Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Int J Mol Sci ; 23(23)2022 Dec 01.
Article in English | MEDLINE | ID: mdl-36499391

ABSTRACT

The SH-SY5Y neuroblastoma cells are a widely used in vitro model approximating neurons for testing the target engagement of therapeutics designed for neurodegenerative diseases and pain disorders. However, their potential as a model for receptor-mediated delivery and uptake of novel modalities, such as antibody-drug conjugates, remains understudied. Investigation of the SH-SY5Y cell surfaceome will aid in greater in vitro to in vivo correlation of delivery and uptake, thereby accelerating drug discovery. So far, the majority of studies have focused on total cell proteomics from undifferentiated and differentiated SH-SY5Y cells. While some studies have investigated the expression of specific proteins in neuroblastoma tissue, a global approach for comparison of neuroblastoma cell surfaceome to the brain and dorsal root ganglion (DRG) neurons remains uninvestigated. Furthermore, an isoform-specific evaluation of cell surface proteins expressed on neuroblastoma cells remains unexplored. In this study, we define a bioinformatic workflow for the identification of high-confidence surface proteins expressed on brain and DRG neurons using tissue proteomic and transcriptomic data. We then delineate the SH-SY5Y cell surfaceome by surface proteomics and show that it significantly overlaps with the human brain and DRG neuronal surface proteome. We find that, for 32% of common surface proteins, SH-SY5Y-specific major isoforms are alternatively spliced, maintaining their protein-coding ability, and are predicted to localize to the cell surface. Validation of these isoforms using surface proteomics confirms a SH-SY5Y-specific alternative NRCAM (neuron-glia related cell adhesion molecule) isoform, which is absent in typical brain neurons, but present in neuroblastomas, making it a receptor of interest for neuroblastoma-specific therapeutics.


Subject(s)
Neuroblastoma , Humans , Neuroblastoma/therapy , Neuroblastoma/drug therapy , Cell Line, Tumor , Proteomics , Neurons/metabolism , Cell Differentiation/physiology , Membrane Proteins/metabolism
2.
MAbs ; 11(5): 861-869, 2019 07.
Article in English | MEDLINE | ID: mdl-31099718

ABSTRACT

Despite recent advances in the development of tools to predict immunogenicity risk of biotherapeutic molecules, the ability of a protein to elicit the formation of anti-drug antibodies (ADA) remains one of the most common causes for termination of clinical development programs. In this study, we use ADA assays to detect and measure pre-existing reactivity or the ability of a molecule to produce an ADA-like response in serum from treatment-naïve, healthy donors. We report herein that the magnitude of pre-existing reactivity evaluated pre-clinically and expressed as the 90th percentile of Tier 2 inhibition correlates with the subsequent rate of ADA emergence in the clinic. Furthermore, a multi-domain biotherapeutic (IgG-scFv bispecific antibody) showed the highest pre-existing reactivity and incidence of treatment-emergent ADA (TE-ADA) (57% and 93%, respectively). Using the components of the multidomain molecule in the Tier 2 step of the ADA assay, we were able to identify the scFv as the target of the serum pre-existing reactivity. Most importantly, the domain specificity of pre-existing ADA was the same as that of the TE-ADA from patients treated with the molecule. Based on these data, we propose the evaluation of the magnitude and of the domain specificity of pre-existing reactivity as a powerful tool to understand the immunogenic potential of novel biotherapeutics.


Subject(s)
Antibodies, Bispecific/immunology , Single-Chain Antibodies/immunology , Adult , Antibodies, Bispecific/adverse effects , Antibodies, Bispecific/blood , Antibody Formation , Biological Therapy/adverse effects , Epitopes/immunology , Female , Humans , Immune Sera/immunology , Male , Middle Aged , Risk , Single-Chain Antibodies/adverse effects , Single-Chain Antibodies/blood , Young Adult
3.
J Am Soc Mass Spectrom ; 25(7): 1228-36, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24845350

ABSTRACT

De novo sequencing by mass spectrometry (MS) allows for the determination of the complete amino acid (AA) sequence of a given protein based on the mass difference of detected ions from MS/MS fragmentation spectra. The technique relies on obtaining specific masses that can be attributed to characteristic theoretical masses of AAs. A major limitation of de novo sequencing by MS is the inability to distinguish between the isobaric residues leucine (Leu) and isoleucine (Ile). Incorrect identification of Ile as Leu or vice versa often results in loss of activity in recombinant antibodies. This functional ambiguity is commonly resolved with costly and time-consuming AA mutation and peptide sequencing experiments. Here, we describe a set of orthogonal biochemical protocols, which experimentally determine the identity of Ile or Leu residues in monoclonal antibodies (mAb) based on the selectivity that leucine aminopeptidase shows for n-terminal Leu residues and the cleavage preference for Leu by chymotrypsin. The resulting observations are combined with germline frequencies and incorporated into a logistic regression model, called Predictor for Xle Sites (PXleS) to provide a statistical likelihood for the identity of Leu at an ambiguous site. We demonstrate that PXleS can generate a probability for an Xle site in mAbs with 96% accuracy. The implementation of PXleS precludes the expression of several possible sequences and, therefore, reduces the overall time and resources required to go from spectra generation to a biologically active sequence for a mAb when an Ile or Leu residue is in question.


Subject(s)
Antibodies, Monoclonal/chemistry , Isoleucine/chemistry , Leucine/chemistry , Sequence Analysis, Protein/methods , Tandem Mass Spectrometry/methods , Amino Acid Sequence , Antibodies, Monoclonal/metabolism , Chymotrypsin/metabolism , Isoleucine/analysis , Leucine/analysis , Leucyl Aminopeptidase/metabolism , Molecular Sequence Data , Protein Aggregates
4.
Methods Mol Biol ; 988: 291-302, 2013.
Article in English | MEDLINE | ID: mdl-23475727

ABSTRACT

Epitope mapping of antibodies is the identification and characterization of binding sites of monoclonal antibodies (mAbs) on target antigens. This knowledge can be useful in generating novel antibodies to a particular target as well as elucidating an antibody mechanism of action. Several techniques are available to identify antibody epitopes among which are preliminary and simple ones like sequence homology analysis ELISA and Western blotting. However, the more widely used robust methods typically involve the use of mass spectrometry to fully analyze and interpret the data and accurately identify the binding site. Such methods include epitope extortion/excision, hydrogen deuterium exchange.


Subject(s)
Antibodies, Monoclonal/chemistry , Epitopes/chemistry , Animals , Biotinylation/methods , CHO Cells , Chromatography, High Pressure Liquid , Cricetinae , Deuterium Exchange Measurement , Epitope Mapping/methods , Epitopes/isolation & purification , Fas Ligand Protein/chemistry , Fas Ligand Protein/isolation & purification , Humans , Immobilized Proteins/chemistry , Immobilized Proteins/isolation & purification , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Proteolysis , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Solutions , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Trypsin/chemistry
5.
Biochemistry ; 48(30): 7251-60, 2009 Aug 04.
Article in English | MEDLINE | ID: mdl-19588926

ABSTRACT

Fas ligand (FasL) is a 40-kDa type II transmembrane protein belonging to the tumor necrosis factor (TNF) family of proteins and binds to its specific receptor, Fas, a member of the TNF receptor family. Membrane-bound FasL can be processed into a soluble form by a metalloprotease similar to that which cleaves TNFalpha. Elevated levels of FasL have been implicated in a wide variety of diseases ranging from cancer to inflammatory abnormalities, which could be targeted by antibody therapy. We generated a fully human high-affinity antibody against FasL that binds to and neutralizes the activity of both soluble and membrane-associated human FasL. In order to elucidate the mechanism of function of this antibody, we have mapped the region and critical residues involved in the recognition of FasL using a combination of homology modeling, immunoprecipitation, hydrogen-deuterium exchange mass spectrometry (H/DXMS), and alanine scanning site-directed mutagenesis. These studies have revealed the antibody binding site on human FasL. Furthermore, through molecular homology modeling, we have proposed a mechanism for the neutralizing activity of this antibody that involves interference with the docking of the ligand to its receptor by the antibody.


Subject(s)
Antibodies , Epitopes/chemistry , Fas Ligand Protein/chemistry , Fas Ligand Protein/immunology , Amino Acid Sequence , Animals , Antibodies/chemistry , Antibodies/immunology , Apoptosis/immunology , Binding Sites , Epitope Mapping , Epitopes/immunology , Fas Ligand Protein/genetics , Humans , Jurkat Cells , Mice , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Protein Binding , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Trypsin/metabolism
6.
J Neurosci ; 29(11): 3603-12, 2009 Mar 18.
Article in English | MEDLINE | ID: mdl-19295164

ABSTRACT

Recent studies suggest that bone marrow-derived macrophages can effectively reduce beta-amyloid (Abeta) deposition in brain. To further elucidate the mechanisms by which macrophages degrade Abeta, we cultured murine macrophages on top of Abeta plaque-bearing brain sections from transgenic mice expressing PDAPP [human amyloid precursor protein (APP) with the APP(717V>F) mutation driven by the platelet-derived growth factor promoter]. Using this ex vivo assay, we found that macrophages from wild-type mice very efficiently degrade both soluble and insoluble Abeta in a time-dependent manner and markedly eliminate thioflavine-S positive amyloid deposits. Because macrophages express and secrete apolipoprotein E (apoE), we compared the efficiency of Abeta degradation by macrophages prepared from apoE-deficient mice or mice expressing human apoE2, apoE3, or apoE4. Macrophages expressing apoE2 were more efficient at degrading Abeta than apoE3-expressing, apoE4-expressing, or apoE-deficient macrophages. Moreover, macrophage-induced degradation of Abeta was effectively blocked by an anti-apoE antibody and receptor-associated protein, an antagonist of the low-density lipoprotein (LDL) receptor family, suggesting involvement of LDL receptors. Measurement of matrix metalloproteinase-9 (MMP-9) activity in the media from human apoE-expressing macrophages cocultured with Abeta-containing brain sections revealed greater levels of MMP-9 activity in apoE2-expressing than in either apoE3- or apoE4-expressing macrophages. Differences in MMP-9 activity appear to contribute to the isoform-specific differences in Abeta degradation by macrophages. These apoE isoform-dependent effects of macrophages on Abeta degradation suggest a novel "peripheral" mechanism for Abeta clearance from brain that may also, in part, explain the isoform-dependent effects of apoE in determining the genetic risk for Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/metabolism , Amyloid/metabolism , Apolipoproteins E/physiology , Macrophages/physiology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Peptides/genetics , Amyloid beta-Protein Precursor/biosynthesis , Amyloid beta-Protein Precursor/genetics , Animals , Apolipoproteins E/genetics , Cells, Cultured , Coculture Techniques/methods , Humans , Macrophages/metabolism , Mice , Mice, Inbred C57BL , Mice, Transgenic , Protein Isoforms/genetics , Protein Isoforms/physiology
7.
Methods Mol Biol ; 425: 53-66, 2008.
Article in English | MEDLINE | ID: mdl-18369886

ABSTRACT

Cerebrospinal fluid (CSF) provides an important source of potential biomarkers for brain disorders and therapeutic drug development. Applications of proteomic technology to the identification and quantification of proteins in CSF are increasing rapidly. Key to obtaining reproducible and reliable data about protein levels in CSF are standardization of methods for sample collection, storage, and subsequent sample processing. Methods are described here for all steps of sample processing for a number of different proteomic approaches.


Subject(s)
Cerebrospinal Fluid/chemistry , Proteins/isolation & purification , Proteomics , Chromatography, Liquid , Electrophoresis, Gel, Two-Dimensional , Humans , Proteins/analysis , Reproducibility of Results , Specimen Handling , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Tandem Mass Spectrometry
8.
Methods Mol Biol ; 428: 209-30, 2008.
Article in English | MEDLINE | ID: mdl-18287776

ABSTRACT

Pharmaceutical companies and regulatory agencies are pursuing biomarkers as a means to increase the productivity of drug development. Quantifying differential levels of proteins from complex biological samples like plasma or cerebrospinal fluid is one specific approach being used to identify markers of drug action, efficacy, toxicity, etc. Academic investigators are also interested in markers that are diagnostic or prognostic of disease states. We report a comprehensive, fully automated, and label-free approach to relative protein quantification including: sample preparation, proteolytic protein digestion, LCMS/MS data acquisition, de-noising, mass and charge state estimation, chromatographic alignment, and peptide quantification via integration of extracted ion chromatograms. Additionally, we describe methods for transformation and normalization of the quantitative peptide levels in multiplexed measurements to improve precision for statistical analysis. Lastly, we outline how the described methods can be used to design and power biomarker discovery studies.


Subject(s)
Biomarkers/analysis , Chromatography, High Pressure Liquid/methods , Proteomics/methods , Tandem Mass Spectrometry/methods , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Data Interpretation, Statistical , Humans , Proteome/analysis , Proteomics/statistics & numerical data
9.
Brief Funct Genomic Proteomic ; 6(2): 149-58, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17584762

ABSTRACT

Immunoprecipitation (IP) combined with matrix-assisted laser desorption ionization (MALDI) time of flight (Tof) mass spectrometry has been used to develop quantitative assays for amyloid-beta (Abeta) peptides in cerebrospinal fluid (CSF). Inclusion of (15)N labelled standard peptides allows for absolute quantification of multiple Abeta isoforms in individual samples. Characterization of variability associated with all steps of the assay indicated that the IP step is the single largest contributor to overall variability. Optimization of the assay resulted in overall coefficient of variation

Subject(s)
Amyloid beta-Peptides/cerebrospinal fluid , Peptide Fragments/cerebrospinal fluid , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Alzheimer Disease/cerebrospinal fluid , Alzheimer Disease/metabolism , Biomarkers/analysis , Biomarkers/cerebrospinal fluid , Enzyme-Linked Immunosorbent Assay , Humans , Immunoprecipitation , Protein Isoforms/cerebrospinal fluid
10.
J Proteome Res ; 6(3): 955-66, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17243662

ABSTRACT

Recognizing specific protein changes in response to drug administration in humans has the potential for significant utility in clinical research. In spite of this, many methodological and practical questions related to assessing such changes are unanswered. We conducted a series of clinical studies to assess the feasibility of measuring changes in proteins related to drug administration using a mass-spectrometry proteomics technique capable of quantifying hundreds of proteins simultaneously in cerebrospinal fluid (CSF) and plasma. Initially, the normal variability of proteins in these compartments was characterized in 16 healthy volunteers over a 2-week period. Drug-associated changes were subsequently assessed in the plasma and CSF proteomes of 11 subjects given atomoxetine, which served as a selective, centrally active probe to test the model. Protein levels in the CSF and plasma were unchanged between visits in the normal variability study. In contrast, statistically significant changes were detected in the CSF protein pattern after drug treatment. These studies suggest that identification of changes in the CSF proteome associated with the administration of centrally active drugs is feasible, and may be of value in the development of new drugs, as well as broader clinical research.


Subject(s)
Blood Proteins/drug effects , Cerebrospinal Fluid Proteins/drug effects , Drug Monitoring/methods , Pharmacokinetics , Atomoxetine Hydrochloride , Biomarkers/analysis , Blood Proteins/analysis , Cerebrospinal Fluid Proteins/analysis , Humans , Pharmaceutical Preparations/administration & dosage , Pilot Projects , Propylamines/administration & dosage , Propylamines/pharmacokinetics , Proteomics/methods , Spectrum Analysis
11.
Rapid Commun Mass Spectrom ; 20(24): 3723-35, 2006.
Article in English | MEDLINE | ID: mdl-17117458

ABSTRACT

The 40 and 42 amino-acid residue forms of amyloid beta (Abeta(1-40) and Abeta(1-42)) in cerebrospinal fluid (CSF) have been proposed as potential biomarkers of Alzheimer's disease (AD). Quantitative analyses of Abeta peptides in CSF have relied almost exclusively on the use of immunoassay-based assays such as the enzyme-linked immunosorbent assay (ELISA) procedure. However, due to the ability of the Abeta peptides to readily self-aggregate or bind to other proteins and glassware, such analyses are extremely challenging. Analyses are further complicated by the potential of the peptides to undergo post-translational modifications and the possibilities for cross-reaction in the ELISA assays with endogenous components of the CSF. An approach based on liquid chromatography/tandem mass spectrometry (LC/MS/MS) has now been developed which overcomes these methodological issues. The key steps in implementing this new approach involved immunoaffinity purification coupled with the use of [15N]-labeled Abeta peptides as internal standards, a basic LC mobile phase, negative ion electrospray ionization, and a basic solvent for dissolving the peptides and washing the injection needle to prevent carryover of analytes during multiple injections on the LC/MS system. The validated method had limits of quantitation of 44 fmol/mL (200 pg/mL) for Abeta(1-42) and 92 fmol/mL (400 pg/mL) for Abeta(1-40). An excellent correlation was found between the LC/MS/MS assay and an ELISA assay for Abeta(1-42) in human CSF (r2 = 0.915), although less correlation was observed for Abeta(1-40) (r2 = 0.644). Mean CSF Abeta(1-42) concentrations for samples collected 2 weeks apart from a limited number of AD patients provided additional confidence in the reproducibility of the LC/MS/MS assay. Concentrations for duplicate samples from AD patients were slightly higher than most previously reported values (mean 1.06 +/- 0.25 ng/mL; n = 7). Abeta(1-40) concentrations in duplicate samples obtained from AD patients were also reproducible but were found to be slightly lower than most previously reported values (mean 6.36 +/- 3.07 ng/mL; n = 7). Consistent with literature reports, mean Abeta(1-42) concentrations were found to be lower in AD patients compared with the normal subjects (mean 1.49 +/- 0.59 ng/mL; n = 7), whereas there was no difference in Abeta(1-40) concentrations between AD patients and normal subjects (mean 5.88 +/- 3.03 ng/mL; n = 7). The accuracy and precision of the LC/MS assay mean that it will be a useful complement to existing ELISA assays for monitoring therapeutic interventions designed to modulate CSF Abeta(1-42) concentrations in individual AD patients. Moreover, the introduction of stable isotope labeled internal standards offers the potential to achieve a more rigorous account of the influence of methodological effects related to sample collection and processing.


Subject(s)
Alzheimer Disease/cerebrospinal fluid , Amyloid beta-Peptides/cerebrospinal fluid , Chromatography, Liquid/methods , Immunoassay/methods , Peptide Fragments/cerebrospinal fluid , Spectrometry, Mass, Electrospray Ionization/methods , Biomarkers/cerebrospinal fluid , Humans , Radioisotope Dilution Technique , Reproducibility of Results , Sensitivity and Specificity
12.
J Proteome Res ; 4(4): 1442-50, 2005.
Article in English | MEDLINE | ID: mdl-16083298

ABSTRACT

Pharmaceutical companies and regulatory agencies are broadly pursuing biomarkers as a means to increase the productivity of drug development. Quantifying differential levels of proteins from complex biological samples such as plasma or cerebrospinal fluid is one specific approach being used to identify markers of drug action, efficacy, toxicity, etc. We have developed a comprehensive, fully automated, and label-free approach to relative protein quantification from LC-MS/MS experiments of proteolytic protein digests including: de-noising, mass and charge state estimation, chromatographic alignment, and peptide quantification via integration of extracted ion chromatograms. Results from a variance components study of the entire method indicate that most of the variability is attributable to the LC-MS injection, with a median peptide LC-MS injection coefficient of variation of 8% on a ThermoFinnigan LTQ mass spectrometer. Spiked recovery results suggest a quantifiable range of approximately 32-fold for a sample protein.


Subject(s)
Chromatography, Liquid/methods , Mass Spectrometry/methods , Proteins/analysis , Amino Acid Sequence , Animals , Biomarkers , Male , Molecular Sequence Data , Rats , Reproducibility of Results , Serum/chemistry , Trypsin Inhibitor, Kunitz Soybean/analysis
13.
Proteomics ; 5(1): 290-6, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15672452

ABSTRACT

Human cerebrospinal fluid (CSF) is in direct contact with the brain extracellular space. Beside the secretion of CSF by the choroid plexus the fluid also derives directly from the brain by the ependymal lining of the ventricular system and the glial membrane and from blood vessels in the arachnoid. Therefore, biochemical change in the brain may be reflected in the CSF. CSF is a potential source of protein molecular indices of central nervous system function and pathology. However, various amounts of blood contamination in CSF may arise during sample acquisition. The concentration of protein in the CSF is only 0.2 to 0.5% that of blood. Minor contamination of CSF with blood during collection of the fluid may dramatically alter the protein profile confounding the identification of potential biomarkers. We have analyzed CSF and CSF spiked with increasing amounts of whole blood using proteomic techniques. We detected at least four blood specific highly abundant proteins: hemoglobin, catalase, peroxiredoxin and carbonic anhydrase I. These proteins can be used as blood contamination markers for proteomic analysis of CSF. Proteins in blood contaminated CSF samples were less stable compared to neat CSF at 37 degrees C suggesting that blood borne protease may induce protein degradation in CSF during sample acquisition. This analysis was aimed at identification of proteins found primarily in CSF, those found primarily in blood and assessment of the impact of blood contamination on those proteins found in both fluids.


Subject(s)
Blood Proteins/chemistry , Cerebrospinal Fluid Proteins/chemistry , Proteome/analysis , Amino Acid Sequence , Biomarkers/cerebrospinal fluid , Blood Proteins/cerebrospinal fluid , Electrophoresis, Gel, Two-Dimensional , Humans , Molecular Sequence Data , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
14.
Biotechniques ; Suppl: 13-7, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16528911

ABSTRACT

A method is described for the quantitative determination of peptides using matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) mass spectrometry. Known limitations imposed by crystal heterogeneity, peptide ionization differences, data handling, and protein quantification with MALDI-TOF mass spectrometry are addressed in this method with a "seed crystal" protocol for analyte-matrix formation, the use of internal protein standards, and a software package called maldi_quant. The seed crystal protocol, a new variation of the fast-evaporation method, minimizes crystal heterogeneity and allows for consistent collection of protein spectra. The software maldi_quant permits rapid and automated analysis of peak intensity data, normalization of peak intensities to internal standards, and peak intensity deconvolution and estimation for vicinal peaks. Using insulin proteins in a background of other unrelated peptides, this method shows an overall coefficient of variance of 4.4%, and a quantitative working range of 0.58-37.5 ng bovine insulin per spot. Coupling of this methodology to powerful analytical procedures such as immunoprecipitation is likely to lead to the rapid and reliable quantification of biologically relevant proteins and their closely related variants.


Subject(s)
Peptides/analysis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Animals , Cattle , Insulin/analysis , Reference Standards , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/standards
15.
Anal Biochem ; 333(1): 174-81, 2004 Oct 01.
Article in English | MEDLINE | ID: mdl-15351294

ABSTRACT

A procedure for reduction and alkylation of cysteine residues in proteins was developed using the volatile reagents triethylphosphine and iodoethanol. These reagents may be used to modify proteins in solution, as well as proteins in gel slices, prior to proteolytic digestion and mass spectral analysis. The procedure eliminates several steps with both types of samples. Samples in solution do not need to be desalted following reduction and alkylation, with excess reagent being removed under vacuum. For gel slices, the procedure combines washing, destaining, reduction and alkylation into a single step. The procedure was applied successfully to samples as complex as serum, and we demonstrated alkylation of cysteines to be quantitative in purified proteins. We also were able to reduce and alkylate proteins with these reagents during the gas phase. Elimination of the need for desalting of samples after reaction raised the possibility of automation of the procedure for liquid samples, which is difficult with conventional reduction and alkylation chemistries.


Subject(s)
Cysteine/chemistry , Muramidase/chemistry , Serum/chemistry , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Alkylating Agents/chemistry , Alkylation , Animals , Chickens , Electrophoresis, Gel, Two-Dimensional , Ethanol/analogs & derivatives , Ethanol/chemistry , Oxidation-Reduction , Peptide Mapping/methods , Phosphines/chemistry , Rats
16.
Science ; 302(5648): 1215-7, 2003 Nov 14.
Article in English | MEDLINE | ID: mdl-14615541

ABSTRACT

A subset of nonsteroidal anti-inflammatory drugs (NSAIDs) has been shown to preferentially reduce the secretion of the highly amyloidogenic, 42-residue amyloid-beta peptide Abeta42. We found that Rho and its effector, Rho-associated kinase, preferentially regulated the amount of Abeta42 produced in vitro and that only those NSAIDs effective as Rho inhibitors lowered Abeta42. Administration of Y-27632, a selective Rock inhibitor, also preferentially lowered brain levels of Abeta42 in a transgenic mouse model of Alzheimer's disease. Thus, the Rho-Rock pathway may regulate amyloid precursor protein processing, and a subset of NSAIDs can reduce Abeta42 through inhibition of Rho activity.


Subject(s)
Amyloid beta-Peptides/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Peptide Fragments/metabolism , Sulindac/analogs & derivatives , rho GTP-Binding Proteins/antagonists & inhibitors , Amides/pharmacology , Amyloid Precursor Protein Secretases , Animals , Aspartic Acid Endopeptidases , Brain/drug effects , Brain/metabolism , Cell Line, Tumor , Endopeptidases/metabolism , Enzyme Inhibitors/pharmacology , Guanosine Triphosphate/metabolism , Humans , Ibuprofen/pharmacology , Intracellular Signaling Peptides and Proteins , Mice , Mice, Transgenic , Polyisoprenyl Phosphates/pharmacology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Protein Serine-Threonine Kinases/metabolism , Pyridines/pharmacology , Sesquiterpenes , Signal Transduction , Sulindac/pharmacology , Transfection , rho GTP-Binding Proteins/metabolism , rho-Associated Kinases , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
17.
Brief Funct Genomic Proteomic ; 2(3): 185-93, 2003 Oct.
Article in English | MEDLINE | ID: mdl-15239922

ABSTRACT

Biomarkers of drug efficacy and toxicity are becoming a key need in the drug development process. Mass spectral-based proteomic technologies are ideally suited for the discovery of protein biomarkers in the absence of any prior knowledge of quantitative changes in protein levels. The success of any biomarker discovery effort will depend upon the quality of samples analysed, the ability to generate quantitative information on relative protein levels and the ability to readily interpret the data generated. This review will focus on the strengths and weaknesses of technologies currently utilised to address these issues.


Subject(s)
Biomarkers/analysis , Proteomics/methods , Animals , Drug Design , Humans , Mass Spectrometry
SELECTION OF CITATIONS
SEARCH DETAIL
...