Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Immunol Rev ; 313(1): 339-357, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36217774

RESUMEN

Dysregulation of the alternative complement pathway predisposes individuals to a number of diseases. It can either be evoked by genetic alterations in or by stabilizing antibodies to important pathway components and typically leads to severe diseases such as paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, C3 glomerulopathy, and age-related macular degeneration. In addition, the alternative pathway may also be involved in many other diseases where its amplifying function for all complement pathways might play a role. To identify specific alternative pathway inhibitors that qualify as therapeutics for these diseases, drug discovery efforts have focused on the two central proteases of the pathway, factor B and factor D. Although drug discovery has been challenging for a number of reasons, potent and selective low-molecular weight (LMW) oral inhibitors have now been discovered for both proteases and several molecules are in clinical development for multiple complement-mediated diseases. While the clinical development of these inhibitors initially focuses on diseases with systemic and/or peripheral tissue complement activation, the availability of LMW inhibitors may also open up the prospect of inhibiting complement in the central nervous system where its activation may also play an important role in several neurodegenerative diseases.


Asunto(s)
Síndrome Hemolítico Urémico Atípico , Vía Alternativa del Complemento , Humanos , Peso Molecular , Proteínas del Sistema Complemento/metabolismo , Activación de Complemento , Factor D del Complemento/metabolismo , Síndrome Hemolítico Urémico Atípico/metabolismo
2.
J Mol Biol ; 433(24): 167309, 2021 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-34687713

RESUMEN

The NLRP3 inflammasome assembles in response to a variety of pathogenic and sterile danger signals, resulting in the production of interleukin-1ß and interleukin-18. NLRP3 is a key component of the innate immune system and has been implicated as a driver of a number of acute and chronic diseases. We report the 2.8 Å crystal structure of the NLRP3 NACHT domain in complex with an inhibitor. The structure defines a binding pocket formed by the four subdomains of the NACHT domain, and shows the inhibitor acts as an intramolecular glue, which locks the protein in an inactive conformation. It provides further molecular insight into our understanding of NLRP3 activation, helps to detail the residues involved in subdomain coordination within the NLRP3 NACHT domain, and gives molecular insights into how gain-of-function mutations de-stabilize the inactive conformation of NLRP3. Finally, it suggests stabilizing the auto-inhibited form of the NACHT domain is an effective way to inhibit NLRP3, and will aid the structure-based development of NLRP3 inhibitors for a range of inflammatory diseases.


Asunto(s)
Inflamasomas/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteína con Dominio Pirina 3 de la Familia NLR/química , Sitios de Unión , Dominio Catalítico , Cristalografía por Rayos X , Furanos/química , Furanos/farmacología , Humanos , Indenos/química , Indenos/farmacología , Inflamasomas/metabolismo , Dominios Proteicos , Sulfonamidas/química , Sulfonamidas/farmacología
3.
Cancer Immunol Res ; 9(7): 790-810, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33990375

RESUMEN

T-cell activation and expansion in the tumor microenvironment (TME) are critical for antitumor immunity. Neutrophils in the TME acquire a complement-dependent T-cell suppressor phenotype that is characterized by inhibition of T-cell proliferation and activation through mechanisms distinct from those of myeloid-derived suppressor cells. In this study, we used ascites fluid supernatants (ASC) from patients with ovarian cancer as an authentic component of the TME to evaluate the effects of ASC on neutrophil function and mechanisms for neutrophil-driven immune suppression. ASC prolonged neutrophil life span, decreased neutrophil density, and induced nuclear hypersegmentation. Mass cytometry analysis showed that ASC induced 15 distinct neutrophil clusters. ASC stimulated complement deposition and signaling in neutrophils, resulting in surface mobilization of granule constituents, including NADPH oxidase. NADPH oxidase activation and phosphatidylserine signaling were required for neutrophil suppressor function, although we did not observe a direct role of extracellular reactive oxygen species in inhibiting T-cell proliferation. Postoperative surgical drainage fluid also induced a complement-dependent neutrophil suppressor phenotype, pointing to this effect as a general response to injury. Like circulating lymphocytes, ASC-activated neutrophils caused complement-dependent suppression of tumor-associated lymphocytes. ASC-activated neutrophils adhered to T cells and caused trogocytosis of T-cell membranes. These injury and signaling cues resulted in T-cell immunoparalysis characterized by impaired NFAT translocation, IL2 production, glucose uptake, mitochondrial function, and mTOR activation. Our results demonstrate that complement-dependent priming of neutrophil effector functions in the TME induces a T-cell nonresponsiveness distinct from established checkpoint pathways and identify targets for immunotherapy.See related Spotlight by Cassatella, p. 725.


Asunto(s)
Neutrófilos/inmunología , Neoplasias Ováricas/inmunología , Linfocitos T/inmunología , Trogocitosis/inmunología , Escape del Tumor , Adulto , Células Cultivadas , Femenino , Humanos , Activación de Linfocitos , Persona de Mediana Edad , Activación Neutrófila , Neutrófilos/metabolismo , Neoplasias Ováricas/sangre , Neoplasias Ováricas/patología , Neoplasias Ováricas/cirugía , Cultivo Primario de Células , Microambiente Tumoral/inmunología , Adulto Joven
4.
Biochem Biophys Res Commun ; 545: 177-182, 2021 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-33561652

RESUMEN

The NLRP3 inflammasome is a critical component of the innate immune response to sterile inflammation. Its regulation involves a priming step, required for up-regulation of inflammasome protagonists and an activation step leading to NLRP3 inflammasome complex assembly, which triggers caspase-1 activity. The IκKß kinase regulates canonical NF-κB, a key pathway involved in transcriptional priming. We found that IκKß also regulates the activation and function of the NLRP3 inflammasome beyond the priming step. Two unrelated IκKß inhibitors, AFN700 and TPCA-1, when applied after priming, fully blocked IL-1ß secretion triggered by nigericin in THP-1 cells. Both inhibitors prevented neither inflammasome assembly, as monitored by measuring the formation of ASC specks, nor the generation of caspase-1 p20, a hallmark of caspase-1 activity, but they impaired the initial cleavage and activation of procaspase-1. These data thus indicate that IκKß activity is required for efficient activation of NLRP3, suggesting that IκKß may fulfill a dual role in coupling priming and activation of the NLRP3 inflammasome.


Asunto(s)
Quinasa I-kappa B/antagonistas & inhibidores , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Amidas/farmacología , Caspasa 1/metabolismo , Humanos , Inmunidad Innata/efectos de los fármacos , Inflamasomas/efectos de los fármacos , Inflamasomas/inmunología , Interleucina-1beta/biosíntesis , FN-kappa B/metabolismo , Nigericina/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Células THP-1 , Tiofenos/farmacología
5.
J Med Chem ; 63(11): 5697-5722, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32073845

RESUMEN

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.


Asunto(s)
Ácido Benzoico/química , Factor B del Complemento/antagonistas & inhibidores , Indoles/química , Síndrome Hemolítico Urémico Atípico/metabolismo , Síndrome Hemolítico Urémico Atípico/patología , Ácido Benzoico/metabolismo , Ácido Benzoico/farmacocinética , Sitios de Unión , Dominio Catalítico , Factor B del Complemento/metabolismo , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Semivida , Humanos , Indoles/metabolismo , Indoles/farmacocinética , Concentración 50 Inhibidora , Degeneración Macular/metabolismo , Degeneración Macular/patología , Simulación de Dinámica Molecular , Relación Estructura-Actividad
6.
Proc Natl Acad Sci U S A ; 116(16): 7926-7931, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30926668

RESUMEN

Dysregulation of the alternative complement pathway (AP) predisposes individuals to a number of diseases including paroxysmal nocturnal hemoglobinuria, atypical hemolytic uremic syndrome, and C3 glomerulopathy. Moreover, glomerular Ig deposits can lead to complement-driven nephropathies. Here we describe the discovery of a highly potent, reversible, and selective small-molecule inhibitor of factor B, a serine protease that drives the central amplification loop of the AP. Oral administration of the inhibitor prevents KRN-induced arthritis in mice and is effective upon prophylactic and therapeutic dosing in an experimental model of membranous nephropathy in rats. In addition, inhibition of factor B prevents complement activation in sera from C3 glomerulopathy patients and the hemolysis of human PNH erythrocytes. These data demonstrate the potential therapeutic value of using a factor B inhibitor for systemic treatment of complement-mediated diseases and provide a basis for its clinical development.


Asunto(s)
Factor B del Complemento/antagonistas & inhibidores , Vía Alternativa del Complemento/efectos de los fármacos , Descubrimiento de Drogas/métodos , Factores Inmunológicos/farmacología , Animales , Modelos Animales de Enfermedad , Glomerulonefritis Membranosa/fisiopatología , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratas Sprague-Dawley
7.
Blood ; 133(13): 1507-1516, 2019 03 28.
Artículo en Inglés | MEDLINE | ID: mdl-30692123

RESUMEN

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.


Asunto(s)
Anticuerpos Monoclonales Humanizados/uso terapéutico , Anticoagulantes/uso terapéutico , Coagulación Sanguínea/efectos de los fármacos , Factor XI/antagonistas & inhibidores , Trombosis/tratamiento farmacológico , Adolescente , Adulto , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticoagulantes/farmacología , Femenino , Humanos , Inmunoglobulina G/farmacología , Inmunoglobulina G/uso terapéutico , Macaca fascicularis , Masculino , Ratones Endogámicos C57BL , Persona de Mediana Edad , Simulación del Acoplamiento Molecular , Trombosis/sangre , Adulto Joven
8.
Nat Rev Drug Discov ; 16(8): 531-543, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28685762

RESUMEN

Phenotypic drug discovery (PDD) approaches do not rely on knowledge of the identity of a specific drug target or a hypothesis about its role in disease, in contrast to the target-based strategies that have been widely used in the pharmaceutical industry in the past three decades. However, in recent years, there has been a resurgence in interest in PDD approaches based on their potential to address the incompletely understood complexity of diseases and their promise of delivering first-in-class drugs, as well as major advances in the tools for cell-based phenotypic screening. Nevertheless, PDD approaches also have considerable challenges, such as hit validation and target deconvolution. This article focuses on the lessons learned by researchers engaged in PDD in the pharmaceutical industry and considers the impact of 'omics' knowledge in defining a cellular disease phenotype in the era of precision medicine, introducing the concept of a chain of translatability. We particularly aim to identify features and areas in which PDD can best deliver value to drug discovery portfolios and can contribute to the identification and the development of novel medicines, and to illustrate the challenges and uncertainties that are associated with PDD in order to help set realistic expectations with regard to its benefits and costs.


Asunto(s)
Diseño de Fármacos , Descubrimiento de Drogas/métodos , Industria Farmacéutica/métodos , Animales , Humanos , Terapia Molecular Dirigida , Fenotipo , Medicina de Precisión/métodos
9.
Nat Chem Biol ; 12(12): 1105-1110, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27775713

RESUMEN

Complement is a key component of the innate immune system, recognizing pathogens and promoting their elimination. Complement component 3 (C3) is the central component of the system. Activation of C3 can be initiated by three distinct routes-the classical, the lectin and the alternative pathways-with the alternative pathway also acting as an amplification loop for the other two pathways. The protease factor D (FD) is essential for this amplification process, which, when dysregulated, predisposes individuals to diverse disorders including age-related macular degeneration and paroxysmal nocturnal hemoglobinuria (PNH). Here we describe the identification of potent and selective small-molecule inhibitors of FD. These inhibitors efficiently block alternative pathway (AP) activation and prevent both C3 deposition onto, and lysis of, PNH erythrocytes. Their oral administration inhibited lipopolysaccharide-induced AP activation in FD-humanized mice. These data demonstrate the feasibility of inhibiting the AP with small-molecule antagonists and support the development of FD inhibitors for the treatment of complement-mediated diseases.


Asunto(s)
Factor D del Complemento/antagonistas & inhibidores , Vía Alternativa del Complemento/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Factor D del Complemento/metabolismo , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/química , Humanos , Lipopolisacáridos/antagonistas & inhibidores , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Modelos Moleculares , Estructura Molecular , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
10.
Nat Commun ; 7: 13166, 2016 10 24.
Artículo en Inglés | MEDLINE | ID: mdl-27774986

RESUMEN

The COP9 signalosome (CSN) is a central component of the activation and remodelling cycle of cullin-RING E3 ubiquitin ligases (CRLs), the largest enzyme family of the ubiquitin-proteasome system in humans. CRLs are implicated in the regulation of numerous cellular processes, including cell cycle progression and apoptosis, and aberrant CRL activity is frequently associated with cancer. Remodelling of CRLs is initiated by CSN-catalysed cleavage of the ubiquitin-like activator NEDD8 from CRLs. Here we describe CSN5i-3, a potent, selective and orally available inhibitor of CSN5, the proteolytic subunit of CSN. The compound traps CRLs in the neddylated state, which leads to inactivation of a subset of CRLs by inducing degradation of their substrate recognition module. CSN5i-3 differentially affects the viability of tumour cell lines and suppresses growth of a human xenograft in mice. Our results provide insights into how CSN regulates CRLs and suggest that CSN5 inhibition has potential for anti-tumour therapy.


Asunto(s)
Antineoplásicos/farmacología , Azepinas/farmacología , Complejo del Señalosoma COP9/antagonistas & inhibidores , Regulación Neoplásica de la Expresión Génica , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Linfoma Anaplásico de Células Grandes/tratamiento farmacológico , Pirazoles/farmacología , Ubiquitina-Proteína Ligasas/genética , Animales , Antineoplásicos/síntesis química , Azepinas/síntesis química , Complejo del Señalosoma COP9/genética , Complejo del Señalosoma COP9/metabolismo , Femenino , Células HCT116 , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Linfoma Anaplásico de Células Grandes/genética , Linfoma Anaplásico de Células Grandes/metabolismo , Linfoma Anaplásico de Células Grandes/patología , Ratones , Ratones SCID , Terapia Molecular Dirigida , Proteína NEDD8/genética , Proteína NEDD8/metabolismo , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo , Procesamiento Proteico-Postraduccional , Proteolisis/efectos de los fármacos , Pirazoles/síntesis química , Células THP-1 , Carga Tumoral/efectos de los fármacos , Ubiquitina-Proteína Ligasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Handb Exp Pharmacol ; 232: 3-22, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26330257

RESUMEN

Drugs discovered by the pharmaceutical industry over the past 100 years have dramatically changed the practice of medicine and impacted on many aspects of our culture. For many years, drug discovery was a target- and mechanism-agnostic approach that was based on ethnobotanical knowledge often fueled by serendipity. With the advent of modern molecular biology methods and based on knowledge of the human genome, drug discovery has now largely changed into a hypothesis-driven target-based approach, a development which was paralleled by significant environmental changes in the pharmaceutical industry. Laboratories became increasingly computerized and automated, and geographically dispersed research sites are now more and more clustered into large centers to capture technological and biological synergies. Today, academia, the regulatory agencies, and the pharmaceutical industry all contribute to drug discovery, and, in order to translate the basic science into new medical treatments for unmet medical needs, pharmaceutical companies have to have a critical mass of excellent scientists working in many therapeutic fields, disciplines, and technologies. The imperative for the pharmaceutical industry to discover breakthrough medicines is matched by the increasing numbers of first-in-class drugs approved in recent years and reflects the impact of modern drug discovery approaches, technologies, and genomics.


Asunto(s)
Descubrimiento de Drogas/tendencias , Genoma Humano , Humanos
12.
Nat Rev Drug Discov ; 13(8): 577-87, 2014 08.
Artículo en Inglés | MEDLINE | ID: mdl-25033734

RESUMEN

Analysis of the origins of new drugs approved by the US Food and Drug Administration (FDA) from 1999 to 2008 suggested that phenotypic screening strategies had been more productive than target-based approaches in the discovery of first-in-class small-molecule drugs. However, given the relatively recent introduction of target-based approaches in the context of the long time frames of drug development, their full impact might not yet have become apparent. Here, we present an analysis of the origins of all 113 first-in-class drugs approved by the FDA from 1999 to 2013, which shows that the majority (78) were discovered through target-based approaches (45 small-molecule drugs and 33 biologics). In addition, of 33 drugs identified in the absence of a target hypothesis, 25 were found through a chemocentric approach in which compounds with known pharmacology served as the starting point, with only eight coming from what we define here as phenotypic screening: testing a large number of compounds in a target-agnostic assay that monitors phenotypic changes. We also discuss the implications for drug discovery strategies, including viewing phenotypic screening as a novel discipline rather than as a neoclassical approach.


Asunto(s)
Descubrimiento de Drogas/métodos , Aprobación de Drogas/historia , Descubrimiento de Drogas/historia , Evaluación Preclínica de Medicamentos/historia , Evaluación Preclínica de Medicamentos/métodos , Industria Farmacéutica/métodos , Historia del Siglo XX , Historia del Siglo XXI , Humanos , Biología de Sistemas/métodos
13.
Proc Natl Acad Sci U S A ; 108(52): 21052-6, 2011 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-22160684

RESUMEN

Hepatitis C virus (HCV) infection is a global health burden with over 170 million people infected worldwide. In a significant portion of patients chronic hepatitis C infection leads to serious liver diseases, including fibrosis, cirrhosis, and hepatocellular carcinoma. The HCV NS3 protein is essential for viral polyprotein processing and RNA replication and hence viral replication. It is composed of an N-terminal serine protease domain and a C-terminal helicase/NTPase domain. For full activity, the protease requires the NS4A protein as a cofactor. HCV NS3/4A protease is a prime target for developing direct-acting antiviral agents. First-generation NS3/4A protease inhibitors have recently been introduced into clinical practice, markedly changing HCV treatment options. To date, crystal structures of HCV NS3/4A protease inhibitors have only been reported in complex with the protease domain alone. Here, we present a unique structure of an inhibitor bound to the full-length, bifunctional protease-helicase NS3/4A and show that parts of the P4 capping and P2 moieties of the inhibitor interact with both protease and helicase residues. The structure sheds light on inhibitor binding to the more physiologically relevant form of the enzyme and supports exploring inhibitor-helicase interactions in the design of the next generation of HCV NS3/4A protease inhibitors. In addition, small angle X-ray scattering confirmed the observed protease-helicase domain assembly in solution.


Asunto(s)
Proteínas Portadoras/química , Hepatitis C/tratamiento farmacológico , Modelos Moleculares , Inhibidores de Proteasas/química , Conformación Proteica , Proteínas no Estructurales Virales/química , Proteínas Portadoras/metabolismo , Cromatografía en Gel , Cristalización , Cristalografía por Rayos X , Escherichia coli , Concentración 50 Inhibidora , Péptidos y Proteínas de Señalización Intracelular , Inhibidores de Proteasas/metabolismo , Inhibidores de Proteasas/uso terapéutico , Dispersión del Ángulo Pequeño , Proteínas no Estructurales Virales/metabolismo
14.
Cancer Res ; 69(11): 4573-6, 2009 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-19487280

RESUMEN

ADAMs (a disintegrin and metalloproteinase) have important roles in development and diseases such as cancer. Previously, an ADAM15 splice variant (ADAM15B), which contains an inserted cytoplasmic Src-binding site, was linked to clinical aggressiveness in breast cancer, yet little was known about how this splice variant affects the function of ADAM15. Here, we show that ADAM15B has enhanced catalytic activity in cell-based assays compared with ADAM15A, which lacks a Src-binding site, using shedding of fibroblast growth factor receptor 2iiib variant as an assay for catalytic activity. Moreover, the enhanced activity of ADAM15B compared with ADAM15A depends on Src because it is abolished by Src-kinase inhibitors and in Src(-/-) cells, but not in Src(-/-) cells rescued with Src. These findings provide insights into the mechanism of how a splice variant linked to clinical agressiveness in breast cancer causes increased activity of ADAM15B, and suggest that inhibitors of the ADAM15 protease activity or of the interaction of ADAM15B with Src could be useful to treat breast cancer in patients with dysregulated ADAM15B.


Asunto(s)
Proteínas ADAM/metabolismo , Neoplasias de la Mama/metabolismo , Genes src/fisiología , Proteínas de la Membrana/metabolismo , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/metabolismo , Proteínas ADAM/genética , Proteínas ADAM/fisiología , Secuencia de Aminoácidos , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Células COS , Dominio Catalítico/fisiología , Células Cultivadas , Chlorocebus aethiops , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/fisiología , Ratones , Datos de Secuencia Molecular , Invasividad Neoplásica , Proteína Oncogénica pp60(v-src)/metabolismo , Unión Proteica , Isoformas de Proteínas/metabolismo , Isoformas de Proteínas/fisiología , Receptor Tipo 2 de Factor de Crecimiento de Fibroblastos/química , Homología de Secuencia de Aminoácido
15.
Curr Pharm Des ; 13(3): 271-85, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17313361

RESUMEN

Aspartic proteases are the smallest class of human proteases with only 15 members. Over the past years, they have received considerable attention as potential targets for pharmaceutical intervention since many have been shown to play important roles in physiological and pathological processes. Despite numerous efforts, however, the only inhibitors for aspartic proteases currently on the market are directed against the HIV protease, an aspartic protease of viral origin. Nevertheless, several inhibitors including those targeting renin, BACE1 and gamma-secretase are in clinical or preclinical development, and some other aspartic proteases are discussed as potential drug target. The crystal structures of seven human aspartic proteases have now been solved and, together with a detailed kinetic understanding of their catalytic mechanism, this has greatly contributed to the design and discovery of novel inhibitors for this protease class. This review describes current aspartic protease drug targets and summarizes the drug discovery efforts in this field. In addition, it highlights recent developments which may lead to a new generation of aspartic protease inhibitors.


Asunto(s)
Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/química , Diseño de Fármacos , Inhibidores de Proteasas/química , Inhibidores de Proteasas/farmacología , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/química , Animales , Ácido Aspártico Endopeptidasas/metabolismo , Diseño Asistido por Computadora , Proteasa del VIH/química , Inhibidores de la Proteasa del VIH/farmacología , Humanos , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Estructura Molecular , Presenilinas/antagonistas & inhibidores , Presenilinas/química , Conformación Proteica , Renina/antagonistas & inhibidores , Renina/química , Relación Estructura-Actividad
16.
Bioorg Med Chem Lett ; 16(1): 108-12, 2006 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-16236504

RESUMEN

The design, synthesis, and the biological evaluation of 2-benzamido-pyrimidines as novel IKK inhibitors are described. By optimization of the lead compound 3, compounds 16 and 24 are identified as good inhibitors of IKK2 with IC(50) values of 40 and 25 nM, respectively. Compound 16 also demonstrated significant in vivo activity in an acute model of cytokine release.


Asunto(s)
Química Farmacéutica/métodos , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , Pirimidinas/síntesis química , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Diseño de Fármacos , Ensayos de Selección de Medicamentos Antitumorales , Células HeLa , Humanos , Concentración 50 Inhibidora , Modelos Químicos , Pirimidinas/química , Pirimidinas/farmacología , Relación Estructura-Actividad
17.
J Mol Biol ; 355(2): 249-61, 2006 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-16305800

RESUMEN

BACE2 is a membrane-bound aspartic protease of the A1 family with a high level of sequence homology to BACE1. While BACE1 is involved in the generation of amyloid plaques in Alzheimer's disease by cleaving Abeta-peptides from the amyloid precursor protein, the physiological function of BACE2 is not well understood. BACE2 appears to be associated with the early onset of dementia in patients with Down's syndrome, and it has been shown to be highly expressed in breast cancers. Further, it may participate in the function of normal and abnormal processes of human muscle biology. Similar to other aspartic proteases, BACE2 is expressed as an inactive zymogen requiring the cleavage of its pro-sequence during the maturation process. We have produced mature BACE2 by expression of pro-BACE2 in Escherichia coli as inclusion bodies, followed by refolding and autocatalytic activation at pH 3.4. Using a C and N-terminally truncated BACE2 variant, we were able to crystallize and determine the crystal structure of mature BACE2 in complex with a hydroxyethylamine transition-state mimetic inhibitor at 3.1 angstroms resolution. The structure of BACE2 follows the general fold of A1 aspartic proteases. However, similar to BACE1, its C-terminal domain is significantly larger than that of the other family members. Furthermore, the structure of BACE2 reveals differences in the S3, S2, S1' and S2' active site substrate pockets as compared to BACE1, and allows, therefore, for a deeper understanding of the structural features that may facilitate the design of selective BACE1 or BACE2 inhibitors.


Asunto(s)
Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/química , Inhibidores Enzimáticos/química , Etanolaminas/química , Secuencia de Aminoácidos , Secretasas de la Proteína Precursora del Amiloide , Ácido Aspártico Endopeptidasas/genética , Sitios de Unión , Cristalización , Diseño de Fármacos , Estabilidad de Enzimas , Escherichia coli/enzimología , Escherichia coli/genética , Humanos , Cuerpos de Inclusión/enzimología , Datos de Secuencia Molecular , Mutación , Estructura Secundaria de Proteína , Relación Estructura-Actividad
18.
J Mol Biol ; 342(3): 889-99, 2004 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-15342244

RESUMEN

Cathepsin E is an intracellular, non-lysosomal aspartic protease expressed in a variety of cells and tissues. The protease has proposed physiological roles in antigen presentation by the MHC class II system, in the biogenesis of the vasoconstrictor peptide endothelin, and in neurodegeneration associated with brain ischemia and aging. Cathepsin E is the only A1 aspartic protease that exists as a homodimer with a disulfide bridge linking the two monomers. Like many other aspartic proteases, it is synthesized as a zymogen which is catalytically inactive towards its natural substrates at neutral pH and which auto-activates in an acidic environment. Here we report the crystal structure of an activation intermediate of human cathepsin E at 2.35A resolution. The overall structure follows the general fold of aspartic proteases of the A1 family, and the intermediate shares many features with the intermediate 2 on the proposed activation pathway of aspartic proteases like pepsin C and cathepsin D. The pro-sequence is cleaved from the protease and remains stably associated with the mature enzyme by forming the outermost sixth strand of the interdomain beta-sheet. However, different from these other aspartic proteases the pro-sequence of cathepsin E remains intact after cleavage from the mature enzyme. In addition, the active site of cathepsin E in the crystal is occupied by N-terminal amino acid residues of the mature protease in the non-primed binding site and by an artificial N-terminal extension of the pro-sequence from a neighboring molecule in the primed site. The crystal structure of the cathepsin E/pro-sequence complex, therefore, provides further insight into the activation mechanism of aspartic proteases.


Asunto(s)
Catepsina E/química , Catepsina E/metabolismo , Secuencia de Aminoácidos , Secuencia de Bases , Dominio Catalítico , Catepsina E/genética , Cristalografía por Rayos X , ADN Recombinante/genética , Activación Enzimática , Precursores Enzimáticos/química , Precursores Enzimáticos/genética , Precursores Enzimáticos/metabolismo , Humanos , Técnicas In Vitro , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Conformación Proteica , Estructura Cuaternaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA