Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Assay Drug Dev Technol ; 16(7): 384-396, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30251873

RESUMO

G-protein-coupled receptors (GPCRs) have varying and diverse physiological roles, transmitting signals from a range of stimuli, including light, chemicals, peptides, and mechanical forces. More than 130 GPCRs are orphan receptors (i.e., their endogenous ligands are unknown), representing a large untapped reservoir of potential therapeutic targets for pharmaceutical intervention in a variety of diseases. Current deorphanization approaches are slow, laborious, and usually require some in-depth knowledge about the receptor pharmacology. In this study we describe a cell-based assay to identify small molecule probes of orphan receptors that requires no a priori knowledge of receptor pharmacology. Built upon the concept of pharmacochaperones, where cell-permeable small molecules facilitate the trafficking of mutant receptors to the plasma membrane, the simple and robust technology is readily accessible by most laboratories and is amenable to high-throughput screening. The assay consists of a target harboring a synthetic point mutation that causes retention of the target in the endoplasmic reticulum. Coupled with a beta-galactosidase enzyme-fragment complementation reporter system, the assay identifies compounds that act as pharmacochaperones causing forward trafficking of the mutant GPCR. The assay can identify compounds with varying mechanisms of action including agonists and antagonists. A universal positive control compound circumvents the need for a target-specific ligand. The veracity of the approach is demonstrated using the beta-2-adrenergic receptor. Together with other existing assay technologies to validate the signaling pathways and the specificity of ligands identified, this pharmacochaperone-based approach can accelerate the identification of ligands for these potentially therapeutically useful receptors.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Sondas Moleculares/análise , Sondas Moleculares/química , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/química , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Humanos , Ligantes , Sondas Moleculares/farmacologia , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/farmacologia , Células Tumorais Cultivadas
2.
Elife ; 62017 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-28498099

RESUMO

Cytokine and growth-factor ligands typically signal through homo- or hetero-dimeric cell surface receptors via Janus Kinase (JAK/TYK), or Receptor Tyrosine Kinase (RTK)-mediated trans-phosphorylation. However, the number of receptor dimer pairings occurring in nature is limited to those driven by natural ligands encoded within our genome. We have engineered synthethic cytokines (synthekines) that drive formation of cytokine receptor dimer pairings that are not formed by endogenous cytokines and that are not found in nature, and which activate distinct signaling programs. We show that a wide range of non-natural cytokine receptor hetero-dimers are competent to elicit a signaling output. We engineered synthekine ligands that assembled IL-2Rß/IL-4Rα or IL-4Rα/IFNAR2 receptor heterodimers, that do not occur naturally, triggering signaling and functional responses distinct from those activated by the endogenous cytokines IL-2, IL-4, and IFN. Furthermore, hybrid synthekine ligands that dimerized a JAK/STAT cytokine receptor with a receptor tyrosine kinase (RTK) also elicited a signaling response. Synthekines represent a new family of synthetic ligands with pre-defined receptors, but 'orphan' functions, that enable the full combinatorial scope of dimeric signaling receptors encoded within the human genome to be exploited for basic research and drug discovery.


Assuntos
Citocinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Receptores de Superfície Celular/metabolismo , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Linhagem Celular , Citocinas/genética , Humanos , Proteínas Recombinantes/genética
3.
Cell ; 168(6): 1041-1052.e18, 2017 03 09.
Artigo em Inglês | MEDLINE | ID: mdl-28283060

RESUMO

Most secreted growth factors and cytokines are functionally pleiotropic because their receptors are expressed on diverse cell types. While important for normal mammalian physiology, pleiotropy limits the efficacy of cytokines and growth factors as therapeutics. Stem cell factor (SCF) is a growth factor that acts through the c-Kit receptor tyrosine kinase to elicit hematopoietic progenitor expansion but can be toxic when administered in vivo because it concurrently activates mast cells. We engineered a mechanism-based SCF partial agonist that impaired c-Kit dimerization, truncating downstream signaling amplitude. This SCF variant elicited biased activation of hematopoietic progenitors over mast cells in vitro and in vivo. Mouse models of SCF-mediated anaphylaxis, radioprotection, and hematopoietic expansion revealed that this SCF partial agonist retained therapeutic efficacy while exhibiting virtually no anaphylactic off-target effects. The approach of biasing cell activation by tuning signaling thresholds and outputs has applications to many dimeric receptor-ligand systems.


Assuntos
Anafilaxia/metabolismo , Células-Tronco Hematopoéticas/imunologia , Mastócitos/metabolismo , Proteínas Proto-Oncogênicas c-kit/metabolismo , Transdução de Sinais , Fator de Células-Tronco/metabolismo , Anafilaxia/imunologia , Animais , Dimerização , Humanos , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Moleculares , Engenharia de Proteínas , Proteínas Proto-Oncogênicas c-kit/agonistas , Proteínas Proto-Oncogênicas c-kit/química , Fator de Células-Tronco/química , Fator de Células-Tronco/genética
4.
Methods Mol Biol ; 1314: 51-61, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26139254

RESUMO

Alternative, antibody-free techniques to western analysis of protein blots can offer reduced assay times for routine analysis of expression of recombinant proteins. We have adapted the commercially available enzyme fragment complementation technology to provide a rapid protein detection method for protein blots based on significantly reducing the number of incubation and washing steps used in traditional approaches, and eliminating the requirement for antibodies. In this chapter, we highlight the use of this assay for measuring recombinant protein expressed in mammalian cells for a range of applications, including dot blot screening of large numbers of different cell samples, assessment of protein integrity through detection of degradation bands, and characterization of posttranslational protein modifications such as glycosylation.


Assuntos
Medições Luminescentes/métodos , Proteínas Recombinantes/análise , Animais , Western Blotting/métodos , Técnicas de Cultura de Células/métodos , Linhagem Celular , Eletroforese em Gel de Poliacrilamida/métodos , Humanos , Medições Luminescentes/economia , Processamento de Proteína Pós-Traducional
5.
J Med Chem ; 58(10): 4220-9, 2015 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-25901762

RESUMO

Allosteric modulators of G protein-coupled receptors (GPCRs) have a number of potential advantages compared to agonists or antagonists that bind to the orthosteric site of the receptor. These include the potential for receptor selectivity, maintenance of the temporal and spatial fidelity of signaling in vivo, the ceiling effect of the allosteric cooperativity which may prevent overdose issues, and engendering bias by differentially modulating distinct signaling pathways. Here we describe the discovery, synthesis, and molecular pharmacology of δ-opioid receptor-selective positive allosteric modulators (δ PAMs). These δ PAMs increase the affinity and/or efficacy of the orthosteric agonists leu-enkephalin, SNC80 and TAN67, as measured by receptor binding, G protein activation, ß-arrestin recruitment, adenylyl cyclase inhibition, and extracellular signal-regulated kinases (ERK) activation. As such, these compounds are useful pharmacological tools to probe the molecular pharmacology of the δ receptor and to explore the therapeutic potential of δ PAMs in diseases such as chronic pain and depression.


Assuntos
Receptores Opioides delta/metabolismo , Relação Estrutura-Atividade , Animais , Arrestinas/metabolismo , Benzamidas/farmacologia , Ligação Competitiva , Células CHO , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Técnicas de Química Sintética , Cricetulus , Descoberta de Drogas , Avaliação Pré-Clínica de Medicamentos/métodos , Encefalina Leucina/farmacologia , Ativação Enzimática/efeitos dos fármacos , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Estrutura Molecular , Terapia de Alvo Molecular , Piperazinas/farmacologia , Ligação Proteica , Quinolinas/farmacologia , beta-Arrestinas
6.
Cell ; 160(6): 1196-208, 2015 Mar 12.
Artigo em Inglês | MEDLINE | ID: mdl-25728669

RESUMO

Most cell-surface receptors for cytokines and growth factors signal as dimers, but it is unclear whether remodeling receptor dimer topology is a viable strategy to "tune" signaling output. We utilized diabodies (DA) as surrogate ligands in a prototypical dimeric receptor-ligand system, the cytokine Erythropoietin (EPO) and its receptor (EpoR), to dimerize EpoR ectodomains in non-native architectures. Diabody-induced signaling amplitudes varied from full to minimal agonism, and structures of these DA/EpoR complexes differed in EpoR dimer orientation and proximity. Diabodies also elicited biased or differential activation of signaling pathways and gene expression profiles compared to EPO. Non-signaling diabodies inhibited proliferation of erythroid precursors from patients with a myeloproliferative neoplasm due to a constitutively active JAK2V617F mutation. Thus, intracellular oncogenic mutations causing ligand-independent receptor activation can be counteracted by extracellular ligands that re-orient receptors into inactive dimer topologies. This approach has broad applications for tuning signaling output for many dimeric receptor systems.


Assuntos
Receptores da Eritropoetina/química , Receptores da Eritropoetina/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/genética , Anticorpos Monoclonais/metabolismo , Linhagem Celular , Cristalografia por Raios X , Dimerização , Eritropoetina/metabolismo , Humanos , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Camundongos , Modelos Moleculares , Simulação de Dinâmica Molecular , Dados de Sequência Molecular , Mutação Puntual , Engenharia de Proteínas , Receptores da Eritropoetina/agonistas , Receptores da Eritropoetina/antagonistas & inibidores , Alinhamento de Sequência
7.
J Biomol Screen ; 19(9): 1255-65, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-25047277

RESUMO

Hetero-oligomeric complexes of G protein-coupled receptors (GPCRs) may represent novel therapeutic targets exhibiting different pharmacology and tissue- or cell-specific site of action compared with receptor monomers or homo-oligomers. An ideal tool for validating this concept pharmacologically would be a hetero-oligomer selective ligand. We set out to develop and execute a 1536-well high-throughput screen of over 1 million compounds to detect potential hetero-oligomer selective ligands using a ß-arrestin recruitment assay in U2OS cells coexpressing recombinant µ- and δ-opioid receptors. Hetero-oligomer selective ligands may bind to orthosteric or allosteric sites, and we might anticipate that the formation of hetero-oligomers may provide novel allosteric binding pockets for ligand binding. Therefore, our goal was to execute the screen in such a way as to identify positive allosteric modulators (PAMs) as well as agonists for µ, δ, and hetero-oligomeric receptors. While no hetero-oligomer selective ligands were identified (based on our selection criteria), this single screen did identify numerous µ- and δ-selective agonists and PAMs as well as nonselective agonists and PAMs. To our knowledge, these are the first µ- and δ-opioid receptor PAMs described in the literature.


Assuntos
Regulação Alostérica/efeitos dos fármacos , Analgésicos Opioides/farmacologia , Ensaios de Triagem em Larga Escala , Receptores Opioides delta/agonistas , Receptores Opioides delta/química , Receptores Opioides mu/agonistas , Receptores Opioides mu/química , Animais , Arrestinas/metabolismo , Células CHO , Colforsina/farmacologia , Cricetinae , Cricetulus , AMP Cíclico/metabolismo , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos , Humanos , Antagonistas de Entorpecentes/farmacologia , Multimerização Proteica , beta-Arrestinas
8.
J Biol Chem ; 288(49): 35039-48, 2013 Dec 06.
Artigo em Inglês | MEDLINE | ID: mdl-24145037

RESUMO

Chemokines display considerable promiscuity with multiple ligands and receptors shared in common, a phenomenon that is thought to underlie their biochemical "redundancy." Their receptors are part of a larger seven-transmembrane receptor superfamily, commonly referred to as G protein-coupled receptors, which have been demonstrated to be able to signal with different efficacies to their multiple downstream signaling pathways, a phenomenon referred to as biased agonism. Biased agonism has been primarily reported as a phenomenon of synthetic ligands, and the biologic prevalence and importance of such signaling are unclear. Here, to assess the presence of biased agonism that may underlie differential signaling by chemokines targeting the same receptor, we performed a detailed pharmacologic analysis of a set of chemokine receptors with multiple endogenous ligands using assays for G protein signaling, ß-arrestin recruitment, and receptor internalization. We found that chemokines targeting the same receptor can display marked differences in their efficacies for G protein- or ß-arrestin-mediated signaling or receptor internalization. This ligand bias correlates with changes in leukocyte migration, consistent with different mechanisms underlying the signaling downstream of these receptors induced by their ligands. These findings demonstrate that biased agonism is a common and likely evolutionarily conserved biological mechanism for generating qualitatively distinct patterns of signaling via the same receptor in response to different endogenous ligands.


Assuntos
Receptores de Quimiocinas/agonistas , Receptores de Quimiocinas/metabolismo , Arrestinas/metabolismo , Quimiocinas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Células HEK293 , Humanos , Cinética , Ligantes , Modelos Biológicos , Transdução de Sinais , beta-Arrestinas
9.
Proc Natl Acad Sci U S A ; 110(26): 10830-5, 2013 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-23754417

RESUMO

µ-Opioid receptors are among the most studied G protein-coupled receptors because of the therapeutic value of agonists, such as morphine, that are used to treat chronic pain. However, these drugs have significant side effects, such as respiratory suppression, constipation, allodynia, tolerance, and dependence, as well as abuse potential. Efforts to fine tune pain control while alleviating the side effects of drugs, both physiological and psychological, have led to the development of a wide variety of structurally diverse agonist ligands for the µ-opioid receptor, as well as compounds that target κ- and δ-opioid receptors. In recent years, the identification of allosteric ligands for some G protein-coupled receptors has provided breakthroughs in obtaining receptor subtype-selectivity that can reduce the overall side effect profiles of a potential drug. However, positive allosteric modulators (PAMs) can also have the specific advantage of only modulating the activity of the receptor when the orthosteric agonist occupies the receptor, thus maintaining spatial and temporal control of receptor signaling in vivo. This second advantage of allosteric modulators may yield breakthroughs in opioid receptor research and could lead to drugs with improved side-effect profiles or fewer tolerance and dependence issues compared with orthosteric opioid receptor agonists. Here, we describe the discovery and characterization of µ-opioid receptor PAMs and silent allosteric modulators, identified from high-throughput screening using a ß-arrestin-recruitment assay.


Assuntos
Receptores Opioides mu/agonistas , Receptores Opioides mu/metabolismo , Sulfonas/farmacologia , Tiazóis/farmacologia , Regulação Alostérica , Sítio Alostérico , Animais , Arrestinas/metabolismo , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Descoberta de Drogas , Ensaios de Triagem em Larga Escala , Humanos , Ligantes , Ratos , Sulfonas/química , Tiazóis/química , beta-Arrestinas
10.
Assay Drug Dev Technol ; 11(4): 237-43, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23557019

RESUMO

Receptor tyrosine kinases (RTKs) are important players in various cellular processes, including proliferation, migration, metabolism, and neuronal development. EphB4 RTK is essential for the development of a functional arterial-venous network in embryonic and adult neoangiogenesis. To develop novel inhibitors of EphB4 that might have applications in severe diseases like cancer and retinopathies, assays need to be in place that resemble, in a most physiological fashion, the activation and downstream function of the kinase. In addition, such assays need to be amenable to high-throughput screening to serve efficiently the modern drug discovery processes in the pharmaceutical industry. The authors have developed an enzyme fragment complementation assay that measures the interaction of a downstream docking protein to the activated and phosphorylated full-length EphB4 kinase in cells. The assay is specific, robust, and amenable to miniaturization and high-throughput screening. It covers most steps in the activation process of EphB4, including ligand binding, autophosphorylation, and docking of a downstream interactor. This assay format can be transferred to other RTKs and adds an important cell-based kinase assay option to researchers in the field.


Assuntos
Descoberta de Drogas/métodos , Ensaios Enzimáticos/métodos , Ensaios de Triagem em Larga Escala/métodos , Mapeamento de Interação de Proteínas/métodos , Receptor EphB4/análise , Receptor EphB4/química , Ativação Enzimática , Especificidade por Substrato
11.
J Biomol Screen ; 18(5): 599-609, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23396314

RESUMO

A variety of G-protein-coupled receptor (GPCR) screening technologies have successfully partnered a number of GPCRs with their cognate ligands. GPCR-mediated ß-arrestin recruitment is now recognized as a distinct intracellular signaling pathway, and ligand-receptor interactions may show a bias toward ß-arrestin over classical GPCR signaling pathways. We hypothesized that the failure to identify native ligands for the remaining orphan GPCRs may be a consequence of biased ß-arrestin signaling. To investigate this, we assembled 10 500 candidate ligands and screened 82 GPCRs using PathHunter ß-arrestin recruitment technology. High-quality screening assays were validated by the inclusion of liganded receptors and the detection and confirmation of these established ligand-receptor pairings. We describe a candidate endogenous orphan GPCR ligand and a number of novel surrogate ligands. However, for the majority of orphan receptors studied, measurement of ß-arrestin recruitment did not lead to the identification of cognate ligands from our screening sets. ß-Arrestin recruitment represents a robust GPCR screening technology, and ligand-biased signaling is emerging as a therapeutically exploitable feature of GPCR biology. The identification of cognate ligands for the orphan GPCRs and the extent to which receptors may exist to preferentially signal through ß-arrestin in response to their native ligand remain to be determined.


Assuntos
Arrestinas/metabolismo , Ensaios de Triagem em Larga Escala/métodos , Receptores Acoplados a Proteínas G/agonistas , Animais , Células CHO , Células Cultivadas , Cricetinae , Cricetulus , Descoberta de Drogas/métodos , Células HEK293 , Humanos , Ligantes , Ligação Proteica/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Saccharomyces cerevisiae , Bibliotecas de Moléculas Pequenas/análise , beta-Arrestinas
12.
Methods Mol Biol ; 897: 171-80, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22674165

RESUMO

More than two-thirds of all known G-protein coupled receptors are known to modulate the function of adenylate cyclase resulting in altered levels of cAMP. In turn, cAMP fluctuations transform agonist binding events into physiological changes in cell behavior. The advent of nonradioactive, homogeneous methods of measuring intracellular cAMP has enabled the rapid growth of drug discovery and research applications for these GPCR targets. In this chapter, we describe a nonradioactive, chemiluminescent cAMP detection method using enzyme fragment complementation technology to detect a wide range of GPCR modulators which is also suitable for high-throughput screening.


Assuntos
Bioensaio/métodos , AMP Cíclico/química , AMP Cíclico/metabolismo , Medições Luminescentes/métodos , Receptores Acoplados a Proteínas G/metabolismo , Regulação Alostérica , Linhagem Celular , Receptores Acoplados a Proteínas G/agonistas
13.
Methods Mol Biol ; 897: 181-203, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22674166

RESUMO

The recruitment of arrestins to activated 7TMRs results in the activation of alternative signaling pathways, quenching of G-protein activation, and coupling to clathrin-mediated endocytosis. The nearly ubiquitous involvement of arrestin in 7TMR signaling has spurred the development of several methods for monitoring this interaction in mammalian cells. Nonetheless, few maintain the reproducibility and precision necessary for drug discovery applications. Enzyme fragment complementation technology (EFC) is an emerging protein-protein interaction technology based on the forced complementation of a split enzyme that has proven to be highly effective in monitoring the formation of GPCR-arrestin complexes. In these systems, the target proteins are fused to two fragments of an enzyme that show little or no spontaneous complementation. Interaction of the two proteins forces the complementation of the enzyme, resulting in an enzymatic measure of the protein interaction. This chapter discusses the utility and methods involved in using the PathHunter ß-galactosidase complementation system to monitor arrestin recruitment and the advantages of exploiting this pathway in the characterization of 7TMR function.


Assuntos
Arrestinas/metabolismo , Descoberta de Drogas/métodos , Receptores Acoplados a Proteínas G/metabolismo , Animais , Linhagem Celular , Humanos , Ligação Proteica , beta-Arrestinas , beta-Galactosidase/metabolismo
14.
Life Sci ; 91(13-14): 544-9, 2012 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-22480514

RESUMO

AIMS: To determine the pharmacology of ET(A)- and ET(B)-mediated ß-arrestin recruitment and compare this to established human pharmacology of these receptors to identify evidence for endothelin receptor biased signalling and pathway specific blockade by antagonists. MAIN METHODS: The ability of ET-1, ET-2, ET-3, sarafotoxin 6b and sarafotoxin 6c to activate ET(A) and ET(B)-mediated ß-arrestin recruitment was determined in CHO-K1 cells. Affinities were obtained for ET(A) selective (BQ123, sitaxentan, ambrisentan), ET(B) selective (BQ788) and mixed (bosentan) antagonists using ET-1 and compared to affinities obtained in competition experiments in human heart and by Schild analysis in human saphenous vein. Agonist dependence of affinities was compared for BQ123 and BQ788 in the ET(A) and ET(B) ß-arrestin assays respectively. KEY FINDINGS: For ß-arrestin recruitment, order of potency was as expected for the ET(A) (ET-1≥ET-2>>ET-3) and ET(B) (ET-1=ET-2=ET-3) receptors. However, at the ET(A) receptor sarafotoxin 6b and ET-3 were partial agonists. Antagonism of ET peptides by selective and mixed antagonists appeared non-competitive. BQ123, but not BQ788, exhibited agonist-dependent affinities. Bosentan was significantly more effective an inhibitor of ß-arrestin recruitment mediated by ET(A) compared to the ET(B) receptor. In the ET(A) vasoconstrictor assay, ET-1, ET-2 and S6b were equipotent, full agonists and antagonists tested behaved in a competitive manner, although affinities were lower than predicted from the competition binding experiments in left ventricle. SIGNIFICANCE: These data suggest that the pharmacology of ET(A) and ET(B) receptors linked to G-protein- and ß-arrestin mediated responses was different and bosentan appeared to show bias, preferentially blocking ET(A) mediated ß-arrestin recruitment.


Assuntos
Arrestinas/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Receptor de Endotelina A/metabolismo , Receptor de Endotelina B/metabolismo , Transdução de Sinais , Animais , Bosentana , Células CHO , Cricetinae , Cricetulus , Antagonistas do Receptor de Endotelina A , Antagonistas do Receptor de Endotelina B , Endotelina-1/farmacologia , Endotelina-2/farmacologia , Endotelina-3/farmacologia , Humanos , Receptor de Endotelina A/agonistas , Receptor de Endotelina B/agonistas , Sulfonamidas/metabolismo , Vasoconstritores/farmacologia , Venenos de Víboras/farmacologia , beta-Arrestinas
15.
Methods Mol Biol ; 574: 249-59, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19685314

RESUMO

Bioluminescence using the reporter enzyme firefly luciferase (Fluc) and the substrate luciferin enables non-invasive optical imaging of living animals with extremely high sensitivity. This type of analysis enables studies of gene expression, tumor growth, and cell migration over time in live animals that were previously not possible. However, a major limitation of this system is that Fluc activity is restricted to the intracellular environment, which precludes important applications of in vivo imaging such as antibody labeling, or serum protein monitoring. In order to expand the application of bioluminescence imaging to other enzymes, we characterized a sequential reporter-enzyme luminescence (SRL) technology for the in vivo detection of beta-galactosidase (beta-gal) activity. The substrate is a "caged" D-luciferin conjugate that must first be cleaved by beta-gal before it can be catalyzed by Fluc in the final, light-emitting step. Hence, luminescence is dependent on and correlates with beta-gal activity. A variety of experiments were performed in order to validate the system and explore potential new applications. We were able to visualize non-invasively over time constitutive beta-gal activity in engineered cells, as well as inducible tissue-specific beta-gal expression in transgenic mice. Since beta-gal, unlike Fluc, retains full activity outside of cells, we were able to show that antibodies conjugated to the recombinant beta-gal enzyme could be used to detect and localize endogenous cells and extracellular antigens in vivo. In addition, we developed a low-affinity beta-gal complementation system that enables inducible, reversible protein interactions to be monitored in real time in vivo, for example, sequential responses to agonists and antagonists of G-protein-coupled receptors (GPCRs). Thus, using SRL, the exquisite luminescent properties of Fluc can be combined with the advantages of another enzyme. Other substrates have been described that extend the scope to endogenous enzymes, such as cytochromes or caspases, potentially enabling additional unprecedented applications.


Assuntos
Genes Reporter , Luminescência , beta-Galactosidase/metabolismo , Animais , Camundongos , Camundongos Nus , beta-Galactosidase/genética
16.
Methods Mol Biol ; 536: 395-405, 2009.
Artigo em Inglês | MEDLINE | ID: mdl-19378077

RESUMO

Alternative, antibody-free techniques to western analysis of protein blots can offer reduced assay times for routine analysis of expression of recombinant proteins. We have adapted the commercially available enzyme fragment complementation technology to provide a rapid protein detection method for protein blots based on significantly reducing the number of incubation and washing steps used in traditional approaches, and eliminating the requirement for antibodies. In this article, we highlight the use of this assay for measuring recombinant protein expressed in mammalian cells for a range of applications, including dot blot screening of large numbers of different cell samples, assessment of protein integrity through detection of degradation bands, and characterization of post-translational protein modifications such as glycosylation.


Assuntos
Anticorpos/química , Western Blotting/métodos , Proteínas Recombinantes/análise , Animais , Células CHO , Linhagem Celular , Cricetinae , Cricetulus , Eletroforese em Gel de Poliacrilamida/métodos , Processamento de Proteína Pós-Traducional
17.
J Biomol Screen ; 13(8): 737-47, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18660457

RESUMO

G-protein-coupled receptors (GPCRs) represent one of the largest gene families in the human genome and have long been regarded as valuable targets for small-molecule drugs. The authors describe a new functional assay that directly monitors GPCR activation. It is based on the interaction between beta-arrestin and ligand-activated GPCRs and uses enzyme fragment complementation technology. In this format, a GPCR of interest is fused to a small (approximately 4 kDa), optimized alpha fragment peptide (termed ProLink) derived from beta-galactosidase, and beta-arrestin is fused to an N-terminal deletion mutant of beta-galactosidase (termed the enzyme acceptor [EA]). Upon activation of the receptor, the beta-arrestin-EA fusion protein binds the activated GPCR. This interaction drives enzyme fragment complementation, resulting in an active beta-galactosidase enzyme, and thus GPCR activation can be determined by quantifying beta-galactosidase activity. In this report, the authors demonstrate the utility of this technology to monitor GPCR activation and validate the approach using a Galphai-coupled GPCR, somatostatin receptor 2. Potential application to high-throughput screens in both agonist and antagonist screening modes is exemplified.


Assuntos
Arrestinas/metabolismo , Bioensaio/métodos , Peptídeos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Somatostatina/metabolismo , Animais , Arrestinas/genética , Linhagem Celular , Humanos , Peptídeos/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Somatostatina/agonistas , Receptores de Somatostatina/antagonistas & inibidores , Receptores de Somatostatina/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Reprodutibilidade dos Testes , Somatostatina/genética , Somatostatina/metabolismo , beta-Arrestinas , beta-Galactosidase/genética , beta-Galactosidase/metabolismo
18.
FASEB J ; 21(14): 3819-26, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17942828

RESUMO

G-protein coupled receptors (GPCRs) are a versatile and ubiquitous family of membrane receptors that transmit extracellular signals to mammalian cells and constitute the most important class of drug targets. Yet, sensitive and specific methods are lacking that would allow quantitative comparisons of pharmacologic properties of these receptors in physiological or pathological settings in live animals. We sought to overcome these limitations by employing low affinity, reversible beta-galactosidase complementation to quantify GPCR activation via interaction with beta-arrestin. A panel of cell lines was engineered expressing different GPCRs together with the reporter system. In vitro evaluation revealed highly sensitive, dynamic, and specific assessment of GPCR agonists and antagonists. Following implantation of the cells into mice, it was possible for the first time to monitor pharmacological GPCR activation and inhibition in their physiological context by noninvasive bioluminescence imaging in living animals. This technology has unique advantages that enable novel applications in the functional investigation of GPCR modulation in live animals in biological research and drug discovery.


Assuntos
Receptores Acoplados a Proteínas G/metabolismo , Animais , Células CHO , Células COS , Chlorocebus aethiops , Cricetinae , Cricetulus , Luciferina de Vaga-Lumes , Humanos , Substâncias Luminescentes , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Receptores Acoplados a Proteínas G/genética
19.
FASEB J ; 21(14): 3827-34, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17942829

RESUMO

G-protein-coupled receptor (GPCR) signaling is involved in a wide range of physiological processes and diseases, and around one-half of currently used drugs target GPCRs. Assays for the signaling of GPCRs have suffered from drawbacks, including low signal-to-noise, temporally transient signals, and difficulty in applying a single assay to a wide range of GPCRs. We have developed a set of assays for G-protein-coupled receptor signaling based on beta-galactosidase enzyme complementation in live mammalian cells. We previously described an assay for GPCR activation by monitoring the binding of beta-arrestin to the receptor. Here we describe a second assay that monitors the internalization of GPCRs to endosomes, an event that follows receptor activation and is critical in desensitizing and resensitizing the receptor. We show that both assays display high signal-to-noise ratios with low variability and are quantitative for a wide range of GPCRs. EC50s obtained with these assays closely match results reported in the literature. Finally, we show that these assays are readily adapted to high-throughput chemical screens. Thus, these two assays for monitoring G-protein-coupled receptor activation and internalization should prove valuable in basic biological studies as well as in high-throughput screens.


Assuntos
Teste de Complementação Genética , Receptores Acoplados a Proteínas G/metabolismo , beta-Galactosidase/genética , Animais , Transporte Biológico Ativo/genética , Linhagem Celular , Membrana Celular/enzimologia , Membrana Celular/genética , Escherichia coli/enzimologia , Escherichia coli/genética , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/análise , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Transdução de Sinais/fisiologia , Temperatura
20.
Neuron ; 53(1): 25-38, 2007 Jan 04.
Artigo em Inglês | MEDLINE | ID: mdl-17196528

RESUMO

Nerve growth factor engages two structurally distinct transmembrane receptors, TrkA and p75, which have been proposed to create a "high-affinity" NGF binding site through formation of a ternary TrkA/NGF/p75 complex. To define a structural basis for the high-affinity site, we have determined the three-dimensional structure of a complete extracellular domain of TrkA complexed with NGF. The complex reveals a crab-shaped homodimeric TrkA structure, but a mechanism for p75 coordination is not obvious. We investigated the heterodimerization of membrane-bound TrkA and p75, on intact mammalian cells, using a beta-gal protein-protein interaction system. We find that NGF dimerizes TrkA and that p75 exists on the cell surface as a preformed oligomer that is not dissociated by NGF. We find no evidence for a direct TrkA/p75 interaction. We propose that TrkA and p75 likely communicate through convergence of downstream signaling pathways and/or shared adaptor molecules, rather than through direct extracellular interactions.


Assuntos
Membrana Celular/metabolismo , Fator de Crescimento Neural/química , Neurônios/metabolismo , Receptor de Fator de Crescimento Neural/química , Receptor trkA/química , Animais , Sítios de Ligação/fisiologia , Linhagem Celular , Membrana Celular/ultraestrutura , Dimerização , Espaço Extracelular/metabolismo , Humanos , Ligantes , Substâncias Macromoleculares/química , Substâncias Macromoleculares/metabolismo , Camundongos , Modelos Moleculares , Fator de Crescimento Neural/metabolismo , Neurônios/ultraestrutura , Estrutura Terciária de Proteína/fisiologia , Receptor de Fator de Crescimento Neural/metabolismo , Receptor trkA/metabolismo , Transdução de Sinais/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...