Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Cell Rep ; 42(8): 112763, 2023 08 29.
Artigo em Inglês | MEDLINE | ID: mdl-37478012

RESUMO

Kynurenine monooxygenase (KMO) blockade protects against multiple organ failure caused by acute pancreatitis (AP), but the link between KMO and systemic inflammation has eluded discovery until now. Here, we show that the KMO product 3-hydroxykynurenine primes innate immune signaling to exacerbate systemic inflammation during experimental AP. We find a tissue-specific role for KMO, where mice lacking Kmo solely in hepatocytes have elevated plasma 3-hydroxykynurenine levels that prime inflammatory gene transcription. 3-Hydroxykynurenine synergizes with interleukin-1ß to cause cellular apoptosis. Critically, mice with elevated 3-hydroxykynurenine succumb fatally earlier and more readily to experimental AP. Therapeutically, blockade with the highly selective KMO inhibitor GSK898 rescues the phenotype, reducing 3-hydroxykynurenine and protecting against critical illness and death. Together, our findings establish KMO and 3-hydroxykynurenine as regulators of inflammation and the innate immune response to sterile inflammation. During critical illness, excess morbidity and death from multiple organ failure can be rescued by systemic KMO blockade.


Assuntos
Cinurenina , Pancreatite , Camundongos , Animais , Estado Terminal , Insuficiência de Múltiplos Órgãos , Doença Aguda , Camundongos Knockout , Inflamação , Quinurenina 3-Mono-Oxigenase/genética
2.
Ann Surg ; 275(2): e453-e462, 2022 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-32487804

RESUMO

OBJECTIVE: Acute Pancreatitis (AP) is sudden onset pancreas inflammation that causes systemic injury with a wide and markedly heterogeneous range of clinical consequences. Here, we hypothesized that this observed clinical diversity corresponds to diversity in molecular subtypes that can be identified in clinical and multiomics data. SUMMARY BACKGROUND DATA: Observational cohort study. n = 57 for the discovery cohort (clinical, transcriptomics, proteomics, and metabolomics data) and n = 312 for the validation cohort (clinical and metabolomics data). METHODS: We integrated coincident transcriptomics, proteomics, and metabolomics data at serial time points between admission to hospital and up to 48 hours after recruitment from a cohort of patients presenting with acute pancreatitis. We systematically evaluated 4 different metrics for patient similarity using unbiased mathematical, biological, and clinical measures of internal and external validity.We next compared the AP molecular endotypes with previous descriptions of endotypes in a critically ill population with acute respiratory distress syndrome (ARDS). RESULTS: Our results identify 4 distinct and stable AP molecular endotypes. We validated our findings in a second independent cohort of patients with AP.We observed that 2 endotypes in AP recapitulate disease endotypes previously reported in ARDS. CONCLUSIONS: Our results show that molecular endotypes exist in AP and reflect biological patterns that are also present in ARDS, suggesting that generalizable patterns exist in diverse presentations of critical illness.


Assuntos
Pancreatite/classificação , Pancreatite/diagnóstico , Estudos de Coortes , Humanos , Metabolômica , Proteômica
3.
Br J Clin Pharmacol ; 88(2): 865-870, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34327739

RESUMO

GSK3335065 is an inhibitor of kynurenine monooxygenase (KMO) being developed for the treatment of acute pancreatitis. Healthy male volunteers were administered ascending doses of GSK3335065 or matched placebo as a single intravenous bolus injection to assess safety, tolerability, pharmacokinetics and pharmacodynamics. GSK3335065 displayed an apparent volume of distribution between 20.6 L and 44.6 L, a clearance between 0.462 L/h and 0.805 L/hr and a terminal half-life between 31.3 and 34.5 hr. In the single subject who received 1.3 mg GSK3335065, changes in tryptophan pathway metabolites were observed consistent with the changes seen in preclinical species suggesting that KMO enzyme activity was partially inhibited. However, a broad complex ventricular tachycardia was observed in this subject, which was judged to be a Serious Adverse Event (SAE) and resulted in early termination of the study. While development of GSK3335065 was subsequently discontinued, significant confounding factors hinder a clear interpretation that the tachycardia was directly related to administration of the compound.


Assuntos
Cinurenina , Pancreatite , Doença Aguda , Método Duplo-Cego , Voluntários Saudáveis , Humanos , Masculino , Oxigenases de Função Mista
5.
Bioorg Med Chem Lett ; 41: 127973, 2021 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-33753261

RESUMO

α1-antitrypsin deficiency is characterised by the misfolding and intracellular polymerisation of mutant α1-antitrypsin protein within the endoplasmic reticulum (ER) of hepatocytes. Small molecules that bind and stabilise Z α1-antitrypsin were identified via a DNA-encoded library screen. A subsequent structure based optimisation led to a series of highly potent, selective and cellular active α1-antitrypsin correctors.


Assuntos
Desenho de Fármacos , Dobramento de Proteína , alfa 1-Antitripsina/metabolismo , Cristalização , Desenvolvimento de Medicamentos/métodos , Avaliação Pré-Clínica de Medicamentos , Retículo Endoplasmático/metabolismo , Biblioteca Gênica , Hepatócitos/metabolismo , Humanos , Modelos Moleculares , Conformação Proteica , alfa 1-Antitripsina/genética
6.
EMBO Mol Med ; 13(3): e13167, 2021 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-33512066

RESUMO

Severe α1 -antitrypsin deficiency results from the Z allele (Glu342Lys) that causes the accumulation of homopolymers of mutant α1 -antitrypsin within the endoplasmic reticulum of hepatocytes in association with liver disease. We have used a DNA-encoded chemical library to undertake a high-throughput screen to identify small molecules that bind to, and stabilise Z α1 -antitrypsin. The lead compound blocks Z α1 -antitrypsin polymerisation in vitro, reduces intracellular polymerisation and increases the secretion of Z α1 -antitrypsin threefold in an iPSC model of disease. Crystallographic and biophysical analyses demonstrate that GSK716 and related molecules bind to a cryptic binding pocket, negate the local effects of the Z mutation and stabilise the bound state against progression along the polymerisation pathway. Oral dosing of transgenic mice at 100 mg/kg three times a day for 20 days increased the secretion of Z α1 -antitrypsin into the plasma by sevenfold. There was no observable clearance of hepatic inclusions with respect to controls over the same time period. This study provides proof of principle that "mutation ameliorating" small molecules can block the aberrant polymerisation that underlies Z α1 -antitrypsin deficiency.


Assuntos
Deficiência de alfa 1-Antitripsina , alfa 1-Antitripsina , Animais , Retículo Endoplasmático , Hepatócitos , Camundongos , alfa 1-Antitripsina/genética
7.
Sci Transl Med ; 12(541)2020 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-32350131

RESUMO

Myotonic dystrophy type 1 (DM1) is an RNA-based disease with no current treatment. It is caused by a transcribed CTG repeat expansion within the 3' untranslated region of the dystrophia myotonica protein kinase (DMPK) gene. Mutant repeat expansion transcripts remain in the nuclei of patients' cells, forming distinct microscopically detectable foci that contribute substantially to the pathophysiology of the condition. Here, we report small-molecule inhibitors that remove nuclear foci and have beneficial effects in the HSALR mouse model, reducing transgene expression, leading to improvements in myotonia, splicing, and centralized nuclei. Using chemoproteomics in combination with cell-based assays, we identify cyclin-dependent kinase 12 (CDK12) as a druggable target for this condition. CDK12 is a protein elevated in DM1 cell lines and patient muscle biopsies, and our results showed that its inhibition led to reduced expression of repeat expansion RNA. Some of the inhibitors identified in this study are currently the subject of clinical trials for other indications and provide valuable starting points for a drug development program in DM1.


Assuntos
Distrofia Miotônica , Animais , Quinases Ciclina-Dependentes , Modelos Animais de Doenças , Humanos , Camundongos , Distrofia Miotônica/tratamento farmacológico , Distrofia Miotônica/genética , RNA , Splicing de RNA/genética , Expansão das Repetições de Trinucleotídeos/genética
8.
J Med Chem ; 63(6): 3348-3358, 2020 03 26.
Artigo em Inglês | MEDLINE | ID: mdl-32109056

RESUMO

ER aminopeptidase 1 (ERAP1) is an intracellular enzyme that generates antigenic peptides and is an emerging target for cancer immunotherapy and the control of autoimmunity. ERAP1 inhibitors described previously target the active site and are limited in selectivity, minimizing their clinical potential. To address this, we targeted the regulatory site of ERAP1 using a high-throughput screen and discovered a small molecule hit that is highly selective for ERAP1. (4aR,5S,6R,8S,8aR)-5-(2-(Furan-3-yl)ethyl)-8-hydroxy-5,6,8a-trimethyl-3,4,4a,5,6,7,8,8a-octahydronaphthalene-1-carboxylic acid is a natural product found in Dodonaea viscosa that constitutes a submicromolar, highly selective, and cell-active modulator of ERAP1. Although the compound activates hydrolysis of small model substrates, it is a competitive inhibitor for physiologically relevant longer peptides. Crystallographic analysis confirmed that the compound targets the regulatory site of the enzyme that normally binds the C-terminus of the peptide substrate. Our findings constitute a novel starting point for the development of selective ERAP1 modulators that have potential for further clinical development.


Assuntos
Aminopeptidases/antagonistas & inibidores , Apresentação de Antígeno/efeitos dos fármacos , Diterpenos Clerodânicos/farmacologia , Epitopos/metabolismo , Peptídeos/metabolismo , Inibidores de Proteases/farmacologia , Sítio Alostérico , Aminopeptidases/química , Aminopeptidases/metabolismo , Animais , Domínio Catalítico , Cristalografia por Raios X , Diterpenos Clerodânicos/química , Diterpenos Clerodânicos/metabolismo , Ativadores de Enzimas/química , Ativadores de Enzimas/metabolismo , Ativadores de Enzimas/farmacologia , Epitopos/química , Células HeLa , Humanos , Camundongos , Antígenos de Histocompatibilidade Menor/química , Antígenos de Histocompatibilidade Menor/metabolismo , Peptídeos/química , Inibidores de Proteases/química , Inibidores de Proteases/metabolismo , Ligação Proteica , Proteólise/efeitos dos fármacos
9.
J Med Chem ; 62(17): 8274-8283, 2019 09 12.
Artigo em Inglês | MEDLINE | ID: mdl-31393717

RESUMO

The wild type protein, transthyretin (TTR), and over 120 genetic TTR variants are amyloidogenic and cause, respectively, sporadic and hereditary systemic TTR amyloidosis. The homotetrameric TTR contains two identical thyroxine binding pockets, occupation of which by specific ligands can inhibit TTR amyloidogenesis in vitro. Ligand binding stabilizes the tetramer, inhibiting its proteolytic cleavage and its dissociation. Here, we show with solution-state NMR that ligand binding induces long-distance conformational changes in the TTR that have not previously been detected by X-ray crystallography, consistently with the inhibition of the cleavage of the DE loop. The NMR findings, coupled with surface plasmon resonance measurements, have identified dynamic exchange processes underlying the negative cooperativity of binding of "monovalent" ligand tafamidis. In contrast, mds84, our prototypic "bivalent" ligand, which is a more potent stabilizer of TTR in vitro that occupies both thyroxine pockets and the intramolecular channel between them, has greater structural effects.


Assuntos
Fenamatos/química , Pré-Albumina/química , Sítios de Ligação , Fenamatos/síntese química , Humanos , Ligantes , Modelos Moleculares , Conformação Molecular , Estrutura Molecular , Pré-Albumina/síntese química , Relação Estrutura-Atividade
10.
Neurosci Lett ; 673: 44-50, 2018 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-29499308

RESUMO

Expression of mutant Huntingtin (HTT) protein is central to the pathophysiology of Huntington's Disease (HD). The E3 ubiquitin ligase MID1 appears to have a key role in facilitating translation of the mutant HTT mRNA suggesting that interference with the function of this complex could be an attractive therapeutic approach. Here we describe a peptide that is able to disrupt the interaction between MID1 and the α4 protein, a regulatory subunit of protein phosphatase 2A (PP2A). By fusing this peptide to a sequence from the HIV-TAT protein we demonstrate that the peptide can disrupt the interaction within cells and show that this results in a decrease in levels of ribosomal S6 phosphorylation and HTT expression in cultures of cerebellar granule neurones derived from HdhQ111/Q7 mice. This data serves to validate this pathway and paves the way for the discovery of small molecule inhibitors of this interaction as potential therapies for HD.


Assuntos
Proteína Huntingtina/metabolismo , Neurônios/metabolismo , Proteínas/metabolismo , Animais , Células HEK293 , Humanos , Proteína Huntingtina/genética , Camundongos , Mutação , Cultura Primária de Células , Ligação Proteica , Proteína Fosfatase 2/metabolismo , Ubiquitina-Proteína Ligases
11.
Nat Commun ; 8: 15827, 2017 06 12.
Artigo em Inglês | MEDLINE | ID: mdl-28604669

RESUMO

Kynurenine-3-monooxygenase (KMO) is a key FAD-dependent enzyme of tryptophan metabolism. In animal models, KMO inhibition has shown benefit in neurodegenerative diseases such as Huntington's and Alzheimer's. Most recently it has been identified as a target for acute pancreatitis multiple organ dysfunction syndrome (AP-MODS); a devastating inflammatory condition with a mortality rate in excess of 20%. Here we report and dissect the molecular mechanism of action of three classes of KMO inhibitors with differentiated binding modes and kinetics. Two novel inhibitor classes trap the catalytic flavin in a previously unobserved tilting conformation. This correlates with picomolar affinities, increased residence times and an absence of the peroxide production seen with previous substrate site inhibitors. These structural and mechanistic insights culminated in GSK065(C1) and GSK366(C2), molecules suitable for preclinical evaluation. Moreover, revising the repertoire of flavin dynamics in this enzyme class offers exciting new opportunities for inhibitor design.


Assuntos
Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Insuficiência de Múltiplos Órgãos/metabolismo , Pancreatite/metabolismo , Animais , Inibidores Enzimáticos/química , Escherichia coli/genética , Humanos , Peróxido de Hidrogênio/metabolismo , Quinurenina 3-Mono-Oxigenase/química , Quinurenina 3-Mono-Oxigenase/metabolismo , Modelos Moleculares , Domínios Proteicos , Células Sf9
12.
J Med Chem ; 60(8): 3383-3404, 2017 04 27.
Artigo em Inglês | MEDLINE | ID: mdl-28398044

RESUMO

Recently, we reported a novel role for KMO in the pathogenesis of acute pancreatitis (AP). A number of inhibitors of kynurenine 3-monooxygenase (KMO) have previously been described as potential treatments for neurodegenerative conditions and particularly for Huntington's disease. However, the inhibitors reported to date have insufficient aqueous solubility relative to their cellular potency to be compatible with the intravenous (iv) dosing route required in AP. We have identified and optimized a novel series of high affinity KMO inhibitors with favorable physicochemical properties. The leading example is exquisitely selective, has low clearance in two species, prevents lung and kidney damage in a rat model of acute pancreatitis, and is progressing into preclinical development.


Assuntos
Inibidores Enzimáticos/farmacologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Pancreatite/tratamento farmacológico , Doença Aguda , Animais , Inibidores Enzimáticos/uso terapêutico , Humanos , Ratos
14.
Sci Rep ; 6: 33951, 2016 Sep 27.
Artigo em Inglês | MEDLINE | ID: mdl-27669975

RESUMO

Inhibition of kynurenine 3-monooxygenase (KMO) protects against multiple organ dysfunction (MODS) in experimental acute pancreatitis (AP). We aimed to precisely define the kynurenine pathway activation in relation to AP and AP-MODS in humans, by carrying out a prospective observational study of all persons presenting with a potential diagnosis of AP for 90 days. We sampled peripheral venous blood at 0, 3, 6, 12, 24, 48, 72 and 168 hours post-recruitment. We measured tryptophan metabolite concentrations and analysed these in the context of clinical data and disease severity indices, cytokine profiles and C-reactive protein (CRP) concentrations. 79 individuals were recruited (median age: 59.6 years; 47 males, 59.5%). 57 met the revised Atlanta definition of AP: 25 had mild, 23 moderate, and 9 severe AP. Plasma 3-hydroxykynurenine concentrations correlated with contemporaneous APACHE II scores (R2 = 0.273; Spearman rho = 0.581; P < 0.001) and CRP (R2 = 0.132; Spearman rho = 0.455, P < 0.001). Temporal profiling showed early tryptophan depletion and contemporaneous 3-hydroxykynurenine elevation. Furthermore, plasma concentrations of 3-hydroxykynurenine paralleled systemic inflammation and AP severity. These findings support the rationale for investigating early intervention with a KMO inhibitor, with the aim of reducing the incidence and severity of AP-associated organ dysfunction.

15.
Nat Med ; 22(2): 202-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26752518

RESUMO

Acute pancreatitis (AP) is a common and devastating inflammatory condition of the pancreas that is considered to be a paradigm of sterile inflammation leading to systemic multiple organ dysfunction syndrome (MODS) and death. Acute mortality from AP-MODS exceeds 20% (ref. 3), and the lifespans of those who survive the initial episode are typically shorter than those of the general population. There are no specific therapies available to protect individuals from AP-MODS. Here we show that kynurenine-3-monooxygenase (KMO), a key enzyme of tryptophan metabolism, is central to the pathogenesis of AP-MODS. We created a mouse strain that is deficient for Kmo (encoding KMO) and that has a robust biochemical phenotype that protects against extrapancreatic tissue injury to the lung, kidney and liver in experimental AP-MODS. A medicinal chemistry strategy based on modifications of the kynurenine substrate led to the discovery of the oxazolidinone GSK180 as a potent and specific inhibitor of KMO. The binding mode of the inhibitor in the active site was confirmed by X-ray co-crystallography at 3.2 Å resolution. Treatment with GSK180 resulted in rapid changes in the levels of kynurenine pathway metabolites in vivo, and it afforded therapeutic protection against MODS in a rat model of AP. Our findings establish KMO inhibition as a novel therapeutic strategy in the treatment of AP-MODS, and they open up a new area for drug discovery in critical illness.


Assuntos
Benzoxazóis/farmacologia , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Insuficiência de Múltiplos Órgãos/genética , Oxazolidinonas/farmacologia , Pancreatite/genética , Propionatos/farmacologia , RNA Mensageiro/metabolismo , Doença Aguda , Animais , Cromatografia Líquida , Cristalografia por Raios X , Modelos Animais de Doenças , Células HEK293 , Hepatócitos/metabolismo , Humanos , Técnicas In Vitro , Rim/metabolismo , Rim/patologia , Quinurenina 3-Mono-Oxigenase/genética , Pulmão/metabolismo , Pulmão/patologia , Camundongos , Camundongos Knockout , Insuficiência de Múltiplos Órgãos/etiologia , Insuficiência de Múltiplos Órgãos/patologia , Pâncreas/metabolismo , Pâncreas/patologia , Pancreatite/complicações , Pancreatite/patologia , Ratos , Espectrometria de Massas em Tandem , Triptofano/metabolismo
16.
J Biomol Screen ; 19(4): 508-15, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24381207

RESUMO

Kynurenine 3-monooxygenase (KMO) is a therapeutically important target on the eukaryotic tryptophan catabolic pathway, where it converts L-kynurenine (Kyn) to 3-hydroxykynurenine (3-HK). We have cloned and expressed the human form of this membrane protein as a full-length GST-fusion in a recombinant baculovirus expression system. An enriched membrane preparation was used for a directed screen of approximately 78,000 compounds using a RapidFire mass spectrometry (RF-MS) assay. The RapidFire platform provides an automated solid-phase extraction system that gives a throughput of approximately 7 s per well to the mass spectrometer, where direct measurement of both the substrate and product allowed substrate conversion to be determined. The RF-MS methodology is insensitive to assay interference, other than where compounds have the same nominal mass as Kyn or 3-HK and produce the same mass transition on fragmentation. These instances could be identified by comparison with the product-only data. The screen ran with excellent performance (average Z' value 0.8) and provided several tractable hit series for further investigation.


Assuntos
Descoberta de Drogas , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala , Quinurenina 3-Mono-Oxigenase/antagonistas & inibidores , Espectrometria de Massas , Animais , Domínio Catalítico , Linhagem Celular , Avaliação Pré-Clínica de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/química , Humanos , Cinética , Quinurenina 3-Mono-Oxigenase/química , Quinurenina 3-Mono-Oxigenase/metabolismo , Espectrometria de Massas/métodos , Ligação Proteica , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo
17.
J Med Chem ; 56(19): 7501-15, 2013 Oct 10.
Artigo em Inglês | MEDLINE | ID: mdl-24015967

RESUMO

The bromo and extra C-terminal domain (BET) family of bromodomains are involved in binding epigenetic marks on histone proteins, more specifically acetylated lysine residues. This paper describes the discovery and structure-activity relationships (SAR) of potent benzodiazepine inhibitors that disrupt the function of the BET family of bromodomains (BRD2, BRD3, and BRD4). This work has yielded a potent, selective compound I-BET762 that is now under evaluation in a phase I/II clinical trial for nuclear protein in testis (NUT) midline carcinoma and other cancers.


Assuntos
Antineoplásicos/farmacologia , Benzodiazepinas/farmacologia , Proteínas Nucleares/antagonistas & inibidores , Fatores de Transcrição/antagonistas & inibidores , Animais , Anti-Inflamatórios/síntese química , Anti-Inflamatórios/farmacocinética , Anti-Inflamatórios/farmacologia , Antineoplásicos/síntese química , Antineoplásicos/farmacocinética , Apolipoproteína A-I/biossíntese , Benzodiazepinas/síntese química , Benzodiazepinas/farmacocinética , Proteínas de Ciclo Celular , Cães , Epigênese Genética , Humanos , Macaca fascicularis , Camundongos , Modelos Moleculares , Permeabilidade , Estrutura Terciária de Proteína , Ratos , Estereoisomerismo , Relação Estrutura-Atividade
18.
J Med Chem ; 54(11): 3827-38, 2011 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-21568322

RESUMO

Epigenetic mechanisms of gene regulation have a profound role in normal development and disease processes. An integral part of this mechanism occurs through lysine acetylation of histone tails which are recognized by bromodomains. While the biological and structural characterization of many bromodomain containing proteins has advanced considerably, the therapeutic tractability of this protein family is only now becoming understood. This paper describes the discovery and molecular characterization of potent (nM) small molecule inhibitors that disrupt the function of the BET family of bromodomains (Brd2, Brd3, and Brd4). By using a combination of phenotypic screening, chemoproteomics, and biophysical studies, we have discovered that the protein-protein interactions between bromodomains and acetylated histones can be antagonized by selective small molecules that bind at the acetylated lysine recognition pocket. X-ray crystal structures of compounds bound into bromodomains of Brd2 and Brd4 elucidate the molecular interactions of binding and explain the precisely defined stereochemistry required for activity.


Assuntos
Apolipoproteína A-I/genética , Benzodiazepinas/metabolismo , Benzodiazepinas/farmacologia , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/química , Acetilação , Sequência de Aminoácidos , Apolipoproteína A-I/química , Apolipoproteína A-I/metabolismo , Benzodiazepinas/síntese química , Benzodiazepinas/química , Sítios de Ligação , Cristalografia por Raios X , Descoberta de Drogas , Epigenômica , Células Hep G2 , Histonas/química , Histonas/genética , Histonas/metabolismo , Humanos , Lisina/química , Lisina/genética , Lisina/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Estrutura Molecular , Terapia de Alvo Molecular , Ligação Proteica , Proteínas Serina-Treonina Quinases/metabolismo , Estereoisomerismo , Fatores de Transcrição , Regulação para Cima
19.
Bioorg Med Chem Lett ; 21(4): 1126-33, 2011 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-21257309

RESUMO

A novel series of indazole non-steroidal glucocorticoid receptor agonist has been discovered. This series features a sulfonamide central core and meta amides which interact with the extended ligand binding domain. This series has produced some of the most potent and least lipophilic agonists of which we are aware such as 20a (NFκB pIC(50) 8.3 (100%), clogP 1.9). Certain analogues in this series also display evidence for modulated pharmacology.


Assuntos
Indazóis/química , Receptores de Glucocorticoides/agonistas , Sulfonamidas/síntese química , Sítios de Ligação , Linhagem Celular Tumoral , Simulação por Computador , Avaliação Pré-Clínica de Medicamentos , Humanos , Interações Hidrofóbicas e Hidrofílicas , Indazóis/síntese química , Indazóis/farmacologia , Receptores de Glucocorticoides/metabolismo , Relação Estrutura-Atividade , Sulfonamidas/farmacologia
20.
J Med Chem ; 53(11): 4531-44, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20469868

RESUMO

Glucocorticoid receptor (GR) agonists have been used for more than half a century as the most effective treatment of acute and chronic inflammatory conditions despite serious side effects that accompany their extended use that include glucose intolerance, muscle wasting, skin thinning, and osteoporosis. As a starting point for the identification of GR ligands with an improved therapeutic index, we wished to discover selective nonsteroidal GR agonists and antagonists with simplified structure compared to known GR ligands to serve as starting points for the optimization of dissociated GR modulators. To do so, we selected multiple chemical series by structure guided docking studies and evaluated GR agonist activity. From these efforts we identified 5-arylindazole compounds that showed moderate binding to the glucocorticoid receptor (GR) with clear opportunities for further development. Structure guided optimization was used to design arrays that led to potent GR agonists and antagonists. Several in vitro and in vivo experiments were utilized to demonstrate that GR agonist 23a (GSK9027) had a profile similar to that of a classical steroidal GR agonist.


Assuntos
Desenho de Fármacos , Indazóis/química , Indazóis/farmacologia , Receptores de Glucocorticoides/agonistas , Receptores de Glucocorticoides/antagonistas & inibidores , Animais , Relação Dose-Resposta a Droga , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Humanos , Indazóis/síntese química , Indazóis/farmacocinética , Masculino , Camundongos , Modelos Moleculares , NF-kappa B/metabolismo , Conformação Proteica , Ratos , Receptores de Glucocorticoides/química , Receptores de Glucocorticoides/metabolismo , Especificidade por Substrato
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA