Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
ACS Omega ; 5(27): 16940-16943, 2020 Jul 14.
Artigo em Inglês | MEDLINE | ID: mdl-32685863

RESUMO

The putative active metabolite of aeruginascin, a naturally occurring tryptamine of "magic mushrooms," has been synthesized and structurally characterized. Competitive radioligand binding assays demonstrate that it has a high affinity at human serotonin receptors 5-HT1A, 5-HT2A, and 5-HT2B, though it does not bind at the 5-HT3 receptor, where activity was previously predicted.

2.
J Mol Cell Cardiol ; 97: 106-13, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27130278

RESUMO

Chronic cardiac hypertrophy is maladaptive and contributes to the pathogenesis of heart failure. The objective of this study was to identify small molecule inhibitors of pathological cardiomyocyte hypertrophy. High content screening was performed with primary neonatal rat ventricular myocytes (NRVMs) cultured on 96-well plates and treated with a library of 3241 distinct small molecules. Non-toxic hit compounds that blocked hypertrophy in response to phenylephrine (PE) and phorbol myristate acetate (PMA) were identified based on their ability to reduce cell size and inhibit expression of atrial natriuretic factor (ANF), which is a biomarker of pathological cardiac hypertrophy. Many of the hit compounds are existing drugs that have not previously been evaluated for benefit in the setting of cardiovascular disease. One such compound, the anti-malarial drug artesunate, blocked left ventricular hypertrophy (LVH) and improved cardiac function in adult mice subjected to transverse aortic constriction (TAC). These findings demonstrate that phenotypic screening with primary cardiomyocytes can be used to discover anti-hypertrophic lead compounds for heart failure drug discovery. Using annotated libraries of compounds with known selectivity profiles, this screening methodology also facilitates chemical biological dissection of signaling networks that control pathological growth of the heart.


Assuntos
Cardiomegalia/metabolismo , Descoberta de Drogas , Ensaios de Triagem em Larga Escala , Animais , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/tratamento farmacológico , Células Cultivadas , Modelos Animais de Doenças , Hemodinâmica/efeitos dos fármacos , Masculino , Camundongos , Imagem Molecular/métodos , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Ratos , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas , Remodelação Ventricular/efeitos dos fármacos
3.
Curr Chem Genom Transl Med ; 8(Suppl 1): 27-35, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24596682

RESUMO

The multi cellular tumor spheroid (MCTS) model has been used for decades with proven superiority over monolayer cell culture models at recapitulating in vivo tumor growth. Yet its use in high-throughput drug discovery has been limited, particularly with image based screening, due to practical and technical hurdles. Here we report a significant advance in utilizing live MCTS models for high-content image based drug discovery. Using a validated GFP reporter (CK5Pro-GFP) of luminal breast cancer stem cells (CSC), we developed an algorithm to quantify changes in CK5Pro-GFP expression levels for individual Z-stack planes (local) or as maximal projections of the summed Z-stacks (global) of MCTS. From these image sets, we can quantify the cross-sectional area of GFP positive cells, the fluorescence intensity of the GFP positive cells, and the percent of spheroid cross-sectional area that expresses CK5Pro-GFP.We demonstrate that acquiring data in this manner can be done in real time and is statistically robust (Z'=0.85) for use in primary high-content screening cancer drug discovery.

4.
Antimicrob Agents Chemother ; 58(1): 386-96, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24165192

RESUMO

One of the most challenging goals of hepatitis C virus (HCV) research is to develop well-tolerated regimens with high cure rates across a variety of patient populations. Such a regimen will likely require a combination of at least two distinct direct-acting antivirals (DAAs). Combining two or more DAAs with different resistance profiles increases the number of mutations required for viral breakthrough. Currently, most DAAs inhibit HCV replication. We recently reported that the combination of two distinct classes of HCV inhibitors, entry inhibitors and replication inhibitors, prolonged reductions in extracellular HCV in persistently infected cells. We therefore sought to identify new inhibitors targeting aspects of the HCV replication cycle other than RNA replication. We report here the discovery of the first small-molecule HCV infectivity inhibitor, GS-563253, also called HCV infectivity inhibitor 1 (HCV II-1). HCV II-1 is a substituted tetrahydroquinoline that selectively inhibits genotype 1 and 2 HCVs with low-nanomolar 50% effective concentrations. It was identified through a high-throughput screen and subsequent chemical optimization. HCV II-1 only permits the production and release of noninfectious HCV particles from cells. Moreover, infectious HCV is rapidly inactivated in its presence. HCV II-1 resistance mutations map to HCV E2. In addition, HCV-II prevents HCV endosomal fusion, suggesting that it either locks the viral envelope in its prefusion state or promotes a viral envelope conformation change incapable of fusion. Importantly, the discovery of HCV II-1 opens up a new class of HCV inhibitors that prolong viral suppression by HCV replication inhibitors in persistently infected cell cultures.


Assuntos
Antivirais/farmacologia , Hepacivirus/efeitos dos fármacos , Antivirais/química , Linhagem Celular , Farmacorresistência Viral , Hepacivirus/metabolismo , Hepatite C/tratamento farmacológico , Humanos , Proteínas não Estruturais Virais/genética , Proteínas não Estruturais Virais/metabolismo , Replicação Viral/efeitos dos fármacos
5.
J Mol Cell Cardiol ; 63: 175-9, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23939492

RESUMO

Cardiac hypertrophy is an independent predictor of adverse outcomes in patients with heart failure, and thus represents an attractive target for novel therapeutic intervention. JQ1, a small molecule inhibitor of bromodomain and extraterminal (BET) acetyl-lysine reader proteins, was identified in a high throughput screen designed to discover novel small molecule regulators of cardiomyocyte hypertrophy. JQ1 dose-dependently blocked agonist-dependent hypertrophy of cultured neonatal rat ventricular myocytes (NRVMs) and reversed the prototypical gene program associated with pathological cardiac hypertrophy. JQ1 also blocked left ventricular hypertrophy (LVH) and improved cardiac function in adult mice subjected to transverse aortic constriction (TAC). The BET family consists of BRD2, BRD3, BRD4 and BRDT. BRD4 protein expression was increased during cardiac hypertrophy, and hypertrophic stimuli promoted recruitment of BRD4 to the transcriptional start site (TSS) of the gene encoding atrial natriuretic factor (ANF). Binding of BRD4 to the ANF TSS was associated with increased phosphorylation of local RNA polymerase II. These findings define a novel function for BET proteins as signal-responsive regulators of cardiac hypertrophy, and suggest that small molecule inhibitors of these epigenetic reader proteins have potential as therapeutics for heart failure.


Assuntos
Cardiomegalia/metabolismo , Proteínas de Transporte/metabolismo , Animais , Azepinas/farmacologia , Cardiomegalia/tratamento farmacológico , Cardiomegalia/patologia , Proteínas de Transporte/química , Descoberta de Drogas , Ensaios de Triagem em Larga Escala , Modelos Biológicos , Ligação Proteica/efeitos dos fármacos , Ratos , Triazóis/farmacologia
6.
Proc Natl Acad Sci U S A ; 110(24): 9806-11, 2013 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-23720316

RESUMO

Cardiac hypertrophy is a strong predictor of morbidity and mortality in patients with heart failure. Small molecule histone deacetylase (HDAC) inhibitors have been shown to suppress cardiac hypertrophy through mechanisms that remain poorly understood. We report that class I HDACs function as signal-dependent repressors of cardiac hypertrophy via inhibition of the gene encoding dual-specificity phosphatase 5 (DUSP5) DUSP5, a nuclear phosphatase that negatively regulates prohypertrophic signaling by ERK1/2. Inhibition of DUSP5 by class I HDACs requires activity of the ERK kinase, mitogen-activated protein kinase kinase (MEK), revealing a self-reinforcing mechanism for promotion of cardiac ERK signaling. In cardiac myocytes treated with highly selective class I HDAC inhibitors, nuclear ERK1/2 signaling is suppressed in a manner that is absolutely dependent on DUSP5. In contrast, cytosolic ERK1/2 activation is maintained under these same conditions. Ectopic expression of DUSP5 in cardiomyocytes results in potent inhibition of agonist-dependent hypertrophy through a mechanism involving suppression of the gene program for hypertrophic growth. These findings define unique roles for class I HDACs and DUSP5 as integral components of a regulatory signaling circuit that controls cardiac hypertrophy.


Assuntos
Cardiomegalia/metabolismo , Fosfatases de Especificidade Dupla/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Histona Desacetilases/metabolismo , Animais , Animais Recém-Nascidos , Benzamidas/farmacologia , Cardiomegalia/genética , Núcleo Celular/enzimologia , Células Cultivadas , Fosfatases de Especificidade Dupla/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Inibidores de Histona Desacetilases/farmacologia , Immunoblotting , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Pirimidinas/farmacologia , Interferência de RNA , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
7.
J Biomol Screen ; 17(9): 1211-20, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22751729

RESUMO

Breast cancers expressing hormone receptors for estrogen (ER) and progesterone (PR) represent ~70% of all cases and are treated with both ER-targeted and chemotherapies, with near 40% becoming resistant. We have previously described that in some ER(+) tumors, the resistant cells express cytokeratin 5 (CK5), a putative marker of breast stem and progenitor cells. CK5(+) cells have lost expression of ER and PR, express the tumor-initiating cell surface marker CD44, and are relatively quiescent. In addition, progestins, which increase breast cancer incidence, expand the CK5(+) subpopulation in ER(+)PR(+) breast cancer cell lines. We have developed models to induce and quantitate CK5(+)ER(-)PR(-) cells, using CK5 promoter-driven luciferase (Fluc) or green fluorescent protein (GFP) reporters stably transduced into T47D breast cancer cells (CK5Pro-GFP or CK5Pro-Luc). We validated the CK5Pro-GFP-T47D model for high-content screening in 96-well microplates and performed a pilot screen using a focused library of 280 compounds from the National Institutes of Health clinical collection. Four hits were obtained that significantly abrogated the progestin-induced CK5(+) cell population, three of which were members of the retinoid family. Hence, this approach will be useful in discovering small molecules that could potentially be developed as combination therapies, preventing the acquisition of a drug-resistant subpopulation.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Descoberta de Drogas/métodos , Resistencia a Medicamentos Antineoplásicos , Células-Tronco Neoplásicas/efeitos dos fármacos , Retinoides/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Acitretina/farmacologia , Antineoplásicos Hormonais/farmacologia , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Genes Reporter , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Humanos , Receptores de Hialuronatos/metabolismo , Isotretinoína/farmacologia , Queratina-5/metabolismo , Luciferases/genética , Luciferases/metabolismo , Miconazol/farmacologia , Células-Tronco Neoplásicas/metabolismo , Progesterona/farmacologia , Progestinas/farmacologia , Regiões Promotoras Genéticas , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Tretinoína/farmacologia
8.
Expert Opin Drug Discov ; 7(9): 819-30, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22788761

RESUMO

INTRODUCTION: For the past 30 years 2D-cell-based assay models have dominated preclinical cancer drug discovery efforts. 2D-cell-based models fail to predict in vivo efficacy, contributing to a lower success rate and higher cost required to translate an investigational new drug to clinical approval. Technological advances in 3D-cell culture models bridge the gap between 2D and in vivo models to improve upon the current success rates of cancer drug discovery. AREAS COVERED: This review focuses on the multicellular tumor spheroid (MCTS), particularly how this model can be utilized for HTS drug discovery. We discuss the current technologies for uniform culture of MCTS suitable for HTS and detection methods utilized for assay development and drug screening. EXPERT OPINION: Substantial hurdles remain before we reach the ultimate goal of robust HTS of large compound libraries with MCTS models. Specifically, we can group these challenges into three categories: MCTS growth, data collection, and data analysis. The MCTS model should be utilized with fluorescent readouts and high-content imaging with a systems biology approach to model human tumors in vitro. Such models will be more predictive of in vivo efficacy, improving on the current success rates of cancer drug discovery from bench to bedside.


Assuntos
Antineoplásicos/farmacologia , Descoberta de Drogas/métodos , Neoplasias/tratamento farmacológico , Esferoides Celulares/efeitos dos fármacos , Animais , Desenho de Fármacos , Descoberta de Drogas/tendências , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Modelos Biológicos , Células Tumorais Cultivadas
9.
Mar Drugs ; 9(11): 2397-2408, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22163192

RESUMO

Type IIα DNA topoisomerase (TopoIIα) is among the most important clinical drug targets for the treatment of cancer. Recently, the DNA repair protein Metnase was shown to enhance TopoIIα activity and increase resistance to TopoIIα poisons. Using in vitro DNA decatenation assays we show that neoamphimedine potently inhibits TopoIIα-dependent DNA decatenation in the presence of Metnase. Cell proliferation assays demonstrate that neoamphimedine can inhibit Metnase-enhanced cell growth with an IC(50) of 0.5 µM. Additionally, we find that the apparent K(m) of TopoIIα for ATP increases linearly with higher concentrations of neoamphimedine, indicating ATP-competitive inhibition, which is substantiated by molecular modeling. These findings support the continued development of neoamphimedine as an anticancer agent, particularly in solid tumors that over-express Metnase.


Assuntos
Acridinas/farmacologia , Trifosfato de Adenosina/metabolismo , Antígenos de Neoplasias/efeitos dos fármacos , DNA Topoisomerases Tipo II/efeitos dos fármacos , Proteínas de Ligação a DNA/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/metabolismo , Acridinas/administração & dosagem , Antígenos de Neoplasias/metabolismo , Proliferação de Células/efeitos dos fármacos , DNA Topoisomerases Tipo II/metabolismo , Proteínas de Ligação a DNA/metabolismo , Relação Dose-Resposta a Droga , Células HEK293 , Humanos , Técnicas In Vitro , Concentração Inibidora 50 , Modelos Moleculares
10.
J Biomol Screen ; 16(7): 724-33, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21521800

RESUMO

Transforming growth factor ß (TGF-ß) type I receptor (activin receptor-like kinase 5, ALK5) has been identified as a promising target for fibrotic diseases. To find a novel inhibitor of ALK5, the authors performed a high-throughput screen of a library of 420,000 compounds using dephosphorylated ALK5. From primary hits of 1521 compounds, 555 compounds were confirmed. In total, 124 compounds were then selected for follow-up based on their unique structures and other properties. Repeated concentration-response testing and final interference assays of the above compounds resulted in the discovery of a structurally novel ALK5 inhibitor (compound 8) (N-(thiophen 2-ylmethyl)-3-(3,4,5 trimethoxyphenyl)imidazo[1,2ß]pyridazin 6-amine) with a low IC(50) value of 0.7 µM. Compound 8 also inhibited the TGF-ß-induced nuclear translocation of SMAD with an EC(50) value of 0.8 µM. Kinetic analysis revealed that compound 8 inhibited ALK5 via mixed-type inhibition, suggesting that it may bind to ALK5 differently than other published adenosine triphosphate site inhibitors.


Assuntos
Ensaios de Triagem em Larga Escala , Inibidores de Proteínas Quinases/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Receptores de Fatores de Crescimento Transformadores beta/antagonistas & inibidores , Difosfato de Adenosina/metabolismo , Linhagem Celular Tumoral , Simulação por Computador , Transferência Ressonante de Energia de Fluorescência , Fluorimunoensaio , Humanos , Cinética , Conformação Molecular , Fosforilação/efeitos dos fármacos , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Receptor do Fator de Crescimento Transformador beta Tipo I , Receptores de Fatores de Crescimento Transformadores beta/metabolismo , Proteínas Smad/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Fator de Crescimento Transformador beta/farmacologia
11.
Biochim Biophys Acta ; 1789(5): 422-31, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19414071

RESUMO

Histone deacetylase 5 (HDAC5) represses expression of nuclear genes that promote cardiac hypertrophy. Agonism of a variety of G protein coupled receptors (GPCRs) triggers phosphorylation-dependent nuclear export of HDAC5 via the CRM1 nuclear export receptor, resulting in derepression of pro-hypertrophic genes. A cell-based high-throughput screen of a commercial compound collection was employed to identify compounds with the ability to preserve the nuclear fraction of GFP-HDAC5 in primary cardiomyocytes exposed to GPCR agonists. A hit compound potently inhibited agonist-induced GFP-HDAC5 nuclear export in cultured neonatal rat ventricular myocytes (NRVMs). A small set of related compounds was designed and synthesized to evaluate structure-activity relationship (SAR). The results demonstrated that inhibition of HDAC5 nuclear export was a result of compounds irreversibly reacting with a key cysteine residue in CRM1 that is required for its function. CRM1 inhibition by the compounds also resulted in potent suppression of cardiomyocyte hypertrophy. These studies define a novel class of anti-hypertrophic compounds that function through irreversible inhibition of CRM1-dependent nuclear export.


Assuntos
Cardiomegalia/tratamento farmacológico , Histona Desacetilases/metabolismo , Carioferinas/antagonistas & inibidores , Miócitos Cardíacos/efeitos dos fármacos , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Amidas/farmacologia , Compostos de Anilina/farmacologia , Animais , Cardiomegalia/metabolismo , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Células Cultivadas , Inibidores de Histona Desacetilases , Histona Desacetilases/química , Humanos , Carioferinas/metabolismo , Microscopia de Fluorescência , Miócitos Cardíacos/metabolismo , Fosforilação , Ratos , Ratos Sprague-Dawley , Receptores Citoplasmáticos e Nucleares/metabolismo , Relação Estrutura-Atividade , Proteína Exportina 1
12.
J Biomol Screen ; 12(2): 220-8, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17351185

RESUMO

After finishing the primary high-throughput screening, the screening team is often faced with thousands of hits to be evaluated further. Effective filtering of these hits is crucial in identifying leads. Mode of inhibition (MOI) study is extremely useful in validating whether the observed compound activity is specific to the biological target. In this article, the authors describe a high-throughput MOI determination method for evaluating thousands of compounds using an existing screening infrastructure. Based on enzyme or receptor kinetics theory, the authors developed the method by measuring the ratio of IC(50) or percent inhibition at 2 carefully chosen substrate or ligand concentrations to define an inhibitor as competitive, uncompetitive, or noncompetitive. This not only facilitates binning of HTS hits according to their MOI but also greatly expands HTS utility in support of the medicinal chemistry team's lead optimization practice. Three case studies are presented to demonstrate how the method was applied successfully in 3 discovery programs targeting either an enzyme or a G-protein-coupled receptor.


Assuntos
Trifosfato de Adenosina/antagonistas & inibidores , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Receptores Acoplados a Proteínas G/antagonistas & inibidores , Animais , Baculoviridae/genética , Sítios de Ligação , Domínio Catalítico , Linhagem Celular , Técnicas de Química Combinatória , Desenho de Fármacos , Avaliação Pré-Clínica de Medicamentos , Escherichia coli/genética , Histidina/química , Humanos , Concentração Inibidora 50 , Cinética , Ligantes , Ligação Proteica , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/química , Proteínas Tirosina Fosfatases/metabolismo , Spodoptera/citologia , Spodoptera/metabolismo
13.
J Biol Chem ; 281(43): 32784-95, 2006 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-16916797

RESUMO

Crystal structures of protein-tyrosine phosphatase 1B in complex with compounds bearing a novel isothiazolidinone (IZD) heterocyclic phosphonate mimetic reveal that the heterocycle is highly complementary to the catalytic pocket of the protein. The heterocycle participates in an extensive network of hydrogen bonds with the backbone of the phosphate-binding loop, Phe(182) of the flap, and the side chain of Arg(221). When substituted with a phenol, the small inhibitor induces the closed conformation of the protein and displaces all waters in the catalytic pocket. Saturated IZD-containing peptides are more potent inhibitors than unsaturated analogs because the IZD heterocycle and phenyl ring directly attached to it bind in a nearly orthogonal orientation with respect to each other, a conformation that is close to the energy minimum of the saturated IZD-phenyl moiety. These results explain why the heterocycle is a potent phosphonate mimetic and an ideal starting point for designing small nonpeptidic inhibitors.


Assuntos
Mimetismo Molecular , Organofosfonatos/farmacologia , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Tirosina Fosfatases/química , Tiazóis/farmacologia , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Escherichia coli/genética , Humanos , Ligação de Hidrogênio , Hidrólise , Concentração Inibidora 50 , Cinética , Modelos Moleculares , Estrutura Molecular , Conformação Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteínas Tirosina Fosfatases/análise , Proteínas Tirosina Fosfatases/isolamento & purificação , Relação Estrutura-Atividade , Especificidade por Substrato , Água/química
14.
J Med Chem ; 48(21): 6544-8, 2005 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-16220970

RESUMO

Structure-based design led to the discovery of novel (S)-isothiazolidinone ((S)-IZD) heterocyclic phosphotyrosine (pTyr) mimetics that when incorporated into dipeptides are exceptionally potent, competitive, and reversible inhibitors of protein tyrosine phosphatase 1B (PTP1B). The crystal structure of PTP1B in complex with our most potent inhibitor 12 revealed that the (S)-IZD heterocycle interacts extensively with the phosphate binding loop precisely as designed in silico. Our data provide strong evidence that the (S)-IZD is the most potent pTyr mimetic reported to date.


Assuntos
Dipeptídeos/síntese química , Fosfotirosina/química , Proteínas Tirosina Fosfatases/antagonistas & inibidores , Proteínas Tirosina Fosfatases/química , Tiazóis/síntese química , Cristalografia por Raios X , Dipeptídeos/química , Desenho de Fármacos , Modelos Moleculares , Mimetismo Molecular , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Relação Quantitativa Estrutura-Atividade , Estereoisomerismo , Tiazóis/química
15.
J Biol Chem ; 280(49): 40838-44, 2005 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-16207718

RESUMO

The hexameric cylindrical Hsp100 chaperone ClpA mediates ATP-dependent unfolding and translocation of recognized substrate proteins into the coaxially associated serine protease ClpP. Each subunit of ClpA is composed of an N-terminal domain of approximately 150 amino acids at the top of the cylinder followed by two AAA+ domains. In earlier studies, deletion of the N-domain was shown to have no effect on the rate of unfolding of substrate proteins bearing a C-terminal ssrA tag, but it did reduce the rate of degradation of these proteins (Lo, J. H., Baker, T. A., and Sauer, R. T. (2001) Protein Sci. 10, 551-559; Singh, S. K., Rozycki, J., Ortega, J., Ishikawa, T., Lo, J., Steven, A. C., and Maurizi, M. R. (2001) J. Biol. Chem. 276, 29420-29429). Here we demonstrate, using both fluorescence resonance energy transfer to measure the arrival of substrate at ClpP and competition between wild-type and an inactive mutant form of ClpP, that this effect on degradation is caused by diminished stability of the ClpA-ClpP complex during translocation and proteolysis, effectively disrupting the targeting of unfolded substrates to the protease. We have also examined two larger ssrA-tagged substrates, CFP-GFP-ssrA and luciferase-ssrA, and observed different behaviors. CFP-GFP-ssrA is not efficiently unfolded by the truncated chaperone whereas luciferase-ssrA is, suggesting that the former requires interaction with the N-domains, likely via the body of the protein, to stabilize its binding. Thus, the N-domains play a key allosteric role in complex formation with ClpP and may also have a critical role in recognizing certain tag elements and binding some substrate proteins.


Assuntos
Endopeptidase Clp/química , Endopeptidase Clp/metabolismo , Sítio Alostérico , Sítios de Ligação , Transporte Biológico , Endopeptidase Clp/genética , Transferência Ressonante de Energia de Fluorescência , Expressão Gênica , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Luciferases/metabolismo , Mutagênese , Fragmentos de Peptídeos/metabolismo , Dobramento de Proteína , RNA Bacteriano , Proteínas Recombinantes de Fusão/metabolismo , Relação Estrutura-Atividade , Especificidade por Substrato
16.
Mol Cell ; 9(3): 673-83, 2002 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11931773

RESUMO

In the bacterial cytosol, ATP-dependent protein degradation is performed by several different chaperone-protease pairs, including ClpAP. The mechanism by which these machines specifically recognize substrates remains unclear. Here, we report the identification of a ClpA cofactor from Escherichia coli, ClpS, which directly influences the ClpAP machine by binding to the N-terminal domain of the chaperone ClpA. The degradation of ClpAP substrates, both SsrA-tagged proteins and ClpA itself, is specifically inhibited by ClpS. In contrast, ClpS enhanced ClpA recognition of two heat-aggregated proteins in vitro and, consequently, the ClpAP-mediated disaggregation and degradation of these substrates. We conclude that ClpS modifies ClpA substrate specificity, potentially redirecting degradation by ClpAP toward aggregated proteins.


Assuntos
Adenosina Trifosfatases/metabolismo , Proteínas de Transporte/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Óperon/genética , Serina Endopeptidases/metabolismo , Adenosina Trifosfatases/genética , Sequência de Aminoácidos , Proteínas de Transporte/genética , Endopeptidase Clp , Escherichia coli/genética , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Temperatura Alta , Luciferases/metabolismo , Malato Desidrogenase/metabolismo , Dados de Sequência Molecular , Ligação Proteica , RNA Bacteriano/metabolismo , Alinhamento de Sequência , Serina Endopeptidases/genética , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...