Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 228
Filtrar
1.
Nat Struct Mol Biol ; 31(9): 1394-1403, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38720088

RESUMEN

Rev1-Polζ-dependent translesion synthesis (TLS) of DNA is crucial for maintaining genome integrity. To elucidate the mechanism by which the two polymerases cooperate in TLS, we determined the cryogenic electron microscopic structure of the Saccharomyces cerevisiae Rev1-Polζ holocomplex in the act of DNA synthesis (3.53 Å). We discovered that a composite N-helix-BRCT module in Rev1 is the keystone of Rev1-Polζ cooperativity, interacting directly with the DNA template-primer and with the Rev3 catalytic subunit of Polζ. The module is positioned akin to the polymerase-associated domain in Y-family TLS polymerases and is set ideally to interact with PCNA. We delineate the full extent of interactions that the carboxy-terminal domain of Rev1 makes with Polζ and identify potential new druggable sites to suppress chemoresistance from first-line chemotherapeutics. Collectively, our results provide fundamental new insights into the mechanism of cooperativity between Rev1 and Polζ in TLS.


Asunto(s)
Microscopía por Crioelectrón , ADN Polimerasa Dirigida por ADN , Modelos Moleculares , Nucleotidiltransferasas , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Nucleotidiltransferasas/metabolismo , Nucleotidiltransferasas/química , Nucleotidiltransferasas/ultraestructura , Nucleotidiltransferasas/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/ultraestructura , Proteínas de Saccharomyces cerevisiae/genética , ADN Polimerasa Dirigida por ADN/metabolismo , ADN Polimerasa Dirigida por ADN/química , Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/genética , Replicación del ADN , Proteínas Nucleares/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/ultraestructura , ADN/metabolismo , ADN/química , ADN de Hongos/metabolismo , ADN de Hongos/química , ADN de Hongos/genética , Unión Proteica , Síntesis Translesional de ADN
2.
Nature ; 629(8010): 219-227, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38570683

RESUMEN

The Integrator complex can terminate RNA polymerase II (Pol II) in the promoter-proximal region of genes. Previous work has shed light on how Integrator binds to the paused elongation complex consisting of Pol II, the DRB sensitivity-inducing factor (DSIF) and the negative elongation factor (NELF) and how it cleaves the nascent RNA transcript1, but has not explained how Integrator removes Pol II from the DNA template. Here we present three cryo-electron microscopy structures of the complete Integrator-PP2A complex in different functional states. The structure of the pre-termination complex reveals a previously unresolved, scorpion-tail-shaped INTS10-INTS13-INTS14-INTS15 module that may use its 'sting' to open the DSIF DNA clamp and facilitate termination. The structure of the post-termination complex shows that the previously unresolved subunit INTS3 and associated sensor of single-stranded DNA complex (SOSS) factors prevent Pol II rebinding to Integrator after termination. The structure of the free Integrator-PP2A complex in an inactive closed conformation2 reveals that INTS6 blocks the PP2A phosphatase active site. These results lead to a model for how Integrator terminates Pol II transcription in three steps that involve major rearrangements.


Asunto(s)
Microscopía por Crioelectrón , Modelos Moleculares , Proteína Fosfatasa 2 , ARN Polimerasa II , ARN Polimerasa II/metabolismo , ARN Polimerasa II/química , ARN Polimerasa II/ultraestructura , Proteína Fosfatasa 2/metabolismo , Proteína Fosfatasa 2/química , Proteína Fosfatasa 2/ultraestructura , Terminación de la Transcripción Genética , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/química , Unión Proteica , Factores de Elongación Transcripcional/metabolismo , Factores de Elongación Transcripcional/química , Proteínas Nucleares/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/ultraestructura , Subunidades de Proteína/metabolismo , Subunidades de Proteína/química
3.
Commun Biol ; 4(1): 1374, 2021 12 08.
Artículo en Inglés | MEDLINE | ID: mdl-34880419

RESUMEN

Huntington's disease results from expansion of a glutamine-coding CAG tract in the huntingtin (HTT) gene, producing an aberrantly functioning form of HTT. Both wildtype and disease-state HTT form a hetero-dimer with HAP40 of unknown functional relevance. We demonstrate in vivo and in cell models that HTT and HAP40 cellular abundance are coupled. Integrating data from a 2.6 Å cryo-electron microscopy structure, cross-linking mass spectrometry, small-angle X-ray scattering, and modeling, we provide a near-atomic-level view of HTT, its molecular interaction surfaces and compacted domain architecture, orchestrated by HAP40. Native mass spectrometry reveals a remarkably stable hetero-dimer, potentially explaining the cellular inter-dependence of HTT and HAP40. The exon 1 region of HTT is dynamic but shows greater conformational variety in the polyglutamine expanded mutant than wildtype exon 1. Our data provide a foundation for future functional and drug discovery studies targeting Huntington's disease and illuminate the structural consequences of HTT polyglutamine expansion.


Asunto(s)
Exones , Proteína Huntingtina/genética , Enfermedad de Huntington/genética , Proteínas Nucleares/genética , Péptidos/metabolismo , Microscopía por Crioelectrón , Humanos , Proteína Huntingtina/metabolismo , Proteína Huntingtina/ultraestructura , Proteínas Nucleares/metabolismo , Proteínas Nucleares/ultraestructura
4.
Nat Commun ; 12(1): 3176, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34039995

RESUMEN

Chromosomes pair and synapse with their homologous partners to segregate correctly at the first meiotic division. Association of telomeres with the LINC (Linker of Nucleoskeleton and Cytoskeleton) complex composed of SUN1 and KASH5 enables telomere-led chromosome movements and telomere bouquet formation, facilitating precise pairwise alignment of homologs. Here, we identify a direct interaction between SUN1 and Speedy A (SPDYA) and determine the crystal structure of human SUN1-SPDYA-CDK2 ternary complex. Analysis of meiosis prophase I process in SPDYA-binding-deficient SUN1 mutant mice reveals that the SUN1-SPDYA interaction is required for the telomere-LINC complex connection and the assembly of a ring-shaped telomere supramolecular architecture at the nuclear envelope, which is critical for efficient homologous pairing and synapsis. Overall, our results provide structural insights into meiotic telomere structure that is essential for meiotic prophase I progression.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Profase Meiótica I , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Telómero/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/aislamiento & purificación , Proteínas de Ciclo Celular/ultraestructura , Línea Celular Tumoral , Cristalografía por Rayos X , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/aislamiento & purificación , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 2 Dependiente de la Ciclina/ultraestructura , Femenino , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/aislamiento & purificación , Proteínas de la Membrana/ultraestructura , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/aislamiento & purificación , Proteínas Asociadas a Microtúbulos/ultraestructura , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/aislamiento & purificación , Proteínas Nucleares/ultraestructura , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura
5.
Nat Struct Mol Biol ; 28(4): 356-364, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33795880

RESUMEN

Ubiquitin-specific protease 1 (USP1) acts together with the cofactor UAF1 during DNA repair processes to specifically remove monoubiquitin signals. One substrate of the USP1-UAF1 complex is the monoubiquitinated FANCI-FANCD2 heterodimer, which is involved in the repair of DNA interstrand crosslinks via the Fanconi anemia pathway. Here we determine structures of human USP1-UAF1 with and without ubiquitin and bound to monoubiquitinated FANCI-FANCD2. The crystal structures of USP1-UAF1 reveal plasticity in USP1 and key differences to USP12-UAF1 and USP46-UAF1, two related proteases. A cryo-EM reconstruction of USP1-UAF1 in complex with monoubiquitinated FANCI-FANCD2 highlights a highly orchestrated deubiquitination process, with USP1-UAF1 driving conformational changes in the substrate. An extensive interface between UAF1 and FANCI, confirmed by mutagenesis and biochemical assays, provides a molecular explanation for the requirement of both proteins, despite neither being directly involved in catalysis. Overall, our data provide molecular details of USP1-UAF1 regulation and substrate recognition.


Asunto(s)
Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/ultraestructura , Proteínas Nucleares/ultraestructura , Proteasas Ubiquitina-Específicas/ultraestructura , Daño del ADN/genética , Reparación del ADN/genética , Enzimas Desubicuitinizantes/química , Enzimas Desubicuitinizantes/genética , Enzimas Desubicuitinizantes/ultraestructura , Anemia de Fanconi/genética , Anemia de Fanconi/patología , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Células HeLa , Humanos , Proteínas Nucleares/genética , Unión Proteica/genética , Conformación Proteica , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación/genética
6.
Curr Genet ; 67(4): 511-518, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-33745061

RESUMEN

This review describes the current models for how the subunit abundance of the Ndc80 complex, a key kinetochore component, is regulated in budding yeast and metazoan meiosis. The past decades of kinetochore research have established the Ndc80 complex to be a key microtubule interactor and a central hub for regulating chromosome segregation. Recent studies further demonstrate that Ndc80 is the limiting kinetochore subunit that dictates the timing of kinetochore activation in budding yeast meiosis. Here, we discuss the molecular circuits that regulate Ndc80 protein synthesis and degradation in budding yeast meiosis and compare the findings with those from metazoans. We envision the regulatory principles discovered in budding yeast to be conserved in metazoans, thereby providing guidance into future investigations on kinetochore regulation in human health and disease.


Asunto(s)
Segregación Cromosómica/genética , Proteínas del Citoesqueleto/ultraestructura , Meiosis/genética , Proteínas Nucleares/genética , Proteínas de Saccharomyces cerevisiae/genética , Proteínas del Citoesqueleto/genética , Humanos , Cinetocoros/ultraestructura , Microtúbulos/genética , Proteínas Nucleares/ultraestructura , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/ultraestructura
7.
Curr Opin Genet Dev ; 67: 130-141, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33524904

RESUMEN

The regulation of genomic function is in part mediated through the physical organization and architecture of the nucleus. Disruption to nuclear organization and architecture is increasingly being recognized by its contribution to many diseases. The LINC complexes - protein structures traversing the nuclear envelope, that physically connect the nuclear interior, and hence the genome, to cytoplasmic cytoskeletal networks are an important component in the physical organization of the genome and its function. This connection, potentially allows for the constant detection of environmental mechanical stimuli, resulting in altered regulation of nuclear architecture and genome function, either directly or via the process of mechanotransduction. Here, we review the influences LINC complexes exert on genome functions and their impact on cellular/organismal health.


Asunto(s)
Genoma/genética , Mecanotransducción Celular/genética , Proteínas Nucleares/genética , ARN Largo no Codificante/genética , Núcleo Celular/genética , Núcleo Celular/ultraestructura , Citoplasma/genética , Citoplasma/ultraestructura , Citoesqueleto/genética , Citoesqueleto/ultraestructura , Humanos , Membrana Nuclear/genética , Membrana Nuclear/ultraestructura , Proteínas Nucleares/ultraestructura , ARN Largo no Codificante/ultraestructura
8.
Int J Mol Sci ; 22(4)2021 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-33572172

RESUMEN

Low complexity regions (LCRs) are very frequent in protein sequences, generally having a lower propensity to form structured domains and tending to be much less evolutionarily conserved than globular domains. Their higher abundance in eukaryotes and in species with more cellular types agrees with a growing number of reports on their function in protein interactions regulated by post-translational modifications. LCRs facilitate the increase of regulatory and network complexity required with the emergence of organisms with more complex tissue distribution and development. Although the low conservation and structural flexibility of LCRs complicate their study, evolutionary studies of proteins across species have been used to evaluate their significance and function. To investigate how to apply this evolutionary approach to the study of LCR function in protein-protein interactions, we performed a detailed analysis for Huntingtin (HTT), a large protein that is a hub for interaction with hundreds of proteins, has a variety of LCRs, and for which partial structural information (in complex with HAP40) is available. We hypothesize that proteins RASA1, SYN2, and KAT2B may compete with HAP40 for their attachment to the core of HTT using similar LCRs. Our results illustrate how evolution might favor the interplay of LCRs with domains, and the possibility of detecting multiple modes of LCR-mediated protein-protein interactions with a large hub such as HTT when enough protein interaction data is available.


Asunto(s)
Evolución Molecular , Proteína Huntingtina/metabolismo , Proteínas Nucleares/metabolismo , Secuencias de Aminoácidos/genética , Secuencia de Aminoácidos/genética , Animales , Humanos , Proteína Huntingtina/química , Proteína Huntingtina/genética , Proteína Huntingtina/ultraestructura , Microscopía Electrónica , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/ultraestructura , Unión Proteica/genética , Conformación Proteica en Hélice alfa/genética , Dominios Proteicos/genética , Mapeo de Interacción de Proteínas , Mapas de Interacción de Proteínas , Alineación de Secuencia , Sinapsinas/química , Sinapsinas/metabolismo , Proteína Activadora de GTPasa p120/química , Proteína Activadora de GTPasa p120/metabolismo , Factores de Transcripción p300-CBP/química , Factores de Transcripción p300-CBP/metabolismo
9.
FEBS J ; 288(9): 2757-2772, 2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-32799420

RESUMEN

Progeroid laminopathies are characterized by the premature appearance of certain signs of physiological aging in a subset of tissues. They are caused by mutations in genes coding for A-type lamins or lamin-binding proteins. Here, we review how different mutations causing progeroid laminopathies alter protein structure or protein-protein interactions and how these impact on mechanisms that protect cell viability and function. One group of progeroid laminopathies, which includes Hutchinson-Gilford progeria syndrome, is characterized by accumulation of unprocessed prelamin A or variants. These are caused by mutations in the A-type lamin gene (LMNA), altering prelamin A itself, or in ZMPSTE24, encoding an endoprotease involved in its processing. The abnormally expressed farnesylated proteins impact on various cellular processes that may contribute to progeroid phenotypes. Other LMNA mutations lead to the production of nonfarnesylated A-type lamin variants with amino acid substitutions in solvent-exposed hot spots located mainly in coil 1B and the immunoglobulin fold domain. Dominant missense mutations might reinforce interactions between lamin domains, thus giving rise to excessively stabilized filament networks. Recessive missense mutations in A-type lamins and barrier-to-autointegration factor (BAF) causing progeroid disorders are found at the interface between these interacting proteins. The amino acid changes decrease the binding affinity of A-type lamins for BAF, which may contribute to lamina disorganization, as well as defective repair of mechanically induced nuclear envelope rupture. Targeting these molecular alterations in A-type lamins and associated proteins identified through structural biology studies could facilitate the design of therapeutic strategies to treat patients with rare but severe progeroid laminopathies.


Asunto(s)
Lamina Tipo A/genética , Laminopatías/genética , Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Progeria/genética , Aminoácidos/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Lamina Tipo A/ultraestructura , Laminopatías/patología , Proteínas de la Membrana/ultraestructura , Metaloendopeptidasas/ultraestructura , Mutación Missense/genética , Proteínas Nucleares/genética , Proteínas Nucleares/ultraestructura , Progeria/patología , Conformación Proteica
10.
Nature ; 590(7846): 498-503, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33361816

RESUMEN

Histone methyltransferases of the nuclear receptor-binding SET domain protein (NSD) family, including NSD1, NSD2 and NSD3, have crucial roles in chromatin regulation and are implicated in oncogenesis1,2. NSD enzymes exhibit an autoinhibitory state that is relieved by binding to nucleosomes, enabling dimethylation of histone H3 at Lys36 (H3K36)3-7. However, the molecular basis that underlies this mechanism is largely unknown. Here we solve the cryo-electron microscopy structures of NSD2 and NSD3 bound to mononucleosomes. We find that binding of NSD2 and NSD3 to mononucleosomes causes DNA near the linker region to unwrap, which facilitates insertion of the catalytic core between the histone octamer and the unwrapped segment of DNA. A network of DNA- and histone-specific contacts between NSD2 or NSD3 and the nucleosome precisely defines the position of the enzyme on the nucleosome, explaining the specificity of methylation to H3K36. Intermolecular contacts between NSD proteins and nucleosomes are altered by several recurrent cancer-associated mutations in NSD2 and NSD3. NSDs that contain these mutations are catalytically hyperactive in vitro and in cells, and their ectopic expression promotes the proliferation of cancer cells and the growth of xenograft tumours. Together, our research provides molecular insights into the nucleosome-based recognition and histone-modification mechanisms of NSD2 and NSD3, which could lead to strategies for therapeutic targeting of proteins of the NSD family.


Asunto(s)
N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/química , Histonas/metabolismo , Proteínas Nucleares/metabolismo , Nucleosomas/química , Nucleosomas/metabolismo , Proteínas Represoras/metabolismo , Sitios de Unión , Biocatálisis , Línea Celular Tumoral , Proliferación Celular , Microscopía por Crioelectrón , Xenoinjertos , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/ultraestructura , Histonas/ultraestructura , Humanos , Metilación , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Mutación , Trasplante de Neoplasias , Neoplasias/genética , Neoplasias/patología , Proteínas Nucleares/genética , Proteínas Nucleares/ultraestructura , Nucleosomas/ultraestructura , Fenotipo , Unión Proteica , Proteínas Represoras/genética , Proteínas Represoras/ultraestructura
11.
Nature ; 587(7835): 683-687, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33208940

RESUMEN

Eukaryotic ribosomes consist of a small 40S and a large 60S subunit that are assembled in a highly coordinated manner. More than 200 factors ensure correct modification, processing and folding of ribosomal RNA and the timely incorporation of ribosomal proteins1,2. Small subunit maturation ends in the cytosol, when the final rRNA precursor, 18S-E, is cleaved at site 3 by the endonuclease NOB13. Previous structures of human 40S precursors have shown that NOB1 is kept in an inactive state by its partner PNO14. The final maturation events, including the activation of NOB1 for the decisive rRNA-cleavage step and the mechanisms driving the dissociation of the last biogenesis factors have, however, remained unresolved. Here we report five cryo-electron microscopy structures of human 40S subunit precursors, which describe the compositional and conformational progression during the final steps of 40S assembly. Our structures explain the central role of RIOK1 in the displacement and dissociation of PNO1, which in turn allows conformational changes and activation of the endonuclease NOB1. In addition, we observe two factors, eukaryotic translation initiation factor 1A domain-containing protein (EIF1AD) and leucine-rich repeat-containing protein 47 (LRRC47), which bind to late pre-40S particles near RIOK1 and the central rRNA helix 44. Finally, functional data shows that EIF1AD is required for efficient assembly factor recycling and 18S-E processing. Our results thus enable a detailed understanding of the last steps in 40S formation in human cells and, in addition, provide evidence for principal differences in small ribosomal subunit formation between humans and the model organism Saccharomyces cerevisiae.


Asunto(s)
Microscopía por Crioelectrón , Subunidades Ribosómicas Pequeñas de Eucariotas/química , Subunidades Ribosómicas Pequeñas de Eucariotas/metabolismo , Activación Enzimática , Células HeLa , Humanos , Modelos Moleculares , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas Nucleares/ultraestructura , Conformación Proteica , Proteínas Serina-Treonina Quinasas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Serina-Treonina Quinasas/ultraestructura , Proteínas/química , Proteínas/metabolismo , Proteínas/ultraestructura , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/ultraestructura , Subunidades Ribosómicas Pequeñas de Eucariotas/ultraestructura , Saccharomyces cerevisiae/química
12.
Nature ; 587(7835): 638-643, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-33208942

RESUMEN

Aquatic birds represent a vast reservoir from which new pandemic influenza A viruses can emerge1. Influenza viruses contain a negative-sense segmented RNA genome that is transcribed and replicated by the viral heterotrimeric RNA polymerase (FluPol) in the context of viral ribonucleoprotein complexes2,3. RNA polymerases of avian influenza A viruses (FluPolA) replicate viral RNA inefficiently in human cells because of species-specific differences in acidic nuclear phosphoprotein 32 (ANP32), a family of essential host proteins for FluPol activity4. Host-adaptive mutations, particularly a glutamic-acid-to-lysine mutation at amino acid residue 627 (E627K) in the 627 domain of the PB2 subunit, enable avian FluPolA to overcome this restriction and efficiently replicate viral RNA in the presence of human ANP32 proteins. However, the molecular mechanisms of genome replication and the interplay with ANP32 proteins remain largely unknown. Here we report cryo-electron microscopy structures of influenza C virus polymerase (FluPolC) in complex with human and chicken ANP32A. In both structures, two FluPolC molecules form an asymmetric dimer bridged by the N-terminal leucine-rich repeat domain of ANP32A. The C-terminal low-complexity acidic region of ANP32A inserts between the two juxtaposed PB2 627 domains of the asymmetric FluPolA dimer, suggesting a mechanism for how the adaptive PB2(E627K) mutation enables the replication of viral RNA in mammalian hosts. We propose that this complex represents a replication platform for the viral RNA genome, in which one of the FluPol molecules acts as a replicase while the other initiates the assembly of the nascent replication product into a viral ribonucleoprotein complex.


Asunto(s)
Microscopía por Crioelectrón , Gammainfluenzavirus/enzimología , Interacciones Huésped-Patógeno , Proteínas Nucleares/química , Proteínas Nucleares/metabolismo , Proteínas de Unión al ARN/química , Proteínas de Unión al ARN/metabolismo , ARN Polimerasa Dependiente del ARN/química , ARN Polimerasa Dependiente del ARN/metabolismo , Animales , Pollos/virología , Genoma Viral/genética , Células HEK293 , Humanos , Gammainfluenzavirus/genética , Modelos Moleculares , Proteínas Nucleares/ultraestructura , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Multimerización de Proteína , ARN Viral/biosíntesis , ARN Viral/genética , Proteínas de Unión al ARN/ultraestructura , ARN Polimerasa Dependiente del ARN/ultraestructura , Células Sf9
13.
J Mol Biol ; 432(23): 6061-6074, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33058883

RESUMEN

The AAA-ATPase VCP/p97 cooperates with the SEP-domain adapters p37, UBXN2A and p47 in stripping inhibitor-3 (I3) from protein phosphatase-1 (PP1) for activation. In contrast to p97-mediated degradative processes, PP1 complex disassembly is ubiquitin-independent. It is therefore unclear how selective targeting is achieved. Using biochemical reconstitution and crosslink mass spectrometry, we show here that SEP-domain adapters use a multivalent substrate recognition strategy. An N-terminal sequence element predicted to form a helix, together with the SEP-domain, binds and engages the direct target I3 in the central pore of p97 for unfolding, while its partner PP1 is held by a linker between SHP box and UBX domain locked onto the peripheral N-domain of p97. Although the I3-binding element is functional in p47, p47 in vitro requires a transplant of the PP1-binding linker from p37 for activity stressing that both sites are essential to control specificity. Of note, unfolding is then governed by an inhibitory segment in the N-terminal region of p47, suggesting a regulatory function. Together, this study reveals how p97 adapters engage a protein complex for ubiquitin-independent disassembly while ensuring selectivity for one subunit.


Asunto(s)
Adenosina Trifosfatasas/química , Complejos Multiproteicos/química , Proteínas Nucleares/química , Conformación Proteica , Proteína Fosfatasa 1/química , Proteínas Adaptadoras Transductoras de Señales/química , Proteínas Adaptadoras Transductoras de Señales/genética , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/ultraestructura , Secuencia de Aminoácidos/genética , Dominio Catalítico/genética , Cristalografía por Rayos X , Humanos , Metaloendopeptidasas/química , Metaloendopeptidasas/genética , Complejos Multiproteicos/genética , Complejos Multiproteicos/ultraestructura , Proteínas Nucleares/genética , Proteínas Nucleares/ultraestructura , Unión Proteica/genética , Proteína Fosfatasa 1/genética , Proteína Fosfatasa 1/ultraestructura , Estructura Terciaria de Proteína , Subunidades de Proteína/química , Ubiquitina/genética , Ubiquitinas/química , Ubiquitinas/genética
14.
Nat Struct Mol Biol ; 27(11): 1086-1093, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32929281

RESUMEN

DUOX1, an NADPH oxidase family member, catalyzes the production of hydrogen peroxide. DUOX1 is expressed in various tissues, including the thyroid and respiratory tract, and plays a crucial role in processes such as thyroid hormone biosynthesis and innate host defense. DUOX1 co-assembles with its maturation factor DUOXA1 to form an active enzyme complex. However, the molecular mechanisms for activation and regulation of DUOX1 remain mostly unclear. Here, I present cryo-EM structures of the mammalian DUOX1-DUOXA1 complex, in the absence and presence of substrate NADPH, as well as DUOX1-DUOXA1 in an unexpected dimer-of-dimers configuration. These structures reveal atomic details of the DUOX1-DUOXA1 interaction, a lipid-mediated NADPH-binding pocket and the electron transfer path. Furthermore, biochemical and structural analyses indicate that the dimer-of-dimers configuration represents an inactive state of DUOX1-DUOXA1, suggesting an oligomerization-dependent regulatory mechanism. Together, my work provides structural bases for DUOX1-DUOXA1 activation and regulation.


Asunto(s)
Oxidasas Duales/metabolismo , Activación Enzimática , Proteínas del Tejido Nervioso/metabolismo , Proteínas Nucleares/metabolismo , Animales , Microscopía por Crioelectrón , Oxidasas Duales/química , Oxidasas Duales/ultraestructura , Ratones , Modelos Moleculares , NADP/metabolismo , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/ultraestructura , Proteínas Nucleares/química , Proteínas Nucleares/ultraestructura , Conformación Proteica
15.
Nature ; 585(7826): 609-613, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32939087

RESUMEN

Breaks in DNA strands recruit the protein PARP1 and its paralogue PARP2 to modify histones and other substrates through the addition of mono- and poly(ADP-ribose) (PAR)1-5. In the DNA damage responses, this post-translational modification occurs predominantly on serine residues6-8 and requires HPF1, an accessory factor that switches the amino acid specificity of PARP1 and PARP2 from aspartate or glutamate to serine9,10. Poly(ADP) ribosylation (PARylation) is important for subsequent chromatin decompaction and provides an anchor for the recruitment of downstream signalling and repair factors to the sites of DNA breaks2,11. Here, to understand the molecular mechanism by which PARP enzymes recognize DNA breaks within chromatin, we determined the cryo-electron-microscopic structure of human PARP2-HPF1 bound to a nucleosome. This showed that PARP2-HPF1 bridges two nucleosomes, with the broken DNA aligned in a position suitable for ligation, revealing the initial step in the repair of double-strand DNA breaks. The bridging induces structural changes in PARP2 that signal the recognition of a DNA break to the catalytic domain, which licenses HPF1 binding and PARP2 activation. Our data suggest that active PARP2 cycles through different conformational states to exchange NAD+ and substrate, which may enable PARP enzymes to act processively while bound to chromatin. The processes of PARP activation and the PARP catalytic cycle we describe can explain mechanisms of resistance to PARP inhibitors and will aid the development of better inhibitors as cancer treatments12-16.


Asunto(s)
Proteínas Portadoras/metabolismo , Roturas del ADN de Doble Cadena , Proteínas Nucleares/metabolismo , Nucleosomas/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Biocatálisis , Proteínas Portadoras/química , Proteínas Portadoras/ultraestructura , Microscopía por Crioelectrón , ADN/metabolismo , Reparación del ADN , Activación Enzimática , Humanos , Modelos Moleculares , NAD/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/ultraestructura , Nucleosomas/química , Nucleosomas/ultraestructura , Poli(ADP-Ribosa) Polimerasas/química , Poli(ADP-Ribosa) Polimerasas/ultraestructura , Dominios Proteicos
16.
Nucleic Acids Res ; 48(18): 10313-10328, 2020 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-32976585

RESUMEN

Transcription of integrated DNA from viruses or transposable elements is tightly regulated to prevent pathogenesis. The Human Silencing Hub (HUSH), composed of Periphilin, TASOR and MPP8, silences transcriptionally active viral and endogenous transgenes. HUSH recruits effectors that alter the epigenetic landscape and chromatin structure, but how HUSH recognizes target loci and represses their expression remains unclear. We identify the physicochemical properties of Periphilin necessary for HUSH assembly and silencing. A disordered N-terminal domain (NTD) and structured C-terminal domain are essential for silencing. A crystal structure of the Periphilin-TASOR minimal core complex shows Periphilin forms an α-helical homodimer, bound by a single TASOR molecule. The NTD forms insoluble aggregates through an arginine/tyrosine-rich sequence reminiscent of low-complexity regions from self-associating RNA-binding proteins. Residues required for TASOR binding and aggregation were required for HUSH-dependent silencing and genome-wide deposition of repressive mark H3K9me3. The NTD was functionally complemented by low-complexity regions from certain RNA-binding proteins and proteins that form condensates or fibrils. Our work suggests the associative properties of Periphilin promote HUSH aggregation at target loci.


Asunto(s)
Antígenos de Neoplasias/ultraestructura , Proteínas Nucleares/ultraestructura , Proteínas de Unión al ARN/química , Transcripción Genética , Antígenos de Neoplasias/química , Antígenos de Neoplasias/genética , Cristalografía por Rayos X , Elementos Transponibles de ADN/genética , Epigénesis Genética/genética , Silenciador del Gen , Humanos , Proteínas Nucleares/química , Proteínas Nucleares/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Agregado de Proteínas/genética , Unión Proteica/genética , Conformación Proteica en Hélice alfa , Dominios Proteicos/genética , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/ultraestructura , Virus/genética
17.
Nat Struct Mol Biol ; 27(8): 743-751, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32661420

RESUMEN

Complexes containing a pair of structural maintenance of chromosomes (SMC) family proteins are fundamental for the three-dimensional (3D) organization of genomes in all domains of life. The eukaryotic SMC complexes cohesin and condensin are thought to fold interphase and mitotic chromosomes, respectively, into large loop domains, although the underlying molecular mechanisms have remained unknown. We used cryo-EM to investigate the nucleotide-driven reaction cycle of condensin from the budding yeast Saccharomyces cerevisiae. Our structures of the five-subunit condensin holo complex at different functional stages suggest that ATP binding induces the transition of the SMC coiled coils from a folded-rod conformation into a more open architecture. ATP binding simultaneously triggers the exchange of the two HEAT-repeat subunits bound to the SMC ATPase head domains. We propose that these steps result in the interconversion of DNA-binding sites in the catalytic core of condensin, forming the basis of the DNA translocation and loop-extrusion activities.


Asunto(s)
Proteínas Portadoras/química , Proteínas Cromosómicas no Histona/química , Proteínas Nucleares/química , Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/ultraestructura , Adenosina Trifosfato/metabolismo , Proteínas Portadoras/metabolismo , Proteínas Portadoras/ultraestructura , Proteínas de Ciclo Celular , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/ultraestructura , Microscopía por Crioelectrón , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/ultraestructura , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Proteínas Nucleares/metabolismo , Proteínas Nucleares/ultraestructura , Conformación Proteica , Pliegue de Proteína , Multimerización de Proteína , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/ultraestructura
18.
Nat Commun ; 11(1): 3656, 2020 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-32694517

RESUMEN

Avian influenza polymerase undergoes host adaptation in order to efficiently replicate in human cells. Adaptive mutants are localised on the C-terminal (627-NLS) domains of the PB2 subunit. In particular, mutation of PB2 residue 627 from E to K rescues polymerase activity in mammalian cells. A host transcription regulator ANP32A, comprising a long C-terminal intrinsically disordered domain (IDD), is responsible for this adaptation. Human ANP32A IDD lacks a 33 residue insertion compared to avian ANP32A, and this deletion restricts avian influenza polymerase activity. We used NMR to determine conformational ensembles of E627 and K627 forms of 627-NLS of PB2 in complex with avian and human ANP32A. Human ANP32A IDD transiently binds to the 627 domain, exploiting multivalency to maximise affinity. E627 interrupts the polyvalency of the interaction, an effect compensated by an avian-unique motif in the IDD. The observed binding mode is maintained in the context of heterotrimeric influenza polymerase, placing ANP32A in the immediate vicinity of known host-adaptive PB2 mutants.


Asunto(s)
Proteínas Aviares/ultraestructura , Subtipo H5N1 del Virus de la Influenza A/patogenicidad , Proteínas Nucleares/ultraestructura , Dominios Proteicos/genética , Proteínas de Unión al ARN/ultraestructura , ARN Polimerasa Dependiente del ARN/ultraestructura , Proteínas Virales/ultraestructura , Animales , Proteínas Aviares/metabolismo , Aves/virología , Humanos , Subtipo H5N1 del Virus de la Influenza A/genética , Subtipo H5N1 del Virus de la Influenza A/metabolismo , Gripe Aviar/virología , Gripe Humana/virología , Mutación , Resonancia Magnética Nuclear Biomolecular , Proteínas Nucleares/metabolismo , Unión Proteica/genética , Proteínas de Unión al ARN/metabolismo , ARN Polimerasa Dependiente del ARN/genética , ARN Polimerasa Dependiente del ARN/metabolismo , Especificidad de la Especie , Proteínas Virales/genética , Proteínas Virales/metabolismo , Replicación Viral
19.
Hum Genet ; 139(12): 1513-1529, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32529326

RESUMEN

Prader-Willi syndrome (PWS) is a neurodevelopmental disorder caused by the loss of function of a set of imprinted genes on chromosome 15q11-15q13. One of these genes, NDN, encodes necdin, a protein that is important for neuronal differentiation and survival. Loss of Ndn in mice causes defects in the formation and function of the nervous system. Necdin is a member of the melanoma-associated antigen gene (MAGE) protein family. The functions of MAGE proteins depend highly on their interactions with other proteins, and in particular MAGE proteins interact with E3 ubiquitin ligases and deubiquitinases to form MAGE-RING E3 ligase-deubiquitinase complexes. Here, we used proximity-dependent biotin identification (BioID) and mass spectrometry (MS) to determine the network of protein-protein interactions (interactome) of the necdin protein. This process yielded novel as well as known necdin-proximate proteins that cluster into a protein network. Next, we used BioID-MS to define the interactomes of necdin proteins carrying coding variants. Variant necdin proteins had interactomes that were distinct from wildtype necdin. BioID-MS is not only a useful tool to identify protein-protein interactions, but also to analyze the effects of variants of unknown significance on the interactomes of proteins involved in genetic disease.


Asunto(s)
Sustitución de Aminoácidos/genética , Proteínas del Tejido Nervioso/genética , Proteínas Nucleares/genética , Mapas de Interacción de Proteínas/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Biotinilación/genética , Diferenciación Celular/genética , Enzimas Desubicuitinizantes/genética , Regulación de la Expresión Génica/genética , Células HEK293 , Humanos , Espectrometría de Masas/métodos , Ratones , Mutación/genética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/ultraestructura , Malformaciones del Sistema Nervioso/genética , Malformaciones del Sistema Nervioso/patología , Neuronas/metabolismo , Proteínas Nucleares/química , Proteínas Nucleares/ultraestructura , Proteínas de Unión a Poli(A)/química , Proteínas de Unión a Poli(A)/genética , Síndrome de Prader-Willi/genética , Conformación Proteica , Relación Estructura-Actividad , Ubiquitina-Proteína Ligasas/química
20.
Mol Cell ; 78(5): 926-940.e13, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32369734

RESUMEN

The eukaryotic replisome, organized around the Cdc45-MCM-GINS (CMG) helicase, orchestrates chromosome replication. Multiple factors associate directly with CMG, including Ctf4 and the heterotrimeric fork protection complex (Csm3/Tof1 and Mrc1), which has important roles including aiding normal replication rates and stabilizing stalled forks. How these proteins interface with CMG to execute these functions is poorly understood. Here we present 3 to 3.5 Å resolution electron cryomicroscopy (cryo-EM) structures comprising CMG, Ctf4, and the fork protection complex at a replication fork. The structures provide high-resolution views of CMG-DNA interactions, revealing a mechanism for strand separation, and show Csm3/Tof1 "grip" duplex DNA ahead of CMG via a network of interactions important for efficient replication fork pausing. Although Mrc1 was not resolved in our structures, we determine its topology in the replisome by cross-linking mass spectrometry. Collectively, our work reveals how four highly conserved replisome components collaborate with CMG to facilitate replisome progression and maintain genome stability.


Asunto(s)
Proteínas de Unión al ADN/ultraestructura , Proteínas de Mantenimiento de Minicromosoma/ultraestructura , Proteínas Nucleares/ultraestructura , Proteínas de Saccharomyces cerevisiae/ultraestructura , Proteínas de Ciclo Celular/metabolismo , Microscopía por Crioelectrón/métodos , ADN Helicasas/genética , Replicación del ADN/genética , Replicación del ADN/fisiología , ADN de Hongos/genética , Proteínas de Unión al ADN/metabolismo , Proteínas de Mantenimiento de Minicromosoma/metabolismo , Proteínas Nucleares/metabolismo , Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA