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










Base de dados
Intervalo de ano de publicação
1.
J Med Chem ; 67(10): 8141-8160, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38728572

RESUMO

Human interleukin-1ß (IL-1ß) is a pro-inflammatory cytokine that plays a critical role in the regulation of the immune response and the development of various inflammatory diseases. In this publication, we disclose our efforts toward the discovery of IL-1ß binders that interfere with IL-1ß signaling. To this end, several technologies were used in parallel, including fragment-based screening (FBS), DNA-encoded library (DEL) technology, peptide discovery platform (PDP), and virtual screening. The utilization of distinct technologies resulted in the identification of new chemical entities exploiting three different sites on IL-1ß, all of them also inhibiting the interaction with the IL-1R1 receptor. Moreover, we identified lysine 103 of IL-1ß as a target residue suitable for the development of covalent, low-molecular-weight IL-1ß antagonists.


Assuntos
Interleucina-1beta , Humanos , Descoberta de Drogas , Interleucina-1beta/metabolismo , Ligantes , Receptores Tipo I de Interleucina-1/metabolismo , Receptores Tipo I de Interleucina-1/antagonistas & inibidores , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Relação Estrutura-Atividade , DNA/química , Biblioteca Gênica
2.
Nat Commun ; 14(1): 5497, 2023 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-37679328

RESUMO

Human interleukin-1ß (hIL-1ß) is a pro-inflammatory cytokine involved in many diseases. While hIL-1ß directed antibodies have shown clinical benefit, an orally available low-molecular weight antagonist is still elusive, limiting the applications of hIL-1ß-directed therapies. Here we describe the discovery of a low-molecular weight hIL-1ß antagonist that blocks the interaction with the IL-1R1 receptor. Starting from a low affinity fragment-based screening hit 1, structure-based optimization resulted in a compound (S)-2 that binds and antagonizes hIL-1ß with single-digit micromolar activity in biophysical, biochemical, and cellular assays. X-ray analysis reveals an allosteric mode of action that involves a hitherto unknown binding site in hIL-1ß encompassing two loops involved in hIL-1R1/hIL-1ß interactions. We show that residues of this binding site are part of a conformationally excited state of the mature cytokine. The compound antagonizes hIL-1ß function in cells, including primary human fibroblasts, demonstrating the relevance of this discovery for future development of hIL-1ß directed therapeutics.


Assuntos
Citocinas , Magreza , Humanos , Interleucina-1beta , Peso Molecular , Sítios de Ligação , Biofísica
3.
J Med Chem ; 63(15): 8088-8113, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32551603

RESUMO

The serine protease factor XI (FXI) is a prominent drug target as it holds promise to deliver efficacious anticoagulation without an enhanced risk of major bleeds. Several efforts have been described targeting the active form of the enzyme, FXIa. Herein, we disclose our efforts to identify potent, selective, and orally bioavailable inhibitors of FXIa. Compound 1, identified from a diverse library of internal serine protease inhibitors, was originally designed as a complement factor D inhibitor and exhibited submicromolar FXIa activity and an encouraging absorption, distribution, metabolism, and excretion (ADME) profile while being devoid of a peptidomimetic architecture. Optimization of interactions in the S1, S1ß, and S1' pockets of FXIa through a combination of structure-based drug design and traditional medicinal chemistry led to the discovery of compound 23 with subnanomolar potency on FXIa, enhanced selectivity over other coagulation proteases, and a preclinical pharmacokinetics (PK) profile consistent with bid dosing in patients.


Assuntos
Fator XIa/antagonistas & inibidores , Fator XIa/genética , Inibidores do Fator Xa/administração & dosagem , Inibidores do Fator Xa/química , Administração Oral , Sequência de Aminoácidos , Animais , Disponibilidade Biológica , Cães , Avaliação Pré-Clínica de Medicamentos/métodos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Ratos , Ratos Sprague-Dawley , Relação Estrutura-Atividade
4.
ACS Med Chem Lett ; 11(2): 188-194, 2020 Feb 13.
Artigo em Inglês | MEDLINE | ID: mdl-32071687

RESUMO

Inhibition of neprilysin (NEP) is widely studied as a therapeutic target for the treatment of hypertension, heart failure, and kidney disease. Sacubitril/valsartan (LCZ696) is a drug approved to reduce the risk of cardiovascular death in heart failure patients with reduced ejection fraction. LBQ657 is the active metabolite of sacubitril and an inhibitor of NEP. Previously, we have reported the crystal structure of NEP bound with LBQ657, whereby we noted the presence of a subsite in S1' that has not been explored before. We were also intrigued by the zinc coordination made by one of the carboxylic acids of LBQ657, leading us to explore alternative linkers to efficiently engage zinc for NEP inhibition. Structure-guided design culminated in the synthesis of selective, orally bioavailable, and subnanomolar inhibitors of NEP. A 17-fold boost in biochemical potency was observed upon addition of a chlorine atom that occupied the newly found subsite in S1'. We report herein the discovery and preclinical profiling of compound 13, which paved the path to our clinical candidate.

5.
J Med Chem ; 63(11): 5697-5722, 2020 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-32073845

RESUMO

The alternative pathway (AP) of the complement system is a key contributor to the pathogenesis of several human diseases including age-related macular degeneration, paroxysmal nocturnal hemoglobinuria (PNH), atypical hemolytic uremic syndrome (aHUS), and various glomerular diseases. The serine protease factor B (FB) is a key node in the AP and is integral to the formation of C3 and C5 convertase. Despite the prominent role of FB in the AP, selective orally bioavailable inhibitors, beyond our own efforts, have not been reported previously. Herein we describe in more detail our efforts to identify FB inhibitors by high-throughput screening (HTS) and leveraging insights from several X-ray cocrystal structures during optimization efforts. This work culminated in the discovery of LNP023 (41), which is currently being evaluated clinically in several diverse AP mediated indications.


Assuntos
Ácido Benzoico/química , Fator B do Complemento/antagonistas & inibidores , Indóis/química , Síndrome Hemolítico-Urêmica Atípica/metabolismo , Síndrome Hemolítico-Urêmica Atípica/patologia , Ácido Benzoico/metabolismo , Ácido Benzoico/farmacocinética , Sítios de Ligação , Domínio Catalítico , Fator B do Complemento/metabolismo , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Meia-Vida , Humanos , Indóis/metabolismo , Indóis/farmacocinética , Concentração Inibidora 50 , Degeneração Macular/metabolismo , Degeneração Macular/patologia , Simulação de Dinâmica Molecular , Relação Estrutura-Atividade
6.
Blood ; 133(13): 1507-1516, 2019 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-30692123

RESUMO

A large unmet medical need exists for safer antithrombotic drugs because all currently approved anticoagulant agents interfere with hemostasis, leading to an increased risk of bleeding. Genetic and pharmacologic evidence in humans and animals suggests that reducing factor XI (FXI) levels has the potential to effectively prevent and treat thrombosis with a minimal risk of bleeding. We generated a fully human antibody (MAA868) that binds the catalytic domain of both FXI (zymogen) and activated FXI. Our structural studies show that MAA868 traps FXI and activated FXI in an inactive, zymogen-like conformation, explaining its equally high binding affinity for both forms of the enzyme. This binding mode allows the enzyme to be neutralized before entering the coagulation process, revealing a particularly attractive anticoagulant profile of the antibody. MAA868 exhibited favorable anticoagulant activity in mice with a dose-dependent protection from carotid occlusion in a ferric chloride-induced thrombosis model. MAA868 also caused robust and sustained anticoagulant activity in cynomolgus monkeys as assessed by activated partial thromboplastin time without any evidence of bleeding. Based on these preclinical findings, we conducted a first-in-human study in healthy subjects and showed that single subcutaneous doses of MAA868 were safe and well tolerated. MAA868 resulted in dose- and time-dependent robust and sustained prolongation of activated partial thromboplastin time and FXI suppression for up to 4 weeks or longer, supporting further clinical investigation as a potential once-monthly subcutaneous anticoagulant therapy.


Assuntos
Anticorpos Monoclonais Humanizados/uso terapêutico , Anticoagulantes/uso terapêutico , Coagulação Sanguínea/efeitos dos fármacos , Fator XI/antagonistas & inibidores , Trombose/tratamento farmacológico , Adolescente , Adulto , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Anticoagulantes/farmacologia , Feminino , Humanos , Imunoglobulina G/farmacologia , Imunoglobulina G/uso terapêutico , Macaca fascicularis , Masculino , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Simulação de Acoplamento Molecular , Trombose/sangue , Adulto Jovem
7.
Sci Rep ; 6: 27909, 2016 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-27302413

RESUMO

Sacubitril is an ethyl ester prodrug of LBQ657, the active neprilysin (NEP) inhibitor, and a component of LCZ696 (sacubitril/valsartan). We report herein the three-dimensional structure of LBQ657 in complex with human NEP at 2 Å resolution. The crystal structure unravels the binding mode of the compound occupying the S1, S1' and S2' sub-pockets of the active site, consistent with a competitive inhibition mode. An induced fit conformational change upon binding of the P1'-biphenyl moiety of the inhibitor suggests an explanation for its selectivity against structurally homologous zinc metallopeptidases.


Assuntos
Aminobutiratos/química , Compostos de Bifenilo/química , Neprilisina/química , Neprilisina/metabolismo , Aminobutiratos/metabolismo , Compostos de Bifenilo/metabolismo , Domínio Catalítico , Cristalografia por Raios X , Combinação de Medicamentos , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Neprilisina/antagonistas & inibidores , Domínios Proteicos , Tetrazóis/metabolismo , Valsartana
8.
MAbs ; 7(6): 1151-60, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26284424

RESUMO

Interleukin-1ß (IL-1ß) plays a key role in autoinflammatory diseases, such as systemic juvenile idiopathic arthritis (sJIA) or cryopyrin-associated periodic syndrome (CAPS). Canakinumab, a human monoclonal anti-IL-1ß antibody, was recently approved for human use under the brand name Ilaris®. Canakinumab does not cross-react with IL-1ß from mouse, rat, rabbit, or macaques. The crystal structure of the canakinumab Fab bound to human IL-1ß was determined in an attempt to rationalize the species specificity. The X-ray analysis reveals a complex surface epitope with an intricate network of well-ordered water molecules at the antibody-antigen interface. The canakinumab paratope is largely pre-organized, as demonstrated by the structure determination of the free Fab. Glu 64 of human IL-1ß is a pivotal epitope residue explaining the exquisite species specificity of canakinumab. We identified marmoset as the only non-human primate species that carries Glu 64 in its IL-1ß and demonstrates full cross-reactivity of canakinumab, thereby enabling toxicological studies in this species. As demonstrated by the X-ray structure of the complex with IL-1ß, canakinumab binds IL-1ß on the opposite side with respect to the IL-1RAcP binding site, and in an approximately orthogonal orientation with respect to IL-1RI. However, the antibody and IL-1RI binding sites slightly overlap and the VH region of canakinumab would sterically interfere with the D1 domain of IL-1RI, as shown by a structural overlay with the IL-1ß:IL-1RI complex. Therefore, direct competition with IL-1RI for IL-1ß binding is the molecular mechanism of neutralization by canakinumab, which is also confirmed by competition assays with recombinant IL-1RI and IL-1RII.


Assuntos
Anticorpos Monoclonais/imunologia , Anticorpos Neutralizantes/imunologia , Especificidade de Anticorpos/imunologia , Interleucina-1beta/imunologia , Animais , Anticorpos Monoclonais/química , Anticorpos Monoclonais/metabolismo , Anticorpos Monoclonais Humanizados , Anticorpos Neutralizantes/metabolismo , Ligação Competitiva/imunologia , Callithrix , Reações Cruzadas/imunologia , Cristalografia por Raios X , Epitopos/química , Epitopos/imunologia , Epitopos/metabolismo , Ácido Glutâmico/química , Ácido Glutâmico/imunologia , Ácido Glutâmico/metabolismo , Humanos , Interleucina-1beta/química , Interleucina-1beta/metabolismo , Macaca , Camundongos , Modelos Moleculares , Ligação Proteica/imunologia , Estrutura Terciária de Proteína , Coelhos , Ratos , Receptores de Interleucina-1/imunologia , Receptores de Interleucina-1/metabolismo , Especificidade da Espécie
9.
Bioorg Med Chem Lett ; 25(3): 438-43, 2015 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-25571794

RESUMO

Adenoviral infections are associated with a wide range of acute diseases, among which ocular viral conjunctivitis (EKC) and disseminated disease in immunocompromised patients. To date, no approved specific anti-adenoviral drug is available, but there is a growing need for an effective treatment of such infections. The adenoviral protease, adenain, plays a crucial role for the viral lifecycle and thus represents an attractive therapeutic target. Structure-guided design with the objective to depeptidize tetrapeptide nitrile 1 led to the novel chemotype 2. Optimization of scaffold 2 resulted in picomolar adenain inhibitors 3a and 3b. In addition, a complementary series of irreversible vinyl sulfone containing inhibitors were rationally designed, prepared and evaluated against adenoviral protease. High resolution X-ray co-crystal structures of representatives of each series proves the successful design of these inhibitors and provides an excellent basis for future medicinal chemistry optimization of these compounds.


Assuntos
Adenoviridae/enzimologia , Antivirais/química , Cisteína Endopeptidases/química , Desenho de Fármacos , Inibidores de Proteases/química , Proteínas Virais/antagonistas & inibidores , Adenoviridae/efeitos dos fármacos , Antivirais/metabolismo , Antivirais/toxicidade , Sítios de Ligação , Cristalografia por Raios X , Cisteína Endopeptidases/metabolismo , Células HEK293 , Humanos , Simulação de Acoplamento Molecular , Inibidores de Proteases/metabolismo , Inibidores de Proteases/toxicidade , Ligação Proteica , Estrutura Terciária de Proteína , Relação Estrutura-Atividade , Proteínas Virais/metabolismo
10.
ACS Med Chem Lett ; 5(8): 937-41, 2014 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-25147618

RESUMO

The cysteine protease adenain is the essential protease of adenovirus and, as such, represents a promising target for the treatment of ocular and other adenoviral infections. Through a concise two-pronged hit discovery approach we identified tetrapeptide nitrile 1 and pyrimidine nitrile 2 as complementary starting points for adenain inhibition. These hits enabled the first high-resolution X-ray cocrystal structures of adenain with inhibitors bound and revealed the binding mode of 1 and 2. The screening hits were optimized by a structure-guided medicinal chemistry strategy into low nanomolar drug-like inhibitors of adenain.

11.
J Biol Chem ; 284(13): 8812-21, 2009 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-19153082

RESUMO

p53 tumor suppressor activity is negatively regulated through binding to the oncogenic proteins Hdm2 and HdmX. The p53 residues Leu(26), Trp(23), and Phe(19) are crucial to mediate these interactions. Inhibiting p53 binding to both Hdm2 and HdmX should be a promising clinical approach to reactivate p53 in the cancer setting, but previous studies have suggested that the discovery of dual Hdm2/HdmX inhibitors will be difficult. We have determined the crystal structures at 1.3 A of the N-terminal domain of HdmX bound to two p53 peptidomimetics without and with a 6-chlorine substituent on the indole (which binds in the same subpocket as Trp(23) of p53). The latter compound is the most potent peptide-based antagonist of the p53-Hdm2 interaction yet to be described. The x-ray structures revealed surprising conformational changes of the binding cleft of HdmX, including an "open conformation" of Tyr(99) and unexpected "cross-talk" between the Trp and Leu pockets. Notably, the 6-chloro p53 peptidomimetic bound with high affinity to both HdmX and Hdm2 (K(d) values of 36 and 7 nm, respectively). Our results suggest that the development of potent dual inhibitors for HdmX and Hdm2 should be feasible. They also reveal possible conformational states of HdmX, which should lead to a better prediction of its interactions with potential biological partners.


Assuntos
Materiais Biomiméticos/química , Complexos Multiproteicos/química , Proteínas Nucleares/química , Peptídeos/química , Proteínas Proto-Oncogênicas/química , Proteína Supressora de Tumor p53/química , Sítios de Ligação/fisiologia , Materiais Biomiméticos/metabolismo , Proteínas de Ciclo Celular , Cristalografia por Raios X , Humanos , Complexos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Peptídeos/metabolismo , Ligação Proteica/fisiologia , Estrutura Quaternária de Proteína/fisiologia , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/química , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo
12.
Anal Chem ; 81(1): 408-19, 2009 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19067573

RESUMO

Protein kinases have emerged as a major drug target in the last years. Since more than 500 kinases are encoded in the human genome, cross-reactivity of a majority of kinase inhibitors causes problems. Tools are required for a rapid classification of inhibitors according to their affinity for a certain target to refine the search for new, more specific lead compounds. Mass spectrometry (MS) is increasingly used in pharmaceutical research and drug discovery to investigate protein-ligand interactions and determination of binding affinities. We present a comparison of different existing nanoelectrospray-MS based methods to quantify binding affinities and qualitatively rank, by competitive experiments, the affinity of several clinical inhibitors. We also present a new competitive method which is derived from our previous work for quantitative assessment of binding strengths (Wortmann et al., J. Mass Spectrom. 2008, 43(5), 600-608). The human kinases studied for this purpose were p38alpha (MAPK14) and LCK (lymphocyte specific kinase), and their interaction with 17 known small molecule kinase inhibitors was probed. Moreover, we present a new method to differentiate type I from type II inhibitors (Liu, Y.; Gray, N. S. Nat. Chem. Biol. 2006, 2(7), 358-364) based on a kinetic experiment with direct MS read-out of the noncovalent complex between the human kinase and the inhibitor. This method was successfully applied to p38alpha binding to BIRB796, as well as to a BIRB796 analogue. Quantitative determination of the binding strength is also described. The results of our competitive experiments for the affinity classification of different inhibitors, as well as the results for the kinetic study, are in good agreement with IC(50) measurements and data found in the literature.


Assuntos
Proteína Tirosina Quinase p56(lck) Linfócito-Específica/antagonistas & inibidores , Proteína Quinase 14 Ativada por Mitógeno/antagonistas & inibidores , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Espectrometria de Massas por Ionização por Electrospray/métodos , Ligação Competitiva , Humanos , Concentração Inibidora 50 , Cinética , Proteína Tirosina Quinase p56(lck) Linfócito-Específica/química , Proteína Quinase 14 Ativada por Mitógeno/química , Nanotecnologia/instrumentação , Nanotecnologia/métodos , Proteômica/instrumentação , Proteômica/métodos , Reprodutibilidade dos Testes , Espectrometria de Massas por Ionização por Electrospray/instrumentação
13.
Protein Expr Purif ; 64(2): 185-93, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19038347

RESUMO

The recombinant expression of eukaryotic proteins in Escherichia coli often results in protein aggregation. Several articles report on improved solubility and increased purification yields of individual proteins upon over-expression of E. coli chaperones but this effect might potentially be protein-specific. To find out whether chaperone over-expression is a generally applicable strategy for the production of human protein kinases in E. coli, we analyzed 10 kinases, mainly as catalytic domain constructs. The kinases studied, namely c-Src, c-Abl, Hck, Lck, Igf1R, InsR, KDR, c-Met, b-Raf and Irak4, belong to the tyrosine and tyrosine kinase-like groups of kinases. Upon over-expression of the E. coli chaperones DnaK/DnaJ/GrpE and GroEL/GroES, the yields of 7 from 10 polyhistidine-tagged kinases were increased up to 5-fold after nickel-affinity purification (IMAC). Additive over-expression of the chaperones ClpB and/or trigger factor showed no further improvement. Co-purification of DnaJ and GroEL indicated incomplete kinase folding, therefore, the oligomerization state of the kinases was determined by size-exclusion chromatography. In our study, kinases behave in three different ways. Kinases where yields are not affected by E. coli chaperone over-expression e.g. c-Src elute in the monomeric fraction (category I). Although IMAC yields increase upon chaperone over-expression, InsR and b-Raf kinase are present as soluble aggregates (category II). Igf1R and c-Met kinase catalytic domains are partially complexed with E. coli chaperones upon over-expression; however, they show approximately 2-fold increased yields of monomer (category III). Together, our results suggest that the benefits of chaperone over-expression on the production of protein kinases in E. coli are indeed case-specific.


Assuntos
Escherichia coli/metabolismo , Chaperonas Moleculares/biossíntese , Fosfotransferases/biossíntese , Proteínas Recombinantes/biossíntese , Cromatografia em Gel , Escherichia coli/genética , Espectrometria de Massas , Chaperonas Moleculares/genética , Chaperonas Moleculares/isolamento & purificação , Fosfotransferases/genética , Fosfotransferases/isolamento & purificação , Engenharia de Proteínas , Dobramento de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/genética , Proteínas Recombinantes/isolamento & purificação , Solubilidade
14.
J Mol Biol ; 380(5): 828-43, 2008 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-18589439

RESUMO

The guanine nucleotide exchange factor (GEF) Vav1 plays an important role in T-cell activation and tumorigenesis. In the GEF superfamily, Vav1 has the ability to interact with multiple families of Rho GTPases. The structure of the Vav1 DH-PH-CRD/Rac1 complex to 2.6 A resolution reveals a unique intramolecular network of contacts between the Vav1 cysteine-rich domain (CRD) and the C-terminal helix of the Vav1 Dbl homology (DH) domain. These unique interactions stabilize the Vav1 DH domain for its intimate association with the Switch II region of Rac1 that is critical for the displacement of the guanine nucleotide. Small angle x-ray scattering (SAXS) studies support this domain arrangement for the complex in solution. Further, mutational analyses confirms that the atypical CRD is critical for maintaining both optimal guanine nucleotide exchange activity and broader specificity of Vav family GEFs. Taken together, the data outline the detailed nature of Vav1's ability to contact a range of Rho GTPases using a novel protein-protein interaction network.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/química , Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteínas Proto-Oncogênicas c-vav/metabolismo , Linfócitos T/química , Alanina/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Cristalografia por Raios X , Ativação Enzimática , Fatores de Troca do Nucleotídeo Guanina/genética , Humanos , Ligação de Hidrogênio , Modelos Moleculares , Dados de Sequência Molecular , Peso Molecular , Conformação Proteica , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-vav/química , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espalhamento a Baixo Ângulo , Homologia de Sequência de Aminoácidos , Solubilidade , Difração de Raios X , Proteínas rac1 de Ligação ao GTP/química , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteínas rho de Ligação ao GTP/metabolismo
15.
Top Curr Chem ; 273: 1-14, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-23605457

RESUMO

Key processes in molecular biology are regulated by interactions between biomolecules. Protein-proteinand protein-ligand interactions, e.g., in signal transduction pathways, rely on the subtle interactionsbetween atoms at the binding interface of the involved molecules. Because biomolecules often havemany interacting partners, these interactions are not necessarily strong. The study of molecularrecognition gives insight into the complex network of signaling in life and is the basis of structure-baseddrug design.In the situation where the interaction is weak, one of the traditional methods that can be appliedto obtain structural information (internuclear distances) of the bound ligand is the so-called transferredNOE (trNOE) method. Recently, it became possible to use transferred cross-correlated relaxation (trCCR)to directly measure dihedral angles. The combined use of these two techniques significantly improvesthe precision of the structure determination of ligands weakly bound to macromolecules.The application of these techniques will be discussed in detail for a peptide derived fromIKKß bound to the protein NEMO that plays an important rolein the NFκB pathway.

16.
Protein Expr Purif ; 53(1): 51-62, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17275330

RESUMO

The signal transduction pathway involving the Vav1 guanine nucleotide exchange factor (GEF) and the Rac1 GTPase plays several key roles in the immune response mediated by the T cell receptor. Vav1 is also a unique member of the GEF family in that it contains a cysteine-rich domain (CRD) that is critical for Rac1 binding and maximal guanine nucleotide exchange activity, and thus may provide a unique protein-protein interface compared to other GEF/GTPase pairs. Here, we have applied a number of remedial structural proteomics strategies, such as construct and expression optimization, surface mutagenesis, limited proteolysis, and protein formulation to successfully express, purify, and crystallize the Vav1-DH-PH-CRD/Rac1 complex in an active conformation. We have also systematically characterized various Vav1 domains in a GEF assay and Rac1 in vitro binding experiments. In the context of Vav1-DH-PH-CRD, the zinc finger motif of the CRD is required for the expression of stable Vav1, as well as for activity in both a GEF assay and in vitro formation of a Vav1/Rac1 complex suitable for biophysical and structural characterization. Our data also indicate that the isolated CRD maintains a low level of specific binding to Rac1, appears to be folded based on 1D NMR analysis and coordinates two zinc ions based on ICP-MS analysis. The protein reagents generated here are essential tools for the determination of a three dimensional Vav1/Rac1 complex crystal structure and possibly for the identification of inhibitors of the Vav1/Rac1 protein-protein interaction with potential to inhibit lymphocyte activation.


Assuntos
Fatores de Troca do Nucleotídeo Guanina/metabolismo , Proteômica , Proteínas Proto-Oncogênicas c-vav/isolamento & purificação , Proteínas Proto-Oncogênicas c-vav/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Sequência de Aminoácidos , Clonagem Molecular , Cristalização , Cisteína/química , DNA Complementar , Glutationa Transferase/metabolismo , Fatores de Troca do Nucleotídeo Guanina/análise , Hidrólise , Cinética , Dados de Sequência Molecular , Mutagênese , Nanotecnologia , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica , Conformação Proteica , Dobramento de Proteína , Estrutura Terciária de Proteína , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-vav/química , Proteínas Proto-Oncogênicas c-vav/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/isolamento & purificação , Proteínas Recombinantes/metabolismo , Zinco/química
17.
ChemMedChem ; 1(2): 267-73, 2006 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-16892359

RESUMO

To understand the structural basis for bisphosphonate therapy of bone diseases, we solved the crystal structures of human farnesyl pyrophosphate synthase (FPPS) in its unliganded state, in complex with the nitrogen-containing bisphosphonate (N-BP) drugs zoledronate, pamidronate, alendronate, and ibandronate, and in the ternary complex with zoledronate and the substrate isopentenyl pyrophosphate (IPP). By revealing three structural snapshots of the enzyme catalytic cycle, each associated with a distinct conformational state, and details about the interactions with N-BPs, these structures provide a novel understanding of the mechanism of FPPS catalysis and inhibition. In particular, the accumulating substrate, IPP, was found to bind to and stabilize the FPPS-N-BP complexes rather than to compete with and displace the N-BP inhibitor. Stabilization of the FPPS-N-BP complex through IPP binding is supported by differential scanning calorimetry analyses of a set of representative N-BPs. Among other factors such as high binding affinity for bone mineral, this particular mode of FPPS inhibition contributes to the exceptional in vivo efficacy of N-BP drugs. Moreover, our data form the basis for structure-guided design of optimized N-BPs with improved pharmacological properties.


Assuntos
Difosfonatos/química , Difosfonatos/farmacologia , Varredura Diferencial de Calorimetria , Cristalografia por Raios X , Humanos , Modelos Moleculares , Estrutura Molecular
18.
J Med Chem ; 48(16): 5175-90, 2005 Aug 11.
Artigo em Inglês | MEDLINE | ID: mdl-16078837

RESUMO

Molecular modeling based on the X-ray crystal structure of the Tang-Ghosh heptapeptide inhibitor 1 (OM99-2) of BACE led to the design and synthesis of a series of constrained P(1)' analogues. A cyclopentane ring was incorporated in 1 spanning the P(1)' Ala methyl group and the adjacent methylene carbon atom of the chain. Progressive truncation at the P(2)'-P(4)' sites led to a potent truncated analogue 5 with good selectivity over Cathepsin D. Using the same backbone replacement concept, a series of cyclopentane, cyclopentanone, tetrahydrofuran, pyrrolidine, and pyrrolidinone analogues were synthesized with considerable variation at the P and P' sites. The cyclopentanone and 2-pyrrolidinone analogues 45 and 57 showed low nM BACE inhibition. X-ray cocrystal structures of two analogues 5 and 45 revealed excellent convergence with the original inhibitor 1 structure while providing new insights into other interactions which could be exploited for future modifications.


Assuntos
Ácido Aspártico Endopeptidases/antagonistas & inibidores , Ciclopentanos/síntese química , Furanos/síntese química , Peptídeos/química , Pirrolidinas/síntese química , Secretases da Proteína Precursora do Amiloide , Ácido Aspártico Endopeptidases/química , Sítios de Ligação , Catepsina D/antagonistas & inibidores , Cristalografia por Raios X , Ciclopentanos/química , Endopeptidases , Furanos/química , Humanos , Modelos Moleculares , Mimetismo Molecular , Pirrolidinas/química , Pirrolidinonas/síntese química , Pirrolidinonas/química , Relação Estrutura-Atividade
19.
J Med Chem ; 48(2): 364-79, 2005 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-15658851

RESUMO

We disclose herein the discovery of estrogen receptor alpha (ERalpha) selective estrogen receptor modulators (SERMs) of the tetrahydroisoquinoline series that incorporate novel conformationally restricted side chains as replacement of the aminoethoxy residue typical of SERMs. Molecular modeling studies used in conjunction with the X-ray crystal structure of the ERalpha ligand binding domain (LBD) with raloxifene (7) suggested a diazadecaline moiety as a viable mimic of the SERM side chain. On the basis of this knowledge, the piperidinylethoxy moiety of our lead compound 60 was replaced by a diazadecaline subunit, providing the novel tetrahydroisoquinoline 29. In addition to exhibiting a binding affinity to ERalpha and antagonistic properties in the estrogen response element and MCF-7 assays similar to those of the parent compound 60, ligand 29 showed a reduced agonist behavior in the MCF-7 assay in the absence of 17beta-estradiol. These data point toward the fact that 29 may have a potential for breast cancer prevention/treatment in vivo, a feature which is particularly attractive in the quest for safe alternatives to hormone replacement therapy. In a pharmacokinetic experiment carried out in rats, 29 displayed an interesting profile, with a bioavailability of 49%. We also disclose the X-ray crystal structure of 29 in complex with ERalpha-LBD, which reveals the preferred configurations of 29 at the two chiral centers and the details of its interactions with the receptor. Finally, our structure-activity relationship studies show that other analogues bearing constrained side chains retain potency and antagonist activity and that a 3-OH substituted phenyl D-ring increases the selectivity of a set of piperazinyl-containing ligands in favor of ERalpha over ERbeta.


Assuntos
Receptor alfa de Estrogênio/efeitos dos fármacos , Isoquinolinas/síntese química , Moduladores Seletivos de Receptor Estrogênico/síntese química , Tetra-Hidroisoquinolinas/síntese química , Animais , Disponibilidade Biológica , Linhagem Celular Tumoral , Cristalografia por Raios X , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Humanos , Concentração Inibidora 50 , Isoquinolinas/química , Isoquinolinas/farmacologia , Ligantes , Modelos Moleculares , Piperazinas/síntese química , Piperazinas/química , Piperazinas/farmacologia , Piperidinas/síntese química , Piperidinas/química , Piperidinas/farmacologia , Ensaio Radioligante , Ratos , Moduladores Seletivos de Receptor Estrogênico/química , Moduladores Seletivos de Receptor Estrogênico/farmacologia , Relação Estrutura-Atividade , Tetra-Hidroisoquinolinas/química , Tetra-Hidroisoquinolinas/farmacologia
20.
Protein Sci ; 12(4): 708-16, 2003 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-12649429

RESUMO

Protein aggregation is commonly observed during protein refolding. To better understand this phenomenon, the intermolecular interactions experienced by a protein during unfolding and refolding are inferred from second virial coefficient (SVC) measurements. It is accepted that a negative SVC is indicative of protein-protein interactions that are attractive, whereas a positive SVC indicates net repulsive interactions. Lysozyme denatured and reduced in guanidinium hydrochloride exhibited a decreasing SVC as the denaturant was diluted, and the SVC approached zero at approximately 3 M GdnHCl. Further dilution of denaturant to renaturation conditions (1.25 M GdnHCl) led to a negative SVC, and significant protein aggregation was observed. The inclusion of 500 mM L-arginine in the renaturation buffer shifted the SVC to positive and suppressed aggregation, thereby increasing refolding yield. The formation of mixed disulfides in the denatured state prior to refolding also increased protein solubility and suppressed aggregation, even without the use of L-arginine. Again, the suppression of aggregation was shown to be caused by a shift from attractive to repulsive intermolecular interactions as reflected in a shift from a negative to a positive SVC value. To the best of our knowledge, this is the first time that SVC data have been reported for renaturation studies. We believe this technique will aid in our understanding of how certain conditions promote renaturation and increase protein solubility, thereby suppressing aggregation. SVC measurements provide a useful link, for protein folding and aggregation, between empirical observation and thermodynamics.


Assuntos
Polímeros/metabolismo , Renaturação Proteica , Animais , Galinhas/metabolismo , Relação Dose-Resposta a Droga , Proteínas do Ovo/metabolismo , Dissulfeto de Glutationa/metabolismo , Guanidina/farmacologia , Muramidase/metabolismo , Desnaturação Proteica/efeitos dos fármacos , Solubilidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...