Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Bioorg Med Chem ; 29: 115865, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33285410

RESUMEN

Recent years have seen a resurgence in drug discovery efforts aimed at the identification of covalent inhibitors which has led to an explosion of literature reports in this area and most importantly new approved therapies. These reports and breakthroughs highlight the significant investments made across the industry in SAR campaigns to optimize inhibitors. The potency of covalent inhibitors is generally considered to be more accurately described by the time-independent kinetic parameter kinact/Ki rather than a by a simple IC50 since the latter is a time-dependent parameter. Enzyme substrate concentrations are an additional important factor to consider when attempting to translate parameters derived from enzymology experiments to phenotypic behavior in a physiologically relevant cell-based system. Theoretical and experimental investigations into the relationship between IC50, time, substrate concentration and Kinact/Ki provided us with an effective approach to provide meaningful data for SAR optimization. The data we generated for our JAK3 irreversible covalent inhibitor program using IC50 values provided by enzyme assays with long incubations (>1h) coupled with physiological substrate concentration provided the medicinal chemist with optimal information in a rapid and efficient manner. We further document the wide applicability of this method by applying it to other enzymes systems where we have run covalent inhibitor programs.


Asunto(s)
Janus Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Relación Dosis-Respuesta a Droga , Humanos , Concentración 50 Inhibidora , Janus Quinasa 3/metabolismo , Estructura Molecular , Inhibidores de Proteínas Quinasas/química , Proteínas Recombinantes , Relación Estructura-Actividad
2.
Structure ; 27(11): 1625-1633.e3, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31693911

RESUMEN

E7820 and indisulam are two examples of aryl sulfonamides that recruit RBM39 to Rbx-Cul4-DDA1-DDB1-DCAF15 E3 ligase complex, leading to its ubiquitination and degradation by the proteasome. To understand their mechanism of action, we performed kinetic analysis on the recruitment of RBM39 to DCAF15 and solved a crystal structure of DDA1-DDB1-DCAF15 in complex with E7820 and the RRM2 domain of RBM39. E7820 packs in a shallow pocket on the surface of DCAF15 and the resulting modified interface binds RBM39 through the α1 helix of the RRM2 domain. Our kinetic studies revealed that aryl sulfonamide and RBM39 bind to DCAF15 in a synergistic manner. The structural and kinetic studies confirm aryl sulfonamides as molecular glues in the recruitment of RBM39 and provide a framework for future efforts to utilize DCAF15 to degrade other proteins of interest.


Asunto(s)
Indoles/química , Péptidos y Proteínas de Señalización Intracelular/química , Proteínas de Unión al ARN/química , Sulfonamidas/química , Sitios de Unión , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Cinética , Simulación del Acoplamiento Molecular , Unión Proteica , Proteínas de Unión al ARN/metabolismo
3.
J Med Chem ; 62(15): 6876-6893, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31282155

RESUMEN

The hypoxia-inducible factor 2α (HIF-2α) is a key oncogenic driver in clear cell renal cell carcinoma (ccRCC). Our first HIF-2α inhibitor PT2385 demonstrated promising proof of concept clinical activity in heavily pretreated advanced ccRCC patients. However, PT2385 was restricted by variable and dose-limited pharmacokinetics resulting from extensive metabolism of PT2385 to its glucuronide metabolite. Herein we describe the discovery of second-generation HIF-2α inhibitor PT2977 with increased potency and improved pharmacokinetic profile achieved by reduction of phase 2 metabolism. Structural modification by changing the geminal difluoro group in PT2385 to a vicinal difluoro group resulted in enhanced potency, decreased lipophilicity, and significantly improved pharmacokinetic properties. In a phase 1 dose-escalation study, the clinical pharmacokinetics for PT2977 supports the hypothesis that attenuating the rate of glucuronidation would improve exposure and reduce variability in patients. Early evidence of clinical activity shows promise for PT2977 in the treatment of ccRCC.


Asunto(s)
Antineoplásicos/uso terapéutico , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Carcinoma de Células Renales/tratamiento farmacológico , Indanos/uso terapéutico , Neoplasias Renales/tratamiento farmacológico , Sulfonas/uso terapéutico , Animales , Antineoplásicos/síntesis química , Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Carcinoma de Células Renales/metabolismo , Perros , Relación Dosis-Respuesta a Droga , Femenino , Haplorrinos , Humanos , Indanos/síntesis química , Indanos/farmacología , Neoplasias Renales/metabolismo , Ratones , Ratones SCID , Ratas , Sulfonas/síntesis química , Sulfonas/farmacología , Resultado del Tratamiento , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
4.
J Med Chem ; 61(23): 10665-10699, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30423248

RESUMEN

Ongoing interest in the discovery of selective JAK3 inhibitors led us to design novel covalent inhibitors that engage the JAK3 residue Cys909 by cyanamide, a structurally and mechanistically differentiated electrophile from other cysteine reacting groups previously incorporated in JAK3 covalent inhibitors. Through crystallography, kinetic, and computational studies, interaction of cyanamide 12 with Cys909 was optimized leading to potent and selective JAK3 inhibitors as exemplified by 32. In relevant cell-based assays and in agreement with previous results from this group, 32 demonstrated that selective inhibition of JAK3 is sufficient to drive JAK1/JAK3-mediated cellular responses. The contribution from extrahepatic processes to the clearance of cyanamide-based covalent inhibitors was also characterized using metabolic and pharmacokinetic data for 12. This work also gave key insights into a productive approach to decrease glutathione/glutathione S-transferase-mediated clearance, a challenge typically encountered during the discovery of covalent kinase inhibitors.


Asunto(s)
Cianamida/química , Cianamida/farmacología , Janus Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Animales , Cianamida/farmacocinética , Evaluación Preclínica de Medicamentos , Humanos , Concentración 50 Inhibidora , Janus Quinasa 3/química , Masculino , Modelos Moleculares , Conformación Proteica , Inhibidores de Proteínas Quinasas/farmacocinética , Ratas , Distribución Tisular
5.
J Med Chem ; 61(21): 9691-9721, 2018 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-30289716

RESUMEN

HIF-2α, a member of the HIF family of transcription factors, is a key oncogenic driver in cancers such as clear cell renal cell carcinoma (ccRCC). A signature feature of these cancers is the overaccumulation of HIF-2α protein, often by inactivation of the E3 ligase VHL (von Hippel-Lindau). Herein we disclose our structure based drug design (SBDD) approach that culminated in the identification of PT2385, the first HIF-2α antagonist to enter clinical trials. Highlights include the use of a putative n → π*Ar interaction to guide early analog design, the conformational restriction of an essential hydroxyl moiety, and the remarkable impact of fluorination near the hydroxyl group. Evaluation of select compounds from two structural classes in a sequence of PK/PD, efficacy, PK, and metabolite profiling identified 10i (PT2385, luciferase EC50 = 27 nM) as the clinical candidate. Finally, a retrospective crystallographic analysis describes the structural perturbations necessary for efficient antagonism.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Carcinoma de Células Renales/patología , Diseño de Fármacos , Indanos/química , Indanos/farmacología , Neoplasias Renales/patología , Sulfonas/química , Sulfonas/farmacología , Animales , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/química , Línea Celular Tumoral , Perros , Indanos/farmacocinética , Ratones , Modelos Moleculares , Conformación Proteica , Ratas , Relación Estructura-Actividad , Sulfonas/farmacocinética , Distribución Tisular
6.
J Med Chem ; 60(5): 1971-1993, 2017 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-28139931

RESUMEN

Significant work has been dedicated to the discovery of JAK kinase inhibitors resulting in several compounds entering clinical development and two FDA approved NMEs. However, despite significant effort during the past 2 decades, identification of highly selective JAK3 inhibitors has eluded the scientific community. A significant effort within our research organization has resulted in the identification of the first orally active JAK3 specific inhibitor, which achieves JAK isoform specificity through covalent interaction with a unique JAK3 residue Cys-909. The relatively rapid resynthesis rate of the JAK3 enzyme presented a unique challenge in the design of covalent inhibitors with appropriate pharmacodynamics properties coupled with limited unwanted off-target reactivity. This effort resulted in the identification of 11 (PF-06651600), a potent and low clearance compound with demonstrated in vivo efficacy. The favorable efficacy and safety profile of this JAK3-specific inhibitor 11 led to its evaluation in several human clinical studies.


Asunto(s)
Janus Quinasa 3/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/química , Pirimidinas/química , Pirroles/química , Transducción de Señal/efectos de los fármacos , Administración Oral , Diseño de Fármacos , Humanos , Janus Quinasa 3/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/administración & dosificación , Pirimidinas/farmacología , Pirroles/administración & dosificación , Pirroles/farmacología
7.
Cancer Res ; 76(18): 5491-500, 2016 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-27635045

RESUMEN

More than 90% of clear cell renal cell carcinomas (ccRCC) exhibit inactivation of the von Hippel-Lindau (pVHL) tumor suppressor, establishing it as the major underlying cause of this malignancy. pVHL inactivation results in stabilization of the hypoxia-inducible transcription factors, HIF1α and HIF2α, leading to expression of a genetic program essential for the initiation and progression of ccRCC. Herein, we describe the potent, selective, and orally active small-molecule inhibitor PT2385 as a specific antagonist of HIF2α that allosterically blocks its dimerization with the HIF1α/2α transcriptional dimerization partner ARNT/HIF1ß. PT2385 inhibited the expression of HIF2α-dependent genes, including VEGF-A, PAI-1, and cyclin D1 in ccRCC cell lines and tumor xenografts. Treatment of tumor-bearing mice with PT2385 caused dramatic tumor regressions, validating HIF2α as a pivotal oncogenic driver in ccRCC. Notably, unlike other anticancer agents that inhibit VEGF receptor signaling, PT2385 exhibited no adverse effect on cardiovascular performance. Thus, PT2385 represents a novel class of therapeutics for the treatment of RCC with potent preclincal efficacy as well as improved tolerability relative to current agents that target the VEGF pathway. Cancer Res; 76(18); 5491-500. ©2016 AACR.


Asunto(s)
Antineoplásicos/farmacología , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/antagonistas & inhibidores , Carcinoma de Células Renales/patología , Neoplasias Renales/patología , Animales , Antineoplásicos/química , Calorimetría , Línea Celular Tumoral , Cristalografía por Rayos X , Humanos , Inmunohistoquímica , Inmunoprecipitación , Ratones , Ratones SCID , Reacción en Cadena de la Polimerasa , Ensayos Antitumor por Modelo de Xenoinjerto
8.
ACS Chem Biol ; 9(7): 1552-8, 2014 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-24814050

RESUMEN

Kinases constitute an important class of therapeutic targets being explored both by academia and the pharmaceutical industry. The major focus of this effort was directed toward the identification of ATP competitive inhibitors. Although it has long been recognized that the intracellular concentration of ATP is very different from the concentrations utilized in biochemical enzyme assays, little thought has been devoted to incorporating this discrepancy into our understanding of translation from enzyme inhibition to cellular function. Significant work has been dedicated to the discovery of JAK kinase inhibitors; however, a disconnect between enzyme and cellular function is prominently displayed in the literature for this class of inhibitors. Herein, we demonstrate utilizing the four JAK family members that the difference in the ATP Km of each individual kinase has a significant impact on the enzyme to cell inhibition translation. We evaluated a large number of JAK inhibitors in enzymatic assays utilizing either 1 mM ATP or Km ATP for the four isoforms as well as in primary cell assays. This data set provided the opportunity to examine individual kinase contributions to the heterodimeric kinase complexes mediating cellular signaling. In contrast to a recent study, we demonstrate that for IL-15 cytokine signaling it is sufficient to inhibit either JAK1 or JAK3 to fully inhibit downstream STAT5 phosphorylation. This additional data thus provides a critical piece of information explaining why JAK1 has incorrectly been thought to have a dominant role over JAK3. Beyond enabling a deeper understanding of JAK signaling, conducting similar analyses for other kinases by taking into account potency at high ATP rather than Km ATP may provide crucial insights into a compound's activity and selectivity in cellular contexts.


Asunto(s)
Adenosina Trifosfato/metabolismo , Quinasas Janus/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal/efectos de los fármacos , Humanos , Janus Quinasa 1/metabolismo , Janus Quinasa 3/metabolismo , Isoformas de Proteínas/metabolismo , Inhibidores de Proteínas Quinasas/química , Factor de Transcripción STAT5/metabolismo
9.
Biochem J ; 460(2): 211-22, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24593284

RESUMEN

ITK (interleukin-2-inducible T-cell kinase) is a critical component of signal transduction in T-cells and has a well-validated role in their proliferation, cytokine release and chemotaxis. ITK is an attractive target for the treatment of T-cell-mediated inflammatory diseases. In the present study we describe the discovery of kinase inhibitors that preferentially bind to an allosteric pocket of ITK. The novel ITK allosteric site was characterized by NMR, surface plasmon resonance, isothermal titration calorimetry, enzymology and X-ray crystallography. Initial screening hits bound to both the allosteric pocket and the ATP site. Successful lead optimization was achieved by improving the contribution of the allosteric component to the overall inhibition. NMR competition experiments demonstrated that the dual-site binders showed higher affinity for the allosteric site compared with the ATP site. Moreover, an optimized inhibitor displayed non-competitive inhibition with respect to ATP as shown by steady-state enzyme kinetics. The activity of the isolated kinase domain and auto-activation of the full-length enzyme were inhibited with similar potency. However, inhibition of the activated full-length enzyme was weaker, presumably because the allosteric site is altered when ITK becomes activated. An optimized lead showed exquisite kinome selectivity and is efficacious in human whole blood and proximal cell-based assays.


Asunto(s)
Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Adenosina Trifosfato/farmacología , Regulación Alostérica , Sitio Alostérico , Cristalización , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Conformación Proteica/efectos de los fármacos , Estructura Terciaria de Proteína , Resonancia por Plasmón de Superficie
10.
J Biol Chem ; 289(15): 10865-10875, 2014 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-24567333

RESUMEN

IRAK4 is a central kinase in innate immunity, but the role of its kinase activity is controversial. The mechanism of activation for IRAK4 is currently unknown, and little is known about the role of IRAK4 kinase in cytokine production, particularly in different human cell types. We show IRAK4 autophosphorylation occurs by an intermolecular reaction and that autophosphorylation is required for full catalytic activity of the kinase. Phosphorylation of any two of the residues Thr-342, Thr-345, and Ser-346 is required for full activity, and the death domain regulates the activation of IRAK4. Using antibodies against activated IRAK4, we demonstrate that IRAK4 becomes phosphorylated in human cells following stimulation by IL-1R and Toll-like receptor agonists, which can be blocked pharmacologically by a dual inhibitor of IRAK4 and IRAK1. Interestingly, in dermal fibroblasts, although complete inhibition of IRAK4 kinase activity does not inhibit IL-1-induced IL-6 production, NF-κB, or MAPK activation, there is complete ablation of these processes in IRAK4-deficient cells. In contrast, the inhibition of IRAK kinase activity in primary human monocytes reduces R848-induced IL-6 production with minimal effect on NF-κB or MAPK activation. Taken together, these studies define the mechanism of IRAK4 activation and highlight the differential role of IRAK4 kinase activity in different human cell types as well as the distinct roles IRAK4 scaffolding and kinase functions play.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Quinasas Asociadas a Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-1/metabolismo , Receptores Toll-Like/metabolismo , Secuencia de Aminoácidos , Animales , Sistema Libre de Células , Clonación Molecular , Citocinas/metabolismo , Inhibidores Enzimáticos/farmacología , Fibroblastos/metabolismo , Células HEK293 , Humanos , Inmunidad Innata , Insectos , Interleucina-6/metabolismo , Sistema de Señalización de MAP Quinasas , Datos de Secuencia Molecular , Monocitos/citología , Mutación , FN-kappa B/metabolismo , Sistemas de Lectura Abierta , Fosforilación , Unión Proteica , Conformación Proteica , Receptores de Interleucina-1/agonistas , Transducción de Señal , Piel/metabolismo , Receptores Toll-Like/agonistas
11.
J Med Chem ; 55(22): 10047-63, 2012 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-23098091

RESUMEN

We wish to report a strategy that targets interleukin-2 inducible T cell kinase (Itk) with covalent inhibitors. Thus far, covalent inhibition of Itk has not been disclosed in the literature. Structure-based drug design was utilized to achieve low nanomolar potency of the disclosed series even at high ATP concentrations. Kinetic measurements confirmed an irreversible binding mode with off-rate half-lives exceeding 24 h and moderate on-rates. The analogues are highly potent in a cellular IP1 assay as well as in a human whole-blood (hWB) assay. Despite a half-life of approximately 2 h in resting primary T cells, the covalent inhibition of Itk resulted in functional silencing of the TCR pathway for more than 24 h. This prolonged effect indicates that covalent inhibition is a viable strategy to target the inactivation of Itk.


Asunto(s)
Interleucina-2/farmacología , Inhibidores de Proteínas Quinasas/sangre , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Linfocitos T/enzimología , Diseño de Fármacos , Semivida , Humanos , Cinética , Modelos Moleculares , Estructura Molecular , Inhibidores de Proteínas Quinasas/síntesis química , Proteínas Tirosina Quinasas/metabolismo , Relación Estructura-Actividad
12.
Bioorg Chem ; 38(6): 252-9, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20709352

RESUMEN

The tautomerase superfamily consists of structurally homologous proteins that are characterized by a ß-α-ß fold and a catalytic amino-terminal proline. 4-Oxalocrotonate tautomerase (4-OT) family members have been identified and categorized into five subfamilies on the basis of multiple sequence alignments and the conservation of key catalytic and structural residues. Representative members from two subfamilies have been cloned, expressed, purified, and subjected to kinetic and structural characterization. The crystal structure of DmpI from Helicobacter pylori (HpDmpI), a 4-OT homolog in subfamily 3, has been determined to high resolution (1.8Å and 2.1Å) in two different space groups. HpDmpI is a homohexamer with an active site cavity that includes Pro-1, but lacks the equivalent of Arg-11 and Arg-39 found in 4-OT. Instead, the side chain of Lys-36 replaces that of Arg-11 in a manner similar to that observed in the trimeric macrophage migration inhibitory factor (MIF), which is the title protein of another family in the superfamily. The electrostatic surface of the active site is also quite different and suggests that HpDmpI might prefer small, monoacid substrates. A kinetic analysis of the enzyme is consistent with the structural analysis, but a biological role for the enzyme remains elusive. The crystal structure of DmpI from Archaeoglobus fulgidus (AfDmpI), a 4-OT homolog in subfamily-4, has been determined to 2.4Å resolution. AfDmpI is also a homohexamer, with a proposed active site cavity that includes Pro-1, but lacks any other residues that are readily identified as catalytic ones related to 4-OT activity. Indeed, the electrostatic potential of the active site differs significantly in that it is mostly neutral, in contrast to the usual electropositive features found in other 4-OT family members, suggesting that AfDmpI might accommodate hydrophobic substrates. A kinetic analysis has been carried out, but does not provide any clues about the type of reaction the enzyme might catalyze.


Asunto(s)
Archaeoglobus fulgidus/enzimología , Helicobacter pylori/enzimología , Isomerasas/química , Archaeoglobus fulgidus/química , Dominio Catalítico , Cristalografía por Rayos X , Helicobacter pylori/química , Isomerasas/metabolismo , Cinética , Modelos Moleculares , Multimerización de Proteína
13.
J Med Chem ; 53(16): 6122-8, 2010 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-20666458

RESUMEN

Acidic mammalian chitinase (AMCase) is a member of the glycosyl hydrolase 18 family (EC 3.2.1.14) that has been implicated in the pathophysiology of allergic airway disease such as asthma. Small molecule inhibitors of AMCase were identified using a combination of high-throughput screening, fragment screening, and virtual screening techniques and characterized by enzyme inhibition and NMR and Biacore binding experiments. X-ray structures of the inhibitors in complex with AMCase revealed that the larger more potent HTS hits, e.g. 5-(4-(2-(4-bromophenoxy)ethyl)piperazine-1-yl)-1H-1,2,4-triazol-3-amine 1, spanned from the active site pocket to a hydrophobic pocket. Smaller fragments identified by FBS occupy both these pockets independently and suggest potential strategies for linking fragments. Compound 1 is a 200 nM AMCase inhibitor which reduced AMCase enzymatic activity in the bronchoalveolar lavage fluid in allergen-challenged mice after oral dosing.


Asunto(s)
Quitinasas/antagonistas & inhibidores , Modelos Moleculares , Piperazinas/síntesis química , Triazoles/síntesis química , Alérgenos/inmunología , Animales , Líquido del Lavado Bronquioalveolar , Dominio Catalítico , Cristalografía por Rayos X , Femenino , Interacciones Hidrofóbicas e Hidrofílicas , Espectroscopía de Resonancia Magnética , Ratones , Ratones Endogámicos C57BL , Piperazinas/química , Piperazinas/farmacología , Unión Proteica , Hipersensibilidad Respiratoria/tratamiento farmacológico , Hipersensibilidad Respiratoria/enzimología , Hipersensibilidad Respiratoria/inmunología , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie , Triazoles/química , Triazoles/farmacología
14.
Biochemistry ; 48(9): 2021-32, 2009 Mar 10.
Artículo en Inglés | MEDLINE | ID: mdl-19206206

RESUMEN

Bruton's tyrosine kinase (Btk) plays a central role in signal transduction pathways regulating survival, activation, proliferation, and differentiation of B-lineage lymphoid cells. A number of cell signaling studies clearly show that Btk is activated by Lyn, a Src family kinase, through phosphorylation on activation loop tyrosine 551 (Y(551)). However, the detailed molecular mechanism regulating Btk activation remains unclear. In particular, we do not fully understand the correlation of kinase activity with Y(551) phosphorylation, and the role of the noncatalytic domains of Btk in the activation process. Insect cell expressed full-length Btk is enzymatically active, but a truncated version of Btk, composed of only the kinase catalytic domain, is largely inactive. Further characterization of both forms of Btk by mass spectrometry showed partial phosphorylation of Y(551) of the full-length enzyme and none of the truncated kinase domain. To determine whether the lack of activity of the kinase domain was due to the absence of Y(551) phosphorylation, we developed an in vitro method to generate Y(551) monophosphorylated Btk kinase domain fragment using the Src family kinase Lyn. Detailed kinetic analyses demonstrated that the in vitro phosphorylated Btk kinase domain has a similar activity as the full-length enzyme while the unphosphorylated kinase domain has a very low k(cat) and is largely inactive. A divalent magnesium metal dependence study established that Btk requires a second magnesium ion for activity. Furthermore, our analysis revealed significant differences in the second metal-binding site among the kinase domain and the full-length enzyme that likely account for the difference in their catalytic profile. Taken together, our study provides important mechanistic insights into Btk kinase activity and phosphorylation-mediated regulation.


Asunto(s)
Proteínas Tirosina Quinasas/metabolismo , Tirosina/metabolismo , Adenosina Trifosfato/metabolismo , Adenilil Imidodifosfato/farmacología , Agammaglobulinemia Tirosina Quinasa , Secuencia de Aminoácidos , Animales , Sitios de Unión , Western Blotting , Catálisis/efectos de los fármacos , Línea Celular , Cromatografía Líquida de Alta Presión , Activación Enzimática , Inhibidores Enzimáticos/farmacología , Humanos , Cinética , Fragmentos de Péptidos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Tirosina Quinasas/química , Proteínas Tirosina Quinasas/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espectrometría de Masa por Ionización de Electrospray , Spodoptera , Especificidad por Sustrato
15.
Protein Sci ; 18(3): 569-78, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19241384

RESUMEN

Acidic mammalian chitinase (AMCase) is a mammalian chitinase that has been implicated in allergic asthma. One of only two active mammalian chinases, AMCase, is distinguished from other chitinases by several unique features. Here, we present the novel structure of the AMCase catalytic domain, both in the apo form and in complex with the inhibitor methylallosamidin, determined to high resolution by X-ray crystallography. These results provide a structural basis for understanding some of the unique characteristics of this enzyme, including the low pH optimum and the preference for the beta-anomer of the substrate. A triad of polar residues in the second-shell is found to modulate the highly conserved chitinase active site. As a novel target for asthma therapy, structural details of AMCase activity will help guide the future design of specific and potent AMCase inhibitors.


Asunto(s)
Quitinasas/química , Quitinasas/metabolismo , Concentración de Iones de Hidrógeno , Acetilglucosamina/análogos & derivados , Acetilglucosamina/metabolismo , Secuencia de Aminoácidos , Animales , Asma/metabolismo , Células CHO , Dominio Catalítico/fisiología , Quitinasas/genética , Cricetinae , Cricetulus , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Especificidad por Sustrato/fisiología , Trisacáridos/metabolismo
16.
Healthc Financ Manage ; 62(9): 128-30, 132, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18782990

RESUMEN

There are several things to consider in selling written-off A/R to a third party: Know how the purchase is being financed. Ensure the hospital has the ability to review collection policies that will be used. Arrive at a price via a sophisticated analysis of the debt, not by an arbitrary number. Insist on receiving detailed reports on collection success on an ongoing basis. Develop some type of profit-sharing arrangement.


Asunto(s)
Contabilidad de Pagos y Cobros , Economía Hospitalaria/organización & administración , Credito y Cobranza a Pacientes/organización & administración , Eficiencia Organizacional/economía , Humanos
17.
J Med Chem ; 51(19): 5958-63, 2008 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-18783200

RESUMEN

The protein kinase C (PKC) family of serine/threonine kinases is implicated in a wide variety of cellular processes. The PKC theta (PKCtheta) isoform is involved in TCR signal transduction and T cell activation and regulates T cell mediated diseases, including lung inflammation and airway hyperresponsiveness. Thus inhibition of PKCtheta enzyme activity by a small molecule represents an attractive strategy for the treatment of asthma. A PKCtheta high-throughput screening (HTS) campaign led to the identification of 4-(3-bromophenylamino)-5-(3,4-dimethoxyphenyl)-3-pyridinecarbonitrile 4a, a low microM ATP competitive PKCtheta inhibitor. Structure based hit-to-lead optimization led to the identification of 5-(3,4-dimethoxyphenyl)-4-(1H-indol-5-ylamino)-3-pyridinecarbonitrile 4p, a 70 nM PKCtheta inhibitor. Compound 4p was selective for inhibition of novel PKC isoforms over a panel of 21 serine/threonine, tyrosine, and phosphoinositol kinases, in addition to the conventional and atypical PKCs, PKCbeta, and PKCzeta, respectively. Compound 4p also inhibited IL-2 production in antiCD3/anti-CD28 activated T cells enriched from splenocytes.


Asunto(s)
Indoles/farmacología , Isoenzimas/antagonistas & inhibidores , Nitrilos/farmacología , Proteína Quinasa C/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Animales , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Evaluación Preclínica de Medicamentos , Femenino , Indoles/síntesis química , Indoles/química , Interleucina-2/antagonistas & inhibidores , Interleucina-2/biosíntesis , Isoenzimas/deficiencia , Isoenzimas/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Moleculares , Estructura Molecular , Nitrilos/síntesis química , Nitrilos/química , Proteína Quinasa C/deficiencia , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C-theta , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Piridinas/síntesis química , Piridinas/química , Bazo/citología , Bazo/efectos de los fármacos , Bazo/inmunología , Estereoisomerismo , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos , Linfocitos T/inmunología
18.
Biochemistry ; 46(42): 11919-29, 2007 Oct 23.
Artículo en Inglés | MEDLINE | ID: mdl-17902707

RESUMEN

YwhB, a 4-oxalocrotonate tautomerase (4-OT) homologue in Bacillus subtilis, has no known biological role, and the gene has no apparent genomic context. The kinetic and stereochemical properties of YwhB have been examined using available enol and dienol compounds. The kinetic analysis shows that YwhB has a relatively nonspecific 1,3- and 1,5-keto-enol tautomerase activity, with the former activity prevailing. Replacement of Pro-1 or Arg-11 with an alanine significantly reduces or abolishes these activities, implicating both residues as critical ones for the activities. In D2O, ketonization of two monoacid substrates (2-hydroxy-2,4-pentadienoate and phenylenolpyruvate) produces a mixture of stereoisomers {2-keto-3-[2H]-4-pentenoate and 3-[2H]-phenylpyruvate}, where the (3R)-isomers predominate. Ketonization of 2-hydroxy-2,4-hexadienedioate, a diacid, in D2O affords mostly the opposite enantiomer, (3S)-2-oxo-[3-2H]-4-hexenedioate. The mono- and diacids apparently bind in different orientations in the active site of YwhB, but the highly stereoselective nature of the YwhB reaction using a diacid suggests that the biological substrate for YwhB may be a diacid. Moreover, of the three dienols examined, 1,3- and 1,5-keto-enol tautomerization reactions are only observed for 2-hydroxy-2,4-hexadienedioate, indicating that the C-3 and C-5 positions are accessible for protonation in this compound. Incubation of 4-OT with 2-hydroxy-2,4-hexadienedioate in D2O results in a racemic mixture of 2-oxo-[3-2H]-4-hexenedioate, suggesting that 4-OT may not catalyze a 1,3-keto-enol tautomerization reaction using this dienol. It has previously been shown that 4-OT catalyzes the near stereospecific conversion of 2-hydroxy-2,4-hexadienedioate to (5S)-[5-2H]-2-oxo-3-hexenedioate in D2O. Taken together, these observations suggest that 4-OT might function as a 1,5-keto-enol tautomerase using 2-hydroxy-2,4-hexadienedioate.


Asunto(s)
Alcanos/química , Alquinos/química , Bacillus subtilis/enzimología , Isomerasas/metabolismo , Alanina/metabolismo , Alcanos/farmacología , Alquinos/farmacología , Sustitución de Aminoácidos , Sitios de Unión , Tampones (Química) , Catálisis , Escherichia coli/genética , Concentración de Iones de Hidrógeno , Isomerasas/genética , Cinética , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Fosfatos/química , Espectrometría de Masa por Ionización de Electrospray , Estereoisomerismo , Especificidad por Sustrato , Temperatura
19.
Bioorg Chem ; 34(4): 183-99, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16780921

RESUMEN

Macrophage migration inhibitory factor (MIF) is an important immunoregulatory protein that has been implicated in several inflammatory diseases. MIF also has a phenylpyruvate tautomerase (PPT) activity, the role of which remains elusive in these biological activities. The acetylene compound, 2-oxo-4-phenyl-3-butynoate (2-OPB), has been synthesized and tested as a potential irreversible inhibitor of its enzymatic activity. Incubation of the compound with MIF results in the rapid and irreversible loss of the PPT activity. Mass spectral analysis established that the amino-terminal proline, previously implicated as a catalytic base in the PPT-catalyzed reaction, is the site of covalent modification. Inactivation of the PPT activity likely occurs by a Michael addition of Pro-1 to C-4 of the inhibitor. Attempts to crystallize the inactivated complex to confirm the structure of the adduct on the covalently modified Pro-1 by X-ray crystallography were not successful. Nor was it possible to unambiguously interpret electron density observed in the active sites of the native crystals soaked with the inhibitor. This may be due to crystal packing in that the side chain of Glu-16 from an adjacent trimer occupies one active site. However, this crystal contact may be partially responsible for the high-resolution quality of these MIF crystals. Nonetheless, because MIF is a member of the tautomerase superfamily, a group of structurally homologous proteins that share a beta-alpha-beta structural motif and a catalytic Pro-1, 2-OPB may find general use as a probe of tautomerase superfamily members that function as PPTs.


Asunto(s)
Alquinos/farmacología , Butiratos/farmacología , Oxidorreductasas Intramoleculares/antagonistas & inhibidores , Factores Inhibidores de la Migración de Macrófagos/farmacología , Secuencia de Bases , Cristalografía por Rayos X , Cartilla de ADN , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
20.
Biochemistry ; 45(14): 4444-54, 2006 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-16584180

RESUMEN

Human acidic mammalian chitinase (AMCase), a member of the family 18 glycosyl hydrolases, is one of the important proteins involved in Th2-mediated inflammation and has been implicated in asthma and allergic diseases. Inhibition of AMCase results in decreased airway inflammation and airway hyper-responsiveness in a mouse asthma model, suggesting that the AMCase activity is a part of the mechanism of Th2 cytokine-driven inflammatory response in asthma. In this paper, we report the first detailed kinetic characterization of recombinant human AMCase. In contrast with mouse AMCase that has been reported to have a major pH optimum at 2 and a secondary pH optimum around 3-6, human AMCase has only one pH optimum for k(cat)/K(m) between pH 4 and 5. Steady state kinetics shows that human AMCase has "low" intrinsic transglycosidase activity, which leads to the observation of apparent substrate inhibition. This slow transglycosylation may provide a mechanism in vivo for feedback regulation of the chitinase activity of human AMCase. HPLC characterization of cleavage of chitooligosaccharides (4-6-mers) suggests that human AMCase prefers the beta anomer of chitooligosaccharides as substrate. Human AMCase also appears to cleave chitooligosaccharides from the nonreducing end primarily by disaccharide units. Ionic strength modulates the enzymatic activity and substrate cleavage pattern of human AMCase against fluorogenic substrates, chitobiose-4-methylumbelliferyl and chitotriose-4-methylumbelliferyl, and enhances activity against chitooligosaccharides. The physiological implications of these results are discussed.


Asunto(s)
Quitinasas/metabolismo , Animales , Células CHO , Cricetinae , Cricetulus , Humanos , Concentración de Iones de Hidrógeno , Cinética , Concentración Osmolar , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...