Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 74
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Chem Biol ; 20(8): 1033-1043, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38302607

RESUMEN

The leaf-cutter ant fungal garden ecosystem is a naturally evolved model system for efficient plant biomass degradation. Degradation processes mediated by the symbiotic fungus Leucoagaricus gongylophorus are difficult to characterize due to dynamic metabolisms and spatial complexity of the system. Herein, we performed microscale imaging across 12-µm-thick adjacent sections of Atta cephalotes fungal gardens and applied a metabolome-informed proteome imaging approach to map lignin degradation. This approach combines two spatial multiomics mass spectrometry modalities that enabled us to visualize colocalized metabolites and proteins across and through the fungal garden. Spatially profiled metabolites revealed an accumulation of lignin-related products, outlining morphologically unique lignin microhabitats. Metaproteomic analyses of these microhabitats revealed carbohydrate-degrading enzymes, indicating a prominent fungal role in lignocellulose decomposition. Integration of metabolome-informed proteome imaging data provides a comprehensive view of underlying biological pathways to inform our understanding of metabolic fungal pathways in plant matter degradation within the micrometer-scale environment.


Asunto(s)
Lignina , Consorcios Microbianos , Lignina/metabolismo , Consorcios Microbianos/fisiología , Animales , Hormigas/metabolismo , Hormigas/microbiología , Ecosistema , Proteómica/métodos , Proteoma/metabolismo , Simbiosis
2.
Mol Cell Proteomics ; 22(8): 100592, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37328065

RESUMEN

The need for a clinically accessible method with the ability to match protein activity within heterogeneous tissues is currently unmet by existing technologies. Our proteomics sample preparation platform, named microPOTS (Microdroplet Processing in One pot for Trace Samples), can be used to measure relative protein abundance in micron-scale samples alongside the spatial location of each measurement, thereby tying biologically interesting proteins and pathways to distinct regions. However, given the smaller pixel/voxel number and amount of tissue measured, standard mass spectrometric analysis pipelines have proven inadequate. Here we describe how existing computational approaches can be adapted to focus on the specific biological questions asked in spatial proteomics experiments. We apply this approach to present an unbiased characterization of the human islet microenvironment comprising the entire complex array of cell types involved while maintaining spatial information and the degree of the islet's sphere of influence. We identify specific functional activity unique to the pancreatic islet cells and demonstrate how far their signature can be detected in the adjacent tissue. Our results show that we can distinguish pancreatic islet cells from the neighboring exocrine tissue environment, recapitulate known biological functions of islet cells, and identify a spatial gradient in the expression of RNA processing proteins within the islet microenvironment.


Asunto(s)
Islotes Pancreáticos , Proteoma , Humanos , Proteoma/metabolismo , Islotes Pancreáticos/metabolismo , Espectrometría de Masas
3.
Anal Chem ; 96(32): 12973-12982, 2024 Aug 13.
Artículo en Inglés | MEDLINE | ID: mdl-39089681

RESUMEN

There is increasing interest in developing in-depth proteomic approaches for mapping tissue heterogeneity in a cell-type-specific manner to better understand and predict the function of complex biological systems such as human organs. Existing spatially resolved proteomics technologies cannot provide deep proteome coverage due to limited sensitivity and poor sample recovery. Herein, we seamlessly combined laser capture microdissection with a low-volume sample processing technology that includes a microfluidic device named microPOTS (microdroplet processing in one pot for trace samples), multiplexed isobaric labeling, and a nanoflow peptide fractionation approach. The integrated workflow allowed us to maximize proteome coverage of laser-isolated tissue samples containing nanogram levels of proteins. We demonstrated that the deep spatial proteomics platform can quantify more than 5000 unique proteins from a small-sized human pancreatic tissue pixel (∼60,000 µm2) and differentiate unique protein abundance patterns in pancreas. Furthermore, the use of the microPOTS chip eliminated the requirement for advanced microfabrication capabilities and specialized nanoliter liquid handling equipment, making it more accessible to proteomic laboratories.


Asunto(s)
Péptidos , Proteoma , Proteómica , Humanos , Proteoma/análisis , Proteómica/métodos , Péptidos/análisis , Péptidos/química , Páncreas/metabolismo , Páncreas/química , Nanotecnología , Técnicas Analíticas Microfluídicas/instrumentación , Captura por Microdisección con Láser/métodos
4.
Mol Cell Proteomics ; 21(12): 100426, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36244662

RESUMEN

Despite their diminutive size, islets of Langerhans play a large role in maintaining systemic energy balance in the body. New technologies have enabled us to go from studying the whole pancreas to isolated whole islets, to partial islet sections, and now to islet substructures isolated from within the islet. Using a microfluidic nanodroplet-based proteomics platform coupled with laser capture microdissection and field asymmetric waveform ion mobility spectrometry, we present an in-depth investigation of protein profiles specific to features within the islet. These features include the islet-acinar interface vascular tissue, inner islet vasculature, isolated endocrine cells, whole islet with vasculature, and acinar tissue from around the islet. Compared to interface vasculature, unique protein signatures observed in the inner vasculature indicate increased innervation and intra-islet neuron-like crosstalk. We also demonstrate the utility of these data for identifying localized structure-specific drug-target interactions using existing protein/drug binding databases.


Asunto(s)
Islotes Pancreáticos , Islotes Pancreáticos/metabolismo , Proteómica/métodos , Proteínas/metabolismo , Captura por Microdisección con Láser
5.
Bioinformatics ; 37(22): 4202-4208, 2021 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-34132786

RESUMEN

MOTIVATION: Viruses infect, reprogram and kill microbes, leading to profound ecosystem consequences, from elemental cycling in oceans and soils to microbiome-modulated diseases in plants and animals. Although metagenomic datasets are increasingly available, identifying viruses in them is challenging due to poor representation and annotation of viral sequences in databases. RESULTS: Here, we establish efam, an expanded collection of Hidden Markov Model (HMM) profiles that represent viral protein families conservatively identified from the Global Ocean Virome 2.0 dataset. This resulted in 240 311 HMM profiles, each with at least 2 protein sequences, making efam >7-fold larger than the next largest, pan-ecosystem viral HMM profile database. Adjusting the criteria for viral contig confidence from 'conservative' to 'eXtremely Conservative' resulted in 37 841 HMM profiles in our efam-XC database. To assess the value of this resource, we integrated efam-XC into VirSorter viral discovery software to discover viruses from less-studied, ecologically distinct oxygen minimum zone (OMZ) marine habitats. This expanded database led to an increase in viruses recovered from every tested OMZ virome by ∼24% on average (up to ∼42%) and especially improved the recovery of often-missed shorter contigs (<5 kb). Additionally, to help elucidate lesser-known viral protein functions, we annotated the profiles using multiple databases from the DRAM pipeline and virion-associated metaproteomic data, which doubled the number of annotations obtainable by standard, single-database annotation approaches. Together, these marine resources (efam and efam-XC) are provided as searchable, compressed HMM databases that will be updated bi-annually to help maximize viral sequence discovery and study from any ecosystem. AVAILABILITY AND IMPLEMENTATION: The resources are available on the iVirus platform at (doi.org/10.25739/9vze-4143). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Microbiota , Virus , Animales , Proteínas Virales , Programas Informáticos , Metagenómica/métodos
6.
J Proteome Res ; 20(1): 1-13, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-32929967

RESUMEN

The throughput efficiency and increased depth of coverage provided by isobaric-labeled proteomics measurements have led to increased usage of these techniques. However, the structure of missing data is different than unlabeled studies, which prompts the need for this review to compare the efficacy of nine imputation methods on large isobaric-labeled proteomics data sets to guide researchers on the appropriateness of various imputation methods. Imputation methods were evaluated by accuracy, statistical hypothesis test inference, and run time. In general, expectation maximization and random forest imputation methods yielded the best performance, and constant-based methods consistently performed poorly across all data set sizes and percentages of missing values. For data sets with small sample sizes and higher percentages of missing data, results indicate that statistical inference with no imputation may be preferable. On the basis of the findings in this review, there are core imputation methods that perform better for isobaric-labeled proteomics data, but great care and consideration as to whether imputation is the optimal strategy should be given for data sets comprised of a small number of samples.


Asunto(s)
Algoritmos , Proteómica
7.
Nat Methods ; 15(7): 554, 2018 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-29899368

RESUMEN

In the version of this article initially published, the authors erroneously reported the search mode that was used for ProSightPC 3.0 in the Online Methods and in Supplementary Table 3.

8.
Mol Cell Proteomics ; 18(8 suppl 1): S26-S36, 2019 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-31227600

RESUMEN

Phosphorylation of proteins is a key way cells regulate function, both at the individual protein level and at the level of signaling pathways. Kinases are responsible for phosphorylation of substrates, generally on serine, threonine, or tyrosine residues. Though particular sequence patterns can be identified that dictate whether a residue will be phosphorylated by a specific kinase, these patterns are not highly predictive of phosphorylation. The availability of large scale proteomic and phosphoproteomic data sets generated using mass-spectrometry-based approaches provides an opportunity to study the important relationship between kinase activity, substrate specificity, and phosphorylation. In this study, we analyze relationships between protein abundance and phosphopeptide abundance across more than 150 tumor samples and show that phosphorylation at specific phosphosites is not well correlated with overall kinase abundance. However, individual kinases show a clear and statistically significant difference in correlation among known phosphosite targets for that kinase and randomly selected phosphosites. We further investigate relationships between phosphorylation of known activating or inhibitory sites on kinases and phosphorylation of their target phosphosites. Combined with motif-based analysis, this approach can predict novel kinase targets and show which subsets of a kinase's target repertoire are specifically active in one condition versus another.


Asunto(s)
Fosfoproteínas/metabolismo , Proteínas Quinasas/metabolismo , Humanos , Neoplasias/metabolismo , Fosforilación , Proteómica
9.
J Proteome Res ; 19(4): 1863-1872, 2020 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-32175737

RESUMEN

Proteins with deamidated/citrullinated amino acids play critical roles in the pathogenesis of many human diseases; however, identifying these modifications in complex biological samples has been an ongoing challenge. Herein we present a method to accurately identify these modifications from shotgun proteomics data generated by a deep proteome profiling study of human pancreatic islets obtained by laser capture microdissection. All MS/MS spectra were searched twice using MSGF+ database matching, with and without a dynamic +0.9840 Da mass shift modification on amino acids asparagine, glutamine, and arginine (NQR). Consequently, each spectrum generates two peptide-to-spectrum matches (PSMs) with MSGF+ scores, which were used for the Delta Score calculation. It was observed that all PSMs with positive Delta Score values were clustered with mass errors around 0 ppm, while PSMs with negative Delta Score values were distributed nearly equally within the defined mass error range (20 ppm) for database searching. To estimate false discovery rate (FDR) of modified peptides, a "target-mock" strategy was applied in which data sets were searched against a concatenated database containing "real-modified" (+0.9840 Da) and "mock-modified" (+1.0227 Da) peptide masses. The FDR was controlled to ∼2% using a Delta Score filter value greater than zero. Manual inspection of spectra showed that PSMs with positive Delta Score values contained deamidated/citrullinated fragments in their MS/MS spectra. Many citrullinated sites identified in this study were biochemically confirmed as autoimmunogenic epitopes of autoimmune diseases in literature. The results demonstrated that in situ deamidated/citrullinated peptides can be accurately identified from shotgun tissue proteomics data using this dual-search Delta Score strategy. Raw MS data is available at ProteomeXchange (PXD010150).


Asunto(s)
Citrulinación , Proteómica , Algoritmos , Bases de Datos de Proteínas , Humanos , Péptidos/metabolismo , Proteínas , Espectrometría de Masas en Tándem
10.
J Proteome Res ; 19(7): 2863-2872, 2020 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-32407631

RESUMEN

Label-free quantitative proteomics has become an increasingly popular tool for profiling global protein abundances. However, one major limitation is the potential performance drift of the LC-MS platform over time, which, in turn, limits its utility for analyzing large-scale sample sets. To address this, we introduce an experimental and data analysis scheme based on a block design with common references within each block for enabling large-scale label-free quantification. In this scheme, a large number of samples (e.g., >100 samples) are analyzed in smaller and more manageable blocks, minimizing instrument drift and variability within individual blocks. Each designated block also contains common reference samples (e.g., controls) for normalization across all blocks. We demonstrated the robustness of this approach by profiling the proteome response of human macrophage THP-1 cells to 11 engineered nanomaterials at two different doses. A total of 116 samples were analyzed in six blocks, yielding an average coverage of 4500 proteins per sample. Following a common reference-based correction, 2537 proteins were quantified with high reproducibility without any imputation of missing values from 116 data sets. The data revealed the consistent quantification of proteins across all six blocks, as illustrated by the highly consistent abundances of house-keeping proteins in all samples and the high levels of correlation among samples from different blocks. The data also demonstrated that label-free quantification is robust and accurate enough to quantify even very subtle abundance changes as well as large fold-changes. Our streamlined workflow is easy to implement and can be readily adapted to other large cohort studies for reproducible label-free proteome quantification.


Asunto(s)
Proteoma , Proteómica , Cromatografía Liquida , Humanos , Espectrometría de Masas , Reproducibilidad de los Resultados , Células THP-1
11.
Nat Methods ; 14(9): 909-914, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28783154

RESUMEN

Top-down proteomics, the analysis of intact proteins in their endogenous form, preserves valuable information about post-translation modifications, isoforms and proteolytic processing. The quality of top-down liquid chromatography-tandem MS (LC-MS/MS) data sets is rapidly increasing on account of advances in instrumentation and sample-processing protocols. However, top-down mass spectra are substantially more complex than conventional bottom-up data. New algorithms and software tools for confident proteoform identification and quantification are needed. Here we present Informed-Proteomics, an open-source software suite for top-down proteomics analysis that consists of an LC-MS feature-finding algorithm, a database search algorithm, and an interactive results viewer. We compare our tool with several other popular tools using human-in-mouse xenograft luminal and basal breast tumor samples that are known to have significant differences in protein abundance based on bottom-up analysis.


Asunto(s)
Cromatografía Líquida de Alta Presión/métodos , Proteoma/análisis , Proteoma/química , Programas Informáticos , Espectrometría de Masas en Tándem/métodos , Interfaz Usuario-Computador , Algoritmos , Lenguajes de Programación , Proteómica/métodos , Integración de Sistemas
12.
Mol Cell Proteomics ; 17(9): 1824-1836, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29666158

RESUMEN

Liquid chromatography-mass spectrometry (LC-MS)-based proteomics studies of large sample cohorts can easily require from months to years to complete. Acquiring consistent, high-quality data in such large-scale studies is challenging because of normal variations in instrumentation performance over time, as well as artifacts introduced by the samples themselves, such as those because of collection, storage and processing. Existing quality control methods for proteomics data primarily focus on post-hoc analysis to remove low-quality data that would degrade downstream statistics; they are not designed to evaluate the data in near real-time, which would allow for interventions as soon as deviations in data quality are detected. In addition to flagging analyses that demonstrate outlier behavior, evaluating how the data structure changes over time can aide in understanding typical instrument performance or identify issues such as a degradation in data quality because of the need for instrument cleaning and/or re-calibration. To address this gap for proteomics, we developed Quality Control Analysis in Real-Time (QC-ART), a tool for evaluating data as they are acquired to dynamically flag potential issues with instrument performance or sample quality. QC-ART has similar accuracy as standard post-hoc analysis methods with the additional benefit of real-time analysis. We demonstrate the utility and performance of QC-ART in identifying deviations in data quality because of both instrument and sample issues in near real-time for LC-MS-based plasma proteomics analyses of a sample subset of The Environmental Determinants of Diabetes in the Young cohort. We also present a case where QC-ART facilitated the identification of oxidative modifications, which are often underappreciated in proteomic experiments.


Asunto(s)
Sistemas de Computación , Proteómica/métodos , Proteómica/normas , Control de Calidad , Espectrometría de Masas en Tándem/métodos , Algoritmos , Estudios de Cohortes , Bases de Datos de Proteínas , Humanos , Marcaje Isotópico , Oxidación-Reducción , Péptidos/metabolismo , Curva ROC , Interfaz Usuario-Computador
13.
Mol Cell Proteomics ; 17(9): 1864-1874, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29941660

RESUMEN

Current mass spectrometry (MS)-based proteomics approaches are ineffective for mapping protein expression in tissue sections with high spatial resolution because of the limited overall sensitivity of conventional workflows. Here we report an integrated and automated method to advance spatially resolved proteomics by seamlessly coupling laser capture microdissection (LCM) with a recently developed nanoliter-scale sample preparation system termed nanoPOTS (Nanodroplet Processing in One pot for Trace Samples). The workflow is enabled by prepopulating nanowells with DMSO, which serves as a sacrificial capture liquid for microdissected tissues. The DMSO droplets efficiently collect laser-pressure catapulted LCM tissues as small as 20 µm in diameter with success rates >87%. We also demonstrate that tissue treatment with DMSO can significantly improve proteome coverage, likely due to its ability to dissolve lipids from tissue and enhance protein extraction efficiency. The LCM-nanoPOTS platform was able to identify 180, 695, and 1827 protein groups on average from 12-µm-thick rat brain cortex tissue sections having diameters of 50, 100, and 200 µm, respectively. We also analyzed 100-µm-diameter sections corresponding to 10-18 cells from three different regions of rat brain and comparatively quantified ∼1000 proteins, demonstrating the potential utility for high-resolution spatially resolved mapping of protein expression in tissues.


Asunto(s)
Captura por Microdisección con Láser , Nanopartículas/química , Proteoma/metabolismo , Proteómica/métodos , Animales , Automatización , Encéfalo/metabolismo , Dimetilsulfóxido/química , Femenino , Humanos , Péptidos/metabolismo , Análisis de Componente Principal , Ratas Sprague-Dawley , Ensayos Antitumor por Modelo de Xenoinjerto
14.
Anal Chem ; 91(15): 9707-9715, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31241912

RESUMEN

Two-dimensional reversed-phase capillary liquid chromatography (2D RPLC) separations have enabled comprehensive proteome profiling of biological systems. However, milligram sample quantities of proteins are typically required due to significant losses during offline fractionation. Such a large sample requirement generally precludes the application samples in the nanogram to low-microgram range. To achieve in-depth proteomic analysis of such small-sized samples, we have developed the nanoFAC (nanoflow Fractionation and Automated Concatenation) 2D RPLC platform, in which the first dimension high-pH fractionation was performed on a 75-µm i.d. capillary column at a 300 nL/min flow rate with automated fraction concatenation, instead of on a typically used 2.1 mm column at a 200 µL/min flow rate with manual concatenation. Each fraction was then fully transferred to the second-dimension low-pH nanoLC separation using an autosampler equipped with a custom-machined syringe. We have found that using a polypropylene 96-well plate as collection device as well as the addition of n-Dodecyl ß-d-maltoside (0.01%) in the collection buffer can significantly improve sample recovery. We have demonstrated the nanoFAC 2D RPLC platform can achieve confident identifications of ∼49,000-94,000 unique peptides, corresponding to ∼6,700-8,300 protein groups using only 100-1000 ng of HeLa tryptic digest (equivalent to ∼500-5,000 cells). Furthermore, by integrating with phosphopeptide enrichment, the nanoFAC 2D RPLC platform can identify ∼20,000 phosphopeptides from 100 µg of MCF-7 cell lysate.


Asunto(s)
Automatización , Cromatografía de Fase Inversa/métodos , Nanotecnología/métodos , Fosfoproteínas/química , Cromatografía de Fase Inversa/instrumentación , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Células MCF-7 , Nanotecnología/instrumentación , Shewanella
15.
Anal Chem ; 91(9): 5794-5801, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-30843680

RESUMEN

Comprehensive phosphoproteomic analysis of small populations of cells remains a daunting task due primarily to the insufficient MS signal intensity from low concentrations of enriched phosphopeptides. Isobaric labeling has a unique multiplexing feature where the "total" peptide signal from all channels (or samples) triggers MS/MS fragmentation for peptide identification, while the reporter ions provide quantitative information. In light of this feature, we tested the concept of using a "boosting" sample (e.g., a biological sample mimicking the study samples but available in a much larger quantity) in multiplexed analysis to enable sensitive and comprehensive quantitative phosphoproteomic measurements with <100 000 cells. This simple boosting to amplify signal with isobaric labeling (BASIL) strategy increased the overall number of quantifiable phosphorylation sites more than 4-fold. Good reproducibility in quantification was demonstrated with a median CV of 15.3% and Pearson correlation coefficient of 0.95 from biological replicates. A proof-of-concept experiment demonstrated the ability of BASIL to distinguish acute myeloid leukemia cells based on the phosphoproteome data. Moreover, in a pilot application, this strategy enabled quantitative analysis of over 20 000 phosphorylation sites from human pancreatic islets treated with interleukin-1ß and interferon-γ. Together, this signal boosting strategy provides an attractive solution for comprehensive and quantitative phosphoproteome profiling of relatively small populations of cells where traditional phosphoproteomic workflows lack sufficient sensitivity.


Asunto(s)
Interferón gamma/farmacología , Interleucina-1beta/farmacología , Islotes Pancreáticos/metabolismo , Fosfopéptidos/metabolismo , Fosfoproteínas/metabolismo , Coloración y Etiquetado/métodos , Espectrometría de Masas en Tándem/métodos , Antivirales/farmacología , Humanos , Islotes Pancreáticos/citología , Islotes Pancreáticos/efectos de los fármacos , Fosforilación
16.
Ann Neurol ; 84(1): 78-88, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29908079

RESUMEN

OBJECTIVE: Previous gene expression analysis identified a network of coexpressed genes that is associated with ß-amyloid neuropathology and cognitive decline in older adults. The current work targeted influential genes in this network with quantitative proteomics to identify potential novel therapeutic targets. METHODS: Data came from 834 community-based older persons who were followed annually, died, and underwent brain autopsy. Uniform structured postmortem evaluations assessed the burden of ß-amyloid and other common age-related neuropathologies. Selected reaction monitoring quantified cortical protein abundance of 12 genes prioritized from a molecular network of aging human brain that is implicated in Alzheimer's dementia. Regression and linear mixed models examined the protein associations with ß-amyloid load and other neuropathological indices as well as cognitive decline over multiple years preceding death. RESULTS: Average age at death was 88.6 years. Overall, 349 participants (41.9%) had Alzheimer's dementia at death. A higher level of PLXNB1 abundance was associated with more ß-amyloid load (p = 1.0 × 10-7 ) and higher PHFtau tangle density (p = 2.3 × 10-7 ), and the association of PLXNB1 with cognitive decline is mediated by these known Alzheimer's disease pathologies. On the other hand, higher IGFBP5, HSPB2, and AK4 and lower ITPK1 levels were associated with faster cognitive decline, and, unlike PLXNB1, these associations were not fully explained by common neuropathological indices, suggesting novel mechanisms leading to cognitive decline. INTERPRETATION: Using targeted proteomics, this work identified cortical proteins involved in Alzheimer's dementia and begins to dissect two different molecular pathways: one affecting ß-amyloid deposition and another affecting resilience without a known pathological footprint. Ann Neurol 2018;83:78-88.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Encéfalo/metabolismo , Proteoma/metabolismo , Proteómica/métodos , Anciano de 80 o más Años , Enfermedad de Alzheimer/complicaciones , Péptidos beta-Amiloides/metabolismo , Autopsia , Trastornos del Conocimiento/etiología , Proteínas de Unión al ADN , Femenino , Proteínas de Choque Térmico HSP27/metabolismo , Humanos , Proteína 5 de Unión a Factor de Crecimiento Similar a la Insulina/metabolismo , Masculino , Proteínas del Tejido Nervioso/metabolismo , Pruebas Neuropsicológicas , Mapas de Interacción de Proteínas , Proteoma/genética , Receptores de Superficie Celular/metabolismo , Características de la Residencia
17.
Analyst ; 144(3): 794-807, 2019 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-30507980

RESUMEN

Mass-spectrometry based omics technologies - namely proteomics, metabolomics and lipidomics - have enabled the molecular level systems biology investigation of organisms in unprecedented detail. There has been increasing interest for gaining a thorough, functional understanding of the biological consequences associated with cellular heterogeneity in a wide variety of research areas such as developmental biology, precision medicine, cancer research and microbiome science. Recent advances in mass spectrometry (MS) instrumentation and sample handling strategies are quickly making comprehensive omics analyses of single cells feasible, but key breakthroughs are still required to push through remaining bottlenecks. In this review, we discuss the challenges faced by single cell MS-based omics analyses and highlight recent technological advances that collectively can contribute to comprehensive and high throughput omics analyses in single cells. We provide a vision of the potential of integrating pioneering technologies such as Structures for Lossless Ion Manipulations (SLIM) for improved sensitivity and resolution, novel peptide identification tactics and standards free metabolomics approaches for future applications in single cell analysis.


Asunto(s)
Genómica/métodos , Espectrometría de Masas/métodos , Metabolómica/métodos , Proteómica/métodos , Análisis de la Célula Individual/métodos , Humanos , Medicina de Precisión , Biología de Sistemas
18.
Anal Bioanal Chem ; 411(19): 4587-4596, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30460388

RESUMEN

Extending proteomics to smaller samples can enable the mapping of protein expression across tissues with high spatial resolution and can reveal sub-group heterogeneity. However, despite the continually improving sensitivity of LC-MS instrumentation, in-depth profiling of samples containing low-nanogram amounts of protein has remained challenging due to analyte losses incurred during preparation and analysis. To address this, we recently developed nanodroplet processing in one pot for trace samples (nanoPOTS), a robotic/microfluidic platform that generates ready-to-analyze peptides from cellular material in ~200 nL droplets with greatly reduced sample losses. In combination with ultrasensitive LC-MS, nanoPOTS has enabled >3000 proteins to be confidently identified from as few as 10 cultured human cells and ~700 proteins from single cells. However, the nanoPOTS platform requires a highly skilled operator and a costly in-house-built robotic nanopipetting instrument. In this work, we sought to evaluate the extent to which the benefits of nanodroplet processing could be preserved when upscaling reagent dispensing volumes by a factor of 10 to those addressable by commercial micropipette. We characterized the resulting platform, termed microdroplet processing in one pot for trace samples (µPOTS), for the analysis of as few as ~25 cultured HeLa cells (4 ng total protein) or 50 µm square mouse liver tissue thin sections and found that ~1800 and ~1200 unique proteins were respectively identified with high reproducibility. The reduced equipment requirements should facilitate broad dissemination of nanoproteomics workflows by obviating the need for a capital-intensive custom liquid handling system.


Asunto(s)
Proteómica/métodos , Flujo de Trabajo , Animales , Cromatografía Liquida/métodos , Células HeLa , Humanos , Hígado/metabolismo , Espectrometría de Masas/métodos , Ratones Endogámicos C57BL , Microfluídica , Reproducibilidad de los Resultados , Extracción en Fase Sólida/métodos
19.
Brain ; 141(9): 2721-2739, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-30137212

RESUMEN

Our hypothesis is that changes in gene and protein expression are crucial to the development of late-onset Alzheimer’s disease. Previously we examined how DNA alleles control downstream expression of RNA transcripts and how those relationships are changed in late-onset Alzheimer’s disease. We have now examined how proteins are incorporated into networks in two separate series and evaluated our outputs in two different cell lines. Our pipeline included the following steps: (i) predicting expression quantitative trait loci; (ii) determining differential expression; (iii) analysing networks of transcript and peptide relationships; and (iv) validating effects in two separate cell lines. We performed all our analysis in two separate brain series to validate effects. Our two series included 345 samples in the first set (177 controls, 168 cases; age range 65–105; 58% female; KRONOSII cohort) and 409 samples in the replicate set (153 controls, 141 cases, 115 mild cognitive impairment; age range 66–107; 63% female; RUSH cohort). Our top target is heat shock protein family A member 2 (HSPA2), which was identified as a key driver in our two datasets. HSPA2 was validated in two cell lines, with overexpression driving further elevation of amyloid-β40 and amyloid-β42 levels in APP mutant cells, as well as significant elevation of microtubule associated protein tau and phosphorylated-tau in a modified neuroglioma line. This work further demonstrates that studying changes in gene and protein expression is crucial to understanding late onset disease and further nominates HSPA2 as a specific key regulator of late-onset Alzheimer’s disease processes.10.1093/brain/awy215_video1awy215media15824729224001.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/fisiopatología , Proteínas HSP70 de Choque Térmico/fisiología , Anciano , Anciano de 80 o más Años , Enfermedad de Alzheimer/metabolismo , Encéfalo/metabolismo , Mapeo Encefálico/métodos , Línea Celular , Femenino , Perfilación de la Expresión Génica/métodos , Células HEK293 , Proteínas HSP70 de Choque Térmico/genética , Proteínas HSP70 de Choque Térmico/metabolismo , Humanos , Masculino , Red Nerviosa/fisiopatología , Procesamiento Proteico-Postraduccional , ARN/análisis , ARN/metabolismo , Transcriptoma/genética
20.
Expert Rev Proteomics ; 15(11): 865-871, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30375896

RESUMEN

INTRODUCTION: Nanoproteomics, which is defined as quantitative proteome profiling of small populations of cells (<5000 cells), can reveal critical information related to rare cell populations, hard-to-obtain clinical specimens, and the cellular heterogeneity of pathological tissues. Areas covered: We present a brief review of the recent technological advances in nanoproteomics. These advances include new technologies or approaches covering major areas of proteomics workflow ranging from sample isolation, sample processing, high-resolution separations, to MS instrumentation. Expert commentary: We comment on the current state of nanoproteomics and discuss perspectives on both future technological directions and potential enabling applications.


Asunto(s)
Nanotecnología/métodos , Proteómica/métodos , Animales , Cromatografía Liquida/instrumentación , Cromatografía Liquida/métodos , Electroforesis Capilar/instrumentación , Electroforesis Capilar/métodos , Captura por Microdisección con Láser/métodos , Mamíferos , Nanotecnología/instrumentación , Procesamiento Proteico-Postraduccional , Proteómica/instrumentación , Análisis de la Célula Individual , Espectrometría de Masa por Ionización de Electrospray/instrumentación , Espectrometría de Masa por Ionización de Electrospray/métodos , Flujo de Trabajo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA