Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Adv Exp Med Biol ; 1163: 253-278, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31707707

RESUMO

Deregulation of protein kinase activity has been linked to many diseases ranging from cancer to AIDS and neurodegenerative diseases. Not surprisingly, drugging the human kinome - the complete set of kinases encoded by the human genome - has been one of the major drug discovery pipelines. Majority of the approved clinical kinase inhibitors target the ATP binding site of kinases. However, the remarkable sequence and structural similarity of ATP binding pockets of kinases make selective inhibition of kinases a daunting task. To circumvent these issues, allosteric inhibitors that target sites other than the orthosteric ATP binding pocket have been developed. The structural diversity of the allosteric sites allows these inhibitors to have higher selectivity, lower toxicity and improved physiochemical properties and overcome drug resistance associated with the use of conventional kinase inhibitors. In this chapter, we will focus on the allosteric inhibitors of selected serine/threonine kinases, outline the benefits of using these inhibitors and discuss the challenges and future opportunities.


Assuntos
Inibidores de Proteínas Quinases , Proteínas Serina-Treonina Quinases , Regulação Alostérica/efeitos dos fármacos , Sítio Alostérico , Humanos , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/metabolismo
3.
Electrophoresis ; 38(19): 2530-2535, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28683171

RESUMO

Herein, a novel assay has been developed for monitoring PreScission protease (His-PSP) mediated enzyme cleavage of ATTO 590 labeled peptide substrate (ATTO-LEV). This novel method is based on combining the use of capillary electrophoresis and fluorescence detection (CE-FL) to dynamically monitor the enzyme cleavage activity. A multivalent peptide substrate was first constructed by immobilizing His-tagged ATTO 590 labeled peptide substrate (ATTO-LEVH6) onto the surface of CdSe/ZnS quantum dots (QDs). Once successfully immobilized, the novel multivalent peptide substrate resulted in the Förster resonance energy transfer (FRET) from QDs to ATTO 590. The ATTO-LEVH6-QD assembly was then incubated with His-PSP to study the proteolytic cleavage of surface bound ATTO-LEVH6 by CE-FL. Our data suggests that PreScission-mediated proteolytic cleavage is enzyme concentration- and incubation time-dependent. By combining capillary electrophoresis, QDs and FRET, our study herein not only provides a new method for the detection and dynamically monitoring of PSP enzyme cleavage activity, but also can be extended to the detection of many other enzymes and proteases.


Assuntos
Eletroforese Capilar/métodos , Peptídeo Hidrolases/química , Peptídeos/química , Pontos Quânticos/química , Proteínas Virais/química , Transferência Ressonante de Energia de Fluorescência/métodos , Corantes Fluorescentes/química , Proteólise , Rhinovirus/enzimologia
4.
PLoS One ; 9(10): e110955, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25354194

RESUMO

Microtubules are a highly validated target in cancer therapy. However, the clinical development of tubulin binding agents (TBA) has been hampered by toxicity and chemoresistance issues and has necessitated the search for new TBAs. Here, we report the identification of a novel cell permeable, tubulin-destabilizing molecule--4,5,6,7-tetrahydro-1H-indazole-3-carboxylic acid [1p-tolyl-meth-(E)-ylidene]-hydrazide (termed as Suprafenacine, SRF). SRF, identified by in silico screening of annotated chemical libraries, was shown to bind microtubules at the colchicine-binding site and inhibit polymerization. This led to G2/M cell cycle arrest and cell death via a mitochondria-mediated apoptotic pathway. Cell death was preceded by loss of mitochondrial membrane potential, JNK-mediated phosphorylation of Bcl-2 and Bad, and activation of caspase-3. Intriguingly, SRF was found to selectively inhibit cancer cell proliferation and was effective against drug-resistant cancer cells by virtue of its ability to bypass the multidrug resistance transporter P-glycoprotein. Taken together, our results suggest that SRF has potential as a chemotherapeutic agent for cancer treatment and provides an alternate scaffold for the development of improved anti-cancer agents.


Assuntos
Antineoplásicos/farmacologia , Hidrazinas/farmacologia , Indazóis/farmacologia , Microtúbulos/efeitos dos fármacos , Sequência de Aminoácidos , Animais , Antineoplásicos/química , Antineoplásicos/isolamento & purificação , Apoptose , Sítios de Ligação , Colchicina/farmacologia , Pontos de Checagem da Fase G2 do Ciclo Celular , Células HeLa , Humanos , Hidrazinas/química , Hidrazinas/isolamento & purificação , Indazóis/química , Indazóis/isolamento & purificação , Potencial da Membrana Mitocondrial , Camundongos , Microtúbulos/química , Microtúbulos/metabolismo , Dados de Sequência Molecular , Células PC12 , Ligação Proteica , Ratos , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia
5.
Curr Med Chem ; 16(34): 4527-43, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19903152

RESUMO

Methods that allow visualisation of proteins in living systems, in real time have been key to our understanding of the molecular underpinnings of life. Although the use of genetically encoded fusions to fluorescent proteins have greatly advanced such studies, the large size of these tags and their ability to perturb protein activity has been major limitations. Attempts to circumvent these issues have seen the genesis of complementary strategies to chemically label/modify proteins. Thus, chemical labelling approaches seek to "decorate" biomolecules in live cells through the site-specific introduction of a small, non-native chemical tag (or reporter group). The introduced tag is minimally invasive such that the activity and/or function of the target molecule in not perturbed/compromised by its inclusion. In most cases, this modification is brought about by fusing target biomolecules to protein domains/peptide tags or via the incorporation of reactive "handles" by either exploiting the cell's biosynthetic machinery or during protein synthesis. Selective tagging of the biomolecule then proceeds via a bioorthogonal chemical reaction following exogenous addition of probe(s). Depending on the nature of the probe, the method can be applied to either visualise/track the dynamics of target molecule(s) in their native cellular milieu or for affinity enrichment for further downstream applications. The versatility of these approaches has been demonstrated by their ability to tag not just proteins but also intractable biomolecules like lipids and glycans. In this review, we summarise the various strategies available to "chemically" tag proteins and provide a comparative analysis their advantages and disadvantages. We also highlight the many creative applications of such methodologies and discuss their future prospects.


Assuntos
Engenharia de Proteínas/métodos , Proteínas/química , Coloração e Rotulagem/métodos , Animais , Biotinilação , Corantes Fluorescentes/química , Humanos , Proteínas/genética , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/genética
6.
Methods Enzymol ; 462: 195-223, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19632476

RESUMO

This section introduces a simple, rapid, high-throughput methodology for the site-specific biotinylation of proteins for the purpose of fabricating functional protein arrays. Step-by-step protocols are provided to generate biotinylated proteins using in vitro, in vivo, or cell-free systems, together with useful hints for troubleshooting. In vitro and in vivo biotinylation rely on the chemoselective native chemical ligation (NCL) reaction between the reactive alpha-thioester group at the C-terminus of target proteins, generated via intein-mediated cleavage, and the added cysteine biotin. The cell-free system uses a low concentration of biotin-conjugated puromycin. The biotinylated proteins can be either purified or directly captured from crude cellular lysates onto an avidin-functionalized slide to afford the corresponding protein array. The methods were designed to preserve the activity of the immobilized protein such that the arrays provide a highly miniaturized platform to simultaneously interrogate the functional activities of thousands of proteins. This is of paramount significance, as new applications of microarray technologies continue to emerge, fueling their growth as an essential tool for high-throughput proteomic studies.


Assuntos
Inteínas , Peptídeos/química , Análise Serial de Proteínas/métodos , Processamento de Proteína , Proteínas/química , Animais , Avidina/metabolismo , Biotina/análogos & derivados , Biotina/química , Biotinilação/métodos , Cisteína/química , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/isolamento & purificação , Humanos , Proteínas Imobilizadas , Estrutura Terciária de Proteína , Proteínas/isolamento & purificação , Proteínas/metabolismo , Puromicina/análogos & derivados , Puromicina/química , Proteínas Recombinantes de Fusão/biossíntese , Proteínas Recombinantes de Fusão/isolamento & purificação , Reagentes de Sulfidrila/química
7.
Bioorg Med Chem ; 17(3): 981-9, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-18261914

RESUMO

The study of dynamic movement and interactions of proteins inside living cells in real time is critical for a better understanding of cellular mechanisms and functions in molecular detail. Genetically encoded fusions to fluorescent protein(s) (FP) have been widely used for this purpose [Annu. Rev. Biochem. 1998, 67, 509-544]. To obviate some of the drawbacks associated with the use of FPs [Curr. Opin. Biotechnol. 2005, 16, 1-6; Nat. Methods2006, 3, 591-596], we report a small molecule-based approach that exploits the unique reactivity between the cysteine residue at the N-terminus of a target protein and cell-permeable, thioester-based small molecule probes resulting in site-specific, covalent tagging of proteins. This approach has been demonstrated by the in vivo labeling of proteins in both bacterial and mammalian systems thereby making it potentially useful for future bioimaging applications.


Assuntos
Sondas Moleculares/química , Proteínas Recombinantes de Fusão/análise , Linhagem Celular , Cisteína/química , Transferência Ressonante de Energia de Fluorescência , Corantes Fluorescentes/química , Humanos , Inteínas , Microscopia de Fluorescência , Sondas Moleculares/metabolismo , Proteínas Recombinantes de Fusão/química , Proteínas Recombinantes de Fusão/metabolismo
9.
Anal Bioanal Chem ; 386(3): 416-26, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16791553

RESUMO

Recent advances in proteomics have provided impetus towards the development of robust technologies for high-throughput studies of enzymes. The term "catalomics" defines an emerging '-omics' field in which high-throughput studies of enzymes are carried out by using advanced chemical proteomics approaches. Of the various available methods, microarrays have emerged as a powerful and versatile platform to accelerate not only the functional annotation but also the substrate and inhibitor specificity (e.g. substrate and inhibitor fingerprinting, respectively) of enzymes. Herein, we review recent developments in the fabrication of various types of microarray technologies (protein-, peptide- and small-molecule-based microarrays) and their applications in high-throughput characterizations of enzymes.


Assuntos
Enzimas/química , Análise em Microsséries/métodos , Análise em Microsséries/tendências , Animais , Inibidores Enzimáticos/química , Enzimas/análise , Humanos , Análise em Microsséries/instrumentação , Especificidade por Substrato
10.
Anal Chim Acta ; 556(1): 69-79, 2006 Jan 18.
Artigo em Inglês | MEDLINE | ID: mdl-17723332

RESUMO

Proteomics deals with the study of proteins, their structures, localizations, posttranslational modifications, functions and interactions with other proteins. The mapping of protein structure-function holds the key to a better understanding of cellular functions under both normal and disease states, which is critical for modern drug discovery. However, the study of human proteome presents scientists with a task much more daunting than the human genome project. In fact, the estimated >100,000 different proteins expressed from 30,000 to 40,000 human genes make it extremely challenging, if not impossible with existing protein analysis techniques, to map the entire cellular functions at the translational level. Consequently, there have been rapid advances in the techniques and methods capable of large-scale proteomic studies. Among them, the recently developed high-throughput screening methods have enabled scientists to analyze proteins quickly and efficiently at an organism-wide scale. Herein, we overview some of these emerging tools for high-throughput protein analysis. In particular, we focus on recent advances in the bioassay development, which has provided sensitive and selective tools for high-throughput identification and characterizations of enzymes. Finally, the recently developed bioimaging techniques to visualize and quantify proteins in living cells are also discussed.

11.
Nat Protoc ; 1(5): 2386-98, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-17406482

RESUMO

This protocol details methodologies for the site-specific biotinylation of proteins using in vitro, in vivo and cell-free systems for the purpose of fabricating functional protein arrays. Biotinylation of recombinant proteins, in vitro as well as in vivo, relies on the chemoselective reaction between cysteine-biotin and a reactive thioester group at the C-terminus of a protein generated via intein-mediated cleavage. The cell-free system utilizes low concentrations of biotin-conjugated puromycin. Unlike other approaches that require tedious and costly downstream steps of protein purification, C-terminal biotinylated proteins can be captured directly onto avidin-functionalized slides from a mixture of other cellular proteins to generate the corresponding protein array. These methods were designed to maintain the integrity and activity of proteins in a microarray format, which potentially allows simultaneous functional assays of thousands of proteins. Assuming that the target proteins have been cloned into the expression vector, transformation of bacterial strain and growth of starter culture would take approximately 2 days. Expression and in vitro protein purification and biotinylation will take approximately 3 days whereas the in vivo method would take approximately 2 days. The cell-free protein biotinylation strategy requires only 6-8 h.


Assuntos
Biotina/química , Análise Serial de Proteínas/métodos , Proteínas/química , Sistema Livre de Células , Cisteína/química , Proteínas/isolamento & purificação
12.
J Am Chem Soc ; 126(44): 14435-46, 2004 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-15521763

RESUMO

The denaturing aspect of current activity-based protein profiling strategies limits the classes of chemical probes to those which irreversibly and covalently modify their targeting enzymes. Herein, we present a complimentary, affinity-based labeling approach to profile enzymes which do not possess covalently bound substrate intermediates. Using a variety of enzymes belonging to the class of metalloproteases, the feasibility of the approach was successfully demonstrated in several proof-of-concept experiments. The design template of affinity-based probes targeting metalloproteases consists of a peptidyl hydroxamate zinc-binding group (ZBG), a fluorescent reporter tag, and a photolabile diazirine group. Photolysis of the photolabile unit in the probe effectively generates a covalent, irreversible linkage between the probe and the target enzyme, rendering the enzyme distinguishable from unlabeled proteins upon separation on a SDS-PAGE gel. A variety of labeling studies were carried out to confirm that the affinity-based approach selectively labeled metalloproteases in the presence of a large excess of other proteins and that the success of the labeling reaction depends intimately upon the catalytic activity of the enzyme. Addition of competitive inhibitors proportionally diminished the extent of enzyme labeling, making the approach useful for potential in situ screening of metalloprotease inhibitors. Using different probes with varying P(1) amino acids, we were able to generate unique "fingerprint" profiles of enzymes which may be used to determine their substrate specificities. Finally, by testing against a panel of yeast metalloproteases, we demonstrated that the affinity-based approach may be used for the large-scale profiling of metalloproteases in future proteomic experiments.


Assuntos
Marcadores de Afinidade/química , Ácidos Hidroxâmicos/química , Metaloproteases/análise , Proteômica/métodos , Bacillus/enzimologia , Metaloproteases/antagonistas & inibidores , Inibidores de Proteases/química , Termolisina/análise , Termolisina/antagonistas & inibidores , Termolisina/química , Leveduras/enzimologia
13.
Chem Commun (Camb) ; (13): 1512-3, 2004 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-15216356

RESUMO

In this article, we report the design and synthesis of a group of novel activity-based probes that target different protease sub-classes based on their substrate specificities, rather than their enzymatic mechanisms. The feasibility of our approach has been demonstrated by using representative members of the different protease sub-classes.


Assuntos
Endopeptidases/análise , Corantes Fluorescentes/síntese química , Amidas/química , Proteômica , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA