Supressora de Tumor p53/antagonistas & inibidores
10.
J Am Chem Soc ; 137(35): 11218-21, 2015 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-26301538

RESUMO

Optical modulation of proteins provides superior spatiotemporal resolution for understanding biological processes, and photoswitches built on light-sensitive proteins have been significantly advancing neuronal and cellular studies. Small molecule photoswitches could complement protein-based switches by mitigating potential interference and affording high specificity for modulation sites. However, genetic encodability and responsiveness to nonultraviolet light, two desired properties possessed by protein photoswitches, are challenging to be engineered into small molecule photoswitches. Here we developed a small molecule photoswitch that can be genetically installed onto proteins in situ and controlled by visible light. A pentafluoro azobenzene-based photoswitchable click amino acid (F-PSCaa) was designed to isomerize in response to visible light. After genetic incorporation into proteins via the expansion of the genetic code, F-PSCaa reacts with a nearby cysteine within the protein generating an azo bridge in situ. The resultant bridge is switchable by visible light and allows conformation and binding of CaM to be regulated by such light. This photoswitch should prove valuable in optobiology for its minimal interference, site flexibility, genetic encodability, and response to the more biocompatible visible light.


Assuntos
Compostos Azo/química , Luz , Optogenética/métodos , Proteínas/química , Proteínas/genética , Aminoácidos/química , Modelos Moleculares , Conformação Proteica , Estereoisomerismo
11.
Cell Rep ; 9(5): 1946-1958, 2014 Dec 11.
Artigo em Inglês | MEDLINE | ID: mdl-25464845

RESUMO

Protein-protein interactions (PPIs) play central roles in orchestrating biological processes. While some PPIs are stable, many important ones are transient and hard to detect with conventional approaches. We developed ReBiL, a recombinase enhanced bimolecular luciferase complementation platform, to enable detection of weak PPIs in living cells. ReBiL readily identified challenging transient interactions between an E3 ubiquitin ligase and an E2 ubiquitin-conjugating enzyme. ReBiL's ability to rapidly interrogate PPIs in diverse conditions revealed that some stapled α-helical peptides, a class of PPI antagonists, induce target-independent cytosolic leakage and cytotoxicity that is antagonized by serum. These results explain the requirement for serum-free conditions to detect stapled peptide activity, and define a required parameter to evaluate for peptide antagonist approaches. ReBiL's ability to expedite PPI analysis, assess target specificity and cell permeability, and reveal off-target effects of PPI modifiers should facilitate the development of effective, cell-permeable PPI therapeutics and the elaboration of diverse biological mechanisms.


Assuntos
Mapeamento de Interação de Proteínas/métodos , Proteínas de Ciclo Celular , Linhagem Celular Tumoral , Genes Reporter , Humanos , Luciferases de Vaga-Lume/biossíntese , Mutação de Sentido Incorreto , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Recombinases/fisiologia , Proteína Supressora de Tumor p53/genética
12.
Angew Chem Int Ed Engl ; 53(15): 3932-6, 2014 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-24615769

RESUMO

The ability to reversibly control protein structure and function with light would offer high spatiotemporal resolution for investigating biological processes. To confer photoresponsiveness on general proteins, we genetically incorporated a set of photoswitchable click amino acids (PSCaas), which contain both a reversible photoswitch and an additional click functional group for further modifications. Orthogonal tRNA-synthetases were evolved to genetically encode PSCaas bearing azobenzene with an alkene, keto, or benzyl chloride group in E. coli and in mammalian cells. After incorporation into calmodulin, the benzyl chloride PSCaa spontaneously generated a covalent protein bridge by reacting with a nearby cysteine residue through proximity-enabled bioreactivity. The resultant azobenzene bridge isomerized in response to light, thereby changing the conformation of calmodulin. These genetically encodable PSCaas will prove valuable for engineering photoswitchable bridges into proteins for reversible optogenetic regulation.


Assuntos
Aminoácidos/química , Escherichia coli/metabolismo , Química Click , Código Genético , Conformação Molecular , Optogenética , Engenharia de Proteínas
13.
Chembiochem ; 13(18): 2657-60, 2012 Dec 21.
Artigo em Inglês | MEDLINE | ID: mdl-23161824

RESUMO

Aggregation of amyloid ß (Aß(1-42)), causing toxicity, is a critical step in Alzheimer's disease (AD). AD studies are difficult to compare because Aß(1-42) aggregation is poorly controllable under physiological conditions. To control aggregation and toxicity, we engineered light-switchable Aß(1-42) analogues that enable controllable conversion of nontoxic fibrils into toxic oligomers simply by illumination.


Assuntos
Peptídeos beta-Amiloides/química , Peptídeos beta-Amiloides/toxicidade , Luz , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/toxicidade , Engenharia de Proteínas , Multimerização Proteica/efeitos da radiação , Sequência de Aminoácidos , Linhagem Celular Tumoral , Humanos , Dados de Sequência Molecular , Estrutura Secundária de Proteína/efeitos da radiação
14.
Beilstein J Org Chem ; 8: 884-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23015838

RESUMO

Photoswitchable click amino acids (PSCaa) are amino acids bearing a side chain consisting of a photoswitchable unit elongated with a functional group that allows for a specific click reaction, such as an alkene that can react with the thiol group of a cysteine residue. An intramolecular click reaction results in the formation of a photoswitchable bridge, which can be used for controlling conformational domains in peptides and proteins. The ability to control conformations as well as the efficiency of the intramolecular bridging depends on the length of the PSCaa side chain and the distance to the cysteine residue to be clicked with. On comparing i,i+4 and i,i+7 spacings of PSCaa and cysteine in a model peptide without a preferred conformation, it was seen that the thiol-ene click reaction takes place efficiently in both cases. Upon induction of an α-helical structure by the addition of trifluoroethanol, the thiol click reaction occurs preferentially with the i,i+4 spacing. Even in the presence of glutathione as an additional thiol the click reaction of the PSCaa occurs intramolecularly with the cysteine rather than with the glutathione, indicating that the click reaction may be used even under reducing conditions occurring in living cells.

15.
Chembiochem ; 12(17): 2555-9, 2011 Nov 25.
Artigo em Inglês | MEDLINE | ID: mdl-21998087

RESUMO

Click the switch: By using a photoswitchable click amino acid (PSCaa) a light-induced intramolecular thiol-ene click reaction with a neighboring cysteine under very mild conditions results in an azobenzene bridge. By expanding the genetic code for PSCaa the specific incorporation of photoswitch units into proteins in living cells can result in an exciting approach for studying light-controllable activity, in vivo.


Assuntos
Aminoácidos/química , Luz , Compostos Azo/química , Química Click , Cisteína/química , Isomerismo , Conformação Molecular , Processos Fotoquímicos , Compostos de Sulfidrila/química , Urocortinas/química
18.
Chemphyschem ; 8(11): 1713-21, 2007 Aug 06.
Artigo em Inglês | MEDLINE | ID: mdl-17614347

RESUMO

The molecule HTI, which combines hemithioindigo and hemistilbene molecular parts, allows reversible switching between two isomeric states. Photochromic behaviour of the HTI molecule is observed by irradiation with UV/Vis light. The photochemical reaction, a Z/E isomerization around the central double bond connecting the two molecular parts, is investigated by transient absorption and emission spectroscopy. For a special HTI molecule, namely, an omega-amino acid, the Z-->E isomerization process occurs on a timescale of 30 ps. In the course of the reaction fast processes on the 1-10 ps timescale are observed which point to motions of the molecule on the potential-energy surface of the excited state. The combination of transient absorption experiments in the visible spectral range with time-resolved fluorescence and infrared measurements reveal a photochemical pathway with three intermediate states. Together with a theoretical modelling procedure the experiments point to a sequential reaction scheme and give indications of the nature of the involved intermediates.


Assuntos
Aminoácidos/efeitos da radiação , Modelos Químicos , Tiofenos/química , Aminoácidos/química , Fotoquímica , Análise Espectral , Estereoisomerismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA