Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
ACS Med Chem Lett ; 15(5): 722-730, 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38746878

RESUMO

Colony stimulating factor-1 receptor (CSF1R or c-FMS), a class III receptor tyrosine kinase expressed on members of the mononuclear phagocyte system (MPS), plays a key role in the proper functioning of macrophages, microglia, and related cells. Aberrant signaling through CSF1R has been associated with a variety of disease states, including cancer, inflammation, and neurodegeneration. In this Letter, we detail our efforts to develop novel CSF1R inhibitors. Drawing on previously described compounds, including GW2580 (4), we have discovered a novel series of compounds based on the imidazo[4,5-b]pyridine scaffold. Initial structure-activity relationship studies culminated in the identification of 36, a lead compound with potent CSF1R biochemical and cellular activity, acceptable in vitro ADME properties, and oral exposure in rat.

2.
Curr Res Struct Biol ; 4: 96-105, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35469152

RESUMO

Clostridium difficile toxins are the primary causative agents for hospital-acquired diarrhea and pseudomembranous colitis. Numerous monoclonal antibodies (mAbs) targeting different domains of Clostridium difficile toxin have been reported. Here we report the crystal structures of two mAbs, B1 and B2, in complex with the glycosyltransferase domain (GTD) of the Clostridium difficile toxin B (TcdB). B2 bound to the N-terminal 4 helix bundle of the GTD, a conserved membrane localization domain (MLD) found in the large clostridial glycosylating toxin family implicated in targeting plasma membrane. B1 bound to a distinct epitope at the hinge region between the MLD and the catalytic subdomain of the GTD. Functional studies revealed the potency of these mAbs in vitro and in vivo to be synergistic when given in combination.

3.
ACS Med Chem Lett ; 11(10): 1899-1904, 2020 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-33062171

RESUMO

Hepatocellular carcinoma (HCC) accounts for a majority of primary liver cancer and is one of the most common forms of cancer worldwide. Aberrant signaling of the FGF19-FGFR4 pathway leads to HCC in mice and is hypothesized to be a driver in FGF19 amplified HCC in humans. Multiple small molecule inhibitors have been pursued as targeted therapies for HCC in recent years, including several selective FGFR4 inhibitors that are currently being evaluated in clinical trials. Herein, we report a novel series of highly selective, covalent 2-amino-6,8-dimethyl-pyrido[2,3-d]pyrimidin-7(8H)-ones that potently and selectively inhibit FGFR4 signaling through covalent modification of Cys552, which was confirmed by X-ray crystallography. Correlative target occupancy and pFGFR4 inhibition were observed in vivo, as well as tumor regression in preclinical models of orthotopic and sorafenib-resistant HCC.

4.
Cell Death Dis ; 11(10): 904, 2020 10 23.
Artigo em Inglês | MEDLINE | ID: mdl-33097690

RESUMO

Microglia serve as the innate immune cells of the central nervous system (CNS) by providing continuous surveillance of the CNS microenvironment and initiating defense mechanisms to protect CNS tissue. Upon injury, microglia transition into an activated state altering their transcriptional profile, transforming their morphology, and producing pro-inflammatory cytokines. These activated microglia initially serve a beneficial role, but their continued activation drives neuroinflammation and neurodegeneration. Multiple sclerosis (MS) is a chronic, inflammatory, demyelinating disease of the CNS, and activated microglia and macrophages play a significant role in mediating disease pathophysiology and progression. Colony-stimulating factor-1 receptor (CSF1R) and its ligand CSF1 are elevated in CNS tissue derived from MS patients. We performed a large-scale RNA-sequencing experiment and identified CSF1R as a key node of disease progression in a mouse model of progressive MS. We hypothesized that modulating microglia and infiltrating macrophages through the inhibition of CSF1R will attenuate deleterious CNS inflammation and reduce subsequent demyelination and neurodegeneration. To test this hypothesis, we generated a novel potent and selective small-molecule CSF1R inhibitor (sCSF1Rinh) for preclinical testing. sCSF1Rinh blocked receptor phosphorylation and downstream signaling in both microglia and macrophages and altered cellular functions including proliferation, survival, and cytokine production. In vivo, CSF1R inhibition with sCSF1Rinh attenuated neuroinflammation and reduced microglial proliferation in a murine acute LPS model. Furthermore, the sCSF1Rinh attenuated a disease-associated microglial phenotype and blocked both axonal damage and neurological impairments in an experimental autoimmune encephalomyelitis (EAE) model of MS. While previous studies have focused on microglial depletion following CSF1R inhibition, our data clearly show that signaling downstream of this receptor can be beneficially modulated in the context of CNS injury. Together, these data suggest that CSF1R inhibition can reduce deleterious microglial proliferation and modulate microglial phenotypes during neuroinflammatory pathogenesis, particularly in progressive MS.


Assuntos
Inflamação/patologia , Esclerose Múltipla/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/patologia , Humanos , Macrófagos/efeitos dos fármacos , Camundongos , Microglia/patologia , Esclerose Múltipla/patologia , Transdução de Sinais/efeitos dos fármacos
5.
Langmuir ; 35(42): 13570-13577, 2019 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-31560544

RESUMO

Although metal phthalocyanines are widely used in optoelectronic devices, e.g., as hole-transport and electron-blocking layers, or as UV-stable dyes, their multilayer growth on metal substrates has surprisingly not been studied very systematically. Even for CuPc, one of the most widely studied representatives of phthalocyanines, contradictory structures are reported for films grown on gold, a common electrode material, suggesting that the influence of actual substrate surface properties on film growth has not been sufficiently considered. In this study, we analyze the growth of CuPc films on gold substrates for thicknesses ranging from the initial seed layer to thick multilayers (50 nm) by combining near-edge X-ray absorption spectroscopy with atomic force microscopy and X-ray diffraction. To study the influence of surface roughness, we compare the formation of CuPc films on well-ordered Au(111) and sputter-deposited polycrystalline gold substrates and also investigate the influence of surface contamination by exposing these gold surfaces to air before film growth. While on clean gold substrates, CuPc molecules exclusively adopt a recumbent orientation and form (112̅)-oriented films, they also grow in an upright orientation on contaminated gold surfaces. On Au(111), this leads to the coexistence of (112̅)- and (100)-oriented regions, whereas only (100)-oriented films are formed on contaminated polycrystalline gold. Remarkably, the (112̅)-oriented films consist of extended but isolated crystalline islands, resulting in large overall roughness, whereas the (100)-oriented films consist of rather small domains but have significantly lower film roughness.

6.
Chem Biol Drug Des ; 73(2): 179-88, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19207420

RESUMO

Aberrant activation of the phosphoinositide 3-kinase pathway because of genetic mutations of essential signalling proteins has been associated with human diseases including cancer and diabetes. The pivotal role of 3-phosphoinositide-dependent kinase-1 in the PI3K signalling cascade has made it an attractive target for therapeutic intervention. The N-terminal lobe of the 3-phosphoinositide-dependent kinase-1 catalytic domain contains a docking site which recognizes the non-catalytic C-terminal hydrophobic motifs of certain substrate kinases. The binding of substrate in this so-called PDK1 Interacting Fragment pocket allows interaction with 3-phosphoinositide-dependent kinase-1 and enhanced phosphorylation of downstream kinases. NMR spectroscopy was used to a screen 3-phosphoinositide-dependent kinase-1 domain construct against a library of chemically diverse fragments in order to identify small, ligand-efficient fragments that might interact at either the ATP site or the allosteric PDK1 Interacting Fragment pocket. While majority of the fragment hits were determined to be ATP-site binders, several fragments appeared to interact with the PDK1 Interacting Fragment pocket. Ligand-induced changes in 1H-15N TROSY spectra acquired using uniformly 15N-enriched PDK1 provided evidence to distinguish ATP-site from PDK1 Interacting Fragment-site binding. Caliper assay data and 19F NMR assay data on the PDK1 Interacting Fragment pocket fragments and structurally related compounds identified them as potential allosteric activators of PDK1 function.


Assuntos
Espectroscopia de Ressonância Magnética/métodos , Proteínas Serina-Treonina Quinases/química , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Sítio Alostérico , Domínio Catalítico , Simulação por Computador , Humanos , Hidrogênio/química , Ligantes , Nitrogênio/química , Conformação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Estrutura Terciária de Proteína
7.
Bioorg Med Chem Lett ; 19(1): 226-9, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19019675

RESUMO

An approach and preliminary results for utilizing legacy MEK inhibitors as templates for a reiterative structural based design and synthesis of novel, type III NCKIs (non-classical kinase inhibitors) is described. Evidence is provided that the MEK-pocket or pockets closely related to it may exist in kinases other than MEK.


Assuntos
Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/síntese química , Domínio Catalítico , Desenho de Fármacos , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia
8.
Chem Biol Drug Des ; 70(6): 540-6, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18005335

RESUMO

Polo-like kinase 1 is an important regulator of cell cycle progression whose over-expression is often associated with oncogenesis. Polo-like kinase 1 hence represents an attractive target for cancer intervention. BI 2536 (Boehringer Ingelheim, Ingelheim, Germany), a Polo-like kinase 1 inhibitor currently in clinical trials, exhibits nanomolar potency against Polo-like kinase isoforms and high selectivity against other kinases. We have previously published the crystal structures of the Polo-like kinase 1 domain in complex with AMPPNP and an Aurora A inhibitor. In this work, we present the co-crystal structure of Polo-like kinase 1 with BI 2536. The structure, in combination with selectivity data for BI 2536 and related compounds, illustrates important features for potency and selectivity. In particular, we show that the methoxy group of BI 2536 is an important specificity determinant against non-Polo-like kinases by taking advantage of a small pocket generated by Leu 132 in the hinge region of Polo-like kinase 1. The work presented here provides a framework for structure-based drug design of Polo-like kinase 1-specific inhibitors.


Assuntos
Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Neoplasias/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Pteridinas/química , Adenilil Imidodifosfato/química , Animais , Ciclo Celular/efeitos dos fármacos , Proteínas de Ciclo Celular/química , Ensaios Clínicos como Assunto , Cristalografia por Raios X , Desenho de Fármacos , Humanos , Modelos Moleculares , Proteínas de Neoplasias/química , Neoplasias/tratamento farmacológico , Neoplasias/enzimologia , Ligação Proteica/fisiologia , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Serina-Treonina Quinases/química , Estrutura Terciária de Proteína/fisiologia , Proteínas Proto-Oncogênicas/química , Pteridinas/farmacologia , Pteridinas/uso terapêutico , Relação Estrutura-Atividade , Quinase 1 Polo-Like
9.
Biochemistry ; 46(20): 5960-71, 2007 May 22.
Artigo em Inglês | MEDLINE | ID: mdl-17461553

RESUMO

Polo-like kinase 1 (Plk1) is an attractive target for the development of anticancer agents due to its importance in regulating cell-cycle progression. Overexpression of Plk1 has been detected in a variety of cancers, and expression levels often correlate with poor prognosis. Despite high interest in Plk1-targeted therapeutics, there is currently no structure publicly available to guide structure-based drug design of specific inhibitors. We determined the crystal structures of the T210V mutant of the kinase domain of human Plk1 complexed with the nonhydrolyzable ATP analogue adenylylimidodiphosphate (AMPPNP) or the pyrrolo-pyrazole inhibitor PHA-680626 at 2.4 and 2.1 A resolution, respectively. Plk1 adopts the typical kinase domain fold and crystallized in a conformation resembling the active state of other kinases. Comparison of the kinetic parameters determined for the (unphosphorylated) wild-type enzyme, as well as the T210V and T210D mutants, shows that the mutations primarily affect the kcat of the reaction, with little change in the apparent Km for the protein or nucleotide substrates (kcat = 0.0094, 0.0376, and 0.0049 s-1 and Km(ATP) = 3.2, 4.0, and 3.0 microM for WT, T210D, and T210V, respectively). The structure highlights features of the active site that can be exploited to obtain Plk1-specific inhibitors with selectivity over other kinases and Plk isoforms. These include the presence of a phenylalanine at the bottom of the ATP pocket, combined with a cysteine (as opposed to the more commonly found leucine) in the roof of the binding site, a pocket created by Leu132 in the hinge region, and a cluster of positively charged residues in the solvent-exposed area outside of the adenine pocket adjacent to the hinge region.


Assuntos
Domínio Catalítico , Proteínas de Ciclo Celular/química , Proteínas Serina-Treonina Quinases/química , Proteínas Proto-Oncogênicas/química , Sequência de Aminoácidos , Sítios de Ligação/genética , Domínio Catalítico/genética , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cristalografia por Raios X , Ativação Enzimática/genética , Estabilidade Enzimática/genética , Humanos , Cinética , Dados de Sequência Molecular , Fosforilação , Conformação Proteica , Dobramento de Proteína , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/metabolismo , Quinase 1 Polo-Like
10.
J Biol Chem ; 280(30): 28127-32, 2005 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-15932873

RESUMO

The enzymatic A1 chain of cholera toxin retrotranslocates across the endoplasmic reticulum membrane into the cytosol, where it induces toxicity. Almost all other retrotranslocation substrates are modified by the attachment of polyubiquitin chains and moved into the cytosol by the ubiquitin-interacting p97 ATPase complex. The cholera toxin A1 chain, however, can induce toxicity in the absence of ubiquitination, and the motive force that drives retrotranslocation is not known. Here, we use adenovirus expressing dominant-negative mutants of p97 to test whether p97 is required for toxin action. We find that cholera toxin still functions with only a small decrease in potency in cells that cannot retrotranslocate other substrates at all. These results suggest that p97 does not provide the primary driving force for extracting the A1 chain from the endoplasmic reticulum, a finding that is consistent with a requirement for polyubiquitination in p97 function.


Assuntos
Adenosina Trifosfatases/fisiologia , Toxina da Cólera/química , Proteínas Nucleares/fisiologia , Transporte Proteico , Adenosina Trifosfatases/química , Adenoviridae/genética , Adenoviridae/metabolismo , Animais , Astrocitoma/metabolismo , Células COS , Linhagem Celular Tumoral , Toxina da Cólera/metabolismo , AMP Cíclico/metabolismo , Citosol/metabolismo , Relação Dose-Resposta a Droga , Eletrofisiologia , Retículo Endoplasmático/metabolismo , Genes Dominantes , Genes MHC Classe I/genética , Humanos , Imunoprecipitação , Mutação , Proteínas Nucleares/química , Ligação Proteica , Dobramento de Proteína , Fatores de Tempo , Ubiquitina/metabolismo
11.
Biochemistry ; 44(23): 8312-25, 2005 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-15938621

RESUMO

Phosphodiesterases (PDEs) modulate signaling by cyclic nucleotides in diverse processes such as cardiac contractility, platelet aggregation, lipolysis, glycogenolysis, and smooth muscle contraction. Cyclic guanosine monophosphate (cGMP) stimulated human phosphodiesterase 2 (PDE2) is expressed mainly in brain and heart tissues. PDE2A is involved in the regulation of blood pressure and fluid homeostasis by the atrial natriuretic peptide (ANP), making PDE2-type enzymes important targets for drug discovery. The design of more potent and selective inhibitors of PDE2A for the treatment of heart disease would be greatly aided by the identification of active site residues in PDE2A that determine substrate and inhibitor selectivity. The identification of active site residues through traditional mutational studies involves the time-consuming and tedious purification of a large number of mutant proteins from overexpressing cells. Here we report an alternative approach to rapidly produce active site mutants of human PDE2A and identify their enzymatic properties using a wheat germ in vitro translation (IVT, also known as cell-free translation) system. We also present the crystal structure of the catalytic domain of human PDE2A determined at 1.7 A resolution, which provided a framework for the rational design of active site mutants. Using a rapid IVT approach for expression of human PDE2A mutants, we identified the roles of active site residues Asp811, Gln812, Ile826, and Tyr827 in inhibitor and substrate selectivity for PDE2A.


Assuntos
3',5'-AMP Cíclico Fosfodiesterases/antagonistas & inibidores , 3',5'-AMP Cíclico Fosfodiesterases/química , Inibidores de Fosfodiesterase/química , Biossíntese de Proteínas , Triticum/química , Triticum/genética , 3',5'-AMP Cíclico Fosfodiesterases/genética , 3',5'-GMP Cíclico Fosfodiesterases/química , Adenina/análogos & derivados , Adenina/química , Sequência de Aminoácidos , Sítios de Ligação/genética , Domínio Catalítico/genética , Sistema Livre de Células , Cristalografia por Raios X , Nucleotídeo Cíclico Fosfodiesterase do Tipo 2 , Nucleotídeo Cíclico Fosfodiesterase do Tipo 3 , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4 , Nucleotídeo Cíclico Fosfodiesterase do Tipo 5 , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Ligação Proteica/genética , Rolipram/química , Alinhamento de Sequência , Especificidade por Substrato/genética
12.
Protein Sci ; 14(1): 37-44, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15576558

RESUMO

Carbamoyl phosphate synthetase synchronizes the utilization of two ATP molecules at duplicated ATP-grasp folds to catalyze carbamoyl phosphate formation. To define the dedicated functional role played by each of the two ATP sites, we have carried out pulse/labeling studies using the synthetases from Aquifex aeolicus and Methanococcus jannaschii, hyperthermophilic organisms that encode the two ATP-grasp folds on separate subunits. These studies allowed us to differentially label each active site with [gamma-(32)P]ATP and determine the fate of the labeled gamma-phosphate in the synthetase reaction. Our results provide the first direct demonstration that enzyme-catalyzed transfer of phosphate from ATP to carbamate occurs on the more C-terminal of the two ATP-grasp folds. These findings rule out one mechanism proposed for carbamoyl phosphate synthetase, where one ATP acts as a molecular switch, and provide additional support for a sequential reaction mechanism where the gamma-phosphate groups of both ATP molecules are transferred to reactants. CP synthesis by subunit C in our single turnover pulse/chase assays did not require subunit N, but subunit N was required for detectable CP synthesis in the traditional continuous assay. These findings suggest that cross-talk between domain N and C is required for product release from subunit C.


Assuntos
Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/química , Carbamoil-Fosfato/síntese química , Trifosfato de Adenosina/química , Bactérias/enzimologia , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/isolamento & purificação , Catálise , Ativação Enzimática , Mathanococcus/enzimologia , Dobramento de Proteína , Estrutura Terciária de Proteína
13.
Protein Sci ; 13(2): 466-75, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-14718657

RESUMO

Synthesis of carbamoyl phosphate by carbamoyl phosphate synthetase (CPS) requires the coordinated utilization of two molecules of ATP per reaction cycle on duplicated nucleotide-binding sites (N and C). To clarify the contributions of sites N and C to the overall reaction, we carried out site-directed mutagenesis aimed at changing the substrate specificity of either of the two sites from ATP to GTP. Mutant design was based in part on an analysis of the nucleotide-binding sites of succinyl-CoA synthetases, which share membership in the ATP-grasp family with CPS and occur as GTP- and ATP-specific isoforms. We constructed and analyzed Escherichia coli CPS single mutations A144Q, D207A, D207N, S209A, I211S, P690Q, D753A, D753N, and F755A, as well as combinations thereof. All of the mutants retained ATP specificity, arguing for a lack of plasticity of the ATP sites of CPS with respect to nucleotide recognition. GTP-specific ATP-grasp proteins appear to accommodate this substrate by a displacement of the base relative to the ATP-bound state, an interaction that is precluded by the architecture of the potassium-binding loop in CPS. Analysis of the ATP-dependent kinetic parameters revealed that mutation of several residues conserved in ATP-grasp proteins and CPSs had surprisingly small effects, whereas constructs containing either A144Q or P690Q exerted the strongest effects on ATP utilization. We propose that these mutations affect proper movement of the lids covering the active sites of CPS, and interfere with access of substrate.


Assuntos
Trifosfato de Adenosina/metabolismo , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/química , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/metabolismo , Proteínas de Transporte/química , Guanosina Trifosfato/metabolismo , Adenina/metabolismo , Animais , Sítios de Ligação , Carbamoil Fosfato Sintase (Glutamina-Hidrolizante)/genética , Columbidae , Sequência Conservada/genética , Escherichia coli/enzimologia , Escherichia coli/genética , Cinética , Proteínas de Membrana , Modelos Moleculares , Mutação/genética , Conformação Proteica , Especificidade por Substrato , Suínos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA