Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
PLoS One ; 13(6): e0198374, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29879184

RESUMO

Protein tyrosine kinase 6 (PTK6, or BRK) is aberrantly expressed in breast cancers, and emerging as an oncogene that promotes tumor cell proliferation, migration and evasion. Both kinase-dependent and -independent functions of PTK6 in driving tumor growth have been described, therefore targeting PTK6 kinase activity by small molecule inhibitors as a therapeutic approach to treat cancers remains to be validated. In this study, we identified novel, potent and selective PTK6 kinase inhibitors as a means to investigate the role of PTK6 kinase activity in breast tumorigenesis. We report here the crystal structures of apo-PTK6 and inhibitor-bound PTK6 complexes, providing the structural basis for small molecule interaction with PTK6. The kinase inhibitors moderately suppress tumor cell growth in 2D and 3D cell cultures. However, the tumor cell growth inhibition shows neither correlation with the PTK6 kinase activity inhibition, nor the total or activated PTK6 protein levels in tumor cells, suggesting that the tumor cell growth is independent of PTK6 kinase activity. Furthermore, in engineered breast tumor cells overexpressing PTK6, the inhibition of PTK6 kinase activity does not parallel the inhibition of tumor cell growth with a >500-fold shift in compound potencies (IC50 values). Overall, these findings suggest that the kinase activity of PTK6 does not play a significant role in tumorigenesis, thus providing important evidence against PTK6 kinase as a potential therapeutic target for breast cancer treatment.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cristalografia por Raios X , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Modelos Moleculares , Proteínas de Neoplasias/genética , Fosforilação , Proteínas Tirosina Quinases/genética , Bibliotecas de Moléculas Pequenas/química , Relação Estrutura-Atividade
2.
J Pharmacokinet Pharmacodyn ; 41(2): 127-39, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24578187

RESUMO

Human Hexokinase IV, or glucokinase (GK), is a regulator of glucose concentrations in the body. It plays a key role in pancreatic insulin secretion as well as glucose biotransformation in the liver, making it a potentially viable target for treatment of Type 2 diabetes. Allosteric activators of GK have been shown to decrease blood glucose concentrations in both animals and humans. Here, the development of a mathematical model is presented that describes glucose modulation in an ob/ob mouse model via administration of a potent GK activator, with the goal of projecting a human efficacious dose and plasma exposure. The model accounts for the allosteric interaction between GK, the activator, and glucose using a modified Hill function. Based on model simulations using data from the ob/ob mouse and in vitro studies, human projections of glucose response to the GK activator are presented, along with dose and regimen predictions to maintain clinically significant decreases in blood glucose in a Type 2 diabetic patient. This effort serves as a basis to build a detailed mechanistic understanding of GK and its role as a therapeutic target for Type 2 diabetes, and it highlights the benefits of using such an approach in a drug discovery setting.


Assuntos
Azetidinas/farmacologia , Benzofuranos/farmacologia , Diabetes Mellitus Tipo 2/metabolismo , Ativadores de Enzimas/farmacocinética , Glucoquinase/metabolismo , Hipoglicemiantes/farmacocinética , Modelos Biológicos , Animais , Azetidinas/farmacocinética , Azetidinas/uso terapêutico , Benzofuranos/farmacocinética , Benzofuranos/uso terapêutico , Glicemia/metabolismo , Diabetes Mellitus Tipo 2/tratamento farmacológico , Ativadores de Enzimas/uso terapêutico , Humanos , Hipoglicemiantes/uso terapêutico , Masculino , Camundongos , Camundongos Obesos
3.
Bioorg Med Chem Lett ; 23(9): 2787-92, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23506825

RESUMO

PI3K, AKT and mTOR, key kinases from a frequently dysregulated PI3K signaling pathway, have been extensively pursued to treat a variety of cancers in oncology. Clinical trials of PF-04691502, a highly potent and selective ATP competitive kinase inhibitor of class 1 PI3Ks and mTOR, from 4-methylpyridopyrimidinone series, led to the discovery of a metabolite with a terminal carboxylic acid, PF-06465603. This paper discusses structure-based drug design, SAR and antitumor activity of the MPP derivatives with a terminal alcohol, a carboxylic acid or a carboxyl amide.


Assuntos
Antineoplásicos/química , Desenho de Fármacos , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/química , Pirimidinonas/química , Serina-Treonina Quinases TOR/antagonistas & inibidores , Animais , Antineoplásicos/síntese química , Sítios de Ligação , Linhagem Celular Tumoral , Cristalografia por Raios X , Humanos , Camundongos , Simulação de Acoplamento Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/síntese química , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Piridonas/química , Pirimidinas/química , Pirimidinonas/síntese química , Transdução de Sinais , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Bioorg Med Chem Lett ; 22(15): 5098-103, 2012 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-22749419
5.
Mol Cancer Ther ; 11(1): 57-65, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22084163

RESUMO

Inhibition of the Smoothened (Smo) represents a promising therapeutic strategy for treating malignant tumors that are dependent on the Hedgehog (Hh) signaling pathway. PF-5274857 is a novel Smo antagonist that specifically binds to Smo with a K(i) of 4.6 ± 1.1 nmol/L and completely blocks the transcriptional activity of the downstream gene Gli1 with an IC(50) of 2.7 ± 1.4 nmol/L in cells. This Smo antagonist showed robust antitumor activity in a mouse model of medulloblastoma with an in vivo IC(50) of 8.9 ± 2.6 nmol/L. The downregulation of Gli1 is closely linked to the tumor growth inhibition in patched(+/-) medulloblastoma mice. Mathematical analysis of the relationship between the drug's pharmacokinetics and Gli1 pharmacodynamics in patched(+/-) medulloblastoma tumor models yielded similar tumor and skin Gli1 IC(50) values, suggesting that skin can be used as a surrogate tissue for the measurement of tumor Gli1 levels. In addition, PF-5274857 was found to effectively penetrate the blood-brain barrier and inhibit Smo activity in the brain of primary medulloblastoma mice, resulting in improved animal survival rates. The brain permeability of PF-5274857 was also confirmed and quantified in nontumor-bearing preclinical species with an intact blood-brain barrier. PF-5274857 was orally available and metabolically stable in vivo. These findings suggest that PF-5274857 is a potentially attractive clinical candidate for the treatment of tumor types including brain tumors and brain metastasis driven by an activated Hh pathway.


Assuntos
2,2'-Dipiridil/análogos & derivados , Proteínas Hedgehog/antagonistas & inibidores , Proteínas Hedgehog/metabolismo , Meduloblastoma/tratamento farmacológico , Piperazinas/farmacologia , Receptores Acoplados a Proteínas G/antagonistas & inibidores , 2,2'-Dipiridil/farmacologia , Animais , Antineoplásicos , Barreira Hematoencefálica/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Linhagem Celular Tumoral , Células HEK293 , Humanos , Fatores de Transcrição Kruppel-Like/genética , Fatores de Transcrição Kruppel-Like/metabolismo , Meduloblastoma/patologia , Camundongos , Camundongos SCID , Camundongos Transgênicos , Receptores Patched , Ratos , Ratos Wistar , Receptores de Superfície Celular/genética , Receptores Acoplados a Proteínas G/metabolismo , Transdução de Sinais/efeitos dos fármacos , Receptor Smoothened , Proteína GLI1 em Dedos de Zinco
6.
Mol Cancer Ther ; 10(11): 2189-99, 2011 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-21750219

RESUMO

Deregulation of the phosphoinositide 3-kinase (PI3K) signaling pathway such as by PTEN loss or PIK3CA mutation occurs frequently in human cancer and contributes to resistance to antitumor therapies. Inhibition of key signaling proteins in the pathway therefore represents a valuable targeting strategy for diverse cancers. PF-04691502 is an ATP-competitive PI3K/mTOR dual inhibitor, which potently inhibited recombinant class I PI3K and mTOR in biochemical assays and suppressed transformation of avian fibroblasts mediated by wild-type PI3K γ, δ, or mutant PI3Kα. In PIK3CA-mutant and PTEN-deleted cancer cell lines, PF-04691502 reduced phosphorylation of AKT T308 and AKT S473 (IC(50) of 7.5-47 nmol/L and 3.8-20 nmol/L, respectively) and inhibited cell proliferation (IC(50) of 179-313 nmol/L). PF-04691502 inhibited mTORC1 activity in cells as measured by PI3K-independent nutrient stimulated assay, with an IC(50) of 32 nmol/L and inhibited the activation of PI3K and mTOR downstream effectors including AKT, FKHRL1, PRAS40, p70S6K, 4EBP1, and S6RP. Short-term exposure to PF-04691502 predominantly inhibited PI3K, whereas mTOR inhibition persisted for 24 to 48 hours. PF-04691502 induced cell cycle G(1) arrest, concomitant with upregulation of p27 Kip1 and reduction of Rb. Antitumor activity was observed in U87 (PTEN null), SKOV3 (PIK3CA mutation), and gefitinib- and erlotinib-resistant non-small cell lung carcinoma xenografts. In summary, PF-04691502 is a potent dual PI3K/mTOR inhibitor with broad antitumor activity. PF-04691502 has entered phase I clinical trials.


Assuntos
Antineoplásicos/farmacologia , Inibidores Enzimáticos/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Piridonas/farmacologia , Pirimidinas/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Trifosfato de Adenosina/metabolismo , Animais , Antineoplásicos/uso terapêutico , Ligação Competitiva , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidores Enzimáticos/uso terapêutico , Feminino , Humanos , Camundongos , Camundongos Nus , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Ligação Proteica/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Bioorg Med Chem Lett ; 21(4): 1270-4, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21269826

RESUMO

Intra-molecular hydrogen bonding was introduced to the quinazoline motif to form a pseudo ring (intra-molecular H-bond scaffold, iMHBS) to mimic our previous published core structures, pyrido[2.3-D]pyrimidin-7-one and pteridinone, as PI3K/mTOR dual inhibitors. This design results in potent PI3K/mTOR dual inhibitors and the purposed intra-molecular hydrogen bonding structure is well supported by co-crystal structure in PI3Kγ enzyme. In addition, a novel synthetic route was developed for these analogs.


Assuntos
Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/química , Quinazolinas/química , Serina-Treonina Quinases TOR/antagonistas & inibidores , Sítios de Ligação , Linhagem Celular Tumoral , Cristalografia por Raios X , Humanos , Ligação de Hidrogênio , Modelos Químicos , Modelos Moleculares , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/síntese química , Quinazolinas/farmacologia , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR/metabolismo
8.
BMC Genomics ; 12 Suppl 3: S3, 2011 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-22369201

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is the fifth most common cancer worldwide. A number of molecular profiling studies have investigated the changes in gene and protein expression that are associated with various clinicopathological characteristics of HCC and generated a wealth of scattered information, usually in the form of gene signature tables. A database of the published HCC gene signatures would be useful to liver cancer researchers seeking to retrieve existing differential expression information on a candidate gene and to make comparisons between signatures for prioritization of common genes. A challenge in constructing such database is that a direct import of the signatures as appeared in articles would lead to a loss or ambiguity of their context information that is essential for a correct biological interpretation of a gene's expression change. This challenge arises because designation of compared sample groups is most often abbreviated, ad hoc, or even missing from published signature tables. Without manual curation, the context information becomes lost, leading to uninformative database contents. Although several databases of gene signatures are available, none of them contains informative form of signatures nor shows comprehensive coverage on liver cancer. Thus we constructed Liverome, a curated database of liver cancer-related gene signatures with self-contained context information. DESCRIPTION: Liverome's data coverage is more than three times larger than any other signature database, consisting of 143 signatures taken from 98 HCC studies, mostly microarray and proteome, and involving 6,927 genes. The signatures were post-processed into an informative and uniform representation and annotated with an itemized summary so that all context information is unambiguously self-contained within the database. The signatures were further informatively named and meaningfully organized according to ten functional categories for guided browsing. Its web interface enables a straightforward retrieval of known differential expression information on a query gene and a comparison of signatures to prioritize common genes. The utility of Liverome-collected data is shown by case studies in which useful biological insights on HCC are produced. CONCLUSION: Liverome database provides a comprehensive collection of well-curated HCC gene signatures and straightforward interfaces for gene search and signature comparison as well. Liverome is available at http://liverome.kobic.re.kr.


Assuntos
Carcinoma Hepatocelular/genética , Bases de Dados Factuais , Neoplasias Hepáticas/genética , Carcinoma Hepatocelular/metabolismo , Humanos , Armazenamento e Recuperação da Informação , Internet , Neoplasias Hepáticas/metabolismo , Transcriptoma , Interface Usuário-Computador
9.
Bioorg Med Chem Lett ; 20(20): 6096-9, 2010 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-20817449

RESUMO

Pteridinones were designed based on a non-selective kinase template. Because of the uniqueness of the PI3K and mTOR binding pockets, a methyl group was introduced to C-4 position of the peteridinone core to give compounds with excellent selectivity for PI3K and mTOR. This series of compounds were further optimized to improve their potency against PI3Kα and mTOR. Finally, orally active compounds with improved solubility and robust in vivo efficacy in tumor growth inhibition were identified as well.


Assuntos
Antineoplásicos/química , Antineoplásicos/uso terapêutico , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/uso terapêutico , Pteridinas/química , Pteridinas/uso terapêutico , Serina-Treonina Quinases TOR/antagonistas & inibidores , Administração Oral , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Glioma/tratamento farmacológico , Humanos , Camundongos , Modelos Moleculares , Neoplasias/tratamento farmacológico , Fosfatidilinositol 3-Quinases/química , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia , Pteridinas/administração & dosagem , Pteridinas/farmacologia , Solubilidade , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR/química , Serina-Treonina Quinases TOR/metabolismo
10.
Biochemistry ; 49(39): 8488-98, 2010 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-20804212

RESUMO

The mammalian target of rapamycin (mTOR) is a Ser/Thr protein kinase and a major controller of cell growth. In cells, mTOR forms two distinct multiprotein complexes, mTORC1 and mTORC2. The mTORC1 complex can phosphorylate 4EBP1 and S6K1, two key regulators of translation initiation, whereas mTORC2 phosphorylates AKT1, an event required for AKT1 activation. Here, we expressed and purified human mTORC1 and mTORC2 from HEK-293 cells using FLAG-M2 affinity chromatography. Western blotting analysis using phospho-specific antibodies indicated that recombinant mTORC1 and mTORC2 exhibit distinct substrate preferences in vitro, consistent with their roles in cells. To improve our understanding of the enzymatic properties of mTOR alone and mTOR in its complex form, steady-state kinetic profiles of truncated mTOR containing the kinase domain (residues 1360-2549) and mTORC1 were determined. The results revealed that mTORC1 is catalytically less active than truncated mTOR, as evidenced by 4.7- and 3.1-fold decreases in catalytic efficiency, k(cat)/K(m), for ATP and 4EBP1, respectively. We also found that truncated mTOR undergoes autophosphorylation through an intramolecular mechanism. Mass spectrometric analysis identified two novel mTOR autophosphorylation sites, Ser2454 and either Thr2473 or Thr2474, in addition to the previously reported Ser2481 site. Truncated mTOR and mTORC1 were completely inhibited by ATP competitive inhibitors PI103 and BEZ235 and partially inhibited by rapamycin/FKBP12 in a noncompetitive fashion toward ATP. All inhibitors tested exhibited similar inhibitory potencies between mTORC1 and truncated mTOR containing the kinase domain. Our studies presented here provide the first detailed kinetic studies of a recombinant mTOR complex.


Assuntos
Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/isolamento & purificação , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Sequência de Aminoácidos , Proteínas de Transporte/antagonistas & inibidores , Proteínas de Transporte/genética , Proteínas de Transporte/isolamento & purificação , Proteínas de Transporte/metabolismo , Linhagem Celular , Inibidores Enzimáticos/farmacologia , Humanos , Peptídeos e Proteínas de Sinalização Intracelular , Cinética , Alvo Mecanístico do Complexo 1 de Rapamicina , Metais/metabolismo , Dados de Sequência Molecular , Complexos Multiproteicos , Fosforilação , Proteínas/antagonistas & inibidores , Proteínas/genética , Proteínas/isolamento & purificação , Proteínas/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Proteína Regulatória Associada a mTOR , Sirolimo/farmacologia , Especificidade por Substrato , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/isolamento & purificação , Regulação para Cima , Homólogo LST8 da Proteína Associada a mTOR
11.
Biochemistry ; 48(11): 2514-21, 2009 Mar 24.
Artigo em Inglês | MEDLINE | ID: mdl-19187021

RESUMO

The glucokinase V62M and G72R mutations are naturally occurring and known to associate with hyperglycemia in humans. Structurally, V62 and G72 residues are located in close proximity to the allosteric site where hypoglycemia-linked activating mutations are clustered. To address the mechanism by which these variants alter the physiological phenotype, we characterized the biochemical and biophysical properties of the enzymes. Recombinant proteins were purified without affinity tags, and their steady-state kinetics and glucose binding affinities were determined. Both enzymes showed reduced rates of turnover (k(cat)) and reduced glucose affinity (i.e., increased K(0.5) and K(D) values). Their thermal stability did not largely differ from that of wild-type glucokinase. However, V62M and G72R lost the stabilizing protein interactions with glucokinase regulatory protein, which may contribute to lower activity in vivo. Both mutants were subject to activation by small molecule activators. In conclusion, the decreased enzyme activities of V62M and G72R observed in this study are consistent with the hyperglycemic phenotype.


Assuntos
Diabetes Mellitus Tipo 2/enzimologia , Glucoquinase/química , Glucoquinase/metabolismo , Mutação de Sentido Incorreto , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Estabilidade Enzimática , Variação Genética , Glucoquinase/genética , Glucoquinase/isolamento & purificação , Glucose/metabolismo , Humanos , Cinética
12.
Biochemistry ; 47(17): 5028-36, 2008 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-18370405

RESUMO

Small molecule activators of glucokinase (GK) were used in kinetic and equilibrium binding studies to probe the biochemical basis for their allosteric effects. These small molecules decreased the glucose K 0.5 ( approximately 1 mM vs approximately 8 mM) and the glucose cooperativity (Hill coefficient of 1.2 vs 1.7) and lowered the k cat to various degrees (62-95% of the control activity). These activators relieved GK's inhibition from glucokinase regulatory protein (GKRP) in a glucose-dependent manner and activated GK to the same extent as control reactions in the absence of GKRP. In equilibrium binding studies, the intrinsic glucose affinity to the activator-bound enzyme was determined and demonstrated a 700-fold increase relative to the apoenzyme. This is consistent with a reduction in apparent glucose K D and the steady-state parameter K 0.5 as a result of enzyme equilibrium shifting to the activator-bound form. The binding of small molecules to GK was dependent on glucose, consistent with the structural evidence for an allosteric binding site which is present in the glucose-induced, active enzyme form of GK and absent in the inactive apoenzyme [Kamata et al. (2004) Structure 12, 429-438]. A mechanistic model that brings together the kinetic and structural data is proposed which allows qualitative and quantitative analysis of the glucose-dependent GK regulation by small molecules. The regulation of GK activation by glucose may have an important implication for the discovery and design of GK activators as potential antidiabetic agents.


Assuntos
Ativadores de Enzimas/química , Ativadores de Enzimas/farmacologia , Glucoquinase/metabolismo , Glucose/metabolismo , Regulação Alostérica , Proteínas de Transporte/metabolismo , Cromatografia em Gel , Ativação Enzimática/efeitos dos fármacos , Ativadores de Enzimas/metabolismo , Cinética , Ligação Proteica
13.
J Biol Chem ; 281(52): 40201-7, 2006 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-17082186

RESUMO

Glucokinase (GK) has several known polymorphic activating mutations that increase the enzyme activity by enhancing glucose binding affinity and/or by alleviating the inhibition of glucokinase regulatory protein (GKRP), a key regulator of GK activity in the liver. Kinetic studies were undertaken to better understand the effect of these mutations on the enzyme mechanism of GK activation and GKRP regulation and to relate the enzyme properties to the associated clinical phenotype of hypoglycemia. Similar to wild type GK, the transient kinetics of glucose binding for activating mutations follows a general two-step mechanism, the formation of an enzyme-glucose complex followed by an enzyme conformational change. However, the kinetics for each step differed from wild type GK and could be grouped into specific types of kinetic changes. Mutations T65I, Y214C, and A456V accelerate glucose binding to the apoenzyme form, whereas W99R, Y214C, and V455M facilitate enzyme isomerization to the active form. Mutations that significantly enhance the glucose binding to the apoenzyme also disrupt the protein-protein interaction with GKRP to a large extent, suggesting these mutations may adopt a more compact conformation in the apoenzyme favorable for glucose binding. Y214C is the most active mutation (11-fold increase in k(cat)/K(0.5)(h)) and exhibits the most severe clinical effects of hypoglycemia. In contrast, moderate activating mutation A456V nearly abolishes the GKRP inhibition (76-fold increase in K(i)) but causes only mild hypoglycemia. This suggests that the alteration in GK enzyme activity may have a more profound biological impact than the alleviation of GKRP inhibition.


Assuntos
Substituição de Aminoácidos/genética , Proteínas de Transporte/fisiologia , Glucoquinase/metabolismo , Mutação Puntual , Sítio Alostérico/genética , Apoenzimas/antagonistas & inibidores , Apoenzimas/genética , Apoenzimas/metabolismo , Ligação Competitiva/genética , Proteínas de Transporte/metabolismo , Ativação Enzimática/genética , Regulação da Expressão Gênica/genética , Glucoquinase/antagonistas & inibidores , Glucoquinase/genética , Glucose/metabolismo , Humanos , Cinética , Polimorfismo Genético , Espectrofotometria
14.
Bioorg Med Chem Lett ; 16(23): 6116-9, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16979341

RESUMO

A series of novel pyridine-2-propanoic acids was synthesized. A structure-activity relationship study of these compounds led to the identification of potent dual PPARalpha/gamma agonists with varied isoform selectivity. Based on the results of efficacy studies in diabetic (db/db) mice, and the desired pharmacokinetic parameters, compound (S)-13 was selected for further profiling.


Assuntos
Hipoglicemiantes/síntese química , Hipoglicemiantes/farmacologia , PPAR alfa/agonistas , PPAR gama/agonistas , Piridinas/química , Piridinas/farmacologia , Animais , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Linhagem Celular Tumoral , Diabetes Mellitus/sangue , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/patologia , Humanos , Hipoglicemiantes/química , Hipoglicemiantes/uso terapêutico , Masculino , Camundongos , Estrutura Molecular , PPAR alfa/metabolismo , PPAR gama/metabolismo , Piridinas/síntese química , Piridinas/uso terapêutico , Relação Estrutura-Atividade
15.
Bioorg Med Chem Lett ; 16(23): 6120-3, 2006 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-16973358

RESUMO

A series of novel pyridine-3-propanoic acids was synthesized. A structure-activity relationship study of these compounds led to the identification of potent dual PPARalpha/gamma agonists with varied isoform selectivity. Based on the results of efficacy studies in diabetic (db/db) mice, and the desired pharmacokinetic parameters, compounds (S)-14 and (S)-19 were selected for further profiling.


Assuntos
Hipoglicemiantes/síntese química , Hipoglicemiantes/farmacologia , PPAR alfa/agonistas , PPAR gama/agonistas , Piridinas/sangue , Piridinas/farmacologia , Animais , Glicemia/metabolismo , Peso Corporal/efeitos dos fármacos , Linhagem Celular Tumoral , Diabetes Mellitus/sangue , Diabetes Mellitus/tratamento farmacológico , Diabetes Mellitus/patologia , Éter/química , Humanos , Hipoglicemiantes/química , Hipoglicemiantes/uso terapêutico , Camundongos , Estrutura Molecular , PPAR alfa/metabolismo , PPAR gama/metabolismo , Piridinas/síntese química , Piridinas/uso terapêutico , Relação Estrutura-Atividade , Tiazolidinedionas/química
16.
Biochemistry ; 45(24): 7553-62, 2006 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-16768451

RESUMO

The transient kinetics of glucose binding to glucokinase (GK) was studied using stopped-flow fluorescence spectrophotometry to investigate the underlying mechanism of positive cooperativity of monomeric GK with glucose. Glucose binding to GK was shown to display biphasic kinetics that fit best to a reversible two-step mechanism. GK initially binds glucose to form a transient intermediate, namely, E* x glucose, followed by a conformational change to a catalytically competent E x glucose complex. The microscopic rate constants for each step were determined as follows: on rate k1 of 557 M(-1) s(-1) and off rate k(-1) of 8.1 s(-1) for E* x glucose formation, and forward rate k2 of 0.45 s(-1) and reverse rate k(-2) of 0.28 s(-1) for the conformational change from E* x glucose to E x glucose. These results suggest that the enzyme conformational change induced by glucose binding is a reversible, slow event that occurs outside the catalytic cycle (kcat = 38 s(-1)). This slow transition between the two enzyme conformations modulated by glucose likely forms the kinetic foundation for the allosteric regulation. Furthermore, the kinetics of the enzyme conformational change was altered in favor of E x glucose formation in D2O, accompanied by a decrease in cooperativity with glucose (Hill slope of 1.3 in D2O vs 1.7 in H2O). The deuterium solvent isotope effects confirm the role of the conformational change in the magnitude of glucose cooperativity. Similar studies were conducted with GK activating mutation Y214C at the allosteric activator site that is likely involved in the protein domain rearrangement associated with glucose binding. The mutation enhanced equilibrium glucose binding by a combination of effects on both the formation of E* x glucose and an enzyme conformational change to E x glucose. Kinetic simulation by KINSIM supports the conclusion that the kinetic cooperativity of GK arises from slow glucose-induced conformational changes in GK.


Assuntos
Glucoquinase/química , Glucose/farmacologia , Regulação Alostérica/efeitos dos fármacos , Apoenzimas/química , Apoenzimas/metabolismo , Sítios de Ligação , Varredura Diferencial de Calorimetria , Relação Dose-Resposta a Droga , Cinética , Modelos Moleculares , Conformação Proteica/efeitos dos fármacos , Espectrometria de Fluorescência
17.
Metab Eng ; 7(1): 38-44, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15721809

RESUMO

In this paper, we described the optimization of a generic binding assay to measure ligand-receptor interactions for peroxisome proliferator-activated receptors (PPARs). The assay is based on scintillation proximity assay, in which a protein is coated on scintillant-incorporated beads, and a radiolabeled ligand stimulates the beads to emit a signal by binding to the immobilized protein. An intrinsic binding affinity of unlabeled ligands is determined by competitive displacement of the radioligand. The protein coating and ligand binding are achieved in one step by simply mixing ligands, protein and beads in sequence. No additional steps of pre-coating and washing of beads are required. Protein is captured on beads effectively by electrostatic interactions, thus no affinity labeling of protein is required. In data analysis, ligands are grouped into two classes based on their binding affinities. For tight binding ligands, an equation is derived to accurately determine the binding affinity. Otherwise a general equation applies. This quantitative and high throughput assay provides a tool to screen a large library of molecules in search of potent ligands.


Assuntos
PPAR gama/análise , PPAR gama/metabolismo , Mapeamento de Interação de Proteínas/métodos , Radioimunoensaio/métodos , Contagem de Cintilação/métodos , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
19.
Anal Biochem ; 307(2): 287-96, 2002 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12202246

RESUMO

A fluorescence polarization (FP) assay was developed to determine the concentration of a c-myc-tagged recombinant protein in a crude cell extract. The basis of the assay was a competition between a c-myc-tagged protein and a fluorescein-labeled c-myc peptide for a c-myc antibody Fab. Fluorescein-labeled c-myc peptide produced a high-fluorescence polarization signal upon binding to the c-myc antibody, which can be inhibited in the presence of a c-myc-tagged protein. Quantitation of a c-myc-tagged protein was realized by measuring the decrease in fluorescence polarization. The observed IC(50) values in the competition FP assay were similar among all monomeric c-myc-tagged proteins tested, indicating that the interaction of the c-myc tag with the antibody was independent of the fusion protein sequence. The c-myc-tagged protein concentrations measured by FP were found to correlate well with values derived from a spike experiment and with values obtained by quantitative immunoblot. This assay was not perturbed by the presence of crude cell lysate, dithiothreitol or detergents, and worked with both native and denatured samples from several expression systems, including Escherichia coli, Pichia, insect cells, and mammalian cells. The assay under the current condition can detect as low as 0.05% expression level of c-myc-tagged protein with regards to total proteins, depending on the expression system. This assay is both quantitative and rapid (less than 15min) and is therefore suitable for the optimization of recombinant protein expression conditions as well as for the monitoring of protein purification procedures.


Assuntos
Extratos Celulares/química , Polarização de Fluorescência/métodos , Proteínas Proto-Oncogênicas c-myc/análise , Proteínas Recombinantes de Fusão/análise , Ligação Competitiva/efeitos dos fármacos , Linhagem Celular , Ditiotreitol/farmacologia , Escherichia coli , Fluoresceína/análise , Humanos , Immunoblotting , Fragmentos Fab das Imunoglobulinas/imunologia , Imunoglobulina G/imunologia , Pichia , Desnaturação Proteica , Proteínas Proto-Oncogênicas c-myc/imunologia , Receptores Citoplasmáticos e Nucleares/análise , Proteínas Recombinantes de Fusão/imunologia , Fatores de Tempo , Fatores de Transcrição/análise
20.
J Steroid Biochem Mol Biol ; 81(3): 217-25, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12163133

RESUMO

The mechanism by which ligands of nuclear receptors show differential effects on gene transcription is not fully understood, but is believed to result in part from the preferential recruitment and/or displacement of coactivators and corepressors. We have explored the interaction of several known ligands and the nuclear receptor (peroxisome proliferator activated receptor alpha, PPARalpha) using scintillation proximity assay (SPA) and the interaction of LXXLL containing peptides derived from three coactivators (SRC-1, CBP and PGC-1) with PPARalpha in the presence of PPARalpha agonist ligands using fluorescence resonance energy transfer (FRET). The EC(50)s of the individual ligands for recruitment showed the same rank order regardless of the coactivator peptide used, with GW2331

Assuntos
Receptores Citoplasmáticos e Nucleares/metabolismo , Fatores de Transcrição/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , DNA Complementar/metabolismo , Transferência de Energia , Escherichia coli/metabolismo , Histona Acetiltransferases , Humanos , Cinética , Ligantes , Coativador 1 de Receptor Nuclear , Peptídeos/química , Peptídeos/metabolismo , Plasmídeos/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas c-myc/metabolismo , Espectrofotometria , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA