Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
J Am Chem Soc ; 146(29): 20045-20058, 2024 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-39001877

RESUMO

G protein-coupled receptor (GPCR) structural studies with in-solution spectroscopic approaches have offered distinctive insights into GPCR activation and signaling that highly complement those yielded from structural snapshots by crystallography or cryo-EM. While most current spectroscopic approaches allow for probing structural changes at selected residues or loop regions, they are not suitable for capturing a holistic view of GPCR conformational rearrangements across multiple domains. Herein, we develop an approach based on limited proteolysis mass spectrometry (LiP-MS) to simultaneously monitor conformational alterations of a large number of residues spanning both flexible loops and structured transmembrane domains for a given GPCR. To benchmark LiP-MS for GPCR conformational profiling, we studied the adenosine 2A receptor (A2AR) in response to different ligand binding (agonist/antagonist/allosteric modulators) and G protein coupling. Systematic and residue-resolved profiling of A2AR conformational rearrangements by LiP-MS precisely captures structural mechanisms in multiple domains underlying ligand engagement, receptor activation, and allostery, and may also reflect local conformational flexibility. Furthermore, these residue-resolution structural fingerprints of the A2AR protein allow us to readily classify ligands of different pharmacology and distinguish the G protein-coupled state. Thus, our study provides a new structural MS approach that would be generalizable to characterizing conformational transition and plasticity for challenging integral membrane proteins.


Assuntos
Espectrometria de Massas , Conformação Proteica , Receptor A2A de Adenosina , Receptor A2A de Adenosina/química , Receptor A2A de Adenosina/metabolismo , Humanos , Ligantes , Modelos Moleculares , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo
2.
Proc Natl Acad Sci U S A ; 120(11): e2220767120, 2023 03 14.
Artigo em Inglês | MEDLINE | ID: mdl-36893261

RESUMO

The recently developed double-click reaction sequence [G. Meng et al., Nature 574, 86-89 (2019)] is expected to vastly expand the number and diversity of synthetically accessible 1,2,3-triazole derivatives. However, it remains elusive how to rapidly navigate the extensive chemical space created by double-click chemistry for bioactive compound discovery. In this study, we selected a particularly challenging drug target, the glucagon-like-peptide-1 receptor (GLP-1R), to benchmark our new platform for the design, synthesis, and screening of double-click triazole libraries. First, we achieved a streamlined synthesis of customized triazole libraries on an unprecedented scale (composed of 38,400 new compounds). By interfacing affinity-selection mass spectrometry and functional assays, we identified a series of positive allosteric modulators (PAMs) with unreported scaffolds that can selectively and robustly enhance the signaling activity of the endogenous GLP-1(9-36) peptide. Intriguingly, we further revealed an unexpected binding mode of new PAMs which likely act as a molecular glue between the receptor and the peptide agonist. We anticipate the merger of double-click library synthesis with the hybrid screening platform allows for efficient and economic discovery of drug candidates or chemical probes for various therapeutic targets.


Assuntos
Receptor do Peptídeo Semelhante ao Glucagon 1 , Peptídeos , Regulação Alostérica , Peptídeo 1 Semelhante ao Glucagon/metabolismo , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Peptídeos/química , Triazóis/química
3.
Proc Natl Acad Sci U S A ; 119(38): e2210769119, 2022 09 20.
Artigo em Inglês | MEDLINE | ID: mdl-36095215

RESUMO

Nanobodies and chemical cross-linking were used to gain information on the identity and positions of flexible domains of PI3Kα. The application of chemical cross-linking mass spectrometry (CXMS) facilitated the identification of the p85 domains BH, cSH2, and SH3 as well as their docking positions on the PI3Kα catalytic core. Binding of individual nanobodies to PI3Kα induced activation or inhibition of enzyme activity and caused conformational changes that could be correlated with enzyme function. Binding of nanobody Nb3-126 to the BH domain of p85α substantially improved resolution for parts of the PI3Kα complex, and binding of nanobody Nb3-159 induced a conformation of PI3Kα that is distinct from known PI3Kα structures. The analysis of CXMS data also provided mechanistic insights into the molecular underpinning of the flexibility of PI3Kα.


Assuntos
Domínio Catalítico , Classe I de Fosfatidilinositol 3-Quinases , Classe Ia de Fosfatidilinositol 3-Quinase , Classe I de Fosfatidilinositol 3-Quinases/química , Classe Ia de Fosfatidilinositol 3-Quinase/química , Humanos , Espectrometria de Massas/métodos , Anticorpos de Domínio Único
4.
Nature ; 604(7907): 779-785, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35418679

RESUMO

Adhesion G protein-coupled receptors (aGPCRs) are essential for a variety of physiological processes such as immune responses, organ development, cellular communication, proliferation and homeostasis1-7. An intrinsic manner of activation that involves a tethered agonist in the N-terminal region of the receptor has been proposed for the aGPCRs8,9, but its molecular mechanism remains elusive. Here we report the G protein-bound structures of ADGRD1 and ADGRF1, which exhibit many unique features with regard to the tethered agonism. The stalk region that proceeds the first transmembrane helix acts as the tethered agonist by forming extensive interactions with the transmembrane domain; these interactions are mostly conserved in ADGRD1 and ADGRF1, suggesting that a common stalk-transmembrane domain interaction pattern is shared by members of the aGPCR family. A similar stalk binding mode is observed in the structure of autoproteolysis-deficient ADGRF1, supporting a cleavage-independent manner of receptor activation. The stalk-induced activation is facilitated by a cascade of inter-helix interaction cores that are conserved in positions but show sequence variability in these two aGPCRs. Furthermore, the intracellular region of ADGRF1 contains a specific lipid-binding site, which proves to be functionally important and may serve as the recognition site for the previously discovered endogenous ADGRF1 ligand synaptamide. These findings highlight the diversity and complexity of the signal transduction mechanisms of the aGPCRs.


Assuntos
Receptores Acoplados a Proteínas G , Transdução de Sinais , Humanos , Ligantes , Proteínas Oncogênicas/agonistas , Proteínas Oncogênicas/metabolismo , Ligação Proteica , Domínios Proteicos , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/metabolismo
5.
Nat Commun ; 12(1): 6685, 2021 11 18.
Artigo em Inglês | MEDLINE | ID: mdl-34795227

RESUMO

Phosphoproteomics integrating data-independent acquisition (DIA) enables deep phosphoproteome profiling with improved quantification reproducibility and accuracy compared to data-dependent acquisition (DDA)-based phosphoproteomics. DIA data mining heavily relies on a spectral library that in most cases is built on DDA analysis of the same sample. Construction of this project-specific DDA library impairs the analytical throughput, limits the proteome coverage, and increases the sample size for DIA phosphoproteomics. Herein we introduce a deep neural network, DeepPhospho, which conceptually differs from previous deep learning models to achieve accurate predictions of LC-MS/MS data for phosphopeptides. By leveraging in silico libraries generated by DeepPhospho, we establish a DIA workflow for phosphoproteome profiling which involves DIA data acquisition and data mining with DeepPhospho predicted libraries, thus circumventing the need of DDA library construction. Our DeepPhospho-empowered workflow substantially expands the phosphoproteome coverage while maintaining high quantification performance, which leads to the discovery of more signaling pathways and regulated kinases in an EGF signaling study than the DDA library-based approach. DeepPhospho is provided as a web server as well as an offline app to facilitate user access to model training, predictions and library generation.


Assuntos
Biologia Computacional/métodos , Aprendizado Profundo , Biblioteca de Peptídeos , Fosfoproteínas/análise , Proteoma/análise , Proteômica/métodos , Algoritmos , Linhagem Celular Tumoral , Cromatografia Líquida/métodos , Simulação por Computador , Mineração de Dados/métodos , Humanos , Fosfopeptídeos/análise , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem/métodos
6.
ACS Chem Biol ; 16(6): 991-1002, 2021 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-34048655

RESUMO

Allosteric ligands provide new opportunities to modulate G protein-coupled receptor (GPCR) function and present therapeutic benefits over orthosteric molecules. Negative allosteric modulators (NAMs) can inhibit the activation of a receptor and downstream signal transduction. Screening NAMs for a GPCR target is particularly challenging because of the difficulty in distinguishing NAMs from antagonists bound to the orthosteric site as they both show inhibitory effects in receptor signaling assays. Here we report an affinity mass spectrometry (MS)-based approach tailored to screening potential NAMs of a GPCR target especially from fragment libraries. Compared to regular surface plasmon resonance or NMR-based methods for fragment screening, our approach features a reduction of the protein and compound consumption by 2-4 orders of magnitude and an increase in the data acquisition speed by 2-3 orders of magnitude. Our affinity MS-based fragment screening led to the identification of a new NAM of the adenosine A2A receptor (A2AAR) bearing an unprecedented azetidine moiety predicted to occupy the allosteric sodium binding site. Molecular dynamics simulations, ligand structure-activity relationship (SAR) studies, and in-solution NMR analyses further revealed the unique binding mode and antagonistic property of this compound that differs considerably from HMA (5-(N,N-hexamethylene)amiloride), a well-characterized NAM of A2AAR. Taken together, our work would facilitate fragment-based screening of allosteric modulators, as well as guide the design of novel NAMs acting at the sodium ion pocket of class A GPCRs.


Assuntos
Agonistas do Receptor A2 de Adenosina/farmacologia , Antagonistas do Receptor A2 de Adenosina/farmacologia , Regulação Alostérica/efeitos dos fármacos , Receptor A2A de Adenosina/metabolismo , Sódio/metabolismo , Agonistas do Receptor A2 de Adenosina/química , Antagonistas do Receptor A2 de Adenosina/química , Sítio Alostérico/efeitos dos fármacos , Sítios de Ligação/efeitos dos fármacos , Descoberta de Drogas , Humanos , Simulação de Acoplamento Molecular , Simulação de Dinâmica Molecular , Receptor A2A de Adenosina/química
7.
Anal Chem ; 91(13): 8162-8169, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31094506

RESUMO

Affinity mass spectrometry (MS) enables rapid screening of compound mixtures for ligands bound to a specific protein target, yet its current throughput is limited to individually assay pools of 400-2000 compounds. Typical affinity MS screens implemented in pharmaceutical industry laboratories identify putative ligands based on qualitative analysis of compound binding to the target whereas no quantitative information is acquired to discriminate high- and low-affinity ligands in the screening phase. Furthermore, these screens require purification of a stabilized form of the protein target, which poses a great challenge for membrane receptor targets. Here, we describe a new, potentially general affinity MS strategy that allows screening of 20,000 compounds in one pool for highly efficient ligand discovery toward a G protein-coupled receptor (GPCR) target. Quantitative measurement of compound binding to the receptor enables high-affinity ligand selection using both the purified receptor and receptor-embedded cell membranes. This high-throughput, label-free and quantitative affinity MS screen resulted in discovery of three new antagonists of the A2A adenosine receptor.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Espectrometria de Massas/métodos , Receptores Acoplados a Proteínas G/metabolismo , Antagonistas do Receptor A2 de Adenosina/análise , Membrana Celular/metabolismo , Humanos , Ligantes , Ligação Proteica , Receptores Acoplados a Proteínas G/antagonistas & inibidores
8.
Analyst ; 144(9): 2881-2890, 2019 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-30788466

RESUMO

Although natural herbs have been a rich source of compounds for drug discovery, identification of bioactive components from natural herbs suffers from low efficiency and prohibitive cost of the conventional bioassay-based screening platforms. Here we develop a new strategy that integrates virtual screening, affinity mass spectrometry (MS) and targeted metabolomics for efficient discovery of herb-derived ligands towards a specific protein target site. Herb-based virtual screening conveniently selects herbs of potential bioactivity whereas affinity MS combined with targeted metabolomics readily screens candidate compounds in a high-throughput manner. This new integrated approach was benchmarked on screening chemical ligands that target the hydrophobic pocket of the nucleoprotein (NP) of Ebola viruses for which no small molecule ligands have been reported. Seven compounds identified by this approach from the crude extracts of three natural herbs were all validated to bind to the NP target in pure ligand binding assays. Among them, three compounds isolated from Piper nigrum (HJ-1, HJ-4 and HJ-6) strongly promoted the formation of large NP oligomers and reduced the protein thermal stability. In addition, cooperative binding between these chemical ligands and an endogenous peptide ligand was observed, and molecular docking was employed to propose a possible mechanism. Taken together, we established a platform integrating in silico and experimental screening approaches for efficient discovery of herb-derived bioactive ligands especially towards non-enzyme protein targets.


Assuntos
Produtos Biológicos/metabolismo , Espectrometria de Massas/métodos , Metabolômica/métodos , Nucleoproteínas/metabolismo , Extratos Vegetais/metabolismo , Proteínas do Core Viral/metabolismo , Sítios de Ligação , Produtos Biológicos/química , Produtos Biológicos/isolamento & purificação , Descoberta de Drogas/métodos , Ebolavirus/química , Ligantes , Simulação de Acoplamento Molecular , Proteínas do Nucleocapsídeo , Nucleoproteínas/química , Ophiopogon/química , Piper nigrum/química , Componentes Aéreos da Planta/química , Extratos Vegetais/química , Extratos Vegetais/isolamento & purificação , Ligação Proteica , Salvia miltiorrhiza/química , Sementes/química , Proteínas do Core Viral/química
9.
Nature ; 560(7720): 666-670, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30135577

RESUMO

Frizzled receptors (FZDs) are class-F G-protein-coupled receptors (GPCRs) that function in Wnt signalling and are essential for developing and adult organisms1,2. As central mediators in this complex signalling pathway, FZDs serve as gatekeeping proteins both for drug intervention and for the development of probes in basic and in therapeutic research. Here we present an atomic-resolution structure of the human Frizzled 4 receptor (FZD4) transmembrane domain in the absence of a bound ligand. The structure reveals an unusual transmembrane architecture in which helix VI is short and tightly packed, and is distinct from all other GPCR structures reported so far. Within this unique transmembrane fold is an extremely narrow and highly hydrophilic pocket that is not amenable to the binding of traditional GPCR ligands. We show that such a pocket is conserved across all FZDs, which may explain the long-standing difficulties in the development of ligands for these receptors. Molecular dynamics simulations on the microsecond timescale and mutational analysis uncovered two coupled, dynamic kinks located at helix VII that are involved in FZD4 activation. The stability of the structure in its ligand-free form, an unfavourable pocket for ligand binding and the two unusual kinks on helix VII suggest that FZDs may have evolved a novel ligand-recognition and activation mechanism that is distinct from that of other GPCRs.


Assuntos
Receptores Frizzled/química , Sítios de Ligação , Cristalografia por Raios X , Cisteína/metabolismo , Proteínas Desgrenhadas/metabolismo , Receptores Frizzled/genética , Humanos , Ligantes , Modelos Moleculares , Simulação de Dinâmica Molecular , Domínios Proteicos , Via de Sinalização Wnt
10.
J Proteomics ; 186: 38-46, 2018 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-30030164

RESUMO

Mass spectrometry (MS) technology has a special advantage in species determination for protein-rich samples which requires identification of species-specific peptides. However, for species discrimination of highly homologous proteins, it remains challenging to select the species unique peptides with routine proteomics approaches. In this work, we chose keratins and keratin-associated proteins (KAPs) present in cashmere fibers from goat and wool fibers from sheep as targets, to develop a dual-protease digestion workflow based on in-silico and experimental analysis. Combined usage of Glu-C and trypsin proteases showed the best digestion performance for MS identification of keratins and KAPs from different species. The parallel reaction monitoring (PRM) technique was implemented to validate and quantify the selected species discriminable peptides. The fiber composition of both blended animal hair fibers and industrial textile fabrics were successfully determined with the PRM assay. Furthermore, we identified over 360 peptides from the cashmere fiber beyond the current Uniprot goat proteome database. We expect our new workflow would improve the identification and quantification of keratin and KAPs, and provide inspiration for distinguishing other highly homologous proteins. We also anticipate the set of species-specific peptides from keratin or KAPs validated in this work would benefit the quality assessment for industrial fiber materials and textile products. SIGNIFICANCE: Discriminating species from highly homologous proteins is challenging for MS-based shotgun proteomics. The large percentage of overlapped protein sequence hinders the identification of the species unique peptides. In this work, we aimed to discriminate sample species between goat and sheep from keratins and keratin-associated proteins (KAPs). A dedicated workflow was developed to boost the exposure and quantification of species discriminable peptides. The dual-proteases digestion approach was optimized based on amino acid sequence analysis and protein in-silico digestion analysis. The PRM assays were established to validate and quantify the selected species unique peptides. Additionally, we have identified about 360 novel candidate peptides complementary to the current goat protein sequence database. We expect our workflow would improve the species discrimination for highly homologous proteins and benefit the proteomics study of keratin and KAPs in the human proteome.


Assuntos
Queratinas/análise , Proteólise , Homologia de Sequência de Aminoácidos , Animais , Cabras , Cabelo/química , Espectrometria de Massas/métodos , Peptídeo Hidrolases/metabolismo , Peptídeos/análise , Especificidade da Espécie , Fibra de Lã/análise
11.
Biochim Biophys Acta Mol Cell Res ; 1864(3): 562-571, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28011284

RESUMO

Posttranslational modifications of certain stress granule (SG) proteins are closely related to the assembly of SGs, a type of cytoplasmic foci structure. Our previous studies revealed that the Tudor staphylococcal nuclease (Tudor-SN) protein participates in the formation of SGs. However, the functional significance of potential Tudor-SN modifications during stress has not been reported. In this study, we demonstrated that the Tudor-SN protein was phosphorylated at threonine 103 (T103) upon stimulation with arsenite. In addition, c-Jun N-terminal kinase (JNK) was found to be responsible for Tudor-SN phosphorylation at the T103 site. We further illustrated that either a T103A mutation or the suppression of phosphorylation of T103 by the JNK inhibitor SP600125 inhibited the efficient recruitment of Tudor-SN into SGs. In addition, the T103A mutation could affect the physical binding of Tudor-SN with the G3BP (Ras-GAP SH3 domain-binding protein) protein but not with the HuR (Hu antigen R) protein and AGTR1-3'UTR (3'-untranslated region of angiotensin II receptor, type 1) mRNA cargo. These data suggested that JNK-enhanced Tudor-SN phosphorylation promotes the interaction between Tudor-SN and G3BP and facilitates the efficient recruitment of Tudor-SN into SGs under conditions of sodium arsenite-induced oxidative stress. This finding provides novel insights into the physiological function of Tudor-SN modification.


Assuntos
Proteínas de Transporte/genética , Grânulos Citoplasmáticos/metabolismo , Proteínas Quinases JNK Ativadas por Mitógeno/genética , Proteínas Nucleares/genética , Processamento de Proteína Pós-Traducional , Antracenos/farmacologia , Arsenitos/farmacologia , Proteínas de Transporte/metabolismo , Grânulos Citoplasmáticos/efeitos dos fármacos , Grânulos Citoplasmáticos/ultraestrutura , DNA Helicases , Proteína Semelhante a ELAV 1/genética , Proteína Semelhante a ELAV 1/metabolismo , Endonucleases , Células HeLa , Humanos , Proteínas Quinases JNK Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Mutação , Proteínas Nucleares/metabolismo , Estresse Oxidativo , Fosforilação/efeitos dos fármacos , Proteínas de Ligação a Poli-ADP-Ribose , Ligação Proteica , Inibidores de Proteínas Quinases/farmacologia , RNA Helicases , Proteínas com Motivo de Reconhecimento de RNA , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Compostos de Sódio/farmacologia , Treonina/metabolismo
12.
Artigo em Inglês | MEDLINE | ID: mdl-26894684

RESUMO

Quantification of energy and redox cofactors is of great value to synthetic biologists to infer the balance of energy metabolism in engineered microbial strains and assess each strain's potential for further improvement. Most currently used methods for intracellular cofactor measurement suffer from incomplete coverage, low reproducibility, suboptimal sensitivity or specificity. In this study, we described an SPE-HILIC/MS approach for simultaneous determination of six cofactor targets (ATP, ADP, NAD, NADH, NADP, NADPH) in Escherichia coli cells. Sufficient linearity, precision and metabolite recoveries of this new approach justified its reliability in targeted cofactor quantification. Our approach was then compared with conventional enzymatic assays to demonstrate its superior performance. We applied the SPE-HILIC/MS approach to profile shift of cofactor balances in several engineered E. coli strains with varying isobutanol production. Our cofactor analysis clearly revealed that optimal energy fitness was achieved in the highest-yield strain through combined modulation of a transhydrogenase and a NAD(+) kinase. Apart from the targeted cofactors, the SPE enrichment procedure also allowed for confident identification of 39 groups of polar metabolites mainly involved in central carbon metabolism in E. coli cells.


Assuntos
Difosfato de Adenosina/análise , Trifosfato de Adenosina/análise , Escherichia coli/química , NADP/análise , NAD/análise , Difosfato de Adenosina/metabolismo , Trifosfato de Adenosina/metabolismo , Butanóis/metabolismo , Cromatografia Líquida/métodos , Escherichia coli/metabolismo , Limite de Detecção , Espectrometria de Massas/métodos , Engenharia Metabólica , NAD/metabolismo , NADP/metabolismo , Extração em Fase Sólida/métodos
13.
PLoS One ; 11(1): e0147044, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-26789629

RESUMO

Cashmere is regarded as a specialty and luxury fiber due to its scarcity and high economic value. For fiber quality assessment, it is technically very challenging to distinguish and quantify the cashmere fiber from yak or wool fibers because of their highly similar physical appearance and substantial protein sequence homology. To address this issue, we propose a workflow combining untargeted and targeted proteomics strategies for selecting, verifying and quantifying biomarkers for cashmere textile authentication. Untargeted proteomic surveys were first applied to identify 174, 157, and 156 proteins from cashmere, wool and yak fibers, respectively. After marker selection at different levels, peptides turned out to afford much higher selectivity than proteins for fiber species discrimination. Subsequently, parallel reaction monitoring (PRM) methods were developed for ten selected peptide markers. The PRM-based targeted analysis of peptide markers enabled accurate determination of fiber species and cashmere percentages in different fiber mixtures. Furthermore, collective use of these peptide makers allowed us to discriminate and quantify cashmere fibers in commercial finished fabrics that have undergone heavy chemical treatments. Cashmere proportion measurement in fabric samples using our proteomic approach was in good agreement with results from traditional light microscopy, yet our method can be more readily standardized to become an objective and robust assay for assessing authenticity of fibers and textiles. We anticipate that the proteomic strategies presented in our study could be further implicated in discovery of quality trait markers for other products containing highly homologous proteomes.


Assuntos
Peptídeos/análise , Proteômica/métodos , Lã/química , Animais , Bovinos , Ovinos
14.
Sci Rep ; 5: 16869, 2015 Nov 19.
Artigo em Inglês | MEDLINE | ID: mdl-26581825

RESUMO

The post-translational modifications (PTMs) occurring on microtubules have been implicated in the regulation of microtubule properties and functions. Acetylated K40 of α-tubulin, a hallmark of long-lived stable microtubules, is known to be negatively controlled by histone deacetylase 6 (HDAC6). However, the vital roles of HDAC6 in microtubule-related processes such as cell motility and cell division cannot be fully explained by the only known target site on tubulin. Here, we attempt to comprehensively map lysine acetylation sites on tubulin purified from mouse brain tissues. Furthermore, mass spectrometry-based quantitative comparison of acetylated peptides from wild-type vs HDAC6 knockout mice allowed us to identify six new deacetylation sites possibly mediated by HDAC6. Thus, adding new sites to the repertoire of HDAC6-mediated tubulin deacetylation events would further our understanding of the multi-faceted roles of HDAC6 in regulating microtubule stability and cellular functions.


Assuntos
Encéfalo/metabolismo , Histona Desacetilases/metabolismo , Espectrometria de Massas em Tandem/métodos , Tubulina (Proteína)/metabolismo , Acetilação , Sequência de Aminoácidos , Animais , Desacetilase 6 de Histona , Humanos , Lisina/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Modelos Moleculares , Dados de Sequência Molecular , Peptídeos/química , Peptídeos/metabolismo , Tubulina (Proteína)/química , Tubulina (Proteína)/isolamento & purificação
15.
Thorac Cancer ; 6(5): 649-54, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26445615

RESUMO

Microtubule-binding proteins (MBPs) are structurally and functionally diverse regulators of microtubule-mediated cellular processes. Alteration of MBPs has been implicated in the pathogenesis of human diseases, including cancer. MBPs can stabilize or destabilize microtubules or move along microtubules to transport various cargoes. In addition, MBPs can control microtubule dynamics through direct interaction with microtubules or coordination with other proteins. To better understand microtubule structure and function, it is necessary to identify additional MBPs. In this study, we isolated microtubules and MBPs from mammalian cells by a taxol-based method and then profiled a panel of MBPs by mass spectrometry. We discovered a number of previously uncharacterized MBPs, including several membrane-associated proteins and proteins involved in post-translational modifications, in addition to several structural components. These results support the notion that microtubules have a wide range of functions and may undergo more exquisite regulation than previously recognized.

16.
J Proteome Res ; 14(8): 3292-304, 2015 Aug 07.
Artigo em Inglês | MEDLINE | ID: mdl-26165356

RESUMO

Tubulin is known to undergo unique post-translational modifications (PTMs), such as detyrosination and polyglutamylation, particularly in the unstructured carboxy-terminal tails (CTTs). However, more conventional PTMs of tubulin and their roles in the regulation of microtubule properties and functions remain poorly defined. Here, we report the comprehensive profiling of tubulin phosphorylation, acetylation, ubiquitylation, and O-GlcNAcylation in HeLa cells with a proteomic approach. Our tubulin-targeted analysis has identified 80 residues bearing single or multiple conventional PTMs including 24 novel PTM sites not covered in previous global proteomic surveys. By using a series of PTM-deficient or PTM-mimicking mutants, we further find that tubulin phosphorylation and acetylation play important roles in the control of microtubule assembly and stability. In addition, these tubulin PTMs have distinct effects on the retrograde transport of adenoviruses along microtubules. These findings thus enlarge the repertoire of tubulin PTMs and foster our understanding of their versatile roles in the regulation of microtubule dynamics and cellular functions.


Assuntos
Microtúbulos/metabolismo , Processamento de Proteína Pós-Traducional , Proteômica/métodos , Tubulina (Proteína)/metabolismo , Acetilação , Sequência de Aminoácidos , Sítios de Ligação/genética , Cromatografia Líquida , Glicosilação , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Células HeLa , Humanos , Immunoblotting , Espectrometria de Massas , Microscopia de Fluorescência , Microtúbulos/genética , Modelos Moleculares , Dados de Sequência Molecular , Fosforilação , Estrutura Terciária de Proteína , Homologia de Sequência de Aminoácidos , Tubulina (Proteína)/química , Tubulina (Proteína)/genética , Ubiquitinação
18.
Cell Cycle ; 13(6): 974-83, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24552808

RESUMO

Cylindromatosis (CYLD), a deubiquitinase involved in inflammation and tumorigenesis via the modulation of cell signaling, has recently been identified as a critical regulator of microtubule dynamics. CYLD has also been shown to stimulate cell migration and thereby contribute to normal physiological processes. However, it remains elusive how the regulation of microtubule dynamic properties by CYLD is connected to its role in mediating cell migration. In this study, we performed yeast 2-hybrid screening with CYLD as bait and identified 7 CYLD-interacting proteins, including end-binding protein 1 (EB1). The CYLD-EB1 interaction was confirmed both in cells and in vitro, and these 2 proteins colocalized at the plus ends of microtubules. Interestingly, the association of CYLD with EB1 was significantly increased upon the stimulation of cell migration. CYLD coordinated with EB1 to orchestrate tail retraction, centrosome reorientation, and leading-edge microtubule stabilization in migratory cells. In addition, CYLD acted in concert with EB1 to regulate microtubule assembly in vitro, microtubule nucleation at the centrosome, and microtubule growth at the cell periphery. These data provide mechanistic insights into the actions of CYLD in the regulation of microtubule dynamics and cell migration. These findings also support the notion that coordinated actions of microtubule-binding proteins are critical for microtubule-mediated cellular events.


Assuntos
Movimento Celular/fisiologia , Proteínas Associadas aos Microtúbulos/metabolismo , Microtúbulos/fisiologia , Proteínas Supressoras de Tumor/metabolismo , Centrossomo/fisiologia , Enzima Desubiquitinante CYLD , Células HeLa , Humanos
19.
Mol Biosyst ; 9(7): 1877-87, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23567825

RESUMO

The current study evaluated the glycoproteomic profile of tissues from colon cancer patients. The lectin microarray was first performed to compare the glycoprotein profiles between colon cancer and matched normal tissues. Level of N-acetylglucosamine (GlcNAc) that Solanum tuberosum lectin (STL) bound was found to be elevated in colon cancer, which was verified through lectin histochemistry. The subsequent glycoproteomic analysis based on STL enrichment of glycoproteins followed by label-free quantitative nano liquid chromatography-mass spectrometry/mass spectrometry (nanoLC-MS/MS) analysis identified 72 proteins in high confidence. Among these proteins, 17 were exclusively detected in cancer tissues, and 14 were significantly upregulated in tumor tissues. Annexin A1 and HSP90ß were chosen for further investigation by immunoprecipitation coupled with lectin blots, western blots and tissue microarrays. Both Annexin A1 and HSP90ß were GlcNAcylated, and their protein expressions were elevated in colon cancer, compared to normal tissues. Moreover, specific changes of GlcNAc abundances in Annexin A1 and HSP90ß suggested that tumor-specific glycan patterns could serve as candidate biomarkers of colon cancer for distinguishing cancer patients from healthy individuals.


Assuntos
Neoplasias do Colo/metabolismo , Glicoproteínas/química , Proteoma/química , Proteômica , Idoso , Idoso de 80 Anos ou mais , Colo/metabolismo , Neoplasias do Colo/patologia , Feminino , Glicoproteínas/análise , Humanos , Lectinas/química , Lectinas/metabolismo , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , Análise Serial de Proteínas , Proteoma/análise , Proteômica/métodos , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem
20.
Cell Cycle ; 12(7): 1133-41, 2013 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-23470638

RESUMO

Parkin, an E3 ubiquitin ligase well known for its role in the pathogenesis of juvenile Parkinson disease, has been considered as a candidate tumor suppressor in certain types of cancer. It remains unknown whether parkin is involved in the development of pancreatic cancer, the fourth leading cause of cancer-related deaths worldwide. Herein, we demonstrate the downregulation and copy number loss of the parkin gene in human pancreatic cancer specimens. The expression of parkin negatively correlates with clinicopathological parameters indicating the malignancy of pancreatic cancer. In addition, knockdown of parkin expression promotes the proliferation and tumorigenic properties of pancreatic cancer cells both in vitro and in mice. We further find that parkin deficiency increases the proportion of cells with spindle multipolarity and multinucleation. Parkin-depleted cells also show a significant increase in spindle misorientation. These findings indicate crucial involvement of parkin deficiency in the pathogenesis of pancreatic cancer.


Assuntos
Neoplasias Pancreáticas/metabolismo , Fuso Acromático/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Proliferação de Células , Regulação para Baixo , Dosagem de Genes , Humanos , Cinesinas/antagonistas & inibidores , Cinesinas/genética , Cinesinas/metabolismo , Camundongos , Camundongos Nus , Neoplasias Pancreáticas/patologia , Interferência de RNA , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Transplante Heterólogo , Células Tumorais Cultivadas , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA