Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
J Biol Chem ; 286(6): 4511-6, 2011 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-21131357

RESUMEN

Perturbed cell adhesion mechanisms are crucial for tumor invasion and metastasis. A cell adhesion protein, TSLC1 (tumor suppressor in lung cancer 1), is inactivated in a majority of metastatic cancers. DAL-1 (differentially expressed in adenocarcinoma of the lung protein), another tumor suppressor, binds through its FERM domain to the TSLC1 C-terminal, 4.1 glycophorin C-like, cytoplasmic domain. However, the molecular basis for this interaction is unknown. Here, we describe the crystal structure of a complex between the DAL-1 FERM domain and a portion of the TSLC1 cytoplasmic domain. DAL-1 binds to TSLC1 through conserved residues in a well defined hydrophobic pocket in the structural C-lobe of the DAL-1 FERM domain. From the crystal structure, it is apparent that Tyr(406) and Thr(408) in the TSLC1 cytoplasmic domain form the most important interactions with DAL-1, and this was also confirmed by surface plasmon resonance studies. Our results refute earlier exon deletion experiments that indicated that glycophorin C interacts with the α-lobe of 4.1 FERM domains.


Asunto(s)
Moléculas de Adhesión Celular/química , Inmunoglobulinas/química , Proteínas de la Membrana/química , Proteínas Supresoras de Tumor/química , Molécula 1 de Adhesión Celular , Moléculas de Adhesión Celular/genética , Moléculas de Adhesión Celular/metabolismo , Cristalografía por Rayos X , Humanos , Inmunoglobulinas/genética , Inmunoglobulinas/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de Microfilamentos , Unión Proteica , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Resonancia por Plasmón de Superficie , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
2.
Biochemistry ; 49(6): 1056-8, 2010 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-20092359

RESUMEN

Poly-ADP-ribose polymerases (PARPs) catalyze transfer of ADP-ribose from NAD(+) to specific residues in their substrate proteins or to growing ADP-ribose chains. PARP activity is involved in processes such as chromatin remodeling, transcription control, and DNA repair. Inhibitors of PARP activity may be useful in cancer therapy. PARP2 is the family member that is most similar to PARP1, and the two can act together as heterodimers. We used X-ray crystallography to determine two structures of the catalytic domain of human PARP2: the complexes with PARP inhibitors 3-aminobenzamide and ABT-888. These results contribute to our understanding of structural features and compound properties that can be employed to develop selective inhibitors of human ADP-ribosyltransferases.


Asunto(s)
Bencimidazoles/química , Dominio Catalítico , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/química , Animales , Benzamidas/química , Dominio Catalítico/efectos de los fármacos , Proteínas de Ciclo Celular/química , Cristalización , Cristalografía por Rayos X , Ácido Glutámico/química , Humanos , Enlace de Hidrógeno/efectos de los fármacos , Ratones , Poli(ADP-Ribosa) Polimerasa-1 , Estructura Secundaria de Proteína/efectos de los fármacos
3.
Protein Expr Purif ; 58(2): 210-21, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18171622

RESUMEN

Bacterial over-expression of proteins is a powerful tool to obtain soluble protein amenable to biochemical, biophysical and/or structural characterization. However, it is well established that many recombinant proteins cannot be produced in a soluble form. Several theoretical and empirical methods to improve soluble production have been suggested, although there is to date no universally accepted protocol. This report describes, and quantitatively analyses, a systematic multi-construct approach to obtain soluble protein. Although commonly used in several laboratories, quantitative analyses of the merits of the strategy applied to a larger number of target proteins are missing from the literature. In this study, typically 10 different protein constructs were tested for each targeted domain of nearly 400 human proteins. Overall, soluble expression was obtained for nearly 50% of the human target proteins upon over-expression in Escherichia coli. The chance of obtaining soluble expression was almost doubled using the multi-construct method as compared to more traditional approaches. Soluble protein constructs were subsequently subjected to crystallization trials and the multi-construct approach yielded a more than fourfold increase, from 15 proteins to 65, for the likelihood of obtaining well-diffracting crystals. The results also demonstrate the value of testing multiple constructs in crystallization trials. Finally, a retrospective analysis of gel filtration profiles indicates that these could be used with caution to prioritize protein targets for crystallization trials.


Asunto(s)
Clonación Molecular/métodos , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/genética , Secuencia de Aminoácidos , Cristalización , Electroforesis en Gel de Poliacrilamida , Escherichia coli/metabolismo , Expresión Génica , Humanos , Proteínas Recombinantes/química , Espectrometría de Masa por Ionización de Electrospray
4.
Artículo en Inglés | MEDLINE | ID: mdl-16820675

RESUMEN

Cytidine triphosphate synthetase (CTPS) is a key enzyme in nucleic acid and phospholipid biosynthesis and its activity is increased in certain human cancers, making it a promising drug target. The crystal structure of the synthetase domain of human CTPS, which represents the first structure of a CTPS from an eukaryote, has been determined. The structure is homotetrameric and each active site is formed by three different subunits. Sulfate ions bound to the active sites indicate the positions of phosphate-binding sites for the substrates ATP and UTP and the feedback inhibitor CTP. Together with earlier structures of bacterial CTPS, the human CTPS structure provides an extended understanding of the structure-function relationship of CTPS-family members. The structure also serves as a basis for structure-based design of anti-proliferative inhibitors.


Asunto(s)
Antineoplásicos , Ligasas de Carbono-Nitrógeno/química , Secuencia de Aminoácidos , Sitios de Unión , Ligasas de Carbono-Nitrógeno/antagonistas & inhibidores , Ligasas de Carbono-Nitrógeno/genética , Ligasas de Carbono-Nitrógeno/aislamiento & purificación , Clonación Molecular , Cristalografía por Rayos X , Escherichia coli/enzimología , Retroalimentación , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas Recombinantes/química
5.
Protein Sci ; 11(2): 313-21, 2002 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-11790841

RESUMEN

A prerequisite for structural genomics and related projects is to standardize the process of gene overexpression and protein solubility screening to enable automation for higher throughput. We have tested a methodology to rapidly subclone a large number of human genes and screen these for expression and protein solubility in Escherichia coli. The methodology, which can be partly automated, was used to compare the effect of six different N-terminal fusion proteins and an N-terminal 6*His tag. As a realistic test set we selected 32 potentially interesting human proteins with unknown structures and sizes suitable for NMR studies. The genes were transferred from cDNA to expression vectors using subcloning by recombination. The subcloning yield was 100% for 27 (of 32) genes for which a PCR fragment of correct size could be obtained. Of these, 26 genes (96%) could be overexpressed at detectable levels and 23 (85%) are detected in the soluble fraction with at least one fusion tag. We find large differences in the effects of fusion protein or tag on expression and solubility. In short, four of seven fusions perform very well, and much better than the 6*His tag, but individual differences motivate the inclusion of several fusions in expression and solubility screening. We also conclude that our methodology and expression vectors can be used for screening of genes for structural studies, and that it should be possible to obtain a large fraction of all NMR-sized and nonmembrane human proteins as soluble fusion proteins in E. coli.


Asunto(s)
Escherichia coli/metabolismo , Biosíntesis de Proteínas , Proteínas Recombinantes de Fusión/biosíntesis , Expresión Génica , Vectores Genéticos , Histidina/química , Humanos , Desnaturalización Proteica , Proteínas/metabolismo , Proteínas Recombinantes de Fusión/aislamiento & purificación , Solubilidad
7.
Structure ; 22(5): 744-55, 2014 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-24704254

RESUMEN

SHIP2, OCRL, and INPP5B belong to inositol polyphosphate 5-phophatase subfamilies involved in insulin regulation and Lowes syndrome. The structural basis for membrane recognition, substrate specificity, and regulation of inositol polyphosphate 5-phophatases is still poorly understood. We determined the crystal structures of human SHIP2, OCRL, and INPP5B, the latter in complex with phosphoinositide substrate analogs, which revealed a membrane interaction patch likely to assist in sequestering substrates from the lipid bilayer. Residues recognizing the 1-phosphate of the substrates are highly conserved among human family members, suggesting similar substrate binding modes. However, 3- and 4-phosphate recognition varies and determines individual substrate specificity profiles. The high conservation of the environment of the scissile 5-phosphate suggests a common reaction geometry for all members of the human 5-phosphatase family.


Asunto(s)
Membrana Celular/metabolismo , Monoéster Fosfórico Hidrolasas/química , Monoéster Fosfórico Hidrolasas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Humanos , Fosfatos de Inositol/química , Fosfatos de Inositol/metabolismo , Modelos Moleculares , Fosfatidilinositol-3,4,5-Trifosfato 5-Fosfatasas , Fosfatidilinositoles/química , Fosfatidilinositoles/metabolismo , Especificidad por Sustrato
9.
J Mol Biol ; 425(22): 4323-33, 2013 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-23816837

RESUMEN

Guanine monophosphate (GMP) synthetase is a bifunctional two-domain enzyme. The N-terminal glutaminase domain generates ammonia from glutamine and the C-terminal synthetase domain aminates xanthine monophosphate (XMP) to form GMP. Mammalian GMP synthetases (GMPSs) contain a 130-residue-long insert in the synthetase domain in comparison to bacterial proteins. We report here the structure of a eukaryotic GMPS. Substrate XMP was bound in the crystal structure of the human GMPS enzyme. XMP is bound to the synthetase domain and covered by a LID motif. The enzyme forms a dimer in the crystal structure with subunit orientations entirely different from the bacterial counterparts. The inserted sub-domain is shown to be involved in substrate binding and dimerization. Furthermore, the structural basis for XMP recognition is revealed as well as a potential allosteric site. Enzymes in the nucleotide metabolism typically display an increased activity in proliferating cells due to the increased need for nucleotides. Many drugs used as immunosuppressants and for treatment of cancer and viral diseases are indeed nucleobase- and nucleoside-based compounds, which are acting on or are activated by enzymes in this pathway. The information obtained from the crystal structure of human GMPS might therefore aid in understanding interactions of nucleoside-based drugs with GMPS and in structure-based design of GMPS-specific inhibitors.


Asunto(s)
Ligasas de Carbono-Nitrógeno/química , Ligasas de Carbono-Nitrógeno/metabolismo , Multimerización de Proteína , Secuencia de Aminoácidos , Escherichia coli/química , Escherichia coli/enzimología , Humanos , Cinética , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Conformación Proteica , Dominios y Motivos de Interacción de Proteínas , Alineación de Secuencia , Especificidad por Sustrato
10.
PLoS One ; 7(1): e30581, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22295093

RESUMEN

Selenium and sulfur are two closely related basic elements utilized in nature for a vast array of biochemical reactions. While toxic at higher concentrations, selenium is an essential trace element incorporated into selenoproteins as selenocysteine (Sec), the selenium analogue of cysteine (Cys). Sec lyases (SCLs) and Cys desulfurases (CDs) catalyze the removal of selenium or sulfur from Sec or Cys and generally act on both substrates. In contrast, human SCL (hSCL) is specific for Sec although the only difference between Sec and Cys is the identity of a single atom. The chemical basis of this selenium-over-sulfur discrimination is not understood. Here we describe the X-ray crystal structure of hSCL and identify Asp146 as the key residue that provides the Sec specificity. A D146K variant resulted in loss of Sec specificity and appearance of CD activity. A dynamic active site segment also provides the structural prerequisites for direct product delivery of selenide produced by Sec cleavage, thus avoiding release of reactive selenide species into the cell. We thus here define a molecular determinant for enzymatic specificity discrimination between a single selenium versus sulfur atom, elements with very similar chemical properties. Our findings thus provide molecular insights into a key level of control in human selenium and selenoprotein turnover and metabolism.


Asunto(s)
Liasas/química , Liasas/metabolismo , Selenio/metabolismo , Azufre/metabolismo , Secuencia de Aminoácidos , Sustitución de Aminoácidos , Animales , Dominio Catalítico , Biología Computacional , Secuencia Conservada , Cristalografía por Rayos X , Humanos , Liasas/genética , Ratones , Modelos Moleculares , Datos de Secuencia Molecular , Ratas , Selenio/química , Especificidad por Sustrato
11.
N Biotechnol ; 29(5): 515-25, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22027370

RESUMEN

The generation of affinity reagents to large numbers of human proteins depends on the ability to express the target proteins as high-quality antigens. The Structural Genomics Consortium (SGC) focuses on the production and structure determination of human proteins. In a 7-year period, the SGC has deposited crystal structures of >800 human protein domains, and has additionally expressed and purified a similar number of protein domains that have not yet been crystallised. The targets include a diversity of protein domains, with an attempt to provide high coverage of protein families. The family approach provides an excellent basis for characterising the selectivity of affinity reagents. We present a summary of the approaches used to generate purified human proteins or protein domains, a test case demonstrating the ability to rapidly generate new proteins, and an optimisation study on the modification of >70 proteins by biotinylation in vivo. These results provide a unique synergy between large-scale structural projects and the recent efforts to produce a wide coverage of affinity reagents to the human proteome.


Asunto(s)
Cromatografía de Afinidad/métodos , Proteoma/química , Proteoma/metabolismo , Proteómica/métodos , Animales , Biotina/metabolismo , Biotinilación , Cristalización , Medios de Cultivo , Genes , Humanos , Espectrometría de Masas , Plásmidos/metabolismo , Estructura Terciaria de Proteína , Proteoma/genética , Proteoma/aislamiento & purificación , Solubilidad
12.
Protein Sci ; 20(3): 597-609, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21308845

RESUMEN

Escherichia coli represents a robust, inexpensive expression host for the production of recombinant proteins. However, one major limitation is that certain protein classes do not express well in a biologically relevant form using standard expression approaches in the cytoplasm of E. coli. To improve the usefulness of the E. coli expression platform we have investigated combinations of promoters and selected N-terminal fusion tags for the extracellular expression of human target proteins. A comparative study was conducted on 24 target proteins fused to outer membrane protein A (OmpA), outer membrane protein F (OmpF) and osmotically inducible protein Y (OsmY). Based on the results of this initial study, we carried out an extended expression screen employing the OsmY fusion and multiple constructs of a more diverse set of human proteins. Using this high-throughput compatible system, we clearly demonstrate that secreted biomedically relevant human proteins can be efficiently retrieved and purified from the growth medium.


Asunto(s)
Sistemas de Secreción Bacterianos/fisiología , Proteínas de Escherichia coli/metabolismo , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas de la Membrana Bacteriana Externa/genética , Proteínas de la Membrana Bacteriana Externa/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Humanos , Espectrometría de Masas/métodos , Proteínas de Unión Periplasmáticas/genética , Proteínas de Unión Periplasmáticas/metabolismo , Porinas/genética , Porinas/metabolismo , Regiones Promotoras Genéticas , Proteínas Recombinantes de Fusión/genética
13.
PLoS One ; 5(9): e12907, 2010 Sep 23.
Artículo en Inglés | MEDLINE | ID: mdl-20957027

RESUMEN

UNLABELLED: The human SnoN is an oncoprotein that interacts with several transcription-regulatory proteins such as the histone-deacetylase, N-CoR containing co-repressor complex and Smad proteins. This study presents the crystal structure of the Dachshund homology domain of human SnoN. The structure reveals a groove composed of conserved residues with characteristic properties of a protein-interaction surface. A comparison of the 12 monomers in the asymmetric unit reveals the presence of two major conformations: an open conformation with a well accessible groove and a tight conformation with a less accessible groove. The variability in the backbone between the open and the tight conformations matches the differences seen in previously determined structures of individual Dachshund homology domains, suggesting a general plasticity within this fold family. The flexibility observed in the putative protein binding groove may enable SnoN to recognize multiple interaction partners. 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)
Péptidos y Proteínas de Señalización Intracelular/química , Proteínas Proto-Oncogénicas/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Alineación de Secuencia
14.
J Mol Biol ; 400(4): 768-82, 2010 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-20510246

RESUMEN

RNA helicases of the DExD/H-box superfamily are critically involved in all RNA-related processes. No crystal structures of human DExH-box domains had been determined previously, and their structures were difficult to predict owing to the low level of homology among DExH-motif-containing proteins from diverse species. Here we present the crystal structures of the conserved domain 1 of the DEIH-motif-containing helicase DHX9 and of the DEAD-box helicase DDX20. Both contain a RecA-like core, but DHX9 differs from DEAD-box proteins in the arrangement of secondary structural elements and is more similar to viral helicases such as NS3. The N-terminus of the DHX9 core contains two long alpha-helices that reside on the surface of the core without contributing to nucleotide binding. The RNA-polymerase-II-interacting minimal transactivation domain sequence forms an extended loop structure that resides in a hydrophobic groove on the surface of the DEIH domain. DHX9 lacks base-selective contacts and forms an unspecific but important stacking interaction with the base of the bound nucleotide, and our biochemical analysis confirms that the protein can hydrolyze ATP, guanosine 5'-triphosphate, cytidine 5'-triphosphate, and uridine 5'-triphosphate. Together, these findings allow the localization of functional motifs within the three-dimensional structure of a human DEIH helicase and show how these enzymes can bind nucleotide with high affinity in the absence of a Q-motif.


Asunto(s)
ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/metabolismo , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Nucleótidos/metabolismo , Secuencia de Aminoácidos , Cristalografía por Rayos X , Proteína 20 DEAD-Box/química , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína , Alineación de Secuencia
15.
J Med Chem ; 53(14): 5352-5, 2010 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-20565110

RESUMEN

We report two crystal structures of the PARP domain of human tankyrase-2 (TNKS2). Tankyrases are involved in fundamental cellular processes such as telomere homeostasis and Wnt signaling. The complex of TNKS2 with the potent inhibitor XAV939 provides insights into the molecular basis of the strong interaction and suggests routes for further development of tankyrase inhibitors.


Asunto(s)
Compuestos Heterocíclicos con 3 Anillos/química , Tanquirasas/química , Proteínas Wnt/fisiología , Cristalografía por Rayos X , Humanos , Modelos Moleculares , Estructura Terciaria de Proteína , Proteínas Recombinantes/química , Transducción de Señal , Tanquirasas/antagonistas & inhibidores
16.
PLoS One ; 5(9)2010 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-20941364

RESUMEN

DEAD-box RNA helicases play various, often critical, roles in all processes where RNAs are involved. Members of this family of proteins are linked to human disease, including cancer and viral infections. DEAD-box proteins contain two conserved domains that both contribute to RNA and ATP binding. Despite recent advances the molecular details of how these enzymes convert chemical energy into RNA remodeling is unknown. We present crystal structures of the isolated DEAD-domains of human DDX2A/eIF4A1, DDX2B/eIF4A2, DDX5, DDX10/DBP4, DDX18/myc-regulated DEAD-box protein, DDX20, DDX47, DDX52/ROK1, and DDX53/CAGE, and of the helicase domains of DDX25 and DDX41. Together with prior knowledge this enables a family-wide comparative structural analysis. We propose a general mechanism for opening of the RNA binding site. This analysis also provides insights into the diversity of DExD/H- proteins, with implications for understanding the functions of individual family members.


Asunto(s)
ARN Helicasas DEAD-box/química , Familia de Multigenes , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Humanos , Modelos Moleculares , Conformación Molecular , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , ARN/metabolismo , Alineación de Secuencia
17.
PLoS One ; 4(10): e6975, 2009 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-19841671

RESUMEN

UNLABELLED: Paraplegin is an m-AAA protease of the mitochondrial inner membrane that is linked to hereditary spastic paraplegias. The gene encodes an FtsH-homology protease domain in tandem with an AAA+ homology ATPase domain. The protein is believed to form a hexamer that uses ATPase-driven conformational changes in its AAA-domain to deliver substrate peptides to its protease domain. We present the crystal structure of the AAA-domain of human paraplegin bound to ADP at 2.2 A. This enables assignment of the roles of specific side chains within the catalytic cycle, and provides the structural basis for understanding the mechanism of disease mutations. 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)
Metaloendopeptidasas/química , ATPasas Asociadas con Actividades Celulares Diversas , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X/métodos , Escherichia coli/metabolismo , Humanos , Enlace de Hidrógeno , Datos de Secuencia Molecular , Péptidos/química , Conformación Proteica , Estructura Terciaria de Proteína , Homología de Secuencia de Aminoácido
18.
J Biol Chem ; 284(16): 10296-300, 2009 Apr 17.
Artículo en Inglés | MEDLINE | ID: mdl-19244245

RESUMEN

DEXD/H-box RNA helicases couple ATP hydrolysis to RNA remodeling by an unknown mechanism. We used x-ray crystallography and biochemical analysis of the human DEXD/H-box protein DDX19 to investigate its regulatory mechanism. The crystal structures of DDX19, in its RNA-bound prehydrolysis and free posthydrolysis state, reveal an alpha-helix that inserts between the conserved domains of the free protein to negatively regulate ATPase activity. This finding was corroborated by biochemical data that confirm an autoregulatory function of the N-terminal region of the protein. This is the first study describing crystal structures of a DEXD/H-box protein in its open and closed cleft conformations.


Asunto(s)
ARN Helicasas DEAD-box/química , Proteínas de Transporte Nucleocitoplasmático/química , Estructura Secundaria de Proteína , Secuencia de Aminoácidos , Sitios de Unión , Cristalografía por Rayos X , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas de Transporte Nucleocitoplasmático/genética , Proteínas de Transporte Nucleocitoplasmático/metabolismo , Estructura Terciaria de Proteína
19.
J Med Chem ; 52(9): 3108-11, 2009 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-19354255

RESUMEN

Poly(ADP-ribose) polymerases (PARPs) activate DNA repair mechanisms upon stress- and cytotoxin-induced DNA damage, and inhibition of PARP activity is a lead in cancer drug therapy. We present a structural and functional analysis of the PARP domain of human PARP-3 in complex with several inhibitors. Of these, KU0058948 is the strongest inhibitor of PARP-3 activity. The presented crystal structures highlight key features for potent inhibitor binding and suggest routes for creating isoenzyme-specific PARP inhibitors.


Asunto(s)
Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/química , Biocatálisis/efectos de los fármacos , Cristalografía por Rayos X , Inhibidores Enzimáticos/metabolismo , Humanos , Modelos Moleculares , Poli(ADP-Ribosa) Polimerasas/metabolismo , Conformación Proteica , Especificidad por Sustrato
20.
J Biol Chem ; 283(18): 11861-5, 2008 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-18332149

RESUMEN

The Toll/interleukin-1 receptor (TIR) domain is a highly conserved signaling domain found in the intracellular regions of Toll-like receptors (TLRs), in interleukin-1 receptors, and in several cytoplasmic adaptor proteins. TIR domains mediate receptor signal transduction through recruitment of adaptor proteins and play critical roles in the innate immune response and inflammation. This work presents the 2.2A crystal structure of the TIR domain of human TLR10, revealing a symmetric dimer in the asymmetric unit. The dimer interaction surface contains residues from the BB-loop, DD-loop, and alphaC-helix, which have previously been identified as important structural motifs for signaling in homologous TLR receptors. The interaction surface is extensive, containing a central hydrophobic patch surrounded by polar residues. The BB-loop forms a tight interaction, where a range of consecutive residues binds in a pocket formed by the reciprocal BB-loop and alphaC-helix. This pocket appears to be well suited for binding peptide substrates, which is consistent with the notion that peptides and peptide mimetics of the BB-loop are inhibitors for TLR signaling. The TLR10 structure is in good agreement with available biochemical data on TLR receptors and is likely to provide a good model for the physiological dimer.


Asunto(s)
Citoplasma/química , Transducción de Señal , Receptor Toll-Like 10/química , Secuencia Conservada , Cristalografía por Rayos X , Dimerización , Humanos , Modelos Moleculares , Mutación/genética , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA