Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mol Cell ; 82(22): 4324-4339.e8, 2022 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-36347259

RESUMEN

ATG9A and ATG2A are essential core members of the autophagy machinery. ATG9A is a lipid scramblase that allows equilibration of lipids across a membrane bilayer, whereas ATG2A facilitates lipid flow between tethered membranes. Although both have been functionally linked during the formation of autophagosomes, the molecular details and consequences of their interaction remain unclear. By combining data from peptide arrays, crosslinking, and hydrogen-deuterium exchange mass spectrometry together with cryoelectron microscopy, we propose a molecular model of the ATG9A-2A complex. Using this integrative structure modeling approach, we identify several interfaces mediating ATG9A-2A interaction that would allow a direct transfer of lipids from ATG2A into the lipid-binding perpendicular branch of ATG9A. Mutational analyses combined with functional activity assays demonstrate their importance for autophagy, thereby shedding light on this protein complex at the heart of autophagy.


Asunto(s)
Autofagosomas , Autofagia , Microscopía por Crioelectrón , Bioensayo , Lípidos
2.
PLoS Pathog ; 20(6): e1012360, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38935780

RESUMEN

The cGMP-dependent protein kinase (PKG) is the sole cGMP sensor in malaria parasites, acting as an essential signalling hub to govern key developmental processes throughout the parasite life cycle. Despite the importance of PKG in the clinically relevant asexual blood stages, many aspects of malarial PKG regulation, including the importance of phosphorylation, remain poorly understood. Here we use genetic and biochemical approaches to show that reduced cGMP binding to cyclic nucleotide binding domain B does not affect in vitro kinase activity but prevents parasite egress. Similarly, we show that phosphorylation of a key threonine residue (T695) in the activation loop is dispensable for kinase activity in vitro but is essential for in vivo PKG function, with loss of T695 phosphorylation leading to aberrant phosphorylation events across the parasite proteome and changes to the substrate specificity of PKG. Our findings indicate that Plasmodium PKG is uniquely regulated to transduce signals crucial for malaria parasite development.


Asunto(s)
Proteínas Quinasas Dependientes de GMP Cíclico , GMP Cíclico , Proteínas Quinasas Dependientes de GMP Cíclico/metabolismo , Proteínas Quinasas Dependientes de GMP Cíclico/genética , Fosforilación , GMP Cíclico/metabolismo , Malaria/parasitología , Malaria/metabolismo , Proteínas Protozoarias/metabolismo , Proteínas Protozoarias/genética , Animales , Plasmodium falciparum/metabolismo , Plasmodium falciparum/genética , Humanos , Transducción de Señal , Eritrocitos/parasitología , Eritrocitos/metabolismo
3.
EMBO J ; 37(24)2018 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-30266824

RESUMEN

Loss-of-function mutations in CDKL5 kinase cause severe neurodevelopmental delay and early-onset seizures. Identification of CDKL5 substrates is key to understanding its function. Using chemical genetics, we found that CDKL5 phosphorylates three microtubule-associated proteins: MAP1S, EB2 and ARHGEF2, and determined the phosphorylation sites. Substrate phosphorylations are greatly reduced in CDKL5 knockout mice, verifying these as physiological substrates. In CDKL5 knockout mouse neurons, dendritic microtubules have longer EB3-labelled plus-end growth duration and these altered dynamics are rescued by reduction of MAP1S levels through shRNA expression, indicating that CDKL5 regulates microtubule dynamics via phosphorylation of MAP1S. We show that phosphorylation by CDKL5 is required for MAP1S dissociation from microtubules. Additionally, anterograde cargo trafficking is compromised in CDKL5 knockout mouse dendrites. Finally, EB2 phosphorylation is reduced in patient-derived human neurons. Our results reveal a novel activity-dependent molecular pathway in dendritic microtubule regulation and suggest a pathological mechanism which may contribute to CDKL5 deficiency disorder.


Asunto(s)
Dendritas/metabolismo , Microtúbulos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Animales , Síndromes Epilépticos/genética , Síndromes Epilépticos/metabolismo , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Microtúbulos/genética , Fosforilación , Proteínas Serina-Treonina Quinasas/genética , Factores de Intercambio de Guanina Nucleótido Rho/genética , Factores de Intercambio de Guanina Nucleótido Rho/metabolismo , Espasmos Infantiles/genética , Espasmos Infantiles/metabolismo
4.
PLoS Pathog ; 16(6): e1008640, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32569299

RESUMEN

Ubiquitylation is a common post translational modification of eukaryotic proteins and in the human malaria parasite, Plasmodium falciparum (Pf) overall ubiquitylation increases in the transition from intracellular schizont to extracellular merozoite stages in the asexual blood stage cycle. Here, we identify specific ubiquitylation sites of protein substrates in three intraerythrocytic parasite stages and extracellular merozoites; a total of 1464 sites in 546 proteins were identified (data available via ProteomeXchange with identifier PXD014998). 469 ubiquitylated proteins were identified in merozoites compared with only 160 in the preceding intracellular schizont stage, suggesting a large increase in protein ubiquitylation associated with merozoite maturation. Following merozoite invasion of erythrocytes, few ubiquitylated proteins were detected in the first intracellular ring stage but as parasites matured through trophozoite to schizont stages the apparent extent of ubiquitylation increased. We identified commonly used ubiquitylation motifs and groups of ubiquitylated proteins in specific areas of cellular function, for example merozoite pellicle proteins involved in erythrocyte invasion, exported proteins, and histones. To investigate the importance of ubiquitylation we screened ubiquitin pathway inhibitors in a parasite growth assay and identified the ubiquitin activating enzyme (UBA1 or E1) inhibitor MLN7243 (TAK-243) to be particularly effective. This small molecule was shown to be a potent inhibitor of recombinant PfUBA1, and a structural homology model of MLN7243 bound to the parasite enzyme highlights avenues for the development of P. falciparum specific inhibitors. We created a genetically modified parasite with a rapamycin-inducible functional deletion of uba1; addition of either MLN7243 or rapamycin to the recombinant parasite line resulted in the same phenotype, with parasite development blocked at the schizont stage. Nuclear division and formation of intracellular structures was interrupted. These results indicate that the intracellular target of MLN7243 is UBA1, and this activity is essential for the final differentiation of schizonts to merozoites.


Asunto(s)
Merozoítos/metabolismo , Plasmodium falciparum/metabolismo , Proteínas Protozoarias/metabolismo , Ubiquitina/metabolismo , Ubiquitinación , Humanos , Plasmodium falciparum/genética , Proteínas Protozoarias/genética , Ubiquitina/genética
5.
RNA Biol ; 18(sup2): 770-781, 2021 11 12.
Artículo en Inglés | MEDLINE | ID: mdl-34719327

RESUMEN

TUT4 and the closely related TUT7 are non-templated poly(U) polymerases required at different stages of development, and their mis-regulation or mutation has been linked to important cancer pathologies. While TUT4(7) interaction with its pre-miRNA targets has been characterized in detail, the molecular bases of the broader target recognition process are unclear. Here, we examine RNA binding by the ZnF domains of the protein. We show that TUT4(7) ZnF2 contains two distinct RNA binding surfaces that are used in the interaction with different RNA nucleobases in different targets, i.e that this small domain encodes diversity in TUT4(7) selectivity and molecular function. Interestingly and unlike other well-characterized CCHC ZnFs, ZnF2 is not physically coupled to the flanking ZnF3 and acts independently in miRNA recognition, while the remaining CCHC ZnF of TUT4(7), ZnF1, has lost its intrinsic RNA binding capability. Together, our data suggest that the ZnFs of TUT4(7) are independent units for RNA and, possibly, protein-protein interactions that underlay the protein's functional flexibility and are likely to play an important role in building its interaction network.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Epistasis Genética , Regulación de la Expresión Génica , MicroARNs/genética , Proteínas de Unión al ARN/metabolismo , Dedos de Zinc , Composición de Base , Proteínas de Unión al ADN/química , Humanos , Espectroscopía de Resonancia Magnética , MicroARNs/química , MicroARNs/metabolismo , Poli U , Dominios y Motivos de Interacción de Proteínas , Proteínas de Unión al ARN/química , Relación Estructura-Actividad
6.
EMBO J ; 35(11): 1204-18, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27154206

RESUMEN

TRIM E3 ubiquitin ligases regulate a wide variety of cellular processes and are particularly important during innate immune signalling events. They are characterized by a conserved tripartite motif in their N-terminal portion which comprises a canonical RING domain, one or two B-box domains and a coiled-coil region that mediates ligase dimerization. Self-association via the coiled-coil has been suggested to be crucial for catalytic activity of TRIMs; however, the precise molecular mechanism underlying this observation remains elusive. Here, we provide a detailed characterization of the TRIM ligases TRIM25 and TRIM32 and show how their oligomeric state is linked to catalytic activity. The crystal structure of a complex between the TRIM25 RING domain and an ubiquitin-loaded E2 identifies the structural and mechanistic features that promote a closed E2~Ub conformation to activate the thioester for ubiquitin transfer allowing us to propose a model for the regulation of activity in the full-length protein. Our data reveal an unexpected diversity in the self-association mechanism of TRIMs that might be crucial for their biological function.


Asunto(s)
Factores de Transcripción/química , Factores de Transcripción/metabolismo , Proteínas de Motivos Tripartitos/química , Proteínas de Motivos Tripartitos/metabolismo , Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Cristalización , Humanos , Conformación Proteica , Multimerización de Proteína , Ubiquitinación
7.
Nature ; 505(7482): 234-8, 2014 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-24336198

RESUMEN

Lentiviruses contain accessory genes that have evolved to counteract the effects of host cellular defence proteins that inhibit productive infection. One such restriction factor, SAMHD1, inhibits human immunodeficiency virus (HIV)-1 infection of myeloid-lineage cells as well as resting CD4(+) T cells by reducing the cellular deoxynucleoside 5'-triphosphate (dNTP) concentration to a level at which the viral reverse transcriptase cannot function. In other lentiviruses, including HIV-2 and related simian immunodeficiency viruses (SIVs), SAMHD1 restriction is overcome by the action of viral accessory protein x (Vpx) or the related viral protein r (Vpr) that target and recruit SAMHD1 for proteasomal degradation. The molecular mechanism by which these viral proteins are able to usurp the host cell's ubiquitination machinery to destroy the cell's protection against these viruses has not been defined. Here we present the crystal structure of a ternary complex of Vpx with the human E3 ligase substrate adaptor DCAF1 and the carboxy-terminal region of human SAMHD1. Vpx is made up of a three-helical bundle stabilized by a zinc finger motif, and wraps tightly around the disc-shaped DCAF1 molecule to present a new molecular surface. This adapted surface is then able to recruit SAMHD1 via its C terminus, making it a competent substrate for the E3 ligase to mark for proteasomal degradation. The structure reported here provides a molecular description of how a lentiviral accessory protein is able to subvert the cell's normal protein degradation pathway to inactivate the cellular viral defence system.


Asunto(s)
Proteínas Portadoras/metabolismo , VIH/química , VIH/fisiología , Proteínas de Unión al GTP Monoméricas/metabolismo , Proteolisis , Proteínas Reguladoras y Accesorias Virales/química , Proteínas Reguladoras y Accesorias Virales/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Portadoras/química , Cercocebus atys/virología , Cristalografía por Rayos X , Interacciones Huésped-Patógeno , Humanos , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas de Unión al GTP Monoméricas/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteínas Serina-Treonina Quinasas , Proteína 1 que Contiene Dominios SAM y HD , Virus de la Inmunodeficiencia de los Simios/química , Virus de la Inmunodeficiencia de los Simios/fisiología , Ubiquitina-Proteína Ligasas , Ubiquitinación , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/química , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo
8.
Nature ; 503(7476): 422-426, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24141947

RESUMEN

Linear ubiquitin chains are important regulators of cellular signalling pathways that control innate immunity and inflammation through nuclear factor (NF)-κB activation and protection against tumour necrosis factor-α-induced apoptosis. They are synthesized by HOIP, which belongs to the RBR (RING-between-RING) family of E3 ligases and is the catalytic component of LUBAC (linear ubiquitin chain assembly complex), a multisubunit E3 ligase. RBR family members act as RING/HECT hybrids, employing RING1 to recognize ubiquitin-loaded E2 while a conserved cysteine in RING2 subsequently forms a thioester intermediate with the transferred or 'donor' ubiquitin. Here we report the crystal structure of the catalytic core of HOIP in its apo form and in complex with ubiquitin. The carboxy-terminal portion of HOIP adopts a novel fold that, together with a zinc-finger, forms a ubiquitin-binding platform that orients the acceptor ubiquitin and positions its α-amino group for nucleophilic attack on the E3∼ubiquitin thioester. The C-terminal tail of a second ubiquitin molecule is located in close proximity to the catalytic cysteine, providing a unique snapshot of the ubiquitin transfer complex containing both donor and acceptor ubiquitin. These interactions are required for activation of the NF-κB pathway in vivo, and they explain the determinants of linear ubiquitin chain specificity by LUBAC.


Asunto(s)
Ubiquitina-Proteína Ligasas/química , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/química , Ubiquitina/metabolismo , Apoproteínas/química , Apoproteínas/metabolismo , Dominio Catalítico , Cristalografía por Rayos X , Células HeLa , Humanos , Modelos Moleculares , Conformación Proteica , Especificidad por Sustrato
9.
Nature ; 480(7377): 379-82, 2011 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-22056990

RESUMEN

SAMHD1, an analogue of the murine interferon (IFN)-γ-induced gene Mg11 (ref. 1), has recently been identified as a human immunodeficiency virus-1 (HIV-1) restriction factor that blocks early-stage virus replication in dendritic and other myeloid cells and is the target of the lentiviral protein Vpx, which can relieve HIV-1 restriction. SAMHD1 is also associated with Aicardi-Goutières syndrome (AGS), an inflammatory encephalopathy characterized by chronic cerebrospinal fluid lymphocytosis and elevated levels of the antiviral cytokine IFN-α. The pathology associated with AGS resembles congenital viral infection, such as transplacentally acquired HIV. Here we show that human SAMHD1 is a potent dGTP-stimulated triphosphohydrolase that converts deoxynucleoside triphosphates to the constituent deoxynucleoside and inorganic triphosphate. The crystal structure of the catalytic core of SAMHD1 reveals that the protein is dimeric and indicates a molecular basis for dGTP stimulation of catalytic activity against dNTPs. We propose that SAMHD1, which is highly expressed in dendritic cells, restricts HIV-1 replication by hydrolysing the majority of cellular dNTPs, thus inhibiting reverse transcription and viral complementary DNA (cDNA) synthesis.


Asunto(s)
VIH-1/fisiología , Proteínas de Unión al GTP Monoméricas/química , Proteínas de Unión al GTP Monoméricas/metabolismo , Nucleósido-Trifosfatasa/química , Nucleósido-Trifosfatasa/metabolismo , Regulación Alostérica , Biocatálisis , Dominio Catalítico , Cristalografía por Rayos X , Células Dendríticas/metabolismo , Células Dendríticas/virología , Nucleótidos de Desoxiadenina/metabolismo , Nucleótidos de Desoxicitosina/metabolismo , Nucleótidos de Desoxiguanina/metabolismo , Humanos , Hidrólisis , Modelos Biológicos , Modelos Moleculares , Proteínas de Unión al GTP Monoméricas/genética , Células Mieloides/virología , Nucleósido-Trifosfatasa/genética , Estructura Terciaria de Proteína , Transcripción Reversa , Proteína 1 que Contiene Dominios SAM y HD , Nucleótidos de Timina/metabolismo , Proteínas Reguladoras y Accesorias Virales/metabolismo , Replicación Viral
10.
Proc Natl Acad Sci U S A ; 109(52): 21474-9, 2012 Dec 26.
Artículo en Inglés | MEDLINE | ID: mdl-23236176

RESUMEN

The hemagglutinin (HA) of influenza A(H3N2) virus responsible for the 1968 influenza pandemic derived from an avian virus. On introduction into humans, its receptor binding properties had changed from a preference for avian receptors (α2,3-linked sialic acid) to a preference for human receptors (α2,6-linked sialic acid). By 2001, the avidity of human H3 viruses for avian receptors had declined, and since then the affinity for human receptors has also decreased significantly. These changes in receptor binding, which correlate with increased difficulties in virus propagation in vitro and in antigenic analysis, have been assessed by virus hemagglutination of erythrocytes from different species and quantified by measuring virus binding to receptor analogs using surface biolayer interferometry. Crystal structures of HA-receptor analog complexes formed with HAs from viruses isolated in 2004 and 2005 reveal significant differences in the conformation of the 220-loop of HA1, relative to the 1968 structure, resulting in altered interactions between the HA and the receptor analog that explain the changes in receptor affinity. Site-specific mutagenesis shows the HA1 Asp-225→Asn substitution to be the key determinant of the decreased receptor binding in viruses circulating since 2005. Our results indicate that the evolution of human influenza A(H3N2) viruses since 1968 has produced a virus with a low propensity to bind human receptor analogs, and this loss of avidity correlates with the marked reduction in A(H3N2) virus disease impact in the last 10 y.


Asunto(s)
Evolución Molecular , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Subtipo H3N2 del Virus de la Influenza A/metabolismo , Receptores Virales/metabolismo , Animales , Sitios de Unión , Glicoproteínas Hemaglutininas del Virus de la Influenza/química , Humanos , Interferometría , Células de Riñón Canino Madin Darby , Modelos Moleculares , Ácido N-Acetilneuramínico/metabolismo , Unión Proteica , Multimerización de Proteína , Electricidad Estática
11.
EMBO Rep ; 13(9): 840-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22791023

RESUMEN

The linear ubiquitin chain assembly complex (LUBAC) is a RING E3 ligase that regulates immune and inflammatory signalling pathways. Unlike classical RING E3 ligases, LUBAC determines the type of ubiquitin chain being formed, an activity normally associated with the E2 enzyme. We show that the RING-in-between-RING (RBR)-containing region of HOIP--the catalytic subunit of LUBAC--is sufficient to generate linear ubiquitin chains. However, this activity is inhibited by the N-terminal portion of the molecule, an inhibition that is released upon complex formation with HOIL-1L or SHARPIN. Furthermore, we demonstrate that HOIP transfers ubiquitin to the substrate through a thioester intermediate formed by a conserved cysteine in the RING2 domain, supporting the notion that RBR ligases act as RING/HECT hybrids.


Asunto(s)
Poliubiquitina/biosíntesis , Ubiquitina-Proteína Ligasas/química , Animales , Dominio Catalítico , Bovinos , Ésteres/química , Poliubiquitina/química , Estructura Cuaternaria de Proteína , Estructura Terciaria de Proteína , Subunidades de Proteína , Compuestos de Sulfhidrilo/química
12.
Nucleic Acids Res ; 39(13): 5715-28, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21415010

RESUMEN

The J-binding protein 1 (JBP1) is essential for biosynthesis and maintenance of DNA base-J (ß-d-glucosyl-hydroxymethyluracil). Base-J and JBP1 are confined to some pathogenic protozoa and are absent from higher eukaryotes, prokaryotes and viruses. We show that JBP1 recognizes J-containing DNA (J-DNA) through a 160-residue domain, DB-JBP1, with 10 000-fold preference over normal DNA. The crystal structure of DB-JBP1 revealed a helix-turn-helix variant fold, a 'helical bouquet' with a 'ribbon' helix encompassing the amino acids responsible for DNA binding. Mutation of a single residue (Asp525) in the ribbon helix abrogates specificity toward J-DNA. The same mutation renders JBP1 unable to rescue the targeted deletion of endogenous JBP1 genes in Leishmania and changes its distribution in the nucleus. Based on mutational analysis and hydrogen/deuterium-exchange mass-spectrometry data, a model of JBP1 bound to J-DNA was constructed and validated by small-angle X-ray scattering data. Our results open new possibilities for targeted prevention of J-DNA recognition as a therapeutic intervention for parasitic diseases.


Asunto(s)
Proteínas de Unión al ADN/química , ADN/química , Glucósidos/química , Proteínas Protozoarias/química , Uracilo/análogos & derivados , Secuencia de Aminoácidos , Arginina/química , Ácido Aspártico/química , Cristalografía por Rayos X , ADN Bacteriano/metabolismo , Proteínas de Unión al ADN/metabolismo , Glucósidos/metabolismo , Lisina/química , Espectrometría de Masas , Modelos Moleculares , Datos de Secuencia Molecular , Unión Proteica , Estructura Terciaria de Proteína , Proteínas Protozoarias/metabolismo , Dispersión del Ángulo Pequeño , Alineación de Secuencia , Uracilo/química , Uracilo/metabolismo , Difracción de Rayos X
13.
J Spinal Disord Tech ; 26(7): E259-64, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23274400

RESUMEN

STUDY DESIGN: Study of the influence of thoracolumbar spinal surgery through a posterior approach to the intercompartmental pressure of the paraspinal muscles. OBJECTIVE: To create waveforms according to the pressure variations up to 24 hours postoperatively and relate these measurements to independent parameters. SUMMARY OF BACKGROUND DATA: The existence of a paraspinal anatomic compartment and a relevant compartment syndrome has been supported theoretically, proven experimentally, and confirmed in clinical cases. The perioperative variations of the intercompartmental pressures remain largely unknown. METHODS: Five measurements were taken from both paraspinal compartments in each operated patient: preoperatively, intraoperatively, immediately after wound closure, and at 6 and 24 hours postoperatively. The recorded pressures were grouped as normal, elevated, or suggestive of a paraspinal compartment syndrome. Abnormal pressures were correlated with patient-related and operation-related parameters. Forty-two patients participated in the study, 21 male and 21 female patients aged 13 to 83 years (mean age 51 y). Seventy compartments were included in the final analysis. RESULTS: Forty-two compartments developed abnormally elevated pressures postoperatively and in 22 of these, pressures suggestive of a compartment syndrome were recorded. In no case was there a clinical presentation of a true compartment syndrome. Different waveforms were created for the normal and elevated pressures group. In compartments with high measurements, pressures were likely to continue to rise at 6 and 24 hours postoperatively. The body mass index was greater in both the elevated pressures and compartment pressures groups. Procedures lasting >2 hours, extended approaches, and instrumented posterior interbody fusion operations were related with lower postoperative pressures. CONCLUSIONS: A large percentage of patients develop increased paraspinal muscle pressures up to 24 hours after posterior thoracolumbar spine surgery. These increases are related to patient-related and operation-related factors and may not present clinically as a compartment syndrome.


Asunto(s)
Síndromes Compartimentales/patología , Síndromes Compartimentales/cirugía , Músculos Paraespinales/patología , Músculos Paraespinales/cirugía , Atención Perioperativa , Presión , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Periodo Posoperatorio , Adulto Joven
14.
Cell Rep Methods ; 3(6): 100508, 2023 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-37426752

RESUMEN

Understanding how the RNA-binding domains of a protein regulator are used to recognize its RNA targets is a key problem in RNA biology, but RNA-binding domains with very low affinity do not perform well in the methods currently available to characterize protein-RNA interactions. Here, we propose to use conservative mutations that enhance the affinity of RNA-binding domains to overcome this limitation. As a proof of principle, we have designed and validated an affinity-enhanced K-homology (KH) domain mutant of the fragile X syndrome protein FMRP, a key regulator of neuronal development, and used this mutant to determine the domain's sequence preference and to explain FMRP recognition of specific RNA motifs in the cell. Our results validate our concept and our nuclear magnetic resonance (NMR)-based workflow. While effective mutant design requires an understanding of the underlying principles of RNA recognition by the relevant domain type, we expect the method will be used effectively in many RNA-binding domains.


Asunto(s)
Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil , ARN , ARN/genética , Proteína de la Discapacidad Intelectual del Síndrome del Cromosoma X Frágil/genética , Proteínas/genética , Mutación , Motivos de Unión al ARN/genética
15.
Acta Orthop Belg ; 78(3): 414-7, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22822587

RESUMEN

Chondroblastoma is a rare benign tumour. Involvement of the femoral head may often lead to a delayed diagnosis. We present the case of a 15-year-old patient with right hip pain which was first attributed to adductor tendinitis. Following aggravation of the symptoms, thorough investigation including a CT-guided biopsy, revealed the diagnosis of chondroblastoma of the femoral head. Removal of the lesion based on the techniques described in literature was not possible, mainly because the articular cartilage was breached. A novel surgical technique was used in order to address the rare location and behaviour of the tumour. This technique offered the patient pain relief and return to his previous every day and sports activities. No recurrence was seen at two years follow-up.


Asunto(s)
Condroblastoma/cirugía , Neoplasias Femorales/cirugía , Procedimientos Ortopédicos/métodos , Adolescente , Cartílago Articular/diagnóstico por imagen , Cartílago Articular/patología , Condroblastoma/diagnóstico por imagen , Condroblastoma/patología , Neoplasias Femorales/diagnóstico por imagen , Neoplasias Femorales/patología , Cabeza Femoral/diagnóstico por imagen , Humanos , Masculino , Radiografía
16.
Front Pharmacol ; 13: 860682, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35548337

RESUMEN

DNA replication initiation requires the loading of MCM2-7 complexes at the origins of replication during G1. Replication licensing renders chromatin competent for DNA replication and its tight regulation is essential to prevent aberrant DNA replication and genomic instability. CDT1 is a critical factor of licensing and its activity is controlled by redundant mechanisms, including Geminin, a protein inhibitor of CDT1. Aberrant CDT1 and Geminin expression have been shown to promote tumorigenesis in vivo and are also evident in multiple human tumors. In this study, we developed an in vitro AlphaScreen™ high-throughput screening (HTS) assay for the identification of small-molecule inhibitors targeting the CDT1/Geminin protein complex. Biochemical characterization of the most potent compound, AF615, provided evidence of specific, dose-dependent inhibition of Geminin binding to CDT1 both in-vitro and in cells. Moreover, compound AF615 induces DNA damage, inhibits DNA synthesis and reduces viability selectively in cancer cell lines, and this effect is CDT1-dependent. Taken together, our data suggest that AF615 may serve as a useful compound to elucidate the role of CDT1/Geminin protein complex in replication licensing and origin firing as well as a scaffold for further medicinal chemistry optimisation.

17.
J Struct Biol ; 175(2): 113-9, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21453775

RESUMEN

High-throughput methods to produce a large number of soluble recombinant protein variants are particularly important in the process of determining the three-dimensional structure of proteins and their complexes. Here, we describe a collection of protein expression vectors for ligation-independent cloning, which allow co-expression strategies by implementing different affinity tags and antibiotic resistances. Since the same PCR product can be inserted in all but one of the vectors, this allows efficiency in versatility while screening for optimal expression strategies. We first demonstrate the use of these vectors for protein expression in Escherichia coli, on a set of proteins belonging to the ubiquitin specific protease (USP) Family. We have selected 35 USPs, created 145 different expression constructs into the pETNKI-His-3C-LIC-kan vector, and obtained 38 soluble recombinant proteins for 21 different USPs. Finally, we exemplify the use of our vectors for bacterial co-expression and for expression in insect cells, with USP4 and USP7 respectively. We conclude that our ligation-independent cloning strategy allows for high-throughput screening for the expression of soluble proteins in a variety of vectors in E. coli and in insect cells. In addition, the same vectors can be used for co-expression studies, at least for simple binary complexes. Application in the family of ubiquitin specific proteases led to a number of soluble USPs that are used for functional and crystallization studies.


Asunto(s)
Clonación Molecular/métodos , Endopeptidasas/genética , Vectores Genéticos , Proteínas Recombinantes/genética , Animales , Automatización de Laboratorios , Baculoviridae , Secuencia de Bases , Línea Celular , Endopeptidasas/metabolismo , Escherichia coli/genética , Humanos , Datos de Secuencia Molecular , Proteínas Recombinantes/metabolismo , Proteasas Ubiquitina-Específicas
18.
Proteins ; 79 Suppl 10: 6-20, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22020785

RESUMEN

One goal of the CASP community wide experiment on the critical assessment of techniques for protein structure prediction is to identify the current state of the art in protein structure prediction and modeling. A fundamental principle of CASP is blind prediction on a set of relevant protein targets, that is, the participating computational methods are tested on a common set of experimental target proteins, for which the experimental structures are not known at the time of modeling. Therefore, the CASP experiment would not have been possible without broad support of the experimental protein structural biology community. In this article, several experimental groups discuss the structures of the proteins which they provided as prediction targets for CASP9, highlighting structural and functional peculiarities of these structures: the long tail fiber protein gp37 from bacteriophage T4, the cyclic GMP-dependent protein kinase Iß dimerization/docking domain, the ectodomain of the JTB (jumping translocation breakpoint) transmembrane receptor, Autotaxin in complex with an inhibitor, the DNA-binding J-binding protein 1 domain essential for biosynthesis and maintenance of DNA base-J (ß-D-glucosyl-hydroxymethyluracil) in Trypanosoma and Leishmania, an so far uncharacterized 73 residue domain from Ruminococcus gnavus with a fold typical for PDZ-like domains, a domain from the phycobilisome core-membrane linker phycobiliprotein ApcE from Synechocystis, the heat shock protein 90 activators PFC0360w and PFC0270w from Plasmodium falciparum, and 2-oxo-3-deoxygalactonate kinase from Klebsiella pneumoniae.


Asunto(s)
Biología Computacional/métodos , Modelos Moleculares , Proteínas/química , Secuencia de Aminoácidos , Animales , Bacteriófago T4/química , Proteínas Quinasas Dependientes de GMP Cíclico/química , Proteínas de Unión al ADN/química , Humanos , Klebsiella pneumoniae/química , Klebsiella pneumoniae/enzimología , Leishmania/química , Datos de Secuencia Molecular , Hidrolasas Diéster Fosfóricas/química , Fosfotransferasas (Aceptor de Grupo Alcohol)/química , Plasmodium falciparum/química , Conformación Proteica , Pliegue de Proteína , Proteínas Protozoarias/química , Trypanosoma/química , Proteínas Virales/química
19.
J Mol Biol ; 433(13): 166964, 2021 06 25.
Artículo en Inglés | MEDLINE | ID: mdl-33781758

RESUMEN

Recent years have seen a dramatic improvement in protein-design methodology. Nevertheless, most methods demand expert intervention, limiting their widespread adoption. By contrast, the PROSS algorithm for improving protein stability and heterologous expression levels has been successfully applied to a range of challenging enzymes and binding proteins. Here, we benchmark the application of PROSS as a stand-alone tool for protein scientists with no or limited experience in modeling. Twelve laboratories from the Protein Production and Purification Partnership in Europe (P4EU) challenged the PROSS algorithm with 14 unrelated protein targets without support from the PROSS developers. For each target, up to six designs were evaluated for expression levels and in some cases, for thermal stability and activity. In nine targets, designs exhibited increased heterologous expression levels either in prokaryotic and/or eukaryotic expression systems under experimental conditions that were tailored for each target protein. Furthermore, we observed increased thermal stability in nine of ten tested targets. In two prime examples, the human Stem Cell Factor (hSCF) and human Cadherin-Like Domain (CLD12) from the RET receptor, the wild type proteins were not expressible as soluble proteins in E. coli, yet the PROSS designs exhibited high expression levels in E. coli and HEK293 cells, respectively, and improved thermal stability. We conclude that PROSS may improve stability and expressibility in diverse cases, and that improvement typically requires target-specific expression conditions. This study demonstrates the strengths of community-wide efforts to probe the generality of new methods and recommends areas for future research to advance practically useful algorithms for protein science.


Asunto(s)
Algoritmos , Estabilidad Proteica , Animales , Escherichia coli/metabolismo , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Modelos Moleculares , Proteínas/química , Proteínas/metabolismo , Solubilidad , Temperatura , Pez Cebra
20.
Sci Adv ; 7(22)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-33888467

RESUMEN

The coronaviral spike is the dominant viral antigen and the target of neutralizing antibodies. We show that SARS-CoV-2 spike binds biliverdin and bilirubin, the tetrapyrrole products of heme metabolism, with nanomolar affinity. Using cryo-electron microscopy and x-ray crystallography, we mapped the tetrapyrrole interaction pocket to a deep cleft on the spike N-terminal domain (NTD). At physiological concentrations, biliverdin significantly dampened the reactivity of SARS-CoV-2 spike with immune sera and inhibited a subset of neutralizing antibodies. Access to the tetrapyrrole-sensitive epitope is gated by a flexible loop on the distal face of the NTD. Accompanied by profound conformational changes in the NTD, antibody binding requires relocation of the gating loop, which folds into the cleft vacated by the metabolite. Our results indicate that SARS-CoV-2 spike NTD harbors a dominant epitope, access to which can be controlled by an allosteric mechanism that is regulated through recruitment of a metabolite.


Asunto(s)
COVID-19/inmunología , Hemo/metabolismo , Glicoproteína de la Espiga del Coronavirus/química , Glicoproteína de la Espiga del Coronavirus/inmunología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Anticuerpos Neutralizantes/inmunología , Bilirrubina/metabolismo , Biliverdina/metabolismo , Microscopía por Crioelectrón , Cristalografía por Rayos X , Epítopos , Humanos , Sueros Inmunes , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA