Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 181(5): 1112-1130.e16, 2020 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-32470399

RESUMEN

Acute physical activity leads to several changes in metabolic, cardiovascular, and immune pathways. Although studies have examined selected changes in these pathways, the system-wide molecular response to an acute bout of exercise has not been fully characterized. We performed longitudinal multi-omic profiling of plasma and peripheral blood mononuclear cells including metabolome, lipidome, immunome, proteome, and transcriptome from 36 well-characterized volunteers, before and after a controlled bout of symptom-limited exercise. Time-series analysis revealed thousands of molecular changes and an orchestrated choreography of biological processes involving energy metabolism, oxidative stress, inflammation, tissue repair, and growth factor response, as well as regulatory pathways. Most of these processes were dampened and some were reversed in insulin-resistant participants. Finally, we discovered biological pathways involved in cardiopulmonary exercise response and developed prediction models revealing potential resting blood-based biomarkers of peak oxygen consumption.


Asunto(s)
Metabolismo Energético/fisiología , Ejercicio Físico/fisiología , Anciano , Biomarcadores/metabolismo , Femenino , Humanos , Insulina/metabolismo , Resistencia a la Insulina , Leucocitos Mononucleares/metabolismo , Estudios Longitudinales , Masculino , Metaboloma , Persona de Mediana Edad , Oxígeno/metabolismo , Consumo de Oxígeno , Proteoma , Transcriptoma
2.
Cell ; 178(5): 1102-1114.e17, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442403

RESUMEN

Caloric restriction is known to improve inflammatory and autoimmune diseases. However, the mechanisms by which reduced caloric intake modulates inflammation are poorly understood. Here we show that short-term fasting reduced monocyte metabolic and inflammatory activity and drastically reduced the number of circulating monocytes. Regulation of peripheral monocyte numbers was dependent on dietary glucose and protein levels. Specifically, we found that activation of the low-energy sensor 5'-AMP-activated protein kinase (AMPK) in hepatocytes and suppression of systemic CCL2 production by peroxisome proliferator-activator receptor alpha (PPARα) reduced monocyte mobilization from the bone marrow. Importantly, we show that fasting improves chronic inflammatory diseases without compromising monocyte emergency mobilization during acute infectious inflammation and tissue repair. These results reveal that caloric intake and liver energy sensors dictate the blood and tissue immune tone and link dietary habits to inflammatory disease outcome.


Asunto(s)
Restricción Calórica , Monocitos/metabolismo , Proteínas Quinasas Activadas por AMP/metabolismo , Adulto , Animales , Antígenos Ly/metabolismo , Células de la Médula Ósea/citología , Células de la Médula Ósea/metabolismo , Quimiocina CCL2/deficiencia , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Femenino , Hepatocitos/citología , Hepatocitos/metabolismo , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/citología , PPAR alfa/deficiencia , PPAR alfa/genética , PPAR alfa/metabolismo
3.
Nat Immunol ; 18(5): 583-593, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28263321

RESUMEN

The immune system is unique in its dynamic interplay between numerous cell types. However, a system-wide view of how immune cells communicate to protect against disease has not yet been established. We applied high-resolution mass-spectrometry-based proteomics to characterize 28 primary human hematopoietic cell populations in steady and activated states at a depth of >10,000 proteins in total. Protein copy numbers revealed a specialization of immune cells for ligand and receptor expression, thereby connecting distinct immune functions. By integrating total and secreted proteomes, we discovered fundamental intercellular communication structures and previously unknown connections between cell types. Our publicly accessible (http://www.immprot.org/) proteomic resource provides a framework for the orchestration of cellular interplay and a reference for altered communication associated with pathology.


Asunto(s)
Células Sanguíneas/fisiología , Inmunidad Celular , Mapas de Interacción de Proteínas , Proteoma , Proteómica , Animales , Secreciones Corporales , Comunicación Celular , Simulación por Computador , Humanos , Espectrometría de Masas , Apoyo Social
4.
Nature ; 569(7758): 663-671, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-31142858

RESUMEN

Type 2 diabetes mellitus (T2D) is a growing health problem, but little is known about its early disease stages, its effects on biological processes or the transition to clinical T2D. To understand the earliest stages of T2D better, we obtained samples from 106 healthy individuals and individuals with prediabetes over approximately four years and performed deep profiling of transcriptomes, metabolomes, cytokines, and proteomes, as well as changes in the microbiome. This rich longitudinal data set revealed many insights: first, healthy profiles are distinct among individuals while displaying diverse patterns of intra- and/or inter-personal variability. Second, extensive host and microbial changes occur during respiratory viral infections and immunization, and immunization triggers potentially protective responses that are distinct from responses to respiratory viral infections. Moreover, during respiratory viral infections, insulin-resistant participants respond differently than insulin-sensitive participants. Third, global co-association analyses among the thousands of profiled molecules reveal specific host-microbe interactions that differ between insulin-resistant and insulin-sensitive individuals. Last, we identified early personal molecular signatures in one individual that preceded the onset of T2D, including the inflammation markers interleukin-1 receptor agonist (IL-1RA) and high-sensitivity C-reactive protein (CRP) paired with xenobiotic-induced immune signalling. Our study reveals insights into pathways and responses that differ between glucose-dysregulated and healthy individuals during health and disease and provides an open-access data resource to enable further research into healthy, prediabetic and T2D states.


Asunto(s)
Biomarcadores/metabolismo , Biología Computacional , Diabetes Mellitus Tipo 2/microbiología , Microbioma Gastrointestinal , Interacciones Microbiota-Huesped/genética , Estado Prediabético/microbiología , Proteoma/metabolismo , Transcriptoma , Adulto , Anciano , Antibacterianos/administración & dosificación , Biomarcadores/análisis , Estudios de Cohortes , Conjuntos de Datos como Asunto , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/metabolismo , Femenino , Glucosa/metabolismo , Voluntarios Sanos , Humanos , Inflamación/metabolismo , Vacunas contra la Influenza/inmunología , Insulina/metabolismo , Resistencia a la Insulina , Estudios Longitudinales , Masculino , Microbiota/fisiología , Persona de Mediana Edad , Estado Prediabético/genética , Estado Prediabético/metabolismo , Infecciones del Sistema Respiratorio/genética , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/virología , Estrés Fisiológico , Vacunación/estadística & datos numéricos
5.
Proc Natl Acad Sci U S A ; 119(11): e2106053119, 2022 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-35275789

RESUMEN

SignificanceDeep profiling of the plasma proteome at scale has been a challenge for traditional approaches. We achieve superior performance across the dimensions of precision, depth, and throughput using a panel of surface-functionalized superparamagnetic nanoparticles in comparison to conventional workflows for deep proteomics interrogation. Our automated workflow leverages competitive nanoparticle-protein binding equilibria that quantitatively compress the large dynamic range of proteomes to an accessible scale. Using machine learning, we dissect the contribution of individual physicochemical properties of nanoparticles to the composition of protein coronas. Our results suggest that nanoparticle functionalization can be tailored to protein sets. This work demonstrates the feasibility of deep, precise, unbiased plasma proteomics at a scale compatible with large-scale genomics enabling multiomic studies.


Asunto(s)
Proteínas Sanguíneas , Aprendizaje Profundo , Nanopartículas , Proteómica , Proteínas Sanguíneas/química , Nanopartículas/química , Corona de Proteínas/química , Proteoma , Proteómica/métodos
6.
Nucleic Acids Res ; 49(21): 12284-12305, 2021 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-34850154

RESUMEN

Neurons critically rely on the functions of RNA-binding proteins to maintain their polarity and resistance to neurotoxic stress. HnRNP R has a diverse range of post-transcriptional regulatory functions and is important for neuronal development by regulating axon growth. Hnrnpr pre-mRNA undergoes alternative splicing giving rise to a full-length protein and a shorter isoform lacking its N-terminal acidic domain. To investigate functions selectively associated with the full-length hnRNP R isoform, we generated a Hnrnpr knockout mouse (Hnrnprtm1a/tm1a) in which expression of full-length hnRNP R was abolished while production of the truncated hnRNP R isoform was retained. Motoneurons cultured from Hnrnprtm1a/tm1a mice did not show any axonal growth defects but exhibited enhanced accumulation of double-strand breaks and an impaired DNA damage response upon exposure to genotoxic agents. Proteomic analysis of the hnRNP R interactome revealed the multifunctional protein Yb1 as a top interactor. Yb1-depleted motoneurons were defective in DNA damage repair. We show that Yb1 is recruited to chromatin upon DNA damage where it interacts with γ-H2AX, a mechanism that is dependent on full-length hnRNP R. Our findings thus suggest a novel role of hnRNP R in maintaining genomic integrity and highlight the function of its N-terminal acidic domain in this context.


Asunto(s)
Cromatina/genética , Daño del ADN , Reparación del ADN/genética , Ribonucleoproteínas Nucleares Heterogéneas/genética , Neuronas Motoras/metabolismo , Proteína 1 de Unión a la Caja Y/genética , Animales , Axones/metabolismo , Línea Celular , Células Cultivadas , Cromatina/metabolismo , Células HEK293 , Ribonucleoproteínas Nucleares Heterogéneas/metabolismo , Humanos , Immunoblotting , Ratones Endogámicos C57BL , Ratones Noqueados , Neuronas Motoras/citología , Unión Proteica , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteína 1 de Unión a la Caja Y/metabolismo
7.
Mol Syst Biol ; 17(7): e10125, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34318608

RESUMEN

Cells signal through rearrangements of protein communities governed by covalent modifications and reversible interactions of distinct sets of proteins. A method that identifies those post-transcriptional modifications regulating signaling complex composition and functional phenotypes in one experimental setup would facilitate an efficient identification of novel molecular signaling checkpoints. Here, we devised modifications, interactions and phenotypes by affinity purification mass spectrometry (MIP-APMS), comprising the streamlined cloning and transduction of tagged proteins into functionalized reporter cells as well as affinity chromatography, followed by MS-based quantification. We report the time-resolved interplay of more than 50 previously undescribed modification and hundreds of protein-protein interactions of 19 immune protein complexes in monocytes. Validation of interdependencies between covalent, reversible, and functional protein complex regulations by knockout or site-specific mutation revealed ISGylation and phosphorylation of TRAF2 as well as ARHGEF18 interaction in Toll-like receptor 2 signaling. Moreover, we identify distinct mechanisms of action for small molecule inhibitors of p38 (MAPK14). Our method provides a fast and cost-effective pipeline for the molecular interrogation of protein communities in diverse biological systems and primary cells.


Asunto(s)
Procesamiento Proteico-Postraduccional , Proteómica , Complejo Antígeno-Anticuerpo , Espectrometría de Masas , Fenotipo
8.
Acc Chem Res ; 54(2): 291-301, 2021 01 19.
Artículo en Inglés | MEDLINE | ID: mdl-33180454

RESUMEN

Understanding the interactions between nanomaterials and biological systems plays a pivotal role in enhancing the efficacy of nanomedicine and advancing the disease diagnosis. The nanoparticle-protein corona, an active biomolecular layer, is formed around nanoparticles (NPs) upon mixing with biological fluid. The surface layer which consists of rapidly exchanged biomolecules is called the "soft" corona. The inner layer which is more stable and tightly packed is called the "hard" corona. It has been suggested that the NP-protein corona has a decisive effect on the in vivo fate of nanomedicine upon intravenously administration into the mouse. Furthermore, the features of the NP-protein corona make it a powerful platform to enrich low-abundance proteins from serum/plasma for downstream mass-spectrometry (MS)-based proteomics for biomarker discovery and disease diagnosis.Herein, we summarize our recent work on the development of nanomedicine and disease detection from the level of nano-bio interactions between nanoparticles and biological systems. Nanomedicine has made substantial progress over the past two decades. However, the significant enhancement of overall patient survival by nanomedicine remains a challenge due to the lack of a deep understanding of nano-bio interactions in the clinical setting. The pharmacokinetic effect of the protein corona on PEGylated NPs during blood circulation indicated that the adsorbed apolipoproteins could prolong the circulation time of NPs. This mechanistic understanding of the protein corona (active biomolecule) formed around polymeric NPs offered insights into enhancing the efficacy of nanomedicine from the biological interactions point of view. Moreover, we discuss the basic rationale for developing bioresponsive cancer nanomedicine by exploiting the pathophysiological environment around the tumor, typically the pH, reactive oxygen species (ROS), and redox-responsive supramolecular motifs based on synthetic amphiphilic polymers. The protein corona in vivo determines the biological fate of NPs, whereas it opens a new avenue to enrich low abundant proteins in a biospecimen ex vivo to render them "visible" for downstream analytical workflows, such as MS-based proteomics. Blood serum/plasma, due to easy accessibility and great potential to uncover and monitor physiological and pathological changes in health and disease, has remained a major source of detecting protein biomarker candidates. Inspired by the features of the NP-protein corona, a Proteograph platform, which integrates multi-NP-protein coronas with MS for large-scale efficient and deep proteome profiling has been developed. Finally, we conclude this Account with a better understanding of nano-bio interactions to accelerate the nanomedicine translation and how MS-based proteomics can boost our understanding of the corona composition and facilitate the identification of disease biomarkers.


Asunto(s)
Nanopartículas/química , Corona de Proteínas/química , Animales , Portadores de Fármacos/química , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Imagen por Resonancia Magnética , Ratones , Microscopía Confocal , Nanomedicina , Nanopartículas/metabolismo , Nanopartículas/uso terapéutico , Neoplasias/diagnóstico , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Oxidación-Reducción , Polietilenglicoles/química , ARN Interferente Pequeño/química , ARN Interferente Pequeño/metabolismo , Especies Reactivas de Oxígeno/metabolismo
9.
Anal Chem ; 93(49): 16369-16378, 2021 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-34859676

RESUMEN

Modern biomarker and translational research as well as personalized health care studies rely heavily on powerful omics' technologies, including metabolomics and lipidomics. However, to translate metabolomics and lipidomics discoveries into a high-throughput clinical setting, standardization is of utmost importance. Here, we compared and benchmarked a quantitative lipidomics platform. The employed Lipidyzer platform is based on lipid class separation by means of differential mobility spectrometry with subsequent multiple reaction monitoring. Quantitation is achieved by the use of 54 deuterated internal standards and an automated informatics approach. We investigated the platform performance across nine laboratories using NIST SRM 1950-Metabolites in Frozen Human Plasma, and three NIST Candidate Reference Materials 8231-Frozen Human Plasma Suite for Metabolomics (high triglyceride, diabetic, and African-American plasma). In addition, we comparatively analyzed 59 plasma samples from individuals with familial hypercholesterolemia from a clinical cohort study. We provide evidence that the more practical methyl-tert-butyl ether extraction outperforms the classic Bligh and Dyer approach and compare our results with two previously published ring trials. In summary, we present standardized lipidomics protocols, allowing for the highly reproducible analysis of several hundred human plasma lipids, and present detailed molecular information for potentially disease relevant and ethnicity-related materials.


Asunto(s)
Laboratorios , Lipidómica , Estudios de Cohortes , Humanos , Estándares de Referencia , Análisis Espectral
10.
Nature ; 526(7573): 443-7, 2015 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-26322584

RESUMEN

Alzheimer disease (AD) is characterized by the accumulation of amyloid plaques, which are predominantly composed of amyloid-ß peptide. Two principal physiological pathways either prevent or promote amyloid-ß generation from its precursor, ß-amyloid precursor protein (APP), in a competitive manner. Although APP processing has been studied in great detail, unknown proteolytic events seem to hinder stoichiometric analyses of APP metabolism in vivo. Here we describe a new physiological APP processing pathway, which generates proteolytic fragments capable of inhibiting neuronal activity within the hippocampus. We identify higher molecular mass carboxy-terminal fragments (CTFs) of APP, termed CTF-η, in addition to the long-known CTF-α and CTF-ß fragments generated by the α- and ß-secretases ADAM10 (a disintegrin and metalloproteinase 10) and BACE1 (ß-site APP cleaving enzyme 1), respectively. CTF-η generation is mediated in part by membrane-bound matrix metalloproteinases such as MT5-MMP, referred to as η-secretase activity. η-Secretase cleavage occurs primarily at amino acids 504-505 of APP695, releasing a truncated ectodomain. After shedding of this ectodomain, CTF-η is further processed by ADAM10 and BACE1 to release long and short Aη peptides (termed Aη-α and Aη-ß). CTFs produced by η-secretase are enriched in dystrophic neurites in an AD mouse model and in human AD brains. Genetic and pharmacological inhibition of BACE1 activity results in robust accumulation of CTF-η and Aη-α. In mice treated with a potent BACE1 inhibitor, hippocampal long-term potentiation was reduced. Notably, when recombinant or synthetic Aη-α was applied on hippocampal slices ex vivo, long-term potentiation was lowered. Furthermore, in vivo single-cell two-photon calcium imaging showed that hippocampal neuronal activity was attenuated by Aη-α. These findings not only demonstrate a major functionally relevant APP processing pathway, but may also indicate potential translational relevance for therapeutic strategies targeting APP processing.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Hipocampo/citología , Metaloproteinasas de la Matriz Asociadas a la Membrana/metabolismo , Neuronas/fisiología , Proteolisis , Proteínas ADAM/metabolismo , Proteína ADAM10 , Enfermedad de Alzheimer/enzimología , Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/antagonistas & inhibidores , Secretasas de la Proteína Precursora del Amiloide/líquido cefalorraquídeo , Secretasas de la Proteína Precursora del Amiloide/deficiencia , Secretasas de la Proteína Precursora del Amiloide/genética , Precursor de Proteína beta-Amiloide/líquido cefalorraquídeo , Precursor de Proteína beta-Amiloide/química , Precursor de Proteína beta-Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/antagonistas & inhibidores , Ácido Aspártico Endopeptidasas/deficiencia , Ácido Aspártico Endopeptidasas/genética , Ácido Aspártico Endopeptidasas/metabolismo , Señalización del Calcio , Modelos Animales de Enfermedad , Femenino , Hipocampo/enzimología , Hipocampo/fisiología , Humanos , Técnicas In Vitro , Potenciación a Largo Plazo , Masculino , Metaloproteinasas de la Matriz Asociadas a la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones , Peso Molecular , Neuritas/enzimología , Neuritas/metabolismo , Neuronas/enzimología , Fragmentos de Péptidos/química , Fragmentos de Péptidos/metabolismo , Placa Amiloide , Procesamiento Proteico-Postraduccional , Análisis de la Célula Individual
11.
Proc Natl Acad Sci U S A ; 115(15): E3446-E3453, 2018 04 10.
Artículo en Inglés | MEDLINE | ID: mdl-29581260

RESUMEN

Huntington's disease is caused by the expansion of a polyglutamine (polyQ) tract in the N-terminal exon of huntingtin (HttEx1), but the cellular mechanisms leading to neurodegeneration remain poorly understood. Here we present in situ structural studies by cryo-electron tomography of an established yeast model system of polyQ toxicity. We find that expression of polyQ-expanded HttEx1 results in the formation of unstructured inclusion bodies and in some cases fibrillar aggregates. This contrasts with recent findings in mammalian cells, where polyQ inclusions were exclusively fibrillar. In yeast, polyQ toxicity correlates with alterations in mitochondrial and lipid droplet morphology, which do not arise from physical interactions with inclusions or fibrils. Quantitative proteomic analysis shows that polyQ aggregates sequester numerous cellular proteins and cause a major change in proteome composition, most significantly in proteins related to energy metabolism. Thus, our data point to a multifaceted toxic gain-of-function of polyQ aggregates, driven by sequestration of endogenous proteins and mitochondrial and lipid droplet dysfunction.


Asunto(s)
Péptidos/metabolismo , Saccharomyces cerevisiae/metabolismo , Humanos , Enfermedad de Huntington/genética , Enfermedad de Huntington/metabolismo , Cuerpos de Inclusión/química , Cuerpos de Inclusión/genética , Cuerpos de Inclusión/metabolismo , Gotas Lipídicas/química , Gotas Lipídicas/metabolismo , Mitocondrias/química , Mitocondrias/metabolismo , Péptidos/química , Péptidos/toxicidad , Proteómica , Saccharomyces cerevisiae/química , Saccharomyces cerevisiae/efectos de los fármacos , Proteínas de Saccharomyces cerevisiae/química , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo
12.
EMBO J ; 35(21): 2350-2370, 2016 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-27621269

RESUMEN

Nuclear clearance of TDP-43 into cytoplasmic aggregates is a key driver of neurodegeneration in amyotrophic lateral sclerosis (ALS) and frontotemporal lobar degeneration (FTLD), but the mechanisms are unclear. Here, we show that TDP-43 knockdown specifically reduces the number and motility of RAB11-positive recycling endosomes in dendrites, while TDP-43 overexpression has the opposite effect. This is associated with delayed transferrin recycling in TDP-43-knockdown neurons and decreased ß2-transferrin levels in patient CSF Whole proteome quantification identified the upregulation of the ESCRT component VPS4B upon TDP-43 knockdown in neurons. Luciferase reporter assays and chromatin immunoprecipitation suggest that TDP-43 represses VPS4B transcription. Preventing VPS4B upregulation or expression of its functional antagonist ALIX restores trafficking of recycling endosomes. Proteomic analysis revealed the broad reduction in surface expression of key receptors upon TDP-43 knockdown, including ErbB4, the neuregulin 1 receptor. TDP-43 knockdown delays the surface delivery of ErbB4. ErbB4 overexpression, but not neuregulin 1 stimulation, prevents dendrite loss upon TDP-43 knockdown. Thus, impaired recycling of ErbB4 and other receptors to the cell surface may contribute to TDP-43-induced neurodegeneration by blocking trophic signaling.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Complejos de Clasificación Endosomal Requeridos para el Transporte/genética , Endosomas/metabolismo , Neuronas/metabolismo , Receptor ErbB-4/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Adenosina Trifosfatasas/genética , Adenosina Trifosfatasas/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/metabolismo , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Complejos de Clasificación Endosomal Requeridos para el Transporte/metabolismo , Degeneración Lobar Frontotemporal/genética , Degeneración Lobar Frontotemporal/metabolismo , Técnicas de Silenciamiento del Gen , Hipocampo/citología , Humanos , Transporte de Proteínas , Ratas , Receptor ErbB-4/genética , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/metabolismo , Transducción de Señal
13.
Nature ; 554(7693): 423, 2018 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-32094961
14.
Nature ; 554(7693): 423, 2018 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-29469129
15.
Diabetologia ; 60(10): 2066-2075, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28669086

RESUMEN

AIMS/HYPOTHESIS: In humans, glucagon-like peptide-1 (GLP-1) is rapidly degraded by dipeptidyl peptidase-4 to a relatively stable metabolite, GLP-1(9-36)NH2, which allows measurement of GLP-1 secretion. However, little is known about the kinetics of the GLP-1 metabolite in mice. We hypothesised that the GLP-1 metabolite is rapidly degraded in this species by neutral endopeptidase(s) (NEP[s]). METHODS: We administered glucose, mixed meal or water orally to 256 mice, and took blood samples before and 2, 6, 10, 20, 30, 60 or 90 min after stimulation. To study the metabolism of the GLP-1 metabolite, i.v. GLP-1(9-36)NH2 (800 fmol) or saline (154 mmol/l NaCl) was administered to 160 mice, some of which had a prior injection of a selective NEP 24.11 ± inhibitor (candoxatril, 5 mg/kg) or saline. Blood was collected before and 1, 2, 4 and 12 min after GLP-1/saline injection. Plasma GLP-1 levels were analysed using a customised single-site C-terminal ELISA, two different two-site ELISAs and MS. RESULTS: GLP-1 secretion profiles after oral glucose administration differed markedly when assayed by C-terminal ELISA compared with sandwich ELISAs, with the former showing a far higher peak value and AUC. In mice injected with GLP-1(9-36)NH2, immunoreactive GLP-1 plasma levels peaked at approximately 75 pmol/l at 1 min when measured with sandwich ELISAs, returning to baseline (~20 pmol/l) after 12 min, but remained elevated using the C-terminal ELISA (~90 pmol/l at 12 min). NEP 24.11 inhibition by candoxatril significantly attenuated GLP-1(9-36)NH2 degradation in vivo and in vitro. MS identified GLP-1 fragments consistent with NEP 24.11 degradation. CONCLUSIONS/INTERPRETATION: In mice, the GLP-1 metabolite is eliminated within a few minutes owing to endoproteolytic cleavage by NEP 24.11. Therefore, accurate measurement of GLP-1 secretion in mice requires assays for NEP 24.11 metabolites. Conventional sandwich ELISAs are inadequate because of endoproteolytic cleavage of the dipeptidyl peptidase-4-generated metabolite.


Asunto(s)
Péptido 1 Similar al Glucagón/sangre , Periodo Posprandial/fisiología , Animales , Femenino , Glucosa/farmacología , Indanos/farmacología , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Masculino , Ratones , Neprilisina/antagonistas & inhibidores , Periodo Posprandial/efectos de los fármacos , Propionatos/farmacología , Inhibidores de Proteasas/farmacología
16.
Mol Cell Proteomics ; 14(7): 2014-29, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25991688

RESUMEN

Hybrid quadrupole time-of-flight (QTOF) mass spectrometry is one of the two major principles used in proteomics. Although based on simple fundamentals, it has over the last decades greatly evolved in terms of achievable resolution, mass accuracy, and dynamic range. The Bruker impact platform of QTOF instruments takes advantage of these developments and here we develop and evaluate the impact II for shotgun proteomics applications. Adaption of our heated liquid chromatography system achieved very narrow peptide elution peaks. The impact II is equipped with a new collision cell with both axial and radial ion ejection, more than doubling ion extraction at high tandem MS frequencies. The new reflectron and detector improve resolving power compared with the previous model up to 80%, i.e. to 40,000 at m/z 1222. We analyzed the ion current from the inlet capillary and found very high transmission (>80%) up to the collision cell. Simulation and measurement indicated 60% transfer into the flight tube. We adapted MaxQuant for QTOF data, improving absolute average mass deviations to better than 1.45 ppm. More than 4800 proteins can be identified in a single run of HeLa digest in a 90 min gradient. The workflow achieved high technical reproducibility (R2 > 0.99) and accurate fold change determination in spike-in experiments in complex mixtures. Using label-free quantification we rapidly quantified haploid against diploid yeast and characterized overall proteome differences in mouse cell lines originating from different tissues. Finally, after high pH reversed-phase fractionation we identified 9515 proteins in a triplicate measurement of HeLa peptide mixture and 11,257 proteins in single measurements of cerebellum-the highest proteome coverage reported with a QTOF instrument so far.


Asunto(s)
Proteómica/instrumentación , Proteómica/métodos , Animales , Línea Celular , Cromatografía Liquida , Diploidia , Haploidia , Células HeLa , Humanos , Concentración de Iones de Hidrógeno , Iones , Espectrometría de Masas , Ratones , Peso Molecular , Péptidos/metabolismo , Proteoma/metabolismo , Reproducibilidad de los Resultados , Saccharomyces cerevisiae/metabolismo , Factores de Tiempo
17.
Proc Natl Acad Sci U S A ; 111(51): 18219-24, 2014 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-25489109

RESUMEN

Expansion of a poly-glutamine (polyQ) repeat in a group of functionally unrelated proteins is the cause of several inherited neurodegenerative disorders, including Huntington's disease. The polyQ length-dependent aggregation and toxicity of these disease proteins can be reproduced in Saccharomyces cerevisiae. This system allowed us to screen for genes that when overexpressed reduce the toxic effects of an N-terminal fragment of mutant huntingtin with 103 Q. Surprisingly, among the identified suppressors were three proteins with Q-rich, prion-like domains (PrDs): glycine threonine serine repeat protein (Gts1p), nuclear polyadenylated RNA-binding protein 3, and minichromosome maintenance protein 1. Overexpression of the PrD of Gts1p, containing an imperfect 28 residue glutamine-alanine repeat, was sufficient for suppression of toxicity. Association with this discontinuous polyQ domain did not prevent 103Q aggregation, but altered the physical properties of the aggregates, most likely early in the assembly pathway, as reflected in their increased SDS solubility. Molecular simulations suggested that Gts1p arrests the aggregation of polyQ molecules at the level of nonfibrillar species, acting as a cap that destabilizes intermediates on path to form large fibrils. Quantitative proteomic analysis of polyQ interactors showed that expression of Gts1p reduced the interaction between polyQ and other prion-like proteins, and enhanced the association of molecular chaperones with the aggregates. These findings demonstrate that short, Q-rich peptides are able to shield the interactive surfaces of toxic forms of polyQ proteins and direct them into nontoxic aggregates.


Asunto(s)
Péptidos/metabolismo , Priones/metabolismo , Unión Proteica , Saccharomyces cerevisiae/genética
18.
Mol Cell Proteomics ; 13(12): 3410-20, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25193168

RESUMEN

The fatal neurodegenerative disorders amyotrophic lateral sclerosis and spinal muscular atrophy are, respectively, the most common motoneuron disease and genetic cause of infant death. Various in vitro model systems have been established to investigate motoneuron disease mechanisms, in particular immortalized cell lines and primary neurons. Using quantitative mass-spectrometry-based proteomics, we compared the proteomes of primary motoneurons to motoneuron-like cell lines NSC-34 and N2a, as well as to non-neuronal control cells, at a depth of 10,000 proteins. We used this resource to evaluate the suitability of murine in vitro model systems for cell biological and biochemical analysis of motoneuron disease mechanisms. Individual protein and pathway analysis indicated substantial differences between motoneuron-like cell lines and primary motoneurons, especially for proteins involved in differentiation, cytoskeleton, and receptor signaling, whereas common metabolic pathways were more similar. The proteins associated with amyotrophic lateral sclerosis also showed distinct differences between cell lines and primary motoneurons, providing a molecular basis for understanding fundamental alterations between cell lines and neurons with respect to neuronal pathways with relevance for disease mechanisms. Our study provides a proteomics resource for motoneuron research and presents a paradigm of how mass-spectrometry-based proteomics can be used to evaluate disease model systems.


Asunto(s)
Esclerosis Amiotrófica Lateral/metabolismo , Neuronas Motoras/metabolismo , Proteoma/metabolismo , Esclerosis Amiotrófica Lateral/genética , Esclerosis Amiotrófica Lateral/patología , Animales , Diferenciación Celular , Línea Celular , Citoesqueleto/genética , Citoesqueleto/metabolismo , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Ratones , Anotación de Secuencia Molecular , Neuronas Motoras/química , Neuronas Motoras/patología , Especificidad de Órganos , Cultivo Primario de Células , Proteoma/genética , Transducción de Señal
19.
Mol Cell Proteomics ; 13(12): 3698-708, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25360005

RESUMEN

The quadrupole Orbitrap mass spectrometer (Q Exactive) made a powerful proteomics instrument available in a benchtop format. It significantly boosted the number of proteins analyzable per hour and has now evolved into a proteomics analysis workhorse for many laboratories. Here we describe the Q Exactive Plus and Q Exactive HF mass spectrometers, which feature several innovations in comparison to the original Q Exactive instrument. A low-resolution pre-filter has been implemented within the injection flatapole, preventing unwanted ions from entering deep into the system, and thereby increasing its robustness. A new segmented quadrupole, with higher fidelity of isolation efficiency over a wide range of isolation windows, provides an almost 2-fold improvement of transmission at narrow isolation widths. Additionally, the Q Exactive HF has a compact Orbitrap analyzer, leading to higher field strength and almost doubling the resolution at the same transient times. With its very fast isolation and fragmentation capabilities, the instrument achieves overall cycle times of 1 s for a top 15 to 20 higher energy collisional dissociation method. We demonstrate the identification of 5000 proteins in standard 90-min gradients of tryptic digests of mammalian cell lysate, an increase of over 40% for detected peptides and over 20% for detected proteins. Additionally, we tested the instrument on peptide phosphorylation enriched samples, for which an improvement of up to 60% class I sites was observed.


Asunto(s)
Espectrometría de Masas/instrumentación , Fosfoproteínas/aislamiento & purificación , Secuencia de Aminoácidos , Filtración , Análisis de Inyección de Flujo , Células HeLa , Humanos , Iones , Espectrometría de Masas/métodos , Datos de Secuencia Molecular , Fosforilación , Sensibilidad y Especificidad , Factores de Tiempo , Tripsina/química
20.
Biophys J ; 106(6): 1318-26, 2014 Mar 18.
Artículo en Inglés | MEDLINE | ID: mdl-24655507

RESUMEN

Many transmembrane helices contain serine and/or threonine residues whose side chains form intrahelical H-bonds with upstream carbonyl oxygens. Here, we investigated the impact of threonine side-chain/main-chain backbonding on the backbone dynamics of the amyloid precursor protein transmembrane helix. This helix consists of a N-terminal dimerization region and a C-terminal cleavage region, which is processed by γ-secretase to a series of products. Threonine mutations within this transmembrane helix are known to alter the cleavage pattern, which can lead to early-onset Alzheimer's disease. Circular dichroism spectroscopy and amide exchange experiments of synthetic transmembrane domain peptides reveal that mutating threonine enhances the flexibility of this helix. Molecular dynamics simulations show that the mutations reduce intrahelical amide H-bonding and H-bond lifetimes. In addition, the removal of side-chain/main-chain backbonding distorts the helix, which alters bending and rotation at a diglycine hinge connecting the dimerization and cleavage regions. We propose that the backbone dynamics of the substrate profoundly affects the way by which the substrate is presented to the catalytic site within the enzyme. Changing this conformational flexibility may thus change the pattern of proteolytic processing.


Asunto(s)
Precursor de Proteína beta-Amiloide/química , Simulación de Dinámica Molecular , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Precursor de Proteína beta-Amiloide/genética , Animales , Humanos , Enlace de Hidrógeno , Datos de Secuencia Molecular , Mutación , Estructura Terciaria de Proteína
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA