Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 40
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 15(1): 3785, 2024 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-38710674

RESUMO

Mutations in human isocitrate dehydrogenase 1 (IDH1) drive tumor formation in a variety of cancers by replacing its conventional activity with a neomorphic activity that generates an oncometabolite. Little is understood of the mechanistic differences among tumor-driving IDH1 mutants. We previously reported that the R132Q mutant unusually preserves conventional activity while catalyzing robust oncometabolite production, allowing an opportunity to compare these reaction mechanisms within a single active site. Here, we employ static and dynamic structural methods and observe that, compared to R132H, the R132Q active site adopts a conformation primed for catalysis with optimized substrate binding and hydride transfer to drive improved conventional and neomorphic activity over R132H. This active site remodeling reveals a possible mechanism of resistance to selective mutant IDH1 therapeutic inhibitors. This work enhances our understanding of fundamental IDH1 mechanisms while pinpointing regions for improving inhibitor selectivity.


Assuntos
Domínio Catalítico , Isocitrato Desidrogenase , Mutação , Isocitrato Desidrogenase/genética , Isocitrato Desidrogenase/metabolismo , Humanos , Cinética , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores Enzimáticos/farmacologia
2.
Res Sq ; 2024 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-38464189

RESUMO

Mutations in human isocitrate dehydrogenase 1 (IDH1) drive tumor formation in a variety of cancers by replacing its conventional activity with a neomorphic activity that generates an oncometabolite. Little is understood of the mechanistic differences among tumor-driving IDH1 mutants. We previously reported that the R132Q mutant uniquely preserves conventional activity while catalyzing robust oncometabolite production, allowing an opportunity to compare these reaction mechanisms within a single active site. Here, we employed static and dynamic structural methods and found that, compared to R132H, the R132Q active site adopted a conformation primed for catalysis with optimized substrate binding and hydride transfer to drive improved conventional and neomorphic activity over R132H. This active site remodeling revealed a possible mechanism of resistance to selective mutant IDH1 therapeutic inhibitors. This work enhances our understanding of fundamental IDH1 mechanisms while pinpointing regions for improving inhibitor selectivity.

3.
bioRxiv ; 2024 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-38260668

RESUMO

Mutations in human isocitrate dehydrogenase 1 (IDH1) drive tumor formation in a variety of cancers by replacing its conventional activity with a neomorphic activity that generates an oncometabolite. Little is understood of the mechanistic differences among tumor-driving IDH1 mutants. We previously reported that the R132Q mutant uniquely preserves conventional activity while catalyzing robust oncometabolite production, allowing an opportunity to compare these reaction mechanisms within a single active site. Here, we employed static and dynamic structural methods and found that, compared to R132H, the R132Q active site adopted a conformation primed for catalysis with optimized substrate binding and hydride transfer to drive improved conventional and neomorphic activity over R132H. This active site remodeling revealed a possible mechanism of resistance to selective mutant IDH1 therapeutic inhibitors. This work enhances our understanding of fundamental IDH1 mechanisms while pinpointing regions for improving inhibitor selectivity.

4.
J Biol Chem ; 298(9): 102349, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35934050

RESUMO

Many transcription factors contain intrinsically disordered transcription activation domains (TADs), which mediate interactions with coactivators to activate transcription. Historically, DNA-binding domains and TADs have been considered as modular units, but recent studies have shown that TADs can influence DNA binding. Whether these results can be generalized to more TADs is not clear. Here, we biophysically characterized the NFκB p50/RelA heterodimer including the RelA TAD and investigated the TAD's influence on NFκB-DNA interactions. In solution, we show the RelA TAD is disordered but compact, with helical tendency in two regions that interact with coactivators. We determined that the presence of the TAD increased the stoichiometry of NFκB-DNA complexes containing promoter DNA sequences with tandem κB recognition motifs by promoting the binding of NFκB dimers in excess of the number of κB sites. In addition, we measured the binding affinity of p50/RelA for DNA containing tandem κB sites and single κB sites. While the presence of the TAD enhanced the binding affinity of p50/RelA for all κB sequences tested, it also increased the affinity for nonspecific DNA sequences by over 10-fold, leading to an overall decrease in specificity for κB DNA sequences. In contrast, previous studies have generally reported that TADs decrease DNA-binding affinity and increase sequence specificity. Our results reveal a novel function of the RelA TAD in promoting binding to nonconsensus DNA, which sheds light on previous observations of extensive nonconsensus DNA binding by NFκB in vivo in response to strong inflammatory signals.


Assuntos
Subunidade p50 de NF-kappa B , Fator de Transcrição RelA , Ativação Transcricional , Sequência de Bases , DNA/química , Subunidade p50 de NF-kappa B/química , Subunidade p50 de NF-kappa B/genética , Ligação Proteica , Domínios Proteicos , Multimerização Proteica , Fator de Transcrição RelA/química , Fator de Transcrição RelA/genética
5.
Nucleic Acids Res ; 49(19): 11211-11223, 2021 11 08.
Artigo em Inglês | MEDLINE | ID: mdl-34614173

RESUMO

Binding and unbinding of transcription factors to DNA are kinetically controlled to regulate the transcriptional outcome. Control of the release of the transcription factor NF-κB from DNA is achieved through accelerated dissociation by the inhibitor protein IκBα. Using single-molecule FRET, we observed a continuum of conformations of NF-κB in free and DNA-bound states interconverting on the subseconds to minutes timescale, comparable to in vivo binding on the seconds timescale, suggesting that structural dynamics directly control binding kinetics. Much of the DNA-bound NF-κB is partially bound, allowing IκBα invasion to facilitate DNA dissociation. IκBα induces a locked conformation where the DNA-binding domains of NF-κB are too far apart to bind DNA, whereas a loss-of-function IκBα mutant retains the NF-κB conformational ensemble. Overall, our results suggest a novel mechanism with a continuum of binding modes for controlling association and dissociation of transcription factors.


Assuntos
DNA/genética , Interferons/genética , Inibidor de NF-kappaB alfa/genética , Fator de Transcrição RelA/genética , Transcrição Gênica , Animais , Avidina/química , Sítios de Ligação , Biotina/química , DNA/metabolismo , Transferência Ressonante de Energia de Fluorescência , Regulação da Expressão Gênica , Humanos , Proteínas Imobilizadas/química , Proteínas Imobilizadas/genética , Proteínas Imobilizadas/metabolismo , Interferons/química , Interferons/metabolismo , Sequências Repetidas Invertidas , Camundongos , Simulação de Dinâmica Molecular , Inibidor de NF-kappaB alfa/química , Inibidor de NF-kappaB alfa/metabolismo , Ligação Proteica , Conformação Proteica em alfa-Hélice , Conformação Proteica em Folha beta , Domínios e Motivos de Interação entre Proteínas , Imagem Individual de Molécula/métodos , Fator de Transcrição RelA/química , Fator de Transcrição RelA/metabolismo
6.
Structure ; 29(8): 781-782, 2021 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-34358463

RESUMO

The bacterial Sec translocase transports unfolded proteins across membranes. In this issue of Structure, Krishnamurthy et al. (2021) report a nexus of conformational dynamics in the translocase motor protein, SecA. Their findings shed light on the Sec activation mechanism and suggest a general role for multi-level dynamics in protein functions.


Assuntos
Proteínas de Bactérias , Proteínas de Membrana Transportadoras , Adenosina Trifosfatases/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Transporte Proteico , Canais de Translocação SEC/genética , Proteínas SecA
7.
Sci Rep ; 9(1): 15092, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31641149

RESUMO

We used hydrogen-deuterium exchange mass spectrometry (HDX-MS) to obtain a comprehensive view of transporter dynamics (85.8% sequence coverage) occurring throughout the multidrug efflux transporter P-glycoprotein (P-gp) in three distinct conformational states: predominantly inward-facing apo P-gp, pre-hydrolytic (E552Q/E1197Q) P-gp bound to Mg+2-ATP, and outward-facing P-gp bound to Mg+2-ADP-VO4-3. Nucleotide affinity was measured with bio-layer interferometry (BLI), which yielded kinetics data that fit a two Mg+2-ATP binding-site model. This model has one high affinity site (3.2 ± 0.3 µM) and one low affinity site (209 ± 25 µM). Comparison of deuterium incorporation profiles revealed asymmetry between the changes undergone at the critical interfaces where nucleotide binding domains (NBDs) contact intracellular helices (ICHs). In the pre-hydrolytic state, both interfaces between ICHs and NBDs decreased exchange to similar extents relative to inward-facing P-gp. In the outward-facing state, the ICH-NBD1 interface showed decreased exchange, while the ICH-NBD2 interface showed less of an effect. The extracellular loops (ECLs) showed reduced deuterium uptake in the pre-hydrolytic state, consistent with an occluded conformation. While in the outward-facing state, increased ECL exchange corresponding to EC domain opening was observed. These findings point toward asymmetry between both NBDs, and they suggest that pre-hydrolytic P-gp occupies an occluded conformation.


Assuntos
Transportadores de Cassetes de Ligação de ATP/química , Simulação de Dinâmica Molecular , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Trifosfato de Adenosina/metabolismo , Animais , Sítios de Ligação , Magnésio/metabolismo , Camundongos , Mutação de Sentido Incorreto , Ligação Proteica
8.
Biochemistry ; 58(21): 2555-2563, 2019 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-31033276

RESUMO

The NFκB transcription factor family members RelA, p50, and cRel form homo- and heterodimers that are inhibited by IκBα, IκBß, and IκBε. These NFκB family members have diverse biological functions, and their expression profiles differ, leading to different concentrations in different tissue types. Here we present definitive biophysical measurements of the NFκB dimer affinities and inhibitor affinities to better understand dimer exchange and how the presence of inhibitors may alter the equilibrium concentrations of NFκB dimers in the cellular context. Fluorescence anisotropy binding experiments were performed at low concentrations to mimic intracellular concentrations. We report binding affinities much stronger than those that had been previously reported by non-equilibrium gel shift and analytical ultracentrifugation assays. The results reveal a wide range of NFκB dimer affinities and a strong preference of each IκB for a small subset of NFκB dimers. Once the preferred IκB is bound, dimer exchange no longer occurs over a period of days. A mathematical model of the cellular distribution of these canonical NFκB transcription factors based on the revised binding affinities recapitulates intracellular observations and provides simple, precise explanations for observed cellular phenomena.


Assuntos
Inibidor de NF-kappaB alfa/química , Subunidade p50 de NF-kappa B/química , Multimerização Proteica , Fator de Transcrição RelA/química , Animais , Citoplasma/metabolismo , Fibroblastos/metabolismo , Polarização de Fluorescência , Meia-Vida , Camundongos , Modelos Teóricos , Ligação Proteica , Proteólise , Fator de Transcrição RelA/antagonistas & inibidores
9.
Nat Commun ; 9(1): 4536, 2018 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-30382093

RESUMO

Stromal interaction molecule 1 (STIM1) monitors ER-luminal Ca2+ levels to maintain cellular Ca2+ balance and to support Ca2+ signalling. The prevailing view has been that STIM1 senses reduced ER Ca2+ through dissociation of bound Ca2+ from a single EF-hand site, which triggers a dramatic loss of secondary structure and dimerization of the STIM1 luminal domain. Here we find that the STIM1 luminal domain has 5-6 Ca2+-binding sites, that binding at these sites is energetically coupled to binding at the EF-hand site, and that Ca2+ dissociation controls a switch to a second structured conformation of the luminal domain rather than protein unfolding. Importantly, the other luminal-domain Ca2+-binding sites interact with the EF-hand site to control physiological activation of STIM1 in cells. These findings fundamentally revise our understanding of physiological Ca2+ sensing by STIM1, and highlight molecular mechanisms that govern the Ca2+ threshold for activation and the steep Ca2+ concentration dependence.


Assuntos
Cálcio/metabolismo , Retículo Endoplasmático/metabolismo , Molécula 1 de Interação Estromal/química , Molécula 1 de Interação Estromal/metabolismo , Animais , Sítios de Ligação , Calorimetria , Cisteína/metabolismo , Medição da Troca de Deutério , Fluorescência , Células HeLa , Humanos , Camundongos , Mutação/genética , Domínios Proteicos , Estrutura Secundária de Proteína , Solubilidade , Relação Estrutura-Atividade
10.
Protein Sci ; 27(9): 1624-1635, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-30133030

RESUMO

The ankyrin repeat (AR) structure is a common protein-protein interaction motif and ankyrin repeat proteins comprise a vast family across a large array of different taxa. Natural AR proteins adopt a conserved fold comprised of several repeats with the N- and C-terminal repeats generally being of more divergent sequences. Obtaining experimental crystal structures for natural ankyrin repeat domains (ARD) can be difficult and often requires complexation with a binding partner. Homology modeling is an attractive method for creating a model of AR proteins due to the highly conserved fold; however, modeling the divergent N- and C-terminal "capping" repeats remains a challenge. We show here that amide hydrogen/deuterium exchange mass spectrometry (HDX-MS), which reports on the presence of secondary structural elements and "foldedness," can aid in the refinement and selection of AR protein homology models when multiple templates are identified with variations between them localizing to these terminal repeats. We report a homology model for the AR protein IκBε from three different templates and use HDX-MS to establish the presence of a seventh AR at the C-terminus identified by only one of the three templates used for modeling.


Assuntos
Proteínas I-kappa B/química , Proteínas Proto-Oncogênicas/química , Repetição de Anquirina , Medição da Troca de Deutério , Humanos , Espectrometria de Massas , Modelos Moleculares , Conformação Proteica
11.
Nat Struct Mol Biol ; 25(8): 687-697, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30076409

RESUMO

The bromodomain and extra-terminal motif (BET) protein BRD4 binds to acetylated histones at enhancers and promoters via its bromodomains (BDs) to regulate transcriptional elongation. In human colorectal cancer cells, we found that BRD4 was recruited to enhancers that were co-occupied by mutant p53 and supported the synthesis of enhancer-directed transcripts (eRNAs) in response to chronic immune signaling. BRD4 selectively associated with eRNAs that were produced from BRD4-bound enhancers. Using biochemical and biophysical methods, we found that BRD4 BDs function cooperatively as docking sites for eRNAs and that the BDs of BRD2, BRD3, BRDT, BRG1, and BRD7 directly interact with eRNAs. BRD4-eRNA interactions increased BRD4 binding to acetylated histones in vitro and augmented BRD4 enhancer recruitment and transcriptional cofactor activities. Our results suggest a mechanism by which eRNAs are directly involved in gene regulation by modulating enhancer interactions and transcriptional functions of BRD4.


Assuntos
Cromatina/metabolismo , Proteínas Nucleares/metabolismo , RNA/metabolismo , Fatores de Transcrição/metabolismo , Ativação Transcricional , Acetilação , Proteínas de Ciclo Celular , Elementos Facilitadores Genéticos , Histonas/metabolismo , Humanos , Proteínas Nucleares/genética , Ligação Proteica , Domínios Proteicos , Transdução de Sinais , Fatores de Transcrição/genética , Proteína Supressora de Tumor p53/metabolismo
12.
J Mol Biol ; 430(10): 1510-1520, 2018 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-29625198

RESUMO

The main nuclear factor kappa B transcription factor family members RelA-p50 heterodimer and RelA homodimer have different biological functions and show different transcriptional activation profiles. To investigate whether the two family members adopt a similar conformation in their free states, we performed hydrogen-deuterium exchange mass spectrometry, all-atom molecular dynamics simulations, and stopped-flow binding kinetics experiments. Surprisingly, the N-terminal DNA-binding domains adopt an open conformation in RelA-p50 but a closed conformation in RelA homodimer. Both hydrogen-deuterium exchange mass spectrometry and molecular dynamics simulations indicate the formation of an interface between the N-terminal DNA-binding domains only in the RelA homodimer. Such an interface would be expected to impede DNA binding, and stopped-flow binding kinetics show that association of DNA is slower for the homodimer as compared to the heterodimer. Our results show that the DNA-binding cavity in the RelA-p50 heterodimer is open for DNA binding, whereas in the RelA homodimer, it is occluded.


Assuntos
Complexos Multiproteicos/química , Subunidade p50 de NF-kappa B/química , Subunidade p50 de NF-kappa B/metabolismo , Fator de Transcrição RelA/química , Fator de Transcrição RelA/metabolismo , Animais , Sítios de Ligação , DNA/metabolismo , Medição da Troca de Deutério , Camundongos , Modelos Moleculares , Simulação de Dinâmica Molecular , Ligação Proteica , Conformação Proteica , Multimerização Proteica
13.
Curr Opin Struct Biol ; 48: 68-73, 2018 02.
Artigo em Inglês | MEDLINE | ID: mdl-29101782

RESUMO

Natural protein molecules are exceptional polymers. Encoded in apparently random strings of amino-acids, these objects perform clear physical tasks that are rare to find by simple chance. Accurate folding, specific binding, powerful catalysis, are examples of basic chemical activities that the great majority of polypeptides do not display, and are thought to be the outcome of the natural history of proteins. Function, a concept genuine to Biology, is at the core of evolution and often conflicts with the physical constraints. Locating the frustration between discrepant goals in a recurrent system leads to fundamental insights about the chances and necessities that shape the encoding of biological information.


Assuntos
Aminoácidos/química , Simulação de Dinâmica Molecular , Proteínas/química , Sequência de Aminoácidos , Animais , Biocatálise , Evolução Molecular , Humanos , Cinética , Ligação Proteica , Dobramento de Proteína , Proteínas/fisiologia , Relação Estrutura-Atividade , Termodinâmica
14.
Nat Commun ; 8(1): 781, 2017 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-28974687

RESUMO

Nipah virus is an emergent paramyxovirus that causes deadly encephalitis and respiratory infections in humans. Two glycoproteins coordinate the infection of host cells, an attachment protein (G), which binds to cell surface receptors, and a fusion (F) protein, which carries out the process of virus-cell membrane fusion. The G protein binds to ephrin B2/3 receptors, inducing G conformational changes that trigger F protein refolding. Using an optical approach based on second harmonic generation, we show that monomeric and dimeric receptors activate distinct conformational changes in G. The monomeric receptor-induced changes are not detected by conformation-sensitive monoclonal antibodies or through electron microscopy analysis of G:ephrinB2 complexes. However, hydrogen/deuterium exchange experiments confirm the second harmonic generation observations and reveal allosteric changes in the G receptor binding and F-activating stalk domains, providing insights into the pathway of receptor-activated virus entry.Nipah virus causes encephalitis in humans. Here the authors use a multidisciplinary approach to study the binding of the viral attachment protein G to its host receptor ephrinB2 and show that monomeric and dimeric receptors activate distinct conformational changes in G and discuss implications for receptor-activated virus entry.


Assuntos
Efrina-B2/metabolismo , Vírus Nipah/metabolismo , Proteínas do Envelope Viral/metabolismo , Regulação Alostérica , Anticorpos Monoclonais/metabolismo , Medição da Troca de Deutério , Células HEK293 , Humanos , Espectrometria de Massas , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestrutura , Coloração Negativa , Ligação Proteica , Multimerização Proteica
15.
Nat Commun ; 8(1): 1171, 2017 10 27.
Artigo em Inglês | MEDLINE | ID: mdl-29079793

RESUMO

Small ubiquitin-like modifier (SUMO) modification regulates numerous cellular processes. Unlike ubiquitin, detection of endogenous SUMOylated proteins is limited by the lack of naturally occurring protease sites in the C-terminal tail of SUMO proteins. Proteome-wide detection of SUMOylation sites on target proteins typically requires ectopic expression of mutant SUMOs with introduced tryptic sites. Here, we report a method for proteome-wide, site-level detection of endogenous SUMOylation that uses α-lytic protease, WaLP. WaLP digestion of SUMOylated proteins generates peptides containing SUMO-remnant diglycyl-lysine (KGG) at the site of SUMO modification. Using previously developed immuno-affinity isolation of KGG-containing peptides followed by mass spectrometry, we identified 1209 unique endogenous SUMO modification sites. We also demonstrate the impact of proteasome inhibition on ubiquitin and SUMO-modified proteomes using parallel quantitation of ubiquitylated and SUMOylated peptides. This methodological advancement enables determination of endogenous SUMOylated proteins under completely native conditions.


Assuntos
Serina Endopeptidases/química , Proteínas Modificadoras Pequenas Relacionadas à Ubiquitina/metabolismo , Células HeLa , Humanos , Lisina/química , Espectrometria de Massas , Mutação , Peptídeos/química , Ligação Proteica , Domínios Proteicos , Processamento de Proteína Pós-Traducional , Proteoma , Proteômica , Transdução de Sinais , Sumoilação , Tripsina/química , Ubiquitina/química
16.
Sci Rep ; 7(1): 3385, 2017 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-28611361

RESUMO

Although trypsin-like serine proteases have flexible surface-exposed loops and are known to adopt higher and lower activity conformations, structural determinants for the different conformations have remained largely obscure. The trypsin-like serine protease, urokinase-type plasminogen activator (uPA), is central in tissue remodeling processes and also strongly implicated in tumor metastasis. We solved five X-ray crystal structures of murine uPA (muPA) in the absence and presence of allosteric molecules and/or substrate-like molecules. The structure of unbound muPA revealed an unsuspected non-chymotrypsin-like protease conformation in which two ß-strands in the core of the protease domain undergoes a major antiparallel-to-parallel conformational transition. We next isolated two anti-muPA nanobodies; an active-site binding nanobody and an allosteric nanobody. Crystal structures of the muPA:nanobody complexes and hydrogen-deuterium exchange mass spectrometry revealed molecular insights about molecular factors controlling the antiparallel-to-parallel equilibrium in muPA. Together with muPA activity assays, the data provide valuable insights into regulatory mechanisms and conformational flexibility of uPA and trypsin-like serine proteases in general.


Assuntos
Conformação Proteica , Anticorpos de Domínio Único/química , Anticorpos de Domínio Único/metabolismo , Ativador de Plasminogênio Tipo Uroquinase/química , Ativador de Plasminogênio Tipo Uroquinase/metabolismo , Animais , Especificidade de Anticorpos , Sítios de Ligação , Domínio Catalítico , Cristalografia por Raios X , Camundongos , Modelos Moleculares
17.
Proc Natl Acad Sci U S A ; 114(8): 1916-1921, 2017 02 21.
Artigo em Inglês | MEDLINE | ID: mdl-28167786

RESUMO

Stress-response transcription factors such as NFκB turn on hundreds of genes and must have a mechanism for rapid cessation of transcriptional activation. We recently showed that the inhibitor of NFκB signaling, IκBα, dramatically accelerates the dissociation of NFκB from transcription sites, a process we have called "stripping." To test the role of the IκBα C-terminal PEST (rich in proline, glutamic acid, serine, and threonine residues) sequence in NFκB stripping, a mutant IκBα was generated in which five acidic PEST residues were mutated to their neutral analogs. This IκBα(5xPEST) mutant was impaired in stripping NFκB from DNA and formed a more stable intermediate ternary complex than that formed from IκBα(WT) because DNA dissociated more slowly. NMR and amide hydrogen-deuterium exchange mass spectrometry showed that the IκBα(5xPEST) appears to be "caught in the act of stripping" because it is not yet completely in the folded and NFκB-bound state. When the mutant was introduced into cells, the rate of postinduction IκBα-mediated export of NFκB from the nucleus decreased markedly.


Assuntos
DNA/metabolismo , Inibidor de NF-kappaB alfa/metabolismo , NF-kappa B/metabolismo , Transdução de Sinais/genética , Ativação Transcricional , Animais , Núcleo Celular/metabolismo , Células Cultivadas , DNA/genética , Fibroblastos , Imunofluorescência , Técnicas de Inativação de Genes , Humanos , Proteínas I-kappa B/genética , Camundongos , Simulação de Acoplamento Molecular , Mutação , Inibidor de NF-kappaB alfa/genética , NF-kappa B/genética , Ressonância Magnética Nuclear Biomolecular , Regiões Promotoras Genéticas , Ligação Proteica , Proteínas Proto-Oncogênicas/genética , Estresse Fisiológico/fisiologia , Fator de Transcrição RelA/genética
18.
Mol Cell Proteomics ; 13(3): 823-35, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24425750

RESUMO

Bottom-up proteomics studies traditionally involve proteome digestion with a single protease, trypsin. However, trypsin alone does not generate peptides that encompass the entire proteome. Alternative proteases have been explored, but most have specificity for charged amino acid side chains. Therefore, additional proteases that improve proteome coverage through cleavage at sequences complementary to trypsin's may increase proteome coverage. We demonstrate the novel application of two proteases for bottom-up proteomics: wild type α-lytic protease (WaLP) and an active site mutant of WaLP, M190A α-lytic protease (MaLP). We assess several relevant factors, including MS/MS fragmentation, peptide length, peptide yield, and protease specificity. When data from separate digestions with trypsin, LysC, WaLP, and MaLP were combined, proteome coverage was increased by 101% relative to that achieved with trypsin digestion alone. To demonstrate how the gained sequence coverage can yield additional post-translational modification information, we show the identification of a number of novel phosphorylation sites in the Schizosaccharomyces pombe proteome and include an illustrative example from the protein MPD2 wherein two novel sites are identified, one in a tryptic peptide too short to identify and the other in a sequence devoid of tryptic sites. The specificity of WaLP and MaLP for aliphatic amino acid side chains was particularly valuable for coverage of membrane protein sequences, which increased 350% when the data from trypsin, LysC, WaLP, and MaLP were combined.


Assuntos
Proteoma/metabolismo , Proteômica/métodos , Serina Endopeptidases/metabolismo , Sequência de Aminoácidos , Aminoácidos/metabolismo , Animais , Bovinos , Ácido Desoxicólico/farmacologia , Elétrons , Guanidina/farmacologia , Espectrometria de Massas , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Peptídeos/química , Peptídeos/metabolismo , Padrões de Referência , Schizosaccharomyces/efeitos dos fármacos , Schizosaccharomyces/metabolismo , Proteínas de Schizosaccharomyces pombe/química , Proteínas de Schizosaccharomyces pombe/metabolismo , Análise de Sequência de Proteína , Dodecilsulfato de Sódio/farmacologia , Especificidade por Substrato/efeitos dos fármacos , Tripsina/metabolismo
19.
Toxicol Sci ; 129(1): 200-12, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22610605

RESUMO

Stress-activated protein kinases and transcription factors are crucial for surviving exposure to cadmium and other environmental toxicants, but their effects on the proteome remain largely unexplored. In this study, isobaric tag for relative and absolute quantitation reveals that cadmium stress triggers rapid proteome remodeling in the fission yeast Schizosaccharomyces pombe. Spc1/Sty1, a mitogen/stress-activated protein kinase homologous to human p38 and Saccharomyces cerevisiae Hog1, controls many of these changes, including enzymes of the oxidative phase of the pentose phosphate pathway and trehalose metabolism. Genetic studies indicate that control of carbohydrate metabolism by Spc1 is required for cadmium tolerance. The bZIP transcription factor Zip1, which is functionally related to human Nrf2 and S. cerevisiae Met4, has a smaller effect on cadmium-induced proteome remodeling, but it is required for production of key proteins involved in sulfur metabolism, which are essential for cadmium resistance. These studies reveal how Spc1 and Zip1 independently reshape the proteome to modulate cellular defense mechanisms against the toxic effects of cadmium.


Assuntos
Cádmio/toxicidade , Proteínas Quinases Ativadas por Mitógeno/fisiologia , Proteoma , Proteínas de Schizosaccharomyces pombe/fisiologia , Schizosaccharomyces/efeitos dos fármacos , Schizosaccharomyces/metabolismo , Transcriptoma , Trealose/metabolismo
20.
IUBMB Life ; 64(6): 499-505, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22573609

RESUMO

The paradigmatic transcription factors of the NFκB family provide an increasingly complex view of the mechanism of signal-mediated transcriptional activation. Although the primary event, phosphorylation and subsequent ubiquitin-dependent degradation of IκBα, the inhibitor of the canonical NFκB (p50/p65), is reasonably well understood, the means whereby the activation is turned off by postinduction repression are less well understood. Recent work highlighted in this review suggests that the inhibitor IκBα participates in the "stripping" of NFκB from the DNA, and that this process relies heavily on the disordered and weakly ordered segments of IκBα. Kinetic and equilibrium measurements in vitro as well as genetic screens in vivo convincingly demonstrate not only that IκBα greatly increases the dissociation rate of NFκB from DNA but also that further control of the process is mediated by the extremely short half-life of free IκBα, doubtless a result of the overall weakly folded nature of the free protein. These studies illustrate the versatility of protein systems that use not only well-structured proteins and protein complexes but also the full range of available weakly structured and disordered states to maximize functional efficiency and metabolic control.


Assuntos
Proteínas I-kappa B/química , Subunidade p50 de NF-kappa B/química , Fator de Transcrição RelA/química , Sequência de Aminoácidos , Animais , Sequência Consenso , Regulação da Expressão Gênica , Humanos , Dados de Sequência Molecular , Sinais de Localização Nuclear , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Estrutura Quaternária de Proteína , Estrutura Secundária de Proteína , Transporte Proteico , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA