Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 215
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38838667

RESUMEN

Telomere maintenance requires the extension of the G-rich telomeric repeat strand by telomerase and the fill-in synthesis of the C-rich strand by Polα/primase. At telomeres, Polα/primase is bound to Ctc1/Stn1/Ten1 (CST), a single-stranded DNA-binding complex. Like mutations in telomerase, mutations affecting CST-Polα/primase result in pathological telomere shortening and cause a telomere biology disorder, Coats plus (CP). We determined cryogenic electron microscopy structures of human CST bound to the shelterin heterodimer POT1/TPP1 that reveal how CST is recruited to telomeres by POT1. Our findings suggest that POT1 hinge phosphorylation is required for CST recruitment, and the complex is formed through conserved interactions involving several residues mutated in CP. Our structural and biochemical data suggest that phosphorylated POT1 holds CST-Polα/primase in an inactive, autoinhibited state until telomerase has extended the telomere ends. We propose that dephosphorylation of POT1 releases CST-Polα/primase into an active state that completes telomere replication through fill-in synthesis.

2.
Cell ; 183(3): 802-817.e24, 2020 10 29.
Artículo en Inglés | MEDLINE | ID: mdl-33053319

RESUMEN

Mammalian SWI/SNF complexes are ATP-dependent chromatin remodeling complexes that regulate genomic architecture. Here, we present a structural model of the endogenously purified human canonical BAF complex bound to the nucleosome, generated using cryoelectron microscopy (cryo-EM), cross-linking mass spectrometry, and homology modeling. BAF complexes bilaterally engage the nucleosome H2A/H2B acidic patch regions through the SMARCB1 C-terminal α-helix and the SMARCA4/2 C-terminal SnAc/post-SnAc regions, with disease-associated mutations in either causing attenuated chromatin remodeling activities. Further, we define changes in BAF complex architecture upon nucleosome engagement and compare the structural model of endogenous BAF to those of related SWI/SNF-family complexes. Finally, we assign and experimentally interrogate cancer-associated hot-spot mutations localizing within the endogenous human BAF complex, identifying those that disrupt BAF subunit-subunit and subunit-nucleosome interfaces in the nucleosome-bound conformation. Taken together, this integrative structural approach provides important biophysical foundations for understanding the mechanisms of BAF complex function in normal and disease states.


Asunto(s)
Enfermedad , Modelos Moleculares , Complejos Multiproteicos/química , Complejos Multiproteicos/metabolismo , Ensamble y Desensamble de Cromatina , Microscopía por Crioelectrón , ADN Helicasas/química , ADN Helicasas/genética , ADN Helicasas/metabolismo , Enfermedad/genética , Humanos , Mutación Missense/genética , Proteínas Nucleares/química , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Nucleosomas/metabolismo , Unión Proteica , Dominios Proteicos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Saccharomyces cerevisiae/metabolismo , Homología Estructural de Proteína , Factores de Transcripción/química , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
Cell ; 175(3): 822-834.e18, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30318141

RESUMEN

Mdn1 is an essential AAA (ATPase associated with various activities) protein that removes assembly factors from distinct precursors of the ribosomal 60S subunit. However, Mdn1's large size (∼5,000 amino acid [aa]) and its limited homology to other well-studied proteins have restricted our understanding of its remodeling function. Here, we present structures for S. pombe Mdn1 in the presence of AMPPNP at up to ∼4 Å or ATP plus Rbin-1, a chemical inhibitor, at ∼8 Å resolution. These data reveal that Mdn1's MIDAS domain is tethered to its ring-shaped AAA domain through an ∼20 nm long structured linker and a flexible ∼500 aa Asp/Glu-rich motif. We find that the MIDAS domain, which also binds other ribosome-assembly factors, docks onto the AAA ring in a nucleotide state-specific manner. Together, our findings reveal how conformational changes in the AAA ring can be directly transmitted to the MIDAS domain and thereby drive the targeted release of assembly factors from ribosomal 60S-subunit precursors.


Asunto(s)
ATPasas Asociadas con Actividades Celulares Diversas/química , Simulación de Dinámica Molecular , Proteínas de Schizosaccharomyces pombe/química , Schizosaccharomyces/enzimología , Secuencias de Aminoácidos , Animales , Sitios de Unión , Microscopía por Crioelectrón , Biogénesis de Organelos , Unión Proteica , Subunidades Ribosómicas Grandes de Eucariotas/metabolismo , Células Sf9 , Spodoptera
4.
Cell ; 161(3): 438-449, 2015 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-25910204

RESUMEN

Cryo-electron microscopy (cryo-EM) of single-particle specimens is used to determine the structure of proteins and macromolecular complexes without the need for crystals. Recent advances in detector technology and software algorithms now allow images of unprecedented quality to be recorded and structures to be determined at near-atomic resolution. However, compared with X-ray crystallography, cryo-EM is a young technique with distinct challenges. This primer explains the different steps and considerations involved in structure determination by single-particle cryo-EM to provide an overview for scientists wishing to understand more about this technique and the interpretation of data obtained with it, as well as a starting guide for new practitioners.


Asunto(s)
Microscopía por Crioelectrón/métodos , Conformación Molecular , Proteínas/ultraestructura , Algoritmos , Microscopía por Crioelectrón/instrumentación , Procesamiento de Imagen Asistido por Computador , Modelos Moleculares , Conformación Proteica , Proteínas/química , Proteínas/aislamiento & purificación
5.
Cell ; 158(4): 778-792, 2014 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-25109876

RESUMEN

Ionotropic glutamate receptors (iGluRs) mediate the majority of fast excitatory signaling in the nervous system. Despite the profound importance of iGluRs to neurotransmission, little is known about the structures and dynamics of intact receptors in distinct functional states. Here, we elucidate the structures of the intact GluA2 AMPA receptor in an apo resting/closed state, in an activated/pre-open state bound with partial agonists and a positive allosteric modulator, and in a desensitized/closed state in complex with fluorowilliardiine. To probe the conformational properties of these states, we carried out double electron-electron resonance experiments on cysteine mutants and cryoelectron microscopy studies. We show how agonist binding modulates the conformation of the ligand-binding domain "layer" of the intact receptors and how, upon desensitization, the receptor undergoes large conformational rearrangements of the amino-terminal and ligand-binding domains. We define mechanistic principles by which to understand antagonism, activation, and desensitization in AMPA iGluRs.


Asunto(s)
Receptores AMPA/química , Receptores AMPA/metabolismo , Animales , Microscopía por Crioelectrón , Cristalografía por Rayos X , Fluorouracilo/análogos & derivados , Fluorouracilo/metabolismo , Técnicas de Inactivación de Genes , Ácido Kaínico/metabolismo , Estructura Molecular , Resonancia Magnética Nuclear Biomolecular , Estructura Terciaria de Proteína , Ratas , Receptores AMPA/agonistas , Receptores AMPA/genética
6.
Cell ; 156(3): 482-94, 2014 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-24485456

RESUMEN

SYG-1 and SYG-2 are multipurpose cell adhesion molecules (CAMs) that have evolved across all major animal taxa to participate in diverse physiological functions, ranging from synapse formation to formation of the kidney filtration barrier. In the crystal structures of several SYG-1 and SYG-2 orthologs and their complexes, we find that SYG-1 orthologs homodimerize through a common, bispecific interface that similarly mediates an unusual orthogonal docking geometry in the heterophilic SYG-1/SYG-2 complex. C. elegans SYG-1's specification of proper synapse formation in vivo closely correlates with the heterophilic complex affinity, which appears to be tuned for optimal function. Furthermore, replacement of the interacting domains of SYG-1 and SYG-2 with those from CAM complexes that assume alternative docking geometries or the introduction of segmental flexibility compromised synaptic function. These results suggest that SYG extracellular complexes do not simply act as "molecular velcro" and that their distinct structural features are important in instructing synaptogenesis. PAPERFLICK:


Asunto(s)
Proteínas de Caenorhabditis elegans/metabolismo , Caenorhabditis elegans/citología , Inmunoglobulinas/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Sinapsis/metabolismo , Secuencia de Aminoácidos , Animales , Caenorhabditis elegans/metabolismo , Proteínas de Caenorhabditis elegans/química , Adhesión Celular , Dimerización , Inmunoglobulinas/química , Modelos Moleculares , Datos de Secuencia Molecular , Proteínas del Tejido Nervioso/química , Neuronas/citología , Neuronas/metabolismo , Estructura Terciaria de Proteína , Alineación de Secuencia , Sinapsis/química
7.
Mol Cell ; 81(2): 281-292.e8, 2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33296676

RESUMEN

Rho is a general transcription termination factor playing essential roles in RNA polymerase (RNAP) recycling, gene regulation, and genomic stability in most bacteria. Traditional models of transcription termination postulate that hexameric Rho loads onto RNA prior to contacting RNAP and then translocates along the transcript in pursuit of the moving RNAP to pull RNA from it. Here, we report the cryoelectron microscopy (cryo-EM) structures of two termination process intermediates. Prior to interacting with RNA, Rho forms a specific "pre-termination complex" (PTC) with RNAP and elongation factors NusA and NusG, which stabilize the PTC. RNA exiting RNAP interacts with NusA before entering the central channel of Rho from the distal C-terminal side of the ring. We map the principal interactions in the PTC and demonstrate their critical role in termination. Our results support a mechanism in which the formation of a persistent PTC is a prerequisite for termination.


Asunto(s)
ARN Polimerasas Dirigidas por ADN/química , Proteínas de Escherichia coli/química , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica , Factores de Elongación de Péptidos/química , Factores de Transcripción/química , Terminación de la Transcripción Genética , Factores de Elongación Transcripcional/química , Secuencia de Aminoácidos , Sitios de Unión , Clonación Molecular , Microscopía por Crioelectrón , ADN Bacteriano/química , ADN Bacteriano/genética , ADN Bacteriano/metabolismo , ARN Polimerasas Dirigidas por ADN/genética , ARN Polimerasas Dirigidas por ADN/metabolismo , Escherichia coli/metabolismo , Proteínas de Escherichia coli/genética , Proteínas de Escherichia coli/metabolismo , Vectores Genéticos/química , Vectores Genéticos/metabolismo , Modelos Moleculares , Factores de Elongación de Péptidos/genética , Factores de Elongación de Péptidos/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , ARN Bacteriano/química , ARN Bacteriano/genética , ARN Bacteriano/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factores de Elongación Transcripcional/genética , Factores de Elongación Transcripcional/metabolismo
8.
Cell ; 152(1-2): 276-89, 2013 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-23273991

RESUMEN

MDA5, a viral double-stranded RNA (dsRNA) receptor, shares sequence similarity and signaling pathways with RIG-I yet plays essential functions in antiviral immunity through distinct specificity for viral RNA. Revealing the molecular basis for the functional divergence, we report here the crystal structure of MDA5 bound to dsRNA, which shows how, using the same domain architecture, MDA5 recognizes the internal duplex structure, whereas RIG-I recognizes the terminus of dsRNA. We further show that MDA5 uses direct protein-protein contacts to stack along dsRNA in a head-to-tail arrangement, and that the signaling domain (tandem CARD), which decorates the outside of the core MDA5 filament, also has an intrinsic propensity to oligomerize into an elongated structure that activates the signaling adaptor, MAVS. These data support a model in which MDA5 uses long dsRNA as a signaling platform to cooperatively assemble the core filament, which in turn promotes stochastic assembly of the tandem CARD oligomers for signaling.


Asunto(s)
ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/metabolismo , ARN Bicatenario/metabolismo , Secuencia de Aminoácidos , Humanos , Helicasa Inducida por Interferón IFIH1 , Modelos Moleculares , Datos de Secuencia Molecular , Estructura Terciaria de Proteína , ARN Bicatenario/química , Receptores de Ácido Retinoico/química , Receptores de Ácido Retinoico/metabolismo , Alineación de Secuencia , Difracción de Rayos X
9.
Mol Cell ; 77(4): 800-809.e6, 2020 02 20.
Artículo en Inglés | MEDLINE | ID: mdl-31810758

RESUMEN

The mammalian pre-mRNA 3'-end-processing machinery consists of cleavage and polyadenylation specificity factor (CPSF), cleavage stimulation factor (CstF), and other proteins, but the overall architecture of this machinery remains unclear. CPSF contains two functionally distinct modules: a cleavage factor (mCF) and a polyadenylation specificity factor (mPSF). Here, we have produced recombinant human CPSF and CstF and examined these factors by electron microscopy (EM). We find that mPSF is the organizational core of the machinery, while the conformations of mCF and CstF and the position of mCF relative to mPSF are highly variable. We have identified by cryo-EM a segment in CPSF100 that tethers mCF to mPSF, and we have named it the PSF interaction motif (PIM). Mutations in the PIM can abolish CPSF formation, indicating that it is a crucial contact in CPSF. We have also obtained reconstructions of mCF and CstF77 by cryo-EM, assembled around the mPSF core.


Asunto(s)
Factor de Especificidad de Desdoblamiento y Poliadenilación/química , Factor de Especificidad de Desdoblamiento y Poliadenilación/genética , Factor de Estimulación del Desdoblamiento/química , Microscopía por Crioelectrón , Humanos , Modelos Moleculares , Mutación , Procesamiento de Término de ARN 3' , Precursores del ARN/metabolismo , ARN Mensajero/metabolismo
10.
Mol Cell ; 80(1): 59-71.e4, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32818430

RESUMEN

Cardiac disease remains the leading cause of morbidity and mortality worldwide. The ß1-adrenergic receptor (ß1-AR) is a major regulator of cardiac functions and is downregulated in the majority of heart failure cases. A key physiological process is the activation of heterotrimeric G-protein Gs by ß1-ARs, leading to increased heart rate and contractility. Here, we use cryo-electron microscopy and functional studies to investigate the molecular mechanism by which ß1-AR activates Gs. We find that the tilting of α5-helix breaks a hydrogen bond between the sidechain of His373 in the C-terminal α5-helix and the backbone carbonyl of Arg38 in the N-terminal αN-helix of Gαs. Together with the disruption of another interacting network involving Gln59 in the α1-helix, Ala352 in the ß6-α5 loop, and Thr355 in the α5-helix, these conformational changes might lead to the deformation of the GDP-binding pocket. Our data provide molecular insights into the activation of G-proteins by G-protein-coupled receptors.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gs/química , Subunidades alfa de la Proteína de Unión al GTP Gs/metabolismo , Isoproterenol/metabolismo , Receptores Adrenérgicos beta 1/química , Receptores Adrenérgicos beta 1/metabolismo , Animales , Sitios de Unión , Bovinos , Línea Celular , Guanosina Difosfato/metabolismo , Guanosina Trifosfato/metabolismo , Modelos Moleculares , Unión Proteica , Dominios Proteicos , Estructura Secundaria de Proteína
11.
Cell ; 150(2): 339-50, 2012 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-22817896

RESUMEN

RIP1 and RIP3 kinases are central players in TNF-induced programmed necrosis. Here, we report that the RIP homotypic interaction motifs (RHIMs) of RIP1 and RIP3 mediate the assembly of heterodimeric filamentous structures. The fibrils exhibit classical characteristics of ß-amyloids, as shown by Thioflavin T (ThT) and Congo red (CR) binding, circular dichroism, infrared spectroscopy, X-ray diffraction, and solid-state NMR. Structured amyloid cores are mapped in RIP1 and RIP3 that are flanked by regions of mobility. The endogenous RIP1/RIP3 complex isolated from necrotic cells binds ThT, is ultrastable, and has a fibrillar core structure, whereas necrosis is partially inhibited by ThT, CR, and another amyloid dye, HBX. Mutations in the RHIMs of RIP1 and RIP3 that are defective in the interaction compromise cluster formation, kinase activation, and programmed necrosis in vivo. The current study provides insight into the structural changes that occur when RIP kinases are triggered to execute different signaling outcomes and expands the realm of amyloids to complex formation and signaling.


Asunto(s)
Necrosis/metabolismo , Proteína Serina-Treonina Quinasas de Interacción con Receptores/metabolismo , Transducción de Señal , Secuencia de Aminoácidos , Amiloide/química , Humanos , Datos de Secuencia Molecular , Dominios y Motivos de Interacción de Proteínas , Proteína Serina-Treonina Quinasas de Interacción con Receptores/química , Alineación de Secuencia
12.
Nature ; 590(7846): 509-514, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33568813

RESUMEN

Mechanosensitive channels sense mechanical forces in cell membranes and underlie many biological sensing processes1-3. However, how exactly they sense mechanical force remains under investigation4. The bacterial mechanosensitive channel of small conductance, MscS, is one of the most extensively studied mechanosensitive channels4-8, but how it is regulated by membrane tension remains unclear, even though the structures are known for its open and closed states9-11. Here we used cryo-electron microscopy to determine the structure of MscS in different membrane environments, including one that mimics a membrane under tension. We present the structures of MscS in the subconducting and desensitized states, and demonstrate that the conformation of MscS in a lipid bilayer in the open state is dynamic. Several associated lipids have distinct roles in MscS mechanosensation. Pore lipids are necessary to prevent ion conduction in the closed state. Gatekeeper lipids stabilize the closed conformation and dissociate with membrane tension, allowing the channel to open. Pocket lipids in a solvent-exposed pocket between subunits are pulled out under sustained tension, allowing the channel to transition to the subconducting state and then to the desensitized state. Our results provide a mechanistic underpinning and expand on the 'force-from-lipids' model for MscS mechanosensation4,11.


Asunto(s)
Microscopía por Crioelectrón , Proteínas de Escherichia coli/metabolismo , Proteínas de Escherichia coli/ultraestructura , Escherichia coli/química , Canales Iónicos/metabolismo , Canales Iónicos/ultraestructura , Membranas Artificiales , Fosfatidilcolinas/metabolismo , Detergentes/farmacología , Escherichia coli/ultraestructura , Proteínas de Escherichia coli/química , Proteínas de Escherichia coli/genética , Interacciones Hidrofóbicas e Hidrofílicas , Canales Iónicos/química , Canales Iónicos/genética , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Mecanotransducción Celular/efectos de los fármacos , Modelos Moleculares , Mutación , Nanoestructuras/química , Nanoestructuras/ultraestructura , Fosfatidilcolinas/química , Fosfatidilcolinas/farmacología , Conformación Proteica/efectos de los fármacos , beta-Ciclodextrinas/farmacología
13.
Trends Biochem Sci ; 47(7): 561-569, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35331611

RESUMEN

Advances in cryogenic electron microscopy (cryo-EM) enabled routine near-atomic structure determination of membrane proteins, while nanodisc technology has provided a way to provide membrane proteins with a native or native-like lipid environment. After giving a brief history of membrane mimetics, we present example structures of membrane proteins in nanodiscs that revealed information not provided by structures obtained in detergent. We describe how the lipid environment surrounding the membrane protein can be custom designed during nanodisc assembly and how it can be modified after assembly to test functional hypotheses. Because nanodiscs most closely replicate the physiologic environment of membrane proteins and often afford novel mechanistic insights, we propose that nanodiscs ought to become the standard for structural studies on membrane proteins.


Asunto(s)
Proteínas de la Membrana , Nanoestructuras , Membrana Dobles de Lípidos/química , Lípidos , Proteínas de la Membrana/metabolismo , Microscopía Electrónica , Modelos Moleculares , Nanoestructuras/química
14.
Cell ; 142(4): 556-67, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20723757

RESUMEN

The monopolin complex regulates different types of kinetochore-microtubule attachments in fungi, ensuring sister chromatid co-orientation in Saccharomyces cerevisiae meiosis I and inhibiting merotelic attachment in Schizosaccharomyces pombe mitosis. In addition, the monopolin complex maintains the integrity and silencing of ribosomal DNA (rDNA) repeats in the nucleolus. We show here that the S. cerevisiae Csm1/Lrs4 monopolin subcomplex has a distinctive V-shaped structure, with two pairs of protein-protein interaction domains positioned approximately 10 nm apart. Csm1 presents a conserved hydrophobic surface patch that binds two kinetochore proteins: Dsn1, a subunit of the outer-kinetochore MIND/Mis12 complex, and Mif2/CENP-C. Csm1 point-mutations that disrupt kinetochore-subunit binding also disrupt sister chromatid co-orientation in S. cerevisiae meiosis I. We further show that the same Csm1 point-mutations affect rDNA silencing, probably by disrupting binding to the rDNA-associated protein Tof2. We propose that Csm1/Lrs4 functions as a molecular clamp, crosslinking kinetochore components to enforce sister chromatid co-orientation in S. cerevisiae meiosis I and to suppress merotelic attachment in S. pombe mitosis, and crosslinking rDNA repeats to aid rDNA silencing.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromosomas Fúngicos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Schizosaccharomyces/metabolismo , Cinetocoros/metabolismo , Meiosis , Mitosis , Modelos Moleculares , Mutación Puntual , Saccharomyces cerevisiae/citología , Saccharomyces cerevisiae/genética , Schizosaccharomyces/citología , Schizosaccharomyces/genética
15.
Mol Cell ; 67(5): 770-782.e6, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28886335

RESUMEN

The mammalian circadian clock is built on a feedback loop in which PER and CRY proteins repress their own transcription. We found that in mouse liver nuclei all three PERs, both CRYs, and Casein Kinase-1δ (CK1δ) are present together in an ∼1.9-MDa repressor assembly that quantitatively incorporates its CLOCK-BMAL1 transcription factor target. Prior to incorporation, CLOCK-BMAL1 exists in an ∼750-kDa complex. Single-particle electron microscopy (EM) revealed nuclear PER complexes purified from mouse liver to be quasi-spherical ∼40-nm structures. In the cytoplasm, PERs, CRYs, and CK1δ were distributed into several complexes of ∼0.9-1.1 MDa that appear to constitute an assembly pathway regulated by GAPVD1, a cytoplasmic trafficking factor. Single-particle EM of two purified cytoplasmic PER complexes revealed ∼20-nm and ∼25-nm structures, respectively, characterized by flexibly tethered globular domains. Our results define the macromolecular assemblies comprising the circadian feedback loop and provide an initial structural view of endogenous eukaryotic clock machinery.


Asunto(s)
Núcleo Celular/metabolismo , Relojes Circadianos , Péptidos y Proteínas de Señalización del Ritmo Circadiano/metabolismo , Ritmo Circadiano , Factores de Transcripción ARNTL/genética , Factores de Transcripción ARNTL/metabolismo , Animales , Quinasa Idelta de la Caseína/metabolismo , Línea Celular , Núcleo Celular/ultraestructura , Péptidos y Proteínas de Señalización del Ritmo Circadiano/deficiencia , Péptidos y Proteínas de Señalización del Ritmo Circadiano/genética , Criptocromos/genética , Criptocromos/metabolismo , Femenino , Genotipo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Electrónica , Complejos Multiproteicos , Tamaño de la Partícula , Proteínas Circadianas Period/genética , Proteínas Circadianas Period/metabolismo , Fenotipo , Interferencia de ARN , Transducción de Señal , Imagen Individual de Molécula , Factores de Tiempo , Transfección
16.
Mol Cell ; 68(2): 388-397.e6, 2017 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-28988932

RESUMEN

Noncoding RNAs (ncRNAs) regulate gene expression in all organisms. Bacterial 6S RNAs globally regulate transcription by binding RNA polymerase (RNAP) holoenzyme and competing with promoter DNA. Escherichia coli (Eco) 6S RNA interacts specifically with the housekeeping σ70-holoenzyme (Eσ70) and plays a key role in the transcriptional reprogramming upon shifts between exponential and stationary phase. Inhibition is relieved upon 6S RNA-templated RNA synthesis. We report here the 3.8 Å resolution structure of a complex between 6S RNA and Eσ70 determined by single-particle cryo-electron microscopy and validation of the structure using footprinting and crosslinking approaches. Duplex RNA segments have A-form C3' endo sugar puckers but widened major groove widths, giving the RNA an overall architecture that mimics B-form promoter DNA. Our results help explain the specificity of Eco 6S RNA for Eσ70 and show how an ncRNA can mimic B-form DNA to directly regulate transcription by the DNA-dependent RNAP.


Asunto(s)
ADN Forma B/metabolismo , ADN Bacteriano/metabolismo , ARN Polimerasas Dirigidas por ADN/metabolismo , Proteínas de Escherichia coli/metabolismo , Escherichia coli/metabolismo , ARN Bacteriano/metabolismo , ARN no Traducido/metabolismo , Factor sigma/metabolismo , ADN Forma B/genética , ADN Bacteriano/genética , ARN Polimerasas Dirigidas por ADN/genética , Escherichia coli/genética , Proteínas de Escherichia coli/genética , ARN Bacteriano/genética , ARN no Traducido/genética , Factor sigma/genética
17.
Proc Natl Acad Sci U S A ; 119(31): e2201662119, 2022 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-35881804

RESUMEN

Human shelterin is a six-subunit complex-composed of TRF1, TRF2, Rap1, TIN2, TPP1, and POT1-that binds telomeres, protects them from the DNA-damage response, and regulates the maintenance of telomeric DNA. Although high-resolution structures have been generated of the individual structured domains within shelterin, the architecture and stoichiometry of the full complex are currently unknown. Here, we report the purification of shelterin subcomplexes and reconstitution of the entire complex using full-length, recombinant subunits. By combining negative-stain electron microscopy (EM), cross-linking mass spectrometry (XLMS), AlphaFold modeling, mass photometry, and native mass spectrometry (MS), we obtain stoichiometries as well as domain-scale architectures of shelterin subcomplexes and determine that they feature extensive conformational heterogeneity. For POT1/TPP1 and POT1/TPP1/TIN2, we observe high variability in the positioning of the POT1 DNA-binding domain, the TPP1 oligonucleotide/oligosaccharide-binding (OB) fold, and the TIN2 TRFH domain with respect to the C-terminal domains of POT1. Truncation of unstructured linker regions in TIN2, TPP1, and POT1 did not reduce the conformational variability of the heterotrimer. Shelterin and TRF1-containing subcomplexes form fully dimeric stoichiometries, even in the absence of DNA substrates. Shelterin and its subcomplexes showed extensive conformational variability, regardless of the presence of DNA substrates. We conclude that shelterin adopts a multitude of conformations and argue that its unusual architectural variability is beneficial for its many functions at telomeres.


Asunto(s)
Complejo Shelterina , Humanos , Espectrometría de Masas , Microscopía Electrónica , Dominios Proteicos , Complejo Shelterina/química
18.
Annu Rev Biochem ; 78: 723-42, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19489732

RESUMEN

Single-particle electron microscopy (EM) can provide structural information for a large variety of biological molecules, ranging from small proteins to large macromolecular assemblies, without the need to produce crystals. The year 2008 has become a landmark year for single-particle EM as for the first time density maps have been produced at a resolution that made it possible to trace protein backbones or even to build atomic models. In this review, we highlight some of the recent successes achieved by single-particle EM and describe the individual steps involved in producing a density map by this technique. We also discuss some of the remaining challenges and areas, in which further advances would have a great impact on the results that can be achieved by single-particle EM.


Asunto(s)
Microscopía Electrónica/métodos , Humanos , Microscopía Electrónica/instrumentación , Complejos Multiproteicos/ultraestructura , Proteínas/ultraestructura , Virus/ultraestructura
19.
Cell ; 139(6): 1119-29, 2009 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-20005805

RESUMEN

Vesicle trafficking requires membrane fusion, mediated by SNARE proteins, and upstream events that probably include "tethering," an initial long-range attachment between a vesicle and its target organelle. Among the factors proposed to mediate tethering are a set of multisubunit tethering complexes (MTCs). The Dsl1 complex, with only three subunits, is the simplest known MTC and is essential for the retrograde traffic of COPI-coated vesicles from the Golgi to the ER. To elucidate structural principles underlying MTC function, we have determined the structure of the Dsl1 complex, revealing a tower containing at its base the binding sites for two ER SNAREs and at its tip a flexible lasso for capturing vesicles. The Dsl1 complex binds to individual SNAREs via their N-terminal regulatory domains and also to assembled SNARE complexes; moreover, it is capable of accelerating SNARE complex assembly. Our results suggest that even the simplest MTC may be capable of orchestrating vesicle capture, uncoating, and fusion.


Asunto(s)
Proteínas de Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/química , Vesículas Transportadoras/metabolismo , Cristalografía por Rayos X , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas SNARE/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo
20.
Cell ; 136(5): 876-90, 2009 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-19249086

RESUMEN

Store-operated Ca(2+) channels activated by the depletion of Ca(2+) from the endoplasmic reticulum (ER) are a major Ca(2+) entry pathway in nonexcitable cells and are essential for T cell activation and adaptive immunity. After store depletion, the ER Ca(2+) sensor STIM1 and the CRAC channel protein Orai1 redistribute to ER-plasma membrane (PM) junctions, but the fundamental issue of how STIM1 activates the CRAC channel at these sites is unresolved. Here, we identify a minimal, highly conserved 107-aa CRAC activation domain (CAD) of STIM1 that binds directly to the N and C termini of Orai1 to open the CRAC channel. Purified CAD forms a tetramer that clusters CRAC channels, but analysis of STIM1 mutants reveals that channel clustering is not sufficient for channel activation. These studies establish a molecular mechanism for store-operated Ca(2+) entry in which the direct binding of STIM1 to Orai1 drives the accumulation and the activation of CRAC channels at ER-PM junctions.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Proteínas de la Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Canales de Calcio/química , Línea Celular , Membrana Celular/metabolismo , Retículo Endoplásmico/metabolismo , Humanos , Proteína ORAI1 , Estructura Terciaria de Proteína , Molécula de Interacción Estromal 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA