Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 104
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 145(4): 513-28, 2011 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-21565611

RESUMEN

Nephronophthisis (NPHP), Joubert (JBTS), and Meckel-Gruber (MKS) syndromes are autosomal-recessive ciliopathies presenting with cystic kidneys, retinal degeneration, and cerebellar/neural tube malformation. Whether defects in kidney, retinal, or neural disease primarily involve ciliary, Hedgehog, or cell polarity pathways remains unclear. Using high-confidence proteomics, we identified 850 interactors copurifying with nine NPHP/JBTS/MKS proteins and discovered three connected modules: "NPHP1-4-8" functioning at the apical surface, "NPHP5-6" at centrosomes, and "MKS" linked to Hedgehog signaling. Assays for ciliogenesis and epithelial morphogenesis in 3D renal cultures link renal cystic disease to apical organization defects, whereas ciliary and Hedgehog pathway defects lead to retinal or neural deficits. Using 38 interactors as candidates, linkage and sequencing analysis of 250 patients identified ATXN10 and TCTN2 as new NPHP-JBTS genes, and our Tctn2 mouse knockout shows neural tube and Hedgehog signaling defects. Our study further illustrates the power of linking proteomic networks and human genetics to uncover critical disease pathways.


Asunto(s)
Enfermedades Renales Quísticas/genética , Proteínas de la Membrana/genética , Transducción de Señal , Animales , Ataxina-10 , Centrosoma/metabolismo , Cilios/metabolismo , Trastornos de la Motilidad Ciliar/genética , Encefalocele/genética , Proteínas Hedgehog/metabolismo , Humanos , Enfermedades Renales Quísticas/metabolismo , Ratones , Células 3T3 NIH , Proteínas del Tejido Nervioso/genética , Enfermedades Renales Poliquísticas/genética , Retinitis Pigmentosa , Pez Cebra
2.
J Biol Chem ; 296: 100194, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33334891

RESUMEN

Cohesin is a multiprotein ring complex that regulates 3D genome organization, sister chromatid cohesion, gene expression, and DNA repair. Cohesin is known to be ubiquitinated, although the mechanism, regulation, and effects of cohesin ubiquitination remain poorly defined. We previously used gene editing to introduce a dual epitope tag into the endogenous allele of each of 11 known components of cohesin in human HCT116 cells. Here we report that mass spectrometry analysis of dual-affinity purifications identified the USP13 deubiquitinase as a novel cohesin-interacting protein. Subsequent immunoprecipitation/Western blots confirmed the endogenous interaction in HCT116, 293T, HeLa, and RPE-hTERT cells; demonstrated that the interaction occurs specifically in the soluble nuclear fraction (not in the chromatin); requires the ubiquitin-binding domains (UBA1/2) of USP13; and occurs preferentially during DNA replication. Reciprocal dual-affinity purification of endogenous USP13 followed by mass spectrometry demonstrated that cohesin is its primary interactor in the nucleus. Ectopic expression and CRISPR knockout of USP13 showed that USP13 is paradoxically required for both deubiquitination and ubiquitination of cohesin subunits in human cells. USP13 was dispensable for sister chromatid cohesion in HCT116 and HeLa cells, whereas it was required for the dissociation of cohesin from chromatin as cells transit through mitosis. Together these results identify USP13 as a new cohesin-interacting protein that regulates the ubiquitination of cohesin and its cell cycle regulated interaction with chromatin.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Proteasas Ubiquitina-Específicas/metabolismo , Ubiquitina/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Cromatina/genética , Proteínas Cromosómicas no Histona/química , Proteínas Cromosómicas no Histona/genética , Segregación Cromosómica , Reparación del ADN , Replicación del ADN , Células HCT116 , Células HeLa , Humanos , Dominios y Motivos de Interacción de Proteínas , Proteasas Ubiquitina-Específicas/química , Proteasas Ubiquitina-Específicas/genética , Ubiquitinación , Cohesinas
3.
J Biol Chem ; 294(22): 8760-8772, 2019 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-31010829

RESUMEN

The cohesin complex regulates sister chromatid cohesion, chromosome organization, gene expression, and DNA repair. Cohesin is a ring complex composed of four core subunits and seven regulatory subunits. In an effort to comprehensively identify additional cohesin-interacting proteins, we used gene editing to introduce a dual epitope tag into the endogenous allele of each of 11 known components of cohesin in cultured human cells, and we performed MS analyses on dual-affinity purifications. In addition to reciprocally identifying all known components of cohesin, we found that cohesin interacts with a panoply of splicing factors and RNA-binding proteins (RBPs). These included diverse components of the U4/U6.U5 tri-small nuclear ribonucleoprotein complex and several splicing factors that are commonly mutated in cancer. The interaction between cohesin and splicing factors/RBPs was RNA- and DNA-independent, occurred in chromatin, was enhanced during mitosis, and required RAD21. Furthermore, cohesin-interacting splicing factors and RBPs followed the cohesin cycle and prophase pathway of cell cycle-regulated interactions with chromatin. Depletion of cohesin-interacting splicing factors and RBPs resulted in aberrant mitotic progression. These results provide a comprehensive view of the endogenous human cohesin interactome and identify splicing factors and RBPs as functionally significant cohesin-interacting proteins.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromosómicas no Histona/metabolismo , Mitosis , Proteómica , Factores de Empalme de ARN/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Ciclo Celular/antagonistas & inhibidores , Proteínas de Ciclo Celular/genética , Línea Celular Tumoral , Cromatina/metabolismo , Proteínas Cromosómicas no Histona/genética , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Microscopía Fluorescente , Unión Proteica , Mapas de Interacción de Proteínas , Interferencia de ARN , Factores de Empalme de ARN/antagonistas & inhibidores , Factores de Empalme de ARN/genética , ARN Interferente Pequeño/metabolismo , Proteínas de Unión al ARN/antagonistas & inhibidores , Proteínas de Unión al ARN/genética , Cohesinas
4.
Genes Dev ; 24(19): 2180-93, 2010 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-20889716

RESUMEN

Primary cilia function as a sensory signaling compartment in processes ranging from mammalian Hedgehog signaling to neuronal control of obesity. Intraflagellar transport (IFT) is an ancient, conserved mechanism required to assemble cilia and for trafficking within cilia. The link between IFT, sensory signaling, and obesity is not clearly defined, but some novel monogenic obesity disorders may be linked to ciliary defects. The tubby mouse, which presents with adult-onset obesity, arises from mutation in the Tub gene. The tubby-like proteins comprise a related family of poorly understood proteins with roles in neural development and function. We find that specific Tubby family proteins, notably Tubby-like protein 3 (TULP3), bind to the IFT-A complex. IFT-A is linked to retrograde ciliary transport, but, surprisingly, we find that the IFT-A complex has a second role directing ciliary entry of TULP3. TULP3 and IFT-A, in turn, promote trafficking of a subset of G protein-coupled receptors (GPCRs), but not Smoothened, to cilia. Both IFT-A and membrane phosphoinositide-binding properties of TULP3 are required for ciliary GPCR localization. TULP3 and IFT-A proteins both negatively regulate Hedgehog signaling in the mouse embryo, and the TULP3-IFT-A interaction suggests how these proteins cooperate during neural tube patterning.


Asunto(s)
Cilios/metabolismo , Proteínas de Unión al GTP/metabolismo , Fosfatidilinositoles/metabolismo , Proteínas/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Secuencia de Aminoácidos , Animales , Línea Celular , Células Cultivadas , Proteínas Hedgehog/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Péptidos y Proteínas de Señalización Intracelular , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Complejos Multiproteicos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Alineación de Secuencia
5.
J Biol Chem ; 290(37): 22795-804, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26221039

RESUMEN

MutS homolog 2 (MSH2) is an essential DNA mismatch repair (MMR) protein. It interacts with MSH6 or MSH3 to form the MutSα or MutSß complex, respectively, which recognize base-base mispairs and insertions/deletions and initiate the repair process. Mutation or dysregulation of MSH2 causes genomic instability that can lead to cancer. MSH2 is acetylated at its C terminus, and histone deacetylase (HDAC6) deacetylates MSH2. However, whether other regions of MSH2 can be acetylated and whether other histone deacetylases (HDACs) and histone acetyltransferases (HATs) are involved in MSH2 deacetylation/acetylation is unknown. Here, we report that MSH2 can be acetylated at Lys-73 near the N terminus. Lys-73 is highly conserved across many species. Although several Class I and II HDACs interact with MSH2, HDAC10 is the major enzyme that deacetylates MSH2 at Lys-73. Histone acetyltransferase HBO1 might acetylate this residue. HDAC10 overexpression in HeLa cells stimulates cellular DNA MMR activity, whereas HDAC10 knockdown decreases DNA MMR activity. Thus, our study identifies an HDAC10-mediated regulatory mechanism controlling the DNA mismatch repair function of MSH2.


Asunto(s)
Reparación de la Incompatibilidad de ADN , ADN/metabolismo , Histona Desacetilasas/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Acetilación , ADN/genética , Células HeLa , Histona Desacetilasas/genética , Humanos , Proteína 2 Homóloga a MutS/genética
6.
J Biol Chem ; 289(11): 7788-98, 2014 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-24448800

RESUMEN

The Doc toxin from bacteriophage P1 (of the phd-doc toxin-antitoxin system) has served as a model for the family of Doc toxins, many of which are harbored in the genomes of pathogens. We have shown previously that the mode of action of this toxin is distinct from the majority derived from toxin-antitoxin systems: it does not cleave RNA; in fact P1 Doc expression leads to mRNA stabilization. However, the molecular triggers that lead to translation arrest are not understood. The presence of a Fic domain, albeit slightly altered in length and at the catalytic site, provided a clue to the mechanism of P1 Doc action, as most proteins with this conserved domain inactivate GTPases through addition of an adenylyl group (also referred to as AMPylation). We demonstrated that P1 Doc added a single phosphate group to the essential translation elongation factor and GTPase, elongation factor (EF)-Tu. The phosphorylation site was at a highly conserved threonine, Thr-382, which was blocked when EF-Tu was treated with the antibiotic kirromycin. Therefore, we have established that Fic domain proteins can function as kinases. This distinct enzymatic activity exhibited by P1 Doc also solves the mystery of the degenerate Fic motif unique to the Doc family of toxins. Moreover, we have established that all characterized Fic domain proteins, even those that phosphorylate, target pivotal GTPases for inactivation through a post-translational modification at a single functionally critical acceptor site.


Asunto(s)
Bacteriófago P1/metabolismo , Proteínas de Escherichia coli/metabolismo , Extensión de la Cadena Peptídica de Translación , Factor Tu de Elongación Peptídica/metabolismo , Proteínas Virales/metabolismo , Secuencias de Aminoácidos , Antibacterianos/química , Sitios de Unión , Proliferación Celular , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica , Espectrometría de Masas , Simulación del Acoplamiento Molecular , Fosforilación , Unión Proteica , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Piridonas/química , ARN Mensajero/metabolismo , Proteínas Recombinantes/química , Treonina/química
7.
Proc Natl Acad Sci U S A ; 109(31): 12710-5, 2012 Jul 31.
Artículo en Inglés | MEDLINE | ID: mdl-22802624

RESUMEN

The Gram-negative bacteria Yersinia pestis, causative agent of plague, is extremely virulent. One mechanism contributing to Y. pestis virulence is the presence of a type-three secretion system, which injects effector proteins, Yops, directly into immune cells of the infected host. One of these Yop proteins, YopJ, is proapoptotic and inhibits mammalian NF-κB and MAP-kinase signal transduction pathways. Although the molecular mechanism remained elusive for some time, recent work has shown that YopJ acts as a serine/threonine acetyl-transferase targeting MAP2 kinases. Using Drosophila as a model system, we find that YopJ inhibits one innate immune NF-κB signaling pathway (IMD) but not the other (Toll). In fact, we show YopJ mediated serine/threonine acetylation and inhibition of dTAK1, the critical MAP3 kinase in the IMD pathway. Acetylation of critical serine/threonine residues in the activation loop of Drosophila TAK1 blocks phosphorylation of the protein and subsequent kinase activation. In addition, studies in mammalian cells show similar modification and inhibition of hTAK1. These data present evidence that TAK1 is a target for YopJ-mediated inhibition.


Asunto(s)
Proteínas Bacterianas/metabolismo , Inmunidad Innata , Quinasas Quinasa Quinasa PAM/metabolismo , Sistema de Señalización de MAP Quinasas , Serina O-Acetiltransferasa/metabolismo , Yersinia pestis/enzimología , Acetilación , Animales , Proteínas Bacterianas/inmunología , Drosophila melanogaster , Células HEK293 , Humanos , Quinasas Quinasa Quinasa PAM/inmunología , FN-kappa B/inmunología , FN-kappa B/metabolismo , Peste/inmunología , Peste/metabolismo , Serina O-Acetiltransferasa/inmunología , Yersinia pestis/inmunología , Yersinia pestis/patogenicidad
8.
J Proteome Res ; 12(10): 4351-65, 2013 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-24004147

RESUMEN

Plasma proteomic experiments performed rapidly and economically using several of the latest high-resolution mass spectrometers were compared. Four quantitative hyperfractionated plasma proteomics experiments were analyzed in replicates by two AB SCIEX TripleTOF 5600 and three Thermo Scientific Orbitrap (Elite/LTQ-Orbitrap Velos/Q Exactive) instruments. Each experiment compared two iTRAQ isobaric-labeled immunodepleted plasma proteomes, provided as 30 labeled peptide fractions, and 480 LC-MS/MS runs delivered >250 GB of data in 2 months. Several analysis algorithms were compared. At 1% false discovery rate, the relative comparative findings concluded that the Thermo Scientific Q Exactive Mass Spectrometer resulted in the highest number of identified proteins and unique sequences with iTRAQ quantitation. The confidence of iTRAQ fold-change for each protein is dependent on the overall ion statistics (Mascot Protein Score) attainable by each instrument. The benchmarking also suggested how to further improve the mass spectrometry parameters and HPLC conditions. Our findings highlight the special challenges presented by the low abundance peptide ions of iTRAQ plasma proteome because the dynamic range of plasma protein abundance is uniquely high compared with cell lysates, necessitating high instrument sensitivity.


Asunto(s)
Proteínas Sanguíneas/química , Espectrometría de Masas en Tándem/métodos , Proteínas Sanguíneas/aislamiento & purificación , Proteínas Sanguíneas/metabolismo , Humanos , Inmunoprecipitación , Mapeo Peptídico , Proteómica , Estándares de Referencia , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Espectrometría de Masas en Tándem/instrumentación , Espectrometría de Masas en Tándem/normas
9.
J Biol Chem ; 287(11): 7834-44, 2012 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-22235124

RESUMEN

The mouse and human TPSB2 and TPSAB1 genes encode tetramer-forming tryptases stored in the secretory granules of mast cells (MCs) ionically bound to heparin-containing serglycin proteoglycans. In mice these genes encode mouse MC protease-6 (mMCP-6) and mMCP-7. The corresponding human genes encode a family of serine proteases that collectively are called hTryptase-ß. We previously showed that the α chain of fibrinogen is a preferred substrate of mMCP-7. We now show that this plasma protein also is highly susceptible to degradation by hTryptase-ß· and mMCP-6·heparin complexes and that Lys(575) is a preferred cleavage site in the protein α chain. Because cutaneous mouse MCs store substantial amounts of mMCP-6·heparin complexes in their secretory granules, the passive cutaneous anaphylaxis reaction was induced in the skin of mMCP-6(+)/mMCP-7(-) and mMCP-6(-)/mMCP-7(-) C57BL/6 mice. In support of the in vitro data, fibrin deposits were markedly increased in the skin of the double-deficient mice 6 h after IgE-sensitized animals were given the relevant antigen. Fibrinogen is a major constituent of the edema fluid that accumulates in tissues when MCs degranulate. Our discovery that mouse and human tetramer-forming tryptases destroy fibrinogen before this circulating protein can be converted to fibrin changes the paradigm of how MCs hinder fibrin deposition and blood coagulation internally. Because of the adverse consequences of fibrin deposits in tissues, our data explain why mice and humans lack a circulating protease inhibitor that rapidly inactivates MC tryptases and why mammals have two genes that encode tetramer-forming serine proteases that preferentially degrade fibrinogen.


Asunto(s)
Coagulación Sanguínea , Fibrina/metabolismo , Fibrinógeno/metabolismo , Heparina/metabolismo , Mastocitos/enzimología , Proteolisis , Vesículas Secretoras/enzimología , Trombina/metabolismo , Triptasas/metabolismo , Anafilaxia/inducido químicamente , Anafilaxia/enzimología , Anafilaxia/genética , Anafilaxia/patología , Animales , Edema/enzimología , Edema/genética , Edema/patología , Fibrina/genética , Fibrinógeno/genética , Heparina/genética , Humanos , Inmunoglobulina E/metabolismo , Mastocitos/patología , Ratones , Ratones Noqueados , Vesículas Secretoras/genética , Piel/enzimología , Piel/patología , Trombina/genética , Triptasas/genética
10.
EMBO J ; 28(24): 3857-67, 2009 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-19816404

RESUMEN

Rapid activation of p53 by ionizing irradiation is a classic DNA damage response mediated by the ATM kinase. However, the major signalling target and mechanism that lead to p53 stabilization are unknown. We show in this report that ATM induces p53 accumulation by phosphorylating the ubiquitin E3 ligase MDM2. Multiple ATM target sites near the MDM2 RING domain function in a redundant manner to provide robust DNA damage signalling. In the absence of DNA damage, the MDM2 RING domain forms oligomers that mediate p53 poly ubiquitination and proteasomal degradation. Phosphorylation by ATM inhibits RING domain oligomerization, specifically suppressing p53 poly ubiquitination. Blocking MDM2 phosphorylation by alanine substitution of all six phosphorylation sites results in constitutive degradation of p53 after DNA damage. These observations show that ATM controls p53 stability by regulating MDM2 RING domain oligomerization and E3 ligase processivity. Promoting or disrupting E3 oligomerization may be a general mechanism by which signalling kinases regulate ubiquitination reactions, and a potential target for therapeutic intervention.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Secuencia de Aminoácidos , Proteínas de la Ataxia Telangiectasia Mutada , Línea Celular , Línea Celular Tumoral , Cromatografía en Gel , Daño del ADN , Humanos , Datos de Secuencia Molecular , Fosforilación , Complejo de la Endopetidasa Proteasomal/metabolismo , Transducción de Señal , Ubiquitina/química
11.
Proc Biol Sci ; 279(1741): 3347-56, 2012 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-22553092

RESUMEN

Temperatures around hydrothermal vents are highly variable, ranging from near freezing up to 300°C. Nevertheless, animals thrive around vents, some of which live near the known limits of animal thermotolerance. Paralvinella sulfincola, an extremely thermotolerant vent polychaete, and Paralvinella palmiformis, a cooler-adapted congener, are found along the Juan de Fuca Ridge in the northwestern Pacific. We conducted shipboard high-pressure thermotolerance experiments on both species to characterize the physiological adaptations underlying P. sulfincola's pronounced thermotolerance. Quantitative proteomics, expressed sequence tag (EST) libraries and glutathione assays revealed that P. sulfincola (i) exhibited an upregulation in the synthesis and recycling of glutathione with increasing temperature, (ii) downregulated nicotinamide adenine dinucleotide (NADH) and succinate dehydrogenases (key enzymes in oxidative phosphorylation) with increasing temperature, and (iii) maintained elevated levels of heat shock proteins (HSPs) across all treatments. In contrast, P. palmiformis exhibited more typical responses to increasing temperatures (e.g. increasing HSPs at higher temperatures). These data reveal differences in how a mesotolerant and extremely thermotolerant eukaryote respond to thermal stress, and suggest that P. sulfincola's capacity to mitigate oxidative stress via increased synthesis of antioxidants and decreased flux through the mitochondrial electron transport chain enable pronounced thermotolerance. Ultimately, oxidative stress may be the key factor in limiting all metazoan thermotolerance.


Asunto(s)
Regulación de la Expresión Génica , Calor , Respiraderos Hidrotermales , Poliquetos/fisiología , Proteínas/metabolismo , Proteómica , Adaptación Fisiológica , Animales , Respuesta al Choque Térmico , Noroeste de Estados Unidos , Estrés Oxidativo , Proteínas/genética
12.
EMBO Rep ; 11(12): 969-76, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20948544

RESUMEN

We have identified the E3 ligase Traf7 as a direct MyoD1 target and show that cell cycle exit-an early event in muscle differentiation-is linked to decreased Traf7 expression. Depletion of Traf7 accelerates myogenesis, in part through downregulation of nuclear factor-κB (NF-κB) activity. We used a proteomic screen to identify NEMO, the NF-κB essential modulator, as a Traf7-interacting protein. Finally, we show that ubiquitylation of NF-κB essential modulator is regulated exclusively by Traf7 activity in myoblasts. Our results suggest a new mechanism by which MyoD1 function is coupled to NF-κB activity through Traf7, regulating the balance between cell cycle progression and differentiation during myogenesis.


Asunto(s)
Desarrollo de Músculos/genética , Proteína MioD/metabolismo , FN-kappa B/metabolismo , Transcripción Genética , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/genética , Animales , Ciclo Celular/genética , Diferenciación Celular/genética , Ciclina D1/metabolismo , Regulación de la Expresión Génica , Quinasa I-kappa B/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Mioblastos/citología , Mioblastos/metabolismo , Fosforilación , Unión Proteica , Proteína de Retinoblastoma/metabolismo , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/deficiencia , Péptidos y Proteínas Asociados a Receptores de Factores de Necrosis Tumoral/metabolismo , Ubiquitina/metabolismo , Ubiquitinación
13.
PLoS Genet ; 5(3): e1000401, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19266019

RESUMEN

Rotifers of Class Bdelloidea are remarkable in having evolved for millions of years, apparently without males and meiosis. In addition, they are unusually resistant to desiccation and ionizing radiation and are able to repair hundreds of radiation-induced DNA double-strand breaks per genome with little effect on viability or reproduction. Because specific histone H2A variants are involved in DSB repair and certain meiotic processes in other eukaryotes, we investigated the histone H2A genes and proteins of two bdelloid species. Genomic libraries were built and probed to identify histone H2A genes in Adineta vaga and Philodina roseola, species representing two different bdelloid families. The expressed H2A proteins were visualized on SDS-PAGE gels and identified by tandem mass spectrometry. We find that neither the core histone H2A, present in nearly all other eukaryotes, nor the H2AX variant, a ubiquitous component of the eukaryotic DSB repair machinery, are present in bdelloid rotifers. Instead, they are replaced by unusual histone H2A variants of higher mass. In contrast, a species of rotifer belonging to the facultatively sexual, desiccation- and radiation-intolerant sister class of bdelloid rotifers, the monogononts, contains a canonical core histone H2A and appears to lack the bdelloid H2A variant genes. Applying phylogenetic tools, we demonstrate that the bdelloid-specific H2A variants arose as distinct lineages from canonical H2A separate from those leading to the H2AX and H2AZ variants. The replacement of core H2A and H2AX in bdelloid rotifers by previously uncharacterized H2A variants with extended carboxy-terminal tails is further evidence for evolutionary diversity within this class of histone H2A genes and may represent adaptation to unusual features specific to bdelloid rotifers.


Asunto(s)
Proteínas del Helminto/genética , Histonas/genética , Filogenia , Rotíferos/clasificación , Rotíferos/genética , Secuencia de Aminoácidos , Animales , Roturas del ADN de Doble Cadena , Evolución Molecular , Variación Genética , Proteínas del Helminto/química , Proteínas del Helminto/metabolismo , Histonas/química , Histonas/metabolismo , Datos de Secuencia Molecular , Rotíferos/química , Rotíferos/metabolismo , Alineación de Secuencia
14.
Proc Natl Acad Sci U S A ; 106(24): 9779-84, 2009 Jun 16.
Artículo en Inglés | MEDLINE | ID: mdl-19497884

RESUMEN

The Drosophila NF-kappaB transcription factor Relish is an essential regulator of antimicrobial peptide gene induction after gram-negative bacterial infection. Relish is a bipartite NF-kappaB precursor protein, with an N-terminal Rel homology domain and a C-terminal IkappaB-like domain, similar to mammalian p100 and p105. Unlike these mammalian homologs, Relish is endoproteolytically cleaved after infection, allowing the N-terminal NF-kappaB module to translocate to the nucleus. Signal-dependent activation of Relish, including cleavage, requires both the Drosophila IkappaB kinase (IKK) and death-related ced-3/Nedd2-like protein (DREDD), the Drosophila caspase-8 like protease. In this report, we show that the IKK complex controls Relish by direct phosphorylation on serines 528 and 529. Surprisingly, these phosphorylation sites are not required for Relish cleavage, nuclear translocation, or DNA binding. Instead they are critical for recruitment of RNA polymerase II and antimicrobial peptide gene induction, whereas IKK functions noncatalytically to support Dredd-mediated cleavage of Relish.


Asunto(s)
Antiinfecciosos , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiología , Regulación de la Expresión Génica , Quinasa I-kappa B/fisiología , Péptidos/genética , Factores de Transcripción/metabolismo , Animales , Drosophila , Proteínas de Drosophila/química , Epistasis Genética , Quinasa I-kappa B/química , Fosforilación , Regiones Promotoras Genéticas , Serina/metabolismo
15.
Proteomics ; 11(8): 1371-81, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21394914

RESUMEN

Resource (core) facilities have played an ever-increasing role in furnishing the scientific community with specialized instrumentation and expertise for proteomics experiments in a cost-effective manner. The Proteomics Research Group (PRG) of the Association of Biomolecular Resource Facilities (ABRF) has sponsored a number of research studies designed to enable participants to try new techniques and assess their capabilities relative to other laboratories analyzing the same samples. Presented here are results from three PRG studies representing different samples that are typically analyzed in a core facility, ranging from simple protein identification to targeted analyses, and include intentional challenges to reflect realistic studies. The PRG2008 study compares different strategies for the qualitative characterization of proteins, particularly the utility of complementary methods for characterizing truncated protein forms. The use of different approaches for determining quantitative differences for several target proteins in human plasma was the focus of the PRG2009 study. The PRG2010 study explored different methods for determining specific constituents while identifying unforeseen problems that could account for unanticipated results associated with the different samples, and included (15) N-labeled proteins as an additional challenge. These studies provide a valuable educational resource to research laboratories and core facilities, as well as a mechanism for establishing good laboratory practices.


Asunto(s)
Técnicas de Laboratorio Clínico , Proteínas/análisis , Proteómica/métodos , Gonadotropina Coriónica/análisis , Glucógeno Fosforilasa/análisis , Humanos , Antígeno Prostático Específico/análisis , Proteómica/educación , Receptor para Productos Finales de Glicación Avanzada , Receptores Inmunológicos/análisis , Proyectos de Investigación
16.
Biochemistry ; 50(12): 2213-22, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21299233

RESUMEN

Proteolysis plays a key role in regulating the levels and activity of peptide hormones. Characterization of the proteolytic pathways that cleave peptide hormones is of basic interest and can, in some cases, spur the development of novel therapeutics. The lack, however, of an efficient approach to identify endogenous fragments of peptide hormones has hindered the elucidation of these proteolytic pathways. Here, we apply a mass spectrometry (MS) based peptidomics approach to characterize the intestinal fragments of peptide histidine isoleucine (PHI), a hormone that promotes glucose-stimulated insulin secretion (GSIS). Our approach reveals a proteolytic pathway in the intestine that truncates PHI at its C-terminus to produce a PHI fragment that is inactive in a GSIS assay, a result that provides a potential mechanism of PHI regulation in vivo. Differences between these in vivo peptidomics studies and in vitro lysate experiments, which showed N- and C-terminal processing of PHI, underscore the effectiveness of this approach to discover physiologically relevant proteolytic pathways. Moreover, integrating this peptidomics approach with bioassays (i.e., GSIS) provides a general strategy to reveal proteolytic pathways that may regulate the activity of peptide hormones.


Asunto(s)
Hormonas Peptídicas/metabolismo , Proteómica/métodos , Secuencia de Aminoácidos , Animales , Unión Competitiva , Dipeptidil Peptidasa 4/metabolismo , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Hormonas Peptídicas/química , Péptido Hidrolasas/metabolismo , Péptido PHI/química , Péptido PHI/metabolismo , Extractos de Tejidos/metabolismo
17.
Nat Chem Biol ; 5(1): 23-5, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19011639

RESUMEN

Peptide metabolism is a complex process that involves many proteins working in concert. Mass spectrometry-based global peptide profiling of mice lacking dipeptidyl peptidase 4 (DPP4) identified endogenous DPP4 substrates and revealed an unrecognized pathway during proline peptide catabolism that interlinks aminopeptidase and DPP4 activities. Together, these studies elucidate specific aspects of DPP4-regulated metabolism and, more generally, highlight the utility of global peptide profiling for studying peptide metabolism in vivo.


Asunto(s)
Dipeptidil Peptidasa 4/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Secuencia de Aminoácidos , Animales , Dipeptidil Peptidasa 4/química , Dipeptidil Peptidasa 4/genética , Inhibidores de la Dipeptidil-Peptidasa IV , Ratones , Ratones Noqueados , Pliegue de Proteína
18.
Nat Cell Biol ; 4(11): 888-93, 2002 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-12389034

RESUMEN

Nuclear factor-kappa B (NF-kappa B) promotes cell survival by upregulating expression of anti-apoptotic genes, a process that is antagonized by inhibitors of kappa B (I kappa B) factors. The only NF-kappa B family member known to be mutated in human cancer is NF-kappa B2 p100 (ref. 2), a factor with I kappa B activity. Here, we report the isolation from irradiated mouse tumour cells of a complex that induces caspase-8 activity in cell-free assays and identify p100 as an essential component of this complex. Expression of p100 profoundly sensitizes cells to death-receptor-mediated apoptosis through a pathway that is independent of I kappa B-like activity. The carboxyl terminus of p100 contains a death domain that is absent from all known tumour-derived mutants. This death domain mediates recruitment of p100 into death machinery complexes after ligand stimulation and is essential for p100's pro-apoptotic activity. p100 also sensitizes NIH3T3 cells to apoptosis triggered by oncogenic Ras, resulting in a marked inhibition of transformation that is rescued by suppression of endogenous caspase-8. These observations thus identify an I kappa B-independent apoptotic activity of NF-kappa B2 p100 and help explain its unique tumour suppressor role.


Asunto(s)
Apoptosis , FN-kappa B/fisiología , Animales , Caspasa 8 , Caspasas/metabolismo , Muerte Celular , Línea Celular Tumoral , Supervivencia Celular , Sistema Libre de Células , Colorimetría , Cicloheximida/farmacología , Relación Dosis-Respuesta a Droga , Electroforesis en Gel de Poliacrilamida , Activación Enzimática , Técnicas de Transferencia de Gen , Genes Supresores de Tumor , Humanos , Immunoblotting , Espectrometría de Masas , Ratones , Mutación , FN-kappa B/metabolismo , Subunidad p52 de NF-kappa B , Células 3T3 NIH , Plásmidos/metabolismo , Unión Proteica , Estructura Terciaria de Proteína , Retroviridae/genética , Factores de Tiempo , Transfección , Regulación hacia Arriba
19.
J Biol Chem ; 284(44): 30016-23, 2009 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-19740738

RESUMEN

Insulin stimulates the translocation of the glucose transporter GLUT4 from intracellular locations to the plasma membrane in adipose and muscle cells. Prior studies have shown that Akt phosphorylation of the Rab GTPase-activating protein, AS160 (160-kDa Akt substrate; also known as TBC1D4), triggers GLUT4 translocation, most likely by suppressing its Rab GTPase-activating protein activity. However, the regulation of a very similar protein, TBC1D1 (TBC domain family, member 1), which is mainly found in muscle, in insulin-stimulated GLUT4 translocation has been unclear. In the present study, we have identified likely Akt sites of insulin-stimulated phosphorylation of TBC1D1 in C2C12 myotubes. We show that a mutant of TBC1D1, in which several Akt sites have been converted to alanine, is considerably more inhibitory to insulin-stimulated GLUT4 translocation than wild-type TBC1D1. This result thus indicates that similar to AS160, Akt phosphorylation of TBC1D1 enables GLUT4 translocation. We also show that in addition to Akt activation, activation of the AMP-dependent protein kinase partially relieves the inhibition of GLUT4 translocation by TBC1D1. Finally, we show that the R125W variant of TBC1D1, which has been genetically associated with obesity, is equally inhibitory to insulin-stimulated GLUT4 translocation, as is wild-type TBC1D1, and that healthy and type 2 diabetic individuals express approximately the same level of TBC1D1 in biopsies of vastus lateralis muscle. In conclusion, phosphorylation of TBC1D1 is required for GLUT4 translocation. Thus, the regulation of TBC1D1 resembles that of its paralog, AS160.


Asunto(s)
Transportador de Glucosa de Tipo 4/metabolismo , Insulina/farmacología , Proteínas Nucleares/metabolismo , Células 3T3-L1 , Animales , Diabetes Mellitus Tipo 2/metabolismo , Proteínas Activadoras de GTPasa/metabolismo , Ratones , Músculo Esquelético/química , Proteínas Nucleares/análisis , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo
20.
Nat Biotechnol ; 25(4): 454-64, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17401360

RESUMEN

Enzymatic removal of blood group ABO antigens to develop universal red blood cells (RBCs) was a pioneering vision originally proposed more than 25 years ago. Although the feasibility of this approach was demonstrated in clinical trials for group B RBCs, a major obstacle in translating this technology to clinical practice has been the lack of efficient glycosidase enzymes. Here we report two bacterial glycosidase gene families that provide enzymes capable of efficient removal of A and B antigens at neutral pH with low consumption of recombinant enzymes. The crystal structure of a member of the alpha-N-acetylgalactosaminidase family reveals an unusual catalytic mechanism involving NAD+. The enzymatic conversion processes we describe hold promise for achieving the goal of producing universal RBCs, which would improve the blood supply while enhancing the safety of clinical transfusions.


Asunto(s)
Bacterias/enzimología , Eritrocitos/metabolismo , Glicósido Hidrolasas/metabolismo , Sistema del Grupo Sanguíneo ABO/química , Sitios de Unión , Tipificación y Pruebas Cruzadas Sanguíneas , Catálisis , Cromatografía en Capa Delgada , Citometría de Flujo , Humanos , Concentración de Iones de Hidrógeno , Cinética , Datos de Secuencia Molecular , Células Procariotas/enzimología , Estructura Secundaria de Proteína , Especificidad por Sustrato , Volumetría , alfa-N-Acetilgalactosaminidasa/química
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA