Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Biotechnol Bioeng ; 119(11): 3210-3220, 2022 11.
Article in English | MEDLINE | ID: mdl-35906818

ABSTRACT

Affinity capture is one of the most attractive strategies for simplifying downstream processing. Although it is a key mainstream approach for antibody purification, the same is not true for other biologics such as vaccines, mainly due to the lack of suitable affinity material. In this study, a novel custom affinity system is introduced permitting widespread adoption of affinity capture for the purification of biologics beyond antibodies. This is illustrated here by the development of a one-step purification process of a mutant form of streptolysin O (SLO), a vaccine candidate against Streptococcus pyogenes infection. The system consists of the association of custom ligands based on the Nanofitin protein scaffold, with Eshmuno® industry-grade chromatography medium. The Nanofitins were selected for their specificity to the target product. The newly developed affinity medium was used at different column sizes to monitor scalability from process development (1 ml) and robustness verification (5 ml) to pilot (133 ml) and technical (469 ml) runs. The single-step affinity purification consistently delivered high purity product (above > 90%) and improved performances compared with the current three-step process: reduced process time and footprint (3 to 1 step) and increased product yields (0.31 g vs. 0.04 g of SLO per kg of harvest broth). The custom affinity system herein described can potentially be applied to any biologic for which a specific Nanofitin is identified, thus establishing a platform with a strong impact on the manufacturing of vaccines and other biological targets.


Subject(s)
Streptococcus pyogenes , Vaccines , Chromatography, Affinity/methods , Ligands , Streptococcus pyogenes/genetics
2.
Sci Rep ; 6: 27996, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27302108

ABSTRACT

During bacterial pathogenesis extensive contacts between the human and the bacterial extracellular proteomes take place. The identification of novel host-pathogen interactions by standard methods using a case-by-case approach is laborious and time consuming. To overcome this limitation, we took advantage of large libraries of human and bacterial recombinant proteins. We applied a large-scale protein microarray-based screening on two important human pathogens using two different approaches: (I) 75 human extracellular proteins were tested on 159 spotted Staphylococcus aureus recombinant proteins and (II) Neisseria meningitidis adhesin (NadA), an important vaccine component against serogroup B meningococcus, was screened against ≈2300 spotted human recombinant proteins. The approach presented here allowed the identification of the interaction between the S. aureus immune evasion protein FLIPr (formyl-peptide receptor like-1 inhibitory protein) and the human complement component C1q, key players of the offense-defense fighting; and of the interaction between meningococcal NadA and human LOX-1 (low-density oxidized lipoprotein receptor), an endothelial receptor. The novel interactions between bacterial and human extracellular proteins here presented might provide a better understanding of the molecular events underlying S. aureus and N. meningitidis pathogenesis.


Subject(s)
Host-Pathogen Interactions , Neisseria meningitidis/physiology , Protein Array Analysis/methods , Staphylococcus aureus/physiology , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/metabolism , Animals , Bacterial Proteins/metabolism , Binding Sites , CHO Cells , Complement C1q/metabolism , Cricetulus , Humans , Protein Binding , Recombinant Proteins/metabolism , Scavenger Receptors, Class E/metabolism
3.
Biochem J ; 455(3): 273-84, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23895222

ABSTRACT

In the human pathogen Staphylococcus aureus, there exists an enormous diversity of proteins containing DUFs (domains of unknown function). In the present study, we characterized the family of conserved staphylococcal antigens (Csa) classified as DUF576 and taxonomically restricted to Staphylococci. The 18 Csa paralogues in S. aureus Newman are highly similar at the sequence level, yet were found to be expressed in multiple cellular locations. Extracellular Csa1A was shown to be post-translationally processed and released. Molecular interaction studies revealed that Csa1A interacts with other Csa paralogues, suggesting that these proteins are involved in the same cellular process. The structures of Csa1A and Csa1B were determined by X-ray crystallography, unveiling a peculiar structure with limited structural similarity to other known proteins. Our results provide the first detailed biological characterization of this family and confirm the uniqueness of this family also at the structural level. We also provide evidence that Csa family members elicit protective immunity in in vivo animal models of staphylococcal infections, indicating a possible important role for these proteins in S. aureus biology and pathogenesis. These findings identify the Csa family as new potential vaccine candidates, and underline the importance of mining the bacterial unknown proteome to identify new targets for preventive vaccines.


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Proteome/chemistry , Staphylococcus aureus/metabolism , Animals , Antigens, Bacterial/immunology , Antigens, Bacterial/metabolism , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Crystallography, X-Ray , Data Mining , Mice , Mice, Inbred Strains , Proteome/genetics , Proteome/metabolism , Staphylococcal Infections/immunology , Staphylococcal Infections/metabolism , Staphylococcus aureus/immunology
4.
Biochem J ; 449(3): 683-93, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23113737

ABSTRACT

Staphylococcus aureus is a human pathogen causing globally significant morbidity and mortality. The development of antibiotic resistance in S. aureus highlights the need for a preventive vaccine. In the present paper we explore the structure and function of FhuD2 (ferric-hydroxamate uptake D2), a staphylococcal surface lipoprotein mediating iron uptake during invasive infection, recently described as a promising vaccine candidate. Differential scanning fluorimetry and calorimetry studies revealed that FhuD2 is stabilized by hydroxamate siderophores. The FhuD2-ferrichrome interaction was of nanomolar affinity in surface plasmon resonance experiments and fully iron(III)-dependent. We determined the X-ray crystallographic structure of ligand-bound FhuD2 at 1.9 Å (1 Å=0.1 nm) resolution, revealing the bilobate fold of class III SBPs (solute-binding proteins). The ligand, ferrichrome, occupies a cleft between the FhuD2 N- and C-terminal lobes. Many FhuD2-siderophore interactions enable the specific recognition of ferrichrome. Biochemical data suggest that FhuD2 does not undergo significant conformational changes upon siderophore binding, supporting the hypothesis that the ligand-bound complex is essential for receptor engagement and uptake. Finally, immunizations with FhuD2 alone or FhuD2 formulated with hydroxamate siderophores were equally protective in a murine staphylococcal infection model, confirming the suitability and efficacy of apo-FhuD2 as a protective antigen, and suggesting that other class III SBPs might also be exploited as vaccine candidates.


Subject(s)
Bacterial Proteins/chemistry , Membrane Transport Proteins/chemistry , Periplasmic Binding Proteins/chemistry , Staphylococcus aureus/metabolism , Virulence Factors/chemistry , Animals , Antigens, Bacterial/chemistry , Antigens, Bacterial/genetics , Bacterial Proteins/genetics , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Crystallography, X-Ray , Ferric Compounds/metabolism , Ferrichrome/metabolism , Genes, Bacterial , Humans , Hydroxamic Acids/metabolism , Membrane Transport Proteins/genetics , Membrane Transport Proteins/immunology , Membrane Transport Proteins/metabolism , Mice , Models, Molecular , Periplasmic Binding Proteins/genetics , Periplasmic Binding Proteins/immunology , Periplasmic Binding Proteins/metabolism , Protein Stability , Siderophores/metabolism , Staphylococcal Vaccines/chemistry , Staphylococcus aureus/genetics , Staphylococcus aureus/immunology , Staphylococcus aureus/pathogenicity , Static Electricity , Transferrin/metabolism , Virulence , Virulence Factors/genetics , Virulence Factors/immunology , Virulence Factors/metabolism
5.
Biochemistry ; 51(34): 6738-52, 2012 Aug 28.
Article in English | MEDLINE | ID: mdl-22834735

ABSTRACT

Neisserial adhesin A (NadA) is a surface exposed trimeric protein present in most hypervirulent meningococcal strains and involved in epithelial cell adhesion and colonization. The expression of nadA is controlled by Neisserial adhesin regulator (NadR), a member of the MarR family, which binds to the nadA promoter and strongly represses the transcription of nadA. It was recently demonstrated that the DNA-binding activity of NadR was attenuated by 4-hydroxyphenylacetic acid (4-HPA), a natural molecule released in human saliva, thus leading to the de-repression of nadA in vivo. To elucidate the mechanism of regulation of NadR by 4-HPA, we used hydrogen-deuterium exchange mass spectrometry in association with in silico docking and site-directed mutagenesis. We show here that 4-HPA binds at the interface between the dimerization and the DNA-binding domains and stabilizes the homodimeric state of NadR without inducing large conformational changes in the DNA-binding lobes. The residues predicted to be in contact with 4-HPA were further selected for mutagenesis to assess their in vitro and in vivo functions in 4-HPA binding. Our results indicate that Arg(40) is critical for DNA-binding and reveal that Tyr(115) plays a key role in the mechanism of regulation of NadR by 4-HPA. Altogether our data suggest that the mechanism of regulation of NadR by 4-HPA mainly involves the stabilization of the dimer in a configuration incompatible with DNA binding.


Subject(s)
Bacterial Proteins/metabolism , Neisseria meningitidis/metabolism , Phenylacetates/metabolism , Repressor Proteins/metabolism , Adhesins, Bacterial/chemistry , Adhesins, Bacterial/genetics , Adhesins, Bacterial/metabolism , Amino Acid Motifs , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Dimerization , Gene Expression Regulation, Bacterial , Ligands , Molecular Conformation , Molecular Sequence Data , Neisseria meningitidis/chemistry , Neisseria meningitidis/genetics , Phenylacetates/chemistry , Protein Binding , Protein Conformation , Repressor Proteins/chemistry , Repressor Proteins/genetics
6.
Bioconjug Chem ; 23(6): 1119-26, 2012 Jun 20.
Article in English | MEDLINE | ID: mdl-22594688

ABSTRACT

Protein array technology is an emerging tool that enables high-throughput screening of protein-protein or protein-lipid interactions and identification of immunodominant antigens during the course of a bacterial or viral infection. In this work, we developed an Influenza virus protein array using the sortase-mediated transpeptidation reaction known as "Sortagging". LPETG-tagged Influenza virus proteins from bacterial and eukaryotic cellular extracts were immobilized at their carboxyl-termini onto a preactivated amine-glass slide coated with a Gly3 linker. Immobilized proteins were revealed by specific antibodies, and the newly generated Sortag-protein chip can be used as a device for antigen and/or antibody screening. The specificity of the Sortase A (SrtA) reaction avoids purification steps in array building and allows immobilization of proteins in an oriented fashion. Previously, this versatile technology has been successfully employed for protein labeling and protein conjugation. Here, the tool is implemented to covalently link proteins of a viral genome onto a solid support. The system could readily be scaled up to proteins of larger genomes in order to develop protein arrays for high-throughput screening.


Subject(s)
Aminoacyltransferases/metabolism , Bacterial Proteins/metabolism , Cysteine Endopeptidases/metabolism , Hemagglutinins, Viral/analysis , Immobilized Proteins/analysis , Influenza A virus/chemistry , Protein Array Analysis/instrumentation , Viral Proteins/analysis , Amino Acid Sequence , Cell Line , Cloning, Molecular , Equipment Design , Escherichia coli/genetics , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/metabolism , Humans , Immobilized Proteins/genetics , Immobilized Proteins/metabolism , Influenza A virus/genetics , Influenza A virus/metabolism , Influenza, Human/virology , Viral Proteins/genetics , Viral Proteins/metabolism
7.
Proc Natl Acad Sci U S A ; 109(14): 5229-34, 2012 Apr 03.
Article in English | MEDLINE | ID: mdl-22431623

ABSTRACT

CRM197 is an enzymatically inactive and nontoxic form of diphtheria toxin that contains a single amino acid substitution (G52E). Being naturally nontoxic, CRM197 is an ideal carrier protein for conjugate vaccines against encapsulated bacteria and is currently used to vaccinate children globally against Haemophilus influenzae, pneumococcus, and meningococcus. To understand the molecular basis for lack of toxicity in CRM197, we determined the crystal structures of the full-length nucleotide-free CRM197 and of CRM197 in complex with the NAD hydrolysis product nicotinamide (NCA), both at 2.0-Å resolution. The structures show for the first time that the overall fold of CRM197 and DT are nearly identical and that the striking functional difference between the two proteins can be explained by a flexible active-site loop that covers the NAD binding pocket. We present the molecular basis for the increased flexibility of the active-site loop in CRM197 as unveiled by molecular dynamics simulations. These structural insights, combined with surface plasmon resonance, NAD hydrolysis, and differential scanning fluorimetry data, contribute to a comprehensive characterization of the vaccine carrier protein, CRM197.


Subject(s)
Bacterial Proteins/toxicity , Mutation , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Models, Molecular , Molecular Dynamics Simulation , NAD/metabolism , Protein Conformation
8.
Sci Transl Med ; 4(123): 123ps5, 2012 Feb 29.
Article in English | MEDLINE | ID: mdl-22378923

ABSTRACT

Meningococcal meningitis is a devastating disease that is often fatal. Vaccines against the five major meningococcal serogroups causing disease are about to become available, a conjugate vaccine against meningococcus A is in use for mass vaccination in Africa, and a protein-based vaccine against meningococcal B is ready for licensure. With the availability of these new vaccines, the world can finally be rid of meningococcal meningitis, thus rewriting a new chapter in medical history.


Subject(s)
Disease Eradication , Meningitis, Meningococcal/prevention & control , Meningococcal Vaccines/therapeutic use , Neisseria meningitidis/immunology , Congresses as Topic , Developing Countries , Drug Design , Global Health , Health Services Accessibility , Humans , Meningitis, Meningococcal/epidemiology , Meningitis, Meningococcal/immunology , Meningitis, Meningococcal/microbiology , Meningococcal Vaccines/immunology , National Health Programs , Neisseria meningitidis/classification , Neisseria meningitidis/pathogenicity
9.
Proteomics ; 10(5): 904-13, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20049858

ABSTRACT

Using antibody-related methods, we recently found that human thyroid cells express various proteins differently depending on whether they are cultured under normal gravity (1g) or simulated microgravity (s-microg). In this study, we performed proteome analysis in order to identify more gravity-sensitive thyroid proteins. Cells cultured under 1g or s-microg conditions were sonicated. Proteins released into the supernatant and those remaining in the cell fragments were fractionated by free-flow IEF. The fractions obtained were further separated by SDS-gel electrophoresis. Selected gel pieces were excised and their proteins were determined by MS. A total of 235 different proteins were found. Out of 235 proteins, 37 appeared to be first identifications in human thyroid cells. Comparing SDS gel lanes of equally numbered free-flow IEF fractions revealed similar patterns with a number of identical bands if proteins of a distinct cell line had been applied, irrespective of whether the cells had been cultured under 1g or s-microg. Most of the identical band pairs contained identical proteins. However, the concentrations of some types of proteins were different within the two pieces of gel. Proteins that concentrated differently in such pieces of gel are considered as candidates for further investigations of gravitational sensitivity.


Subject(s)
Isoelectric Focusing/methods , Proteome/analysis , Weightlessness , Blotting, Western , Cell Extracts , Cell Line, Tumor , Electrophoresis, Polyacrylamide Gel , Humans , Hydrogen-Ion Concentration , Mass Spectrometry , Peptides/analysis , Silver Staining , Sonication
10.
Arch Physiol Biochem ; 115(5): 267-78, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19883214

ABSTRACT

Adiponectin is an important adipocytokine hormone which circulates in blood as homo-oligomers (trimer, hexamer and high molecular weight (HMW) forms) as well as a truncated form corresponding to the globular domain. Free flow electrophoresis (FFE) used in zone electrophoresis mode revealed the presence of isoforms within these oligomeric forms in plasma. HMW adiponectin oligomer showed two isoforms which carry different charge density at pH 4.7, only one of which is susceptible to dissociation by SDS. The adiponectin hexamer was shown to consist of a doublet and also shown to have at least two isoforms. A truncated form of adiponectin was identified as the main constituent of adiponectin in plasma and appeared to circulate bound to a basic protein, potentially one of the chemokines reported to bind to the globular domain. Analysis of the monomer composition of the oligomers revealed differences in monomeric isoforms used to build up the oligomers.


Subject(s)
Adiponectin/blood , Electrophoresis/methods , Humans , Protein Isoforms/chemistry , Protein Multimerization
11.
Proteomics ; 9(21): 4991-6, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19810032

ABSTRACT

Human umbilical vein endothelial cells are the most widely used in vitro model for endothelial cells. Their secreted proteins, however, have not been comprehensively analysed so far. In this study, we accomplished to map the secretome of human umbilical vein endothelial cells by combining free-flow electrophoresis with nanoflow LC-MS/MS. This comprehensive analysis provides a basis for future comparative studies of protein secretion by endothelial cells in response to cardiovascular risk factors and is available on our website http://www.vascular-proteomics.com.


Subject(s)
Chromatography, Liquid/methods , Electrophoresis/methods , Endothelial Cells/metabolism , Proteins/analysis , Proteomics/methods , Tandem Mass Spectrometry/methods , Umbilical Veins/metabolism , Chromatography, Liquid/instrumentation , Electrophoresis/instrumentation , Endothelial Cells/chemistry , Humans , Proteins/metabolism , Proteomics/instrumentation , Tandem Mass Spectrometry/instrumentation , Umbilical Veins/chemistry
12.
Mol Cell Proteomics ; 8(6): 1424-35, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19193607

ABSTRACT

Fetal growth restriction (FGR) is a common disorder in which a fetus is unable to achieve its genetically determined potential size. High concentrations of insulin-like growth factor-binding protein-1 (IGFBP-1) have been associated with FGR. Phosphorylation of IGFBP-1 is a mechanism by which insulin-like growth factor-I (IGF-I) bioavailability can be modulated in FGR. In this study a novel strategy was designed to determine a link between IGF-I affinity and the concomitant phosphorylation state characteristics of IGFBP-1 phosphoisoforms. Using free flow electrophoresis (FFE), multiple IGFBP-1 phosphoisoforms in amniotic fluid were resolved within pH 4.43-5.09. The binding of IGFBP-1 for IGF-I in each FFE fraction was determined with BIAcore biosensor analysis. The IGF-I affinity (K(D)) for different IGFBP-1 isoforms ranged between 1.12e-08 and 4.59e-07. LC-MS/MS characterization revealed four phosphorylation sites, Ser(P)(98), Ser(P)(101), Ser(P)(119), and Ser(P)(169), of which Ser(P)(98) was new. Although the IGF-I binding affinity for IGFBP-1 phosphoisoforms across the FFE fractions did not correlate with phosphopeptide intensities for Ser(P)(101), Ser(P)(98), and Ser(P)(169) sites, a clear association was recorded with Ser(P)(119). Our data demonstrate that phosphorylation at Ser(119) plays a significant role in modulating affinity of IGFBP-1 for IGF-I. In addition, an altered profile of IGFBP-1 phosphoisoforms was revealed between FGR and healthy pregnancies that may result from potential site-specific phosphorylation. This study provides a strong basis for use of this novel approach in establishing the linkage between phosphorylation of IGFBP-1 and FGR. This overall strategy will also be broadly applicable to other phosphoproteins with clinical and functional significance.


Subject(s)
Insulin-Like Growth Factor Binding Protein 1/metabolism , Isoelectric Focusing/methods , Protein Isoforms/metabolism , Biosensing Techniques , Blotting, Western , Electrophoresis, Polyacrylamide Gel , Humans , Phosphorylation
13.
Expert Rev Proteomics ; 5(4): 571-87, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18761468

ABSTRACT

Due to ease of accessibility, plasma has become the sample of choice for proteomics studies directed towards biomarker discovery intended for use in diagnostics, prognostics and even in theranostics. The result of these extensive efforts is a long list of potential biomarkers, very few of which have led to clinical utility. Why have so many potential biomarkers failed validation? Herein, we address certain issues encountered, which complicate biomarker discovery efforts originating from plasma. The advantages of stabilizing the sample at collection by the addition of protease inhibitors are discussed. The principles of free-flow electrophoresis (FFE) separation are provided together with examples applying to various studies. Finally, particular attention is given to plasma or serum analysis using multidimensional separation strategies into which the FFE is incorporated. The advantages of using FFE separation in these workflows are discussed.


Subject(s)
Blood Proteins/analysis , Blood Proteins/isolation & purification , Electrophoresis/methods , Proteomics , Electrophoresis/instrumentation , Humans
15.
Methods Mol Biol ; 484: 131-44, 2008.
Article in English | MEDLINE | ID: mdl-18592177

ABSTRACT

Prefractionation of complex protein samples prior to mass spectrometry provides a method for the isolation of low-abundance proteins into specific fractions thereby enabling their identification. Free-flow electrophoresis in the isoelectric focusing mode (IEF-FFE) presents a complementary approach to established prefractionation methodologies. Proteins are separated in solution according to their isoelectric point (pI) with a high throughput of sample volume. The separation may be performed under denaturing or nondenaturing conditions and detergents may be added to promote protein solubilization. A protocol covering the pH range from pH 3 to 9 under denaturing conditions was used to illustrate the method of IEF-FFE including sample preparation prior to reversed-phase liquid chromatography and tandem mass spectrometry. The IEF-FFE separation was applied to a sample of human urine.


Subject(s)
Electrophoresis/methods , Proteome/analysis , Proteomics/methods , Urine/chemistry , Electrophoresis/instrumentation , Humans , Isoelectric Focusing/instrumentation , Isoelectric Focusing/methods , Mass Spectrometry/instrumentation , Mass Spectrometry/methods , Proteomics/instrumentation
16.
Proteomics ; 8(14): 2798-808, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18655049

ABSTRACT

High-efficiency prefractionation of complex protein mixtures is critical for top-down proteomics, i.e., the analysis of intact proteins by MS. Free-flow electrophoresis (FFE) can be used for IEF to separate proteins within a pH gradient according to their pIs. In an FFE system, this separation is performed entirely in the liquid phase, without the need for particulate chromatographic media, gels, or membranes. Herein, we demonstrated the compatibility of IEF-FFE with ESI-Fourier transform ICR MS (ESI-FTICR-MS) for top-down experiments. We demonstrated that IEF-FFE of intact proteins were highly reproducible between FFE instruments, between laboratories, and between analyses. Applying native (0.2% hydroxypropylmethyl cellulose) IEF-FFE to an enzyme resulted in no decrease in enzyme activity; applying either native or denaturing (8 M urea) IEF-FFE to a four-protein mixture with different pIs resulted in isolation of each protein into separate fractions in a 96-well plate. After desalting, each protein was sequenced by top-down MS/MS. As an application of this technique, chicken erythrocyte histone H2A-IV and its major modified forms were enriched by IEF-FFE. Top-down analysis revealed Lys-5 to be a major acetylation site, in addition to N-terminal acetylation.


Subject(s)
Cyclotrons , Fourier Analysis , Isoelectric Focusing/methods , Proteomics/methods , Spectrometry, Mass, Electrospray Ionization , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Amino Acid Sequence , Animals , Cattle , Chickens , Horses , Hydrogen-Ion Concentration , Molecular Sequence Data , Proteomics/instrumentation , Spectrometry, Mass, Electrospray Ionization/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
17.
Methods Mol Biol ; 424: 287-300, 2008.
Article in English | MEDLINE | ID: mdl-18369870

ABSTRACT

This chapter describes the technology of free flow electrophoresis (FFE) and protocols to separate human plasma for proteome analysis. FFE is a highly versatile technology applied in the field of proteomics because of its continuous processing of sample and high resolution in separation of most kinds of charged or chargeable particles including ions, proteins peptides, organelles, and whole cells. FFE is carried out in an aqueous medium without inducing any solid matrix, such as acrylamide, so that it simplifies complex sample for the downstream analysis. Two FFE protocols are described to separate human plasma proteins under native and denaturing conditions. Plasma separated under native conditions was pooled into acidic-, alkaline-, and albumin- fractions that were furthered for gel-based analysis. Under denaturing condition plasma proteins were separated into 96 fractions. Each fraction can be supplied for in-solution digestion and further LC-MS/MS analysis. From a single FFE fraction 46 different proteins (protein family) have been identified, demonstrating FFE as a high efficient separation tool for human plasma proteome studies.


Subject(s)
Blood Proteins/analysis , Electrophoresis/methods , Plasma/chemistry , Proteome/analysis , Proteomics/methods , Blood Protein Electrophoresis/methods , Blood Proteins/chemistry , Humans , Proteome/chemistry
18.
Proteomics ; 7(23): 4218-27, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17973290

ABSTRACT

Blood plasma is the most complex human-derived proteome, containing other tissue proteomes as subsets. This proteome has only been partially characterized due to the extremely wide dynamic range of the plasma proteins of more than ten orders of magnitude. Thus, the reduction in sample complexity prior to mass spectrometric analysis is particularly important and alternative separation methodologies are required to more effectively mine the lower abundant plasma proteins. Here, we demonstrated a novel separation approach using 2-D free-flow electrophoresis (FFE) separating proteins and peptides in solution according to their pI prior to LC-MS/MS. We used the combination of sequential protein and peptide separation by first separating the plasma proteins into specific FFE fractions. Tryptic digests of the separated proteins were generated and subsequently separated using FFE. The protein separation medium was optimized to segregate albumin into specific fractions containing only few other proteins. An optimization of throughput for the protein separation reduced the separation time of 1 mL of plasma to approximately 3 h providing sufficient material for digestion and the subsequent peptide separation. Our approach revealed low-abundant proteins (e.g., L-selectin at 17 ng/mL and vascular endothelial-cadherin precursor at 30 ng/mL) and several tissue leakage products, thus providing a powerful orthogonal separation step in the proteomics workflow.


Subject(s)
Blood Proteins/analysis , Proteome/analysis , Proteomics/methods , Blood Protein Electrophoresis/instrumentation , Blood Protein Electrophoresis/methods , Blood Proteins/chemistry , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Humans , Hydrogen-Ion Concentration , Isoelectric Focusing/instrumentation , Isoelectric Focusing/methods , Isoelectric Point , Peptide Fragments/analysis , Peptide Fragments/chemistry , Proteome/chemistry , Proteomics/instrumentation , Serum Albumin/chemistry , Tandem Mass Spectrometry , Trypsin/chemistry
19.
Mol Cell Proteomics ; 6(11): 2021-31, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17660509

ABSTRACT

The proteasome-dependent protein degradation participates in multiple essential cellular processes. Modulation of proteasomal activities may alter cardiac function and disease phenotypes. However, cardiovascular studies reported thus far have yielded conflicting results. We hypothesized that a contributing factor to the contradicting literature may be caused by existing proteasome heterogeneity in the myocardium. In this investigation, we provide the very first direct demonstration of distinct proteasome subpopulations in murine hearts. The cardiac proteasome subpopulations differ in their molecular compositions and proteolytic activities. Furthermore they were distinguished from proteasome subpopulations identified in murine livers. The study was facilitated by the development of novel protocols for in-solution isoelectric focusing of multiprotein complexes in a laminar flow that support an average resolution of 0.04 pH units. Utilizing these protocols, the majority of cardiac proteasome complexes displayed an isoelectric point of 5.26 with additional subpopulations focusing in the range from pH 5.10 to 5.33. In contrast, the majority of hepatic 20 S proteasomes had a pI of 5.05 and focused from pH 5.01 to 5.29. Importantly proteasome subpopulations degraded specific model peptides with different turnover rates. Among cardiac subpopulations, proteasomes with an approximate pI of 5.21 showed 40% higher trypsin-like activity than those with pI 5.28. Distinct proteasome assembly may be a contributing factor to variations in proteolytic activities because proteasomes with pI 5.21 contained 58% less of the inducible subunit beta 2i compared with those with pI 5.28. In addition, dephosphorylation of 20 S proteasomes demonstrated that besides molecular composition posttranslational modifications largely contribute to their pI values. These data suggest the possibility of mixed 20 S proteasome assembly, a departure from the currently hypothesized two subpopulations: constitutive and immuno forms. The identification of multiple distinct proteasome subpopulations in heart provides key mechanistic insights for achieving selective and targeted regulation of this essential protein degradation machinery. Thus, proteasome subpopulations may serve as novel therapeutic targets in the myocardium.


Subject(s)
Cell Fractionation/methods , Isoelectric Focusing/methods , Myocardium/chemistry , Proteasome Endopeptidase Complex/chemistry , Animals , Hydrolysis , Liver/chemistry , Mice
20.
Anal Chem ; 76(7): 2040-5, 2004 Apr 01.
Article in English | MEDLINE | ID: mdl-15053669

ABSTRACT

We present a new procedure for in-gel digestion of proteins introducing a combination of two different 96-well microplates. The two plates have incorporated small capillaries with a length of 2.4 mm in each well, one of which has 75-microm-inner diameter capillaries, whereas the second plate has reversed-phase-type capillaries fixed to it. The initial steps of the in-gel digestion process, comprising destaining, reduction/alkylation, dehydration, and digestion, was carried out in the plate containing 75-microm capillaries. Capillaries containing C18 reversed-phase modified monolithic silica rods of a 200-microm diameter were used for the second plate in which extraction and cleanup of peptides were carried out. Peptides were eluted directly from the solid-phase extraction plate onto the MALDI sample support. The separation of the process into two plates led to increased process stability, without compromising sensitivity, i.e. peptide recovery, making it suitable for true high-throughput protein identification. The handling of proteinases could easily be optimized, and no restrictions were made on chosen pH range through the absence of the solid phase in the initial steps of the protocol. Efficient binding of peptides to the solid phase and subsequent direct elution onto the MALDI sample support led to sensitivities in the attomole range. Performance of the process was demonstrated with tryptic digests of proteins stained with colloidal coomassie blue, silver, and the fluorescent stain SYPRO Ruby.


Subject(s)
Proteins/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/instrumentation , Animals , Electrophoresis, Gel, Two-Dimensional , Fungal Proteins/chemistry , Fungal Proteins/metabolism , Hydrolysis , Liver/chemistry , Liver/enzymology , Peptides/isolation & purification , Proteins/chemistry , Proteins/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
SELECTION OF CITATIONS
SEARCH DETAIL
...