Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 84
Filtrar
1.
RSC Med Chem ; 15(4): 1274-1282, 2024 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-38665825

RESUMEN

We recently reported novel purine-based CK2α inhibitors using the solvent ordering-based method as virtual screening. Among these, the X-ray crystal structure of a complex with CK2α was determined. The results showed that the crystalline water molecules observed in many previously reported complex structures of CK2α and its inhibitors had been eliminated. We then proposed a structure-based drug design. Since the removal of water molecules would be detrimental to inhibitor binding, new groups of compounds were designed by changing the position of the carboxy group located at the point where a water molecule would be present so as not to eliminate it. Compounds with (E)-2-carboxyethenyl and 3-carboxyphenyl substituted at the 2-position on the purine scaffold showed much higher inhibitory potency than 4-carboxyphenyl derivatives. Furthermore, in the presence of a 4-fluorophenyl group at the 9-position on the purine scaffold, the inhibitory activity of the 3-carboxyphenyl derivative against CK2α was 0.18 µM, a 167-fold improvement compared to the 4-carboxyphenyl derivative. The strategy of leaving crystalline water can significantly increase inhibitory activity.

2.
Biochem Biophys Res Commun ; 704: 149707, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38428305

RESUMEN

Mitogen-activated protein kinases (MAPKs), including extracellular signal-regulated kinase 2 (ERK2) and p38α MAP kinase (p38α MAPK), regulate various cellular responses. ERK2 is a drug target for treating many diseases, such as cancer, whereas p38α has attracted much attention as a promising drug target for treating inflammatory disorders. ERK2 is a critical off-target for p38α MAPK and vice versa. In this study, an allosteric ERK2 inhibitor with a benzothiazole moiety (compound 1) displayed comparable inhibitory activity against p38α MAPK. Crystal structures of these MAPKs showed that compound 1 bound to the allosteric site of ERK2 and p38α MAPK in distinct manners. Compound 1 formed a covalent bond with Cys162 of p38α MAPK, whereas this covalent bond was absent in the ERK2 complex even though the corresponding cysteine is conserved in ERK2. Structural dissection combined with computational simulations indicated that an amino acid difference in the allosteric site is responsible for the distinct binding modes of compound 1 with ERK2 and p38α MAPK. These structural insights underline the feasibility of developing highly selective and potent ERK2 and p38α MAPK inhibitors.


Asunto(s)
Proteína Quinasa 14 Activada por Mitógenos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Benzotiazoles/farmacología
3.
Bioorg Med Chem Lett ; 93: 129431, 2023 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-37544371

RESUMEN

Extracellular signal-regulated kinase 2 (ERK2), a mitogen-activated protein kinase (MAPK), plays an essential role in physiological cellular processes and is a drug target for treating cancers and type 2 diabetes. A previous in silico screening study focusing on an allosteric site that plays a crucial role in substrate anchoring conferred an ERK2 inhibitor (compound 1). In this report, compound 1 was found to show high selectivity toward ERK2 compared with the nearest off-target p38α MAPK, and the crystal structure revealed that compound 1 binds to the allosteric site of ERK2. Fragment molecular orbital calculations based upon this crystal structure provided the structural basis to improve potency of compound 1 derivatives. Further computational studies uncovered that the low entropic cost of binding conferred the high selectivity of compound 1 toward ERK2 over p38α MAPK. These findings demonstrate the feasibility of developing potent and selective ERK2 inhibitors.


Asunto(s)
Diabetes Mellitus Tipo 2 , Proteína Quinasa 1 Activada por Mitógenos , Humanos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Sitio Alostérico
4.
Chem Pharm Bull (Tokyo) ; 71(7): 558-565, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37394605

RESUMEN

Protein kinase CK2 (CK2) is involved in the suppression of gene expression, protein synthesis, cell proliferation, and apoptosis, thus making it a target protein for the development of therapeutics toward cancer, nephritis, and coronavirus disease 2019. Using the solvent dipole ordering-based method for virtual screening, we identified and designed new candidate CK2α inhibitors containing purine scaffolds. Virtual docking experiments supported by experimental structure-activity relationship studies identified the importance of the 4-carboxyphenyl group at the 2-position, a carboxamide group at the 6-position, and an electron-rich phenyl group at the 9-position of the purine scaffold. Docking studies based on the crystal structures of CK2α and inhibitor (PDBID: 5B0X) successfully predicted the binding mode of 4-(6-carbamoyl-8-oxo-9-phenyl-8,9-dihydro-7H-purin-2-yl) benzoic acid (11), and the results were used to design stronger small molecule targets for CK2α inhibition. Interaction energy analysis suggested that 11 bound around the hinge region without the water molecule (W1) near Trp176 and Glu81 that is frequently reported in crystal structures of CK2α inhibitor complexes. X-ray crystallographic data for 11 bound to CK2α was in very good agreement with the docking experiments, and consistent with activity. From the structure-activity relationship (SAR) studies presented here, 4-(6-Carbamoyl-9-(4-(dimethylamino)phenyl)-8-oxo-8,9-dihydro-7H-purin-2-yl) benzoic acid (12) was identified as an improved active purine-based CK2α inhibitor with an IC50 of 4.3 µM. These active compounds with an unusual binding mode are expected to inspire new CK2α inhibitors and the development of therapeutics targeting CK2 inhibition.


Asunto(s)
COVID-19 , Inhibidores de Proteínas Quinasas , Humanos , Inhibidores de Proteínas Quinasas/farmacología , Inhibidores de Proteínas Quinasas/química , Quinasa de la Caseína II/genética , Quinasa de la Caseína II/metabolismo , Relación Estructura-Actividad , Ácido Benzoico , Purinas
5.
Biochem Biophys Res Commun ; 630: 30-35, 2022 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-36130444

RESUMEN

Casein kinase 2 (CK2) is a vital protein kinase that consists of two catalytic subunits (CK2α1 and/or CK2α2) and two regulatory subunits (CK2ß). CK2α1 is a drug target for nephritis and cancers, while CK2α2 is a serious off-target because its inhibition causes testicular toxicity. High similarity between the isozymes CK2α1 and CK2α2 make it difficult to design CK2α1-specific inhibitors. Herein, the crystal structures of CK2α1 and CK2α2 complexed with a 3-amino-pyrazole inhibitor revealed the remarkable differences in the protein-inhibitor interaction modes. This inhibitor bound to the ATP binding sites of both isozymes in apparently distinct orientations. In addition, another molecule of this inhibitor bound to CK2α1, but not to CK2α2, at the CK2ß protein-protein interface. Binding energy calculations and biochemical experiments suggested that this inhibitor possesses the conventional ATP-competitive characteristics with moderate allosteric function in a molecular glue mechanism. These results will assist the potential design of potent and selective CK2α1 inhibitors.


Asunto(s)
Quinasa de la Caseína II , Isoenzimas , Pirazoles/farmacología , Adenosina Trifosfato , Quinasa de la Caseína II/metabolismo , Isoenzimas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología
6.
Biochem Biophys Res Commun ; 593: 73-78, 2022 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-35063772

RESUMEN

Extracellular signal-regulated kinase 2 (ERK2) controls vital physiological processes involving proliferation and differentiation and is a drug target molecule for many diseases such as cancers. In silico screening focusing on an allosteric site that plays a crucial role in substrate anchoring conferred an ERK2 inhibitor (compound 1). However, a competitive binding assay indicated that compound 1 did not bind to the allosteric site. Here, the crystal structure of ERK2 in complex with compound 1 revealed a novel binding site. This finding demonstrates the feasibility of developing new types of ERK2 inhibitors.


Asunto(s)
Adenosina Trifosfato/metabolismo , Proteína Quinasa 1 Activada por Mitógenos/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Sitio Alostérico , Sitios de Unión , Unión Competitiva , Cristalografía por Rayos X , Humanos , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Fosforilación , Conformación Proteica
7.
Bioorg Med Chem Lett ; 43: 128056, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33892104

RESUMEN

p38α mitogen activated protein kinase (MAPK) plays important roles in multiple cellular functions by phosphorylating a wide variety of substrates, and therefore, p38α MAPK has been considered as an important drug target. In this study, we designed peptide-based inhibitors for p38α MAPK, which can only inhibit the Smad3 phosphorylation specifically, by targeting the KIM binding site of p38α MAPK. Peptide 6 showed a significant inhibitory potency for the Smad3 phosphorylation by p38α MAPK. Peptide 6 showed no ATP dependency, and did not inhibit the phosphorylation of other substrates by p38α MAPK. The discovery of peptide 6 by targeting the KIM binding site likely provide an opportunity for the discovery of a novel class of allosteric and substrate-specific p38α MAPK inhibitors.


Asunto(s)
Péptidos/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteína smad3/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Regulación Alostérica/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Humanos , Modelos Moleculares , Estructura Molecular , Péptidos/síntesis química , Péptidos/química , Fosforilación/efectos de los fármacos , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteína smad3/metabolismo , Relación Estructura-Actividad , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
Bioorg Med Chem Lett ; 30(22): 127546, 2020 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-32931911

RESUMEN

Mitogen-activated protein kinase kinase 7 (MAP2K7) in the c-Jun N-terminal kinase signal cascade is an attractive drug target for a variety of diseases. The selectivity of MAP2K7 inhibitors against off-target kinases is a major barrier in drug development. We report a crystal structure of MAP2K7 complexed with a potent covalent inhibitor bearing an acrylamide moiety as an electrophile, which discloses a structural basis for producing selective and potent MAP2K7 inhibitors.


Asunto(s)
Acrilamida/farmacología , MAP Quinasa Quinasa 7/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Acrilamida/síntesis química , Acrilamida/química , Relación Dosis-Respuesta a Droga , Humanos , MAP Quinasa Quinasa 7/metabolismo , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Relación Estructura-Actividad
9.
Bioorg Med Chem Lett ; 30(2): 126837, 2020 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-31859160

RESUMEN

Casein kinase 2 catalytic subunit (CK2α) is classified into two subtypes CK2α1 and CK2α2. CK2α1 is a drug discovery target, whereas CK2α2 is an off-target of CK2α1 inhibitors. High amino acid sequence homology between these subtypes hampers efforts to produce ATP competitive inhibitors that are highly selective to CK2α1. Hematein was identified previously as a non-ATP-competitive inhibitor for CK2α1, whereas this compound acts as an ATP competitive CK2α2 inhibitor. Crystal structures of CK2α1 and CK2α2 in complex with hematein revealed distinct binding features that provide structural insights for producing CK2α1-selective inhibitors.


Asunto(s)
Secuencia de Aminoácidos , Quinasa de la Caseína II/antagonistas & inhibidores , Humanos , Modelos Moleculares
10.
Bioorg Med Chem Lett ; 30(1): 126775, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31699609

RESUMEN

The production of TrkA-selective inhibitors is considerably difficult because the kinase domains of TrkA and its isoforms TrkB/C have highly homologous amino acid sequences. Here we describe the structural basis for the acquisition of selectivity for a isoform-selective TrkA inhibitor, namely compound V1. The X-ray structure revealed that V1 acts as a molecular glue to stabilize the symmetrical dimer of the TrkA kinase domains. V1 binds to the ATP-binding site and simultaneously engages in the dimeric interface of TrkA. The region of the dimeric interface in TrkA is not conserved in TrkB/C; thus, dimer formation may be a novel mechanism for the production of selective TrkA inhibitors. The biochemical and biophysical assay results confirmed that V1 selectively inhibited TrkA and induced the dimer formation of TrkA, but not TrkB. The binding pocket at the TrkA dimer interface can be used for the production of new isoform-selective TrkA inhibitors.


Asunto(s)
Isoformas de Proteínas/metabolismo , Receptor trkA/metabolismo , Secuencia de Aminoácidos , Humanos , Modelos Moleculares
11.
Biochem Biophys Res Commun ; 521(1): 106-112, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31635803

RESUMEN

Mitogen-activated protein kinase kinase 4 (MAP2K4) plays a critical role in regulating the stress-activated protein kinase signaling cascade. A small angle X-ray scattering experiment, a powerful technique for analyzing a solution structure cleared from the structural artifacts due to crystal packing, provided the ensemble structures of human non-phosphorylated MAP2K4 in three states involving the apo form, the binary complex with an ATP analogue, and the ternary complex with the ATP analogue and substrate peptide. These ensemble structures provided more detailed mechanisms for regulating MAP2K4 in addition to those delineated only by the crystal structures in three states.


Asunto(s)
MAP Quinasa Quinasa 4/análisis , MAP Quinasa Quinasa 4/metabolismo , Adenosina Trifosfato/química , Adenosina Trifosfato/metabolismo , Humanos , Conformación Proteica , Dispersión del Ángulo Pequeño , Difracción de Rayos X
12.
Adv Exp Med Biol ; 1163: 45-64, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31707699

RESUMEN

This chapter focuses on protein kinases that transfer the phosphate group of ATP to the hydroxyl group of a substrate protein. Five hundred eighteen human protein kinases are classified into serine/threonine kinases and tyrosine kinases and individually or synergistically transduce physiologic stimuli into cell to promote cell proliferation or apoptosis, etc. Protein kinases are identified as drug targets because dysfunction of kinases leads to severe diseases such as cancers and autoimmune diseases. A large number of the crystal structures of the protein kinase inhibitor complex are available in Protein Data Bank and facilitated the drug discovery targeting protein kinases. The protein kinase inhibitors are classified into categories, Type-I, Type-II, Type-III, Type-IV, and Type-V, and as a separate class, covalent-type inhibitors. In any type, a protein kinase inhibitor bound to the allosteric region is advantageous in terms of selectivity compared to the traditional ATP-competitive one. In the following sections, the successful and promising examples of the partially or fully allosteric protein kinase inhibitors are illustrated in the following pages.


Asunto(s)
Diseño de Fármacos , Inhibidores de Proteínas Quinasas , Proteínas Quinasas , Adenosina Trifosfato , Descubrimiento de Drogas , Activación Enzimática/efectos de los fármacos , Humanos , Unión Proteica/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo
13.
Acta Crystallogr F Struct Biol Commun ; 75(Pt 7): 515-519, 2019 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-31282872

RESUMEN

Protein kinase CK2a1 is a serine/threonine kinase that plays a crucial role in the growth, proliferation and survival of cells and is a well known target for tumour and glomerulonephritis therapies. Here, the crystal structure of the kinase domain of CK2a1 complexed with 5-iodotubercidin (5IOD), an ATP-mimetic inhibitor, was determined at 1.78 Šresolution. The structure shows distinct structural features and, in combination with a comparison of the crystal structures of five off-target kinases complexed with 5IOD, provides valuable information for the development of highly selective inhibitors.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/química , Secuencia de Aminoácidos , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/química , Quinasa de la Caseína II/metabolismo , Cristalografía por Rayos X , Humanos , Proteínas Serina-Treonina Quinasas/metabolismo , Estructura Secundaria de Proteína , Electricidad Estática , Tubercidina/análogos & derivados , Tubercidina/química , Tubercidina/metabolismo
14.
Sci Adv ; 5(1): eaau9060, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30746467

RESUMEN

Compounds targeting the circadian clock have been identified as potential treatments for clock-related diseases, including cancer. Our cell-based phenotypic screen revealed uncharacterized clock-modulating compounds. Through affinity-based target deconvolution, we identified GO289, which strongly lengthened circadian period, as a potent and selective inhibitor of CK2. Phosphoproteomics identified multiple phosphorylation sites inhibited by GO289 on clock proteins, including PER2 S693. Furthermore, GO289 exhibited cell type-dependent inhibition of cancer cell growth that correlated with cellular clock function. The x-ray crystal structure of the CK2α-GO289 complex revealed critical interactions between GO289 and CK2-specific residues and no direct interaction of GO289 with the hinge region that is highly conserved among kinases. The discovery of GO289 provides a direct link between the circadian clock and cancer regulation and reveals unique design principles underlying kinase selectivity.


Asunto(s)
Carcinoma de Células Renales/metabolismo , Proliferación Celular/efectos de los fármacos , Relojes Circadianos/efectos de los fármacos , Ritmo Circadiano/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales/métodos , Neoplasias Renales/metabolismo , Animales , Proteínas CLOCK/metabolismo , Carcinoma de Células Renales/patología , Quinasa de la Caseína II/antagonistas & inhibidores , Línea Celular Tumoral , Cristalografía por Rayos X , Células HEK293 , Humanos , Neoplasias Renales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosforilación/efectos de los fármacos
15.
Cell Chem Biol ; 26(1): 98-108.e5, 2019 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-30449673

RESUMEN

The c-Jun NH2-terminal kinase (JNK) signaling pathway is central to the cell response to stress, inflammatory signals, and toxins. While selective inhibitors are known for JNKs and for various upstream MAP3Ks, no selective inhibitor is reported for MKK7--one of two direct MAP2Ks that activate JNK. Here, using covalent virtual screening, we identify selective MKK7 covalent inhibitors. We optimized these compounds to low-micromolar inhibitors of JNK phosphorylation in cells. The crystal structure of a lead compound bound to MKK7 demonstrated that the binding mode was correctly predicted by docking. We asserted the selectivity of our inhibitors on a proteomic level and against a panel of 76 kinases, and validated an on-target effect using knockout cell lines. Lastly, we show that the inhibitors block activation of primary mouse B cells by lipopolysaccharide. These MKK7 tool compounds will enable better investigation of JNK signaling and may serve as starting points for therapeutics.


Asunto(s)
MAP Quinasa Quinasa 7/antagonistas & inhibidores , Simulación del Acoplamiento Molecular , Inhibidores de Proteínas Quinasas/farmacología , Células 3T3 , Animales , Supervivencia Celular/efectos de los fármacos , Células HEK293 , Humanos , MAP Quinasa Quinasa 7/genética , MAP Quinasa Quinasa 7/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de Proteínas Quinasas/química
16.
J Mol Biol ; 430(24): 5094-5104, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30359582

RESUMEN

Casein kinase 2 (CK2) has broad phosphorylation activity against various regulatory proteins, which are important survival factors in eukaryotic cells. To clarify the hydration structure and catalytic mechanism of CK2, we determined the crystal structure of the alpha subunit of human CK2 containing hydrogen and deuterium atoms using joint neutron (1.9 Šresolution) and X-ray (1.1 Šresolution) crystallography. The analysis revealed the structure of conserved water molecules at the active site and a long potential hydrogen bonding network originating from the catalytic Asp156 that is well known to enhance the nucleophilicity of the substrate OH group to the γ-phospho group of ATP by proton elimination. His148 and Asp214 conserved in the protein kinase family are located in the middle of the network. The water molecule forming a hydrogen bond with Asp214 appears to be deformed. In addition, mutational analysis of His148 in CK2 showed significant reductions by 40%-75% in the catalytic efficiency with similar affinity for ATP. Likewise, remarkable reductions to less than 5% were shown by corresponding mutations on His131 in death-associated protein kinase 1, which belongs to a group different from that of CK2. These findings shed new light on the catalytic mechanism of protein kinases in which the hydrogen bond network through the C-terminal domain may assist the general base catalyst to extract a proton with a link to the bulk solvent via intermediates of a pair of residues.


Asunto(s)
Mutación , Agua/química , Sitios de Unión , Quinasa de la Caseína II/química , Quinasa de la Caseína II/genética , Dominio Catalítico , Cristalografía por Rayos X , Deuterio , Humanos , Enlace de Hidrógeno , Modelos Moleculares , Dominios Proteicos
17.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 5): 288-293, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29717996

RESUMEN

The catalytic subunits of protein kinase CK2 are classified into two subtypes: CK2α1 and CK2α2. CK2α1 is an attractive drug-discovery target for various diseases such as cancers and nephritis. CK2α2 is defined as an off-target of CK2α1 and is a potential target in the development of male contraceptive drugs. High-resolution crystal structures of both isozymes are likely to provide crucial clues for the design of selective inhibitors of CK2α1 and/or CK2α2. To date, several crystal structures of CK2α1 have been solved at high resolutions of beyond 1.5 Å. However, crystal structures of CK2α2 have barely achieved a low resolution of around 3 Šbecause of the formation of needle-shaped crystals. In this study, new crystal forms were exploited and one provided a crystal structure of CK2α2 at 1.89 Šresolution. This result, together with the structure of CK2α1, will assist in the development of highly selective inhibitors for both isozymes.


Asunto(s)
Quinasa de la Caseína II/química , Quinasa de la Caseína II/metabolismo , Cristalización/métodos , Humanos , Isoenzimas/química , Isoenzimas/metabolismo , Estructura Secundaria de Proteína , Trometamina/metabolismo
18.
Clin Cancer Res ; 24(10): 2357-2369, 2018 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-29463555

RESUMEN

Purpose: Rearrangement of the neurotrophic tropomyosin receptor kinase 1 (NTRK1) gene, which encodes tyrosine receptor kinase A (TRK-A), occurs in various cancers, including colon cancer. Although entrectinib is effective in the treatment of central nervous system (CNS) metastases that express NTRK1 fusion proteins, acquired resistance inevitably results in recurrence. The CNS is a sanctuary for targeted drugs; however, the mechanism by which CNS metastases become entrectinib-resistant remains elusive and must be clarified to develop better therapeutics.Experimental Design: The entrectinib-resistant cell line KM12SM-ER was developed by continuous treatment with entrectinib in the brain metastasis-mimicking model inoculated with the entrectinib-sensitive human colon cancer cell line KM12SM, which harbors the TPM3-NTRK1 gene fusion. The mechanism of entrectinib resistance in KM12SM-ER cells was examined by next-generation sequencing. Compounds that overcame entrectinib resistance were screened from a library of 122 kinase inhibitors.Results: KM12SM-ER cells, which showed moderate resistance to entrectinib in vitro, had acquired the G667C mutation in NTRK1 The kinase inhibitor foretinib inhibited TRK-A phosphorylation and the viability of KM12SM-ER cells bearing the NTRK1-G667C mutation in vitro Moreover, foretinib markedly inhibited the progression of entrectinib-refractory KM12SM-ER-derived liver metastases and brain tumors in animal models, predominantly through inhibition of TRK-A phosphorylation.Conclusions: These results suggest that foretinib may be effective in overcoming entrectinib resistance associated with the NTRK1-G667C mutation in NTRK1 fusion-positive tumors in various organs, including the brain, and provide a rationale for clinical trials of foretinib in cancer patients with entrectinib-resistant tumors harboring the NTRK1-G667C mutation, including patients with brain metastases. Clin Cancer Res; 24(10); 2357-69. ©2018 AACR.


Asunto(s)
Anilidas/farmacología , Benzamidas/farmacología , Neoplasias Encefálicas/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Indazoles/farmacología , Mutación , Proteínas de Fusión Oncogénica/genética , Quinolinas/farmacología , Receptor trkA/genética , Sustitución de Aminoácidos , Anilidas/química , Animales , Benzamidas/química , Neoplasias Encefálicas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Xenoinjertos , Humanos , Indazoles/química , Ratones , Modelos Moleculares , Quinolinas/química , Receptor trkA/química , Relación Estructura-Actividad
19.
Biochem Biophys Res Commun ; 493(1): 313-317, 2017 11 04.
Artículo en Inglés | MEDLINE | ID: mdl-28890347

RESUMEN

Mitogen-activated protein kinase kinase 7 (MAP2K7) regulates stress and inflammatory responses, and is an attractive drug discovery target for several diseases including arthritis and cardiac hypertrophy. Intracellular proteins such as MAP2K7 are prone to aggregation due to cysteine-driven oxidation in in vitro experiments. MAP2K7 instability due to the four free cysteine residues on the molecular surface abrogated the crystal growth and led to a low-resolution structure with large residual errors. To acquire a higher resolution structure for promoting rational drug discovery, we explored stable mutants of MAP2K7 by replacing the surface cysteine residues, Cys147, Cys218, Cys276 and Cys296. Single-site mutations, except for Cys147, maintained the specific activity and increased the protein yield, while all the multi-site mutations massively reduced the activity. The C218S mutation drastically augmented the protein production and crystallographic resolution. Furthermore, the C218S crystals grown under microgravity in a space environment yielded a 1.3 Å resolution structure, providing novel insights for drug discovery: the precisely assigned water molecules in the active site, the double conformations in the flexible region and the C-terminal extension bound to the N-terminal region of the adjacent molecules. The latter insight is likely to promote the production of allosteric MAP2K7 inhibitors.


Asunto(s)
MAP Quinasa Quinasa 7/química , MAP Quinasa Quinasa 7/ultraestructura , Regulación Alostérica , Sitios de Unión , Simulación por Computador , Activación Enzimática , Modelos Químicos , Modelos Moleculares , Unión Proteica , Conformación Proteica , Relación Estructura-Actividad
20.
Bioorg Med Chem Lett ; 27(5): 1233-1236, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28159414

RESUMEN

Although numerous crystal structures for protein kinases have been reported, many include only the kinase domain but not the juxtamembrane (JM) region, a critical activity-controlling segment of receptor tyrosine kinases (RTKs). In this study, we determined the X-ray crystal structure of the tropomyosin receptor kinase (Trk) A selective inhibitor A1 complexed with the TrkA kinase domain and the JM region. This structure revealed that the unique inhibitor-binding pocket created by a novel JM configuration yields significant potency and high selectivity against TrkB and TrkC. Moreover, we validated the importance of the JM region for the potency of A1 using in vitro assays. The introduction of moieties that interact with the JM region will be one of the most effective strategies for producing highly selective RTK inhibitors.


Asunto(s)
Proteínas de la Membrana/química , Modelos Moleculares , Inhibidores de Proteínas Quinasas/química , Receptor trkA/antagonistas & inhibidores , Receptor trkA/química , Sitios de Unión , Bioensayo , Membrana Celular/enzimología , Cristalografía por Rayos X , Activación Enzimática/efectos de los fármacos , Enlace de Hidrógeno , Concentración 50 Inhibidora , Proteínas de la Membrana/metabolismo , Estructura Molecular , Inhibidores de Proteínas Quinasas/farmacología , Estructura Terciaria de Proteína , Receptor trkA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...