Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
2.
ACS Med Chem Lett ; 9(9): 935-940, 2018 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-30258544

RESUMO

The X-ray structure of the previously reported PPARδ modulator 1 bound to the ligand binding domain (LBD) revealed that the amide moiety in 1 exists in the thermodynamically disfavored cis-amide orientation. Isosteric replacement of the cis-amide with five-membered heterocycles led to the identification of imidazole 17 (MA-0204), a potent, selective PPARδ modulator with good pharmacokinetic properties. MA-0204 was tested in vivo in mice and in vitro in patient-derived muscle myoblasts (from Duchenne Muscular Dystrophy (DMD) patients); 17 altered the expression of PPARδ target genes and improved fatty acid oxidation, which supports the therapeutic hypothesis for the study of MA-0204 in DMD patients.

3.
Bioorg Med Chem Lett ; 28(15): 2655-2659, 2018 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-29935771

RESUMO

Mitophagy is one of the processes that cells use to maintain overall health. An E3 ligase, parkin, ubiquitinates mitochondrial proteins prior to their degradation by autophagasomes. USP30 is an enzyme that de-ubiquitinates mitochondrial proteins; therefore, inhibiting this enzyme could foster mitophagy. Herein, we disclose the structure-activity relationships (SAR) within a novel series of highly selective USP30 inhibitors. Two structurally similar compounds, MF-094 (a potent and selective USP30 inhibitor) and MF-095 (a significantly less potent USP30 inhibitor), serve as useful controls for biological evaluation. We show that MF-094 increases protein ubiquitination and accelerates mitophagy.


Assuntos
Proteínas Mitocondriais/antagonistas & inibidores , Mitofagia/efeitos dos fármacos , Inibidores de Proteases/farmacologia , Tioléster Hidrolases/antagonistas & inibidores , Animais , Camundongos , Mitocôndrias/enzimologia , Mitocôndrias/metabolismo , Proteínas Mitocondriais/metabolismo , Inibidores de Proteases/química , Relação Estrutura-Atividade , Tioléster Hidrolases/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação
4.
Bioorg Med Chem Lett ; 28(3): 533-536, 2018 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29275935

RESUMO

Compound 1 regulates significantly fewer genes than the PPARδ modulator, GW501516. Both compounds are efficacious in a thermal injury model of muscle regeneration. The restricted gene profile of 1 relative to GW501516 suggests that 1 may be pharmacoequivalent to GW501516 with fewer PPAR-related safety concerns.


Assuntos
PPAR delta/metabolismo , Tiazóis/farmacologia , Animais , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Camundongos , Estrutura Molecular , Ratos , Relação Estrutura-Atividade
5.
Bioorg Med Chem Lett ; 27(23): 5230-5234, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29103972

RESUMO

Optimization of benzamide PPARδ modulator 1 led to (E)-6-(2-((4-(furan-2-yl)-N-methylbenzamido)methyl)phenoxy)-4-methylhex-4-enoic acid (18), a potent selective PPARδ modulator with significantly improved exposure in multiple species following oral administration.


Assuntos
Benzamidas/farmacocinética , Administração Oral , Animais , Benzamidas/administração & dosagem , Benzamidas/sangue , Relação Dose-Resposta a Droga , Humanos , Injeções Intravenosas , Macaca fascicularis , Masculino , Camundongos , Estrutura Molecular , Ratos Wistar , Relação Estrutura-Atividade
6.
Bioorg Med Chem Lett ; 25(22): 5309-14, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26421993

RESUMO

Hepsin, a type II transmembrane serine protease, is upregulated in prostate cancer and known to be involved in the progression of metastasis. Here we report a structure-guided approach, which resulted in the discovery of 2-aryl/pyridin-2-yl-1H-indole derivatives as potent and selective inhibitors of hepsin. Potent and selective inhibition of hepsin by compound 8 is likely due to interactions of the amidine group at the S1 site with the cyclohexyl ring from the 2-aryl group projecting towards the S1' site and the tert-hydroxyl group interacting with His57 side-chain as revealed by X-ray crystallography. Compounds 8 and 10, showed Ki of 0.1 µM for hepsin, and exhibited inhibition of invasion and migration of hepsin-overexpressing cell line. Compounds described here could serve as useful tool reagents to investigate the role of hepsin as a potential therapeutic target in cancer.


Assuntos
Antineoplásicos/farmacologia , Cicloexanos/farmacologia , Indóis/farmacologia , Piridinas/farmacologia , Serina Endopeptidases/metabolismo , Inibidores de Serina Proteinase/farmacologia , Animais , Antineoplásicos/síntese química , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Cicloexanos/síntese química , Humanos , Indóis/síntese química , Camundongos , Microssomos Hepáticos/metabolismo , Simulação de Acoplamento Molecular , Invasividade Neoplásica , Piridinas/síntese química , Inibidores de Serina Proteinase/síntese química
7.
Bioorg Med Chem Lett ; 25(4): 887-92, 2015 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-25599839

RESUMO

Kappa opioid receptor (KOR) is an important mediator of pain signaling and it is targeted for the treatment of various pains. Pharmacophore based mining of databases led to the identification of 2-aminobenzimidazole derivative as KOR agonists with selectivity over the other opioid receptors DOR and MOR. A short SAR exploration with the objective of identifying more polar and hence less brain penetrant agonists is described herewith. Modeling studies of the recently published structures of KOR, DOR and MOR are used to explain the receptor selectivity. The synthesis, biological evaluation and SAR of novel benzimidazole derivatives as KOR agonists are described. The in vivo proof of principle for anti-nociceptive effect with a lead compound from this series is exemplified.


Assuntos
Benzimidazóis/farmacologia , Receptores Opioides kappa/agonistas , Sequência de Aminoácidos , Simulação por Computador , Humanos , Dados de Sequência Molecular , Receptores Opioides kappa/química , Homologia de Sequência de Aminoácidos , Relação Estrutura-Atividade
8.
Biochem J ; 405(2): 307-17, 2007 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-17447891

RESUMO

Mutations in the LRRK2 (leucine-rich repeat kinase-2) gene cause late-onset PD (Parkinson's disease). LRRK2 contains leucine-rich repeats, a GTPase domain, a COR [C-terminal of Roc (Ras of complex)] domain, a kinase and a WD40 (Trp-Asp 40) motif. Little is known about how LRRK2 is regulated, what its physiological substrates are or how mutations affect LRRK2 function. Thus far LRRK2 activity has only been assessed by autophosphorylation and phosphorylation of MBP (myelin basic protein), which is catalysed rather slowly. We undertook a KESTREL (kinase substrate tracking and elucidation) screen in rat brain extracts to identify proteins that were phosphorylated by an activated PD mutant of LRRK2 (G2019S). This led to the discovery that moesin, a protein which anchors the actin cytoskeleton to the plasma membrane, is efficiently phosphorylated by LRRK2, at Thr558, a previously identified in-vivo-phosphorylation site that regulates the ability of moesin to bind actin. LRRK2 also phosphorylated ezrin and radixin, which are related to moesin, at the residue equivalent to Thr558, as well as a peptide (LRRKtide: RLGRDKYKTLRQIRQ) encompassing Thr558. We exploited these findings to determine how nine previously reported PD mutations of LRRK2 affected kinase activity. Only one of the mutations analysed, namely G2019S, stimulated kinase activity. Four mutations inhibited LRRK2 kinase activity (R1941H, I2012T, I2020T and G2385R), whereas the remainder (R1441C, R1441G, Y1699C and T2356I) did not influence activity. Therefore the manner in which LRRK2 mutations induce PD is more complex than previously imagined and is not only caused by an increase in LRRK2 kinase activity. Finally, we show that the minimum catalytically active fragment of LRRK2 requires an intact GTPase, COR and kinase domain, as well as a WD40 motif and a C-terminal tail. The results of the present study suggest that moesin, ezrin and radixin may be LRRK2 substrates, findings that have been exploited to develop the first robust quantitative assay to measure LRRK2 kinase activity.


Assuntos
Proteínas dos Microfilamentos/metabolismo , Doença de Parkinson/genética , Proteínas Serina-Treonina Quinases/metabolismo , Treonina/metabolismo , Sequência de Aminoácidos , Animais , Proteínas do Citoesqueleto/metabolismo , Humanos , Serina-Treonina Proteína Quinase-2 com Repetições Ricas em Leucina , Proteínas de Membrana/metabolismo , Doença de Parkinson/metabolismo , Proteínas Serina-Treonina Quinases/genética , Ratos
9.
Biochemistry ; 45(6): 1888-98, 2006 Feb 14.
Artigo em Inglês | MEDLINE | ID: mdl-16460035

RESUMO

The role of the kinase homology domain (KHD) in receptor guanylyl cyclases is to regulate the activity of the catalytic guanylyl cyclase domain. The KHD lacks many of the amino acids required for phosphotransfer activity and, therefore, is not expected to possess kinase activity. Guanylyl cyclase activity of the receptor guanylyl cyclase C (GC-C) is modulated by ATP, and computational modeling showed that the KHD can adopt a structure similar to protein kinases, suggesting that the KHD is the site for ATP interaction. A monoclonal antibody, GCC:4D7, raised to the KHD of GC-C, fails to react with GC-C in the presence of ATP and ATP analogues that regulate GC-C catalytic activity, indicating that a conformational change occurs in the KHD on ATP binding. Mapping of the epitope of the antibody through the use of recombinant protein constructs and phage display showed that the epitope for GC-C:4D7 lies immediately C-terminal to a critical lysine residue (Lys516 in GC-C), required for ATP interaction in protein kinases. By employing a novel approach utilizing ATP-agarose affinity chromatography, we demonstrate that the intracellular domain of GC-C and the KHD bind ATP. Mutation of Lys516 to Ala abolishes ATP binding. Thus, this report is the first to show direct ATP binding to the pseudokinase domain of receptor guanylyl cyclase C, as well as to identify dramatic conformational changes that occur in this domain on ATP binding, akin to those seen in catalytically active protein kinases.


Assuntos
Nucleotídeos de Adenina/metabolismo , Trifosfato de Adenosina/metabolismo , Guanilato Ciclase/metabolismo , Nucleotídeos de Adenina/química , Nucleotídeos de Adenina/genética , Sequência de Aminoácidos , Animais , Anticorpos Monoclonais/imunologia , Sequência de Bases , Sítios de Ligação , Cromatografia de Afinidade , Epitopos/genética , Epitopos/metabolismo , Guanilato Ciclase/química , Guanilato Ciclase/genética , Lisina/genética , Dados de Sequência Molecular , Mutação , Fosfotransferases/metabolismo , Estrutura Terciária de Proteína
10.
Biochem J ; 394(Pt 3): 545-55, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16396636

RESUMO

Recent work indicates that the LKB1 tumour suppressor protein kinase, which is mutated in Peutz-Jeghers cancer syndrome, phosphorylates and activates a group of protein kinases that are related to AMPK (AMP-activated protein kinase). Ten of the 14 AMPK-related protein kinases activated by LKB1, including SIK (salt-induced kinase), MARK (microtubule-affinity-regulating kinase) and BRSK (brain-specific kinase) isoforms, possess a ubiquitin-associated (UBA) domain immediately C-terminal to the kinase catalytic domain. These are the only protein kinases in the human genome known to possess a UBA domain, but their roles in regulating AMPK-related kinases are unknown. We have investigated the roles that the UBA domain may play in regulating these enzymes. Limited proteolysis of MARK2 revealed that the kinase and UBA domains were contained within a fragment that was resistant to trypsin proteolysis. SAXS (small-angle X-ray scattering) analysis of inactive and active LKB1-phosphorylated MARK2 revealed that activation of MARK2 is accompanied by a significant conformational change that alters the orientation of the UBA domain with respect to the catalytic domain. Our results indicate that none of the UBA domains found in AMPK-related kinases interact with polyubiquitin or other ubiquitin-like molecules. Instead, the UBA domains appear to play an essential conformational role and are required for the LKB1-mediated phosphorylation and activation of AMPK-related kinases. This is based on the findings that mutation or removal of the UBA domains of several AMPK-related kinases, including isoforms of MARK, SIK and BRSK, markedly impaired the catalytic activity and LKB1-mediated phosphorylation of these enzymes. We also provide evidence that the UBA domains do not function as LKB1-STRAD (STE20-related adaptor)-MO25 (mouse protein 25) docking/interacting sites and that mutations in the UBA domain of SIK suppressed the ability of SIK to localize within punctate regions of the nucleus. Taken together, these findings suggest that the UBA domains of AMPK-related kinases play an important role in regulating the conformation, activation and localization of these enzymes.


Assuntos
Complexos Multienzimáticos/química , Complexos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/metabolismo , Ubiquitina/metabolismo , Quinases Proteína-Quinases Ativadas por AMP , Proteínas Quinases Ativadas por AMP , Sequência de Aminoácidos , Linhagem Celular , Ativação Enzimática , Escherichia coli , Regulação da Expressão Gênica , Humanos , Modelos Moleculares , Dados de Sequência Molecular , Complexos Multienzimáticos/genética , Fosforilação , Proteínas Serina-Treonina Quinases/genética , Estrutura Terciária de Proteína , Transporte Proteico , Alinhamento de Sequência , Homologia de Sequência de Aminoácidos
11.
FEBS Lett ; 579(6): 1417-23, 2005 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-15733851

RESUMO

Recent work has shown that the LKB1 tumour suppressor protein kinase phosphorylates and activates protein kinases belonging to the AMP activated kinase (AMPK) subfamily. In this study, we identify the sucrose non-fermenting protein (SNF1)-related kinase (SNRK), a largely unstudied AMPK subfamily member, as a novel substrate for LKB1. We demonstrate that LKB1 activates SNRK by phosphorylating the T-loop residue (Thr173), and that the LKB1 regulatory subunits STRAD and MO25 are required for LKB1 to activate SNRK. We find that SNRK is not active when expressed in HeLa cells that lack expression of LKB1, and its activity is restored by expression of wild type LKB1, but not catalytically deficient LKB1. We also present evidence that two other AMPK-related kinases more distantly related to AMPK than SNRK, namely NIM1 and testis-specific serine/threonine kinase-1 (TSSK1) are not substrates for LKB1. Tissue distribution analysis indicates that SNRK protein is mainly expressed in testis, similar to TSSK isoforms, whereas NIM1 is more widely expressed. These results provide evidence that SNRK could mediate some of the physiological effects of LKB1.


Assuntos
Proteínas Serina-Treonina Quinases/metabolismo , Sacarose/metabolismo , Proteínas Quinases Ativadas por AMP , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Sequência de Aminoácidos , Animais , Catálise , Células Cultivadas , Ativação Enzimática , Fermentação , Células HeLa , Humanos , Camundongos , Camundongos Knockout , Dados de Sequência Molecular , Fosforilação , Proteínas Serina-Treonina Quinases/química , Proteínas Serina-Treonina Quinases/classificação , Alinhamento de Sequência , Especificidade por Substrato
12.
Biochemistry ; 43(25): 8247-55, 2004 Jun 29.
Artigo em Inglês | MEDLINE | ID: mdl-15209521

RESUMO

Guanylyl cyclase C (GC-C), the receptor for guanylin, uroguanylin, and the heat-stable enterotoxin, regulates fluid balance in the intestine and extraintestinal tissues. The receptor has an extracellular domain, a single transmembrane spanning domain, and an intracellular domain that harbors a region homologous to protein kinases, followed by the C-terminal guanylyl cyclase domain. Adenine nucleotides can regulate the guanylyl cyclase activity of GC-C by binding to the intracellular kinase homology domain (KHD). In this study, we have tested the effect of several protein kinase inhibitors on GC-C activity and find that the tyrphostins, known to be tyrosine kinase inhibitors, could inhibit GC-C activity in vitro. Tyrphostin A25 (AG82) was the most potent inhibitor with an IC(50) of approximately 15 microM. The mechanism of inhibition was found to be noncompetitive with respect to both the substrate MnGTP and the metal cofactor. Interestingly, the activity of the catalytic domain of GC-C (lacking the KHD) expressed in insect cells was also inhibited by tyrphostin A25 with an IC(50) of approximately 5 microM. As with the full-length receptor, inhibition was found to be noncompetitive with respect to MnGTP. Inhibition was reversible, ruling out a covalent modification of the receptor. Structurally similar proteins such as the soluble guanylyl cyclase and the adenylyl cyclases were also inhibited by tyrphostin A25. Evaluation of a number of tyrphostins allowed us to identify the requirement of two vicinal hydroxyl groups in the tyrphostin for effective inhibition of cyclase activity. Therefore, our studies are the first to report that nucleotide cyclases are inhibited by tyrphostins and suggest that novel inhibitors based on the tyrphostin scaffold can be developed, which could aid in a greater understanding of nucleotide cyclase structure and function.


Assuntos
Inibidores de Adenilil Ciclases , Inibidores Enzimáticos/farmacologia , Guanilato Ciclase/antagonistas & inibidores , Tirfostinas/farmacologia , Adenilil Ciclases/genética , Adenilil Ciclases/metabolismo , Animais , Domínio Catalítico , Linhagem Celular , GMP Cíclico/metabolismo , Citosol/enzimologia , Cães , Escherichia coli/metabolismo , Guanosina Trifosfato/química , Guanosina Trifosfato/metabolismo , Guanilato Ciclase/química , Guanilato Ciclase/genética , Guanilato Ciclase/metabolismo , Humanos , Concentração Inibidora 50 , Cinética , Manganês/química , Manganês/farmacologia , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/metabolismo , Proteínas Tirosina Quinases/antagonistas & inibidores , Coelhos , Ensaio Radioligante , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase , Receptores de Peptídeos/antagonistas & inibidores , Receptores de Peptídeos/química , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Proteínas Recombinantes/antagonistas & inibidores , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Spodoptera/citologia , Relação Estrutura-Atividade , Tirfostinas/química , Tirfostinas/metabolismo
13.
Biol Reprod ; 67(6): 1975-80, 2002 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-12444076

RESUMO

Guanylyl cyclase C (GC-C) is a membrane-associated form of guanylyl cyclase and serves as the receptor for the heat-stable enterotoxin (ST) peptide and endogenous ligands guanylin, uroguanylin, and lymphoguanylin. The major site of expression of GC-C is the intestinal epithelial cell, although GC-C is also expressed in extraintestinal tissue such as the kidney, airway epithelium, perinatal liver, stomach, brain, and adrenal glands. Binding of ligands to GC-C leads to accumulation of intracellular cGMP, the activation of protein kinases G and A, and phosphorylation of the cystic fibrosis transmembrane conductance regulator (CFTR), a chloride channel that regulates salt and water secretion. We examined the expression of GC-C and its ligands in various tissues of the reproductive tract of the rat. Using reverse transcriptase and the polymerase chain reaction, we demonstrated the presence of GC-C, uroguanylin, and guanylin mRNA in both male and female reproductive organs. Western blot analysis using a monoclonal antibody to GC-C revealed the presence of differentially glycosylated forms of GC-C in the caput and cauda epididymis. Exogenous addition of uroguanylin to minced epididymal tissue resulted in cGMP accumulation, suggesting an autocrine or endocrine activation of GC-C in this tissue. Immunohistochemical analyses demonstrated expression of GC-C in the tubular epithelial cells of both the caput epididymis and cauda epididymis. Our results suggest that the GC-C signaling pathway could converge on CFTR in the epididymis and perhaps control fluid and ion balance for optimal sperm maturation and storage in this tissue.


Assuntos
Epididimo/enzimologia , Expressão Gênica , Guanilato Ciclase/genética , Guanilato Ciclase/fisiologia , Receptores de Peptídeos/genética , Receptores de Peptídeos/fisiologia , Transdução de Sinais , Animais , Anticorpos Monoclonais , GMP Cíclico/biossíntese , Regulador de Condutância Transmembrana em Fibrose Cística/metabolismo , Feminino , Hormônios Gastrointestinais/genética , Imuno-Histoquímica , Ligantes , Masculino , Peptídeos Natriuréticos , Especificidade de Órgãos , Peptídeos/genética , Fosforilação , RNA Mensageiro/análise , Ratos , Receptores de Enterotoxina , Receptores Acoplados a Guanilato Ciclase , Reação em Cadeia da Polimerase Via Transcriptase Reversa
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...