Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Front Immunol ; 11: 296, 2020.
Article in English | MEDLINE | ID: mdl-32184781

ABSTRACT

The strong links between (Human Leukocyte Antigen) HLA, infection and autoimmunity combine to implicate T-cells as primary triggers of autoimmune disease (AD). T-cell crossreactivity between microbially-derived peptides and self-peptides has been shown to break tolerance and trigger AD in experimental animal models. Detailed examination of the potential for T-cell crossreactivity to trigger human AD will require means of predicting which peptides might be recognised by autoimmune T-cell receptors (TCRs). Recent developments in high throughput sequencing and bioinformatics mean that it is now possible to link individual TCRs to specific pathologies for the first time. Deconvolution of TCR function requires knowledge of TCR specificity. Positional Scanning Combinatorial Peptide Libraries (PS-CPLs) can be used to predict HLA-restriction and define antigenic peptides derived from self and pathogen proteins. In silico search of the known terrestrial proteome with a prediction algorithm that ranks potential antigens in order of recognition likelihood requires complex, large-scale computations over several days that are infeasible on a personal computer. We decreased the time required for peptide searching to under 30 min using multiple blocks on graphics processing units (GPUs). This time-efficient, cost-effective hardware accelerator was used to screen bacterial and fungal human pathogens for peptide sequences predicted to activate a T-cell clone, InsB4, that was isolated from a patient with type 1 diabetes and recognised the insulin B-derived epitope HLVEALYLV in the context of disease-risk allele HLA A*0201. InsB4 was shown to kill HLA A*0201+ human insulin producing ß-cells demonstrating that T-cells with this specificity might contribute to disease. The GPU-accelerated algorithm and multispecies pathogen proteomic databases were validated to discover pathogen-derived peptide sequences that acted as super-agonists for the InsB4 T-cell clone. Peptide-MHC tetramer binding and surface plasmon resonance were used to confirm that the InsB4 TCR bound to the highest-ranked peptide agonists derived from infectious bacteria and fungi. Adoption of GPU-accelerated prediction of T-cell agonists has the capacity to revolutionise our understanding of AD by identifying potential targets for autoimmune T-cells. This approach has further potential for dissecting T-cell responses to infectious disease and cancer.


Subject(s)
Epitopes, T-Lymphocyte/metabolism , Insulin/metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , Clone Cells , Combinatorial Chemistry Techniques , Computational Biology , Cross Reactions , Epitope Mapping , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , High-Throughput Nucleotide Sequencing , Host-Pathogen Interactions , Insulin/immunology , Molecular Mimicry , Pathogen-Associated Molecular Pattern Molecules/immunology , Peptide Library , T-Cell Antigen Receptor Specificity
2.
PLoS One ; 13(6): e0199323, 2018.
Article in English | MEDLINE | ID: mdl-29924875

ABSTRACT

Coxsackie B Virus (CBV) infection has been linked to the aetiology of type 1 diabetes (T1D) and vaccination has been proposed as prophylaxis for disease prevention. Serum neutralising antibodies and the presence of viral protein and RNA in tissues have been common tools to examine this potential disease relationship, whilst the role of anti-CBV cytotoxic T cell responses and their targets have not been studied. To address this knowledge gap, we augmented conventional HLA-binding predictive algorithm-based epitope discovery by cross-referencing epitopes with sites of positive natural selection within the CBV3 viral genome, identified using mixed effects models of evolution. Eight epitopes for the common MHC class I allele HLA-A*0201 occur at sites that appear to be positively selected. Furthermore, such epitopes span the viral genome, indicating that effective anti-viral responses may not be restricted to the capsid region. To assess the spectrum of IFNy responses in non-diabetic subjects and recently diagnosed type 1 diabetes (T1D) patients, we stimulated PBMC ex vivo with pools of synthetic peptides based on component-restricted sequences identified in silico. We found responders were more likely to recognize multiple rather than a single CBV peptide pool, indicating that the natural course of infection results in multiple targets for effector memory responses, rather than immunodominant epitopes or viral components. The finding that anti-CBV CD8 T cell immunity is broadly targeted has implications for vaccination strategies and studies on the pathogenesis of CBV-linked diseases.


Subject(s)
Capsid/metabolism , Computer Simulation , Enterovirus B, Human/metabolism , Immune System/virology , Amino Acid Sequence , Base Sequence , Case-Control Studies , Diabetes Mellitus/virology , Enterovirus B, Human/genetics , Epitopes/chemistry , Epitopes/immunology , Evolution, Molecular , Genome, Viral , Histocompatibility Antigens Class I/metabolism , Humans , Interferon-gamma/metabolism , Peptides/chemistry , Peptides/metabolism , Phylogeny , Selection, Genetic , Serotyping
3.
Diabetes ; 64(3): 916-925, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25249579

ABSTRACT

Autoreactive CD8 T cells play a central role in the destruction of pancreatic islet ß-cells that leads to type 1 diabetes, yet the key features of this immune-mediated process remain poorly defined. In this study, we combined high-definition polychromatic flow cytometry with ultrasensitive peptide-human leukocyte antigen class I tetramer staining to quantify and characterize ß-cell-specific CD8 T cell populations in patients with recent-onset type 1 diabetes and healthy control subjects. Remarkably, we found that ß-cell-specific CD8 T cell frequencies in peripheral blood were similar between subject groups. In contrast to healthy control subjects, however, patients with newly diagnosed type 1 diabetes displayed hallmarks of antigen-driven expansion uniquely within the ß-cell-specific CD8 T cell compartment. Molecular analysis of selected ß-cell-specific CD8 T cell populations further revealed highly skewed oligoclonal T cell receptor repertoires comprising exclusively private clonotypes. Collectively, these data identify novel and distinctive features of disease-relevant CD8 T cells that inform the immunopathogenesis of type 1 diabetes.


Subject(s)
Autoantigens/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , Adult , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation/physiology , Female , Flow Cytometry , Glutamate Decarboxylase/immunology , Humans , Insulin-Secreting Cells/cytology , Male , Receptor-Like Protein Tyrosine Phosphatases, Class 8/immunology
4.
J Immunol ; 190(9): 4542-52, 2013 May 01.
Article in English | MEDLINE | ID: mdl-23547116

ABSTRACT

A major goal of immunotherapy remains the control of pathogenic T cell responses that drive autoimmunity and allograft rejection. Adherent progenitor cells, including mesenchymal stromal cells (MSCs) and multipotent adult progenitor cells (MAPCs), represent attractive immunomodulatory cell therapy candidates currently active in clinical trials. MAPCs can be distinguished from MSCs on the basis of cellular phenotype, size, transcriptional profile, and expansion capacity. However, despite their ongoing evaluation in autoimmune and allogeneic solid organ transplantation settings, data supporting the immune regulatory potential of clinical-grade MAPCs are limited. In this study, we used allogeneic islet transplantation as a model indication to assess the ability of clinical-grade MAPCs to control T cell responses that drive immunopathology in human autoimmune disease and allograft rejection. MAPCs suppressed T cell proliferation and Th1 and Th17 cytokine production while increasing secretion of IL-10 and were able to suppress effector functions of bona fide autoreactive T cells from individuals with type 1 diabetes mellitus, including killing of human islets. Furthermore, MAPCs favored the proliferation of regulatory T cells during homeostatic expansion driven by γ-chain cytokines and exerted a durable, yet reversible, control of T cell function. MAPC suppression required licensing and proceeded via IDO-mediated tryptophan catabolism. Therefore, the common immune modulatory characteristics of clinical-grade MAPCs shown in this study suggest that they can be regarded as an alternative source of adult progenitor cells with similar clinical usefulness to MSCs. Taken collectively, these findings may guide the successful deployment of both MSCs and MAPCs for the amelioration of human autoimmunity and allograft rejection.


Subject(s)
Autoimmunity/immunology , Islets of Langerhans Transplantation/immunology , Lymphocyte Activation/immunology , Stem Cells/immunology , T-Lymphocytes, Regulatory/immunology , Th1 Cells/immunology , Th17 Cells/immunology , Adult , Adult Stem Cells/immunology , Cell Proliferation , Cells, Cultured , Diabetes Mellitus, Type 1/immunology , Graft Rejection/immunology , Humans , Immunomodulation/immunology , Interleukin-10/immunology , Male , Tryptophan/immunology , Young Adult
5.
Diabetes ; 62(1): 205-13, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22936177

ABSTRACT

The end-stage immunopathology of type 1 diabetes resulting in ß-cell destruction appears to be strongly dominated by cytotoxic CD8 T lymphocytes (CD8 T cells). However, the mechanism of cytotoxicity used by autoreactive CD8 T cells in the human setting remains unknown. Using type 1 diabetes patient-derived preproinsulin-specific CD8 T-cell clones recognizing either an HLA-A2 (A*0201) or HLA-A24 (A*2402)-restricted epitope (peptide of preproinsulin [PPI](15-24), ALWGPDPAAA; or PPI(3-11), LWMRLLPLL), we assessed the use of conventional mediators of cytotoxicity in the destruction of human ß-cells in vitro compared with virus-specific cytotoxic CD8 T-cell clones. We show that PPI-specific CD8 T-cell clones are mainly reliant upon cytotoxic degranulation for inducing ß-cell death. Furthermore, we find that in comparison with virus-specific CD8 T cells, there are differences in the killing potency of PPI-specific CD8 T cells that are not due to cell-intrinsic differences, but rather are mediated by differences in strength of signaling by peptide-HLA ligands. The study highlights the regulation of ß-cell killing as a potential point for therapeutic control, including the possibility of blocking autoreactive CD8 T-cell function without impacting upon general immune competence.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cell Degranulation , Cytotoxicity, Immunologic , Insulin-Secreting Cells/pathology , Insulin/immunology , Protein Precursors/immunology , Receptors, Antigen, T-Cell/physiology , Cell Line , Fas Ligand Protein/physiology , Humans , Tumor Necrosis Factor-alpha/physiology , fas Receptor/physiology
6.
Diabetes ; 61(7): 1752-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22522618

ABSTRACT

Type 1 diabetes results from T cell-mediated ß-cell destruction. The HLA-A*24 class I gene confers significant risk of disease and early onset. We tested the hypothesis that HLA-A24 molecules on islet cells present preproinsulin (PPI) peptide epitopes to CD8 cytotoxic T cells (CTLs). Surrogate ß-cell lines secreting proinsulin and expressing HLA-A24 were generated and their peptide ligandome examined by mass spectrometry to discover naturally processed and HLA-A24-presented PPI epitopes. A novel PPI epitope was identified and used to generate HLA-A24 tetramers and examine the frequency of PPI-specific T cells in new-onset HLA-A*24(+) patients and control subjects. We identified a novel naturally processed and HLA-A24-presented PPI signal peptide epitope (PPI(3-11); LWMRLLPLL). HLA-A24 tetramer analysis reveals a significant expansion of PPI(3-11)-specific CD8 T cells in the blood of HLA-A*24(+) recent-onset patients compared with HLA-matched control subjects. Moreover, a patient-derived PPI(3-11)-specific CD8 T-cell clone shows a proinflammatory phenotype and kills surrogate ß-cells and human HLA-A*24(+) islet cells in vitro. These results indicate that the type 1 diabetes susceptibility molecule HLA-A24 presents a naturally processed PPI signal peptide epitope. PPI-specific, HLA-A24-restricted CD8 T cells are expanded in patients with recent-onset disease. Human islet cells process and present PPI(3-11), rendering themselves targets for CTL-mediated killing.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , HLA-A24 Antigen/immunology , Insulin-Secreting Cells/immunology , Insulin/immunology , Protein Precursors/immunology , Protein Sorting Signals , Adult , Autoantibodies/blood , Autoantibodies/immunology , Cell Death/immunology , Cell Line , Epitopes, T-Lymphocyte/immunology , Female , Glutamate Decarboxylase/immunology , Humans , Insulin/blood , Male , Middle Aged , Protein Precursors/blood , Young Adult
7.
Nat Immunol ; 13(3): 283-9, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22245737

ABSTRACT

The structural characteristics of the engagement of major histocompatibility complex (MHC) class II-restricted self antigens by autoreactive T cell antigen receptors (TCRs) is established, but how autoimmune TCRs interact with complexes of self peptide and MHC class I has been unclear. Here we examined how CD8(+) T cells kill human islet beta cells in type 1 diabetes via recognition of a human leukocyte antigen HLA-A*0201-restricted glucose-sensitive preproinsulin peptide by the autoreactive TCR 1E6. Rigid 'lock-and-key' binding underpinned the 1E6-HLA-A*0201-peptide interaction, whereby 1E6 docked similarly to most MHC class I-restricted TCRs. However, this interaction was extraordinarily weak because of limited contacts with MHC class I. TCR binding was highly peptide centric, dominated by two residues of the complementarity-determining region 3 (CDR3) loops that acted as an 'aromatic-cap' over the complex of peptide and MHC class I (pMHCI). Thus, highly focused peptide-centric interactions associated with suboptimal TCR-pMHCI binding affinities might lead to thymic escape and potential CD8(+) T cell-mediated autoreactivity.


Subject(s)
Apoptosis , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Insulin-Secreting Cells/immunology , CD8-Positive T-Lymphocytes/chemistry , Histocompatibility Antigens/immunology , Humans , Insulin-Secreting Cells/pathology , Models, Molecular , Protein Structure, Tertiary , Receptors, Antigen, T-Cell/chemistry , Receptors, Antigen, T-Cell/immunology
8.
Physiol Meas ; 32(6): 619-35, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21493981

ABSTRACT

The QT interval in the electrocardiogram (ECG) is a measure of total duration of depolarization and repolarization. Correction for heart rate is necessary to provide a single intrinsic physiological value that can be compared between subjects and within the same subject under different conditions. Standard formulas for the corrected QT (QTc) do not fully reproduce the complexity of the dependence in the preceding interbeat intervals (RR) and inter-subject variability. In this paper, a subject-specific, nonlinear, transfer function-based correction method is formulated to compute the QTc from Holter ECG recordings. The model includes five parameters: three describing the static QT-RR relationship and two representing memory/hysteresis effects that intervene in the calculation of effective RR values. The parameter identification procedure is designed to minimize QTc fluctuations and enforce zero correlation between QTc and effective RR. Weighted regression is used to better handle unbalanced or skewed RR distributions. The proposed optimization approach provides a general mathematical framework for further extensions of the model. Validation, robustness evaluation and comparison with existing QT correction formulas is performed on ECG signals recorded during sinus rhythm, atrial pacing, tilt-table tests, stress tests and atrial flutter (29 subjects in total). The resulting average modeling error on the QTc is 4.9 ± 1.1 ms with a sampling interval of 2 ms, which outperforms correction formulas currently used. The results demonstrate the benefits of subject-specific rate correction and hysteresis reduction.


Subject(s)
Electrocardiography/methods , Heart Conduction System/physiology , Nonlinear Dynamics , Atrial Flutter/physiopathology , Heart Rate/physiology , Humans , Models, Cardiovascular , Pacemaker, Artificial , Reproducibility of Results
9.
PLoS Biol ; 5(7): e156, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17579514

ABSTRACT

The adult human intestine contains trillions of bacteria, representing hundreds of species and thousands of subspecies. Little is known about the selective pressures that have shaped and are shaping this community's component species, which are dominated by members of the Bacteroidetes and Firmicutes divisions. To examine how the intestinal environment affects microbial genome evolution, we have sequenced the genomes of two members of the normal distal human gut microbiota, Bacteroides vulgatus and Bacteroides distasonis, and by comparison with the few other sequenced gut and non-gut Bacteroidetes, analyzed their niche and habitat adaptations. The results show that lateral gene transfer, mobile elements, and gene amplification have played important roles in affecting the ability of gut-dwelling Bacteroidetes to vary their cell surface, sense their environment, and harvest nutrient resources present in the distal intestine. Our findings show that these processes have been a driving force in the adaptation of Bacteroidetes to the distal gut environment, and emphasize the importance of considering the evolution of humans from an additional perspective, namely the evolution of our microbiomes.


Subject(s)
Bacteroides/genetics , Evolution, Molecular , Intestines/microbiology , Symbiosis/genetics , Adaptation, Physiological , Bacteriophages/genetics , Bacteroides/physiology , Bacteroides/virology , Conjugation, Genetic , DNA Transposable Elements , Ecosystem , Gene Duplication , Gene Transfer, Horizontal , Genetic Variation , Genome, Bacterial , Humans , Molecular Sequence Data , Phylogeny , Polysaccharides, Bacterial/biosynthesis , Polysaccharides, Bacterial/genetics , RNA, Bacterial/genetics , RNA, Ribosomal, 16S/genetics , Species Specificity
10.
PLoS Biol ; 4(5): e127, 2006 May.
Article in English | MEDLINE | ID: mdl-16620150

ABSTRACT

The genetic code provides the translation table necessary to transform the information contained in DNA into the language of proteins. In this table, a correspondence between each codon and each amino acid is established: tRNA is the main adaptor that links the two. Although the genetic code is nearly universal, several variants of this code have been described in a wide range of nuclear and organellar systems, especially in metazoan mitochondria. These variants are generally found by searching for conserved positions that consistently code for a specific alternative amino acid in a new species. We have devised an accurate computational method to automate these comparisons, and have tested it with 626 metazoan mitochondrial genomes. Our results indicate that several arthropods have a new genetic code and translate the codon AGG as lysine instead of serine (as in the invertebrate mitochondrial genetic code) or arginine (as in the standard genetic code). We have investigated the evolution of the genetic code in the arthropods and found several events of parallel evolution in which the AGG codon was reassigned between serine and lysine. Our analyses also revealed correlated evolution between the arthropod genetic codes and the tRNA-Lys/-Ser, which show specific point mutations at the anticodons. These rather simple mutations, together with a low usage of the AGG codon, might explain the recurrence of the AGG reassignments.


Subject(s)
Arthropods/genetics , DNA, Mitochondrial/genetics , Evolution, Molecular , Genes, Mitochondrial/genetics , Genetic Code/genetics , Animals , Codon , Computer Simulation , Models, Genetic , Phylogeny , RNA, Transfer/genetics
11.
J Proteome Res ; 5(3): 709-19, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16512687

ABSTRACT

An important strategy for "shotgun proteomics" profiling involves solution proteolysis of proteins, followed by peptide separation using multidimensional liquid chromatography and automated sequencing by mass spectrometry (LC-MS/MS). Several protocols for extracting and handling membrane proteins for shotgun proteomics experiments have been reported, but few direct comparisons of different protocols have been reported. We compare four methods for preparing membrane proteins from human cells, using acid labile surfactants (ALS), urea, and mixed organic-aqueous solvents. These methods were compared with respect to their efficiency of protein solubilization and proteolysis, peptide and protein recovery, membrane protein enrichment, and peptide coverage of transmembrane proteins. Overall, approximately 50-60% of proteins recovered were membrane-associated, identified from Gene Ontology annotations and transmembrane prediction software. Samples extracted with ALS, extracted with urea followed by dilution, or extracted with urea followed by desalting yielded comparable peptide recoveries and sequence coverage of transmembrane proteins. In contrast, suboptimal proteolysis was observed with organic solvent. Urea extraction followed by desalting may be a particularly useful approach, as it is less costly than ALS and yields satisfactory protein denaturation and proteolysis under conditions that minimize reactivity with urea-derived cyanate. Spectral counting was used to compare datasets of proteins from membrane samples with those of soluble proteins from K562 cells, and to estimate fold differences in protein abundances. Proteins most highly abundant in the membrane samples showed enrichment of integral membrane protein identifications, consistent with their isolation by differential centrifugation.


Subject(s)
Cell Extracts/analysis , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Membrane Proteins/analysis , Neoplasm Proteins/chemistry , Chromatography, Liquid , Electrophoresis, Polyacrylamide Gel , Humans , K562 Cells , Neoplasm Proteins/analysis , Tandem Mass Spectrometry
12.
Proc Natl Acad Sci U S A ; 102(31): 11070-5, 2005 Aug 02.
Article in English | MEDLINE | ID: mdl-16033867

ABSTRACT

We have analyzed 5,088 bacterial 16S rRNA gene sequences from the distal intestinal (cecal) microbiota of genetically obese ob/ob mice, lean ob/+ and wild-type siblings, and their ob/+ mothers, all fed the same polysaccharide-rich diet. Although the majority of mouse gut species are unique, the mouse and human microbiota(s) are similar at the division (superkingdom) level, with Firmicutes and Bacteroidetes dominating. Microbial-community composition is inherited from mothers. However, compared with lean mice and regardless of kinship, ob/ob animals have a 50% reduction in the abundance of Bacteroidetes and a proportional increase in Firmicutes. These changes, which are division-wide, indicate that, in this model, obesity affects the diversity of the gut microbiota and suggest that intentional manipulation of community structure may be useful for regulating energy balance in obese individuals. The sequences reported in this paper have been deposited in the GenBank database [accession nos. DQ 014552--DQ 015671 (mothers) and AY 989911--AY 993908 (offspring)].


Subject(s)
Intestines/microbiology , Obesity/microbiology , Animals , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Base Sequence , Cyanobacteria/classification , Cyanobacteria/genetics , Cyanobacteria/isolation & purification , DNA/genetics , Ecosystem , Female , Genes, Bacterial , Humans , Mice , Mice, Inbred C57BL , Mice, Obese , Molecular Sequence Data , Obesity/etiology , Pregnancy , RNA, Ribosomal, 16S/genetics
13.
J Am Vet Med Assoc ; 226(12): 2039-43, 2002, 2005 Jun 15.
Article in English | MEDLINE | ID: mdl-15989188

ABSTRACT

Five horses were evaluated because of severe cutaneous burn injuries following a barn fire. Gross hemolysis and morphologic changes in RBCs consistent with oxidative damage were detected in all of the horses. Of these horses, 4 became azotemic. The overall goals of treatment included wound care, correction of dehydration and provision of diuresis, control of inflammation, pain management, and prophylaxis against sepsis. After treatment, 2 horses survived and were discharged from the hospital. Red blood cell damage and hemolysis following cutaneous burn injury have been investigated in other species and appear to be a result of the release of oxygen radicals from complement-activated neutrophils. Early intervention with aggressive fluid therapy is recommended in the treatment of human burn patients and is likely to be of benefit in horses with burn injuries; a beneficial role of free radical scavengers and xanthine oxidase inhibitors has also been suggested.


Subject(s)
Burns/veterinary , Hemolysis , Horse Diseases/etiology , Horses/injuries , Skin/injuries , Animals , Anti-Inflammatory Agents/therapeutic use , Burns/blood , Burns/complications , Burns/drug therapy , Horse Diseases/drug therapy , Male , Uremia/drug therapy , Uremia/etiology , Uremia/veterinary
14.
Anal Chem ; 76(13): 3556-68, 2004 Jul 01.
Article in English | MEDLINE | ID: mdl-15228325

ABSTRACT

Identifying proteins in cell extracts by shotgun proteomics involves digesting the proteins, sequencing the resulting peptides by data-dependent mass spectrometry (MS/MS), and searching protein databases to identify the proteins from which the peptides are derived. Manual analysis and direct spectral comparison reveal that scores from two commonly used search programs (Sequest and Mascot) validate less than half of potentially identifiable MS/MS spectra (class positive) from shotgun analyses of the human erythroleukemia K562 cell line. Here we demonstrate increased sensitivity and accuracy using a focused search strategy along with a peptide sequence validation script that does not rely exclusively on XCorr or Mowse scores generated by Sequest or Mascot, but uses consensus between the search programs, along with chemical properties and scores describing the nature of the fragmentation spectrum (ion score and RSP). The approach yielded 4.2% false positive and 8% false negative frequencies in peptide assignments. The protein profile is then assembled from peptide assignments using a novel peptide-centric protein nomenclature that more accurately reports protein variants that contain identical peptide sequences. An Isoform Resolver algorithm ensures that the protein count is not inflated by variants in the protein database, eliminating approximately 25% of redundant proteins. Analysis of soluble proteins from a human K562 cells identified 5130 unique proteins, with approximately 100 false positive protein assignments.


Subject(s)
Proteins/chemistry , Proteomics/methods , Cell Line, Tumor , Humans , K562 Cells , Mass Spectrometry/methods , Peptides/chemistry , Reproducibility of Results , Sensitivity and Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...