Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 263
Filtrar
1.
Nature ; 610(7933): 761-767, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36261523

RESUMEN

Stimulator of interferon genes (STING) functions downstream of cyclic GMP-AMP synthase in DNA sensing or as a direct receptor for bacterial cyclic dinucleotides and small molecules to activate immunity during infection, cancer and immunotherapy1-10. Precise regulation of STING is essential to ensure balanced immune responses and prevent detrimental autoinflammation11-16. After activation, STING, a transmembrane protein, traffics from the endoplasmic reticulum to the Golgi, where its phosphorylation by the protein kinase TBK1 enables signal transduction17-20. The mechanism that ends STING signalling at the Golgi remains unknown. Here we show that adaptor protein complex 1 (AP-1) controls the termination of STING-dependent immune activation. We find that AP-1 sorts phosphorylated STING into clathrin-coated transport vesicles for delivery to the endolysosomal system, where STING is degraded21. We identify a highly conserved dileucine motif in the cytosolic C-terminal tail (CTT) of STING that, together with TBK1-dependent CTT phosphorylation, dictates the AP-1 engagement of STING. A cryo-electron microscopy structure of AP-1 in complex with phosphorylated STING explains the enhanced recognition of TBK1-activated STING. We show that suppression of AP-1 exacerbates STING-induced immune responses. Our results reveal a structural mechanism of negative regulation of STING and establish that the initiation of signalling is inextricably associated with its termination to enable transient activation of immunity.


Asunto(s)
Complejo 1 de Proteína Adaptadora , Clatrina , Complejo 1 de Proteína Adaptadora/química , Complejo 1 de Proteína Adaptadora/metabolismo , Complejo 1 de Proteína Adaptadora/ultraestructura , Clatrina/metabolismo , Microscopía por Crioelectrón , ADN/metabolismo , Inmunidad Innata , Proteínas Serina-Treonina Quinasas , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Secuencias de Aminoácidos , Endosomas/metabolismo , Lisosomas/metabolismo , Fosforilación
2.
Nature ; 608(7924): 803-807, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35859168

RESUMEN

Stimulator of interferon genes (STING) is an antiviral signalling protein that is broadly conserved in both innate immunity in animals and phage defence in prokaryotes1-4. Activation of STING requires its assembly into an oligomeric filament structure through binding of a cyclic dinucleotide4-13, but the molecular basis of STING filament assembly and extension remains unknown. Here we use cryogenic electron microscopy to determine the structure of the active Toll/interleukin-1 receptor (TIR)-STING filament complex from a Sphingobacterium faecium cyclic-oligonucleotide-based antiphage signalling system (CBASS) defence operon. Bacterial TIR-STING filament formation is driven by STING interfaces that become exposed on high-affinity recognition of the cognate cyclic dinucleotide signal c-di-GMP. Repeating dimeric STING units stack laterally head-to-head through surface interfaces, which are also essential for human STING tetramer formation and downstream immune signalling in mammals5. The active bacterial TIR-STING structure reveals further cross-filament contacts that brace the assembly and coordinate packing of the associated TIR NADase effector domains at the base of the filament to drive NAD+ hydrolysis. STING interface and cross-filament contacts are essential for cell growth arrest in vivo and reveal a stepwise mechanism of activation whereby STING filament assembly is required for subsequent effector activation. Our results define the structural basis of STING filament formation in prokaryotic antiviral signalling.


Asunto(s)
Proteínas Bacterianas , Microscopía por Crioelectrón , Proteínas de la Membrana , Receptores de Interleucina-1 , Sphingobacterium , Receptores Toll-Like , Animales , Antivirales/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/inmunología , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/ultraestructura , Bacteriófagos/inmunología , Fosfatos de Dinucleósidos/metabolismo , Humanos , Inmunidad Innata , Proteínas de la Membrana/química , Proteínas de la Membrana/inmunología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Operón/genética , Receptores de Interleucina-1/química , Receptores de Interleucina-1/inmunología , Receptores de Interleucina-1/metabolismo , Receptores de Interleucina-1/ultraestructura , Sphingobacterium/química , Sphingobacterium/genética , Sphingobacterium/ultraestructura , Sphingobacterium/virología , Receptores Toll-Like/química , Receptores Toll-Like/inmunología , Receptores Toll-Like/metabolismo , Receptores Toll-Like/ultraestructura
3.
Biochem Biophys Res Commun ; 590: 163-168, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-34979317

RESUMEN

Liquid-phase electron microscopy is highly desirable for observing biological samples in their native liquid state at high resolution. We developed liquid imaging approaches for biological cells using scanning electron microscopy. Novel approaches included scanning transmission electron imaging using a liquid-cell apparatus (LC-STEM), as well as correlative cathodoluminescence and electron microscopy (CCLEM) imaging. LC-STEM enabled imaging at a ∼2 nm resolution and excellent contrast for the precise recognition of localization, distribution, and configuration of individually labeled membrane proteins on the native cells in solution. CCLEM improved the resolution of fluorescent images down to 10 nm. Liquid SEM technologies will bring unique and wide applications to the study of the structure and function of cells and membrane proteins in their near-native states at the monomolecular level.


Asunto(s)
Proteínas de la Membrana/ultraestructura , Microscopía Electrónica de Rastreo , Línea Celular Tumoral , Receptores ErbB/ultraestructura , Fluorescencia , Humanos
4.
PLoS Biol ; 19(12): e3001474, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34879065

RESUMEN

Endoplasmic reticulum-associated degradation (ERAD) is a protein quality control pathway of fundamental importance to cellular homeostasis. Although multiple ERAD pathways exist for targeting topologically distinct substrates, all pathways require substrate ubiquitination. Here, we characterize a key role for the UBE2G2 Binding Region (G2BR) of the ERAD accessory protein ancient ubiquitous protein 1 (AUP1) in ERAD pathways. This 27-amino acid (aa) region of AUP1 binds with high specificity and low nanomolar affinity to the backside of the ERAD ubiquitin-conjugating enzyme (E2) UBE2G2. The structure of the AUP1 G2BR (G2BRAUP1) in complex with UBE2G2 reveals an interface that includes a network of salt bridges, hydrogen bonds, and hydrophobic interactions essential for AUP1 function in cells. The G2BRAUP1 shares significant structural conservation with the G2BR found in the E3 ubiquitin ligase gp78 and in vitro can similarly allosterically activate ubiquitination in conjunction with ERAD E3s. In cells, AUP1 is uniquely required to maintain normal levels of UBE2G2; this is due to G2BRAUP1 binding to the E2 and preventing its rapid degradation. In addition, the G2BRAUP1 is required for both ER membrane recruitment of UBE2G2 and for its activation at the ER membrane. Thus, by binding to the backside of a critical ERAD E2, G2BRAUP1 plays multiple critical roles in ERAD.


Asunto(s)
Degradación Asociada con el Retículo Endoplásmico/genética , Proteínas de la Membrana/fisiología , Enzimas Ubiquitina-Conjugadoras/fisiología , Secuencia de Aminoácidos/genética , Línea Celular Tumoral , Retículo Endoplásmico/metabolismo , Degradación Asociada con el Retículo Endoplásmico/fisiología , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Unión Proteica/genética , Dominios Proteicos/genética , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/metabolismo , Enzimas Ubiquitina-Conjugadoras/ultraestructura , Ubiquitinación
5.
Mol Cell ; 81(24): 5025-5038.e10, 2021 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-34890564

RESUMEN

The Sonic Hedgehog (SHH) morphogen pathway is fundamental for embryonic development and stem cell maintenance and is implicated in various cancers. A key step in signaling is transfer of a palmitate group to the SHH N terminus, catalyzed by the multi-pass transmembrane enzyme Hedgehog acyltransferase (HHAT). We present the high-resolution cryo-EM structure of HHAT bound to substrate analog palmityl-coenzyme A and a SHH-mimetic megabody, revealing a heme group bound to HHAT that is essential for HHAT function. A structure of HHAT bound to potent small-molecule inhibitor IMP-1575 revealed conformational changes in the active site that occlude substrate binding. Our multidisciplinary analysis provides a detailed view of the mechanism by which HHAT adapts the membrane environment to transfer an acyl chain across the endoplasmic reticulum membrane. This structure of a membrane-bound O-acyltransferase (MBOAT) superfamily member provides a blueprint for other protein-substrate MBOATs and a template for future drug discovery.


Asunto(s)
Aciltransferasas/antagonistas & inhibidores , Aciltransferasas/metabolismo , Inhibidores Enzimáticos/farmacología , Proteínas Hedgehog/metabolismo , Proteínas de la Membrana/metabolismo , Acilación , Aciltransferasas/genética , Aciltransferasas/ultraestructura , Regulación Alostérica , Animales , Células COS , Dominio Catalítico , Chlorocebus aethiops , Microscopía por Crioelectrón , Células HEK293 , Hemo/metabolismo , Humanos , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Simulación de Dinámica Molecular , Palmitoil Coenzima A/metabolismo , Conformación Proteica , Transducción de Señal , Relación Estructura-Actividad
6.
Genes (Basel) ; 12(10)2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34681018

RESUMEN

In recent years, there has been tremendous enthusiasm with respect to detailing the genetic basis of many neoplasms, including conjunctival melanoma (CM). We aim to analyze five proteins associated with CM, namely BRAF, NRAS, c-KIT, NF1, and PTEN. We evaluated each protein for its intrinsically disordered protein regions (IDPRs) and its protein-protein interactions (PPI) with the Predictor of Natural Disordered Protein Regions (PONDR®) and the Search Tool for the Retrieval of Interacting Genes (STRING®). Our PONDR® analysis found high levels of IDPRs in all five proteins with mutations linked to CM. The highest levels of IDPRs were in BRAF (45.95%), followed by PTEN (31.76%), NF1 (22.19%), c-KIT (21.82%), and NRAS (14.81%). Our STRING analysis found that each of these five proteins had more predicted interactions then expected (p-value < 1.0 × 10-16). Our analysis demonstrates that the mutations linked to CM likely affected IDPRs and possibly altered their highly complex PPIs. Quantifying IDPRs in BRAF, NRAS, c-KIT, NF1, and PTEN and understanding these protein regions are important processes as IDPRs can be possible drug targets for novel targeted therapies for treating CM.


Asunto(s)
Neoplasias de la Conjuntiva/genética , Proteínas Intrínsecamente Desordenadas/genética , Melanoma/genética , Conformación Proteica , Neoplasias de la Conjuntiva/patología , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/ultraestructura , Humanos , Proteínas Intrínsecamente Desordenadas/ultraestructura , Melanoma/patología , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Mutación/genética , Neurofibromina 1/genética , Neurofibromina 1/ultraestructura , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/ultraestructura , Mapas de Interacción de Proteínas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/ultraestructura , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/ultraestructura , Transducción de Señal
7.
Nat Commun ; 12(1): 4893, 2021 08 12.
Artículo en Inglés | MEDLINE | ID: mdl-34385445

RESUMEN

The Tweety homologs (TTYHs) are members of a conserved family of eukaryotic membrane proteins that are abundant in the brain. The three human paralogs were assigned to function as anion channels that are either activated by Ca2+ or cell swelling. To uncover their unknown architecture and its relationship to function, we have determined the structures of human TTYH1-3 by cryo-electron microscopy. All structures display equivalent features of a dimeric membrane protein that contains five transmembrane segments and an extended extracellular domain. As none of the proteins shows attributes reminiscent of an anion channel, we revisited functional experiments and did not find any indication of ion conduction. Instead, we find density in an extended hydrophobic pocket contained in the extracellular domain that emerges from the lipid bilayer, which suggests a role of TTYH proteins in the interaction with lipid-like compounds residing in the membrane.


Asunto(s)
Canales de Cloruro/ultraestructura , Microscopía por Crioelectrón/métodos , Proteínas de la Membrana/ultraestructura , Proteínas de Neoplasias/ultraestructura , Canales de Cloruro/química , Canales de Cloruro/metabolismo , Humanos , Canales Iónicos/química , Canales Iónicos/metabolismo , Canales Iónicos/ultraestructura , Membrana Dobles de Lípidos/metabolismo , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Modelos Moleculares , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Unión Proteica , Conformación Proteica
8.
Brief Bioinform ; 22(6)2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34143202

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), a causative agent of the coronavirus disease (COVID-19), is a part of the $\beta $-Coronaviridae family. The virus contains five major protein classes viz., four structural proteins [nucleocapsid (N), membrane (M), envelop (E) and spike glycoprotein (S)] and replicase polyproteins (R), synthesized as two polyproteins (ORF1a and ORF1ab). Due to the severity of the pandemic, most of the SARS-CoV-2-related research are focused on finding therapeutic solutions. However, studies on the sequences and structure space throughout the evolutionary time frame of viral proteins are limited. Besides, the structural malleability of viral proteins can be directly or indirectly associated with the dysfunctionality of the host cell proteins. This dysfunctionality may lead to comorbidities during the infection and may continue at the post-infection stage. In this regard, we conduct the evolutionary sequence-structure analysis of the viral proteins to evaluate their malleability. Subsequently, intrinsic disorder propensities of these viral proteins have been studied to confirm that the short intrinsically disordered regions play an important role in enhancing the likelihood of the host proteins interacting with the viral proteins. These interactions may result in molecular dysfunctionality, finally leading to different diseases. Based on the host cell proteins, the diseases are divided in two distinct classes: (i) proteins, directly associated with the set of diseases while showing similar activities, and (ii) cytokine storm-mediated pro-inflammation (e.g. acute respiratory distress syndrome, malignancies) and neuroinflammation (e.g. neurodegenerative and neuropsychiatric diseases). Finally, the study unveils that males and postmenopausal females can be more vulnerable to SARS-CoV-2 infection due to the androgen-mediated protein transmembrane serine protease 2.


Asunto(s)
COVID-19/genética , Genoma Viral/genética , Conformación Proteica , SARS-CoV-2/ultraestructura , COVID-19/virología , Proteínas de la Envoltura de Coronavirus/genética , Proteínas de la Envoltura de Coronavirus/ultraestructura , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Proteínas de la Nucleocápside/genética , Proteínas de la Nucleocápside/ultraestructura , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Glicoproteína de la Espiga del Coronavirus/genética , Glicoproteína de la Espiga del Coronavirus/ultraestructura , Proteinas del Complejo de Replicasa Viral/genética , Proteinas del Complejo de Replicasa Viral/ultraestructura , Proteínas Estructurales Virales/genética , Proteínas Estructurales Virales/ultraestructura
9.
Nat Commun ; 12(1): 3176, 2021 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-34039995

RESUMEN

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


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Profase Meiótica I , Proteínas de la Membrana/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares/metabolismo , Telómero/metabolismo , Animales , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/aislamiento & purificación , Proteínas de Ciclo Celular/ultraestructura , Línea Celular Tumoral , Cristalografía por Rayos X , Quinasa 2 Dependiente de la Ciclina/genética , Quinasa 2 Dependiente de la Ciclina/aislamiento & purificación , Quinasa 2 Dependiente de la Ciclina/metabolismo , Quinasa 2 Dependiente de la Ciclina/ultraestructura , Femenino , Células HEK293 , Humanos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/aislamiento & purificación , Proteínas de la Membrana/ultraestructura , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/aislamiento & purificación , Proteínas Asociadas a Microtúbulos/ultraestructura , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/aislamiento & purificación , Proteínas Nucleares/ultraestructura , Proteínas Recombinantes/genética , Proteínas Recombinantes/aislamiento & purificación , Proteínas Recombinantes/metabolismo , Proteínas Recombinantes/ultraestructura
10.
Elife ; 102021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33904393

RESUMEN

Many of the world's warm-blooded species are chronically infected with Toxoplasma gondii tissue cysts, including an estimated one-third of the global human population. The cellular processes that permit long-term persistence within the cyst are largely unknown for T. gondii and related coccidian parasites that impact human and animal health. Herein, we show that genetic ablation of TgATG9 substantially reduces canonical autophagy and compromises bradyzoite viability. Transmission electron microscopy revealed numerous structural abnormalities occurring in ∆atg9 bradyzoites. Intriguingly, abnormal mitochondrial networks were observed in TgATG9-deficient bradyzoites, some of which contained numerous different cytoplasmic components and organelles. ∆atg9 bradyzoite fitness was drastically compromised in vitro and in mice, with very few brain cysts identified in mice 5 weeks post-infection. Taken together, our data suggests that TgATG9, and by extension autophagy, is critical for cellular homeostasis in bradyzoites and is necessary for long-term persistence within the cyst of this coccidian parasite.


Asunto(s)
Autofagia , Encéfalo/parasitología , Proteínas de la Membrana/metabolismo , Proteínas Protozoarias/metabolismo , Toxoplasma/metabolismo , Toxoplasmosis Cerebral/parasitología , Animales , Encéfalo/patología , Línea Celular , Modelos Animales de Enfermedad , Femenino , Interacciones Huésped-Parásitos , Humanos , Estadios del Ciclo de Vida , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Ratones Endogámicos CBA , Mitocondrias/genética , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Proteínas Protozoarias/genética , Proteínas Protozoarias/ultraestructura , Factores de Tiempo , Toxoplasma/genética , Toxoplasma/patogenicidad , Toxoplasma/ultraestructura , Toxoplasmosis Cerebral/patología , Vacuolas/genética , Vacuolas/metabolismo , Vacuolas/ultraestructura , Virulencia
11.
Genes (Basel) ; 12(2)2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33672023

RESUMEN

P53, P63, and P73 proteins belong to the P53 family of transcription factors, sharing a common gene organization that, from the P1 and P2 promoters, produces two groups of mRNAs encoding proteins with different N-terminal regions; moreover, alternative splicing events at C-terminus further contribute to the generation of multiple isoforms. P53 family proteins can influence a plethora of cellular pathways mainly through the direct binding to specific DNA sequences known as response elements (REs), and the transactivation of the corresponding target genes. However, the transcriptional activation by P53 family members can be regulated at multiple levels, including the DNA topology at responsive promoters. Here, by using a yeast-based functional assay, we evaluated the influence that a G-quadruplex (G4) prone sequence adjacent to the p53 RE derived from the apoptotic PUMA target gene can exert on the transactivation potential of full-length and N-terminal truncated P53 family α isoforms (wild-type and mutant). Our results show that the presence of a G4 prone sequence upstream or downstream of the P53 RE leads to significant changes in the relative activity of P53 family proteins, emphasizing the potential role of structural DNA features as modifiers of P53 family functions at target promoter sites.


Asunto(s)
ADN/genética , G-Cuádruplex , Proteínas de la Membrana/genética , Proteína Tumoral p73/genética , Proteína p53 Supresora de Tumor/genética , Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/genética , ADN/ultraestructura , Humanos , Proteínas de la Membrana/ultraestructura , Conformación de Ácido Nucleico , Regiones Promotoras Genéticas/genética , Proteínas Proto-Oncogénicas/genética , Elementos de Respuesta/genética , Saccharomyces cerevisiae/genética , Proteína Tumoral p73/ultraestructura , Proteína p53 Supresora de Tumor/ultraestructura
12.
Am J Med Genet A ; 185(4): 1081-1090, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33403770

RESUMEN

Pathogenic variants in Steroid 5 alpha reductase type 3 (SRD5A3) cause rare inherited congenital disorder of glycosylation known as SRD5A3-CDG (MIM# 612379). To date, 43 affected individuals have been reported. Despite the development of various dysmorphic features in significant number of patients, facial recognition entity has not yet been established for SRD5A3-CDG. Herein, we reported a novel SRD5A3 missense pathogenic variant c.460 T > C p.(Ser154Pro). The 3D structural modeling of the SRD5A3 protein revealed additional transmembrane α-helices and predicted that the p.(Ser154Pro) variant is located in a potential active site and is capable of reducing its catalytic efficiency. Based on phenotypes of our patients and all published SRD5A3-CDG cases, we identified the most common clinical features as well as some recurrent dysmorphic features such as arched eyebrows, wide eyes, shallow nasal bridge, short nose, and large mouth. Based on facial digital 2D images, we successfully designed and validated a SRD5A3-CDG computer based dysmorphic facial analysis, which achieved 92.5% accuracy. The current work integrates genotypic, 3D structural modeling and phenotypic characteristics of CDG-SRD5A3 cases with the successful development of computer tool for accurate facial recognition of CDG-SRD5A3 complex cases to assist in the diagnosis of this particular disorder globally.


Asunto(s)
3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/genética , Anomalías Múltiples/genética , Catarata/genética , Trastornos Congénitos de Glicosilación/genética , Proteínas de la Membrana/genética , Atrofia Muscular/genética , 3-Oxo-5-alfa-Esteroide 4-Deshidrogenasa/ultraestructura , Anomalías Múltiples/patología , Adolescente , Catarata/complicaciones , Catarata/patología , Niño , Preescolar , Trastornos Congénitos de Glicosilación/complicaciones , Trastornos Congénitos de Glicosilación/patología , Ojo/patología , Reconocimiento Facial , Facies , Femenino , Humanos , Proteínas de la Membrana/ultraestructura , Atrofia Muscular/complicaciones , Atrofia Muscular/patología , Mutación Missense/genética
13.
FEBS J ; 288(11): 3507-3529, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33305529

RESUMEN

N-myc downstream-regulated gene 1 (NDRG1) is a tumour suppressor involved in vesicular trafficking and stress response. NDRG1 participates in peripheral nerve myelination, and mutations in the NDRG1 gene lead to Charcot-Marie-Tooth neuropathy. The 43-kDa NDRG1 is considered as an inactive member of the α/ß hydrolase superfamily. In addition to a central α/ß hydrolase fold domain, NDRG1 consists of a short N terminus and a C-terminal region with three 10-residue repeats. We determined the crystal structure of the α/ß hydrolase domain of human NDRG1 and characterised the structure and dynamics of full-length NDRG1. The structure of the α/ß hydrolase domain resembles the canonical α/ß hydrolase fold with a central ß sheet surrounded by α helices. Small-angle X-ray scattering and CD spectroscopy indicated a variable conformation for the N- and C-terminal regions. NDRG1 binds to various types of lipid vesicles, and the conformation of the C-terminal region is modulated upon lipid interaction. Intriguingly, NDRG1 interacts with metal ions, such as nickel, but is prone to aggregation in their presence. Our results uncover the structural and dynamic features of NDRG1, as well as elucidate its interactions with metals and lipids, and encourage studies to identify a putative hydrolase activity of NDRG1. DATABASES: The coordinates and structure factors for the crystal structure of human NDRG1 were deposited to PDB (PDB ID: 6ZMM).


Asunto(s)
Proteínas de Ciclo Celular/ultraestructura , Vaina de Mielina/genética , Neoplasias/genética , Proteínas Supresoras de Tumor/genética , Proteínas Portadoras/genética , Proteínas de Ciclo Celular/genética , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Lípidos/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Metales/química , Mutación/genética , Neoplasias/patología , Organogénesis/genética , Unión Proteica/genética , Conformación Proteica
14.
Biochim Biophys Acta Proteins Proteom ; 1869(2): 140558, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33130089

RESUMEN

OMA1 is a mitochondrial protease. Among its substrates are DELE1, a signaling peptide, which can elicit the integrated stress response, as well as the membrane-shaping dynamin-related GTPase OPA1, which can drive mitochondrial outer membrane permeabilization. OMA1 is dormant under physiological conditions but rapidly activated upon mitochondrial stress, such as loss of membrane potential or excessive reactive oxygen species. Accordingly, OMA1 was found to be activated in a number of disease conditions, including cancer and neurodegeneration. OMA1 has a predicted transmembrane domain and is believed to be tethered to the mitochondrial inner membrane. Yet, its structure has not been resolved and its context-dependent regulation remains obscure. Here, I review the literature with focus on OMA1's biochemistry. I provide a good homology model of OMA1's active site with a root-mean-square deviation of 0.9 Šand a DALI Z-score of 19.8. And I build a case for OMA1 actually being an integral membrane protease based on OMA1's role in the generation of small signaling peptides, its functional overlap with PARL, and OMA1's homology with ZMPSTE24. The refined understanding of this important enzyme can help with the design of tool compounds and development of chemical probes in the future.


Asunto(s)
Proteínas de la Membrana/genética , Metaloendopeptidasas/genética , Metaloendopeptidasas/ultraestructura , Mitocondrias/genética , Apoptosis/genética , Humanos , Proteínas de la Membrana/ultraestructura , Mitocondrias/enzimología , Membranas Mitocondriales/química , Membranas Mitocondriales/metabolismo , Péptido Hidrolasas/genética , Transducción de Señal/genética , Homología Estructural de Proteína
15.
Nat Commun ; 11(1): 5080, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-33033258

RESUMEN

Natural transformation is the process by which bacteria take up genetic material from their environment and integrate it into their genome by homologous recombination. It represents one mode of horizontal gene transfer and contributes to the spread of traits like antibiotic resistance. In Vibrio cholerae, a type IVa pilus (T4aP) is thought to facilitate natural transformation by extending from the cell surface, binding to exogenous DNA, and retracting to thread this DNA through the outer membrane secretin, PilQ. Here, we use a functional tagged allele of VcPilQ purified from native V. cholerae cells to determine the cryoEM structure of the VcPilQ secretin in amphipol to ~2.7 Å. We use bioinformatics to examine the domain architecture and gene neighborhood of T4aP secretins in Proteobacteria in comparison with VcPilQ. This structure highlights differences in the architecture of the T4aP secretin from the type II and type III secretion system secretins. Based on our cryoEM structure, we design a series of mutants to reversibly regulate VcPilQ gate dynamics. These experiments support the idea of VcPilQ as a potential druggable target and provide insight into the channel that DNA likely traverses to promote the spread of antibiotic resistance via horizontal gene transfer by natural transformation.


Asunto(s)
Sistemas de Secreción Bacterianos/ultraestructura , Microscopía por Crioelectrón , Fimbrias Bacterianas/ultraestructura , Secretina/química , Vibrio cholerae/metabolismo , Vibrio cholerae/ultraestructura , Cisteína/genética , Proteínas de la Membrana/ultraestructura , Modelos Moleculares , Mutación/genética , Filogenia , Dominios Proteicos , Transformación Bacteriana
16.
J Mol Biol ; 432(23): 6028-6041, 2020 11 20.
Artículo en Inglés | MEDLINE | ID: mdl-33058875

RESUMEN

Linker of nucleoskeleton and cytoskeleton (LINC) complexes are molecular tethers that span the nuclear envelope (NE) and physically connect the nucleus to the cytoskeleton. They transmit mechanical force across the NE in processes such as nuclear anchorage, nuclear migration, and homologous chromosome pairing during meiosis. LINC complexes are composed of KASH proteins traversing the outer nuclear membrane, and SUN proteins crossing the inner nuclear membrane. Humans have several SUN- and KASH-containing proteins, yet what governs their proper engagement is poorly understood. To investigate this question, we solved high resolution crystal structures of human SUN2 in complex with the KASH-peptides of Nesprin3, Nesprin4, and KASH5. In comparison to the published structures of SUN2-KASH1/2 we observe alternative binding modes for these KASH peptides. While the core interactions between SUN and the C-terminal residues of the KASH peptide are similar in all five complexes, the extended KASH-peptide adopts at least two different conformations. The much-improved resolution allows for a more detailed analysis of other elements critical for KASH interaction, including the KASH-lid and the cation loop, and a possible self-locked state for unbound SUN. In summary, we observe distinct differences between the examined SUN-KASH complexes. These differences may have an important role in regulating the SUN-KASH network.


Asunto(s)
Proteínas de Ciclo Celular/ultraestructura , Proteínas de la Membrana/ultraestructura , Proteínas de Microfilamentos/ultraestructura , Complejos Multiproteicos/ultraestructura , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Nucléolo Celular/genética , Nucléolo Celular/ultraestructura , Emparejamiento Cromosómico/genética , Cristalografía por Rayos X , Citoesqueleto/genética , Citoesqueleto/ultraestructura , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Meiosis/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Complejos Multiproteicos/química , Complejos Multiproteicos/genética , Membrana Nuclear/genética , Membrana Nuclear/ultraestructura , Matriz Nuclear/genética , Matriz Nuclear/ultraestructura , Péptidos/química , Péptidos/genética , Conformación Proteica
17.
Mol Biol Cell ; 31(25): 2816-2825, 2020 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-33026967

RESUMEN

The action of guanine nucleotide exchange factors (GEFs) on the ADP-ribosylation factor (ARF) family of small GTPases initiates intracellular transport pathways. This role requires ARF GEFs to be recruited from the cytosol to intracellular membrane compartments. An ARF GEF known as General receptor for 3-phosphoinositides 1 (Grp1) is recruited to the plasma membrane through its pleckstrin homology (PH) domain that recognizes phosphatidylinositol 3,4,5-trisphosphate (PIP3). Here, we find that the phosphorylation of Grp1 induces its PH domain to recognize instead phosphatidylinositol 4-phosphate (PI4P). This phosphorylation also releases an autoinhibitory mechanism that results in the coil-coil (CC) domain of Grp1 engaging two peripheral membrane proteins of the recycling endosome. Because the combination of these actions results in Grp1 being recruited preferentially to the recycling endosome rather than to the plasma membrane, our findings reveal the complexity of recruitment mechanisms that need to be coordinated in localizing an ARF GEF to an intracellular compartment to initiate a transport pathway. Our elucidation is also remarkable for having revealed that phosphoinositide recognition by a PH domain can be switched through its phosphorylation.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/metabolismo , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/fisiología , Fosfatos de Fosfatidilinositol/metabolismo , Células 3T3-L1 , Factores de Ribosilacion-ADP/metabolismo , Secuencia de Aminoácidos/genética , Animales , Membrana Celular/metabolismo , Factores de Intercambio de Guanina Nucleótido/metabolismo , Fosfatos de Inositol/metabolismo , Membranas Intracelulares/metabolismo , Proteínas de la Membrana/ultraestructura , Ratones , Fosfatidilinositoles/metabolismo , Fosforilación , Receptores Citoplasmáticos y Nucleares/metabolismo
18.
Life Sci Alliance ; 3(11)2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32958604

RESUMEN

Tetraspanins are eukaryotic membrane proteins that contribute to a variety of signaling processes by organizing partner-receptor molecules in the plasma membrane. How tetraspanins bind and cluster partner receptors into tetraspanin-enriched microdomains is unknown. Here, we present crystal structures of the large extracellular loop of CD9 bound to nanobodies 4C8 and 4E8 and, the cryo-EM structure of 4C8-bound CD9 in complex with its partner EWI-F. CD9-EWI-F displays a tetrameric arrangement with two central EWI-F molecules, dimerized through their ectodomains, and two CD9 molecules, one bound to each EWI-F transmembrane helix through CD9-helices h3 and h4. In the crystal structures, nanobodies 4C8 and 4E8 bind CD9 at loops C and D, which is in agreement with the 4C8 conformation in the CD9-EWI-F complex. The complex varies from nearly twofold symmetric (with the two CD9 copies nearly anti-parallel) to ca. 50° bent arrangements. This flexible arrangement of CD9-EWI-F with potential CD9 homo-dimerization at either end provides a "concatenation model" for forming short linear or circular assemblies, which may explain the occurrence of tetraspanin-enriched microdomains.


Asunto(s)
Tetraspanina 29/metabolismo , Tetraspanina 29/ultraestructura , Antígenos CD/química , Antígenos CD/metabolismo , Antígenos CD/ultraestructura , Proteínas Portadoras/metabolismo , Línea Celular Tumoral , Membrana Celular/metabolismo , Humanos , Glicoproteínas de Membrana/química , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Tetraspanina 28/metabolismo , Tetraspanina 28/ultraestructura , Tetraspanina 29/fisiología , Tetraspaninas/metabolismo , Tetraspaninas/fisiología , Tetraspaninas/ultraestructura , Factores de Transcripción/metabolismo
19.
J Biol Chem ; 295(28): 9502-9512, 2020 07 10.
Artículo en Inglés | MEDLINE | ID: mdl-32409586

RESUMEN

Six-transmembrane epithelial antigen of the prostate 1 (STEAP1) is an integral membrane protein that is highly up-regulated on the cell surface of several human cancers, making it a promising therapeutic target to manage these diseases. It shares sequence homology with three enzymes (STEAP2-STEAP4) that catalyze the NADPH-dependent reduction of iron(III). However, STEAP1 lacks an intracellular NADPH-binding domain and does not exhibit cellular ferric reductase activity. Thus, both the molecular function of STEAP1 and its role in cancer progression remain elusive. Here, we present a ∼3.0-Šcryo-EM structure of trimeric human STEAP1 bound to three antigen-binding fragments (Fabs) of the clinically used antibody mAb120.545. The structure revealed that STEAP1 adopts a reductase-like conformation and interacts with the Fabs through its extracellular helices. Enzymatic assays in human cells revealed that STEAP1 promotes iron(III) reduction when fused to the intracellular NADPH-binding domain of its family member STEAP4, suggesting that STEAP1 functions as a ferric reductase in STEAP heterotrimers. Our work provides a foundation for deciphering the molecular mechanisms of STEAP1 and may be useful in the design of new therapeutic strategies to target STEAP1 in cancer.


Asunto(s)
Antígenos de Neoplasias , Proteínas de Neoplasias , Neoplasias/enzimología , Oxidorreductasas , Antígenos de Neoplasias/química , Antígenos de Neoplasias/metabolismo , Antígenos de Neoplasias/ultraestructura , Antineoplásicos Inmunológicos/química , Microscopía por Crioelectrón , Células HEK293 , Humanos , Fragmentos Fab de Inmunoglobulinas/química , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/ultraestructura , Proteínas de Neoplasias/química , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/ultraestructura , Neoplasias/ultraestructura , Oxidorreductasas/química , Oxidorreductasas/metabolismo , Oxidorreductasas/ultraestructura , Dominios Proteicos
20.
Nat Struct Mol Biol ; 27(4): 392-399, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32251413

RESUMEN

The endosomal sorting complexes required for transport (ESCRTs) mediate diverse membrane remodeling events. These typically require ESCRT-III proteins to stabilize negatively curved membranes; however, recent work has indicated that certain ESCRT-IIIs also participate in positive-curvature membrane-shaping reactions. ESCRT-IIIs polymerize into membrane-binding filaments, but the structural basis for negative versus positive membrane remodeling by these proteins remains poorly understood. To learn how certain ESCRT-IIIs shape positively curved membranes, we determined structures of human membrane-bound CHMP1B-only, membrane-bound CHMP1B + IST1, and IST1-only filaments by cryo-EM. Our structures show how CHMP1B first polymerizes into a single-stranded helical filament, shaping membranes into moderate-curvature tubules. Subsequently, IST1 assembles a second strand on CHMP1B, further constricting the membrane tube and reducing its diameter nearly to the fission point. Each step of constriction thins the underlying bilayer, lowering the barrier to membrane fission. Our structures reveal how a two-component, sequential polymerization mechanism drives membrane tubulation, constriction and bilayer thinning.


Asunto(s)
Membrana Celular/ultraestructura , Complejos de Clasificación Endosomal Requeridos para el Transporte/ultraestructura , Proteínas Oncogénicas/ultraestructura , Membrana Celular/química , Membrana Celular/genética , Citocinesis/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/química , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Endosomas/química , Endosomas/genética , Endosomas/ultraestructura , Humanos , Proteínas de la Membrana/genética , Proteínas de la Membrana/ultraestructura , Proteínas Oncogénicas/química , Proteínas Oncogénicas/genética , Polimerizacion , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA