Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Life Sci Alliance ; 3(1)2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31826928

RESUMEN

Host interferon-induced transmembrane proteins (IFITMs) are broad-spectrum antiviral restriction factors. Of these, IFITM3 potently inhibits viruses that enter cells through acidic endosomes, many of which are zoonotic and emerging viruses with bats (order Chiroptera) as their natural hosts. We previously demonstrated that microbat IFITM3 is antiviral. Here, we show that bat IFITMs are characterized by strong adaptive evolution and identify a highly variable and functionally important site-codon 70-within the conserved CD225 domain of IFITMs. Mutation of this residue in microbat IFITM3 impairs restriction of representatives of four different virus families that enter cells via endosomes. This mutant shows altered subcellular localization and reduced S-palmitoylation, a phenotype copied by mutation of conserved cysteine residues in microbat IFITM3. Furthermore, we show that microbat IFITM3 is S-palmitoylated on cysteine residues C71, C72, and C105, mutation of each cysteine individually impairs virus restriction, and a triple C71A-C72A-C105A mutant loses all restriction activity, concomitant with subcellular re-localization of microbat IFITM3 to Golgi-associated sites. Thus, we propose that S-palmitoylation is critical for Chiropteran IFITM3 function and identify a key molecular determinant of IFITM3 S-palmitoylation.


Asunto(s)
Quirópteros/genética , Lipoilación/genética , Proteínas de la Membrana/genética , Polimorfismo Genético , Dominios Proteicos/genética , Proteínas de Unión al ARN/genética , Células A549 , Animales , Antígenos de Diferenciación/genética , Codón/genética , Codón/metabolismo , Endosomas/metabolismo , Endosomas/virología , Evolución Molecular , Humanos , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/metabolismo , Gripe Humana/virología , Proteínas de la Membrana/metabolismo , Filogenia , Proteínas de Unión al ARN/metabolismo , Transducción Genética , Internalización del Virus , Virus Zika/fisiología , Infección por el Virus Zika/metabolismo , Infección por el Virus Zika/virología
3.
J Lipid Res ; 55(9): 1925-32, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24927729

RESUMEN

Hepatic conversion to bile acids is a major elimination route for cholesterol in mammals. CYP7A1 catalyzes the first and rate-limiting step in classic bile acid biosynthesis, converting cholesterol to 7α-hydroxycholesterol. To identify the structural determinants that govern the stereospecific hydroxylation of cholesterol, we solved the crystal structure of CYP7A1 in the ligand-free state. The structure-based mutation T104L in the B' helix, corresponding to the nonpolar residue of CYP7B1, was used to obtain crystals of complexes with cholest-4-en-3-one and with cholesterol oxidation product 7-ketocholesterol (7KCh). The structures reveal a motif of residues that promote cholest-4-en-3-one binding parallel to the heme, thus positioning the C7 atom for hydroxylation. Additional regions of the binding cavity (most distant from the access channel) are involved to accommodate the elongated conformation of the aliphatic side chain. Structural complex with 7KCh shows an active site rigidity and provides an explanation for its inhibitory effect. Based on our previously published data, we proposed a model of cholesterol abstraction from the membrane by CYP7A1 for metabolism. CYP7A1 structural data provide a molecular basis for understanding of the diversity of 7α-hydroxylases, on the one hand, and cholesterol-metabolizing enzymes adapted for their specific activity, on the other hand.


Asunto(s)
Colesterol 7-alfa-Hidroxilasa/química , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Dominio Catalítico , Colesterol 7-alfa-Hidroxilasa/genética , Cristalografía por Rayos X , Inhibidores Enzimáticos/química , Humanos , Enlace de Hidrógeno , Hidroxilación , Cetocolesteroles/química , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Secundaria de Proteína
4.
FEBS J ; 281(6): 1700-13, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24491228

RESUMEN

Oxysterols and neurosteroids are important signaling molecules produced by monooxygenases of the cytochrome P450 family that realize their effect through nuclear receptors. CYP7B1 catalyzes the 6- or 7-hydroxylation of both steroids and oxysterols and thus is involved in the metabolism of neurosteroids and bile acid synthesis, respectively. The dual physiological role of CYP7B1 is evidenced from different diseases, liver failure and progressive neuropathy, caused by enzyme malfunction. Here we present biochemical characterization of CYP7B1 at the molecular level to understand substrate specificity and susceptibility to azole drugs. Based on our experiments with purified enzyme, the requirements for CYP7B1 hydroxylation of steroid molecules are as follows: C5 hydrogen in the α-configuration (or double bond at C5), a polar group at C17, a hydroxyl group at C3, and the absence of the hydroxyl group at C20-C24 in the C27-sterol side chain. 21-hydroxy-pregnenolone was identified as a new substrate, and overall low activity toward pregnanes could be related to the increased potency of 7-hydroxy derivatives produced by CYP7B1. Metabolic conversion (deactivation) of oxysterols by CYP7B1 in a reconstituted system proceeds via two sequential hydroxylations. Two mutations that are found in patients with diseases, Gly57Arg and Phe216Ser, result in apo-P450 (devoid of heme) protein formation. Our CYP7B1 homology model provides a rationale for understanding clinical mutations and relatively broad substrate specificity for steroid hydroxylase.


Asunto(s)
Esteroide Hidroxilasas/química , Esteroide Hidroxilasas/metabolismo , 17-alfa-Hidroxipregnenolona/metabolismo , Sustitución de Aminoácidos , Azoles/metabolismo , Dominio Catalítico , Familia 7 del Citocromo P450 , Humanos , Cinética , Modelos Moleculares , Mutagénesis Sitio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Pliegue de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Espectrofotometría , Esteroide Hidroxilasas/genética , Homología Estructural de Proteína , Especificidad por Sustrato
5.
PLoS One ; 7(3): e33943, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22470497

RESUMEN

Kinesin-1 transports various cargos along the axon by interacting with the cargos through its light chain subunit. Kinesin light chains (KLC) utilize its tetratricopeptide repeat (TPR) domain to interact with over 10 different cargos. Despite a high sequence identity between their TPR domains (87%), KLC1 and KLC2 isoforms exhibit differential binding properties towards some cargos. We determined the structures of human KLC1 and KLC2 tetratricopeptide repeat (TPR) domains using X-ray crystallography and investigated the different mechanisms by which KLCs interact with their cargos. Using isothermal titration calorimetry, we attributed the specific interaction between KLC1 and JNK-interacting protein 1 (JIP1) cargo to residue N343 in the fourth TRP repeat. Structurally, the N343 residue is adjacent to other asparagines and lysines, creating a positively charged polar patch within the groove of the TPR domain. Whereas, KLC2 with the corresponding residue S328 did not interact with JIP1. Based on these finding, we propose that N343 of KLC1 can form "a carboxylate clamp" with its neighboring asparagine to interact with JIP1, similar to that of HSP70/HSP90 organizing protein-1's (HOP1) interaction with heat shock proteins. For the binding of cargos shared by KLC1 and KLC2, we propose a different site located within the groove but not involving N343. We further propose a third binding site on KLC1 which involves a stretch of polar residues along the inter-TPR loops that may form a network of hydrogen bonds to JIP3 and JIP4. Together, these results provide structural insights into possible mechanisms of interaction between KLC TPR domains and various cargo proteins.


Asunto(s)
Cinesinas/química , Modelos Moleculares , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Datos de Secuencia Molecular , Isoformas de Proteínas/química , Isoformas de Proteínas/metabolismo , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Alineación de Secuencia
6.
PLoS One ; 7(2): e30375, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22363433

RESUMEN

SMN (Survival motor neuron protein) was characterized as a dimethyl-arginine binding protein over ten years ago. TDRD3 (Tudor domain-containing protein 3) and SPF30 (Splicing factor 30 kDa) were found to bind to various methyl-arginine proteins including Sm proteins as well later on. Recently, TDRD3 was shown to be a transcriptional coactivator, and its transcriptional activity is dependent on its ability to bind arginine-methylated histone marks. In this study, we systematically characterized the binding specificity and affinity of the Tudor domains of these three proteins quantitatively. Our results show that TDRD3 preferentially recognizes asymmetrical dimethylated arginine mark, and SMN is a very promiscuous effector molecule, which recognizes different arginine containing sequence motifs and preferentially binds symmetrical dimethylated arginine. SPF30 is the weakest methyl-arginine binder, which only binds the GAR motif sequences in our library. In addition, we also reported high-resolution crystal structures of the Tudor domain of TDRD3 in complex with two small molecules, which occupy the aromatic cage of TDRD3.


Asunto(s)
Arginina/análogos & derivados , Proteínas/química , Proteínas del Complejo SMN/química , Secuencia de Aminoácidos , Arginina/metabolismo , Cristalografía por Rayos X , Polarización de Fluorescencia , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Proteínas/metabolismo , Factores de Empalme de ARN , Proteínas del Complejo SMN/metabolismo , Homología Estructural de Proteína
7.
PLoS One ; 7(12): e52977, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23285239

RESUMEN

SUV39H1, the first identified histone lysine methyltransferase in human, is involved in chromatin modification and gene regulation. SUV39H1 contains a chromodomain in its N-terminus, which potentially plays a role in methyl-lysine recognition and SUV39H1 targeting. In this study, the structure of the chromodomain of human SUV39H1 was determined by X-ray crystallography. The SUV39H1 chromodomain displays a generally conserved structure fold compared with other solved chromodomains. However, different from other chromodomains, the SUV39H1 chromodomain possesses a much longer helix at its C-terminus. Furthermore, the SUV39H1 chromodomain was shown to recognize histone H3K9me2/3 specifically.


Asunto(s)
Histonas/química , Histonas/metabolismo , Metiltransferasas/química , Proteínas Represoras/química , Secuencia de Aminoácidos , Cristalización , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Lisina/química , Lisina/metabolismo , Metilación , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica/fisiología , Dominios y Motivos de Interacción de Proteínas/fisiología , Mapas de Interacción de Proteínas , Estructura Secundaria de Proteína , Homología de Secuencia de Aminoácido
8.
J Biol Chem ; 287(4): 2317-27, 2012 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-22069321

RESUMEN

The aggresome pathway is activated when proteasomal clearance of misfolded proteins is hindered. Misfolded polyubiquitinated protein aggregates are recruited and transported to the aggresome via the microtubule network by a protein complex consisting of histone deacetylase 6 (HDAC6) and the dynein motor complex. The current model suggests that HDAC6 recognizes protein aggregates by binding directly to polyubiquitinated proteins. Here, we show that there are substantial amounts of unanchored ubiquitin in protein aggregates with solvent-accessible C termini. The ubiquitin-binding domain (ZnF-UBP) of HDAC6 binds exclusively to the unanchored C-terminal diglycine motif of ubiquitin instead of conjugated polyubiquitin. The unanchored ubiquitin C termini in the aggregates are generated in situ by aggregate-associated deubiquitinase ataxin-3. These results provide structural and mechanistic bases for the role of HDAC6 in aggresome formation and further suggest a novel ubiquitin-mediated signaling pathway, where the exposure of ubiquitin C termini within protein aggregates enables HDAC6 recognition and transport to the aggresome.


Asunto(s)
Histona Desacetilasas/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Transducción de Señal/fisiología , Ubiquitina/metabolismo , Proteínas Ubiquitinadas/metabolismo , Ataxina-3 , Línea Celular , Histona Desacetilasa 6 , Histona Desacetilasas/genética , Humanos , Complejos Multiproteicos/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Estructura Terciaria de Proteína , Transporte de Proteínas/fisiología , Proteínas Represoras/genética , Ubiquitina/genética , Proteínas Ubiquitinadas/genética
9.
BMC Genomics ; 12: 478, 2011 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-21962082

RESUMEN

BACKGROUND: Hundreds of millions of people are infected with cryptosporidiosis annually, with immunocompromised individuals suffering debilitating symptoms and children in socioeconomically challenged regions at risk of repeated infections. There is currently no effective drug available. In order to facilitate the pursuit of anti-cryptosporidiosis targets and compounds, our study spans the classification of the Cryptosporidium parvum kinome and the structural and biochemical characterization of representatives from the CDPK family and a MAP kinase. RESULTS: The C. parvum kinome comprises over 70 members, some of which may be promising drug targets. These C. parvum protein kinases include members in the AGC, Atypical, CaMK, CK1, CMGC, and TKL groups; however, almost 35% could only be classified as OPK (other protein kinases). In addition, about 25% of the kinases identified did not have any known orthologues outside of Cryptosporidium spp. Comparison of specific kinases with their Plasmodium falciparum and Toxoplasma gondii orthologues revealed some distinct characteristics within the C. parvum kinome, including potential targets and opportunities for drug design. Structural and biochemical analysis of 4 representatives of the CaMK group and a MAP kinase confirms features that may be exploited in inhibitor design. Indeed, screening CpCDPK1 against a library of kinase inhibitors yielded a set of the pyrazolopyrimidine derivatives (PP1-derivatives) with IC50 values of < 10 nM. The binding of a PP1-derivative is further described by an inhibitor-bound crystal structure of CpCDPK1. In addition, structural analysis of CpCDPK4 identified an unprecedented Zn-finger within the CDPK kinase domain that may have implications for its regulation. CONCLUSIONS: Identification and comparison of the C. parvum protein kinases against other parasitic kinases shows how orthologue- and family-based research can be used to facilitate characterization of promising drug targets and the search for new drugs.


Asunto(s)
Cryptosporidium parvum/enzimología , Proteínas Quinasas/análisis , Proteínas Protozoarias/análisis , Cryptosporidium parvum/genética , Bases de Datos de Proteínas , Plasmodium falciparum/enzimología , Proteínas Quinasas/clasificación , Proteínas Quinasas/genética , Estructura Terciaria de Proteína , Proteínas Protozoarias/clasificación , Proteínas Protozoarias/genética , Toxoplasma/enzimología
10.
Biochem Biophys Res Commun ; 413(1): 58-61, 2011 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-21888896

RESUMEN

DPF2 is an evolutionary highly conserved member of the d4-protein family characterized by an N-terminal 2/3 domain, a C2H2-type zinc finger (ZF), and a C-terminal tandem PHD zinc finger. DPF2 is identified as a transcription factor and may be related with some cancers in human. Here, we report the crystal structure of the C2H2-type zinc finger domain of human DPF2 with a canonical C2H2 fold, which contains two beta strands and one alpha helix. Several conserved residues, including Lys207, Lys216 and Arg217, constitute a positively charged surface in C2H2 domain, which implicates that it has the potential to bind DNA. The side chains of the residues Y209, C211, C214, K216, Y218, L224, H227 and H232 form the hydrophobic core of C2H2 domain, which indicates a potential-binding surface in the human DPF2.


Asunto(s)
Proteínas de Unión al ADN/química , Dedos de Zinc , Secuencia de Aminoácidos , Cristalografía por Rayos X , Cisteína/química , Histidina/química , Humanos , Datos de Secuencia Molecular , Pliegue de Proteína , Factores de Transcripción
11.
Proc Natl Acad Sci U S A ; 108(25): 10139-43, 2011 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-21636783

RESUMEN

In humans, the precursor to all steroid hormones, pregnenolone, is synthesized from cholesterol by an enzyme complex comprising adrenodoxin reductase (AdR), adrenodoxin (Adx), and a cytochrome P450 (P450scc or CYP11A1). This complex not only plays a key role in steroidogenesis, but also has long been a model to study electron transfer, multistep catalysis, and C-C bond cleavage performed by monooxygenases. Detailed mechanistic understanding of these processes has been hindered by a lack of structural information. Here we present the crystal structure of the complex of human Adx and CYP11A1--the first of a complex between a eukaryotic CYP and its redox partner. The structures with substrate and a series of reaction intermediates allow us to define the mechanism underlying sequential hydroxylations of the cholesterol and suggest the mechanism of C-C bond cleavage. In the complex the [2Fe-2S] cluster of Adx is positioned 17.4 Å away from the heme iron of CYP11A1. This structure suggests that after an initial protein-protein association driven by electrostatic forces, the complex adopts an optimized geometry between the redox centers. Conservation of the interaction interface suggests that this mechanism is common for all mitochondrial P450s.


Asunto(s)
Adrenodoxina/química , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/química , Mitocondrias/enzimología , Oxigenasas de Función Mixta/metabolismo , Pregnenolona/biosíntesis , Conformación Proteica , Adrenodoxina/metabolismo , Animales , Dominio Catalítico , Bovinos , Colesterol/metabolismo , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Humanos , Modelos Moleculares , Oxidación-Reducción , Unión Proteica , Electricidad Estática
12.
Proc Natl Acad Sci U S A ; 107(47): 20346-51, 2010 Nov 23.
Artículo en Inglés | MEDLINE | ID: mdl-21057110

RESUMEN

Phosphatidylinositol 3,4,5-triphosphate (PIP3) plays a key role in neuronal polarization and axon formation. PIP3-containing vesicles are transported to axon tips by the kinesin KIF13B via an adaptor protein, centaurin α1 (CENTA1). KIF13B interacts with CENTA1 through its forkhead-associated (FHA) domain. We solved the crystal structures of CENTA1 in ligand-free, KIF13B-FHA domain-bound, and PIP3 head group (IP4)-bound conformations, and the CENTA1/KIF13B-FHA/IP4 ternary complex. The first pleckstrin homology (PH) domain of CENTA1 specifically binds to PIP3, while the second binds to both PIP3 and phosphatidylinositol 3,4-biphosphate (PI(3,4)P(2)). The FHA domain of KIF13B interacts with the PH1 domain of one CENTA1 molecule and the ArfGAP domain of a second CENTA1 molecule in a threonine phosphorylation-independent fashion. We propose that full-length KIF13B and CENTA1 form heterotetramers that can bind four phosphoinositide molecules in the vesicle and transport it along the microtubule.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/química , Axones/metabolismo , Cinesinas/química , Modelos Moleculares , Proteínas del Tejido Nervioso/química , Neuronas/citología , Fosfatos de Fosfatidilinositol/metabolismo , Conformación Proteica , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Transporte Biológico/fisiología , Calorimetría , Cromatografía de Afinidad , Cromatografía en Gel , Clonación Molecular , Biología Computacional , Cristalografía , Electroforesis en Gel de Poliacrilamida , Glutatión Transferasa , Humanos , Cinesinas/genética , Cinesinas/metabolismo , Modelos Químicos , Mutagénesis Sitio-Dirigida , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Vesículas Transportadoras/metabolismo
13.
Proc Natl Acad Sci U S A ; 107(45): 19266-71, 2010 Nov 09.
Artículo en Inglés | MEDLINE | ID: mdl-20974918

RESUMEN

The polycomb repressive complex 2 (PRC2) is the major methyltransferase for H3K27 methylation, a modification critical for maintaining repressed gene expression programs throughout development. It has been previously shown that PRC2 maintains histone methylation patterns during DNA replication in part through its ability to bind to H3K27me3. However, the mechanism by which PRC2 recognizes H3K27me3 is unclear. Here we show that the WD40 domain of EED, a PRC2 component, is a methyllysine histone-binding domain. The crystal structures of apo-EED and EED in complex respectively with five different trimethyllysine histone peptides reveal that EED binds these peptides via the top face of its ß-propeller architecture. The ammonium group of the trimethyllysine is accommodated by an aromatic cage formed by three aromatic residues, while its aliphatic chain is flanked by a fourth aromatic residue. Our structural data provide an explanation for the preferential recognition of the Ala-Arg-Lys-Ser motif-containing trimethylated H3K27, H3K9, and H1K26 marks by EED over lower methylation states and other histone methyllysine marks. More importantly, we found that binding of different histone marks by EED differentially regulates the activity and specificity of PRC2. Whereas the H3K27me3 mark stimulates the histone methyltransferase activity of PRC2, the H1K26me3 mark inhibits PRC2 methyltransferase activity on the nucleosome. Moreover, H1K26me3 binding switches the specificity of PRC2 from methylating H3K27 to EED. In addition to determining the molecular basis of EED-methyllysine recognition, our work provides the biochemical characterization of how the activity of a histone methyltransferase is oppositely regulated by two histone marks.


Asunto(s)
Histonas/metabolismo , Proteínas Represoras/metabolismo , Sitios de Unión , Proteínas Portadoras/metabolismo , Línea Celular , Cristalografía por Rayos X , Proteínas de Unión al ADN/metabolismo , Proteína Potenciadora del Homólogo Zeste 2 , Humanos , Metilación , Metiltransferasas/metabolismo , Proteínas de Neoplasias , Proteínas Nucleares/metabolismo , Complejo Represivo Polycomb 2 , Unión Proteica , Conformación Proteica , Proteínas Represoras/química , Proteína 4 de Unión a Retinoblastoma/metabolismo , Proteína 7 de Unión a Retinoblastoma/metabolismo , Factores de Transcripción/metabolismo
14.
Proc Natl Acad Sci U S A ; 107(43): 18398-403, 2010 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-20937909

RESUMEN

Arginine methylation modulates diverse cellular processes and represents a molecular signature of germ-line-specific Piwi family proteins. A subset of Tudor domains recognize arginine methylation modifications, but the binding mechanism has been lacking. Here we establish that, like other germ-line Tudor proteins, the ancestral staphylococcal nuclease domain-containing 1 (SND1) polypeptide is expressed and associates with PIWIL1/Miwi in germ cells. We find that human SND1 binds PIWIL1 in an arginine methylation-dependent manner with a preference for symmetrically dimethylated arginine. The entire Tudor domain and a bifurcated SN domain are required for this binding activity, whereas the canonical Tudor domain alone is insufficient for methylarginine ligand binding. Crystal structures show that the intact SND1 extended Tudor domain forms a wide and negatively charged binding groove, which can accommodate distinct symmetrically dimethylated arginine peptides from PIWIL1 in different orientations. This analysis explains how SND1 preferentially recognizes symmetrical dimethylarginine via an aromatic cage and conserved hydrogen bonds, and provides a general paradigm for the binding mechanisms of methylarginine-containing peptides by extended Tudor domains.


Asunto(s)
Proteínas/química , Secuencia de Aminoácidos , Animales , Arginina/química , Proteínas Argonautas , Cristalografía por Rayos X , Endonucleasas , Humanos , Técnicas In Vitro , Masculino , Metilación , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Complejos Multiproteicos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas/genética , Proteínas/metabolismo , ARN Interferente Pequeño/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Testículo/metabolismo
15.
PLoS Biol ; 8(7): e1000439, 2010 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-20676357

RESUMEN

Peptidyl-prolyl isomerases catalyze the conversion between cis and trans isomers of proline. The cyclophilin family of peptidyl-prolyl isomerases is well known for being the target of the immunosuppressive drug cyclosporin, used to combat organ transplant rejection. There is great interest in both the substrate specificity of these enzymes and the design of isoform-selective ligands for them. However, the dearth of available data for individual family members inhibits attempts to design drug specificity; additionally, in order to define physiological functions for the cyclophilins, definitive isoform characterization is required. In the current study, enzymatic activity was assayed for 15 of the 17 human cyclophilin isomerase domains, and binding to the cyclosporin scaffold was tested. In order to rationalize the observed isoform diversity, the high-resolution crystallographic structures of seven cyclophilin domains were determined. These models, combined with seven previously solved cyclophilin isoforms, provide the basis for a family-wide structure:function analysis. Detailed structural analysis of the human cyclophilin isomerase explains why cyclophilin activity against short peptides is correlated with an ability to ligate cyclosporin and why certain isoforms are not competent for either activity. In addition, we find that regions of the isomerase domain outside the proline-binding surface impart isoform specificity for both in vivo substrates and drug design. We hypothesize that there is a well-defined molecular surface corresponding to the substrate-binding S2 position that is a site of diversity in the cyclophilin family. Computational simulations of substrate binding in this region support our observations. Our data indicate that unique isoform determinants exist that may be exploited for development of selective ligands and suggest that the currently available small-molecule and peptide-based ligands for this class of enzyme are insufficient for isoform specificity.


Asunto(s)
Ciclofilinas/química , Ciclofilinas/metabolismo , Familia de Multigenes , Isomerasa de Peptidilprolil/química , Isomerasa de Peptidilprolil/metabolismo , Secuencia de Aminoácidos , Dominio Catalítico , Humanos , Modelos Biológicos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Secundaria de Proteína
16.
Nat Struct Mol Biol ; 17(5): 596-601, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20436473

RESUMEN

Calcium-dependent protein kinases (CDPKs) have pivotal roles in the calcium-signaling pathway in plants, ciliates and apicomplexan parasites and comprise a calmodulin-dependent kinase (CaMK)-like kinase domain regulated by a calcium-binding domain in the C terminus. To understand this intramolecular mechanism of activation, we solved the structures of the autoinhibited (apo) and activated (calcium-bound) conformations of CDPKs from the apicomplexan parasites Toxoplasma gondii and Cryptosporidium parvum. In the apo form, the C-terminal CDPK activation domain (CAD) resembles a calmodulin protein with an unexpected long helix in the N terminus that inhibits the kinase domain in the same manner as CaMKII. Calcium binding triggers the reorganization of the CAD into a highly intricate fold, leading to its relocation around the base of the kinase domain to a site remote from the substrate binding site. This large conformational change constitutes a distinct mechanism in calcium signal-transduction pathways.


Asunto(s)
Calcio/metabolismo , Cryptosporidium parvum/enzimología , Proteínas Quinasas/química , Proteínas Quinasas/metabolismo , Toxoplasma/enzimología , Calcio/química , Cristalografía por Rayos X , Activación Enzimática , Modelos Moleculares , Unión Proteica , Conformación Proteica
17.
J Biol Chem ; 285(21): 16330-40, 2010 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-20299452

RESUMEN

Human choline kinase (ChoK) catalyzes the first reaction in phosphatidylcholine biosynthesis and exists as ChoKalpha (alpha1 and alpha2) and ChoKbeta isoforms. Recent studies suggest that ChoK is implicated in tumorigenesis and emerging as an attractive target for anticancer chemotherapy. To extend our understanding of the molecular mechanism of ChoK inhibition, we have determined the high resolution x-ray structures of the ChoKalpha1 and ChoKbeta isoforms in complex with hemicholinium-3 (HC-3), a known inhibitor of ChoK. In both structures, HC-3 bound at the conserved hydrophobic groove on the C-terminal lobe. One of the HC-3 oxazinium rings complexed with ChoKalpha1 occupied the choline-binding pocket, providing a structural explanation for its inhibitory action. Interestingly, the HC-3 molecule co-crystallized with ChoKbeta was phosphorylated in the choline binding site. This phosphorylation, albeit occurring at a very slow rate, was confirmed experimentally by mass spectroscopy and radioactive assays. Detailed kinetic studies revealed that HC-3 is a much more potent inhibitor for ChoKalpha isoforms (alpha1 and alpha2) compared with ChoKbeta. Mutational studies based on the structures of both inhibitor-bound ChoK complexes demonstrated that Leu-401 of ChoKalpha2 (equivalent to Leu-419 of ChoKalpha1), or the corresponding residue Phe-352 of ChoKbeta, which is one of the hydrophobic residues neighboring the active site, influences the plasticity of the HC-3-binding groove, thereby playing a key role in HC-3 sensitivity and phosphorylation.


Asunto(s)
Colina Quinasa/antagonistas & inhibidores , Colina Quinasa/química , Inhibidores Enzimáticos/química , Hemicolinio 3/química , Dominio Catalítico , Colina Quinasa/genética , Colina Quinasa/metabolismo , Colinérgicos/química , Colinérgicos/metabolismo , Inhibidores Enzimáticos/metabolismo , Hemicolinio 3/metabolismo , Humanos , Interacciones Hidrofóbicas e Hidrofílicas , Isoenzimas , Espectrometría de Masas , Mutación Missense , Fosforilación
18.
PLoS One ; 4(10): e7165, 2009 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-19841673

RESUMEN

BACKGROUND: UDP-GlcNAc 2-epimerase/ManNAc 6-kinase, GNE, is a bi-functional enzyme that plays a key role in sialic acid biosynthesis. Mutations of the GNE protein cause sialurea or autosomal recessive inclusion body myopathy/Nonaka myopathy. GNE is the only human protein that contains a kinase domain belonging to the ROK (repressor, ORF, kinase) family. PRINCIPAL FINDINGS: We solved the structure of the GNE kinase domain in the ligand-free state. The protein exists predominantly as a dimer in solution, with small populations of monomer and higher-order oligomer in equilibrium with the dimer. Crystal packing analysis reveals the existence of a crystallographic hexamer, and that the kinase domain dimerizes through the C-lobe subdomain. Mapping of disease-related missense mutations onto the kinase domain structure revealed that the mutation sites could be classified into four different groups based on the location - dimer interface, interlobar helices, protein surface, or within other secondary structural elements. CONCLUSIONS: The crystal structure of the kinase domain of GNE provides a structural basis for understanding disease-causing mutations and a model of hexameric wild type full length enzyme. ENHANCED VERSION: This article can also be viewed as an enhanced version in which the text of the article is integrated with interactive 3D representations and animated transitions. Please note that a web plugin is required to access this enhanced functionality. Instructions for the installation and use of the web plugin are available in Text S1.


Asunto(s)
Carbohidrato Epimerasas/química , Carbohidratos/química , Cristalografía por Rayos X/métodos , Dimerización , Humanos , Ligandos , Conformación Molecular , Enfermedades Musculares/metabolismo , Mutación , Mutación Missense , Sistemas de Lectura Abierta , Conformación Proteica , Pliegue de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
19.
PLoS One ; 4(10): e7274, 2009 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-19841675

RESUMEN

BACKGROUND: The Polycomb group (PcG) of proteins is a family of important developmental regulators. The respective members function as large protein complexes involved in establishment and maintenance of transcriptional repression of developmental control genes. MBTD1, Malignant Brain Tumor domain-containing protein 1, is one such PcG protein. MBTD1 contains four MBT repeats. METHODOLOGY/PRINCIPAL FINDINGS: We have determined the crystal structure of MBTD1 (residues 130-566aa covering the 4 MBT repeats) at 2.5 A resolution by X-ray crystallography. The crystal structure of MBTD1 reveals its similarity to another four-MBT-repeat protein L3MBTL2, which binds lower methylated lysine histones. Fluorescence polarization experiments confirmed that MBTD1 preferentially binds mono- and di-methyllysine histone peptides, like L3MBTL1 and L3MBTL2. All known MBT-peptide complex structures characterized to date do not exhibit strong histone peptide sequence selectivity, and use a "cavity insertion recognition mode" to recognize the methylated lysine with the deeply buried methyl-lysine forming extensive interactions with the protein while the peptide residues flanking methyl-lysine forming very few contacts [1]. Nevertheless, our mutagenesis data based on L3MBTL1 suggested that the histone peptides could not bind to MBT repeats in any orientation. CONCLUSIONS: The four MBT repeats in MBTD1 exhibits an asymmetric rhomboid architecture. Like other MBT repeat proteins characterized so far, MBTD1 binds mono- or dimethylated lysine histones through one of its four MBT repeats utilizing a semi-aromatic cage. ENHANCED VERSION: This article can also be viewed as an enhanced version in which the text of the article is integrated with interactive 3D representations and animated transitions. Please note that a web plugin is required to access this enhanced functionality. Instructions for the installation and use of the web plugin are available in Text S1.


Asunto(s)
Proteínas Cromosómicas no Histona/química , Proteínas Represoras/química , Secuencia de Aminoácidos , Arginina/química , Cromatina/química , Histonas/química , Humanos , Lisina/química , Metilación , Conformación Molecular , Datos de Secuencia Molecular , Nucleosomas/metabolismo , Proteínas del Grupo Polycomb , Conformación Proteica , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
20.
FEBS J ; 275(9): 2283-95, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18397323

RESUMEN

Cyclophilins comprise one of the three classes of peptidylprolyl isomerases found in all eukaryotic and prokaryotic organisms, as well as viruses. Many of the 17 annotated human cyclophilins contain the catalytic domain in tandem with other domains, and many of the specific functions of a particular cyclophilin or its associated domains remain unknown. The structure of the isomerase domain from a spliceosome-associated cyclophilin, PPWD1 (peptidylprolyl isomerase containing WD40 repeat), has been solved to 1.65 A. In the crystal, the N-terminus of one isomerase domain is bound in the active site of a neighboring isomerase molecule in a manner analogous to substrate. NMR solution studies show that this sequence binds to the active site of the cyclophilin, but cannot be turned over by the enzyme. A pseudo-substrate immediately N-terminal to the cyclophilin domain in PPWD1 could have wider implications for the function of this cyclophilin in the spliceosome, where it is located in human cells.


Asunto(s)
Isomerasa de Peptidilprolil/química , Secuencia de Aminoácidos , Sitios de Unión , Catálisis , Dominio Catalítico , Cristalografía por Rayos X , Ciclofilinas/química , Ciclofilinas/clasificación , Ciclofilinas/metabolismo , Humanos , Enlace de Hidrógeno , Modelos Químicos , Modelos Moleculares , Datos de Secuencia Molecular , Resonancia Magnética Nuclear Biomolecular , Isomerasa de Peptidilprolil/genética , Unión Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido , Espectrometría Raman , Empalmosomas/química , Empalmosomas/metabolismo , Agua/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...