Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Biochem Soc Trans ; 48(4): 1323-1336, 2020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32794575

RESUMEN

The proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates plasma LDL cholesterol levels by binding to the liver LDL receptor (LDLR) and promoting its degradation. Therefore, PCSK9 has become a compelling new therapeutic target for lipid lowering and the prevention of cardiovascular disease. PCSK9 contains two regions of conformational flexibility, the N-terminal regions of the prodomain and of the catalytic domain. The recognition that the latter region, the so-called P' helix, is able to transition from an α-helical to a disordered state gave rise to new strategies to develop small molecule inhibitors of PCSK9 for lipid lowering. In the ordered state the P' helix is buried in a groove of the PCSK9 catalytic domain located next to the main LDLR binding site. The transition to a disordered state leaves the groove site vacated and accessible for compounds to antagonize LDLR binding. By use of a groove-directed phage display strategy we were able to identify several groove-binding peptides. Based on structural information of PCSK9-peptide complexes, a minimized groove-binding peptide was generated and utilized as an anchor to extend towards the adjacent main LDLR binding site, either by use of a phage-displayed peptide extension library, or by appending organic moieties to yield organo-peptides. Both strategies led to antagonists with pharmacologic activities in cell-based assays. The intricate bipartite mechanism of the potent organo-peptide inhibitors was revealed by structural studies, showing that the core peptide occupies the N-terminal groove, while the organic moiety interacts with the LDLR binding site to create antagonism. These findings validate the PCSK9 groove as an attractive target site and should inspire the development of a new class of small molecule antagonists of PCSK9.


Asunto(s)
Anticolesterolemiantes/química , LDL-Colesterol/sangre , Diseño de Fármacos , Proproteína Convertasa 9/metabolismo , Inhibidores de Serina Proteinasa/química , Animales , Anticolesterolemiantes/farmacología , Sitios de Unión , Humanos , Inhibidores de PCSK9 , Proproteína Convertasa 9/química , Receptores de LDL/metabolismo , Inhibidores de Serina Proteinasa/farmacología
2.
Artículo en Inglés | MEDLINE | ID: mdl-30104274

RESUMEN

There is a critical need for new antibacterial strategies to counter the growing problem of antibiotic resistance. In Gram-negative bacteria, the outer membrane (OM) provides a protective barrier against antibiotics and other environmental insults. The outer leaflet of the outer membrane is primarily composed of lipopolysaccharide (LPS). Outer membrane biogenesis presents many potentially compelling drug targets as this pathway is absent in higher eukaryotes. Most proteins involved in LPS biosynthesis and transport are essential; however, few compounds have been identified that inhibit these proteins. The inner membrane ABC transporter MsbA carries out the first essential step in the trafficking of LPS to the outer membrane. We conducted a biochemical screen for inhibitors of MsbA and identified a series of quinoline compounds that kill Escherichia coli through inhibition of its ATPase and transport activity, with no loss of activity against clinical multidrug-resistant strains. Identification of these selective inhibitors indicates that MsbA is a viable target for new antibiotics, and the compounds we identified serve as useful tools to further probe the LPS transport pathway in Gram-negative bacteria.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , Lipopolisacáridos/metabolismo , Antibacterianos/farmacología , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Escherichia coli/efectos de los fármacos
3.
J Biol Chem ; 289(2): 942-55, 2014 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-24225950

RESUMEN

PCSK9 (proprotein convertase subtilisin/kexin type 9) is a negative regulator of the hepatic LDL receptor, and clinical studies with PCSK9-inhibiting antibodies have demonstrated strong LDL-c-lowering effects. Here we screened phage-displayed peptide libraries and identified the 13-amino acid linear peptide Pep2-8 as the smallest PCSK9 inhibitor with a clearly defined mechanism of inhibition that has been described. Pep2-8 bound to PCSK9 with a KD of 0.7 µm but did not bind to other proprotein convertases. It fully restored LDL receptor surface levels and LDL particle uptake in PCSK9-treated HepG2 cells. The crystal structure of Pep2-8 bound to C-terminally truncated PCSK9 at 1.85 Å resolution showed that the peptide adopted a strand-turn-helix conformation, which is remarkably similar to its solution structure determined by NMR. Consistent with the functional binding site identified by an Ala scan of PCSK9, the structural Pep2-8 contact region of about 400 Å(2) largely overlapped with that contacted by the EGF(A) domain of the LDL receptor, suggesting a competitive inhibition mechanism. Consistent with this, Pep2-8 inhibited LDL receptor and EGF(A) domain binding to PCSK9 with IC50 values of 0.8 and 0.4 µm, respectively. Remarkably, Pep2-8 mimicked secondary structural elements of the EGF(A) domain that interact with PCSK9, notably the ß-strand and a discontinuous short α-helix, and it engaged in the same ß-sheet hydrogen bonds as EGF(A) does. Although Pep2-8 itself may not be amenable to therapeutic applications, this study demonstrates the feasibility of developing peptidic inhibitors to functionally relevant sites on PCSK9.


Asunto(s)
Oligopéptidos/farmacología , Proproteína Convertasas/metabolismo , Receptores de LDL/metabolismo , Serina Endopeptidasas/metabolismo , Secuencia de Aminoácidos , Animales , Sitios de Unión , Unión Competitiva/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/metabolismo , Inhibidores Enzimáticos/farmacología , Células Hep G2 , Humanos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Oligopéptidos/química , Oligopéptidos/metabolismo , Biblioteca de Péptidos , Proproteína Convertasa 9 , Proproteína Convertasas/química , Proproteína Convertasas/genética , Unión Proteica/efectos de los fármacos , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de LDL/química , Receptores de LDL/genética , Serina Endopeptidasas/química , Serina Endopeptidasas/genética
4.
Proc Natl Acad Sci U S A ; 109(14): 5299-304, 2012 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-22431598

RESUMEN

The Ras gene is frequently mutated in cancer, and mutant Ras drives tumorigenesis. Although Ras is a central oncogene, small molecules that bind to Ras in a well-defined manner and exert inhibitory effects have not been uncovered to date. Through an NMR-based fragment screen, we identified a group of small molecules that all bind to a common site on Ras. High-resolution cocrystal structures delineated a unique ligand-binding pocket on the Ras protein that is adjacent to the switch I/II regions and can be expanded upon compound binding. Structure analysis predicts that compound-binding interferes with the Ras/SOS interactions. Indeed, selected compounds inhibit SOS-mediated nucleotide exchange and prevent Ras activation by blocking the formation of intermediates of the exchange reaction. The discovery of a small-molecule binding pocket on Ras with functional significance provides a new direction in the search of therapeutically effective inhibitors of the Ras oncoprotein.


Asunto(s)
Nucleótidos/metabolismo , Proteínas Son Of Sevenless/metabolismo , Proteínas ras/metabolismo , Sitios de Unión , Línea Celular , Humanos , Ligandos , Modelos Moleculares , Resonancia Magnética Nuclear Biomolecular , Proteínas ras/química
5.
Proc Natl Acad Sci U S A ; 109(47): 19368-73, 2012 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-23134728

RESUMEN

The protein kinase v-akt murine thymoma viral oncogene homolog (AKT), a key regulator of cell survival and proliferation, is frequently hyperactivated in human cancers. Intramolecular pleckstrin homology (PH) domain-kinase domain (KD) interactions are important in maintaining AKT in an inactive state. AKT activation proceeds after a conformational change that dislodges the PH from the KD. To understand these autoinhibitory interactions, we generated mutations at the PH-KD interface and found that most of them lead to constitutive activation of AKT. Such mutations are likely another mechanism by which activation may occur in human cancers and other diseases. In support of this likelihood, we found somatic mutations in AKT1 at the PH-KD interface that have not been previously described in human cancers. Furthermore, we show that the AKT1 somatic mutants are constitutively active, leading to oncogenic signaling. Additionally, our studies show that the AKT1 mutants are not effectively inhibited by allosteric AKT inhibitors, consistent with the requirement for an intact PH-KD interface for allosteric inhibition. These results have important implications for therapeutic intervention in patients with AKT mutations at the PH-KD interface.


Asunto(s)
Neoplasias/enzimología , Neoplasias/genética , Oncogenes/genética , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/genética , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/genética , Animales , Línea Celular Tumoral , Membrana Celular/efectos de los fármacos , Membrana Celular/enzimología , Transformación Celular Neoplásica/efectos de los fármacos , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Activación Enzimática/efectos de los fármacos , Humanos , Ratones , Modelos Moleculares , Proteínas Mutantes/metabolismo , Mutación/genética , Células 3T3 NIH , Unión Proteica/efectos de los fármacos , Unión Proteica/genética , Inhibidores de Proteínas Quinasas/farmacología , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
6.
Bioorg Med Chem Lett ; 24(3): 954-62, 2014 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-24433859

RESUMEN

The fragment-based identification of two novel and potent biochemical inhibitors of the nicotinamide phosphoribosyltransferase (NAMPT) enzyme is described. These compounds (51 and 63) incorporate an amide moiety derived from 3-aminopyridine, and are thus structurally distinct from other known anti-NAMPT agents. Each exhibits potent inhibition of NAMPT biochemical activity (IC50=19 and 15 nM, respectively) as well as robust antiproliferative properties in A2780 cell culture experiments (IC50=121 and 99 nM, respectively). However, additional biological studies indicate that only inhibitor 51 exerts its A2780 cell culture effects via a NAMPT-mediated mechanism. The crystal structures of both 51 and 63 in complex with NAMPT are also independently described.


Asunto(s)
Amidas/síntesis química , Amidas/farmacología , Aminopiridinas/síntesis química , Citocinas/antagonistas & inhibidores , Inhibidores Enzimáticos/síntesis química , Inhibidores Enzimáticos/farmacología , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Amidas/química , Aminopiridinas/química , Aminopiridinas/farmacología , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Cristalografía por Rayos X , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/química , Humanos , Concentración 50 Inhibidora , Modelos Moleculares , Relación Estructura-Actividad
7.
ACS Med Chem Lett ; 15(6): 864-872, 2024 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-38894924

RESUMEN

We were attracted to the therapeutic potential of inhibiting Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b), a RING E3 ligase that plays a critical role in regulating the activation of T cells. However, given that only protein-protein interactions were involved, it was unclear whether inhibition by a small molecule would be a viable approach. After screening an ∼6 billion member DNA-encoded library (DEL) using activated Cbl-b, we identified compound 1 as a hit for which the cis-isomer (2) was confirmed by biochemical and surface plasmon resonance (SPR) assays. Our hit optimization effort was greatly accelerated when we obtained a cocrystal structure of 2 with Cbl-b, which demonstrated induced binding at the substrate binding site, namely, the Src homology-2 (SH2) domain. This was quite noteworthy given that there are few reports of small molecule inhibitors that bind to SH2 domains and block protein-protein interactions. Structure- and property-guided optimization led to compound 27, which demonstrated measurable cell activity, albeit only at high concentrations.

8.
Bioorg Med Chem Lett ; 21(8): 2410-4, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21392984

RESUMEN

A novel series of spirochromane pan-Akt inhibitors is reported. SAR optimization furnished compounds with improved enzyme potencies and excellent selectivity over the related AGC kinase PKA. Attempted replacement of the phenol hinge binder provided compounds with excellent Akt enzyme and cell activities but greatly diminished selectivity over PKA.


Asunto(s)
Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Sitios de Unión , Cristalografía por Rayos X , Evaluación Preclínica de Medicamentos , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Relación Estructura-Actividad
9.
Bioorg Med Chem Lett ; 21(8): 2335-40, 2011 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-21420856
10.
Bioorg Med Chem Lett ; 20(23): 7037-41, 2010 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-20971641

RESUMEN

Herein we report the discovery and synthesis of a novel series of dihydrothieno- and dihydrofuropyrimidines (2 and 3) as potent pan Akt inhibitors. Utilizing previous SAR and analysis of the amino acid sequences in the binding site we have designed inhibitors displaying increased PKA and general kinase selectivity with improved tolerability compared to the progenitor pyrrolopyrimidine (1). A representative dihydrothieno compound (34) was advanced into a PC3-NCI prostate mouse tumor model in which it demonstrated a dose-dependent reduction in tumor growth and stasis when dosed orally daily at 200 mg/kg.


Asunto(s)
Neoplasias de la Próstata/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Pirimidinas/química , Animales , Sitios de Unión , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Masculino , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Relación Estructura-Actividad , Carga Tumoral/efectos de los fármacos
11.
Bioorg Med Chem Lett ; 20(19): 5607-12, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20810279
12.
ACS Chem Biol ; 15(6): 1392-1400, 2020 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-32302100

RESUMEN

Ubiquitin specific protease 7 (USP7) regulates the protein stability of key cellular regulators in pathways ranging from apoptosis to neuronal development, making it a promising therapeutic target. Here we used an engineered, activated variant of the USP7 catalytic domain to perform structure-activity studies of electrophilic peptidomimetic inhibitors. Employing this USP7 variant, we found that inhibitors with a cyanopyrrolidine warhead unexpectedly promoted a ß-elimination reaction of the initial covalent adducts, thereby converting the active-site cysteine residue to dehydroalanine. We determined that this phenomenon is specific for the USP7 catalytic cysteine and that structural features of the inhibitor and protein microenvironment impact elimination rates. Using comprehensive docking studies, we propose that the characteristic conformational dynamics of USP7 allow access to conformations that promote the ligand-induced elimination. Unlike in conventional reversible-covalent inhibition, the compounds described here irreversibly destroy a catalytic residue while simultaneously converting the inhibitor to a nonelectrophilic byproduct. Accordingly, this unexpected finding expands the scope of covalent inhibitor modalities and offers intriguing insights into enzyme-inhibitor dynamics.


Asunto(s)
Dominio Catalítico/efectos de los fármacos , Pirrolidinas/química , Pirrolidinas/farmacología , Peptidasa Específica de Ubiquitina 7/antagonistas & inhibidores , Cisteína/química , Cisteína/metabolismo , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Humanos , Simulación del Acoplamiento Molecular , Peptidomiméticos/química , Peptidomiméticos/farmacología , Peptidasa Específica de Ubiquitina 7/química , Peptidasa Específica de Ubiquitina 7/metabolismo
13.
ACS Chem Biol ; 15(2): 425-436, 2020 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-31962046

RESUMEN

Proprotein convertase subtilisin/kexin 9 (PCSK9) has become an important therapeutic target for lipid lowering, since it regulates low-density lipoprotein cholesterol (LDL-c) levels by binding to liver LDL receptors (LDLR) and effecting their intracellular degradation. However, the development of small molecule inhibitors is hampered by the lack of attractive PCSK9 target sites. We recently discovered helical peptides that are able to bind to a cryptic groove site on PCSK9, which is situated in proximity to the main LDLR binding site. Here, we designed potent bipartite PCSK9 inhibitors by appending organic moieties to a helical groove-binding peptide to reach a hydrophobic pocket in the proximal LDLR binding region. The ultimately designed 1-amino-4-phenylcyclohexane-1-carbonyl extension improved the peptide affinity by >100-fold, yielding organo-peptide antagonists that potently inhibited PCSK9 binding to LDLR and preserved cellular LDLR. These new bipartite antagonists have reduced mass and improved potency compared to the first-generation peptide antagonists, further validating the PCSK9 groove as a viable therapeutic target site.


Asunto(s)
Inhibidores de PCSK9 , Péptidos/farmacología , Inhibidores de Serina Proteinasa/farmacología , Sitios de Unión , Cristalografía por Rayos X , Diseño de Fármacos , Células Hep G2 , Humanos , Estructura Molecular , Péptidos/química , Péptidos/metabolismo , Proproteína Convertasa 9/química , Proproteína Convertasa 9/metabolismo , Unión Proteica , Inhibidores de Serina Proteinasa/química , Inhibidores de Serina Proteinasa/metabolismo
14.
Structure ; 26(1): 72-84.e7, 2018 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-29249604

RESUMEN

Ubiquitin-specific protease 7 (USP7) deubiquitinase activity is controlled by a number of regulatory factors, including stimulation by intramolecular accessory domains. Alone, the USP7 catalytic domain (USP7cd) shows limited activity and apo USP7cd crystal structures reveal a disrupted catalytic triad. By contrast, ubiquitin-conjugated USP7cd structures demonstrate the canonical cysteine protease active-site geometry; however, the structural features of the USP7cd that stabilize the inactive conformation and the mechanism of transition between inactive and active states remain unclear. Here we use comparative structural analyses, molecular dynamics simulations, and in silico sequence re-engineering via directed sampling by RosettaDesign to identify key molecular determinants of USP7cd activation and successfully engineer USP7cd for improved activity. Full kinetic analysis and multiple X-ray crystal structures of our designs indicate that electrostatic interactions in the distal "switching loop" region and local packing in the hydrophobic core mediate subtle but significant conformational changes that modulate USP7cd activation.


Asunto(s)
Inhibidores Enzimáticos/química , Mutación , Peptidomiméticos/química , Peptidasa Específica de Ubiquitina 7/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Dominio Catalítico , Clonación Molecular , Cristalografía por Rayos X , Activación Enzimática , Inhibidores Enzimáticos/síntesis química , Escherichia coli/genética , Escherichia coli/metabolismo , Expresión Génica , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Cinética , Simulación de Dinámica Molecular , Peptidomiméticos/síntesis química , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Electricidad Estática , Especificidad por Sustrato , Termodinámica , Peptidasa Específica de Ubiquitina 7/antagonistas & inhibidores , Peptidasa Específica de Ubiquitina 7/genética , Peptidasa Específica de Ubiquitina 7/metabolismo
15.
Protein Sci ; 16(11): 2454-71, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17962403

RESUMEN

High-temperature requirement A (HtrA) and its homologs contain a serine protease domain followed by one or two PDZ domains. Bacterial HtrA proteins and the mitochondrial protein HtrA2/Omi maintain cell function by acting as both molecular chaperones and proteases to manage misfolded proteins. The biological roles of the mammalian family members HtrA1 and HtrA3 are less clear. We report a detailed structural and functional analysis of the PDZ domains of human HtrA1 and HtrA3 using peptide libraries and affinity assays to define specificity, structural studies to view the molecular details of ligand recognition, and alanine scanning mutagenesis to investigate the energetic contributions of individual residues to ligand binding. In common with HtrA2/Omi, we show that the PDZ domains of HtrA1 and HtrA3 recognize hydrophobic polypeptides, and while C-terminal sequences are preferred, internal sequences are also recognized. However, the details of the interactions differ, as different domains rely on interactions with different residues within the ligand to achieve high affinity binding. The results suggest that mammalian HtrA PDZ domains interact with a broad range of hydrophobic binding partners. This promiscuous specificity resembles that of bacterial HtrA family members and suggests a similar function for recognizing misfolded polypeptides with exposed hydrophobic sequences. Our results support a common activation mechanism for the HtrA family, whereby hydrophobic peptides bind to the PDZ domain and induce conformational changes that activate the protease. Such a mechanism is well suited to proteases evolved for the recognition and degradation of misfolded proteins.


Asunto(s)
Serina Endopeptidasas/química , Secuencia de Aminoácidos , Chaperoninas/química , Escherichia coli/metabolismo , Serina Peptidasa A1 que Requiere Temperaturas Altas , Humanos , Ligandos , Datos de Secuencia Molecular , Dominios PDZ , Péptidos/química , Unión Proteica , Conformación Proteica , Pliegue de Proteína , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
16.
Sci Rep ; 7(1): 12524, 2017 10 02.
Artículo en Inglés | MEDLINE | ID: mdl-28970566

RESUMEN

Fibroblast Activation Protein (FAP) is a membrane-bound serine protease whose expression is often elevated in activated fibroblasts associated with tissue remodeling in various common diseases such as cancer, arthritis and fibrosis. Like the closely related dipeptidyl peptidase DPPIV, the extracellular domain of FAP can be released into circulation as a functional enzyme, and limited studies suggest that the circulating level of FAP correlates with the degree of tissue fibrosis. Here we describe a novel homogeneous fluorescence intensity assay for circulating FAP activity based on a recently identified natural substrate, FGF21. This assay is unique in that it can effectively distinguish endopeptidase activity of FAP from that of other related enzymes such as prolyl endopeptidase (PREP) and was validated using Fap-deficient mice. Structural modeling was used to elucidate the mechanistic basis for the observed specificity in substrate recognition by FAP, but not by DPPIV or PREP. Finally, the assay was used to detect elevated FAP activity in human patients diagnosed with liver cirrhosis and to determine the effectiveness of a chemical inhibitor for FAP in mice. We propose that the assay presented here could thus be utilized for diagnosis of FAP-related pathologies and for the therapeutic development of FAP inhibitors.


Asunto(s)
Factores de Crecimiento de Fibroblastos/genética , Fibrosis/genética , Gelatinasas/genética , Cirrosis Hepática/genética , Proteínas de la Membrana/genética , Serina Endopeptidasas/genética , Animales , Endopeptidasas/genética , Fibroblastos/enzimología , Fibroblastos/metabolismo , Fibrosis/enzimología , Fibrosis/patología , Regulación Enzimológica de la Expresión Génica , Humanos , Cirrosis Hepática/enzimología , Cirrosis Hepática/patología , Ratones , Prolil Oligopeptidasas , Especificidad por Sustrato
17.
Nat Struct Mol Biol ; 24(10): 848-856, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28825733

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) regulates plasma LDL cholesterol (LDL-c) levels by promoting the degradation of liver LDL receptors (LDLRs). Antibodies that inhibit PCSK9 binding to the EGF(A) domain of the LDLR are effective in lowering LDL-c. However, the discovery of small-molecule therapeutics is hampered by difficulty in targeting the relatively flat EGF(A)-binding site on PCSK9. Here we demonstrate that it is possible to target this site, based on the finding that the PCSK9 P' helix displays conformational flexibility. As a consequence, the vacated N-terminal groove of PCSK9, which is adjacent to the EGF(A)-binding site, is in fact accessible to small peptides. In phage-display experiments, the EGF(A)-mimicking peptide Pep2-8 was used as an anchor peptide for the attachment of an extension peptide library directed toward the groove site. Guided by structural information, we further engineered the identified groove-binding peptides into antagonists, which encroach on the EGF(A)-binding site and inhibit LDLR binding.


Asunto(s)
Inhibidores Enzimáticos/metabolismo , Inhibidores de PCSK9 , Péptidos/metabolismo , Proproteína Convertasa 9/metabolismo , Sitios de Unión , Inhibidores Enzimáticos/aislamiento & purificación , Humanos , Simulación del Acoplamiento Molecular , Biblioteca de Péptidos , Péptidos/aislamiento & purificación
18.
Cancer Cell ; 29(4): 477-493, 2016 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-26996308

RESUMEN

Activating mutations in protein kinases drive many cancers. While how recurring point mutations affect kinase activity has been described, the effect of in-frame deletions is not well understood. We show that oncogenic deletions within the ß3-αC loop of HER2 and BRAF are analogous to the recurrent EGFR exon 19 deletions. We identify pancreatic carcinomas with BRAF deletions mutually exclusive with KRAS mutations. Crystal structures of BRAF deletions reveal the truncated loop restrains αC in an active "in" conformation, imparting resistance to inhibitors like vemurafenib that bind the αC "out" conformation. Characterization of loop length explains the prevalence of five amino acid deletions in BRAF, EGFR, and HER2 and highlights the importance of this region for kinase activity and inhibitor efficacy.


Asunto(s)
Genes erbB-1 , Genes erbB-2 , Mutación , Proteínas de Neoplasias/genética , Neoplasias/genética , Proteínas Proto-Oncogénicas B-raf/genética , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Antineoplásicos/farmacología , Emparejamiento Base/genética , Secuencia Conservada , Dimerización , Resistencia a Antineoplásicos/genética , Activación Enzimática/genética , Receptores ErbB/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Neoplasias/enzimología , Conformación Proteica , Mapeo de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Estructura Secundaria de Proteína , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Proteínas Proto-Oncogénicas B-raf/metabolismo , Receptor ErbB-2/antagonistas & inhibidores , Receptor ErbB-2/metabolismo , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Relación Estructura-Actividad
19.
J Mol Biol ; 316(5): 1111-25, 2002 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-11884148

RESUMEN

Display of peptide libraries on filamentous phage has led to the identification of peptides of the form X(2-5)CX(2)GPXTWXCX(2-5) (where X is a variable residue) that bind to the extra-cellular portion of the erythropoietin receptor (EPO-R). These peptides adopt beta-hairpin conformations when co-crystallized with EPO-R. Solution NMR studies reveal that the peptide is conformationally heterogeneous in the absence of receptor due to cis-trans isomerization about the Gly-Pro peptide bond. Replacement of the conserved threonine residue with glycine at the turn i+3 position produces a stable beta-hairpin conformation in solution, although this peptide no longer has activity in an EPO-R-dependent cell proliferation assay. A truncated form of the EPO-R-binding peptide (containing the i+3 glycine residue) also forms a highly populated, monomeric beta-hairpin. In contrast, phage-derived peptide antagonists of insulin-like growth factor binding protein 1 (IGFBP-1) have a high level of sequence identity with the truncated EPO-R peptide (eight of 12 residues) yet adopt a turn-alpha-helix conformation in solution. Peptides containing all possible pairwise amino acid substitutions between the EPO-R and IGFBP-1 peptides have been analyzed to assess the degree to which the non-conserved residues stabilize the hairpin or helix conformation. All four residues present in the original sequence are required for maximum population of either the beta-hairpin or alpha-helix conformation, although some substitutions have a more dominant effect. The results demonstrate that, within a given sequence, the observed conformation can be dictated by a small subset of the residues (in this case four out of 12).


Asunto(s)
Biblioteca de Péptidos , Péptidos/química , Péptidos/metabolismo , Receptores de Eritropoyetina/agonistas , Proteínas Virales/química , Proteínas Virales/metabolismo , Secuencia de Aminoácidos , División Celular/efectos de los fármacos , Línea Celular , Dicroismo Circular , Biología Computacional , Bases de Datos de Proteínas , Humanos , Enlace de Hidrógeno , Ligandos , Espectroscopía de Resonancia Magnética , Modelos Moleculares , Datos de Secuencia Molecular , Oxidación-Reducción , Péptidos/farmacología , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Receptores de Eritropoyetina/química , Receptores de Eritropoyetina/genética , Receptores de Eritropoyetina/metabolismo , Alineación de Secuencia , Eliminación de Secuencia , Termodinámica
20.
Chem Biol ; 9(4): 495-505, 2002 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11983338

RESUMEN

A panel of 22 naïve peptide libraries was constructed in a polyvalent phage display format and sorted against insulin-like growth factor-1 (IGF-1). The libraries were pooled to achieve a total diversity of 4.4 x 10(11). After three rounds of selection, the majority of the phage clones bound specifically to IGF-1, with a disulfide-constrained CX(9)C scaffold dominating the selection. Four monovalently displayed sub-libraries were designed on the basis of these conserved motifs. Sub-library maturation in a monovalent format yielded an antagonistic peptide that inhibited the interactions between IGF-1 and two cell-surface receptors and those between IGF-1 and two soluble IGF binding proteins with micromolar potency. NMR analysis revealed that the peptide is highly structured in the absence of IGF-1, and peptides that preorganize the binding elements were selected during the sorting.


Asunto(s)
Factor I del Crecimiento Similar a la Insulina/antagonistas & inhibidores , Biblioteca de Péptidos , Péptidos/farmacología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , Unión Competitiva , Humanos , Proteínas de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Datos de Secuencia Molecular , Péptidos/química , Péptidos/metabolismo , Estructura Secundaria de Proteína , Receptores de Superficie Celular/metabolismo , Solubilidad , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA