Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Anal Chem ; 95(7): 3629-3637, 2023 02 21.
Article in English | MEDLINE | ID: mdl-36745752

ABSTRACT

Multidimensional liquid chromatography (mD-LC) is becoming a powerful tool for complete characterization of individual peaks and protein variants through separation methods such as nondenaturing ion exchange (IEC) or size-exclusion chromatography coupled to reversed-phase (RP) chromatography. The flexibility of commercially available and customized mD-LC systems is still limited in terms of enzymatic peak processing between chromatographic dimensions. In this regard, only a few column-immobilized proteases are available for detailed peak characterization by mD-LC coupled to mass spectrometry (mD-LC-MS). Here, we present a purpose-built and automated multiple heart-cutting mD-LC design with a novel analytical workflow involving in-loop enzymatic heart-cut digestion between the first-dimensional column and transfer to the second dimension before MS or MS/MS analyses. The setup facilitates the spike-in of any enzyme to multiple heart-cuts for multilevel analysis, for example, for peptide mapping, fragment generation, or deglycosylation, to reduce heterogeneity and provide maximum flexibility in terms of incubation time for optimal peak characterization. We demonstrate the application of IEC coupled to RP-LC-MS and automated in-loop deglycosylation and on-column reduction of an IgG antibody combined with upper hinge region cleavage for Fab generation. We further employ mD-LC-MS and mD-LC-MS/MS to assess post-translational modifications of a bispecific antibody and to support molecule selection by evaluating the best downstream purification strategy. The novel design and automated workflow of the mD-LC system described here offers enhanced flexibility for in-solution processing and real-time monitoring of multiple heart-cuts enabling streamlined characterization of unknown biotherapeutic charge and size variants.


Subject(s)
Chromatography, Reverse-Phase , Tandem Mass Spectrometry , Chromatography, Liquid/methods , Workflow , Chromatography, Reverse-Phase/methods , Chromatography, Gel
2.
Sci Rep ; 9(1): 6634, 2019 04 29.
Article in English | MEDLINE | ID: mdl-31036840

ABSTRACT

Huntington's disease (HD) is a devastating hereditary movement disorder, characterized by degeneration of neurons in the striatum and cortex. Studies in human patients and mouse HD models suggest that disturbances of neuronal function in the neocortex play an important role in disease onset and progression. However, the precise nature and time course of cortical alterations in HD have remained elusive. Here, we use chronic in vivo two-photon calcium imaging to longitudinally monitor the activity of identified single neurons in layer 2/3 of the primary motor cortex in awake, behaving R6/2 transgenic HD mice and wildtype littermates. R6/2 mice show age-dependent changes in cortical network function, with an increase in activity that affects a large fraction of cells and occurs rather abruptly within one week, preceeding the onset of motor defects. Furthermore, quantitative proteomics demonstrate a pronounced downregulation of synaptic proteins in the cortex, and histological analyses in R6/2 mice and human HD autopsy cases reveal a reduction in perisomatic inhibitory synaptic contacts on layer 2/3 pyramidal cells. Taken together, our study provides a time-resolved description of cortical network dysfunction in behaving HD mice and points to disturbed excitation/inhibition balance as an important pathomechanism in HD.


Subject(s)
Huntington Disease/pathology , Motor Disorders/pathology , Motor Disorders/physiopathology , Animals , Disease Models, Animal , Female , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/metabolism , Huntington Disease/physiopathology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Disorders/metabolism , Promoter Regions, Genetic/genetics
3.
Neuron ; 96(3): 558-571, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29096073

ABSTRACT

The enormous complexity of the central nervous system has impeded its systemic exploration for decades but powerful "omic" technologies are now pushing forward the frontiers of neuroscience research at an increasing pace. This Primer reviews the most recent progress in mass spectrometry (MS)-based proteomics, focusing on the analysis of whole proteomes, protein-based interactions, and post-translational modifications. We also discuss how advanced workflows help to unravel spatial, regulatory, and temporal aspects of neuronal systems. These tools and approaches have already led to detailed and quantitative proteomic maps of the brain and its signaling architecture, generating new insights into health and disease. We predict that these new approaches will also accelerate biomarker discovery and contribute to novel therapeutics for neurodegenerative and other brain-related diseases.


Subject(s)
Brain/physiology , Neurons/physiology , Neurosciences/methods , Proteomics/methods , Animals , Brain/cytology , Chromatography, Liquid/methods , Humans , Mass Spectrometry/methods , Neurosciences/trends , Proteomics/trends
4.
Cell Rep ; 21(8): 2291-2303, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-29166617

ABSTRACT

Aggregation of polyglutamine-expanded huntingtin exon 1 (HttEx1) in Huntington's disease (HD) proceeds from soluble oligomers to late-stage inclusions. The nature of the aggregates and how they lead to neuronal dysfunction is not well understood. We employed mass spectrometry (MS)-based quantitative proteomics to dissect spatiotemporal mechanisms of neurodegeneration using the R6/2 mouse model of HD. Extensive remodeling of the soluble brain proteome correlated with insoluble aggregate formation during disease progression. In-depth and quantitative characterization of the aggregates uncovered an unprecedented complexity of several hundred proteins. Sequestration to aggregates depended on protein expression levels and sequence features such as low-complexity regions or coiled-coil domains. In a cell-based HD model, overexpression of a subset of the sequestered proteins in most cases rescued viability and reduced aggregate size. Our spatiotemporally resolved proteome resource of HD progression indicates that widespread loss of cellular protein function contributes to aggregate-mediated toxicity.


Subject(s)
Huntington Disease/genetics , Inclusion Bodies/metabolism , Neurons/metabolism , Nuclear Proteins/metabolism , Animals , Disease Models, Animal , Gene Expression Profiling/methods , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Peptides/genetics , Proteomics/methods
5.
Cell Metab ; 26(3): 475-492.e7, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-28877454

ABSTRACT

Obesity and type 2 diabetes are associated with metabolic defects and adipose tissue inflammation. Foxp3+ regulatory T cells (Tregs) control tissue homeostasis by counteracting local inflammation. However, if and how T cells interlink environmental influences with adipocyte function remains unknown. Here, we report that enhancing sympathetic tone by cold exposure, beta3-adrenergic receptor (ADRB3) stimulation or a short-term high-calorie diet enhances Treg induction in vitro and in vivo. CD4+ T cell proteomes revealed higher expression of Foxp3 regulatory networks in response to cold or ADRB3 stimulation in vivo reflecting Treg induction. Specifically, Ragulator-interacting protein C17orf59, which limits mTORC1 activity, was upregulated in CD4+ T cells by either ADRB3 stimulation or cold exposure, suggesting contribution to Treg induction. By loss- and gain-of-function studies, including Treg depletion and transfers in vivo, we demonstrated that a T cell-specific Stat6/Pten axis links cold exposure or ADRB3 stimulation with Foxp3+ Treg induction and adipose tissue function. Our findings offer a new mechanistic model in which tissue-specific Tregs maintain adipose tissue function.


Subject(s)
Adipose Tissue, Brown/metabolism , Adipose Tissue, White/metabolism , PTEN Phosphohydrolase/metabolism , STAT6 Transcription Factor/metabolism , Animals , Cold Temperature , Female , Forkhead Transcription Factors/metabolism , Mice, Inbred BALB C , Proteome/metabolism , Receptors, Adrenergic, beta/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , Uncoupling Protein 1/metabolism
6.
Methods Mol Biol ; 1653: 65-81, 2017.
Article in English | MEDLINE | ID: mdl-28822126

ABSTRACT

Photorespiratory enzymes in different cellular compartments have been reported to be posttranslational modified by phosphorylation, disulfide formation, S-nitrosylation, glutathionylation, and lysine acetylation. However, not much is known yet about the function of these modifications to regulate the activities, localizations, or interactions of the proteins in this metabolic pathway. Hence, it will be of great importance to study these modifications and their temporal and conditional occurrence in more detail. Here, we focus on the analysis of lysine acetylation as a relatively newly discovered modification on plant metabolic enzymes. The acetylation of lysine residues within proteins is a highly conserved and reversible posttranslational modification which occurs in all living organisms. First discovered on histones and implied in the regulation of gene expression, lysine acetylation also occurs on a diverse set of cellular proteins in different subcellular compartments and is particularly abundant on metabolic enzymes. Upon lysine acetylation, the function of proteins can be modulated due to the loss of the positive charge of the lysine residue. Lysine acetylation was also discovered on proteins involved in photosynthesis and novel tools are needed to study the regulation of this modification in dependence on the environmental conditions, tissues, or plant genotype. This chapter describes a method for the identification and relative quantification of lysine-acetylated proteins in plant tissues using a dimethyl labeling technique combined with an anti-acetyl lysine antibody enrichment strategy. Here, we describe the protein purification, labeling of trypsinated peptides, as well as immuno-enrichment of lysine-acetylated peptides followed by liquid chromatography tandem mass spectrometry (LC-MS/MS) data acquisition and analysis.


Subject(s)
Arabidopsis/metabolism , Lysine/metabolism , Peptides/isolation & purification , Plant Proteins/isolation & purification , Protein Processing, Post-Translational , Staining and Labeling/methods , Acetylation , Antibodies/chemistry , Arabidopsis/chemistry , Arabidopsis/genetics , Chromatography, Liquid/methods , Oryza/chemistry , Oryza/genetics , Oryza/metabolism , Oxidation-Reduction , Oxygen Consumption/physiology , Pisum sativum/chemistry , Pisum sativum/genetics , Pisum sativum/metabolism , Peptides/chemistry , Peptides/metabolism , Photosynthesis/physiology , Plant Leaves/chemistry , Plant Leaves/genetics , Plant Leaves/metabolism , Plant Proteins/chemistry , Plant Proteins/genetics , Plant Proteins/metabolism , Signal Transduction , Tandem Mass Spectrometry/methods , Triticum/chemistry , Triticum/genetics , Triticum/metabolism
7.
Mitochondrion ; 33: 58-71, 2017 Mar.
Article in English | MEDLINE | ID: mdl-27476757

ABSTRACT

Cellular signaling pathways are regulated in a highly dynamic fashion in order to quickly adapt to distinct environmental conditions. Acetylation of lysine residues represents a central process that orchestrates cellular metabolism and signaling. In mitochondria, acetylation seems to be the most prevalent post-translational modification, presumably linked to the compartmentation and high turnover of acetyl-CoA in this organelle. Similarly, the elevated pH and the higher concentration of metabolites in mitochondria seem to favor non-enzymatic lysine modifications, as well as other acylations. Hence, elucidating the mechanisms for metabolic control of protein acetylation is crucial for our understanding of cellular processes. Recent advances in mass spectrometry-based proteomics have considerably increased our knowledge of the regulatory scope of acetylation. Here, we review the current knowledge and functional impact of mitochondrial protein acetylation across species. We first cover the experimental approaches to identify and analyze lysine acetylation on a global scale, we then explore both commonalities and specific differences of plant and animal acetylomes and the evolutionary conservation of protein acetylation, as well as its particular impact on metabolism and diseases. Important future directions and technical challenges are discussed, and it is pointed out that the transfer of knowledge between species and diseases, both in technology and biology, is of particular importance for further advancements in this field.


Subject(s)
Acetyl Coenzyme A/metabolism , Lysine/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Protein Processing, Post-Translational , Acetylation , Animals , Computational Biology , Mass Spectrometry , Plants , Proteomics
8.
Mol Cell ; 63(6): 951-64, 2016 09 15.
Article in English | MEDLINE | ID: mdl-27570076

ABSTRACT

Huntington's disease is one of several neurodegenerative disorders characterized by the aggregation of polyglutamine (polyQ)-expanded mutant protein. How polyQ aggregation leads to cellular dysfunction is not well understood. Here, we analyzed aberrant protein interactions of soluble oligomers and insoluble inclusions of mutant huntingtin using in-cell single molecule fluorescence spectroscopy and quantitative proteomics. We find that the interactome of soluble oligomers is highly complex, with an enrichment of RNA-binding proteins as well as proteins functioning in ribosome biogenesis, translation, transcription, and vesicle transport. The oligomers frequently target proteins containing extended low-complexity sequences, potentially interfering with key cellular pathways. In contrast, the insoluble inclusions are less interactive and associate strongly with protein quality control components, such as Hsp40 chaperones and factors of the ubiquitin-proteasome system. Our results suggest a "multiple hit" model for the pathogenic effects of mutant huntingtin, with soluble forms engaging more extensively in detrimental interactions than insoluble aggregates.


Subject(s)
Huntingtin Protein/metabolism , Neurons/metabolism , Peptides/metabolism , Single Molecule Imaging/methods , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Cell Line, Tumor , Gene Expression , Gene Ontology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HSP40 Heat-Shock Proteins/genetics , HSP40 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Huntingtin Protein/genetics , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Mice , Molecular Sequence Annotation , Mutation , Neurons/pathology , Peptides/chemistry , Peptides/genetics , Protein Aggregates , Protein Interaction Mapping , Protein Multimerization , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Solubility , Spectrometry, Fluorescence , Red Fluorescent Protein
9.
Cell Rep ; 11(7): 1134-46, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25959826

ABSTRACT

Several proteins have been linked to neurodegenerative disorders (NDDs), but their molecular function is not completely understood. Here, we used quantitative interaction proteomics to identify binding partners of Amyloid beta precursor protein (APP) and Presenilin-1 (PSEN1) for Alzheimer's disease (AD), Huntingtin (HTT) for Huntington's disease, Parkin (PARK2) for Parkinson's disease, and Ataxin-1 (ATXN1) for spinocerebellar ataxia type 1. Our network reveals common signatures of protein degradation and misfolding and recapitulates known biology. Toxicity modifier screens and comparison to genome-wide association studies show that interaction partners are significantly linked to disease phenotypes in vivo. Direct comparison of wild-type proteins and disease-associated variants identified binders involved in pathogenesis, highlighting the value of differential interactome mapping. Finally, we show that the mitochondrial protein LRPPRC interacts preferentially with an early-onset AD variant of APP. This interaction appears to induce mitochondrial dysfunction, which is an early phenotype of AD.


Subject(s)
Neurodegenerative Diseases/metabolism , Proteomics/methods , Animals , Chromatography, Liquid , Genome-Wide Association Study , Humans , Immunoprecipitation , Phenotype , Tandem Mass Spectrometry
10.
Mol Cell Proteomics ; 14(7): 2030-41, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25887394

ABSTRACT

The field of proteomics has evolved hand-in-hand with technological advances in LC-MS/MS systems, now enabling the analysis of very deep proteomes in a reasonable time. However, most applications do not deal with full cell or tissue proteomes but rather with restricted subproteomes relevant for the research context at hand or resulting from extensive fractionation. At the same time, investigation of many conditions or perturbations puts a strain on measurement capacity. Here, we develop a high-throughput workflow capable of dealing with large numbers of low or medium complexity samples and specifically aim at the analysis of 96-well plates in a single day (15 min per sample). We combine parallel sample processing with a modified liquid chromatography platform driving two analytical columns in tandem, which are coupled to a quadrupole Orbitrap mass spectrometer (Q Exactive HF). The modified LC platform eliminates idle time between measurements, and the high sequencing speed of the Q Exactive HF reduces required measurement time. We apply the pipeline to the yeast chromatin remodeling landscape and demonstrate quantification of 96 pull-downs of chromatin complexes in about 1 day. This is achieved with only 500 µg input material, enabling yeast cultivation in a 96-well format. Our system retrieved known complex-members and the high throughput allowed probing with many bait proteins. Even alternative complex compositions were detectable in these very short gradients. Thus, sample throughput, sensitivity and LC/MS-MS duty cycle are improved severalfold compared with established workflows. The pipeline can be extended to different types of interaction studies and to other medium complexity proteomes.


Subject(s)
Chromatography, Liquid/methods , Protein Interaction Mapping/methods , Saccharomyces cerevisiae/metabolism , Tandem Mass Spectrometry/methods , Chromatin Assembly and Disassembly , Chromatography, High Pressure Liquid , Reproducibility of Results , Saccharomyces cerevisiae Proteins/metabolism , Time Factors
11.
Mol Cell Proteomics ; 11(6): M111.011767, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22199231

ABSTRACT

RNA polymerase II (RNAPII) transcribes protein-coding genes in eukaryotes and interacts with factors involved in chromatin remodeling, transcriptional activation, elongation, and RNA processing. Here, we present the isolation of native RNAPII complexes using mild extraction conditions and immunoaffinity purification. RNAPII complexes were extracted from mitotic cells, where they exist dissociated from chromatin. The proteomic content of native complexes in total and size-fractionated extracts was determined using highly sensitive LC-MS/MS. Protein associations with RNAPII were validated by high-resolution immunolocalization experiments in both mitotic cells and in interphase nuclei. Functional assays of transcriptional activity were performed after siRNA-mediated knockdown. We identify >400 RNAPII associated proteins in mitosis, among these previously uncharacterized proteins for which we show roles in transcriptional elongation. We also identify, as novel functional RNAPII interactors, two proteins involved in human disease, ALMS1 and TFG, emphasizing the importance of gene regulation for normal development and physiology.


Subject(s)
Mitosis , RNA Polymerase II/metabolism , Chromatography, Gel , Disease , Gene Knockdown Techniques , HeLa Cells , Humans , Immunoprecipitation , Interphase , Nuclear Proteins/genetics , Nuclear Proteins/isolation & purification , Nuclear Proteins/metabolism , Protein Interaction Mapping , Protein Subunits/genetics , Protein Subunits/isolation & purification , Protein Subunits/metabolism , Proteome/genetics , Proteome/isolation & purification , Proteome/metabolism , Proteomics , RNA Interference , RNA Polymerase II/isolation & purification , Ribonucleoproteins/genetics , Ribonucleoproteins/isolation & purification , Ribonucleoproteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/isolation & purification , Ribosomal Proteins/metabolism , Transcription, Genetic
12.
Methods ; 54(4): 387-95, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21382495

ABSTRACT

Since most cellular processes depend on interactions between proteins, information about protein-protein interactions (PPIs) provide valuable insights into protein function. Over the last years, quantitative affinity purification followed by mass spectrometry (q-AP-MS) has become a powerful approach to investigate PPIs in an unbiased manner. In q-AP-MS the protein of interest is biochemically enriched together with its interaction partners. In parallel, a control experiment is performed to control for non-specific binding. Quantitative mass spectrometry is then employed to compare protein levels in both samples and to exclude non-specific contaminants. Here, we provide two detailed q-AP-MS protocols for pull-downs with immobilized bait proteins or transient transfection of tagged expression constructs. We discuss benefits and limitations of q-AP-MS and highlight critical parameters that need to be considered. The protocols and background information presented here allow the reader to adapt the generic q-AP-MS strategy for a wide range of biological questions.


Subject(s)
Protein Interaction Mapping/methods , Proteins/chemistry , Tandem Mass Spectrometry/methods , Chromatography, Liquid/methods , HEK293 Cells , HeLa Cells , Humans , Isotope Labeling
13.
J Biol Chem ; 285(43): 32810-32817, 2010 Oct 22.
Article in English | MEDLINE | ID: mdl-20720017

ABSTRACT

Cysteine synthesis in bacteria and plants is catalyzed by serine acetyltransferase (SAT) and O-acetylserine (thiol)-lyase (OAS-TL), which form the hetero-oligomeric cysteine synthase complex (CSC). In plants, but not in bacteria, the CSC is assumed to control cellular sulfur homeostasis by reversible association of the subunits. Application of size exclusion chromatography, analytical ultracentrifugation, and isothermal titration calorimetry revealed a hexameric structure of mitochondrial SAT from Arabidopsis thaliana (AtSATm) and a 2:1 ratio of the OAS-TL dimer to the SAT hexamer in the CSC. Comparable results were obtained for the composition of the cytosolic SAT from A. thaliana (AtSATc) and the cytosolic SAT from Glycine max (Glyma16g03080, GmSATc) and their corresponding CSCs. The hexameric SAT structure is also supported by the calculated binding energies between SAT trimers. The interaction sites of dimers of AtSATm trimers are identified using peptide arrays. A negative Gibbs free energy (ΔG = -33 kcal mol(-1)) explains the spontaneous formation of the AtCSCs, whereas the measured SAT:OAS-TL affinity (K(D) = 30 nm) is 10 times weaker than that of bacterial CSCs. Free SAT from bacteria is >100-fold more sensitive to feedback inhibition by cysteine than AtSATm/c. The sensitivity of plant SATs to cysteine is further decreased by CSC formation, whereas the feedback inhibition of bacterial SAT by cysteine is not affected by CSC formation. The data demonstrate highly similar quaternary structures of the CSCs from bacteria and plants but emphasize differences with respect to the affinity of CSC formation (K(D)) and the regulation of cysteine sensitivity of SAT within the CSC.


Subject(s)
Arabidopsis/enzymology , Cysteine Synthase/chemistry , Mitochondria/enzymology , Plant Proteins/chemistry , Bacteria/enzymology , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Cysteine/chemistry , Cysteine/metabolism , Cysteine Synthase/metabolism , Plant Proteins/metabolism , Protein Structure, Quaternary , Glycine max/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...