Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nucleic Acids Res ; 50(5): 2549-2565, 2022 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-35188579

RESUMEN

The Isw1b chromatin-remodeling complex is specifically recruited to gene bodies to help retain pre-existing histones during transcription by RNA polymerase II. Recruitment is dependent on H3K36 methylation and the Isw1b subunit Ioc4, which contains an N-terminal PWWP domain. Here, we present the crystal structure of the Ioc4-PWWP domain, including a detailed functional characterization of the domain on its own as well as in the context of full-length Ioc4 and the Isw1b remodeler. The Ioc4-PWWP domain preferentially binds H3K36me3-containing nucleosomes. Its ability to bind DNA is required for nucleosome binding. It is also furthered by the unique insertion motif present in Ioc4-PWWP. The ability to bind H3K36me3 and DNA promotes the interaction of full-length Ioc4 with nucleosomes in vitro and they are necessary for its recruitment to gene bodies in vivo. Furthermore, a fully functional Ioc4-PWWP domain promotes efficient remodeling by Isw1b and the maintenance of ordered chromatin in vivo, thereby preventing the production of non-coding RNAs.


Asunto(s)
Ensamble y Desensamble de Cromatina , Código de Histonas , Cromatina , ADN/química , Metilación , Nucleosomas/genética , Unión Proteica
2.
Proc Natl Acad Sci U S A ; 118(32)2021 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-34362845

RESUMEN

Excessive activation of T cells and microglia represents a hallmark of the pathogenesis of human multiple sclerosis (MS). However, the regulatory molecules overactivating these immune cells remain to be identified. Previously, we reported that extracellular IFP35 family proteins, including IFP35 and NMI, activated macrophages as proinflammatory molecules in the periphery. Here, we investigated their functions in the process of neuroinflammation both in the central nervous system (CNS) and the periphery. Our analysis of clinical transcriptomic data showed that expression of IFP35 family proteins was up-regulated in patients with MS. Additional in vitro studies demonstrated that IFP35 and NMI were released by multiple cells. IFP35 and NMI subsequently triggered nuclear factor kappa B-dependent activation of microglia via the TLR4 pathway. Importantly, we showed that both IFP35 and NMI activated dendritic cells and promoted naïve T cell differentiation into Th1 and Th17 cells. Nmi-/- , Ifp35-/- , or administration of neutralizing antibodies against IFP35 alleviated the immune cells' infiltration and demyelination in the CNS, thus reducing the severity of experimental autoimmune encephalomyelitis. Together, our findings reveal a hitherto unknown mechanism by which IFP35 family proteins facilitate overactivation of both T cells and microglia and propose avenues to study the pathogenesis of MS.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/sangre , Péptidos y Proteínas de Señalización Intracelular/genética , Esclerosis Múltiple/patología , Enfermedades Neuroinflamatorias/patología , Animales , Anticuerpos Neutralizantes/farmacología , Estudios de Casos y Controles , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/inmunología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lisofosfatidilcolinas/toxicidad , Ratones Endogámicos C57BL , Ratones Mutantes , Microglía/metabolismo , Microglía/patología , Esclerosis Múltiple/genética , Enfermedades Neuroinflamatorias/genética , Células Th17/inmunología , Células Th17/metabolismo
3.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33431678

RESUMEN

Nuclear factor κB (NF-κB)-mediated signaling pathway plays a crucial role in the regulation of inflammatory process, innate and adaptive immune responses. The hyperactivation of inflammatory response causes host cell death, tissue damage, and autoinflammatory disorders, such as sepsis and inflammatory bowel disease. However, how these processes are precisely controlled is still poorly understood. In this study, we demonstrated that ankyrin repeat and suppressor of cytokine signaling box containing 1 (ASB1) is involved in the positive regulation of inflammatory responses by enhancing the stability of TAB2 and its downstream signaling pathways, including NF-κB and mitogen-activated protein kinase pathways. Mechanistically, unlike other members of the ASB family that induce ubiquitination-mediated degradation of their target proteins, ASB1 associates with TAB2 to inhibit K48-linked polyubiquitination and thereby promote the stability of TAB2 upon stimulation of cytokines and lipopolysaccharide (LPS), which indicates that ASB1 plays a noncanonical role to further stabilize the target protein rather than induce its degradation. The deficiency of Asb1 protects mice from Salmonella typhimurium- or LPS-induced septic shock and increases the survival of mice. Moreover, Asb1-deficient mice exhibited less severe colitis and intestinal inflammation induced by dextran sodium sulfate. Given the crucial role of ASB proteins in inflammatory signaling pathways, our study offers insights into the immune regulation in pathogen infection and inflammatory disorders with therapeutic implications.


Asunto(s)
Colitis/inmunología , FN-kappa B/inmunología , Procesamiento Proteico-Postraduccional , Infecciones por Salmonella/inmunología , Choque Séptico/inmunología , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Colitis/inducido químicamente , Colitis/genética , Colitis/mortalidad , Sulfato de Dextran , Genes Reporteros , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Lipopolisacáridos , Luciferasas/genética , Luciferasas/inmunología , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Ratones , Ratones Noqueados , FN-kappa B/genética , Unión Proteica , Infecciones por Salmonella/genética , Infecciones por Salmonella/microbiología , Infecciones por Salmonella/mortalidad , Salmonella typhimurium/inmunología , Salmonella typhimurium/patogenicidad , Choque Séptico/inducido químicamente , Choque Séptico/genética , Choque Séptico/mortalidad , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/genética , Análisis de Supervivencia , Ubiquitinación
4.
Mol Cell ; 57(5): 925-935, 2015 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-25620561

RESUMEN

Replication and transcription of influenza virus genome mainly depend on its RNA-dependent RNA polymerase (RdRP), composed of the PA, PB1, and PB2 subunits. Although extensively studied, the underlying mechanism of the RdRP complex is still unclear. Here we report the biochemical characterization of influenza RdRP subcomplex comprising PA, PB1, and N terminus of PB2, which exist as dimer in solution and can assemble into a tetramer state, regulated by vRNA promoter. Using single-particle cryo-electron microscopy, we have reconstructed the RdRP tetramer complex at 4.3 Å, highlighting the assembly and interfaces between monomers within the tetrameric structure. The individual RdRP subcomplex contains all the characterized motifs and appears as a cage-like structure. High-throughput mutagenesis profiling revealed that residues involved in the oligomer state formation are critical for viral life cycle. Our results lay a solid base for understanding the mechanism of replication of influenza and other negative-stranded RNA viruses.


Asunto(s)
Microscopía por Crioelectrón/métodos , Orthomyxoviridae/enzimología , ARN Polimerasa Dependiente del ARN/ultraestructura , Proteínas Virales/ultraestructura , Secuencia de Aminoácidos , Animales , Línea Celular , Células HEK293 , Humanos , Imagenología Tridimensional , Modelos Moleculares , Datos de Secuencia Molecular , Mutación , Orthomyxoviridae/genética , Multimerización de Proteína , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/genética , Homología de Secuencia de Aminoácido , Proteínas Virales/química , Proteínas Virales/genética
5.
Biochem Biophys Res Commun ; 621: 25-31, 2022 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-35809344

RESUMEN

Acute liver injury caused by overdose usage of acetaminophen (APAP) is an intractable clinical problem. Necrotic hepatocytes release large amounts of intracellular components including damage-associated molecular patterns (DAMPs) which contribute to liver failure and may serve as therapeutic targets. However, the pathogenic mechanisms of DAMPs in APAP-induced liver injury (AILI) are remain largely uncovered. Here, we found that a recently identified DAMP, interferon-induced protein 35 (IFP35), is involved in the early phase of AILI. Our data demonstrated that although the expression level of IFP35 is not significantly increased in either patients or mice with AILI, it is released from necrotic hepatocytes. Within 24 h post APAP injection, mice lacking Ifp35 are resistant to APAP-induced toxicity, and induce less inflammatory response than that of wild-type mice, including reduced AST/ALT level, pro-inflammatory cytokines production and neutrophils infiltration. More importantly, antibody of IFP35 reduces the expression level of inflammatory factors and chemokines. This study brings new knowledge into the pathogenic mechanism of AILI.


Asunto(s)
Acetaminofén , Enfermedad Hepática Inducida por Sustancias y Drogas , Péptidos y Proteínas de Señalización Intracelular , Acetaminofén/toxicidad , Animales , Enfermedad Hepática Inducida por Sustancias y Drogas/patología , Interferones/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Hígado/metabolismo , Ratones , Ratones Endogámicos C57BL , Necrosis/patología
6.
J Med Virol ; 94(12): 5943-5953, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36000451

RESUMEN

Dengue virus (DENV) is a critical public health concern in tropical and subtropical regions worldwide. Thus, immunocompetent murine models of DENV infection with robust viremia are required for vaccine studies. Diabetes is highly prevalent worldwide, making it frequent comorbidity in patients with dengue fever. Therefore, murine models are needed to understand viral pathogenesis and disease progression. Acquired-induced and inherently diabetic C57BL/6 and db/db mice were inoculated with DENV-3 via the tail vein. After infection, both the diabetic C57BL/6 and db/db mice showed obvious weight loss with clinical manifestations. Quantitative reverse-transcription polymerase chain reaction revealed robust and replicable viremia in the two types of diabetic mice. Immunohistochemical detection showed persistent DENV-3 infection in the liver. Enzyme-linked immunosorbent assay for cytokine detection revealed that diabetic mice showed more severe inflammatory responses than did nondiabetic mice, and significant histological alterations were observed in diabetic mice. Thus, the diabetic mice were more susceptible to DENV infection than the nondiabetic mice. Taken together, we established two types of immunocompetent diabetic mice for DENV infection, which can be used to further study the mechanisms of dengue pathogenesis in diabetes and to develop antiviral pharmaceuticals and treatments.


Asunto(s)
Virus del Dengue , Dengue , Diabetes Mellitus Experimental , Animales , Antivirales/uso terapéutico , Citocinas , Diabetes Mellitus Experimental/complicaciones , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Viremia
7.
Plant Physiol ; 184(2): 762-776, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32719110

RESUMEN

Lys deacylases are essential regulators of cell biology in many contexts. Here, we have identified CddA (cyanobacterial deacetylase/depropionylase), a Lys deacylase enzyme expressed in the cyanobacterium Synechococcus sp. PCC 7002 that has both deacetylase and depropionylase activity. Loss of the gene cddA led to slower growth and impaired linear and cyclic photosynthetic electron transfer. We determined the crystal structure of this depropionylase/deacetylase at 2.1 Å resolution and established that it has a unique and characteristically folded α/ß structure. We detected an acyl binding site within CddA via site-directed mutagenesis and demonstrated that this site is essential for the deproprionylase activity of this enzyme. Through a proteomic approach, we identified a total of 598 Lys residues across 382 proteins that were capable of undergoing propionylation. These propionylated proteins were highly enriched for photosynthetic and metabolic functionality. We additionally demonstrated that CddA was capable of catalyzing in vivo and in vitro Lys depropionylation and deacetylation of Fru-1,6-bisphosphatase, thereby regulating its enzymatic activity. Our identification of a Lys deacylase provides insight into the mechanisms globally regulating photosynthesis and carbon metabolism in cyanobacteria and potentially in other photosynthetic organisms as well.


Asunto(s)
Lisina/metabolismo , Synechococcus/enzimología , Fotosíntesis , Synechococcus/genética , Synechococcus/crecimiento & desarrollo
8.
Biochem Biophys Res Commun ; 526(4): 993-998, 2020 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-32295713

RESUMEN

Minichromosome maintenance 8 (MCM8) is a recently identified member of the minichromosome maintenance family, which possesses helicase and ATPase activity. It interacts with MCM9 and participates in homologous recombination repair. The structure of MCM8 is unclear now. Here, we report the crystal structure of the winged-helix domain of human MCM8 (MCM8-WHD) at 1.21 Å resolution. MCM8-WHD adopts a conserved winged-helix architecture. Structure analysis and biochemical study results showed the DNA binding ability and crucial residues of MCM8-WHD. Our results are helpful to understand the function of MCM8.


Asunto(s)
Proteínas de Mantenimiento de Minicromosoma/química , Secuencia de Aminoácidos , Cristalografía por Rayos X , ADN/química , Células HEK293 , Humanos , Modelos Moleculares , Unión Proteica , Dominios Proteicos , RecQ Helicasas/química
9.
Genes Dev ; 26(12): 1339-50, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22713871

RESUMEN

During apoptosis, apoptotic cells are removed by professional phagocytes or neighboring engulfing cells either directly through phagocytic receptors or indirectly through bridging molecules that cross-link dying cells to phagocytes. However, how bridging molecules recognize "eat me" signals and phagocytic receptors to mediate engulfment remains unclear. Here, we report the structural and functional studies of Caenorhabditis elegans TTR-52, a recently identified bridging molecule that cross-links surface-exposed phosphatidylserine (PtdSer) on apoptotic cells to the CED-1 receptor on phagocytes. Crystal structure studies show that TTR-52 has an open ß-barrel-like structure with some similarities to the PKCα-C2 domain. TTR-52 is proposed to bind PtdSer via an "ion-mediating" PtdSer-binding mode. Intensive functional studies show that CED-1 binds TTR-52 through its N-terminal EMI domain and that the hydrophobic region of the TTR-52 C terminus is involved in this interaction. In addition, unlike other PtdSer-binding domains, TTR-52 forms dimers, and its dimerization is important for its function in vivo. Our results reveal the first full-length structure of a bridging molecule and the mechanism underlying bridging molecule-mediated apoptotic cell recognition.


Asunto(s)
Apoptosis , Proteínas de Caenorhabditis elegans/química , Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/citología , Caenorhabditis elegans/metabolismo , Fagocitosis , Animales , Animales Modificados Genéticamente , Sitios de Unión , Proteínas Fluorescentes Verdes/metabolismo , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/metabolismo , Mutación/genética , Fosfatidilserinas/metabolismo , Prealbúmina/metabolismo , Unión Proteica , Multimerización de Proteína , Proteínas Recombinantes de Fusión/metabolismo , Relación Estructura-Actividad
10.
J Biol Chem ; 293(36): 14155-14164, 2018 09 07.
Artículo en Inglés | MEDLINE | ID: mdl-30012887

RESUMEN

Endosomal transport represents the primary mode for intracellular trafficking and signaling transduction and thus has to be tightly controlled. The molecular processes controlling the endosomal positioning utilize several large protein complexes, one of which contains the small GTPase Rab7, Rab-interacting lysosomal protein (RILP), and oxysterol-binding protein-related protein 1 (ORP1L). Rab7 is known to interact with RILP through a canonical binding site termed the effector-interacting switch region, but it is not clear how Rab7 interacts with ORP1L, limiting our understanding of the overall process. Here, we report structural and biochemical investigation of the Rab7-ORP1L interaction. We found that, contrary to prior studies, the interaction between Rab7 and the N-terminal ankyrin repeat domain (ARDN) of ORP1L is independent of Rab7's GTP- or GDP-binding state. Moreover, we show that Rab7 interacts with ORP1L ARDN via a unique region consisting of helix3 (α3) and 310-helix 2 (η2). This architecture leaves the canonical effector-interacting switch regions available for RILP binding and thus allows formation of the ORP1L-Rab7-RILP tripartite complex. Mutational disruption of the interacting interface between ORP1L and Rab7 compromised the ability of ORP1L-Rab7-RILP to regulate the late endosome positioning. Collectively, our results again manifested the versatility in the interaction between GTPase and its effector.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Endosomas/metabolismo , Complejos Multiproteicos/biosíntesis , Receptores de Esteroides/metabolismo , Proteínas de Unión al GTP rab/metabolismo , Sitios de Unión , Transporte Biológico , Células HeLa , Humanos , Complejos Multiproteicos/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión a GTP rab7
11.
J Biol Chem ; 293(4): 1192-1202, 2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29203529

RESUMEN

Protein-disulfide isomerase-like protein of the testis (PDILT), a member of the protein-disulfide isomerase family, is a chaperone essential for the folding of spermatogenesis-specific proteins in male postmeiotic germ cells. However, the structural mechanisms that regulate the chaperone function of PDILTs are unknown. Here, we report the structures of human PDILT (hPDILT) determined by X-ray crystallography to 2.4 Å resolution and small-angle X-ray scattering (SAXS). Distinct from previously reported U-like structures of related PDI family proteins, our structures revealed that hPDILT folds into a compact L-like structure in crystals and into an extended chain-like structure in solution. The hydrophobic regions and the hydrophobic pockets in hPDILT, which are important for substrate recognition, were clearly delineated in the crystal structure. Moreover, our results of the SAXS analysis and of structure-based substitutions and truncations indicated that the C-terminal tail in hPDILT is required for suppression of aggregation of denatured proteins, suggesting that the tail is crucial for the chaperone activity of PDILT. Taken together, our findings have identified the critical regions and conformational changes of PDILT that enable and control its activity. These results advance our understanding of the structural mechanisms involved in the chaperone activity of PDILT.


Asunto(s)
Proteína Disulfuro Isomerasas/química , Cristalografía por Rayos X , Humanos , Dominios Proteicos , Relación Estructura-Actividad
12.
Biochem Biophys Res Commun ; 510(4): 601-605, 2019 03 19.
Artículo en Inglés | MEDLINE | ID: mdl-30739782

RESUMEN

AlpK is an essential monooxygenase involved in the biosynthesis of kinamycin. It catalyzes the C5-hyfroxylattion of the crucial benzo[b]-fluorence intermediate in kinamycin synthesis. However, the structure and mechanism of AlpK is unclear. Here, we report the first structure of AlpK in complex with FAD. Our structure sheds light on the catalytic mechanism of AlpK.


Asunto(s)
Proteínas Bacterianas/química , Oxigenasas de Función Mixta/química , Streptomyces/enzimología , Secuencia de Aminoácidos , Proteínas Bacterianas/metabolismo , Cristalografía por Rayos X , Oxigenasas de Función Mixta/metabolismo , Modelos Moleculares , Conformación Proteica , Multimerización de Proteína , Quinonas/metabolismo , Alineación de Secuencia , Streptomyces/química , Streptomyces/metabolismo
13.
J Biol Chem ; 292(23): 9627-9636, 2017 06 09.
Artículo en Inglés | MEDLINE | ID: mdl-28381552

RESUMEN

DNA replication in eukaryotic cells is performed by a multiprotein complex called the replisome, which consists of helicases, polymerases, and adaptor molecules. Human acidic nucleoplasmic DNA-binding protein 1 (AND-1), also known as WD repeat and high mobility group (HMG)-box DNA-binding protein 1 (WDHD1), is an adaptor molecule crucial for DNA replication. Although structural information for the AND-1 yeast ortholog is available, the mechanistic details for how human AND-1 protein anchors the lagging-strand DNA polymerase α (pol α) to the DNA helicase complex (Cdc45-MCM2-7-GINS, CMG) await elucidation. Here, we report the structures of the N-terminal WD40 and SepB domains of human AND-1, as well as a biochemical analysis of the C-terminal HMG domain. We show that AND-1 exists as a homotrimer mediated by the SepB domain. Mutant study results suggested that a positively charged groove within the SepB domain provides binding sites for pol α. Different from its ortholog protein in budding yeast, human AND-1 is recruited to the CMG complex, mediated by unknown participants other than Go Ichi Ni San. In addition, we show that AND-1 binds to DNA in vitro, using its C-terminal HMG domain. In conclusion, our findings provide important insights into the mechanistic details of human AND-1 function, advancing our understanding of replisome formation during eukaryotic replication.


Asunto(s)
ADN Helicasas/química , ADN Polimerasa I/química , Proteínas de Unión al ADN/química , ADN/química , Complejos Multienzimáticos/química , Multimerización de Proteína , ADN/biosíntesis , ADN/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , ADN Polimerasa I/genética , ADN Polimerasa I/metabolismo , Replicación del ADN/fisiología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Humanos , Complejos Multienzimáticos/genética , Complejos Multienzimáticos/metabolismo , Dominios Proteicos
14.
Nucleic Acids Res ; 44(11): 5083-94, 2016 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-26908650

RESUMEN

Chromatin assembly factor 1 (CAF-1) is a histone H3-H4 chaperone that deposits newly synthesized histone (H3-H4)2 tetramers during replication-coupled nucleosome assembly. However, how CAF-1 functions in this process is not yet well understood. Here, we report the crystal structure of C terminus of Cac1 (Cac1C), a subunit of yeast CAF-1, and the function of this domain in stabilizing CAF-1 at replication forks. We show that Cac1C forms a winged helix domain (WHD) and binds DNA in a sequence-independent manner. Mutations in Cac1C that abolish DNA binding result in defects in transcriptional silencing and increased sensitivity to DNA damaging agents, and these defects are exacerbated when combined with Cac1 mutations deficient in PCNA binding. Similar phenotypes are observed for corresponding mutations in mouse CAF-1. These results reveal a mechanism conserved in eukaryotic cells whereby the ability of CAF-1 to bind DNA is important for its association with the DNA replication forks and subsequent nucleosome assembly.


Asunto(s)
Factor 1 de Ensamblaje de la Cromatina/metabolismo , Replicación del ADN , Antígeno Nuclear de Célula en Proliferación/metabolismo , Dominios y Motivos de Interacción de Proteínas , Origen de Réplica , Animales , Factor 1 de Ensamblaje de la Cromatina/química , Factor 1 de Ensamblaje de la Cromatina/genética , Histonas/metabolismo , Ratones , Modelos Moleculares , Mutación , Conformación de Ácido Nucleico , Antígeno Nuclear de Célula en Proliferación/química , Unión Proteica , Conformación Proteica , Proteínas Recombinantes de Fusión , Relación Estructura-Actividad
15.
J Gen Virol ; 97(9): 2149-2156, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27411929

RESUMEN

Orthomyxoviruses are a family of ssRNA virus, including influenza virus, infectious salmon anaemia virus and Thogoto virus. The matrix proteins of orthomyxoviruses play crucial roles in some essential processes of the viral life cycle. However, the mechanisms of the matrix proteins involved in these processes remain incompletely understood. Currently, only the structure and function of the matrix protein from influenza virus have been studied. Here, we present the crystal structures of the N-terminal domain of matrix protein from Thogoto virus at pH 7.0 and 4.5. By analysing the structures, we identified the conformational changes of monomers and dimers in different pH conditions, mainly caused by two flexible loops, L3 and L5. These structural deviations would reflect the basis of viral capsid assembly or disassembly.


Asunto(s)
Thogotovirus/fisiología , Proteínas de la Matriz Viral/química , Ensamble de Virus , Desencapsidación Viral , Cristalografía por Rayos X , Concentración de Iones de Hidrógeno , Modelos Moleculares , Conformación Proteica
16.
Nat Methods ; 9(7): 727-9, 2012 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-22581370

RESUMEN

Monomeric (m)Eos2 is an engineered photoactivatable fluorescent protein widely used for super-resolution microscopy. We show that mEos2 forms oligomers at high concentrations and forms aggregates when labeling membrane proteins, limiting its application as a fusion partner. We solved the crystal structure of tetrameric mEos2 and rationally designed improved versions, mEos3.1 and mEos3.2, that are truly monomeric, are brighter, mature faster and exhibit higher photon budget and label density.


Asunto(s)
Proteínas Fluorescentes Verdes , Proteínas Luminiscentes , Microscopía Fluorescente/métodos , Ingeniería de Proteínas/métodos , Animales , Células COS , Chlorocebus aethiops , Cromatografía en Gel , Proteínas Fluorescentes Verdes/química , Proteínas Fluorescentes Verdes/genética , Células HEK293 , Células HeLa , Humanos , Proteínas Luminiscentes/química , Proteínas Luminiscentes/genética , Modelos Moleculares , Procesos Fotoquímicos , Plásmidos , Conformación Proteica , Transfección , Proteína Fluorescente Roja
17.
Nature ; 458(7240): 909-13, 2009 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-19194458

RESUMEN

The heterotrimeric influenza virus polymerase, containing the PA, PB1 and PB2 proteins, catalyses viral RNA replication and transcription in the nucleus of infected cells. PB1 holds the polymerase active site and reportedly harbours endonuclease activity, whereas PB2 is responsible for cap binding. The PA amino terminus is understood to be the major functional part of the PA protein and has been implicated in several roles, including endonuclease and protease activities as well as viral RNA/complementary RNA promoter binding. Here we report the 2.2 ångström (A) crystal structure of the N-terminal 197 residues of PA, termed PA(N), from an avian influenza H5N1 virus. The PA(N) structure has an alpha/beta architecture and reveals a bound magnesium ion coordinated by a motif similar to the (P)DX(N)(D/E)XK motif characteristic of many endonucleases. Structural comparisons and mutagenesis analysis of the motif identified in PA(N) provide further evidence that PA(N) holds an endonuclease active site. Furthermore, functional analysis with in vivo ribonucleoprotein reconstitution and direct in vitro endonuclease assays strongly suggest that PA(N) holds the endonuclease active site and has critical roles in endonuclease activity of the influenza virus polymerase, rather than PB1. The high conservation of this endonuclease active site among influenza strains indicates that PA(N) is an important target for the design of new anti-influenza therapeutics.


Asunto(s)
Endonucleasas/química , Endonucleasas/metabolismo , Subtipo H5N1 del Virus de la Influenza A/enzimología , Gripe Aviar/virología , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/metabolismo , Proteínas Virales/química , Proteínas Virales/metabolismo , Animales , Aves/virología , Dominio Catalítico , Cristalografía por Rayos X , Endonucleasas/genética , Modelos Moleculares , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Polimerasa Dependiente del ARN/genética , Proteínas Virales/genética
18.
Nature ; 454(7208): 1123-6, 2008 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-18615018

RESUMEN

The recent emergence of highly pathogenic avian influenza A virus strains with subtype H5N1 pose a global threat to human health. Elucidation of the underlying mechanisms of viral replication is critical for development of anti-influenza virus drugs. The influenza RNA-dependent RNA polymerase (RdRp) heterotrimer has crucial roles in viral RNA replication and transcription. It contains three proteins: PA, PB1 and PB2. PB1 harbours polymerase and endonuclease activities and PB2 is responsible for cap binding; PA is implicated in RNA replication and proteolytic activity, although its function is less clearly defined. Here we report the 2.9 ångström structure of avian H5N1 influenza A virus PA (PA(C), residues 257-716) in complex with the PA-binding region of PB1 (PB1(N), residues 1-25). PA(C) has a fold resembling a dragon's head with PB1(N) clamped into its open 'jaws'. PB1(N) is a known inhibitor that blocks assembly of the polymerase heterotrimer and abolishes viral replication. Our structure provides details for the binding of PB1(N) to PA(C) at the atomic level, demonstrating a potential target for novel anti-influenza therapeutics. We also discuss a potential nucleotide binding site and the roles of some known residues involved in polymerase activity. Furthermore, to explore the role of PA in viral replication and transcription, we propose a model for the influenza RdRp heterotrimer by comparing PA(C) with the lambda3 reovirus polymerase structure, and docking the PA(C) structure into an available low resolution electron microscopy map.


Asunto(s)
Aves/virología , Subtipo H5N1 del Virus de la Influenza A/enzimología , ARN Polimerasa Dependiente del ARN/química , Proteínas Virales/química , Proteínas Virales/metabolismo , Animales , Sitios de Unión , Cristalografía por Rayos X , Modelos Moleculares , Complejos Multienzimáticos/química , Complejos Multienzimáticos/metabolismo , Nucleótidos/metabolismo , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Unión Proteica , Estructura Cuaternaria de Proteína , ARN Polimerasa Dependiente del ARN/metabolismo , Replicación Viral
19.
Proc Natl Acad Sci U S A ; 108(18): 7373-8, 2011 May 03.
Artículo en Inglés | MEDLINE | ID: mdl-21502537

RESUMEN

The Origin Recognition Complex (ORC) is a six-subunit protein important for the initiation of DNA replication in eukaryotic cells. Orc6 is the smallest and the least conserved among ORC subunits. It is required for the DNA replication but also has a function in cytokinesis in metazoan species, however, the mechanisms of Orc6 action in these processes are not clear. Here we report a structure of the middle domain of human Orc6. This domain has an overall fold similar to the corresponding helical domain of transcription factor TFIIB. Based on these findings, a model of Orc6 binding to DNA is produced. We have identified amino acids of Orc6 which are directly involved in DNA binding. Alterations of these amino acids abolish DNA binding ability of Orc6 and also result in reduced levels of DNA replication in vitro and in cultured cells. Our data indicate that Orc6 is one of the DNA binding subunits of ORC in metazoan species. We propose that Orc6 may participate in positioning of ORC at the origins of DNA replication similar to the role of TFIIB in positioning transcription preinitiation complex at the promoter.


Asunto(s)
Replicación del ADN/genética , Modelos Moleculares , Complejo de Reconocimiento del Origen/genética , Unión Proteica , Conformación Proteica , Factor de Transcripción TFIIB/genética , Secuencia de Aminoácidos , Animales , Bromodesoxiuridina , Cromatografía en Gel , Clonación Molecular , Cristalización , Drosophila , Electroforesis en Gel de Poliacrilamida , Proteínas Fluorescentes Verdes , Humanos , Datos de Secuencia Molecular , Complejo de Reconocimiento del Origen/química , Alineación de Secuencia , Homología de Secuencia , Xenopus
20.
Proc Natl Acad Sci U S A ; 108(51): 20538-43, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22143770

RESUMEN

Symmetric and asymmetric dimethylation of arginine are isomeric protein posttranslational modifications with distinct biological effects, evidenced by the methylation of arginine 3 of histone H4 (H4R3): symmetric dimethylation of H4R3 leads to repression of gene expression, while asymmetric dimethylation of H4R3 is associated with gene activation. The enzymes catalyzing these modifications share identifiable sequence similarities, but the relationship between their catalytic mechanisms is unknown. Here we analyzed the structure of a prototypic symmetric arginine dimethylase, PRMT5, and discovered that a conserved phenylalanine in the active site is critical for specifying symmetric addition of methyl groups. Changing it to a methionine significantly elevates the overall methylase activity, but also converts PRMT5 to an enzyme that catalyzes both symmetric and asymmetric dimethylation of arginine. Our results demonstrate a common catalytic mechanism intrinsic to both symmetric and asymmetric arginine dimethylases, and show that steric constrains in the active sites play an essential role in determining the product specificity of arginine methylases. This discovery also implies a potentially regulatable outcome of arginine dimethylation that may provide versatile control of eukaryotic gene expression.


Asunto(s)
Proteínas de Caenorhabditis elegans/química , Regulación Enzimológica de la Expresión Génica , Proteína Metiltransferasas/química , Proteína-Arginina N-Metiltransferasas/química , Secuencia de Aminoácidos , Animales , Arginina/química , Caenorhabditis elegans , Catálisis , Cristalografía por Rayos X/métodos , Escherichia coli/metabolismo , Células HEK293 , Humanos , Cinética , Metilación , Conformación Molecular , Datos de Secuencia Molecular , Empalme del ARN , Ratas , Homología de Secuencia de Aminoácido , Transcripción Genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA