Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 105
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 175(3): 809-821.e19, 2018 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-30270044

RESUMEN

Approximately 10% of human protein kinases are believed to be inactive and named pseudokinases because they lack residues required for catalysis. Here, we show that the highly conserved pseudokinase selenoprotein-O (SelO) transfers AMP from ATP to Ser, Thr, and Tyr residues on protein substrates (AMPylation), uncovering a previously unrecognized activity for a member of the protein kinase superfamily. The crystal structure of a SelO homolog reveals a protein kinase-like fold with ATP flipped in the active site, thus providing a structural basis for catalysis. SelO pseudokinases localize to the mitochondria and AMPylate proteins involved in redox homeostasis. Consequently, SelO activity is necessary for the proper cellular response to oxidative stress. Our results suggest that AMPylation may be a more widespread post-translational modification than previously appreciated and that pseudokinases should be analyzed for alternative transferase activities.


Asunto(s)
Adenosina Monofosfato/metabolismo , Dominio Catalítico , Procesamiento Proteico-Postraduccional , Selenoproteínas/metabolismo , Secuencia Conservada , Humanos , Proteínas Mitocondriales/genética , Proteínas Mitocondriales/metabolismo , Estrés Oxidativo , Selenoproteínas/química
2.
Cell ; 161(7): 1619-32, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26091039

RESUMEN

The existence of extracellular phosphoproteins has been acknowledged for over a century. However, research in this area has been undeveloped largely because the kinases that phosphorylate secreted proteins have escaped identification. Fam20C is a kinase that phosphorylates S-x-E/pS motifs on proteins in milk and in the extracellular matrix of bones and teeth. Here, we show that Fam20C generates the majority of the extracellular phosphoproteome. Using CRISPR/Cas9 genome editing, mass spectrometry, and biochemistry, we identify more than 100 secreted phosphoproteins as genuine Fam20C substrates. Further, we show that Fam20C exhibits broader substrate specificity than previously appreciated. Functional annotations of Fam20C substrates suggest roles for the kinase beyond biomineralization, including lipid homeostasis, wound healing, and cell migration and adhesion. Our results establish Fam20C as the major secretory pathway protein kinase and serve as a foundation for new areas of investigation into the role of secreted protein phosphorylation in human biology and disease.


Asunto(s)
Quinasa de la Caseína I/química , Quinasa de la Caseína I/metabolismo , Proteínas de la Matriz Extracelular/química , Proteínas de la Matriz Extracelular/metabolismo , Secuencia de Aminoácidos , Proteínas Sanguíneas/metabolismo , Quinasa de la Caseína I/genética , Adhesión Celular , Movimiento Celular , Proteínas del Líquido Cefalorraquídeo/metabolismo , Proteínas de la Matriz Extracelular/genética , Técnicas de Inactivación de Genes , Ontología de Genes , Humanos , Datos de Secuencia Molecular , Fosfoproteínas/análisis , Vías Secretoras , Especificidad por Sustrato
3.
Cell ; 154(5): 1085-1099, 2013 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-23954414

RESUMEN

The molecular mechanism of autophagy and its relationship to other lysosomal degradation pathways remain incompletely understood. Here, we identified a previously uncharacterized mammalian-specific protein, Beclin 2, which, like Beclin 1, functions in autophagy and interacts with class III PI3K complex components and Bcl-2. However, Beclin 2, but not Beclin 1, functions in an additional lysosomal degradation pathway. Beclin 2 is required for ligand-induced endolysosomal degradation of several G protein-coupled receptors (GPCRs) through its interaction with GASP1. Beclin 2 homozygous knockout mice have decreased embryonic viability, and heterozygous knockout mice have defective autophagy, increased levels of brain cannabinoid 1 receptor, elevated food intake, and obesity and insulin resistance. Our findings identify Beclin 2 as a converging regulator of autophagy and GPCR turnover and highlight the functional and mechanistic diversity of Beclin family members in autophagy, endolysosomal trafficking, and metabolism.


Asunto(s)
Autofagia , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Secuencia de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis/química , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Humanos , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/genética , Lisosomas/metabolismo , Masculino , Proteínas de la Membrana/química , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Datos de Secuencia Molecular , Obesidad/metabolismo , Alineación de Secuencia
4.
Proc Natl Acad Sci U S A ; 121(6): e2312291121, 2024 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-38294943

RESUMEN

A missense variant in patatin-like phospholipase domain-containing protein 3 [PNPLA3(I148M)] is the most impactful genetic risk factor for fatty liver disease (FLD). We previously showed that PNPLA3 is ubiquitylated and subsequently degraded by proteasomes and autophagosomes and that the PNPLA3(148M) variant interferes with this process. To define the machinery responsible for PNPLA3 turnover, we used small interfering (si)RNAs to inactivate components of the ubiquitin proteasome system. Inactivation of bifunctional apoptosis regulator (BFAR), a membrane-bound E3 ubiquitin ligase, reproducibly increased PNPLA3 levels in two lines of cultured hepatocytes. Conversely, overexpression of BFAR decreased levels of endogenous PNPLA3 in HuH7 cells. BFAR and PNPLA3 co-immunoprecipitated when co-expressed in cells. BFAR promoted ubiquitylation of PNPLA3 in vitro in a reconstitution assay using purified, epitope-tagged recombinant proteins. To confirm that BFAR targets PNPLA3, we inactivated Bfar in mice. Levels of PNPLA3 protein were increased twofold in hepatic lipid droplets of Bfar-/- mice with no associated increase in PNPLA3 mRNA levels. Taken together these data are consistent with a model in which BFAR plays a role in the post-translational degradation of PNPLA3. The identification of BFAR provides a potential target to enhance PNPLA3 turnover and prevent FLD.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Proteínas Reguladoras de la Apoptosis , Proteínas de la Membrana , Enfermedad del Hígado Graso no Alcohólico , Animales , Ratones , Aciltransferasas , Hepatocitos/metabolismo , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Fosfolipasas A2 Calcio-Independiente/genética , Ubiquitina , Ubiquitina-Proteína Ligasas/genética , Proteínas de la Membrana/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Humanos , Línea Celular Tumoral
5.
Proc Natl Acad Sci U S A ; 120(12): e2214069120, 2023 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-36917664

RESUMEN

Recent advances in protein structure prediction have generated accurate structures of previously uncharacterized human proteins. Identifying domains in these predicted structures and classifying them into an evolutionary hierarchy can reveal biological insights. Here, we describe the detection and classification of domains from the human proteome. Our classification indicates that only 62% of residues are located in globular domains. We further classify these globular domains and observe that the majority (65%) can be classified among known folds by sequence, with a smaller fraction (33%) requiring structural data to refine the domain boundaries and/or to support their homology. A relatively small number (966 domains) cannot be confidently assigned using our automatic pipelines, thus demanding manual inspection. We classify 47,576 domains, of which only 23% have been included in experimental structures. A portion (6.3%) of these classified globular domains lack sequence-based annotation in InterPro. A quarter (23%) have not been structurally modeled by homology, and they contain 2,540 known disease-causing single amino acid variations whose pathogenesis can now be inferred using AF models. A comparison of classified domains from a series of model organisms revealed expansions of several immune response-related domains in humans and a depletion of olfactory receptors. Finally, we use this classification to expand well-known protein families of biological significance. These classifications are presented on the ECOD website (http://prodata.swmed.edu/ecod/index_human.php).


Asunto(s)
Aminoácidos , Proteoma , Humanos , Proteoma/genética , Alineación de Secuencia , Bases de Datos de Proteínas
6.
PLoS Comput Biol ; 20(2): e1011586, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38416793

RESUMEN

Protein structure prediction has now been deployed widely across several different large protein sets. Large-scale domain annotation of these predictions can aid in the development of biological insights. Using our Evolutionary Classification of Protein Domains (ECOD) from experimental structures as a basis for classification, we describe the detection and cataloging of domains from 48 whole proteomes deposited in the AlphaFold Database. On average, we can provide positive classification (either of domains or other identifiable non-domain regions) for 90% of residues in all proteomes. We classified 746,349 domains from 536,808 proteins comprised of over 226,424,000 amino acid residues. We examine the varying populations of homologous groups in both eukaryotes and bacteria. In addition to containing a higher fraction of disordered regions and unassigned domains, eukaryotes show a higher proportion of repeated proteins, both globular and small repeats. We enumerate those highly populated domains that are shared in both eukaryotes and bacteria, such as the Rossmann domains, TIM barrels, and P-loop domains. Additionally, we compare the sampling of homologous groups from this whole proteome set against our stable ECOD reference and discuss groups that have been enriched by structure predictions. Finally, we discuss the implication of these results for protein target selection for future classification strategies for very large protein sets.


Asunto(s)
Evolución Biológica , Proteoma , Dominios Proteicos , Evolución Molecular , Bacterias , Bases de Datos de Proteínas
7.
Proc Natl Acad Sci U S A ; 119(24): e2203176119, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35648808

RESUMEN

Bacterial signal transduction systems sense changes in the environment and transmit these signals to control cellular responses. The simplest one-component signal transduction systems include an input sensor domain and an output response domain encoded in a single protein chain. Alternatively, two-component signal transduction systems transmit signals by phosphorelay between input and output domains from separate proteins. The membrane-tethered periplasmic bile acid sensor that activates the Vibrio parahaemolyticus type III secretion system adopts an obligate heterodimer of two proteins encoded by partially overlapping VtrA and VtrC genes. This co-component signal transduction system binds bile acid using a lipocalin-like domain in VtrC and transmits the signal through the membrane to a cytoplasmic DNA-binding transcription factor in VtrA. Using the domain and operon organization of VtrA/VtrC, we identify a fast-evolving superfamily of co-component systems in enteric bacteria. Accurate machine learning­based fold predictions for the candidate co-components support their homology in the twilight zone of rapidly evolving sequences and provide mechanistic hypotheses about previously unrecognized lipid-sensing functions.


Asunto(s)
Proteínas Bacterianas , Regulación Bacteriana de la Expresión Génica , Islas Genómicas , Proteínas de la Membrana , Sistemas de Secreción Tipo III , Vibrio parahaemolyticus , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Ácidos y Sales Biliares/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Multimerización de Proteína , Transducción de Señal , Factores de Transcripción/metabolismo , Sistemas de Secreción Tipo III/genética , Vibrio parahaemolyticus/genética , Vibrio parahaemolyticus/patogenicidad , Virulencia/genética
8.
Proteins ; 2024 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-38775337

RESUMEN

A propeptide is removed from a precursor protein to generate its active or mature form. Propeptides play essential roles in protein folding, transportation, and activation and are present in about 2.3% of reviewed proteins in the UniProt database. They are often found in secreted or membrane-bound proteins including proteolytic enzymes, hormones, and toxins. We identified a variety of globular and nonglobular Pfam domains in protein sequences designated as propeptides, some of which form intramolecular interactions with other domains in the mature proteins. Propeptide-containing enzymes mostly function as proteases, as they are depleted in other enzyme classes such as hydrolases acting on DNA and RNA, isomerases, and lyases. We applied AlphaFold to generate structural models for over 7000 proteins with propeptides having no less than 20 residues. Analysis of residue contacts in these models revealed conformational changes for over 300 proteins before and after the cleavage of the propeptide. Examples of conformation change occur in several classes of proteolytic enzymes in the families of subtilisins, trypsins, aspartyl proteases, and thermolysin-like metalloproteases. In most of the observed cases, cleavage of the propeptide releases the constraints imposed by the covalent bond between the propeptide and the mature protein, and cleavage enables stronger interactions between the propeptide and the mature protein. These findings suggest that post-cleavage propeptides could play critical roles in regulating the activity of mature proteins.

9.
Mol Cell ; 63(3): 420-32, 2016 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-27425409

RESUMEN

Recent studies suggest that the microprocessor (Drosha-DGCR8) complex can be recruited to chromatin to catalyze co-transcriptional processing of primary microRNAs (pri-miRNAs) in mammalian cells. However, the molecular mechanism of co-transcriptional miRNA processing is poorly understood. Here we find that HP1BP3, a histone H1-like chromatin protein, specifically associates with the microprocessor and promotes global miRNA biogenesis in human cells. Chromatin immunoprecipitation (ChIP) studies reveal genome-wide co-localization of HP1BP3 and Drosha and HP1BP3-dependent Drosha binding to actively transcribed miRNA loci. Moreover, HP1BP3 specifically binds endogenous pri-miRNAs and facilitates the Drosha/pri-miRNA association in vivo. Knockdown of HP1BP3 compromises pri-miRNA processing by causing premature release of pri-miRNAs from the chromatin. Taken together, these studies suggest that HP1BP3 promotes co-transcriptional miRNA processing via chromatin retention of nascent pri-miRNA transcripts. This work significantly expands the functional repertoire of the H1 family of proteins and suggests the existence of chromatin retention factors for widespread co-transcriptional miRNA processing.


Asunto(s)
Cromatina/metabolismo , MicroARNs/biosíntesis , Proteínas Nucleares/metabolismo , Procesamiento Postranscripcional del ARN , Transcripción Genética , Animales , Sitios de Unión , Cromatina/genética , Inmunoprecipitación de Cromatina , ADN Polimerasa II/genética , ADN Polimerasa II/metabolismo , Proteínas de Unión al ADN , Drosophila melanogaster/genética , Drosophila melanogaster/metabolismo , Genoma Humano , Células HeLa , Humanos , MicroARNs/genética , Proteínas Nucleares/genética , Unión Proteica , Interferencia de ARN , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Transfección
10.
Mol Biol Evol ; 38(5): 2166-2176, 2021 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-33502509

RESUMEN

Centuries of zoological studies have amassed billions of specimens in collections worldwide. Genomics of these specimens promises to reinvigorate biodiversity research. However, because DNA degrades with age in historical specimens, it is a challenge to obtain genomic data for them and analyze degraded genomes. We developed experimental and computational protocols to overcome these challenges and applied our methods to resolve a series of long-standing controversies involving a group of butterflies. We deduced the geographical origins of several historical specimens of uncertain provenance that are at the heart of these debates. Here, genomics tackles one of the greatest problems in zoology: countless old specimens that serve as irreplaceable embodiments of species concepts cannot be confidently assigned to extant species or population due to the lack of diagnostic morphological features and clear documentation of the collection locality. The ability to determine where they were collected will resolve many on-going disputes. More broadly, we show the utility of applying genomics to historical museum specimens to delineate the boundaries of species and populations, and to hypothesize about genotypic determinants of phenotypic traits.


Asunto(s)
Mariposas Diurnas/genética , ADN Antiguo/análisis , Genómica/métodos , Adaptación Biológica/genética , Altitud , Animales , Pigmentación/genética
11.
Nat Chem Biol ; 16(3): 337-344, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31932719

RESUMEN

Infection by the fungal pathogen Cryptococcus neoformans causes lethal meningitis, primarily in immune-compromised individuals. Colonization of the brain by C. neoformans is dependent on copper (Cu) acquisition from the host, which drives critical virulence mechanisms. While C. neoformans Cu+ import and virulence are dependent on the Ctr1 and Ctr4 proteins, little is known concerning extracellular Cu ligands that participate in this process. We identified a C. neoformans gene, BIM1, that is strongly induced during Cu limitation and which encodes a protein related to lytic polysaccharide monooxygenases (LPMOs). Surprisingly, bim1 mutants are Cu deficient, and Bim1 function in Cu accumulation depends on Cu2+ coordination and cell-surface association via a glycophosphatidyl inositol anchor. Bim1 participates in Cu uptake in concert with Ctr1 and expression of this pathway drives brain colonization in mouse infection models. These studies demonstrate a role for LPMO-like proteins as a critical factor for Cu acquisition in fungal meningitis.


Asunto(s)
Cobre/metabolismo , Cryptococcus neoformans/metabolismo , Oxigenasas de Función Mixta/metabolismo , Animales , Criptococosis/metabolismo , Cryptococcus neoformans/patogenicidad , Modelos Animales de Enfermedad , Femenino , Proteínas Fúngicas/genética , Proteínas Fúngicas/metabolismo , Meningitis/metabolismo , Meningitis/fisiopatología , Ratones , Ratones Endogámicos A , Polisacáridos/metabolismo , Virulencia
12.
Nature ; 533(7604): 561-4, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27144356

RESUMEN

ATP binding cassette (ABC) transporters play critical roles in maintaining sterol balance in higher eukaryotes. The ABCG5/ABCG8 heterodimer (G5G8) mediates excretion of neutral sterols in liver and intestines. Mutations disrupting G5G8 cause sitosterolaemia, a disorder characterized by sterol accumulation and premature atherosclerosis. Here we use crystallization in lipid bilayers to determine the X-ray structure of human G5G8 in a nucleotide-free state at 3.9 Å resolution, generating the first atomic model of an ABC sterol transporter. The structure reveals a new transmembrane fold that is present in a large and functionally diverse superfamily of ABC transporters. The transmembrane domains are coupled to the nucleotide-binding sites by networks of interactions that differ between the active and inactive ATPases, reflecting the catalytic asymmetry of the transporter. The G5G8 structure provides a mechanistic framework for understanding sterol transport and the disruptive effects of mutations causing sitosterolaemia.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Lipoproteínas/química , Esteroles/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 5 , Transportador de Casete de Unión a ATP, Subfamilia G, Miembro 8 , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Adenosina Trifosfatasas/metabolismo , Secuencia de Aminoácidos , Sitios de Unión , Biocatálisis , Cristalografía por Rayos X , Humanos , Hipercolesterolemia/genética , Enfermedades Intestinales/genética , Membrana Dobles de Lípidos/química , Membrana Dobles de Lípidos/metabolismo , Errores Innatos del Metabolismo Lipídico/genética , Lipoproteínas/genética , Lipoproteínas/metabolismo , Modelos Moleculares , Datos de Secuencia Molecular , Nucleótidos , Fitosteroles/efectos adversos , Fitosteroles/genética , Pliegue de Proteína , Multimerización de Proteína , Estructura Terciaria de Proteína
13.
Proteins ; 89(12): 1618-1632, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34350630

RESUMEN

An evolutionary-based definition and classification of target evaluation units (EUs) is presented for the 14th round of the critical assessment of structure prediction (CASP14). CASP14 targets included 84 experimental models submitted by various structural groups (designated T1024-T1101). Targets were split into EUs based on the domain organization of available templates and performance of server groups. Several targets required splitting (19 out of 25 multidomain targets) due in part to observed conformation changes. All in all, 96 CASP14 EUs were defined and assigned to tertiary structure assessment categories (Topology-based FM or High Accuracy-based TBM-easy and TBM-hard) considering their evolutionary relationship to existing ECOD fold space: 24 family level, 50 distant homologs (H-group), 12 analogs (X-group), and 10 new folds. Principal component analysis and heatmap visualization of sequence and structure similarity to known templates as well as performance of servers highlighted trends in CASP14 target difficulty. The assigned evolutionary levels (i.e., H-groups) and assessment classes (i.e., FM) displayed overlapping clusters of EUs. Many viral targets diverged considerably from their template homologs and thus were more difficult for prediction than other homology-related targets. On the other hand, some targets did not have sequence-identifiable templates, but were predicted better than expected due to relatively simple arrangements of secondary structural elements. An apparent improvement in overall server performance in CASP14 further complicated traditional classification, which ultimately assigned EUs into high-accuracy modeling (27 TBM-easy and 31 TBM-hard), topology (23 FM), or both (15 FM/TBM).


Asunto(s)
Modelos Moleculares , Conformación Proteica , Proteínas , Secuencia de Aminoácidos , Biología Computacional , Evolución Molecular , Proteínas/química , Proteínas/genética , Análisis de Secuencia de Proteína , Programas Informáticos
14.
Proteins ; 89(12): 1673-1686, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34240477

RESUMEN

This report describes the tertiary structure prediction assessment of difficult modeling targets in the 14th round of the Critical Assessment of Structure Prediction (CASP14). We implemented an official ranking scheme that used the same scores as the previous CASP topology-based assessment, but combined these scores with one that emphasized physically realistic models. The top performing AlphaFold2 group outperformed the rest of the prediction community on all but two of the difficult targets considered in this assessment. They provided high quality models for most of the targets (86% over GDT_TS 70), including larger targets above 150 residues, and they correctly predicted the topology of almost all the rest. AlphaFold2 performance was followed by two manual Baker methods, a Feig method that refined Zhang-server models, two notable automated Zhang server methods (QUARK and Zhang-server), and a Zhang manual group. Despite the remarkable progress in protein structure prediction of difficult targets, both the prediction community and AlphaFold2, to a lesser extent, faced challenges with flexible regions and obligate oligomeric assemblies. The official ranking of top-performing methods was supported by performance generated PCA and heatmap clusters that gave insight into target difficulties and the most successful state-of-the-art structure prediction methodologies.


Asunto(s)
Biología Computacional/métodos , Modelos Moleculares , Conformación Proteica , Pliegue de Proteína , Programas Informáticos , Bases de Datos de Proteínas , Proteínas/química , Proteínas/metabolismo , Análisis de Secuencia de Proteína
15.
Immunity ; 36(6): 959-73, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22705106

RESUMEN

RIG-I and MDA5 detect viral RNA in the cytoplasm and activate signaling cascades leading to the production of type-I interferons. RIG-I is activated through sequential binding of viral RNA and unanchored lysine-63 (K63) polyubiquitin chains, but how polyubiquitin activates RIG-I and whether MDA5 is activated through a similar mechanism remain unresolved. Here, we showed that the CARD domains of MDA5 bound to K63 polyubiquitin and that this binding was essential for MDA5 to activate the transcription factor IRF3. Mutations of conserved residues in MDA5 and RIG-I that disrupt their ubiquitin binding also abrogated their ability to activate IRF3. Polyubiquitin binding induced the formation of a large complex consisting of four RIG-I and four ubiquitin chains. This hetero-tetrameric complex was highly potent in activating the antiviral signaling cascades. These results suggest a unified mechanism of RIG-I and MDA5 activation and reveal a unique mechanism by which ubiquitin regulates cell signaling and immune response.


Asunto(s)
ARN Helicasas DEAD-box/fisiología , Virus de la Encefalomiocarditis/fisiología , Poliubiquitina/metabolismo , Animales , Sistema Libre de Células , Proteína 58 DEAD Box , ARN Helicasas DEAD-box/química , ARN Helicasas DEAD-box/genética , Virus de la Encefalomiocarditis/genética , Fibroblastos/metabolismo , Fibroblastos/virología , Células HEK293/metabolismo , Células HEK293/virología , Humanos , Factor 3 Regulador del Interferón/metabolismo , Helicasa Inducida por Interferón IFIH1 , Interferón beta/biosíntesis , Interferón beta/genética , Ratones , Complejos Multiproteicos , Mutagénesis Sitio-Dirigida , Unión Proteica , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , ARN Viral/metabolismo , Receptores Inmunológicos , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/fisiología , Transducción de Señal/fisiología , Relación Estructura-Actividad , Ubiquitinación
16.
PLoS Comput Biol ; 16(5): e1007775, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32413045

RESUMEN

The human genome harbors a variety of genetic variations. Single-nucleotide changes that alter amino acids in protein-coding regions are one of the major causes of human phenotypic variation and diseases. These single-amino acid variations (SAVs) are routinely found in whole genome and exome sequencing. Evaluating the functional impact of such genomic alterations is crucial for diagnosis of genetic disorders. We developed DeepSAV, a deep-learning convolutional neural network to differentiate disease-causing and benign SAVs based on a variety of protein sequence, structural and functional properties. Our method outperforms most stand-alone programs, and the version incorporating population and gene-level information (DeepSAV+PG) has similar predictive power as some of the best available. We transformed DeepSAV scores of rare SAVs in the human population into a quantity termed "mutation severity measure" for each human protein-coding gene. It reflects a gene's tolerance to deleterious missense mutations and serves as a useful tool to study gene-disease associations. Genes implicated in cancer, autism, and viral interaction are found by this measure as intolerant to mutations, while genes associated with a number of other diseases are scored as tolerant. Among known disease-associated genes, those that are mutation-intolerant are likely to function in development and signal transduction pathways, while those that are mutation-tolerant tend to encode metabolic and mitochondrial proteins.


Asunto(s)
Enfermedad/genética , Predicción/métodos , Genoma Humano/genética , Alelos , Secuencia de Aminoácidos/genética , Biología Computacional/métodos , Aprendizaje Profundo , Redes Reguladoras de Genes/genética , Humanos , Mutación/genética , Mutación Missense/genética , Red Nerviosa , Sistemas de Lectura Abierta/genética , Análisis de Secuencia/métodos , Secuenciación del Exoma/métodos
17.
J Lipid Res ; 61(5): 746-757, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32188638

RESUMEN

The autosomal dominant disorder Schnyder corneal dystrophy (SCD) is caused by mutations in UbiA prenyltransferase domain-containing protein-1 (UBIAD1), which uses geranylgeranyl pyrophosphate (GGpp) to synthesize the vitamin K2 subtype menaquinone-4 (MK-4). SCD is characterized by opacification of the cornea, owing to aberrant build-up of cholesterol in the tissue. We previously discovered that sterols stimulate association of UBIAD1 with ER-localized HMG-CoA reductase, which catalyzes a rate-limiting step in the synthesis of cholesterol and nonsterol isoprenoids, including GGpp. Binding to UBIAD1 inhibits sterol-accelerated ER-associated degradation (ERAD) of reductase and permits continued synthesis of GGpp in cholesterol-replete cells. GGpp disrupts UBIAD1-reductase binding and thereby allows for maximal ERAD of reductase as well as ER-to-Golgi translocation of UBIAD1. SCD-associated UBIAD1 is refractory to GGpp-mediated dissociation from reductase and remains sequestered in the ER to inhibit ERAD. Here, we report development of a biochemical assay for UBIAD1-mediated synthesis of MK-4 in isolated membranes and intact cells. Using this assay, we compared enzymatic activity of WT UBIAD1 with that of SCD-associated variants. Our studies revealed that SCD-associated UBIAD1 exhibited reduced MK-4 synthetic activity, which may result from its reduced affinity for GGpp. Sequestration in the ER protects SCD-associated UBIAD1 from autophagy and allows intracellular accumulation of the mutant protein, which amplifies the inhibitory effect on reductase ERAD. These findings have important implications not only for the understanding of SCD etiology but also for the efficacy of cholesterol-lowering statin therapy, which becomes limited, in part, because of UBIAD1-mediated inhibition of reductase ERAD.


Asunto(s)
Autofagia/genética , Distrofias Hereditarias de la Córnea/genética , Dimetilaliltranstransferasa/genética , Dimetilaliltranstransferasa/metabolismo , Variación Genética , Proteolisis , Vitamina K 2/análogos & derivados , Línea Celular , Humanos , Espacio Intracelular/metabolismo , Transporte de Proteínas , Vitamina K 2/metabolismo
18.
J Biol Chem ; 294(16): 6405-6415, 2019 04 19.
Artículo en Inglés | MEDLINE | ID: mdl-30733336

RESUMEN

Upon phagocytosis into macrophages, the intracellular bacterial pathogen Legionella pneumophila secretes effector proteins that manipulate host cell components, enabling it to evade lysosomal degradation. However, the bacterial proteins involved in this evasion are incompletely characterized. Here we show that the L. pneumophila effector protein RavD targets host membrane compartments and contributes to the molecular mechanism the pathogen uses to prevent encounters with lysosomes. Protein-lipid binding assays revealed that RavD selectively binds phosphatidylinositol-3-phosphate (PI(3)P) in vitro We further determined that a C-terminal RavD region mediates the interaction with PI(3)P and that this interaction requires Arg-292. In transiently transfected mammalian cells, mCherry-RavD colocalized with the early endosome marker EGFP-Rab5 as well as the PI(3)P biosensor EGFP-2×FYVE. However, treatment with the phosphoinositide 3-kinase inhibitor wortmannin did not disrupt localization of mCherry-RavD to endosomal compartments, suggesting that RavD's interaction with PI(3)P is not necessary to anchor RavD to endosomal membranes. Using superresolution and immunogold transmission EM, we observed that, upon translocation into macrophages, RavD was retained onto the Legionella-containing vacuole and was also present on small vesicles adjacent to the vacuole. We also report that despite no detectable effects on intracellular growth of L. pneumophila within macrophages or amebae, the lack of RavD significantly increased the number of vacuoles that accumulate the late endosome/lysosome marker LAMP-1 during macrophage infection. Together, our findings suggest that, although not required for intracellular replication of L. pneumophila, RavD is a part of the molecular mechanism that steers the Legionella-containing vacuole away from endolysosomal maturation pathways.


Asunto(s)
Proteínas Bacterianas/metabolismo , Endosomas/metabolismo , Legionella pneumophila/metabolismo , Enfermedad de los Legionarios/metabolismo , Lisosomas/metabolismo , Macrófagos/metabolismo , Vacuolas/metabolismo , Proteínas Bacterianas/genética , Endosomas/genética , Endosomas/ultraestructura , Células HEK293 , Células HeLa , Humanos , Legionella pneumophila/genética , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/genética , Enfermedad de los Legionarios/patología , Proteínas de Membrana de los Lisosomas/genética , Proteínas de Membrana de los Lisosomas/metabolismo , Lisosomas/genética , Lisosomas/ultraestructura , Macrófagos/microbiología , Macrófagos/ultraestructura , Fosfatidilinositol 3-Quinasas/genética , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatos de Fosfatidilinositol/antagonistas & inhibidores , Fosfatos de Fosfatidilinositol/genética , Fosfatos de Fosfatidilinositol/metabolismo , Células U937 , Vacuolas/genética , Vacuolas/microbiología , Vacuolas/ultraestructura , Wortmanina/farmacología , Proteínas de Unión al GTP rab5/genética , Proteínas de Unión al GTP rab5/metabolismo
19.
J Biol Chem ; 294(15): 6054-6061, 2019 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-30808712

RESUMEN

Adopting a proper topology is crucial for transmembrane proteins to perform their functions. We previously reported that ceramide regulates a transmembrane protein called TM4SF20 (transmembrane 4 L six family member 20) through topological inversion by altering the direction through which the protein is translocated across membranes during translation. This regulatory mechanism, denoted regulated alternative translocation (RAT), depends on a GXXXN motif present in the first transmembrane helix of TM4SF20. Here, using site-directed mutagenesis, we show that Asn-26 in the motif is crucial for RAT of TM4SF20, as it cannot be replaced even by Gln. In contrast, Gly-22 in the motif could be substituted by other small residues such as Ala and Ser without affecting RAT of TM4SF20. We further demonstrate that the GXXXN motif alone is insufficient to induce RAT of a transmembrane protein because TM4SF4, a relative of TM4SF20 that also contains the motif in the first transmembrane helix, did not undergo RAT. Using TM4SF40-TM4SF20 chimeras, we identified Pro-29 of TM4SF20 as another important element required for RAT of the protein. Substituting Pro-29 alone did not affect RAT of TM4SF20, whereas replacing Pro-29 together with either Leu-25 or Val-17 of TM4SF20 with the corresponding residues of TM4SF4 abolished RAT of TM4SF20. Because Val-17, Gly-22, Leu-25, Asn-26, and Pro-29 are predicted to reside along the same surface of the transmembrane helix, our results suggest that interactions with other proteins mediated by this surface during translocation may be critical for RAT of TM4SF20.


Asunto(s)
Tetraspaninas , Células A549 , Secuencias de Aminoácidos , Sustitución de Aminoácidos , Humanos , Glicoproteínas de Membrana/química , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Mutagénesis Sitio-Dirigida , Mutación Missense , Transporte de Proteínas , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/genética , Proteínas Recombinantes de Fusión/metabolismo , Tetraspaninas/química , Tetraspaninas/genética , Tetraspaninas/metabolismo
20.
Proteins ; 88(11): 1513-1527, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32543729

RESUMEN

Protein domains exist by themselves or in combination with other domains to form complex multidomain proteins. Defining domain boundaries in proteins is essential for understanding their evolution and function but is not trivial. More specifically, partitioning domains that interact by forming a single ß-sheet is known to be particularly troublesome for automatic structure-based domain decomposition pipelines. Here, we study edge-to-edge ß-strand interactions between domains in a protein chain, to help define the boundaries for some more difficult cases where a single ß-sheet spanning over two domains gives an appearance of one. We give a number of examples where ß-strands belonging to a single ß-sheet do not belong to a single domain and highlight the difficulties of automatic domain parsers on these examples. This work can be used as a baseline for defining domain boundaries in homologous proteins or proteins with similar domain interactions in the future.


Asunto(s)
Isomerasas de Aminoácido/química , Proteínas de Unión a las Penicilinas/química , Dominios y Motivos de Interacción de Proteínas , Racemasas y Epimerasas/química , Isomerasas de Aminoácido/metabolismo , Secuencia de Aminoácidos , Animales , Bacterias/química , Sitios de Unión , Bases de Datos de Proteínas , Conjuntos de Datos como Asunto , Humanos , Modelos Moleculares , Proteínas de Unión a las Penicilinas/metabolismo , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Racemasas y Epimerasas/metabolismo , Termodinámica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA