Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 87
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
EMBO J ; 41(23): e108970, 2022 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-36281581

RESUMEN

Phagocytosis is a key process in innate immunity and homeostasis. After particle uptake, newly formed phagosomes mature by acquisition of endolysosomal enzymes. Macrophage activation by interferon gamma (IFN-γ) increases microbicidal activity, but delays phagosomal maturation by an unknown mechanism. Using quantitative proteomics, we show that phagosomal proteins harbour high levels of typical and atypical ubiquitin chain types. Moreover, phagosomal ubiquitylation of vesicle trafficking proteins is substantially enhanced upon IFN-γ activation of macrophages, suggesting a role in regulating phagosomal functions. We identified the E3 ubiquitin ligase RNF115, which is enriched on phagosomes of IFN-γ activated macrophages, as an important regulator of phagosomal maturation. Loss of RNF115 protein or ligase activity enhanced phagosomal maturation and increased cytokine responses to bacterial infection, suggesting that both innate immune signalling from the phagosome and phagolysosomal trafficking are controlled through ubiquitylation. RNF115 knock-out mice show less tissue damage in response to S. aureus infection, indicating a role of RNF115 in inflammatory responses in vivo. In conclusion, RNF115 and phagosomal ubiquitylation are important regulators of innate immune functions during bacterial infections.


Asunto(s)
Infecciones Bacterianas , Fagosomas , Ubiquitina-Proteína Ligasas , Animales , Ratones , Infecciones Bacterianas/metabolismo , Interferón gamma/metabolismo , Fagocitosis , Fagosomas/metabolismo , Staphylococcus aureus , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
2.
Annu Rev Physiol ; 81: 43-62, 2019 02 10.
Artículo en Inglés | MEDLINE | ID: mdl-30354932

RESUMEN

In the nervous system, ATP is co-stored in vesicles with classical transmitters and released in a regulated manner. ATP from the intracellular compartment can also exit the cell through hemichannels and following shear stress or membrane damage. In the past 30 years, the action of ATP as an extracellular transmitter at cell-surface receptors has evolved from somewhat of a novelty that was treated with skepticism to purinergic transmission being accepted as having widespread important functional roles mediated by ATP-gated ionotropic P2X receptors (P2XRs). This review focuses on work published in the last five years and provides an overview of ( a) structural studies, ( b) the molecular basis of channel properties and regulation of P2XRs, and ( c) the physiological and pathophysiological roles of ATP acting at defined P2XR subtypes.


Asunto(s)
Adenosina Trifosfato/metabolismo , Canales Iónicos/metabolismo , Receptores Purinérgicos P2X/metabolismo , Animales , Humanos
3.
Mol Pharmacol ; 101(1): 33-44, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34718224

RESUMEN

The P2X4 receptor is a ligand-gated ion channel activated by extracellular ATP. P2X4 activity is associated with neuropathic pain, vasodilation, and pulmonary secretion and is therefore of therapeutic interest. The structure-activity relationship of P2X4 antagonists is poorly understood. Here we elucidate the structure-activity of 5-(3-bromophenyl)-1,3-dihydro-2H-benzofuro[3,2-e]-1,4-diazepin-2-one (5-BDBD) at human P2X4 by combining pharmacology, electrophysiology, molecular modeling, and medicinal chemistry. 5-BDBD antagonized P2X4 in a noncompetitive manner but lacked effect at human P2X2. Molecular modeling and site-directed mutagenesis suggested an allosteric binding site for 5-BDBD located between two subunits in the body region of P2X4, with M109, F178, Y300, and I312 on one subunit and R301 on the neighboring subunit as key residues involved in antagonist binding. The bromine group of 5-BDBD was redundant for the antagonist activity of 5-BDBD, although an interaction between the carbonyl group of 5-BDBD and R301 in P2X4 was associated with 5-BDBD activity. 5-BDBD could inhibit the closed channel but poorly inhibited the channel in the open/desensitizing state. We hypothesize that this is due to constriction of the allosteric site after transition from closed to open channel state. We propose that M109, F178, Y300, R301, and I312 are key residues for 5-BDBD binding; provide a structural explanation of how they contribute to 5-BDBD antagonism; and highlight that the limited action of 5-BDBD on open versus closed channels is due to a conformational change in the allosteric site. SIGNIFICANCE STATEMENT: Activity of P2X4 receptor is associated with neuropathic pain, inflammation, and vasodilatation. Molecular information regarding small-molecule interaction with P2X4 is very limited. Here, this study provides a structural explanation for the action of the small-molecule antagonist 5-BDBD at the human P2X4 receptor.


Asunto(s)
Benzodiazepinonas/química , Benzodiazepinonas/metabolismo , Antagonistas del Receptor Purinérgico P2X/química , Antagonistas del Receptor Purinérgico P2X/metabolismo , Receptores Purinérgicos P2X4/química , Receptores Purinérgicos P2X4/metabolismo , Regulación Alostérica/efectos de los fármacos , Regulación Alostérica/fisiología , Benzodiazepinonas/farmacología , Células HEK293 , Humanos , Simulación de Dinámica Molecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Antagonistas del Receptor Purinérgico P2X/farmacología
4.
J Chem Phys ; 156(12): 124504, 2022 Mar 28.
Artículo en Inglés | MEDLINE | ID: mdl-35364890

RESUMEN

We use a degenerated Ising model to describe nucleation and crystallization from solution in a confined two-component system. The free energy is calculated using metadynamics simulation with coordination numbers as the reaction coordinates. We deploy nudged elastic band simulation to determine the minimum energy path and give properties of the crystallization path. In this confined system, depletion effects, which could also be caused by slow material transport in the solution, prevent the post-critical cluster from further growth, and the crystalline state would only be stable at larger cluster sizes. Fluctuation of the higher coupling strength of the crystalline state enables further growth until the crystalline cluster is in equilibrium with the solvent, and this way, a second barrier is crossed. From the parameters and setup, we find necessary conditions for the occurrence of two-step nucleation in our system. These findings can be adapted to real systems as biomineralization, colloidal crystallization, and the solidification of metals.

5.
J Biol Chem ; 295(38): 13277-13286, 2020 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-32723862

RESUMEN

The EAG (ether-à-go-go) family of voltage-gated K+ channels are important regulators of neuronal and cardiac action potential firing (excitability) and have major roles in human diseases such as epilepsy, schizophrenia, cancer, and sudden cardiac death. A defining feature of EAG (Kv10-12) channels is a highly conserved domain on the N terminus, known as the eag domain, consisting of a Per-ARNT-Sim (PAS) domain capped by a short sequence containing an amphipathic helix (Cap domain). The PAS and Cap domains are both vital for the normal function of EAG channels. Using heme-affinity pulldown assays and proteomics of lysates from primary cortical neurons, we identified that an EAG channel, hERG3 (Kv11.3), binds to heme. In whole-cell electrophysiology experiments, we identified that heme inhibits hERG3 channel activity. In addition, we expressed the Cap and PAS domain of hERG3 in Escherichia coli and, using spectroscopy and kinetics, identified the PAS domain as the location for heme binding. The results identify heme as a regulator of hERG3 channel activity. These observations are discussed in the context of the emerging role for heme as a regulator of ion channel activity in cells.


Asunto(s)
Corteza Cerebral/química , Canales de Potasio Éter-A-Go-Go/química , Hemo/química , Neuronas/química , Corteza Cerebral/metabolismo , Canales de Potasio Éter-A-Go-Go/metabolismo , Hemo/metabolismo , Humanos , Neuronas/metabolismo , Unión Proteica , Dominios Proteicos
6.
Biochem Biophys Res Commun ; 523(1): 190-195, 2020 02 26.
Artículo en Inglés | MEDLINE | ID: mdl-31843194

RESUMEN

P2X receptors are trimeric ATP-gated ion channels. In response to ATP binding, conformational changes lead to opening of the channel and ion flow. Current flow can decline during continued ATP binding in a process called desensitisation. The rate and extent of desensitisation is affected by multiple factors, for instance the T18A mutation in P2X2 makes the ion channel fast desensitising. We have used this mutation to investigate whether the gate restricting ion flow is different in the desensitised and the closed state, by combining molecular modelling and cysteine modification using MTSET (2-(Trimethylammonium)ethyl methanethiosulfonate). Homology modelling of the P2X2 receptor and negative space imaging of the channel suggested a movement of the restriction gate with residue T335 being solvent accessible in the desensitised, but not the closed state. This was confirmed experimentally by probing the accessibility of T335C in the P2X2 T18A/T335C (fast desensitisation) and T335C (slow desensitisation) mutants with MTSET which demonstrates that the barrier to ion flow is different in the closed and the desensitised states. To investigate the T18A induced switch in desensitisation we compared molecular dynamics simulations of the wild type and T18A P2X2 receptor which suggest that the differences in time course of desensitisation are due to structural destabilization of a hydrogen bond network of conserved residues in the proximity of T18.


Asunto(s)
Adenosina Trifosfato/metabolismo , Receptores Purinérgicos P2X2/química , Receptores Purinérgicos P2X2/metabolismo , Humanos , Modelos Moleculares , Mutación , Receptores Purinérgicos P2X2/genética
7.
Mol Pharmacol ; 96(3): 355-363, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31263019

RESUMEN

The P2X7 receptor is a trimeric ligand-gated ion channel activated by ATP. It is implicated in the cellular response to trauma/disease and considered to have significant therapeutic potential. Using chimeras and point mutants we have mapped the binding site of the P2X7R-selective antagonist AZ11645373 to the known allosteric binding pocket at the interface between two subunits, in proximity to, but separated from the ATP binding site. Our structural model of AZ11645373 binding is consistent with effects of mutations on antagonist sensitivity, and the proposed binding mode explains variation in antagonist sensitivity between the human and rat P2X7 receptors. We have also determined the site of action for the P2X7R-selective antagonists ZINC58368839, brilliant blue G, KN-62, and calmidazolium. The effect of intersubunit allosteric pocket "signature mutants" F88A, T90V, D92A, F103A, and V312A on antagonist sensitivity suggests that ZINC58368839 comprises a binding mode similar to AZ11645373 and other previously characterized antagonists. For the larger antagonists, brilliant blue G, KN-62, and calmidazolium, our data imply an overlapping but distinct binding mode involving the central upper vestibule of the receptor in addition to the intersubunit allosteric pocket. Our work explains the site of action for a series of P2X7R antagonists and establishes "signature mutants" for P2X7R binding-mode characterization.


Asunto(s)
Mutación Puntual , Antagonistas del Receptor Purinérgico P2X/farmacología , Receptores Purinérgicos P2X7/química , Receptores Purinérgicos P2X7/metabolismo , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/química , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Adenosina Trifosfato/metabolismo , Sitio Alostérico , Amidas/química , Amidas/farmacología , Sitios de Unión , Humanos , Imidazoles/química , Imidazoles/farmacología , Indoles/química , Indoles/farmacología , Modelos Moleculares , Simulación del Acoplamiento Molecular , Antagonistas del Receptor Purinérgico P2X/química , Receptores Purinérgicos P2X7/genética , Colorantes de Rosanilina/química , Colorantes de Rosanilina/farmacología , Tiazoles/química , Tiazoles/farmacología
8.
J Biol Chem ; 293(33): 12820-12831, 2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-29997254

RESUMEN

ATP is the native agonist for cell-surface ligand-gated P2X receptor (P2XR) cation channels. The seven mammalian subunits (P2X1-7) form homo- and heterotrimeric P2XRs having significant physiological and pathophysiological roles. Pyridoxalphosphate-6-azophenyl-2',4'-disulfonic acid (PPADS) is an effective antagonist at most mammalian P2XRs. Lys-249 in the extracellular domain of P2XR has previously been shown to contribute to PPADS action. To map this antagonist site, we generated human P2X1R cysteine substitutions within a circle centered at Lys-249 (with a radius of 13 Å equal to the length of PPADS). We hypothesized that cysteine substitutions of residues involved in PPADS binding would (i) reduce cysteine accessibility (measured by MTSEA-biotinylation), (ii) exhibit altered PPADS affinity, and (iii) quench the fluorescence of cysteine residues modified with MTS-TAMRA. Of the 26 residues tested, these criteria were met by only four (Lys-70, Asp-170, Lys-190, and Lys-249), defining the antagonist site, validating molecular docking results, and thereby providing the first experimentally supported model of PPADS binding. This binding site overlapped with the ATP-binding site, indicating that PPADS sterically blocks agonist access. Moreover, PPADS induced a conformational change at the cysteine-rich head (CRH) region adjacent to the orthosteric ATP-binding pocket. The importance of this movement was confirmed by demonstrating that substitution introducing positive charge present in the CRH of the hP2X1R causes PPADS sensitivity at the normally insensitive rat P2X4R. This study provides a template for developing P2XR subtype selectivity based on the differences among the mammalian subunits around the orthosteric P2XR-binding site and the CRH.


Asunto(s)
Modelos Moleculares , Antagonistas del Receptor Purinérgico P2X/química , Fosfato de Piridoxal/análogos & derivados , Receptores Purinérgicos P2X1/química , Animales , Sitios de Unión , Humanos , Fosfato de Piridoxal/química , Ratas , Receptores Purinérgicos P2X1/genética , Receptores Purinérgicos P2X4/química , Receptores Purinérgicos P2X4/genética , Xenopus laevis
9.
Neurobiol Dis ; 127: 242-252, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30872158

RESUMEN

Niemann-Pick type C disease (NPCD) is a neurodegenerative disease associated with increases in cellular cholesterol and glycolipids and most commonly caused by defective NPC1, a late endosomal protein. Using ratiometric probes we find that NPCD cells show increased endolysosomal pH. In addition U18666A, an inhibitor of NPC1, was found to increase endolysosomal pH, and the number, size and heterogeneity of endolysosomal vesicles. NPCD fibroblasts and cells treated with U18666A also show disrupted targeting of fluorescent lipid BODIPY-LacCer to high pH vesicles. Inhibiting non-lysosomal glucocerebrosidase (GBA2) reversed increases in endolysosomal pH and restored disrupted BODIPY-LacCer trafficking in NPCD fibroblasts. GBA2 KO cells also show decreased endolysosomal pH. NPCD fibroblasts also show increased expression of a key subunit of the lysosomal proton pump vATPase on GBA2 inhibition. The results are consistent with a model where both endolysosomal pH and Golgi targeting of BODIPY-LacCer are dependent on adequate levels of cytosolic-facing GlcCer, which are reduced in NPC disease.


Asunto(s)
Citosol/metabolismo , Endosomas/metabolismo , Glucosilceramidas/metabolismo , Lisosomas/metabolismo , Enfermedad de Niemann-Pick Tipo C/metabolismo , Androstenos/farmacología , Animales , Citosol/efectos de los fármacos , Endosomas/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Humanos , Lisosomas/efectos de los fármacos , Ratones , Proteína Niemann-Pick C1/antagonistas & inhibidores
10.
J Chem Phys ; 150(5): 054504, 2019 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-30736690

RESUMEN

We employ the well-tempered parallel-bias metadynamics algorithm to study the stability of nanoparticles in a lattice gas for crystallization from solution. The model allows us to give a description for the transition from amorphous to crystalline nanoparticles by introducing parameters directly related to the surface tensions of the two phases and also the differences of the entropy per particle in each phase. By examining the parameter space, we find a critical cluster size of crystalline stability, whose temperature and size dependencies follow the Gibbs-Thomson equation. An additional melting point depression due to cluster surface fluctuations is observed, leading to a non-classical nucleation barrier of cluster growth.

11.
Proc Natl Acad Sci U S A ; 113(14): 3785-90, 2016 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-27006498

RESUMEN

Heme iron has many and varied roles in biology. Most commonly it binds as a prosthetic group to proteins, and it has been widely supposed and amply demonstrated that subtle variations in the protein structure around the heme, including the heme ligands, are used to control the reactivity of the metal ion. However, the role of heme in biology now appears to also include a regulatory responsibility in the cell; this includes regulation of ion channel function. In this work, we show that cardiac KATP channels are regulated by heme. We identify a cytoplasmic heme-binding CXXHX16H motif on the sulphonylurea receptor subunit of the channel, and mutagenesis together with quantitative and spectroscopic analyses of heme-binding and single channel experiments identified Cys628 and His648 as important for heme binding. We discuss the wider implications of these findings and we use the information to present hypotheses for mechanisms of heme-dependent regulation across other ion channels.


Asunto(s)
Hemo/metabolismo , Canales KATP/metabolismo , Receptores de Sulfonilureas/química , Secuencias de Aminoácidos/genética , Animales , Línea Celular , Células HEK293 , Humanos , Canales KATP/genética , Miocardio/metabolismo , Unión Proteica/genética , Estructura Terciaria de Proteína , Ratas , Ratas Wistar , Receptores de Sulfonilureas/genética
12.
Int J Mol Sci ; 20(3)2019 Feb 07.
Artículo en Inglés | MEDLINE | ID: mdl-30736449

RESUMEN

The accumulation of lipids in the late endosomes and lysosomes of Niemann⁻Pick type C disease (NPCD) cells is a consequence of the dysfunction of one protein (usually NPC1) but induces dysfunction in many proteins. We used molecular docking to propose (a) that NPC1 exports not just cholesterol, but also sphingosine, (b) that the cholesterol sensitivity of big potassium channel (BK) can be traced to a previously unappreciated site on the channel's voltage sensor, (c) that transient receptor potential mucolipin 1 (TRPML1) inhibition by sphingomyelin is likely an indirect effect, and (d) that phosphoinositides are responsible for both the mislocalization of annexin A2 (AnxA2) and a soluble NSF (N-ethylmaleimide Sensitive Fusion) protein attachment receptor (SNARE) recycling defect. These results are set in the context of existing knowledge of NPCD to sketch an account of the endolysosomal pathology key to this disease.


Asunto(s)
Lípidos/química , Simulación del Acoplamiento Molecular , Simulación de Dinámica Molecular , Proteínas/química , Membrana Celular/química , Membrana Celular/metabolismo , Colesterol/química , Colesterol/metabolismo , Lisosomas , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/metabolismo , Conformación Molecular , Enfermedad de Niemann-Pick Tipo C , Fosfatidilinositoles/química , Fosfatidilinositoles/metabolismo , Unión Proteica , Relación Estructura-Actividad Cuantitativa , Proteínas SNARE/química , Proteínas SNARE/metabolismo
13.
Mol Pharmacol ; 93(5): 553-562, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29535152

RESUMEN

P2X7 receptor (P2X7R) activation requires ∼100-fold higher concentrations of ATP than other P2X receptor (P2XR) subtypes. Such high levels are found during cellular stress, and P2X7Rs consequently contribute to a range of pathophysiological conditions. We have used chimeric and mutant P2X7Rs, coupled with molecular modeling, to produce a validated model of the binding mode of the subtype-selective antagonist A438079 at an intersubunit allosteric site. Within the allosteric site large effects on antagonist action were found for point mutants of residues F88A, D92A, F95A, and F103A that were conserved or similar between sensitive/insensitive P2XR subtypes, suggesting that these side-chain interactions were not solely responsible for high-affinity antagonist binding. Antagonist sensitivity was increased with mutations that remove the bulk of side chains around the center of the binding pocket, suggesting that the dimensions of the pocket make a significant contribution to selectivity. Chimeric receptors swapping the left flipper (around the orthosteric site) reduced both ATP and antagonist sensitivity. Point mutations within this region highlighted the contribution of a P2X7R-specific aspartic acid residue (D280) that modeling suggests forms a salt bridge with the lower body region of the receptor. The D280A mutant removing this charge increased ATP potency 15-fold providing a new insight into the low ATP sensitivity of the P2X7R. The ortho- and allosteric binding sites form either side of the ß-strand Y291-E301 adjacent to the left flipper. This structural linking may explain the contribution of the left flipper to both agonist and antagonist action.


Asunto(s)
Acetamidas/farmacología , Antagonistas del Receptor Purinérgico P2X/farmacología , Piridinas/farmacología , Quinolinas/farmacología , Receptores Purinérgicos P2X7/efectos de los fármacos , Tetrazoles/farmacología , Regulación Alostérica , Sitio Alostérico , Secuencia de Aminoácidos , Sitios de Unión , Humanos , Ligandos , Simulación del Acoplamiento Molecular , Mutación Puntual , Receptores Purinérgicos P2X7/química , Receptores Purinérgicos P2X7/genética , Homología de Secuencia de Aminoácido
15.
J Biol Chem ; 290(3): 1559-69, 2015 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-25425641

RESUMEN

P2X receptor subtype-selective antagonists are promising candidates for treatment of a range of pathophysiological conditions. However, in contrast to high resolution structural understanding of agonist action in the receptors, comparatively little is known about the molecular basis of antagonist binding. We have generated chimeras and point mutations in the extracellular ligand-binding loop of the human P2X1 receptor, which is inhibited by NF449, suramin, and pyridoxal-phosphate-6-azophenyl-2,4-disulfonate, with residues from the rat P2X4 receptor, which is insensitive to these antagonists. There was little or no effect on sensitivity to suramin and pyridoxal-phosphate-6-azophenyl-2,4-disulfonate in chimeric P2X1/4 receptors, indicating that a significant number of residues required for binding of these antagonists are present in the P2X4 receptor. Sensitivity to the P2X1 receptor-selective antagonist NF449 was reduced by ∼60- and ∼135-fold in chimeras replacing the cysteine-rich head, and the dorsal fin region below it in the adjacent subunit, respectively. Point mutants identified the importance of four positively charged residues at the base of the cysteine-rich head and two variant residues in the dorsal fin for high affinity NF449 binding. These six residues were used as the starting area for molecular docking. The four best potential NF449-binding poses were then discriminated by correspondence with the mutagenesis data and an additional mutant to validate the binding of one lobe of NF449 within the core conserved ATP-binding pocket and the other lobes coordinated by positive charge on the cysteine-rich head region and residues in the adjacent dorsal fin.


Asunto(s)
Adenosina Trifosfato/química , Bencenosulfonatos/química , Mutación Puntual , Antagonistas del Receptor Purinérgico P2X/química , Animales , Sitios de Unión , Cisteína/química , Diseño de Fármacos , Humanos , Ligandos , Lisina/química , Ratones , Modelos Moleculares , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Oocitos/metabolismo , Unión Proteica , Ratas , Suramina/química , Xenopus laevis
16.
PLoS Comput Biol ; 11(10): e1004548, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26513470

RESUMEN

Separases are large proteins that mediate sister chromatid disjunction in all eukaryotes. They belong to clan CD of cysteine peptidases and contain a well-conserved C-terminal catalytic protease domain similar to caspases and gingipains. However, unlike other well-characterized groups of clan CD peptidases, there are no high-resolution structures of separases and the details of their regulation and substrate recognition are poorly understood. Here we undertook an in-depth bioinformatical analysis of separases from different species with respect to their similarity in amino acid sequence and protein fold in comparison to caspases, MALT-1 proteins (mucosa-associated lymphoidtissue lymphoma translocation protein 1) and gingipain-R. A comparative model of the single C-terminal caspase-like domain in separase from C. elegans suggests similar binding modes of substrate peptides between these protein subfamilies, and enables differences in substrate specificity of separase proteins to be rationalised. We also modelled a newly identified putative death domain, located N-terminal to the caspase-like domain. The surface features of this domain identify potential sites of protein-protein interactions. Notably, we identified a novel conserved region with the consensus sequence WWxxRxxLD predicted to be exposed on the surface of the death domain, which we termed the WR motif. We envisage that findings from our study will guide structural and functional studies of this important protein family.


Asunto(s)
Caspasas/química , Caspasas/ultraestructura , Simulación del Acoplamiento Molecular , Receptores de Muerte Celular/química , Separasa/química , Separasa/ultraestructura , Adhesinas Bacterianas/química , Adhesinas Bacterianas/ultraestructura , Secuencia de Aminoácidos , Sitios de Unión , Cisteína Endopeptidasas/química , Cisteína Endopeptidasas/ultraestructura , Activación Enzimática , Cisteína-Endopeptidasas Gingipaínas , Modelos Químicos , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Receptores de Muerte Celular/ultraestructura , Relación Estructura-Actividad , Especificidad por Sustrato
17.
Nucleic Acids Res ; 41(22): 10170-84, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24038356

RESUMEN

Meiosis requires conserved transcriptional changes, but it is not known whether there is a corresponding set of RNA splicing switches. Here, we used RNAseq of mouse testis to identify changes associated with the progression from mitotic spermatogonia to meiotic spermatocytes. We identified ∼150 splicing switches, most of which affect conserved protein-coding exons. The expression of many key splicing regulators changed in the course of meiosis, including downregulation of polypyrimidine tract binding protein (PTBP1) and heterogeneous nuclear RNP A1, and upregulation of nPTB, Tra2ß, muscleblind, CELF proteins, Sam68 and T-STAR. The sequences near the regulated exons were significantly enriched in target sites for PTB, Tra2ß and STAR proteins. Reporter minigene experiments investigating representative exons in transfected cells showed that PTB binding sites were critical for splicing of a cassette exon in the Ralgps2 mRNA and a shift in alternative 5' splice site usage in the Bptf mRNA. We speculate that nPTB might functionally replace PTBP1 during meiosis for some target exons, with changes in the expression of other splicing factors helping to establish meiotic splicing patterns. Our data suggest that there are substantial changes in the determinants and patterns of alternative splicing in the mitotic-to-meiotic transition of the germ cell cycle.


Asunto(s)
Empalme Alternativo , Meiosis/genética , Testículo/metabolismo , Animales , Secuencia de Bases , Exones , Masculino , Ratones , Datos de Secuencia Molecular , Isoformas de ARN/metabolismo , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Análisis de Secuencia de ARN , Espermatocitos/metabolismo , Espermatogonias/metabolismo , Transcriptoma
18.
Proc Natl Acad Sci U S A ; 109(12): 4663-7, 2012 Mar 20.
Artículo en Inglés | MEDLINE | ID: mdl-22393010

RESUMEN

P2X receptors for ATP have a wide range of physiological roles and comprise a structurally distinct family of ligand-gated trimeric ion channels. The crystal structure of a P2X4 receptor, in combination with mutagenesis studies, has provided a model of the intersubunit ATP-binding sites and identified an extracellular lateral portal, adjacent to the membrane, that funnels ions to the channel pore. However, little is known about the extent of ATP-induced conformational changes in the extracellular domain of the receptor. To address this issue, we have used MTSEA-biotinylation (N-Biotinoylaminoethyl methanethiosulfonate) to show ATP-sensitive accessibility of cysteine mutants at the human P2X1 receptor. Mapping these data to a P2X1 receptor homology model identifies significant conformational rearrangement. Electron microscopy of purified P2X1 receptors showed marked changes in structure on ATP binding, and introducing disulphide bonds between adjacent subunits to restrict intersubunit movements inhibited channel function. These results are consistent with agonist-induced rotation of the propeller-head domain of the receptor, sliding of adjacent subunits leading to restricted access to the upper vestibule, movement in the ion conducting lateral portals, and gating of the channel pore.


Asunto(s)
Receptores Purinérgicos P2X1/química , Adenosina Trifosfato/química , Animales , Sitios de Unión , Biotinilación , Disulfuros/química , Humanos , Iones/química , Microscopía Electrónica/métodos , Conformación Molecular , Mutagénesis , Oocitos/metabolismo , Unión Proteica , Conformación Proteica , Estructura Terciaria de Proteína , Xenopus
19.
J Cell Biochem ; 115(3): 557-65, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24123053

RESUMEN

Phospholipase C-η (PLCη) enzymes are a class of phosphatidylinositol 4,5-bisphosphate-hydrolyzing enzymes involved in intracellular signaling. PLCη2 can sense Ca(2+) (stimulated by ∼1 µM free Ca(2+) ) suggesting that it can amplify transient Ca(2+) signals. PLCη enzymes possess an EF-hand domain composed of two EF-loops; a canonical 12-residue loop (EF-loop 1) and a non-canonical 13-residue loop (EF-loop 2). Ca(2+) -binding to synthetic peptides corresponding to EF-loops 1 and 2 of PLCη2 and EF-loop 1 of calmodulin (as a control) was examined by 2D-[(1) H,(1) H] TOCSY NMR. Both PLCη2 EF-loop peptides bound Ca(2+) in a similar manner to that of the canonical calmodulin EF-loop 1, particularly at their N-terminus. A molecular model of the PLCη2 EF-hand domain, constructed based upon the structure of calmodulin, suggested both EF-loops may participate in Ca(2+) -binding. To determine whether the EF-hand is responsible for Ca(2+) -sensing, inositol phosphate accumulation was measured in COS7 cells transiently expressing wild-type or mutant PLCη2 proteins. Addition of 70 µM monensin (a Na(+) /H(+) antiporter that increases intracellular Ca(2+) ) induced a 4- to 7-fold increase in wild-type PLCη2 activity. In permeabilized cells, PLCη2 exhibited a ∼4-fold increase in activity in the presence of 1 µM free Ca(2+) . The D256A (EF-loop1) mutant exhibited a ∼10-fold reduction in Ca(2+) -sensitivity and was not activated by monensin, highlighting the involvement of EF-loop 1 in Ca(2+) -sensing. Involvement of EF-loop 2 was examined using D292A, H296A, Q297A, and E304A mutants. Interestingly, the monensin responses and Ca(2+) -sensitivities were largely unaffected by the mutations, indicating that the non-canonical EF-loop 2 is not involved in Ca(2+) -sensing.


Asunto(s)
Calcio/metabolismo , Modelos Moleculares , Fosfoinositido Fosfolipasa C/química , Conformación Proteica , Secuencia de Aminoácidos , Animales , Células COS , Calcio/química , Calmodulina/química , Chlorocebus aethiops , Motivos EF Hand/genética , Humanos , Inositol/farmacología , Mutación/genética , Fosfoinositido Fosfolipasa C/metabolismo , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Transducción de Señal , Relación Estructura-Actividad
20.
Nat Genet ; 36(12): 1259-67, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15543149

RESUMEN

The phylum Nematoda occupies a huge range of ecological niches, from free-living microbivores to human parasites. We analyzed the genomic biology of the phylum using 265,494 expressed-sequence tag sequences, corresponding to 93,645 putative genes, from 30 species, including 28 parasites. From 35% to 70% of each species' genes had significant similarity to proteins from the model nematode Caenorhabditis elegans. More than half of the putative genes were unique to the phylum, and 23% were unique to the species from which they were derived. We have not yet come close to exhausting the genomic diversity of the phylum. We identified more than 2,600 different known protein domains, some of which had differential abundances between major taxonomic groups of nematodes. We also defined 4,228 nematode-specific protein families from nematode-restricted genes: this class of genes probably underpins species- and higher-level taxonomic disparity. Nematode-specific families are particularly interesting as drug and vaccine targets.


Asunto(s)
Evolución Molecular , Etiquetas de Secuencia Expresada , Variación Genética , Genoma , Nematodos/genética , Estructura Terciaria de Proteína/genética , Animales , Secuencia de Bases , Mapeo Cromosómico , Biología Computacional , Secuencia Conservada/genética , Bases de Datos Genéticas , Datos de Secuencia Molecular , Filogenia , Análisis de Secuencia de ADN , Especificidad de la Especie
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA