Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
1.
Mol Syst Biol ; 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38907068

RESUMEN

Mass spectrometry has revolutionized cell signaling research by vastly simplifying the analysis of many thousands of phosphorylation sites in the human proteome. Defining the cellular response to perturbations is crucial for further illuminating the functionality of the phosphoproteome. Here we describe µPhos ('microPhos'), an accessible phosphoproteomics platform that permits phosphopeptide enrichment from 96-well cell culture and small tissue amounts in <8 h total processing time. By greatly minimizing transfer steps and liquid volumes, we demonstrate increased sensitivity, >90% selectivity, and excellent quantitative reproducibility. Employing highly sensitive trapped ion mobility mass spectrometry, we quantify ~17,000 Class I phosphosites in a human cancer cell line using 20 µg starting material, and confidently localize ~6200 phosphosites from 1 µg. This depth covers key signaling pathways, rendering sample-limited applications and perturbation experiments with hundreds of samples viable. We employ µPhos to study drug- and time-dependent response signatures in a leukemia cell line, and by quantifying 30,000 Class I phosphosites in the mouse brain we reveal distinct spatial kinase activities in subregions of the hippocampal formation.

2.
Mol Cell Proteomics ; 22(3): 100503, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36682716

RESUMEN

Acute myeloid leukemia (AML) is the most common and aggressive form of acute leukemia, with a 5-year survival rate of just 24%. Over a third of all AML patients harbor activating mutations in kinases, such as the receptor tyrosine kinases FLT3 (receptor-type tyrosine-protein kinase FLT3) and KIT (mast/stem cell growth factor receptor kit). FLT3 and KIT mutations are associated with poor clinical outcomes and lower remission rates in response to standard-of-care chemotherapy. We have recently identified that the core kinase of the non-homologous end joining DNA repair pathway, DNA-PK (DNA-dependent protein kinase), is activated downstream of FLT3; and targeting DNA-PK sensitized FLT3-mutant AML cells to standard-of-care therapies. Herein, we investigated DNA-PK as a possible therapeutic vulnerability in KIT mutant AML, using isogenic FDC-P1 mouse myeloid progenitor cell lines transduced with oncogenic mutant KIT (V560G and D816V) or vector control. Targeted quantitative phosphoproteomic profiling identified phosphorylation of DNA-PK in the T2599/T2605/S2608/S2610 cluster in KIT mutant cells, indicative of DNA-PK activation. Accordingly, proliferation assays revealed that KIT mutant FDC-P1 cells were more sensitive to the DNA-PK inhibitors M3814 or NU7441, compared with empty vector controls. DNA-PK inhibition combined with inhibition of KIT signaling using the kinase inhibitors dasatinib or ibrutinib, or the protein phosphatase 2A activators FTY720 or AAL(S), led to synergistic cell death. Global phosphoproteomic analysis of KIT-D816V cells revealed that dasatinib and M3814 single-agent treatments inhibited extracellular signal-regulated kinase and AKT (RAC-alpha serine/threonine-protein kinase)/MTOR (serine/threonine-protein kinase mTOR) activity, with greater inhibition of both pathways when used in combination. Combined dasatinib and M3814 treatment also synergistically inhibited phosphorylation of the transcriptional regulators MYC and MYB. This study provides insight into the oncogenic pathways regulated by DNA-PK beyond its canonical role in DNA repair and demonstrates that DNA-PK is a promising therapeutic target for KIT mutant cancers.


Asunto(s)
Proteína Quinasa Activada por ADN , Leucemia Mieloide Aguda , Animales , Ratones , Apoptosis , Línea Celular Tumoral , Dasatinib , ADN , Proteína Quinasa Activada por ADN/genética , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/tratamiento farmacológico , Mutación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas Receptoras , Serina , Transducción de Señal , Treonina , Serina-Treonina Quinasas TOR , Tirosina
3.
Biochem J ; 479(11): 1237-1256, 2022 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-35594055

RESUMEN

Trafficking regulator of GLUT4-1, TRARG1, positively regulates insulin-stimulated GLUT4 trafficking and insulin sensitivity. However, the mechanism(s) by which this occurs remain(s) unclear. Using biochemical and mass spectrometry analyses we found that TRARG1 is dephosphorylated in response to insulin in a PI3K/Akt-dependent manner and is a novel substrate for GSK3. Priming phosphorylation of murine TRARG1 at serine 84 allows for GSK3-directed phosphorylation at serines 72, 76 and 80. A similar pattern of phosphorylation was observed in human TRARG1, suggesting that our findings are translatable to human TRARG1. Pharmacological inhibition of GSK3 increased cell surface GLUT4 in cells stimulated with a submaximal insulin dose, and this was impaired following Trarg1 knockdown, suggesting that TRARG1 acts as a GSK3-mediated regulator in GLUT4 trafficking. These data place TRARG1 within the insulin signaling network and provide insights into how GSK3 regulates GLUT4 trafficking in adipocytes.


Asunto(s)
Glucógeno Sintasa Quinasa 3 , Fosfatidilinositol 3-Quinasas , Adipocitos/metabolismo , Animales , Membrana Celular/metabolismo , Glucosa/metabolismo , Transportador de Glucosa de Tipo 4/genética , Transportador de Glucosa de Tipo 4/metabolismo , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Humanos , Insulina/metabolismo , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina/metabolismo
4.
Elife ; 102021 07 13.
Artículo en Inglés | MEDLINE | ID: mdl-34253290

RESUMEN

The phosphoinositide 3-kinase (PI3K)-Akt network is tightly controlled by feedback mechanisms that regulate signal flow and ensure signal fidelity. A rapid overshoot in insulin-stimulated recruitment of Akt to the plasma membrane has previously been reported, which is indicative of negative feedback operating on acute timescales. Here, we show that Akt itself engages this negative feedback by phosphorylating insulin receptor substrate (IRS) 1 and 2 on a number of residues. Phosphorylation results in the depletion of plasma membrane-localised IRS1/2, reducing the pool available for interaction with the insulin receptor. Together these events limit plasma membrane-associated PI3K and phosphatidylinositol (3,4,5)-trisphosphate (PIP3) synthesis. We identified two Akt-dependent phosphorylation sites in IRS2 at S306 (S303 in mouse) and S577 (S573 in mouse) that are key drivers of this negative feedback. These findings establish a novel mechanism by which the kinase Akt acutely controls PIP3 abundance, through post-translational modification of the IRS scaffold.


For the body to work properly, cells must constantly 'talk' to each other using signalling molecules. Receiving a chemical signal triggers a series of molecular events in a cell, a so-called 'signal transduction pathway' that connects a signal with a precise outcome. Disturbing cell signalling can trigger disease, and strict control mechanisms are therefore in place to ensure that communication does not break down or become erratic. For instance, just as a thermostat turns off the heater once the right temperature is reached, negative feedback mechanisms in cells switch off signal transduction pathways when the desired outcome has been achieved. The hormone insulin is a signal for growth that increases in the body following a meal to promote the storage of excess blood glucose (sugar) in muscle and fat cells. The hormone binds to insulin receptors at the cell surface and switches on a signal transduction pathway that makes the cell take up glucose from the bloodstream. If the signal is not engaged diseases such as diabetes develop. Conversely, if the signal cannot be adequately switched of cancer can develop. Determining exactly how insulin works would help to understand these diseases better and to develop new treatments. Kearney et al. therefore set out to examine the biochemical 'fail-safes' that control insulin signalling. Experiments using computer simulations of the insulin signalling pathway revealed a potential new mechanism for negative feedback, which centred on a molecule known as Akt. The models predicted that if the negative feedback were removed, then Akt would become hyperactive and accumulate at the cell's surface after stimulation with insulin. Further manipulation of the 'virtual' insulin signalling pathway and studies of live cells in culture confirmed that this was indeed the case. The cell biology experiments also showed how Akt, once at the cell surface, was able to engage the negative feedback and shut down further insulin signalling. Akt did this by inactivating a protein required to pass the signal from the insulin receptor to the rest of the cell. Overall, this work helps to understand cell communication by revealing a previously unknown, and critical component of the insulin signalling pathway.


Asunto(s)
Fosfatidilinositol 3-Quinasa/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptor de Insulina/metabolismo , Animales , Antígenos CD , Membrana Celular/metabolismo , Biología Computacional , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina/metabolismo , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Fosforilación , Transducción de Señal/fisiología
5.
Cell Mol Life Sci ; 78(8): 4035-4052, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33834258

RESUMEN

The mechanistic target of rapamycin complex 1 (mTORC1) is an important regulator of cellular metabolism that is commonly hyperactivated in cancer. Recent cancer genome screens have identified multiple mutations in Ras-homolog enriched in brain (Rheb), the primary activator of mTORC1 that might act as driver oncogenes by causing hyperactivation of mTORC1. Here, we show that a number of recurrently occurring Rheb mutants drive hyperactive mTORC1 signalling through differing levels of insensitivity to the primary inactivator of Rheb, tuberous sclerosis complex. We show that two activated mutants, Rheb-T23M and E40K, strongly drive increased cell growth, proliferation and anchorage-independent growth resulting in enhanced tumour growth in vivo. Proteomic analysis of cells expressing the mutations revealed, surprisingly, that these two mutants promote distinct oncogenic pathways with Rheb-T23M driving an increased rate of anaerobic glycolysis, while Rheb-E40K regulates the translation factor eEF2 and autophagy, likely through differential interactions with 5' AMP-activated protein kinase (AMPK) which modulate its activity. Our findings suggest that unique, personalized, combination therapies may be utilised to treat cancers according to which Rheb mutant they harbour.


Asunto(s)
Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Neoplasias/genética , Mutación Puntual , Proteína Homóloga de Ras Enriquecida en el Cerebro/genética , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteína 2 del Complejo de la Esclerosis Tuberosa/metabolismo , Animales , Células HEK293 , Células HeLa , Humanos , Ratones , Modelos Moleculares , Células 3T3 NIH , Neoplasias/metabolismo , Proteoma/metabolismo , Proteómica , Proteína Homóloga de Ras Enriquecida en el Cerebro/metabolismo , Transducción de Señal
6.
Cell Death Discov ; 7(1): 81, 2021 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-33863878

RESUMEN

Both tumour suppressive and oncogenic functions have been reported for dual-specificity tyrosine phosphorylation-regulated kinase 1A (DYRK1A). Herein, we performed a detailed investigation to delineate the role of DYRK1A in glioblastoma. Our phosphoproteomic and mechanistic studies show that DYRK1A induces degradation of cyclin B by phosphorylating CDC23, which is necessary for the function of the anaphase-promoting complex, a ubiquitin ligase that degrades mitotic proteins. DYRK1A inhibition leads to the accumulation of cyclin B and activation of CDK1. Importantly, we established that the phenotypic response of glioblastoma cells to DYRK1A inhibition depends on both retinoblastoma (RB) expression and the degree of residual DYRK1A activity. Moderate DYRK1A inhibition leads to moderate cyclin B accumulation, CDK1 activation and increased proliferation in RB-deficient cells. In RB-proficient cells, cyclin B/CDK1 activation in response to DYRK1A inhibition is neutralized by the RB pathway, resulting in an unchanged proliferation rate. In contrast, complete DYRK1A inhibition with high doses of inhibitors results in massive cyclin B accumulation, saturation of CDK1 activity and cell cycle arrest, regardless of RB status. These findings provide new insights into the complexity of context-dependent DYRK1A signalling in cancer cells.

7.
Cell Rep ; 34(8): 108771, 2021 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-33626354

RESUMEN

Mass spectrometry (MS)-based phosphoproteomics has revolutionized our ability to profile phosphorylation-based signaling in cells and tissues on a global scale. To infer the action of kinases and signaling pathways in phosphoproteomic experiments, we present PhosR, a set of tools and methodologies implemented in a suite of R packages facilitating comprehensive analysis of phosphoproteomic data. By applying PhosR to both published and new phosphoproteomic datasets, we demonstrate capabilities in data imputation and normalization by using a set of "stably phosphorylated sites" and in functional analysis for inferring active kinases and signaling pathways. In particular, we introduce a "signalome" construction method for identifying a collection of signaling modules to summarize and visualize the interaction of kinases and their collective actions on signal transduction. Together, our data and findings demonstrate the utility of PhosR in processing and generating biological knowledge from MS-based phosphoproteomic data.


Asunto(s)
Hígado/metabolismo , Espectrometría de Masas , Fibras Musculares Esqueléticas/metabolismo , Proteoma , Proteómica , Transducción de Señal , Diseño de Software , Proteínas Quinasas Activadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacología , Animales , Línea Celular Tumoral , Activación Enzimática , Insulina/farmacología , Hígado/efectos de los fármacos , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Fosforilación , Proteoma/efectos de los fármacos , Ratas , Ribonucleótidos/farmacología , Transducción de Señal/efectos de los fármacos
8.
J Neurochem ; 153(2): 173-188, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31742704

RESUMEN

Sphingosine 1-phosphate (S1P) is an essential lipid metabolite that signals through a family of five G protein-coupled receptors, S1PR1-S1PR5, to regulate cell physiology. The multiple sclerosis drug Fingolimod (FTY720) is a potent S1P receptor agonist that causes peripheral lymphopenia. Recent research has demonstrated direct neuroprotective properties of FTY720 in several neurodegenerative paradigms; however, neuroprotective properties of the native ligand S1P have not been established. We aimed to establish the significance of neurotrophic factor up-regulation by S1P for neuroprotection, comparing S1P with FTY720. S1P induced brain-derived neurotrophic factor (BDNF), leukemia inhibitory factor (LIF), platelet-derived growth factor B (PDGFB), and heparin-binding EGF-like growth factor (HBEGF) gene expression in primary human and murine astrocytes, but not in neurons, and to a much greater extent than FTY720. Accordingly, S1P but not FTY720 protected cultured neurons against excitotoxic cell death in a primary murine neuron-glia coculture model, and a neutralizing antibody to LIF blocked this S1P-mediated neuroprotection. Antagonists of S1PR1 and S1PR2 both inhibited S1P-mediated neurotrophic gene induction in human astrocytes, indicating that simultaneous activation of both receptors is required. S1PR2 signaling was transduced through Gα13 and the small GTPase Rho, and was necessary for the up-regulation and activation of the transcription factors FOS and JUN, which regulate LIF, BDNF, and HBEGF transcription. In summary, we show that S1P protects hippocampal neurons against excitotoxic cell death through up-regulation of neurotrophic gene expression, particularly LIF, in astrocytes. This up-regulation requires both S1PR1 and S1PR2 signaling. FTY720 does not activate S1PR2, explaining its relative inefficacy compared to S1P.


Asunto(s)
Astrocitos/metabolismo , Clorhidrato de Fingolimod/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Lisofosfolípidos/farmacología , Factores de Crecimiento Nervioso/biosíntesis , Neuronas/metabolismo , Esfingosina/análogos & derivados , Animales , Astrocitos/efectos de los fármacos , Muerte Celular/efectos de los fármacos , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Humanos , Ratones , Neuronas/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Esfingosina/farmacología , Moduladores de los Receptores de fosfatos y esfingosina 1/farmacología
9.
Nat Commun ; 10(1): 5486, 2019 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-31792197

RESUMEN

Protein oxidation sits at the intersection of multiple signalling pathways, yet the magnitude and extent of crosstalk between oxidation and other post-translational modifications remains unclear. Here, we delineate global changes in adipocyte signalling networks following acute oxidative stress and reveal considerable crosstalk between cysteine oxidation and phosphorylation-based signalling. Oxidation of key regulatory kinases, including Akt, mTOR and AMPK influences the fidelity rather than their absolute activation state, highlighting an unappreciated interplay between these modifications. Mechanistic analysis of the redox regulation of Akt identified two cysteine residues in the pleckstrin homology domain (C60 and C77) to be reversibly oxidized. Oxidation at these sites affected Akt recruitment to the plasma membrane by stabilizing the PIP3 binding pocket. Our data provide insights into the interplay between oxidative stress-derived redox signalling and protein phosphorylation networks and serve as a resource for understanding the contribution of cellular oxidation to a range of diseases.


Asunto(s)
Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adipocitos/metabolismo , Animales , Cisteína/genética , Cisteína/metabolismo , Humanos , Ratones , Oxidación-Reducción , Estrés Oxidativo , Fosfoproteínas/química , Fosfoproteínas/genética , Fosforilación , Dominios Proteicos , Proteoma/química , Proteoma/genética , Proteoma/metabolismo , Proteómica , Proteínas Proto-Oncogénicas c-akt/química , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
10.
J Proteome Res ; 18(8): 3219-3222, 2019 08 02.
Artículo en Inglés | MEDLINE | ID: mdl-31268342

RESUMEN

Limiting contamination of liquid chromatography/tandem mass spectrometry (LC-MS) systems and reducing the downtime associated with maintenance and cleaning is essential for productivity. We developed a simple device that creates a gas curtain barrier to prevent ions from entering the MS inlet. The gas can be quickly and easily applied when certain contaminant ions are known to elute. We show that the device can prevent the buildup of contaminants on the heated transfer capillary following >100 injections of a crude tissue lysate and improves peptide identifications. The device may provide a promising approach toward improving instrument robustness.


Asunto(s)
Cromatografía Liquida/normas , Péptidos/química , Proteómica/normas , Espectrometría de Masas en Tándem/normas , Cromatografía Líquida de Alta Presión , Electroforesis Capilar , Péptidos/aislamiento & purificación
11.
Proteomics ; 19(13): e1900068, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31099962

RESUMEN

The increasing role played by liquid chromatography-mass spectrometry (LC-MS)-based proteomics in biological discovery has led to a growing need for quality control (QC) on the LC-MS systems. While numerous quality control tools have been developed to track the performance of LC-MS systems based on a pre-defined set of performance factors (e.g., mass error, retention time), the precise influence and contribution of the performance factors and their generalization property to different biological samples are not as well characterized. Here, a web-based application (QCMAP) is developed for interactive diagnosis and prediction of the performance of LC-MS systems across different biological sample types. Leveraging on a standardized HeLa cell sample run as QC within a multi-user facility, predictive models are trained on a panel of commonly used performance factors to pinpoint the precise conditions to a (un)satisfactory performance in three LC-MS systems. It is demonstrated that the learned model can be applied to predict LC-MS system performance for brain samples generated from an independent study. By compiling these predictive models into our web-application, QCMAP allows users to benchmark the performance of their LC-MS systems using their own samples and identify key factors for instrument optimization. QCMAP is freely available from: http://shiny.maths.usyd.edu.au/QCMAP/.


Asunto(s)
Cromatografía Liquida/métodos , Proteómica/métodos , Control de Calidad , Espectrometría de Masas en Tándem/métodos , Línea Celular Tumoral , Células HeLa , Humanos , Internet
12.
Mol Metab ; 25: 83-94, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31105056

RESUMEN

OBJECTIVE: Insulin suppresses adipose tissue lipolysis after a meal, playing a key role in metabolic homeostasis. This is mediated via the kinase Akt and its substrate phosphodiesterase 3B (PDE3B). Once phosphorylated and activated, PDE3B hydrolyses cAMP leading to the inactivation of cAMP-dependent protein kinase (PKA) and suppression of lipolysis. However, several gaps have emerged in this model. Here we investigated the role of the PDE3B-interacting protein, α/ß-hydrolase ABHD15 in this process. METHODS: Lipolysis, glucose uptake, and signaling were assessed in ABHD15 knock down and knock out adipocytes and fat explants in response to insulin and/or ß-adrenergic receptor agonist. Glucose and fatty acid metabolism were determined in wild type and ABHD15-/- littermate mice. RESULTS: Deletion of ABHD15 in adipocytes resulted in a significant defect in insulin-mediated suppression of lipolysis with no effect on insulin-mediated glucose uptake. ABHD15 played a role in suppressing PKA signaling as phosphorylation of the PKA substrate Perilipin-1 remained elevated in response to insulin upon ABHD15 deletion. ABHD15-/- mice had normal glucose metabolism but defective fatty acid metabolism: plasma fatty acids were elevated upon fasting and in response to insulin, and this was accompanied by elevated liver triglycerides upon ß-adrenergic receptor activation. This is likely due to hyperactive lipolysis as evident by the larger triglyceride depletion in brown adipose tissue in these mice. Finally, ABHD15 protein levels were reduced in adipocytes from mice fed a Western diet, further implicating this protein in metabolic homeostasis. CONCLUSIONS: Collectively, ABHD15 regulates adipocyte lipolysis and liver lipid accumulation, providing novel therapeutic opportunities for modulating lipid homeostasis in disease.


Asunto(s)
Tejido Adiposo/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Producto de la Acumulación de Lípidos/fisiología , Lipólisis/fisiología , Hígado/metabolismo , Proteínas de la Membrana/metabolismo , Células 3T3-L1 , Adipocitos/metabolismo , Tejido Adiposo/efectos de los fármacos , Animales , Metabolismo de los Hidratos de Carbono , Hidrolasas de Éster Carboxílico/genética , Hidrolasas de Éster Carboxílico/farmacología , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Ayuno , Ácidos Grasos/sangre , Glucosa/metabolismo , Homeostasis , Insulina/metabolismo , Metabolismo de los Lípidos , Lipólisis/efectos de los fármacos , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/farmacología , Ratones , Ratones Noqueados , Perilipina-1/metabolismo , Fosforilación , Transducción de Señal , Triglicéridos
13.
J Proteome Res ; 18(5): 2228-2240, 2019 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-30892045

RESUMEN

Intermittent fasting (IF) increases lifespan and decreases metabolic disease phenotypes and cancer risk in model organisms, but the health benefits of IF in humans are less clear. Human plasma derived from clinical trials is one of the most difficult sample sets to analyze using mass spectrometry-based proteomics due to the extensive sample preparation required and the need to process many samples to achieve statistical significance. Here, we describe an optimized and accessible device (Spin96) to accommodate up to 96 StageTips, a widely used sample preparation medium enabling efficient and consistent processing of samples prior to LC-MS/MS. We have applied this device to the analysis of human plasma from a clinical trial of IF. In this longitudinal study employing 8-weeks IF, we identified significant abundance differences induced by the IF intervention, including increased apolipoprotein A4 (APOA4) and decreased apolipoprotein C2 (APOC2) and C3 (APOC3). These changes correlated with a significant decrease in plasma triglycerides after the IF intervention. Given that these proteins have a role in regulating apolipoprotein particle metabolism, we propose that IF had a positive effect on lipid metabolism through modulation of HDL particle size and function. In addition, we applied a novel human protein variant database to detect common protein variants across the participants. We show that consistent detection of clinically relevant peptides derived from both alleles of many proteins is possible, including some that are associated with human metabolic phenotypes. Together, these findings illustrate the power of accessible workflows for proteomics analysis of clinical samples to yield significant biological insight.


Asunto(s)
Apolipoproteína C-III/sangre , Apolipoproteína C-II/sangre , Apolipoproteínas A/sangre , Ayuno/sangre , Metabolismo de los Lípidos/genética , Proteómica/métodos , Adulto , Anciano , Apolipoproteína C-II/genética , Apolipoproteína C-III/genética , Apolipoproteínas A/genética , Cromatografía Liquida , Bases de Datos de Proteínas , Femenino , Expresión Génica , Humanos , Lipoproteínas HDL/sangre , Lipoproteínas HDL/genética , Estudios Longitudinales , Persona de Mediana Edad , Tamaño de la Partícula , Impresión Tridimensional/instrumentación , Proteómica/instrumentación , Extracción en Fase Sólida , Manejo de Especímenes/métodos , Espectrometría de Masas en Tándem , Triglicéridos/sangre
14.
Nat Protoc ; 13(9): 1897-1916, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30190555

RESUMEN

Mass spectrometry has transformed the field of cell signaling by enabling global studies of dynamic protein phosphorylation ('phosphoproteomics'). Recent developments are enabling increasingly sophisticated phosphoproteomics studies, but practical challenges remain. The EasyPhos workflow addresses these and is sufficiently streamlined to enable the analysis of hundreds of phosphoproteomes at a depth of >10,000 quantified phosphorylation sites. Here we present a detailed and updated workflow that further ensures high performance in sample-limited conditions while also reducing sample preparation time. By eliminating protein precipitation steps and performing the entire protocol, including digestion, in a single 96-well plate, we now greatly minimize opportunities for sample loss and variability. This results in very high reproducibility and a small sample size requirement (≤200 µg of protein starting material). After cell culture or tissue collection, the protocol takes 1 d, whereas mass spectrometry measurements require ~1 h per sample. Applied to glioblastoma cells acutely treated with epidermal growth factor (EGF), EasyPhos quantified 20,132 distinct phosphopeptides from 200 µg of protein in less than 1 d of measurement time, revealing thousands of EGF-regulated phosphorylation events.


Asunto(s)
Espectrometría de Masas/métodos , Fosfoproteínas/análisis , Proteoma/análisis , Proteómica/métodos , Línea Celular Tumoral , Humanos
15.
Science ; 360(6395)2018 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-29930108

RESUMEN

A systems view of G protein-coupled receptor (GPCR) signaling in its native environment is central to the development of GPCR therapeutics with fewer side effects. Using the kappa opioid receptor (KOR) as a model, we employed high-throughput phosphoproteomics to investigate signaling induced by structurally diverse agonists in five mouse brain regions. Quantification of 50,000 different phosphosites provided a systems view of KOR in vivo signaling, revealing novel mechanisms of drug action. Thus, we discovered enrichment of the mechanistic target of rapamycin (mTOR) pathway by U-50,488H, an agonist causing aversion, which is a typical KOR-mediated side effect. Consequently, mTOR inhibition during KOR activation abolished aversion while preserving beneficial antinociceptive and anticonvulsant effects. Our results establish high-throughput phosphoproteomics as a general strategy to investigate GPCR in vivo signaling, enabling prediction and modulation of behavioral outcomes.


Asunto(s)
Encéfalo/metabolismo , Ensayos Analíticos de Alto Rendimiento , Fosfoproteínas/metabolismo , Proteómica/métodos , Receptores Opioides kappa/metabolismo , Transducción de Señal , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/metabolismo , 3,4-Dicloro-N-metil-N-(2-(1-pirrolidinil)-ciclohexil)-bencenacetamida, (trans)-Isómero/farmacología , Analgésicos no Narcóticos/farmacología , Animales , Anticonvulsivantes/farmacología , Arrestinas/metabolismo , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Línea Celular Tumoral , Diterpenos de Tipo Clerodano/metabolismo , Diterpenos de Tipo Clerodano/farmacología , Humanos , Ligandos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenetilaminas/metabolismo , Fenetilaminas/farmacología , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Receptores Opioides kappa/agonistas , Receptores Opioides kappa/genética , Transducción de Señal/efectos de la radiación , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo
16.
Nat Commun ; 7: 13250, 2016 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-27841257

RESUMEN

Insulin-secreting beta cells play an essential role in maintaining physiological blood glucose levels, and their dysfunction leads to the development of diabetes. To elucidate the signalling events regulating insulin secretion, we applied a recently developed phosphoproteomics workflow. We quantified the time-resolved phosphoproteome of murine pancreatic cells following their exposure to glucose and in combination with small molecule compounds that promote insulin secretion. The quantitative phosphoproteome of 30,000 sites clustered into three main groups in concordance with the modulation of the three key kinases: PKA, PKC and CK2A. A high-resolution time course revealed key novel regulatory sites, revealing the importance of methyltransferase DNMT3A phosphorylation in the glucose response. Remarkably a significant proportion of these novel regulatory sites is significantly downregulated in diabetic islets. Control of insulin secretion is embedded in an unexpectedly broad and complex range of cellular functions, which are perturbed by drugs in multiple ways.


Asunto(s)
Glucosa/farmacología , Células Secretoras de Insulina/efectos de los fármacos , Insulina/metabolismo , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular Tumoral , Análisis por Conglomerados , Diabetes Mellitus Tipo 1/metabolismo , Secreción de Insulina , Células Secretoras de Insulina/metabolismo , Islotes Pancreáticos/efectos de los fármacos , Islotes Pancreáticos/metabolismo , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Fosfoproteínas/clasificación , Fosforilación/efectos de los fármacos , Proteómica/métodos
17.
Cell Rep ; 17(1): 29-36, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27681418

RESUMEN

FGF21 improves the metabolic profile of obese animals through its actions on adipocytes. To elucidate the signaling network responsible for mediating these effects, we quantified dynamic changes in the adipocyte phosphoproteome following acute exposure to FGF21. FGF21 regulated a network of 821 phosphosites on 542 proteins. A major FGF21-regulated signaling node was mTORC1/S6K. In contrast to insulin, FGF21 activated mTORC1 via MAPK rather than through the canonical PI3K/AKT pathway. Activation of mTORC1/S6K by FGF21 was surprising because this is thought to contribute to deleterious metabolic effects such as obesity and insulin resistance. Rather, mTORC1 mediated many of the beneficial actions of FGF21 in vitro, including UCP1 and FGF21 induction, increased adiponectin secretion, and enhanced glucose uptake without any adverse effects on insulin action. This study provides a global view of FGF21 signaling and suggests that mTORC1 may act to facilitate FGF21-mediated health benefits in vivo.


Asunto(s)
Adipocitos/efectos de los fármacos , Adiponectina/genética , Factores de Crecimiento de Fibroblastos/farmacología , Complejos Multiproteicos/genética , Fosfoproteínas/genética , Proteínas Quinasas S6 Ribosómicas 90-kDa/genética , Serina-Treonina Quinasas TOR/genética , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Adiponectina/agonistas , Adiponectina/metabolismo , Animales , Diferenciación Celular , Desoxiglucosa/metabolismo , Factores de Crecimiento de Fibroblastos/genética , Factores de Crecimiento de Fibroblastos/metabolismo , Regulación de la Expresión Génica , Redes Reguladoras de Genes/efectos de los fármacos , Inyecciones Intraperitoneales , Marcaje Isotópico , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Ratones Endogámicos C57BL , Complejos Multiproteicos/agonistas , Complejos Multiproteicos/metabolismo , Fosfoproteínas/metabolismo , Proteoma/genética , Proteoma/metabolismo , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Proteínas Quinasas S6 Ribosómicas 90-kDa/metabolismo , Transducción de Señal , Sirolimus/farmacología , Grasa Subcutánea Abdominal/citología , Grasa Subcutánea Abdominal/efectos de los fármacos , Grasa Subcutánea Abdominal/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteína Desacopladora 1/agonistas , Proteína Desacopladora 1/genética , Proteína Desacopladora 1/metabolismo
18.
Cell Rep ; 12(6): 937-43, 2015 Aug 11.
Artículo en Inglés | MEDLINE | ID: mdl-26235620

RESUMEN

The mechanistic target of rapamycin complex 2 (mTORC2) regulates cell survival and cytoskeletal organization by phosphorylating its AGC kinase substrates; however, little is known about the regulation of mTORC2 itself. It was previously reported that Akt phosphorylates the mTORC2 subunit SIN1 at T86, activating mTORC2 through a positive feedback loop, though another study reported that S6K phosphorylates SIN1 at the same site, inhibiting mTORC2 activity. We performed extensive analysis of SIN1 phosphorylation upon inhibition of Akt, S6K, and mTOR under diverse cellular contexts, and we found that, in all cell lines and conditions studied, Akt is the major kinase responsible for SIN1 phosphorylation. These findings refine the activation mechanism of the Akt-mTORC2 signaling branch as follows: PDK1 phosphorylates Akt at T308, increasing Akt kinase activity. Akt phosphorylates SIN1 at T86, enhancing mTORC2 kinase activity, which leads to phosphorylation of Akt S473 by mTORC2, thereby catalyzing full activation of Akt.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Línea Celular , Inhibidores Enzimáticos/farmacología , Células HEK293 , Células HeLa , Humanos , Diana Mecanicista del Complejo 2 de la Rapamicina , Complejos Multiproteicos/genética , Fosforilación/efectos de los fármacos , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Pirimidinas/farmacología , Proteínas Quinasas S6 Ribosómicas 70-kDa/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal/genética , Transducción de Señal/fisiología , Serina-Treonina Quinasas TOR/genética
19.
Sci Signal ; 8(380): rs6, 2015 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-26060331

RESUMEN

A major goal in signaling biology is the establishment of high-throughput quantitative methods for measuring changes in protein phosphorylation of entire signal transduction pathways across many different samples comprising temporal or dose data or patient samples. Data-independent acquisition (DIA) mass spectrometry (MS) methods, which involve tandem MS scans that are collected independently of precursor ion information and then are followed by targeted searching for known peptides, may achieve this goal. We applied DIA-MS to systematically quantify phosphorylation of components in the insulin signaling network in response to insulin as well as in stimulated cells exposed to a panel of kinase inhibitors targeting key downstream effectors in the network. We accurately quantified the effect of insulin on phosphorylation of 86 protein targets in the insulin signaling network using either stable isotope standards (SIS) or label-free quantification (LFQ) and mapped signal transmission through this network. By matching kinases to specific phosphorylation events (based on linear consensus motifs and temporal phosphorylation) to the quantitative phosphoproteomic data from cells exposed to inhibitors, we investigated predicted kinase-substrate relationships of AKT and mTOR in a targeted fashion. Furthermore, we applied this approach to show that AKT2-dependent phosphorylation of GAB2 promoted insulin signaling but inhibited epidermal growth factor (EGF) signaling in a manner dependent on 14-3-3 binding. Because DIA-MS can increase throughput and improve the reproducibility of peptide detection across multiple samples, this approach should facilitate more accurate, comprehensive, and quantitative assessment of signaling networks under various experimental conditions than are possible using other MS proteomic methods.


Asunto(s)
Insulina/metabolismo , Fosfoproteínas/metabolismo , Proteómica/métodos , Transducción de Señal , Espectrometría de Masas en Tándem/métodos , Proteínas 14-3-3/metabolismo , Células 3T3-L1 , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Western Blotting , Inhibidores Enzimáticos/farmacología , Factor de Crecimiento Epidérmico/metabolismo , Factor de Crecimiento Epidérmico/farmacología , Células HEK293 , Humanos , Insulina/farmacología , Ratones , Fosfopéptidos/metabolismo , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina/metabolismo , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Treonina/metabolismo
20.
Cell Metab ; 17(6): 1009-1020, 2013 Jun 04.
Artículo en Inglés | MEDLINE | ID: mdl-23684622

RESUMEN

A major challenge of the post-genomics era is to define the connectivity of protein phosphorylation networks. Here, we quantitatively delineate the insulin signaling network in adipocytes by high-resolution mass spectrometry-based proteomics. These data reveal the complexity of intracellular protein phosphorylation. We identified 37,248 phosphorylation sites on 5,705 proteins in this single-cell type, with approximately 15% responding to insulin. We integrated these large-scale phosphoproteomics data using a machine learning approach to predict physiological substrates of several diverse insulin-regulated kinases. This led to the identification of an Akt substrate, SIN1, a core component of the mTORC2 complex. The phosphorylation of SIN1 by Akt was found to regulate mTORC2 activity in response to growth factors, revealing topological insights into the Akt/mTOR signaling network. The dynamic phosphoproteome described here contains numerous phosphorylation sites on proteins involved in diverse molecular functions and should serve as a useful functional resource for cell biologists.


Asunto(s)
Adipocitos/metabolismo , Proteínas Portadoras/metabolismo , Complejos Multiproteicos/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Células 3T3 , Animales , Línea Celular , Insulina/metabolismo , Espectrometría de Masas , Diana Mecanicista del Complejo 2 de la Rapamicina , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Fosforilación , Proteoma , Proteómica , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA