Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Elife ; 132024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38829031

RESUMEN

Connexins allow intercellular communication by forming gap junction channels (GJCs) between juxtaposed cells. Connexin26 (Cx26) can be regulated directly by CO2. This is proposed to be mediated through carbamylation of K125. We show that mutating K125 to glutamate, mimicking the negative charge of carbamylation, causes Cx26 GJCs to be constitutively closed. Through cryo-EM we observe that the K125E mutation pushes a conformational equilibrium towards the channel having a constricted pore entrance, similar to effects seen on raising the partial pressure of CO2. In previous structures of connexins, the cytoplasmic loop, important in regulation and where K125 is located, is disordered. Through further cryo-EM studies we trap distinct states of Cx26 and observe density for the cytoplasmic loop. The interplay between the position of this loop, the conformations of the transmembrane helices and the position of the N-terminal helix, which controls the aperture to the pore, provides a mechanism for regulation.


Asunto(s)
Dióxido de Carbono , Conexina 26 , Microscopía por Crioelectrón , Conformación Proteica , Humanos , Dióxido de Carbono/metabolismo , Conexina 26/metabolismo , Conexina 26/genética , Conexinas/metabolismo , Conexinas/genética , Conexinas/química , Uniones Comunicantes/metabolismo , Mutación
2.
Elife ; 122023 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-37963091

RESUMEN

The bile acid sodium symporter (BASS) family transports a wide array of molecules across membranes, including bile acids in humans, and small metabolites in plants. These transporters, many of which are sodium-coupled, have been shown to use an elevator mechanism of transport, but exactly how substrate binding is coupled to sodium ion binding and transport is not clear. Here, we solve the crystal structure at 2.3 Å of a transporter from Neisseria meningitidis (ASBTNM) in complex with pantoate, a potential substrate of ASBTNM. The BASS family is characterised by two helices that cross-over in the centre of the protein in an arrangement that is intricately held together by two sodium ions. We observe that the pantoate binds, specifically, between the N-termini of two of the opposing helices in this cross-over region. During molecular dynamics simulations the pantoate remains in this position when sodium ions are present but is more mobile in their absence. Comparison of structures in the presence and absence of pantoate demonstrates that pantoate elicits a conformational change in one of the cross-over helices. This modifies the interface between the two domains that move relative to one another to elicit the elevator mechanism. These results have implications, not only for ASBTNM but for the BASS family as a whole and indeed other transporters that work through the elevator mechanism.


Asunto(s)
Simportadores , Humanos , Simportadores/metabolismo , Sodio/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Simulación de Dinámica Molecular , Iones/metabolismo
3.
bioRxiv ; 2023 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-37645971

RESUMEN

The Bile Acid Sodium Symporter (BASS) family transports a wide array of molecules across membranes, including bile acids in humans, and small metabolites in plants. These transporters, many of which are sodium-coupled, have been shown to use an elevator mechanism of transport, but exactly how substrate binding is coupled to sodium ion binding and transport is not clear. Here we solve the crystal structure at 2.3 Å of a transporter from Neisseria Meningitidis (ASBTNM) in complex with pantoate, a potential substrate of ASBTNM. The BASS family is characterised by two helices that cross-over in the centre of the protein in an arrangement that is intricately held together by two sodium ions. We observe that the pantoate binds, specifically, between the N-termini of two of the opposing helices in this cross-over region. During molecular dynamics simulations the pantoate remains in this position when sodium ions are present but is more mobile in their absence. Comparison of structures in the presence and absence of pantoate demonstrates that pantoate elicits a conformational change in one of the cross-over helices. This modifies the interface between the two domains that move relative to one another to elicit the elevator mechanism. These results have implications, not only for ASBTNM but for the BASS family as a whole and indeed other transporters that work through the elevator mechanism.

4.
EMBO J ; 41(16): e110527, 2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-35775318

RESUMEN

CodB is a cytosine transporter from the Nucleobase-Cation-Symport-1 (NCS1) transporter family, a member of the widespread LeuT superfamily. Previous experiments with the nosocomial pathogen Pseudomonas aeruginosa have shown CodB as also important for the uptake of 5-fluorocytosine, which has been suggested as a novel drug to combat antimicrobial resistance by suppressing virulence. Here we solve the crystal structure of CodB from Proteus vulgaris, at 2.4 Å resolution in complex with cytosine. We show that CodB carries out the sodium-dependent uptake of cytosine and can bind 5-fluorocytosine. Comparison of the substrate-bound structures of CodB and the hydantoin transporter Mhp1, the only other NCS1 family member for which the structure is known, highlight the importance of the hydrogen bonds that the substrates make with the main chain at the breakpoint in the discontinuous helix, TM6. In contrast to other LeuT superfamily members, neither CodB nor Mhp1 makes specific interactions with residues on TM1. Comparison of the structures provides insight into the intricate mechanisms of how these proteins transport substrates across the plasma membrane.


Asunto(s)
Simportadores , Transporte Biológico , Cationes , Citosina , Flucitosina , Proteínas de Transporte de Membrana , Simportadores/genética
5.
Structure ; 30(5): 697-706.e4, 2022 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-35276081

RESUMEN

Connexins form large-pore channels that function either as dodecameric gap junctions or hexameric hemichannels to allow the regulated movement of small molecules and ions across cell membranes. Opening or closing of the channels is controlled by a variety of stimuli, and dysregulation leads to multiple diseases. An increase in the partial pressure of carbon dioxide (PCO2) has been shown to cause connexin26 (Cx26) gap junctions to close. Here, we use cryoelectron microscopy (cryo-EM) to determine the structure of human Cx26 gap junctions under increasing levels of PCO2. We show a correlation between the level of PCO2 and the size of the aperture of the pore, governed by the N-terminal helices that line the pore. This indicates that CO2 alone is sufficient to cause conformational changes in the protein. Analysis of the conformational states shows that movements at the N terminus are linked to both subunit rotation and flexing of the transmembrane helices.


Asunto(s)
Dióxido de Carbono , Conexinas , Dióxido de Carbono/metabolismo , Membrana Celular/metabolismo , Conexina 26 , Conexinas/química , Conexinas/metabolismo , Microscopía por Crioelectrón , Uniones Comunicantes/metabolismo , Humanos
6.
Proc Natl Acad Sci U S A ; 110(49): 19754-9, 2013 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-24248379

RESUMEN

Bromodomains have emerged as attractive candidates for the development of inhibitors targeting gene transcription. Inhibitors of the bromo and extraterminal (BET) family recently showed promising activity in diverse disease models. However, the pleiotropic nature of BET proteins regulating tissue-specific transcription has raised safety concerns and suggested that attempts should be made for domain-specific targeting. Here, we report that RVX-208, a compound currently in phase II clinical trials, is a BET bromodomain inhibitor specific for second bromodomains (BD2s). Cocrystal structures revealed binding modes of RVX-208 and its synthetic precursor, and fluorescent recovery after photobleaching demonstrated that RVX-208 displaces BET proteins from chromatin. However, gene-expression data showed that BD2 inhibition only modestly affects BET-dependent gene transcription. Our data demonstrate the feasibility of specific targeting within the BET family resulting in different transcriptional outcomes and highlight the importance of BD1 in transcriptional regulation.


Asunto(s)
Modelos Moleculares , Quinazolinas/química , Quinazolinas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/química , Cristalización , Recuperación de Fluorescencia tras Fotoblanqueo , Células Hep G2 , Humanos , Estructura Molecular , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , Estructura Terciaria de Proteína/fisiología , Quinazolinonas
7.
J Pharmacol Exp Ther ; 339(1): 132-42, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21778281

RESUMEN

The therapeutic and toxic effects of drugs are often generated through effects on distinct cell types in the body. Selective delivery of drugs to specific cells or cell lineages would, therefore, have major advantages, in particular, the potential to significantly improve the therapeutic window of an agent. Cells of the monocyte-macrophage lineage represent an important target for many therapeutic agents because of their central involvement in a wide range of diseases including inflammation, cancer, atherosclerosis, and diabetes. We have developed a versatile chemistry platform that is designed to enhance the potency and delivery of small-molecule drugs to intracellular molecular targets. One facet of the technology involves the selective delivery of drugs to cells of the monocyte-macrophage lineage, using the intracellular carboxylesterase, human carboxylesterase-1 (hCE-1), which is expressed predominantly in these cells. Here, we demonstrate selective delivery of many types of intracellularly targeted small molecules to monocytes and macrophages by attaching a small esterase-sensitive chemical motif (ESM) that is selectively hydrolyzed within these cells to a charged, pharmacologically active drug. ESM versions of histone deacetylase (HDAC) inhibitors, for example, are extremely potent anticytokine and antiarthritic agents with a wider therapeutic window than conventional HDAC inhibitors. In human blood, effects on monocytes (hCE-1-positive) are seen at concentrations 1000-fold lower than those that affect other cell types (hCE-1-negative). Chemical conjugates of this type, by limiting effects on other cells, could find widespread applicability in the treatment of human diseases where monocyte-macrophages play a key role in disease pathology.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Esterasas/antagonistas & inhibidores , Esterasas/química , Macrófagos/efectos de los fármacos , Monocitos/efectos de los fármacos , Aminoácidos/química , Animales , Anisomicina/farmacología , Artritis/inmunología , Carboxilesterasa/antagonistas & inhibidores , Carboxilesterasa/química , Carboxilesterasa/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Citocinas/biosíntesis , Citocinas/sangre , Citocinas/genética , Inhibidores Enzimáticos/farmacología , Esterasas/genética , Ésteres/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lipopolisacáridos/farmacología , Espectroscopía de Resonancia Magnética , Ratones , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/sangre , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
8.
J Med Chem ; 53(24): 8663-78, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-21080647

RESUMEN

A novel series of HDAC inhibitors demonstrating class I subtype selectivity and good oral bioavailability is described. The compounds are potent enzyme inhibitors (IC50 values less than 100 nM), and improved activity in cell proliferation assays was achieved by modulation of polar surface area (PSA) through the introduction of novel linking groups. Employing oral pharmacokinetic studies in mice, comparing drug levels in spleen to plasma, we selected compounds that were tested for efficacy in human tumor xenograft studies based on their potential to distribute into tumor. One compound, 21r (CHR-3996), showed good oral activity in these models, including dose-related activity in a LoVo xenograft. In addition 21r showed good activity in combination with other anticancer agents in in vitro studies. On the basis of these results, 21r was nominated for clinical development.


Asunto(s)
Antineoplásicos/síntesis química , Compuestos de Azabiciclo/síntesis química , Inhibidores de Histona Desacetilasas/síntesis química , Pirimidinas/síntesis química , Animales , Antineoplásicos/farmacocinética , Antineoplásicos/farmacología , Compuestos de Azabiciclo/farmacocinética , Compuestos de Azabiciclo/farmacología , Línea Celular Tumoral , Perros , Ensayos de Selección de Medicamentos Antitumorales , Sinergismo Farmacológico , Femenino , Inhibidores de Histona Desacetilasas/farmacocinética , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Técnicas In Vitro , Ratones , Ratones Desnudos , Microsomas Hepáticos/metabolismo , Modelos Moleculares , Trasplante de Neoplasias , Pirimidinas/farmacocinética , Pirimidinas/farmacología , Ratas , Estereoisomerismo , Relación Estructura-Actividad , Distribución Tisular , Trasplante Heterólogo
9.
Nat Chem Biol ; 4(6): 357-65, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18469809

RESUMEN

Cdc7 is an essential kinase that promotes DNA replication by activating origins of replication. Here, we characterized the potent Cdc7 inhibitor PHA-767491 (1) in biochemical and cell-based assays, and we tested its antitumor activity in rodents. We found that the compound blocks DNA synthesis and affects the phosphorylation of the replicative DNA helicase at Cdc7-dependent phosphorylation sites. Unlike current DNA synthesis inhibitors, PHA-767491 prevents the activation of replication origins but does not impede replication fork progression, and it does not trigger a sustained DNA damage response. Treatment with PHA-767491 results in apoptotic cell death in multiple cancer cell types and tumor growth inhibition in preclinical cancer models. To our knowledge, PHA-767491 is the first molecule that directly affects the mechanisms controlling initiation as opposed to elongation in DNA replication, and its activities suggest that Cdc7 kinase inhibition could be a new strategy for the development of anticancer therapeutics.


Asunto(s)
Antineoplásicos/farmacología , Proteínas de Ciclo Celular/antagonistas & inhibidores , Replicación del ADN/efectos de los fármacos , ADN/efectos de los fármacos , Piperidonas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Pirroles/farmacología , Animales , Antineoplásicos/química , Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN/biosíntesis , Relación Dosis-Respuesta a Droga , Fibroblastos/efectos de los fármacos , Células HeLa , Humanos , Ratones , Ratones Desnudos , Ratones SCID , Componente 2 del Complejo de Mantenimiento de Minicromosoma , Estructura Molecular , Proteínas Nucleares/antagonistas & inhibidores , Proteínas Nucleares/química , Fosforilación , Piperidonas/química , Inhibidores de Proteínas Quinasas/química , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/genética , Pirroles/química , Ratas , Bibliotecas de Moléculas Pequeñas , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Biol Chem ; 282(1): 208-15, 2007 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-17062569

RESUMEN

Cdc7 kinase promotes and regulates DNA replication in eukaryotic organisms. Multiple mechanisms modulating kinase activity in response to DNA replication stress have been reported, supporting the opposing notions that Cdc7 either plays an active role under these conditions or, conversely, is a final target inactivated by a checkpoint response. We have developed new immnunological reagents to study the properties of human Cdc7 kinase in cells challenged with the ribonucleotide reductase inhibitor hydroxyurea or with the DNA topoisomerase II inhibitor etoposide. We show that Cdc7.Dbf4 and Cdc7.Drf1 complexes are stable and active in multiple cell lines upon drug treatment, with Cdc7.Dbf4 accumulating on chromatin-enriched fractions. Cdc7 depletion by small interfering RNA in hydroxyurea and etoposide impairs hyper-phosphorylation of Mcm2 at specific Cdc7-dependent phosphorylation sites and drug-induced hyper-phosphorylation of chromatin-bound Mcm4. Furthermore, sustained inhibition of Cdc7 in the presence of these drugs increases cell death supporting the notion that the Cdc7 kinase plays a role in maintaining cell viability during replication stress.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Apoptosis , Proteínas de Ciclo Celular/fisiología , Supervivencia Celular , Cromatina/química , ADN/química , Etopósido/química , Etopósido/farmacología , Células HeLa , Humanos , Hidroxiurea/química , Hidroxiurea/farmacología , Componente 2 del Complejo de Mantenimiento de Minicromosoma , Proteínas Nucleares/fisiología , Fosforilación , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Ribonucleótido Reductasas/antagonistas & inhibidores
11.
J Biol Chem ; 281(15): 10281-90, 2006 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-16446360

RESUMEN

Minichromosome maintenance 2-7 proteins play a pivotal role in replication of the genome in eukaryotic organisms. Upon entry into S-phase several subunits of the MCM hexameric complex are phosphorylated. It is thought that phosphorylation activates the intrinsic MCM DNA helicase activity, thus allowing formation of active replication forks. Cdc7, Cdk2, and ataxia telangiectasia and Rad3-related kinases regulate S-phase entry and S-phase progression and are known to phosphorylate the Mcm2 subunit. In this work, by in vitro kinase reactions and mass spectrometry analysis of the products, we have mapped phosphorylation sites in the N terminus of Mcm2 by Cdc7, Cdk2, Cdk1, and CK2. We found that Cdc7 phosphorylates Mcm2 in at least three different sites, one of which corresponds to a site also reported to be phosphorylated by ataxia telangiectasia and Rad3-related. Three serine/proline sites were identified for Cdk2 and Cdk1, and a unique site was phosphorylated by CK2. We raised specific anti-phosphopeptide antibodies and found that all the sites identified in vitro are also phosphorylated in cells. Importantly, although all the Cdc7-dependent Mcm2 phosphosites fluctuate during the cell cycle with kinetics similar to Cdc7 kinase activity and Cdc7 protein levels, phosphorylation of Mcm2 in the putative cyclin-dependent kinase (Cdk) consensus sites is constant during the cell cycle. Furthermore, our analysis indicates that the majority of the Mcm2 isoforms phosphorylated by Cdc7 are not stably associated with chromatin. This study forms the basis for understanding how MCM functions are regulated by multiple kinases within the cell cycle and in response to external perturbations.


Asunto(s)
Proteínas de Ciclo Celular/fisiología , Proteínas Nucleares/fisiología , Proteínas Quinasas Asociadas a Fase-S/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Western Blotting , Proteína Quinasa CDC2/metabolismo , Quinasa de la Caseína II/metabolismo , Ciclo Celular , Proteínas de Ciclo Celular/metabolismo , Cromatina/química , Cromatografía Liquida , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasas Ciclina-Dependientes/metabolismo , ADN Helicasas/química , Electroforesis en Gel de Poliacrilamida , Fibroblastos/metabolismo , Células HeLa , Humanos , Iones , Luciferasas/metabolismo , Espectrometría de Masas , Microscopía Fluorescente , Componente 2 del Complejo de Mantenimiento de Minicromosoma , Datos de Secuencia Molecular , Proteínas Nucleares/metabolismo , Péptidos/química , Fosforilación , Prolina/química , Isoformas de Proteínas , Estructura Terciaria de Proteína , Proteínas Recombinantes de Fusión/química , Serina/química , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Timidina/química , Transfección , Tripsina/farmacología , Quinasa Activadora de Quinasas Ciclina-Dependientes
12.
Cancer Res ; 64(19): 7110-6, 2004 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-15466207

RESUMEN

Cdc7 is an evolutionarily conserved kinase that regulates S phase by promoting replication origin activation. Down-regulation of Cdc7 by small interfering RNA in a variety of tumor cell lines causes an abortive S phase, leading to cell death by either p53-independent apoptosis or aberrant mitosis. Unlike replication fork blockade, Cdc7-depleted tumor cells do not elicit a robust checkpoint response; thus, inhibitory signals preventing additional cell cycle progression are not generated. In normal fibroblasts, however, a p53-dependent pathway actively prevents progression through a lethal S phase in the absence of sufficient Cdc7 kinase. We show that in this experimental system, p53 is required for the lasting maintenance of this checkpoint and for cell viability. With this work we reveal and begin to characterize a novel mechanism that regulates DNA synthesis in human cells, and we suggest that inhibition of Cdc7 kinase represents a promising approach for the development of a new generation of anticancer agents.


Asunto(s)
Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Fase S/fisiología , Proteína p53 Supresora de Tumor/fisiología , Apoptosis/fisiología , Ciclo Celular/fisiología , Proteínas de Ciclo Celular/genética , Progresión de la Enfermedad , Regulación hacia Abajo , Células HeLa , Humanos , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , ARN Interferente Pequeño/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...