Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 183
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 160(5): 952-962, 2015 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-25723169

RESUMEN

Bacteria use rapid contraction of a long sheath of the type VI secretion system (T6SS) to deliver effectors into a target cell. Here, we present an atomic-resolution structure of a native contracted Vibrio cholerae sheath determined by cryo-electron microscopy. The sheath subunits, composed of tightly interacting proteins VipA and VipB, assemble into a six-start helix. The helix is stabilized by a core domain assembled from four ß strands donated by one VipA and two VipB molecules. The fold of inner and middle layers is conserved between T6SS and phage sheaths. However, the structure of the outer layer is distinct and suggests a mechanism of interaction of the bacterial sheath with an accessory ATPase, ClpV, that facilitates multiple rounds of effector delivery. Our results provide a mechanistic insight into assembly of contractile nanomachines that bacteria and phages use to translocate macromolecules across membranes.


Asunto(s)
Proteínas Bacterianas/química , Sistemas de Secreción Bacterianos , Vibrio cholerae/metabolismo , Secuencia de Aminoácidos , Microscopía por Crioelectrón , Modelos Moleculares , Datos de Secuencia Molecular , Alineación de Secuencia , Vibrio cholerae/química , Vibrio cholerae/citología , Vibrio cholerae/ultraestructura
2.
Mol Cell ; 82(15): 2754-2768.e5, 2022 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-35835111

RESUMEN

Type I CRISPR-Cas systems typically rely on a two-step process to degrade DNA. First, an RNA-guided complex named Cascade identifies the complementary DNA target. The helicase-nuclease fusion enzyme Cas3 is then recruited in trans for processive DNA degradation. Contrary to this model, here, we show that type I-A Cascade and Cas3 function as an integral effector complex. We provide four cryoelectron microscopy (cryo-EM) snapshots of the Pyrococcus furiosus (Pfu) type I-A effector complex in different stages of DNA recognition and degradation. The HD nuclease of Cas3 is autoinhibited inside the effector complex. It is only allosterically activated upon full R-loop formation, when the entire targeted region has been validated by the RNA guide. The mechanistic insights inspired us to convert Pfu Cascade-Cas3 into a high-sensitivity, low-background, and temperature-activated nucleic acid detection tool. Moreover, Pfu CRISPR-Cas3 shows robust bi-directional deletion-editing activity in human cells, which could find usage in allele-specific inactivation of disease-causing mutations.


Asunto(s)
Proteínas Asociadas a CRISPR , Proteínas Asociadas a CRISPR/genética , Proteínas Asociadas a CRISPR/metabolismo , Sistemas CRISPR-Cas , Microscopía por Crioelectrón , ADN/genética , ADN/metabolismo , Endonucleasas/genética , Edición Génica , Humanos , ARN
3.
Nature ; 617(7959): 176-184, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37100904

RESUMEN

Physical interactions between proteins are essential for most biological processes governing life1. However, the molecular determinants of such interactions have been challenging to understand, even as genomic, proteomic and structural data increase. This knowledge gap has been a major obstacle for the comprehensive understanding of cellular protein-protein interaction networks and for the de novo design of protein binders that are crucial for synthetic biology and translational applications2-9. Here we use a geometric deep-learning framework operating on protein surfaces that generates fingerprints to describe geometric and chemical features that are critical to drive protein-protein interactions10. We hypothesized that these fingerprints capture the key aspects of molecular recognition that represent a new paradigm in the computational design of novel protein interactions. As a proof of principle, we computationally designed several de novo protein binders to engage four protein targets: SARS-CoV-2 spike, PD-1, PD-L1 and CTLA-4. Several designs were experimentally optimized, whereas others were generated purely in silico, reaching nanomolar affinity with structural and mutational characterization showing highly accurate predictions. Overall, our surface-centric approach captures the physical and chemical determinants of molecular recognition, enabling an approach for the de novo design of protein interactions and, more broadly, of artificial proteins with function.


Asunto(s)
Simulación por Computador , Aprendizaje Profundo , Unión Proteica , Proteínas , Humanos , Proteínas/química , Proteínas/metabolismo , Proteómica , Mapas de Interacción de Proteínas , Sitios de Unión , Biología Sintética
4.
PLoS Pathog ; 19(4): e1011206, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37018380

RESUMEN

Investigation of potential hosts of the severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is crucial to understanding future risks of spillover and spillback. SARS-CoV-2 has been reported to be transmitted from humans to various animals after requiring relatively few mutations. There is significant interest in describing how the virus interacts with mice as they are well adapted to human environments, are used widely as infection models and can be infected. Structural and binding data of the mouse ACE2 receptor with the Spike protein of newly identified SARS-CoV-2 variants are needed to better understand the impact of immune system evading mutations present in variants of concern (VOC). Previous studies have developed mouse-adapted variants and identified residues critical for binding to heterologous ACE2 receptors. Here we report the cryo-EM structures of mouse ACE2 bound to trimeric Spike ectodomains of four different VOC: Beta, Omicron BA.1, Omicron BA.2.12.1 and Omicron BA.4/5. These variants represent the oldest to the newest variants known to bind the mouse ACE2 receptor. Our high-resolution structural data complemented with bio-layer interferometry (BLI) binding assays reveal a requirement for a combination of mutations in the Spike protein that enable binding to the mouse ACE2 receptor.


Asunto(s)
COVID-19 , SARS-CoV-2 , Animales , Humanos , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/virología , Microscopía por Crioelectrón , Especificidad del Huésped , Mutación , Unión Proteica , SARS-CoV-2/genética , Glicoproteína de la Espiga del Coronavirus/genética
5.
Brain ; 2024 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-38696728

RESUMEN

Multiple System Atrophy is characterized pathologically by the accumulation of alpha-synuclein (aSyn) into glial cytoplasmic inclusions (GCIs). The mechanism underlying the formation of GCIs is not well understood. In this study, correlative light and electron microscopy was employed to investigate aSyn pathology in the substantia nigra and putamen of post-mortem multiple system atrophy brain donors. Three distinct types of aSyn immuno-positive inclusions were identified in oligodendrocytes, neurons and dark cells presumed to be dark microglia. Oligodendrocytes contained fibrillar GCIs that were consistently enriched with lysosomes and peroxisomes, supporting the involvement of the autophagy pathway in aSyn aggregation in multiple system atrophy. Neuronal cytoplasmic inclusions exhibited ultrastructural heterogeneity resembling both fibrillar and membranous inclusions, linking multiple systems atrophy and Parkinson's disease. The novel aSyn pathology identified in the dark cells, displayed GCI-like fibrils or non-GCI-like ultrastructures suggesting various stages of aSyn accumulation in these cells. The observation of GCI-like fibrils within dark cells suggests these cells may be an important contributor to the origin or spread of pathological aSyn in multiple system atrophy. Our results suggest a complex interplay between multiple cell types that may underlie the formation of aSyn pathology in multiple system atrophy brain and highlight the need for further investigation into cell-specific disease pathologies in multiple system atrophy.

6.
EMBO J ; 39(22): e106246, 2020 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-32954513

RESUMEN

Centrioles are evolutionarily conserved barrels of microtubule triplets that form the core of the centrosome and the base of the cilium. While the crucial role of the proximal region in centriole biogenesis has been well documented, its native architecture and evolutionary conservation remain relatively unexplored. Here, using cryo-electron tomography of centrioles from four evolutionarily distant species, we report on the architectural diversity of the centriole's proximal cartwheel-bearing region. Our work reveals that the cartwheel central hub is constructed from a stack of paired rings with cartwheel inner densities inside. In both Paramecium and Chlamydomonas, the repeating structural unit of the cartwheel has a periodicity of 25 nm and consists of three ring pairs, with 6 radial spokes emanating and merging into a single bundle that connects to the microtubule triplet via the D2-rod and the pinhead. Finally, we identified that the cartwheel is indirectly connected to the A-C linker through the triplet base structure extending from the pinhead. Together, our work provides unprecedented evolutionary insights into the architecture of the centriole proximal region, which underlies centriole biogenesis.


Asunto(s)
Centriolos/fisiología , Centriolos/ultraestructura , Tomografía con Microscopio Electrónico/métodos , Centrosoma , Chlamydomonas reinhardtii/fisiología , Cilios , Humanos , Microtúbulos , Modelos Moleculares , Naegleria/fisiología , Paramecium tetraurelia/fisiología
7.
EMBO J ; 39(19): e104743, 2020 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-32779739

RESUMEN

Focal adhesion kinase (FAK) is a key component of the membrane proximal signaling layer in focal adhesion complexes, regulating important cellular processes, including cell migration, proliferation, and survival. In the cytosol, FAK adopts an autoinhibited state but is activated upon recruitment into focal adhesions, yet how this occurs or what induces structural changes is unknown. Here, we employ cryo-electron microscopy to reveal how FAK associates with lipid membranes and how membrane interactions unlock FAK autoinhibition to promote activation. Intriguingly, initial binding of FAK to the membrane causes steric clashes that release the kinase domain from autoinhibition, allowing it to undergo a large conformational change and interact itself with the membrane in an orientation that places the active site toward the membrane. In this conformation, the autophosphorylation site is exposed and multiple interfaces align to promote FAK oligomerization on the membrane. We show that interfaces responsible for initial dimerization and membrane attachment are essential for FAK autophosphorylation and resulting cellular activity including cancer cell invasion, while stable FAK oligomerization appears to be needed for optimal cancer cell proliferation in an anchorage-independent manner. Together, our data provide structural details of a key membrane bound state of FAK that is primed for efficient autophosphorylation and activation, hence revealing the critical event in integrin mediated FAK activation and signaling at focal adhesions.


Asunto(s)
Proteínas Aviares/química , Proteína-Tirosina Quinasas de Adhesión Focal/química , Membranas/química , Multimerización de Proteína , Animales , Proteínas Aviares/metabolismo , Pollos , Activación Enzimática , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Células HEK293 , Humanos , Membranas/enzimología , Relación Estructura-Actividad
8.
Nature ; 558(7710): 470-474, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29899443

RESUMEN

Acetyl-CoA carboxylase catalyses the ATP-dependent carboxylation of acetyl-CoA, a rate-limiting step in fatty acid biosynthesis1,2. Eukaryotic acetyl-CoA carboxylases are large, homodimeric multienzymes. Human acetyl-CoA carboxylase occurs in two isoforms: the metabolic, cytosolic ACC1, and ACC2, which is anchored to the outer mitochondrial membrane and controls fatty acid ß-oxidation1,3. ACC1 is regulated by a complex interplay of phosphorylation, binding of allosteric regulators and protein-protein interactions, which is further linked to filament formation1,4-8. These filaments were discovered in vitro and in vivo 50 years ago7,9,10, but the structural basis of ACC1 polymerization and regulation remains unknown. Here, we identify distinct activated and inhibited ACC1 filament forms. We obtained cryo-electron microscopy structures of an activated filament that is allosterically induced by citrate (ACC-citrate), and an inactivated filament form that results from binding of the BRCT domains of the breast cancer type 1 susceptibility protein (BRCA1). While non-polymeric ACC1 is highly dynamic, filament formation locks ACC1 into different catalytically competent or incompetent conformational states. This unique mechanism of enzyme regulation via large-scale conformational changes observed in ACC1 has potential uses in engineering of switchable biosynthetic systems. Dissecting the regulation of acetyl-CoA carboxylase opens new paths towards counteracting upregulation of fatty acid biosynthesis in disease.


Asunto(s)
Acetil-CoA Carboxilasa/química , Acetil-CoA Carboxilasa/ultraestructura , Microscopía por Crioelectrón , Acetil-CoA Carboxilasa/metabolismo , Animales , Proteína BRCA1/química , Proteína BRCA1/farmacología , Biopolímeros/química , Biopolímeros/metabolismo , Línea Celular , Ácido Cítrico/farmacología , Humanos , Modelos Moleculares , Polimerizacion/efectos de los fármacos , Dominios Proteicos/efectos de los fármacos , Estructura Cuaternaria de Proteína/efectos de los fármacos , Spodoptera , Relación Estructura-Actividad
9.
Nature ; 563(7731): 426-430, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30405239

RESUMEN

ABCG2 is a transporter protein of the ATP-binding-cassette (ABC) family that is expressed in the plasma membrane in cells of various tissues and tissue barriers, including the blood-brain, blood-testis and maternal-fetal barriers1-4. Powered by ATP, it translocates endogenous substrates, affects the pharmacokinetics of many drugs and protects against a wide array of xenobiotics, including anti-cancer drugs5-12. Previous studies have revealed the architecture of ABCG2 and the structural basis of its inhibition by small molecules and antibodies13,14. However, the mechanisms of substrate recognition and ATP-driven transport are unknown. Here we present high-resolution cryo-electron microscopy (cryo-EM) structures of human ABCG2 in a substrate-bound pre-translocation state and an ATP-bound post-translocation state. For both structures, we used a mutant containing a glutamine replacing the catalytic glutamate (ABCG2EQ), which resulted in reduced ATPase and transport rates and facilitated conformational trapping for structural studies. In the substrate-bound state, a single molecule of estrone-3-sulfate (E1S) is bound in a central, hydrophobic and cytoplasm-facing cavity about halfway across the membrane. Only one molecule of E1S can bind in the observed binding mode. In the ATP-bound state, the substrate-binding cavity has collapsed while an external cavity has opened to the extracellular side of the membrane. The ATP-induced conformational changes include rigid-body shifts of the transmembrane domains, pivoting of the nucleotide-binding domains (NBDs), and a change in the relative orientation of the NBD subdomains. Mutagenesis and in vitro characterization of transport and ATPase activities demonstrate the roles of specific residues in substrate recognition, including a leucine residue that forms a 'plug' between the two cavities. Our results show how ABCG2 harnesses the energy of ATP binding to extrude E1S and other substrates, and suggest that the size and binding affinity of compounds are important for distinguishing substrates from inhibitors.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/ultraestructura , Adenosina Trifosfato/metabolismo , Microscopía por Crioelectrón , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestructura , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/ultraestructura , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Sitios de Unión , Humanos , Modelos Moleculares , Proteínas Mutantes/química , Proteínas Mutantes/genética , Mutación , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Unión Proteica , Conformación Proteica , Especificidad por Sustrato
10.
EMBO J ; 38(18): e100825, 2019 09 16.
Artículo en Inglés | MEDLINE | ID: mdl-31403721

RESUMEN

Protein translocation by the bacterial type VI secretion system (T6SS) is driven by a rapid contraction of a sheath assembled around a tube with associated effectors. Here, we show that TssA-like or TagA-like proteins with a conserved N-terminal domain and varying C-terminal domains can be grouped into at least three distinct classes based on their role in sheath assembly. The proteins of the first class increase speed and frequency of sheath assembly and form a stable dodecamer at the distal end of a polymerizing sheath. The proteins of the second class localize to the cell membrane and block sheath polymerization upon extension across the cell. This prevents excessive sheath polymerization and bending, which may result in sheath destabilization and detachment from its membrane anchor and thus result in failed secretion. The third class of these proteins localizes to the baseplate and is required for initiation of sheath assembly. Our work shows that while various proteins share a conserved N-terminal domain, their roles in T6SS biogenesis are fundamentally different.


Asunto(s)
Proteínas Bacterianas/metabolismo , Bacterias Gramnegativas/metabolismo , Lipoproteínas/metabolismo , Sistemas de Secreción Tipo VI/metabolismo , Proteínas Bacterianas/química , Membrana Celular/metabolismo , Lipoproteínas/química , Modelos Moleculares , Conformación Proteica , Dominios Proteicos
11.
EMBO Rep ; 22(12): e53877, 2021 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-34806807

RESUMEN

Morphologically distinct TDP-43 aggregates occur in clinically different FTLD-TDP subtypes, yet the mechanism of their emergence and contribution to clinical heterogeneity are poorly understood. Several lines of evidence suggest that pathological TDP-43 follows a prion-like cascade, but the molecular determinants of this process remain unknown. We use advanced microscopy techniques to compare the seeding properties of pathological FTLD-TDP-A and FTLD-TDP-C aggregates. Upon inoculation of patient-derived aggregates in cells, FTLD-TDP-A seeds amplify in a template-dependent fashion, triggering neoaggregation more efficiently than those extracted from FTLD-TDP-C patients, correlating with the respective disease progression rates. Neoaggregates are sequentially phosphorylated with N-to-C directionality and with subtype-specific timelines. The resulting FTLD-TDP-A neoaggregates are large and contain densely packed fibrils, reminiscent of the pure compacted fibrils present within cytoplasmic inclusions in postmortem brains. In contrast, FTLD-TDP-C dystrophic neurites show less dense fibrils mixed with cellular components, and their respective neoaggregates are small, amorphous protein accumulations. These cellular seeding models replicate aspects of the patient pathological diversity and will be a useful tool in the quest for subtype-specific therapeutics.


Asunto(s)
Demencia Frontotemporal , Priones , Encéfalo/metabolismo , Demencia Frontotemporal/metabolismo , Humanos , Cuerpos de Inclusión/metabolismo , Priones/metabolismo
12.
Nature ; 546(7659): 504-509, 2017 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-28554189

RESUMEN

ABCG2 is a constitutively expressed ATP-binding cassette (ABC) transporter that protects many tissues against xenobiotic molecules. Its activity affects the pharmacokinetics of commonly used drugs and limits the delivery of therapeutics into tumour cells, thus contributing to multidrug resistance. Here we present the structure of human ABCG2 determined by cryo-electron microscopy, providing the first high-resolution insight into a human multidrug transporter. We visualize ABCG2 in complex with two antigen-binding fragments of the human-specific, inhibitory antibody 5D3 that recognizes extracellular loops of the transporter. We observe two cholesterol molecules bound in the multidrug-binding pocket that is located in a central, hydrophobic, inward-facing translocation pathway between the transmembrane domains. Combined with functional in vitro analyses, our results suggest a multidrug recognition and transport mechanism of ABCG2, rationalize disease-causing single nucleotide polymorphisms and the allosteric inhibition by the 5D3 antibody, and provide the structural basis of cholesterol recognition by other G-subfamily ABC transporters.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/química , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/ultraestructura , Microscopía por Crioelectrón , Proteínas de Neoplasias/química , Proteínas de Neoplasias/ultraestructura , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/antagonistas & inhibidores , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Adenosina Trifosfatasas/química , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Adenosina Trifosfatasas/ultraestructura , Secuencia de Aminoácidos , Anticuerpos/química , Anticuerpos/inmunología , Anticuerpos/ultraestructura , Sitios de Unión , Transporte Biológico , Colesterol/química , Colesterol/metabolismo , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Fragmentos Fab de Inmunoglobulinas/inmunología , Fragmentos Fab de Inmunoglobulinas/ultraestructura , Modelos Moleculares , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Polimorfismo de Nucleótido Simple/genética , Dominios Proteicos
13.
EMBO J ; 37(4)2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29255010

RESUMEN

The bacterial Type VI secretion system (T6SS) assembles from three major parts: a membrane complex that spans inner and outer membranes, a baseplate, and a sheath-tube polymer. The baseplate assembles around a tip complex with associated effectors and connects to the membrane complex by TssK. The baseplate assembly initiates sheath-tube polymerization, which in some organisms requires TssA. Here, we analyzed both ends of isolated non-contractile Vibrio cholerae sheaths by cryo-electron microscopy. Our analysis suggests that the baseplate, solved to an average 8.0 Å resolution, is composed of six subunits of TssE/F2/G and the baseplate periphery is decorated by six TssK trimers. The VgrG/PAAR tip complex in the center of the baseplate is surrounded by a cavity, which may accommodate up to ~450 kDa of effector proteins. The distal end of the sheath, resolved to an average 7.5 Å resolution, shows sixfold symmetry; however, its protein composition is unclear. Our structures provide an important step toward an atomic model of the complete T6SS assembly.


Asunto(s)
Proteínas Bacterianas/química , Microscopía por Crioelectrón/métodos , Proteínas de la Membrana/química , Sistemas de Secreción Tipo VI/ultraestructura , Vibrio cholerae/ultraestructura , Vibrio cholerae/citología , Vibrio cholerae/metabolismo
14.
15.
Nature ; 533(7603): 346-52, 2016 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-27193680

RESUMEN

Several systems, including contractile tail bacteriophages, the type VI secretion system and R-type pyocins, use a multiprotein tubular apparatus to attach to and penetrate host cell membranes. This macromolecular machine resembles a stretched, coiled spring (or sheath) wound around a rigid tube with a spike-shaped protein at its tip. A baseplate structure, which is arguably the most complex part of this assembly, relays the contraction signal to the sheath. Here we present the atomic structure of the approximately 6-megadalton bacteriophage T4 baseplate in its pre- and post-host attachment states and explain the events that lead to sheath contraction in atomic detail. We establish the identity and function of a minimal set of components that is conserved in all contractile injection systems and show that the triggering mechanism is universally conserved.


Asunto(s)
Bacteriófago T4/química , Bacteriófago T4/ultraestructura , Proteínas Estructurales Virales/química , Proteínas Estructurales Virales/ultraestructura , Microscopía por Crioelectrón , Cristalografía por Rayos X , Modelos Moleculares , Conformación Proteica
16.
Nature ; 531(7596): 598-603, 2016 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-27029275

RESUMEN

The cullin-RING ubiquitin E3 ligase (CRL) family comprises over 200 members in humans. The COP9 signalosome complex (CSN) regulates CRLs by removing their ubiquitin-like activator NEDD8. The CUL4A-RBX1-DDB1-DDB2 complex (CRL4A(DDB2)) monitors the genome for ultraviolet-light-induced DNA damage. CRL4A(DBB2) is inactive in the absence of damaged DNA and requires CSN to regulate the repair process. The structural basis of CSN binding to CRL4A(DDB2) and the principles of CSN activation are poorly understood. Here we present cryo-electron microscopy structures for CSN in complex with neddylated CRL4A ligases to 6.4 Å resolution. The CSN conformers defined by cryo-electron microscopy and a novel apo-CSN crystal structure indicate an induced-fit mechanism that drives CSN activation by neddylated CRLs. We find that CSN and a substrate cannot bind simultaneously to CRL4A, favouring a deneddylated, inactive state for substrate-free CRL4 complexes. These architectural and regulatory principles appear conserved across CRL families, allowing global regulation by CSN.


Asunto(s)
Biocatálisis , Complejos Multiproteicos/metabolismo , Complejos Multiproteicos/ultraestructura , Péptido Hidrolasas/metabolismo , Péptido Hidrolasas/ultraestructura , Regulación Alostérica , Apoproteínas/química , Apoproteínas/metabolismo , Apoproteínas/ultraestructura , Sitios de Unión , Complejo del Señalosoma COP9 , Proteínas Portadoras/química , Proteínas Portadoras/metabolismo , Proteínas Portadoras/ultraestructura , Microscopía por Crioelectrón , Cristalografía por Rayos X , Proteínas Cullin/química , Proteínas Cullin/metabolismo , Proteínas Cullin/ultraestructura , Daño del ADN , Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Proteínas de Unión al ADN/ultraestructura , Humanos , Cinética , Modelos Moleculares , Complejos Multiproteicos/química , Péptido Hidrolasas/química , Unión Proteica , Ubiquitinación , Ubiquitinas/metabolismo
17.
Proc Natl Acad Sci U S A ; 116(30): 15007-15012, 2019 07 23.
Artículo en Inglés | MEDLINE | ID: mdl-31292253

RESUMEN

High-resolution structural information is essential to understand protein function. Protein-structure determination needs a considerable amount of protein, which can be challenging to produce, often involving harsh and lengthy procedures. In contrast, the several thousand to a few million protein particles required for structure determination by cryogenic electron microscopy (cryo-EM) can be provided by miniaturized systems. Here, we present a microfluidic method for the rapid isolation of a target protein and its direct preparation for cryo-EM. Less than 1 µL of cell lysate is required as starting material to solve the atomic structure of the untagged, endogenous human 20S proteasome. Our work paves the way for high-throughput structure determination of proteins from minimal amounts of cell lysate and opens more opportunities for the isolation of sensitive, endogenous protein complexes.


Asunto(s)
Microscopía por Crioelectrón/métodos , Procesamiento de Imagen Asistido por Computador/estadística & datos numéricos , Complejo de la Endopetidasa Proteasomal/ultraestructura , Subunidades de Proteína/química , Biotinilación , Microscopía por Crioelectrón/instrumentación , Células HeLa , Humanos , Imagenología Tridimensional , Fragmentos Fab de Inmunoglobulinas/química , Técnicas Analíticas Microfluídicas/métodos , Complejo de la Endopetidasa Proteasomal/química , Complejo de la Endopetidasa Proteasomal/aislamiento & purificación , Conformación Proteica , Subunidades de Proteína/aislamiento & purificación , Vitrificación
18.
Alzheimers Dement ; 18(12): 2481-2492, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-35142027

RESUMEN

Abnormal tau protein aggregates constitute a hallmark of Alzheimer's disease. The mechanisms underlying the initiation of tau aggregation in sporadic neurodegeneration remain unclear. Here we investigate whether a non-human prion can seed tau aggregation. Due to their structural similarity with tau aggregates, we chose Sup35NM yeast prion domain fibrils for explorative tau seedings. Upon in vitro incubation with tau monomers, Sup35NM fibrils promoted the formation of morphologically distinct tau fibril strains. In vivo, intrahippocampal inoculation of Sup35NM fibrils accentuated tau pathology in P301S tau transgenic mice. Thus, our results provide first in vivo evidence for heterotypic cross-species seeding of a neurodegenerative human prion-like protein by a yeast prion. This opens up the conceptual perspective that non-mammalian prions present in the human microbiome could be involved in the initiation of protein misfolding in neurodegenerative disorders, a mechanism for which we propose the term "trans-seeding."


Asunto(s)
Enfermedad de Alzheimer , Priones , Tauopatías , Ratones , Animales , Humanos , Proteínas tau/metabolismo , Priones/metabolismo , Enfermedad de Alzheimer/metabolismo , Tauopatías/patología , Saccharomyces cerevisiae/metabolismo , Ratones Transgénicos
19.
J Biol Chem ; 295(26): 8692-8705, 2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32265298

RESUMEN

Myelin protein P2 is a peripheral membrane protein of the fatty acid-binding protein family that functions in the formation and maintenance of the peripheral nerve myelin sheath. Several P2 gene mutations cause human Charcot-Marie-Tooth neuropathy, but the mature myelin sheath assembly mechanism is unclear. Here, cryo-EM of myelin-like proteolipid multilayers revealed an ordered three-dimensional (3D) lattice of P2 molecules between stacked lipid bilayers, visualizing supramolecular assembly at the myelin major dense line. The data disclosed that a single P2 layer is inserted between two bilayers in a tight intermembrane space of ∼3 nm, implying direct interactions between P2 and two membrane surfaces. X-ray diffraction from P2-stacked bicelle multilayers revealed lateral protein organization, and surface mutagenesis of P2 coupled with structure-function experiments revealed a role for both the portal region of P2 and its opposite face in membrane interactions. Atomistic molecular dynamics simulations of P2 on model membrane surfaces suggested that Arg-88 is critical for P2-membrane interactions, in addition to the helical lid domain. Negatively charged lipid headgroups stably anchored P2 on the myelin-like bilayer surface. Membrane binding may be accompanied by opening of the P2 ß-barrel structure and ligand exchange with the apposing bilayer. Our results provide an unprecedented view into an ordered, multilayered biomolecular membrane system induced by the presence of a peripheral membrane protein from human myelin. This is an important step toward deciphering the 3D assembly of a mature myelin sheath at the molecular level.


Asunto(s)
Proteína P2 de Mielina/química , Proteína P2 de Mielina/ultraestructura , Colesterol/metabolismo , Microscopía por Crioelectrón , Ácidos Grasos/metabolismo , Humanos , Membrana Dobles de Lípidos/metabolismo , Simulación de Dinámica Molecular , Proteína P2 de Mielina/genética , Proteína P2 de Mielina/metabolismo , Mutación Puntual , Unión Proteica , Conformación Proteica , Difracción de Rayos X
20.
FASEB J ; 34(10): 13918-13934, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32860273

RESUMEN

Interphotoreceptor retinoid-binding protein (IRBP) is a highly expressed protein secreted by rod and cone photoreceptors that has major roles in photoreceptor homeostasis as well as retinoid and polyunsaturated fatty acid transport between the neural retina and retinal pigment epithelium. Despite two crystal structures reported on fragments of IRBP and decades of research, the overall structure of IRBP and function within the visual cycle remain unsolved. Here, we studied the structure of native bovine IRBP in complex with a monoclonal antibody (mAb5) by cryo-electron microscopy, revealing the tertiary and quaternary structure at sufficient resolution to clearly identify the complex components. Complementary mass spectrometry experiments revealed the structure and locations of N-linked carbohydrate post-translational modifications. This work provides insight into the structure of IRBP, displaying an elongated, flexible three-dimensional architecture not seen among other retinoid-binding proteins. This work is the first step in elucidation of the function of this enigmatic protein.


Asunto(s)
Proteínas del Ojo/química , Proteínas de Unión al Retinol/química , Animales , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/inmunología , Complejo Antígeno-Anticuerpo/química , Bovinos , Microscopía por Crioelectrón , Proteínas del Ojo/inmunología , Femenino , Ratones , Ratones Endogámicos C57BL , Proteínas de Unión al Retinol/inmunología , Imagen Individual de Molécula
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA