Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 3.948
Filtrar
Más filtros

Publication year range
1.
Cell ; 2024 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-38996527

RESUMEN

Latent membrane protein 1 (LMP1) is the primary oncoprotein of Epstein-Barr virus (EBV) and plays versatile roles in the EBV life cycle and pathogenesis. Despite decades of extensive research, the molecular basis for LMP1 folding, assembly, and activation remains unclear. Here, we report cryo-electron microscopy structures of LMP1 in two unexpected assemblies: a symmetric homodimer and a higher-order filamentous oligomer. LMP1 adopts a non-canonical and unpredicted fold that supports the formation of a stable homodimer through tight and antiparallel intermolecular packing. LMP1 dimers further assemble side-by-side into higher-order filamentous oligomers, thereby allowing the accumulation and specific organization of the flexible cytoplasmic tails for efficient recruitment of downstream factors. Super-resolution microscopy and cellular functional assays demonstrate that mutations at both dimeric and oligomeric interfaces disrupt LMP1 higher-order assembly and block multiple LMP1-mediated signaling pathways. Our research provides a framework for understanding the mechanism of LMP1 and for developing potential therapies targeting EBV-associated diseases.

2.
Annu Rev Biochem ; 91: 629-649, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35287474

RESUMEN

Transient receptor potential (TRP) ion channels are sophisticated signaling machines that detect a wide variety of environmental and physiological signals. Every cell in the body expresses one or more members of the extended TRP channel family, which consists of over 30 subtypes, each likely possessing distinct pharmacological, biophysical, and/or structural attributes. While the function of some TRP subtypes remains enigmatic, those involved in sensory signaling are perhaps best characterized and have served as models for understanding how these excitatory ion channels serve as polymodal signal integrators. With the recent resolution revolution in cryo-electron microscopy, these and other TRP channel subtypes are now yielding their secrets to detailed atomic analysis, which is beginning to reveal structural underpinnings of stimulus detection and gating, ion permeation, and allosteric mechanisms governing signal integration. These insights are providing a framework for designing and evaluating modality-specific pharmacological agents for treating sensory and other TRP channel-associated disorders.


Asunto(s)
Canales de Potencial de Receptor Transitorio , Microscopía por Crioelectrón , Transducción de Señal , Canales de Potencial de Receptor Transitorio/química , Canales de Potencial de Receptor Transitorio/genética , Canales de Potencial de Receptor Transitorio/metabolismo
3.
Cell ; 185(7): 1143-1156.e13, 2022 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-35294859

RESUMEN

Transmembrane ß barrel proteins are folded into the outer membrane (OM) of Gram-negative bacteria by the ß barrel assembly machinery (BAM) via a poorly understood process that occurs without known external energy sources. Here, we used single-particle cryo-EM to visualize the folding dynamics of a model ß barrel protein (EspP) by BAM. We found that BAM binds the highly conserved "ß signal" motif of EspP to correctly orient ß strands in the OM during folding. We also found that the folding of EspP proceeds via "hybrid-barrel" intermediates in which membrane integrated ß sheets are attached to the essential BAM subunit, BamA. The structures show an unprecedented deflection of the membrane surrounding the EspP intermediates and suggest that ß sheets progressively fold toward BamA to form a ß barrel. Along with in vivo experiments that tracked ß barrel folding while the OM tension was modified, our results support a model in which BAM harnesses OM elasticity to accelerate ß barrel folding.


Asunto(s)
Proteínas de la Membrana Bacteriana Externa/ultraestructura , Pliegue de Proteína , Proteínas de la Membrana Bacteriana Externa/metabolismo , Microscopía por Crioelectrón , Escherichia coli/metabolismo , Proteínas de Escherichia coli/metabolismo
4.
Cell ; 185(26): 4971-4985.e16, 2022 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-36462505

RESUMEN

Intraflagellar transport (IFT) trains are massive molecular machines that traffic proteins between cilia and the cell body. Each IFT train is a dynamic polymer of two large complexes (IFT-A and -B) and motor proteins, posing a formidable challenge to mechanistic understanding. Here, we reconstituted the complete human IFT-A complex and obtained its structure using cryo-EM. Combined with AlphaFold prediction and genome-editing studies, our results illuminate how IFT-A polymerizes, interacts with IFT-B, and uses an array of ß-propeller and TPR domains to create "carriages" of the IFT train that engage TULP adaptor proteins. We show that IFT-A⋅TULP carriages are essential for cilia localization of diverse membrane proteins, as well as ICK-the key kinase regulating IFT train turnaround. These data establish a structural link between IFT-A's distinct functions, provide a blueprint for IFT-A in the train, and shed light on how IFT evolved from a proto-coatomer ancestor.


Asunto(s)
Cilios , Cinesinas , Humanos , Cilios/metabolismo , Transporte Biológico , Cinesinas/metabolismo , Dineínas/metabolismo , Proteínas de la Membrana/metabolismo , Transporte de Proteínas , Flagelos/metabolismo
5.
Cell ; 185(20): 3739-3752.e18, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36113465

RESUMEN

Lysosomal amino acid efflux by proton-driven transporters is essential for lysosomal homeostasis, amino acid recycling, mTOR signaling, and maintaining lysosomal pH. To unravel the mechanisms of these transporters, we focus on cystinosin, a prototypical lysosomal amino acid transporter that exports cystine to the cytosol, where its reduction to cysteine supplies this limiting amino acid for diverse fundamental processes and controlling nutrient adaptation. Cystinosin mutations cause cystinosis, a devastating lysosomal storage disease. Here, we present structures of human cystinosin in lumen-open, cytosol-open, and cystine-bound states, which uncover the cystine recognition mechanism and capture the key conformational states of the transport cycle. Our structures, along with functional studies and double electron-electron resonance spectroscopic investigations, reveal the molecular basis for the transporter's conformational transitions and protonation switch, show conformation-dependent Ragulator-Rag complex engagement, and demonstrate an unexpected activation mechanism. These findings provide molecular insights into lysosomal amino acid efflux and a potential therapeutic strategy.


Asunto(s)
Cistina , Protones , Sistemas de Transporte de Aminoácidos/metabolismo , Cisteína/metabolismo , Cistina/metabolismo , Humanos , Lisosomas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
6.
Cell ; 185(19): 3551-3567.e39, 2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36055250

RESUMEN

Interactions between cells are indispensable for signaling and creating structure. The ability to direct precise cell-cell interactions would be powerful for engineering tissues, understanding signaling pathways, and directing immune cell targeting. In humans, intercellular interactions are mediated by cell adhesion molecules (CAMs). However, endogenous CAMs are natively expressed by many cells and tend to have cross-reactivity, making them unsuitable for programming specific interactions. Here, we showcase "helixCAM," a platform for engineering synthetic CAMs by presenting coiled-coil peptides on the cell surface. helixCAMs were able to create specific cell-cell interactions and direct patterned aggregate formation in bacteria and human cells. Based on coiled-coil interaction principles, we built a set of rationally designed helixCAM libraries, which led to the discovery of additional high-performance helixCAM pairs. We applied this helixCAM toolkit for various multicellular engineering applications, such as spherical layering, adherent cell targeting, and surface patterning.


Asunto(s)
Bacterias , Péptidos , Humanos , Péptidos/química
7.
Annu Rev Biochem ; 90: 559-579, 2021 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-33492991

RESUMEN

Microorganisms contend with numerous and unusual chemical threats and have evolved a catalog of resistance mechanisms in response. One particularly ancient, pernicious threat is posed by fluoride ion (F-), a common xenobiotic in natural environments that causes broad-spectrum harm to metabolic pathways. This review focuses on advances in the last ten years toward understanding the microbial response to cytoplasmic accumulation of F-, with a special emphasis on the structure and mechanisms of the proteins that microbes use to export fluoride: the CLCF family of F-/H+ antiporters and the Fluc/FEX family of F- channels.


Asunto(s)
Antiportadores/química , Antiportadores/metabolismo , Fluoruros/metabolismo , Canales Iónicos/química , Canales Iónicos/metabolismo , Canales de Cloruro/química , Canales de Cloruro/metabolismo , Citoplasma/metabolismo , Fluoruros/toxicidad , Transporte Iónico , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Conformación Proteica , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo
8.
Annu Rev Biochem ; 90: 535-558, 2021 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-33556281

RESUMEN

Members of the mitochondrial carrier family [solute carrier family 25 (SLC25)] transport nucleotides, amino acids, carboxylic acids, fatty acids, inorganic ions, and vitamins across the mitochondrial inner membrane. They are important for many cellular processes, such as oxidative phosphorylation of lipids and sugars, amino acid metabolism, macromolecular synthesis, ion homeostasis, cellular regulation, and differentiation. Here, we describe the functional elements of the transport mechanism of mitochondrial carriers, consisting of one central substrate-binding site and two gates with salt-bridge networks on either side of the carrier. Binding of the substrate during import causes three gate elements to rotate inward, forming the cytoplasmic network and closing access to the substrate-binding site from the intermembrane space. Simultaneously, three core elements rock outward, disrupting the matrix network and opening the substrate-binding site to the matrix side of the membrane. During export, substrate binding triggers conformational changes involving the same elements but operating in reverse.


Asunto(s)
Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Agrecanos/química , Agrecanos/genética , Agrecanos/metabolismo , Secuencia de Aminoácidos , Aminoácidos/química , Aminoácidos/metabolismo , Sitios de Unión , Transporte Biológico , Calcio/metabolismo , Cardiolipinas/metabolismo , Secuencia Conservada , Citoplasma/metabolismo , Humanos , Translocasas Mitocondriales de ADP y ATP/química , Translocasas Mitocondriales de ADP y ATP/metabolismo , Mutación , Conformación Proteica , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/metabolismo
9.
Cell ; 184(18): 4669-4679.e13, 2021 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-34390643

RESUMEN

Hearing involves two fundamental processes: mechano-electrical transduction and signal amplification. Despite decades of studies, the molecular bases for both remain elusive. Here, we show how prestin, the electromotive molecule of outer hair cells (OHCs) that senses both voltage and membrane tension, mediates signal amplification by coupling conformational changes to alterations in membrane surface area. Cryoelectron microscopy (cryo-EM) structures of human prestin bound with chloride or salicylate at a common "anion site" adopt contracted or expanded states, respectively. Prestin is ensconced within a perimeter of well-ordered lipids, through which it induces dramatic deformation in the membrane and couples protein conformational changes to the bulk membrane. Together with computational studies, we illustrate how the anion site is allosterically coupled to changes in the transmembrane domain cross-sectional area and the surrounding membrane. These studies provide insight into OHC electromotility by providing a structure-based mechanism of the membrane motor prestin.


Asunto(s)
Fenómenos Electrofisiológicos , Transportadores de Sulfato/metabolismo , Aniones , Sitios de Unión , Cloruros/metabolismo , Microscopía por Crioelectrón , Células HEK293 , Humanos , Membrana Dobles de Lípidos/metabolismo , Modelos Moleculares , Simulación de Dinámica Molecular , Dominios Proteicos , Multimerización de Proteína , Estabilidad Proteica , Ácido Salicílico/metabolismo , Homología Estructural de Proteína , Transportadores de Sulfato/química , Transportadores de Sulfato/ultraestructura
10.
Cell ; 181(3): 665-673.e10, 2020 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-32289252

RESUMEN

A growing number of bacteria are recognized to conduct electrons across their cell envelope, and yet molecular details of the mechanisms supporting this process remain unknown. Here, we report the atomic structure of an outer membrane spanning protein complex, MtrAB, that is representative of a protein family known to transport electrons between the interior and exterior environments of phylogenetically and metabolically diverse microorganisms. The structure is revealed as a naturally insulated biomolecular wire possessing a 10-heme cytochrome, MtrA, insulated from the membrane lipidic environment by embedding within a 26 strand ß-barrel formed by MtrB. MtrAB forms an intimate connection with an extracellular 10-heme cytochrome, MtrC, which presents its hemes across a large surface area for electrical contact with extracellular redox partners, including transition metals and electrodes.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/ultraestructura , Proteínas de la Membrana Bacteriana Externa/ultraestructura , Proteínas Bacterianas/ultraestructura , Proteínas de Unión al ARN/ultraestructura , Factores de Transcripción/ultraestructura , Transportadoras de Casetes de Unión a ATP/metabolismo , Membrana Externa Bacteriana/metabolismo , Proteínas de la Membrana Bacteriana Externa/metabolismo , Proteínas Bacterianas/metabolismo , Membrana Celular/metabolismo , Citocromos/metabolismo , Transporte de Electrón/fisiología , Electrones , Hemo/metabolismo , Complejos Multiproteicos/ultraestructura , Oxidación-Reducción , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo
11.
Annu Rev Biochem ; 88: 515-549, 2019 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-30901262

RESUMEN

F1Fo ATP synthases produce most of the ATP in the cell. F-type ATP synthases have been investigated for more than 50 years, but a full understanding of their molecular mechanisms has become possible only with the recent structures of complete, functionally competent complexes determined by electron cryo-microscopy (cryo-EM). High-resolution cryo-EM structures offer a wealth of unexpected new insights. The catalytic F1 head rotates with the central γ-subunit for the first part of each ATP-generating power stroke. Joint rotation is enabled by subunit δ/OSCP acting as a flexible hinge between F1 and the peripheral stalk. Subunit a conducts protons to and from the c-ring rotor through two conserved aqueous channels. The channels are separated by ∼6 Šin the hydrophobic core of Fo, resulting in a strong local field that generates torque to drive rotary catalysis in F1. The structure of the chloroplast F1Fo complex explains how ATPase activity is turned off at night by a redox switch. Structures of mitochondrial ATP synthase dimers indicate how they shape the inner membrane cristae. The new cryo-EM structures complete our picture of the ATP synthases and reveal the unique mechanism by which they transform an electrochemical membrane potential into biologically useful chemical energy.


Asunto(s)
Adenosina Trifosfato/metabolismo , ATPasas de Translocación de Protón/metabolismo , Adenosina Trifosfato/biosíntesis , Animales , Bacterias/enzimología , Bacterias/metabolismo , ATPasas de Translocación de Protón de Cloroplastos/química , ATPasas de Translocación de Protón de Cloroplastos/metabolismo , ATPasas de Translocación de Protón de Cloroplastos/ultraestructura , Cloroplastos/enzimología , Microscopía por Crioelectrón , Eucariontes/enzimología , Eucariontes/metabolismo , Humanos , Mitocondrias/enzimología , ATPasas de Translocación de Protón Mitocondriales/química , ATPasas de Translocación de Protón Mitocondriales/metabolismo , ATPasas de Translocación de Protón Mitocondriales/ultraestructura , Conformación Proteica , Subunidades de Proteína , ATPasas de Translocación de Protón/química , ATPasas de Translocación de Protón/ultraestructura
12.
Cell ; 178(4): 993-1003.e12, 2019 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-31353218

RESUMEN

Voltage-gated sodium (NaV) channels initiate action potentials in nerve, muscle, and other electrically excitable cells. The structural basis of voltage gating is uncertain because the resting state exists only at deeply negative membrane potentials. To stabilize the resting conformation, we inserted voltage-shifting mutations and introduced a disulfide crosslink in the VS of the ancestral bacterial sodium channel NaVAb. Here, we present a cryo-EM structure of the resting state and a complete voltage-dependent gating mechanism. The S4 segment of the VS is drawn intracellularly, with three gating charges passing through the transmembrane electric field. This movement forms an elbow connecting S4 to the S4-S5 linker, tightens the collar around the S6 activation gate, and prevents its opening. Our structure supports the classical "sliding helix" mechanism of voltage sensing and provides a complete gating mechanism for voltage sensor function, pore opening, and activation-gate closure based on high-resolution structures of a single sodium channel protein.


Asunto(s)
Potenciales de Acción/fisiología , Membrana Externa Bacteriana/metabolismo , Escherichia coli/metabolismo , Activación del Canal Iónico/fisiología , Canales de Sodio Activados por Voltaje/metabolismo , Animales , Línea Celular , Microscopía por Crioelectrón , Cristalografía por Rayos X , Mutación , Conformación Proteica en Hélice alfa , Sodio/metabolismo , Spodoptera/citología , Canales de Sodio Activados por Voltaje/química
13.
Cell ; 172(1-2): 68-80.e12, 2018 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-29290469

RESUMEN

Signaling across cellular membranes, the 826 human G protein-coupled receptors (GPCRs) govern a wide range of vital physiological processes, making GPCRs prominent drug targets. X-ray crystallography provided GPCR molecular architectures, which also revealed the need for additional structural dynamics data to support drug development. Here, nuclear magnetic resonance (NMR) spectroscopy with the wild-type-like A2A adenosine receptor (A2AAR) in solution provides a comprehensive characterization of signaling-related structural dynamics. All six tryptophan indole and eight glycine backbone 15N-1H NMR signals in A2AAR were individually assigned. These NMR probes provided insight into the role of Asp522.50 as an allosteric link between the orthosteric drug binding site and the intracellular signaling surface, revealing strong interactions with the toggle switch Trp 2466.48, and delineated the structural response to variable efficacy of bound drugs across A2AAR. The present data support GPCR signaling based on dynamic interactions between two semi-independent subdomains connected by an allosteric switch at Asp522.50.


Asunto(s)
Regulación Alostérica , Receptor de Adenosina A2A/química , Transducción de Señal , Agonistas del Receptor de Adenosina A2/química , Agonistas del Receptor de Adenosina A2/farmacología , Sitio Alostérico , Animales , Simulación del Acoplamiento Molecular , Pichia , Unión Proteica , Receptor de Adenosina A2A/metabolismo , Células Sf9 , Spodoptera
14.
Cell ; 175(5): 1365-1379.e25, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30445040

RESUMEN

The exchange of metabolites between the mitochondrial matrix and the cytosol depends on ß-barrel channels in the outer membrane and α-helical carrier proteins in the inner membrane. The essential translocase of the inner membrane (TIM) chaperones escort these proteins through the intermembrane space, but the structural and mechanistic details remain elusive. We have used an integrated structural biology approach to reveal the functional principle of TIM chaperones. Multiple clamp-like binding sites hold the mitochondrial membrane proteins in a translocation-competent elongated form, thus mimicking characteristics of co-translational membrane insertion. The bound preprotein undergoes conformational dynamics within the chaperone binding clefts, pointing to a multitude of dynamic local binding events. Mutations in these binding sites cause cell death or growth defects associated with impairment of carrier and ß-barrel protein biogenesis. Our work reveals how a single mitochondrial "transfer-chaperone" system is able to guide α-helical and ß-barrel membrane proteins in a "nascent chain-like" conformation through a ribosome-free compartment.


Asunto(s)
Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Chaperonas Moleculares/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Membranas Intracelulares/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/genética , Simulación de Dinámica Molecular , Mutagénesis Sitio-Dirigida , Unión Proteica , Dominios Proteicos , Precursores de Proteínas/química , Precursores de Proteínas/metabolismo , Estructura Secundaria de Proteína , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Alineación de Secuencia
15.
Cell ; 173(7): 1636-1649.e16, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29754813

RESUMEN

Hydrogen gas-evolving membrane-bound hydrogenase (MBH) and quinone-reducing complex I are homologous respiratory complexes with a common ancestor, but a structural basis for their evolutionary relationship is lacking. Here, we report the cryo-EM structure of a 14-subunit MBH from the hyperthermophile Pyrococcus furiosus. MBH contains a membrane-anchored hydrogenase module that is highly similar structurally to the quinone-binding Q-module of complex I while its membrane-embedded ion-translocation module can be divided into a H+- and a Na+-translocating unit. The H+-translocating unit is rotated 180° in-membrane with respect to its counterpart in complex I, leading to distinctive architectures for the two respiratory systems despite their largely conserved proton-pumping mechanisms. The Na+-translocating unit, absent in complex I, resembles that found in the Mrp H+/Na+ antiporter and enables hydrogen gas evolution by MBH to establish a Na+ gradient for ATP synthesis near 100°C. MBH also provides insights into Mrp structure and evolution of MBH-based respiratory enzymes.


Asunto(s)
Proteínas Arqueales/metabolismo , Hidrogenasas/metabolismo , Pyrococcus furiosus/metabolismo , Secuencia de Aminoácidos , Proteínas Arqueales/química , Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Sitios de Unión , Membrana Celular/química , Membrana Celular/metabolismo , Microscopía por Crioelectrón , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/metabolismo , Evolución Molecular , Hidrógeno/metabolismo , Hidrogenasas/química , Hidrogenasas/genética , Mutagénesis , Estructura Cuaternaria de Proteína , Subunidades de Proteína/química , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/aislamiento & purificación , Alineación de Secuencia , Sodio/química , Sodio/metabolismo , Intercambiadores de Sodio-Hidrógeno/química , Intercambiadores de Sodio-Hidrógeno/metabolismo
16.
Cell ; 169(1): 96-107.e12, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28340354

RESUMEN

Transporters shuttle molecules across cell membranes by alternating among distinct conformational states. Fundamental questions remain about how transporters transition between states and how such structural rearrangements regulate substrate translocation. Here, we capture the translocation process by crystallography and unguided molecular dynamics simulations, providing an atomic-level description of alternating access transport. Simulations of a SWEET-family transporter initiated from an outward-open, glucose-bound structure reported here spontaneously adopt occluded and inward-open conformations. Strikingly, these conformations match crystal structures, including our inward-open structure. Mutagenesis experiments further validate simulation predictions. Our results reveal that state transitions are driven by favorable interactions formed upon closure of extracellular and intracellular "gates" and by an unfavorable transmembrane helix configuration when both gates are closed. This mechanism leads to tight allosteric coupling between gates, preventing them from opening simultaneously. Interestingly, the substrate appears to take a "free ride" across the membrane without causing major structural rearrangements in the transporter.


Asunto(s)
Bacterias/química , Proteínas Bacterianas/química , Proteínas de Transporte de Membrana/química , Bacterias/clasificación , Cristalografía por Rayos X , Modelos Moleculares , Simulación de Dinámica Molecular , Conformación Proteica
17.
Cell ; 170(6): 1234-1246.e14, 2017 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-28823560

RESUMEN

AMPA receptors mediate fast excitatory neurotransmission in the mammalian brain and transduce the binding of presynaptically released glutamate to the opening of a transmembrane cation channel. Within the postsynaptic density, however, AMPA receptors coassemble with transmembrane AMPA receptor regulatory proteins (TARPs), yielding a receptor complex with altered gating kinetics, pharmacology, and pore properties. Here, we elucidate structures of the GluA2-TARP γ2 complex in the presence of the partial agonist kainate or the full agonist quisqualate together with a positive allosteric modulator or with quisqualate alone. We show how TARPs sculpt the ligand-binding domain gating ring, enhancing kainate potency and diminishing the ensemble of desensitized states. TARPs encircle the receptor ion channel, stabilizing M2 helices and pore loops, illustrating how TARPs alter receptor pore properties. Structural and computational analysis suggests the full agonist and modulator complex harbors an ion-permeable channel gate, providing the first view of an activated AMPA receptor.


Asunto(s)
Canales de Calcio/química , Receptores AMPA/química , Animales , Microscopía por Crioelectrón , Agonistas de Aminoácidos Excitadores/química , Agonistas de Aminoácidos Excitadores/farmacología , Ácido Kaínico/química , Ácido Kaínico/farmacología , Modelos Moleculares , Ácido Quiscuálico/química , Ácido Quiscuálico/farmacología , Ratas , Receptores AMPA/agonistas
18.
Mol Cell ; 84(10): 1917-1931.e15, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38723633

RESUMEN

Many multi-spanning membrane proteins contain poorly hydrophobic transmembrane domains (pTMDs) protected from phospholipid in mature structure. Nascent pTMDs are difficult for translocon to recognize and insert. How pTMDs are discerned and packed into mature, muti-spanning configuration remains unclear. Here, we report that pTMD elicits a post-translational topogenesis pathway for its recognition and integration. Using six-spanning protein adenosine triphosphate-binding cassette transporter G2 (ABCG2) and cultured human cells as models, we show that ABCG2's pTMD2 can pass through translocon into the endoplasmic reticulum (ER) lumen, yielding an intermediate with inserted yet mis-oriented downstream TMDs. After translation, the intermediate recruits P5A-ATPase ATP13A1, which facilitates TMD re-orientation, allowing further folding and the integration of the remaining lumen-exposed pTMD2. Depleting ATP13A1 or disrupting pTMD-characteristic residues arrests intermediates with mis-oriented and exposed TMDs. Our results explain how a "difficult" pTMD is co-translationally skipped for insertion and post-translationally buried into the final correct structure at the late folding stage to avoid excessive lipid exposure.


Asunto(s)
Retículo Endoplásmico , Pliegue de Proteína , Humanos , Retículo Endoplásmico/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/química , ATPasas de Translocación de Protón/metabolismo , ATPasas de Translocación de Protón/genética , ATPasas de Translocación de Protón/química , Células HEK293 , Dominios Proteicos , Interacciones Hidrofóbicas e Hidrofílicas , Procesamiento Proteico-Postraduccional , Transportadoras de Casetes de Unión a ATP/metabolismo , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/química
19.
Mol Cell ; 84(6): 1101-1119.e9, 2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38428433

RESUMEN

Mitochondrial outer membrane ⍺-helical proteins play critical roles in mitochondrial-cytoplasmic communication, but the rules governing the targeting and insertion of these biophysically diverse proteins remain unknown. Here, we first defined the complement of required mammalian biogenesis machinery through genome-wide CRISPRi screens using topologically distinct membrane proteins. Systematic analysis of nine identified factors across 21 diverse ⍺-helical substrates reveals that these components are organized into distinct targeting pathways that act on substrates based on their topology. NAC is required for the efficient targeting of polytopic proteins, whereas signal-anchored proteins require TTC1, a cytosolic chaperone that physically engages substrates. Biochemical and mutational studies reveal that TTC1 employs a conserved TPR domain and a hydrophobic groove in its C-terminal domain to support substrate solubilization and insertion into mitochondria. Thus, the targeting of diverse mitochondrial membrane proteins is achieved through topological triaging in the cytosol using principles with similarities to ER membrane protein biogenesis systems.


Asunto(s)
Membranas Mitocondriales , Proteínas de Saccharomyces cerevisiae , Animales , Membranas Mitocondriales/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Chaperonas Moleculares/genética , Chaperonas Moleculares/metabolismo , Mutación , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Transporte de Proteínas , Proteínas de Saccharomyces cerevisiae/metabolismo , Mamíferos/metabolismo
20.
Mol Cell ; 84(10): 1932-1947.e10, 2024 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-38703769

RESUMEN

Mutations in transporters can impact an individual's response to drugs and cause many diseases. Few variants in transporters have been evaluated for their functional impact. Here, we combine saturation mutagenesis and multi-phenotypic screening to dissect the impact of 11,213 missense single-amino-acid deletions, and synonymous variants across the 554 residues of OCT1, a key liver xenobiotic transporter. By quantifying in parallel expression and substrate uptake, we find that most variants exert their primary effect on protein abundance, a phenotype not commonly measured alongside function. Using our mutagenesis results combined with structure prediction and molecular dynamic simulations, we develop accurate structure-function models of the entire transport cycle, providing biophysical characterization of all known and possible human OCT1 polymorphisms. This work provides a complete functional map of OCT1 variants along with a framework for integrating functional genomics, biophysical modeling, and human genetics to predict variant effects on disease and drug efficacy.


Asunto(s)
Simulación de Dinámica Molecular , Humanos , Células HEK293 , Relación Estructura-Actividad , Mutación Missense , Farmacogenética , Fenotipo , Transportador 1 de Catión Orgánico/genética , Transportador 1 de Catión Orgánico/metabolismo , Mutación , Conformación Proteica , Transporte Biológico , Factor 1 de Transcripción de Unión a Octámeros
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda