Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
PNAS Nexus ; 3(2): pgae023, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38312223

RESUMO

The ability to monitor the response of metabolic enzymes to drug exposure in individuals is highly appealing and critical to personalized medicine. Although pharmacogenomics assesses genotypic differences, it does not report changes in metabolic enzyme activities due to environmental factors such as drug interactions. Here, we report a quantitative proteomics strategy to monitor drug metabolic pathways by profiling metabolic enzymes in circulating extracellular vesicles (EVs) upon drug exposure. Mass spectrometry (MS)-based measurement revealed that changes in metabolic enzyme abundance in EVs paralleled those in hepatic cells isolated from liver tissue. Coupling with multiplexed isotopic labeling, we temporally quantified 34 proteins involved in drug absorption, distribution, metabolism, and excretion (ADME) pathways. Out of 44 known ADME proteins in plasma EVs, previously annotated mouse cytochrome P450 3A11 (Cyp3a11), homolog to human CYP3A4, and uridine 5'-diphospho (UDP) glucuronosyltransferase 2A3 (Ugt2a3), increased upon daily rifampicin dosage. Dasatinib, a tyrosine kinase inhibitor to treat leukemia, also elevated Cyp3a11 levels in plasma EVs, but to a lesser extent. Altogether, this study demonstrates that measuring drug enzymes in circulating EVs as an effective surrogate is highly feasible and may transform today's drug discovery and development for personalized medicine.

2.
Trends Analyt Chem ; 1632023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37215489

RESUMO

Protein phosphorylation is an essential post-translational modification that regulates many aspects of cellular physiology, and dysregulation of pivotal phosphorylation events is often responsible for disease onset and progression. Clinical analysis on disease-relevant phosphoproteins, while quite challenging, provides unique information for precision medicine and targeted therapy. Among various approaches, mass spectrometry (MS)-centered characterization features discovery-driven, high-throughput and in-depth identification of phosphorylation events. This review highlights advances in sample preparation and instrument in MS-based phosphoproteomics and recent clinical applications. We emphasize the preeminent data-independent acquisition method in MS as one of the most promising future directions and biofluid-derived extracellular vesicles as an intriguing source of the phosphoproteome for liquid biopsy.

3.
Mol Cell Proteomics ; 22(5): 100536, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36997065

RESUMO

Translating the research capability and knowledge in cancer signaling into clinical settings has been slow and ineffective. Recently, extracellular vesicles (EVs) have emerged as a promising source for developing disease phosphoprotein markers to monitor disease status. This study focuses on the development of a robust data-independent acquisition (DIA) using mass spectrometry to profile urinary EV phosphoproteomics for renal cell cancer (RCC) grades differentiation. We examined gas-phase fractionated library, direct DIA (library-free), forbidden zones, and several different windowing schemes. After the development of a DIA mass spectrometry method for EV phosphoproteomics, we applied the strategy to identify and quantify urinary EV phosphoproteomes from 57 individuals representing low-grade clear cell RCC, high-grade clear cell RCC, chronic kidney disease, and healthy control individuals. Urinary EVs were efficiently isolated by functional magnetic beads, and EV phosphopeptides were subsequently enriched by PolyMAC. We quantified 2584 unique phosphosites and observed that multiple prominent cancer-related pathways, such as ErbB signaling, renal cell carcinoma, and regulation of actin cytoskeleton, were only upregulated in high-grade clear cell RCC. These results show that EV phosphoproteome analysis utilizing our optimized procedure of EV isolation, phosphopeptide enrichment, and DIA method provides a powerful tool for future clinical applications.


Assuntos
Carcinoma de Células Renais , Vesículas Extracelulares , Neoplasias Renais , Humanos , Carcinoma de Células Renais/metabolismo , Cromatografia de Afinidade/métodos , Transdução de Sinais , Neoplasias Renais/metabolismo , Vesículas Extracelulares/metabolismo
4.
Adv Clin Chem ; 112: 119-153, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36642482

RESUMO

We are currently experiencing a rapidly developing era in terms of translational and clinical medical sciences. The relatively mature state of nucleic acid examination has significantly improved our understanding of disease mechanism and therapeutic potential of personalized treatment, but misses a large portion of phenotypic disease information. Proteins, in particular phosphorylation events that regulates many cellular functions, could provide real-time information for disease onset, progression and treatment efficacy. The technical advances in liquid chromatography and mass spectrometry have realized large-scale and unbiased proteome and phosphoproteome analyses with disease relevant samples such as tissues. However, tissue biopsy still has multiple shortcomings, such as invasiveness of sample collection, potential health risk for patients, difficulty in protein preservation and extreme heterogeneity. Recently, extracellular vesicles (EVs) have offered a great promise as a unique source of protein biomarkers for non-invasive liquid biopsy. Membranous EVs provide stable preservation of internal proteins and especially labile phosphoproteins, which is essential for effective routine biomarker detection. To aid efficient EV proteomic and phosphoproteomic analyses, recent developments showcase clinically-friendly EV techniques, facilitating diagnostic and therapeutic applications. Ultimately, we envision that with streamlined sample preparation from tissues and EVs proteomics and phosphoproteomics analysis will become routine in clinical settings.


Assuntos
Vesículas Extracelulares , Espectrometria de Massas em Tandem , Humanos , Proteômica/métodos , Vesículas Extracelulares/metabolismo , Cromatografia Líquida/métodos , Fosfoproteínas/análise , Fosfoproteínas/química , Fosfoproteínas/metabolismo , Biomarcadores/análise , Proteoma/análise
5.
Methods Mol Biol ; 2504: 147-156, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35467285

RESUMO

A successful phosphoproteomics analysis of extracellular vesicles (EVs) requires a unique approach, fine-tuned to address the challenges that have plagued plasma-based biomarker discovery. Here, I detail a procedure, which combines EVtrap-based high-recovery EV isolation, phase-transfer surfactant method for protein extraction, and PolyMAC-based enrichment of phosphopeptides. The combination of these methods provides a highly effective strategy for EV-based phosphoproteome analysis and leads to the discovery of novel phospho-markers previously undetectable.


Assuntos
Vesículas Extracelulares , Biomarcadores/metabolismo , Vesículas Extracelulares/metabolismo , Fosfopeptídeos/metabolismo , Plasma/química , Proteoma/metabolismo
6.
Anal Chem ; 92(21): 14790-14797, 2020 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-33074658

RESUMO

Extracellular vesicles (EVs) are attracting increasing interest with their intriguing role in intercellular communications. Protein phosphorylation in EVs is of great importance for understanding intercellular signaling processes. However, the study of EV phosphoproteomics is impeded by their relatively low amount in limited clinical sample volumes, and it is necessary to have a sensitive and efficient enrichment method for EV phosphopeptides. Herein, a novel Ti(IV)-functionalized and glass fiber-supported hybrid monolithic spin tip, termed PhosTip, was prepared for enriching phosphopeptides from urinary EVs. Glass fiber as the stationary phase positions the hybrid monolith in a standard pipet tip and prevents the monolith from distortion during experiments. The preparation procedure for the new PhosTip is simple and time-saving. The hybrid monolithic PhosTip provides excellent enrichment efficiency of low-abundance phosphopeptides from cell digests and urinary EVs with minimum contamination and sample loss. Using the PhosTip, we demonstrate that 5373 and 336 unique phosphopeptides were identified from 100 and 1 µg of cell lysates, while 3919 and 217 unique phosphopeptides were successfully identified from 10 and 1 mL of urinary samples, respectively. The PhosTip was finally applied to enrich phosphopeptides in urine EVs from prostate cancer patients and healthy controls and quantify 118 up-regulated proteins with phosphosites in prostate cancer samples. These results demonstrated that the PhosTip could be a simple and convenient tool for enriching phosphopeptides from clinical samples and for broader applications in biomarker discovery.


Assuntos
Métodos Analíticos de Preparação de Amostras/instrumentação , Vesículas Extracelulares/metabolismo , Vidro , Fosfopeptídeos/urina , Humanos , Masculino , Fosfopeptídeos/química , Neoplasias da Próstata/urina , Titânio/química
7.
Nat Protoc ; 15(1): 161-180, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31863077

RESUMO

Extracellular vesicles (EVs) are increasingly being recognized as important vehicles for intercellular communication and as promising sources for biomarker discovery. Because the state of protein post-translational modifications (PTMs) such as phosphorylation and glycosylation can be a key determinant of cellular physiology, comprehensive characterization of protein PTMs in EVs can be particularly valuable for early-stage diagnostics and monitoring of disease status. However, the analysis of PTMs in EVs has been complicated by limited amounts of purified EVs, low-abundance PTM proteins, and interference from proteins and metabolites in biofluids. Recently, we developed an approach to isolate phosphoproteins and glycoproteins in EVs from small volumes of human plasma that enabled us to identify nearly 10,000 unique phosphopeptides and 1,500 unique N-glycopeptides. The approach demonstrated the feasibility of using these data to identify potential markers to differentiate disease from healthy states. Here we present an updated workflow to sequentially isolate phosphopeptides and N-glycopeptides, enabling multiple PTM analyses of the same clinical samples. In this updated workflow, we have improved the reproducibility and efficiency of EV isolation, protein extraction, and phosphopeptide/N-glycopeptide enrichment to achieve sensitive analyses of low-abundance PTMs in EVs isolated from 1 mL of plasma. The modularity of the workflow also allows for the characterization of phospho- or glycopeptides only and enables additional analysis of total proteomes and other PTMs of interest. After blood collection, the protocol takes 2 d, including EV isolation, PTM/peptide enrichment, mass spectrometry analysis, and data quantification.


Assuntos
Vesículas Extracelulares/metabolismo , Glicoproteínas/metabolismo , Fosfoproteínas/metabolismo , Plasma/citologia , Proteômica/métodos , Humanos , Processamento de Proteína Pós-Traducional
8.
Anal Chem ; 90(10): 6307-6313, 2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29629753

RESUMO

Glycoproteins comprise more than half of current FDA-approved protein cancer markers, but the development of new glycoproteins as disease biomarkers has been stagnant. Here we present a pipeline to develop glycoproteins from extracellular vesicles (EVs) through integrating quantitative glycoproteomics with a novel reverse phase glycoprotein array and then apply it to identify novel biomarkers for breast cancer. EV glycoproteomics show promise in circumventing the problems plaguing current serum/plasma glycoproteomics and allowed us to identify hundreds of glycoproteins that have not been identified in blood. We identified 1,453 unique glycopeptides representing 556 glycoproteins in EVs, among which 20 were verified significantly higher in individual breast cancer patients. We further applied a novel glyco-specific reverse phase protein array to quantify a subset of the candidates. Together, this study demonstrates the great potential of this integrated pipeline for biomarker discovery.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias da Mama/diagnóstico por imagem , Vesículas Extracelulares/química , Glicoproteínas/sangue , Cromatografia Líquida , Feminino , Humanos , Espectrometria de Massas em Tandem
9.
Proc Natl Acad Sci U S A ; 114(12): 3175-3180, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28270605

RESUMO

The state of protein phosphorylation can be a key determinant of cellular physiology such as early-stage cancer, but the development of phosphoproteins in biofluids for disease diagnosis remains elusive. Here we demonstrate a strategy to isolate and identify phosphoproteins in extracellular vesicles (EVs) from human plasma as potential markers to differentiate disease from healthy states. We identified close to 10,000 unique phosphopeptides in EVs isolated from small volumes of plasma samples. Using label-free quantitative phosphoproteomics, we identified 144 phosphoproteins in plasma EVs that are significantly higher in patients diagnosed with breast cancer compared with healthy controls. Several biomarkers were validated in individual patients using paralleled reaction monitoring for targeted quantitation. This study demonstrates that the development of phosphoproteins in plasma EV as disease biomarkers is highly feasible and may transform cancer screening and monitoring.


Assuntos
Biomarcadores Tumorais , Neoplasias da Mama/metabolismo , Vesículas Extracelulares/metabolismo , Fosfoproteínas/metabolismo , Biomarcadores , Proteínas Sanguíneas , Neoplasias da Mama/sangue , Estudos de Casos e Controles , Análise por Conglomerados , Biologia Computacional/métodos , Exossomos/metabolismo , Feminino , Humanos , Fosfopeptídeos/metabolismo , Proteoma , Proteômica/métodos , Reprodutibilidade dos Testes , Espectrometria de Massas em Tandem , Fluxo de Trabalho
10.
Curr Protoc Chem Biol ; 7(1): 17-25, 2015 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-25727060

RESUMO

This article describes methods for a new, non-antibody phosphorylation detection reagent, termed pIMAGO (phospho-imaging). This novel reagent takes advantage not only of the unique properties of the soluble nanoparticles, but also of the multiple functionalities of the molecule, allowing for highly selective, sensitive, and quantitative assessment of protein phosphorylation without using radioactive isotopes or phospho-specific antibodies. The methods allow for multiplexed detection of phosphorylation and total protein amount simultaneously. The straightforward and routine detection and quantitation of general phosphorylation on any site of any protein can be performed in western blot and ELISA formats.


Assuntos
Imagem Molecular/métodos , Fosforilação , Proteínas/análise , Biotina/química , Dendrímeros/química , Fosfatos/química , Proteínas/química , Titânio/química
11.
Anal Chem ; 86(13): 6363-71, 2014 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-24905233

RESUMO

Engagement of the B cell receptor for antigen (BCR) leads to immune responses through a cascade of intracellular signaling events. Most studies to date have focused on the BCR and protein tyrosine phosphorylation. Because spleen tyrosine kinase, Syk, is an upstream kinase in multiple BCR-regulated signaling pathways, it also affects many downstream events that are modulated through the phosphorylation of proteins on serine and threonine residues. Here, we report a novel phosphopeptide enrichment strategy and its application to a comprehensive quantitative phosphoproteomics analysis of Syk-dependent downstream signaling events in B cells, focusing on serine and threonine phosphorylation. Using a combination of the Syk inhibitor piceatannol, SILAC quantification, peptide fractionation, and complementary PolyMAC-Ti and PolyMAC-Zr enrichment techniques, we analyzed changes in BCR-stimulated protein phosphorylation that were dependent on the activity of Syk. We identified and quantified over 13,000 unique phosphopeptides, with a large percentage dependent on Syk activity in BCR-stimulated B cells. Our results not only confirmed many known functions of Syk, but more importantly, suggested many novel roles, including in the ubiquitin proteasome pathway, that warrant further exploration.


Assuntos
Linfócitos B/imunologia , Dendrímeros/química , Peptídeos e Proteínas de Sinalização Intracelular/química , Peptídeos e Proteínas de Sinalização Intracelular/imunologia , Fosfopeptídeos/análise , Proteínas Tirosina Quinases/química , Proteínas Tirosina Quinases/imunologia , Zircônio/química , Linhagem Celular , Fracionamento Químico/métodos , Humanos , Imunoglobulina M/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Íons/química , Metais/química , Fosfopeptídeos/isolamento & purificação , Fosforilação , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteômica/métodos , Transdução de Sinais , Quinase Syk , Titânio/química
12.
Electrophoresis ; 35(24): 3430-40, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24890697

RESUMO

Phosphoproteomics is the systematic study of one of the most common protein modifications in high throughput with the aim of providing detailed information of the control, response, and communication of biological systems in health and disease. Advances in analytical technologies and strategies, in particular the contributions of high-resolution mass spectrometers, efficient enrichments of phosphopeptides, and fast data acquisition and annotation, have catalyzed dramatic expansion of signaling landscapes in multiple systems during the past decade. While phosphoproteomics is an essential inquiry to map high-resolution signaling networks and to find relevant events among the apparently ubiquitous and widespread modifications of proteome, it presents tremendous challenges in separation sciences to translate it from discovery to clinical practice. In this mini-review, we summarize the analytical tools currently utilized for phosphoproteomic analysis (with focus on MS), progresses made on deciphering clinically relevant kinase-substrate networks, MS uses for biomarker discovery and validation, and the potential of phosphoproteomics for disease diagnostics and personalized medicine.


Assuntos
Espectrometria de Massas/métodos , Fosfopeptídeos/química , Fosfoproteínas/química , Proteômica/métodos , Pesquisa Translacional Biomédica/métodos , Animais , Humanos , Fosfopeptídeos/análise , Fosfopeptídeos/isolamento & purificação , Fosfoproteínas/análise , Fosfoproteínas/isolamento & purificação , Medicina de Precisão
13.
Electrophoresis ; 35(24): 3463-9, 2014 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24723360

RESUMO

Altered protein phosphorylation is a feature of many human cancers that can be targeted therapeutically. Phosphopeptide enrichment is a critical step for maximizing the depth of phosphoproteome coverage by MS, but remains challenging for tissue specimens because of their high complexity. We describe the first analysis of a tissue phosphoproteome using polymer-based metal ion affinity capture (PolyMAC), a nanopolymer that has excellent yield and specificity for phosphopeptide enrichment, on a transgenic mouse model of HER2-driven breast cancer. By combining phosphotyrosine immunoprecipitation with PolyMAC, 411 unique peptides with 139 phosphotyrosine, 45 phosphoserine, and 29 phosphothreonine sites were identified from five LC-MS/MS runs. Combining reverse phase liquid chromatography fractionation at pH 8.0 with PolyMAC identified 1571 unique peptides with 1279 phosphoserine, 213 phosphothreonine, and 21 phosphotyrosine sites from eight LC-MS/MS runs. Linear motif analysis indicated that many of the phosphosites correspond to well-known phosphorylation motifs. Analysis of the tyrosine phosphoproteome with the Drug Gene Interaction database uncovered a network of potential therapeutic targets centered on Src family kinases with inhibitors that are either FDA-approved or in clinical development. These results demonstrate that PolyMAC is well suited for phosphoproteomic analysis of tissue specimens.


Assuntos
Neoplasias Mamárias Experimentais/metabolismo , Fosfoaminoácidos/análise , Fosfopeptídeos/análise , Proteômica/métodos , Análise Serial de Tecidos/métodos , Animais , Cromatografia de Afinidade/métodos , Feminino , Masculino , Neoplasias Mamárias Experimentais/química , Neoplasias Mamárias Experimentais/enzimologia , Camundongos , Camundongos Transgênicos , Fosfoaminoácidos/metabolismo , Fosfopeptídeos/química , Fosfopeptídeos/metabolismo , Receptor ErbB-2/biossíntese , Espectrometria de Massas em Tandem
14.
Clin Chim Acta ; 420: 23-7, 2013 May.
Artigo em Inglês | MEDLINE | ID: mdl-23159844

RESUMO

BACKGROUND: For many diseases such as cancer where phosphorylation-dependent signaling is the foundation of disease onset and progression, single-gene testing and genomic profiling alone are not sufficient in providing most critical information. The reason for this is that in these activated pathways the signaling changes and drug resistance are often not directly correlated with changes in protein expression levels. In order to obtain the essential information needed to evaluate pathway activation or the effects of certain drugs and therapies on the molecular level, the analysis of changes in protein phosphorylation is critical. METHODS: Existing approaches do not differentiate clinical disease subtypes on the protein and signaling pathway level, and therefore hamper the predictive management of the disease and the selection of therapeutic targets. CONCLUSIONS: The mini-review examines the impact of emerging systems biology tools and the possibility of applying phosphoproteomics to clinical research.


Assuntos
Pesquisa Biomédica/tendências , Proteômica/tendências , Humanos , Fosfoproteínas/química , Fosforilação
16.
Mol Cell Proteomics ; 9(10): 2162-72, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20562096

RESUMO

The ability to obtain in-depth understanding of signaling networks in cells is a key objective of systems biology research. Such ability depends largely on unbiased and reproducible analysis of phosphoproteomes. We present here a novel proteomics tool, polymer-based metal ion affinity capture (PolyMAC), for the highly efficient isolation of phosphopeptides to facilitate comprehensive phosphoproteome analyses. This approach uses polyamidoamine dendrimers multifunctionalized with titanium ions and aldehyde groups to allow the chelation and subsequent isolation of phosphopeptides in a homogeneous environment. Compared with current strategies based on solid phase micro- and nanoparticles, PolyMAC demonstrated outstanding reproducibility, exceptional selectivity, fast chelation times, and high phosphopeptide recovery from complex mixtures. Using the PolyMAC method combined with antibody enrichment, we identified 794 unique sites of tyrosine phosphorylation in malignant breast cancer cells, 514 of which are dependent on the expression of Syk, a protein-tyrosine kinase with unusual properties of a tumor suppressor. The superior sensitivity of PolyMAC allowed us to identify novel components in a variety of major signaling networks, including cell migration and apoptosis. PolyMAC offers a powerful and widely applicable tool for phosphoproteomics and molecular signaling.


Assuntos
Nanoestruturas , Fosfoproteínas/química , Polímeros/química , Proteoma , Titânio/química , Western Blotting , Linhagem Celular Tumoral , Humanos , Imunoprecipitação , Reprodutibilidade dos Testes , Solubilidade , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz
17.
J Biol Chem ; 283(16): 10396-407, 2008 Apr 18.
Artigo em Inglês | MEDLINE | ID: mdl-18287090

RESUMO

The anaphase-promoting complex (APC) regulates the eukaryotic cell cycle by targeting specific proteins for proteasomal degradation. Its activity must be strictly controlled to ensure proper cell cycle progression. The co-activator proteins Cdc20 and Cdh1 are required for APC activity and are important regulatory targets. Recently, budding yeast Acm1 was identified as a Cdh1 binding partner and APC(Cdh1) inhibitor. Acm1 disappears in late mitosis when APC(Cdh1) becomes active and contains conserved degron-like sequences common to APC substrates, suggesting it could be both an inhibitor and substrate. Surprisingly, we found that Acm1 proteolysis is independent of APC. A major determinant of Acm1 stability is phosphorylation at consensus cyclin-dependent kinase sites. Acm1 is a substrate of Cdc28 cyclin-dependent kinase and Cdc14 phosphatase both in vivo and in vitro. Mutation of Cdc28 phosphorylation sites or conditional inactivation of Cdc28 destabilizes Acm1. In contrast, inactivation of Cdc14 prevents Acm1 dephosphorylation and proteolysis. Cdc28 stabilizes Acm1 in part by promoting binding of the 14-3-3 proteins Bmh1 and Bmh2. We conclude that the opposing actions of Cdc28 and Cdc14 are primary factors limiting Acm1 to the interval from G(1)/S to late mitosis and are capable of establishing APC-independent expression patterns similar to APC substrates.


Assuntos
Proteína Quinase CDC28 de Saccharomyces cerevisiae/fisiologia , Proteínas de Ciclo Celular/fisiologia , Regulação Fúngica da Expressão Gênica , Proteínas Tirosina Fosfatases/fisiologia , Proteínas Repressoras/fisiologia , Proteínas de Saccharomyces cerevisiae/fisiologia , Complexos Ubiquitina-Proteína Ligase , Proteínas 14-3-3/metabolismo , Sequência de Aminoácidos , Ciclossomo-Complexo Promotor de Anáfase , Mitose , Modelos Biológicos , Dados de Sequência Molecular , Fosforilação , Ligação Proteica , Proteínas de Saccharomyces cerevisiae/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...