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

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Cell ; 145(4): 513-28, 2011 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-21565611

RESUMO

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.


Assuntos
Doenças Renais Císticas/genética , Proteínas de Membrana/genética , Transdução de Sinais , Animais , Ataxina-10 , Centrossomo/metabolismo , Cílios/metabolismo , Transtornos da Motilidade Ciliar/genética , Encefalocele/genética , Proteínas Hedgehog/metabolismo , Humanos , Doenças Renais Císticas/metabolismo , Camundongos , Células NIH 3T3 , Proteínas do Tecido Nervoso/genética , Doenças Renais Policísticas/genética , Retinose Pigmentar , Peixe-Zebra
2.
J Biol Chem ; 296: 100194, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33334891

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Proteases Específicas de Ubiquitina/metabolismo , Ubiquitina/metabolismo , Proteínas de Ciclo Celular/química , Proteínas de Ciclo Celular/genética , Cromatina/genética , Proteínas Cromossômicas não Histona/química , Proteínas Cromossômicas não Histona/genética , Segregação de Cromossomos , Reparo do DNA , Replicação do DNA , Células HCT116 , Células HeLa , Humanos , Domínios e Motivos de Interação entre Proteínas , Proteases Específicas de Ubiquitina/química , Proteases Específicas de Ubiquitina/genética , Ubiquitinação , Coesinas
3.
J Biol Chem ; 294(22): 8760-8772, 2019 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-31010829

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Mitose , Proteômica , Fatores de Processamento de RNA/metabolismo , Proteínas de Ligação a RNA/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Cromatina/metabolismo , Proteínas Cromossômicas não Histona/genética , Proteínas de Ligação a DNA/antagonistas & inibidores , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Humanos , Microscopia de Fluorescência , Ligação Proteica , Mapas de Interação de Proteínas , Interferência de RNA , Fatores de Processamento de RNA/antagonistas & inibidores , Fatores de Processamento de RNA/genética , RNA Interferente Pequeno/metabolismo , Proteínas de Ligação a RNA/antagonistas & inibidores , Proteínas de Ligação a RNA/genética , Coesinas
4.
Genes Dev ; 24(19): 2180-93, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20889716

RESUMO

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.


Assuntos
Cílios/metabolismo , Proteínas de Ligação ao GTP/metabolismo , Fosfatidilinositóis/metabolismo , Proteínas/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Sequência de Aminoácidos , Animais , Linhagem Celular , Células Cultivadas , Proteínas Hedgehog/metabolismo , Humanos , Peptídeos e Proteínas de Sinalização Intercelular , Peptídeos e Proteínas de Sinalização Intracelular , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Complexos Multiproteicos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Transporte Proteico , Alinhamento de Sequência
5.
J Biol Chem ; 290(37): 22795-804, 2015 Sep 11.
Artigo em Inglês | MEDLINE | ID: mdl-26221039

RESUMO

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.


Assuntos
Reparo de Erro de Pareamento de DNA , DNA/metabolismo , Histona Desacetilases/metabolismo , Proteína 2 Homóloga a MutS/metabolismo , Acetilação , DNA/genética , Células HeLa , Histona Desacetilases/genética , Humanos , Proteína 2 Homóloga a MutS/genética
6.
J Biol Chem ; 289(11): 7788-98, 2014 Mar 14.
Artigo em Inglês | MEDLINE | ID: mdl-24448800

RESUMO

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.


Assuntos
Bacteriófago P1/metabolismo , Proteínas de Escherichia coli/metabolismo , Elongação Traducional da Cadeia Peptídica , Fator Tu de Elongação de Peptídeos/metabolismo , Proteínas Virais/metabolismo , Motivos de Aminoácidos , Antibacterianos/química , Sítios de Ligação , Proliferação de Células , Escherichia coli/metabolismo , Regulação Bacteriana da Expressão Gênica , Espectrometria de Massas , Simulação de Acoplamento Molecular , Fosforilação , Ligação Proteica , Processamento de Proteína Pós-Traducional , Estrutura Terciária de Proteína , Piridonas/química , RNA Mensageiro/metabolismo , Proteínas Recombinantes/química , Treonina/química
7.
Proc Natl Acad Sci U S A ; 109(31): 12710-5, 2012 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-22802624

RESUMO

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.


Assuntos
Proteínas de Bactérias/metabolismo , Imunidade Inata , MAP Quinase Quinase Quinases/metabolismo , Sistema de Sinalização das MAP Quinases , Serina O-Acetiltransferase/metabolismo , Yersinia pestis/enzimologia , Acetilação , Animais , Proteínas de Bactérias/imunologia , Drosophila melanogaster , Células HEK293 , Humanos , MAP Quinase Quinase Quinases/imunologia , NF-kappa B/imunologia , NF-kappa B/metabolismo , Peste/imunologia , Peste/metabolismo , Serina O-Acetiltransferase/imunologia , Yersinia pestis/imunologia , Yersinia pestis/patogenicidade
8.
J Proteome Res ; 12(10): 4351-65, 2013 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-24004147

RESUMO

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.


Assuntos
Proteínas Sanguíneas/química , Espectrometria de Massas em Tandem/métodos , Proteínas Sanguíneas/isolamento & purificação , Proteínas Sanguíneas/metabolismo , Humanos , Imunoprecipitação , Mapeamento de Peptídeos , Proteômica , Padrões de Referência , Reprodutibilidade dos Testes , Sensibilidade e Especificidade , Espectrometria de Massas em Tandem/instrumentação , Espectrometria de Massas em Tandem/normas
9.
J Biol Chem ; 287(11): 7834-44, 2012 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-22235124

RESUMO

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.


Assuntos
Coagulação Sanguínea , Fibrina/metabolismo , Fibrinogênio/metabolismo , Heparina/metabolismo , Mastócitos/enzimologia , Proteólise , Vesículas Secretórias/enzimologia , Trombina/metabolismo , Triptases/metabolismo , Anafilaxia/induzido quimicamente , Anafilaxia/enzimologia , Anafilaxia/genética , Anafilaxia/patologia , Animais , Edema/enzimologia , Edema/genética , Edema/patologia , Fibrina/genética , Fibrinogênio/genética , Heparina/genética , Humanos , Imunoglobulina E/metabolismo , Mastócitos/patologia , Camundongos , Camundongos Knockout , Vesículas Secretórias/genética , Pele/enzimologia , Pele/patologia , Trombina/genética , Triptases/genética
10.
EMBO J ; 28(24): 3857-67, 2009 Dec 16.
Artigo em Inglês | MEDLINE | ID: mdl-19816404

RESUMO

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.


Assuntos
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Sequência de Aminoácidos , Proteínas Mutadas de Ataxia Telangiectasia , Linhagem Celular , Linhagem Celular Tumoral , Cromatografia em Gel , Dano ao DNA , Humanos , Dados de Sequência Molecular , Fosforilação , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Ubiquitina/química
11.
Proc Biol Sci ; 279(1741): 3347-56, 2012 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-22553092

RESUMO

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.


Assuntos
Regulação da Expressão Gênica , Temperatura Alta , Fontes Hidrotermais , Poliquetos/fisiologia , Proteínas/metabolismo , Proteômica , Adaptação Fisiológica , Animais , Resposta ao Choque Térmico , Noroeste dos Estados Unidos , Estresse Oxidativo , Proteínas/genética
12.
EMBO Rep ; 11(12): 969-76, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20948544

RESUMO

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.


Assuntos
Desenvolvimento Muscular/genética , Proteína MyoD/metabolismo , NF-kappa B/metabolismo , Transcrição Gênica , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/genética , Animais , Ciclo Celular/genética , Diferenciação Celular/genética , Ciclina D1/metabolismo , Regulação da Expressão Gênica , Quinase I-kappa B/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Camundongos , Mioblastos/citologia , Mioblastos/metabolismo , Fosforilação , Ligação Proteica , Proteína do Retinoblastoma/metabolismo , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/deficiência , Peptídeos e Proteínas Associados a Receptores de Fatores de Necrose Tumoral/metabolismo , Ubiquitina/metabolismo , Ubiquitinação
13.
PLoS Genet ; 5(3): e1000401, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19266019

RESUMO

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.


Assuntos
Proteínas de Helminto/genética , Histonas/genética , Filogenia , Rotíferos/classificação , Rotíferos/genética , Sequência de Aminoácidos , Animais , Quebras de DNA de Cadeia Dupla , Evolução Molecular , Variação Genética , Proteínas de Helminto/química , Proteínas de Helminto/metabolismo , Histonas/química , Histonas/metabolismo , Dados de Sequência Molecular , Rotíferos/química , Rotíferos/metabolismo , Alinhamento de Sequência
14.
Proc Natl Acad Sci U S A ; 106(24): 9779-84, 2009 Jun 16.
Artigo em Inglês | MEDLINE | ID: mdl-19497884

RESUMO

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.


Assuntos
Anti-Infecciosos , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/fisiologia , Regulação da Expressão Gênica , Quinase I-kappa B/fisiologia , Peptídeos/genética , Fatores de Transcrição/metabolismo , Animais , Drosophila , Proteínas de Drosophila/química , Epistasia Genética , Quinase I-kappa B/química , Fosforilação , Regiões Promotoras Genéticas , Serina/metabolismo
15.
Proteomics ; 11(8): 1371-81, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21394914

RESUMO

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.


Assuntos
Técnicas de Laboratório Clínico , Proteínas/análise , Proteômica/métodos , Gonadotropina Coriônica/análise , Glicogênio Fosforilase/análise , Humanos , Antígeno Prostático Específico/análise , Proteômica/educação , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/análise , Projetos de Pesquisa
16.
Biochemistry ; 50(12): 2213-22, 2011 Mar 29.
Artigo em Inglês | MEDLINE | ID: mdl-21299233

RESUMO

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.


Assuntos
Hormônios Peptídicos/metabolismo , Proteômica/métodos , Sequência de Aminoácidos , Animais , Ligação Competitiva , Dipeptidil Peptidase 4/metabolismo , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Hormônios Peptídicos/química , Peptídeo Hidrolases/metabolismo , Peptídeo PHI/química , Peptídeo PHI/metabolismo , Extratos de Tecidos/metabolismo
17.
Nat Chem Biol ; 5(1): 23-5, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19011639

RESUMO

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.


Assuntos
Dipeptidil Peptidase 4/metabolismo , Perfilação da Expressão Gênica , Regulação da Expressão Gênica/fisiologia , Sequência de Aminoácidos , Animais , Dipeptidil Peptidase 4/química , Dipeptidil Peptidase 4/genética , Inibidores da Dipeptidil Peptidase IV , Camundongos , Camundongos Knockout , Dobramento de Proteína
18.
Nat Cell Biol ; 4(11): 888-93, 2002 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-12389034

RESUMO

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.


Assuntos
Apoptose , NF-kappa B/fisiologia , Animais , Caspase 8 , Caspases/metabolismo , Morte Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Sistema Livre de Células , Colorimetria , Cicloeximida/farmacologia , Relação Dose-Resposta a Droga , Eletroforese em Gel de Poliacrilamida , Ativação Enzimática , Técnicas de Transferência de Genes , Genes Supressores de Tumor , Humanos , Immunoblotting , Espectrometria de Massas , Camundongos , Mutação , NF-kappa B/metabolismo , Subunidade p52 de NF-kappa B , Células NIH 3T3 , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Retroviridae/genética , Fatores de Tempo , Transfecção , Regulação para Cima
19.
J Biol Chem ; 284(44): 30016-23, 2009 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-19740738

RESUMO

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.


Assuntos
Transportador de Glucose Tipo 4/metabolismo , Insulina/farmacologia , Proteínas Nucleares/metabolismo , Células 3T3-L1 , Animais , Diabetes Mellitus Tipo 2/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Camundongos , Músculo Esquelético/química , Proteínas Nucleares/análise , Fosforilação , Transporte Proteico/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-akt/metabolismo
20.
Nat Biotechnol ; 25(4): 454-64, 2007 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-17401360

RESUMO

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.


Assuntos
Bactérias/enzimologia , Eritrócitos/metabolismo , Glicosídeo Hidrolases/metabolismo , Sistema ABO de Grupos Sanguíneos/química , Sítios de Ligação , Tipagem e Reações Cruzadas Sanguíneas , Catálise , Cromatografia em Camada Fina , Citometria de Fluxo , Humanos , Concentração de Íons de Hidrogênio , Cinética , Dados de Sequência Molecular , Células Procarióticas/enzimologia , Estrutura Secundária de Proteína , Especificidade por Substrato , Titulometria , alfa-N-Acetilgalactosaminidase/química
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA