Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
J Cereb Blood Flow Metab ; 36(7): 1281-94, 2016 07.
Article in English | MEDLINE | ID: mdl-26661166

ABSTRACT

Endothelial cells of the blood-brain barrier form a structural and functional barrier maintaining brain homeostasis via paracellular tight junctions and specific transporters such as P-glycoprotein. The blood-brain barrier is responsible for negligible bioavailability of many neuroprotective drugs. In Alzheimer's disease, current treatment approaches include inhibitors of BACE-1 (ß-site of amyloid precursor protein cleaving enzyme), a proteinase generating neurotoxic ß-amyloid. It is known that BACE-1 is highly expressed in endosomes and membranes of neurons and glia. We now provide evidence that BACE-1 is expressed in blood-brain barrier endothelial cells of human, mouse, and bovine origin. We further show its predominant membrane localization by 3D-dSTORM super-resolution microscopy, and by biochemical fractionation that further shows an abluminal distribution of BACE-1 in brain microvessels. We confirm its functionality in processing APP in primary mouse brain endothelial cells. In an Alzheimer's disease mouse model we show that BACE-1 is upregulated at the blood-brain barrier compared to healthy controls. We therefore suggest a critical role for BACE-1 at the blood-brain barrier in ß-amyloid generation and in vascular aspects of Alzheimer's disease, particularly in the development of cerebral amyloid angiopathy.


Subject(s)
Alzheimer Disease/metabolism , Amyloid Precursor Protein Secretases/genetics , Aspartic Acid Endopeptidases/genetics , Blood-Brain Barrier/metabolism , Endothelium, Vascular/metabolism , Amyloid beta-Protein Precursor/genetics , Animals , Blotting, Western , Cattle , Cells, Cultured , Disease Models, Animal , Endothelial Cells/metabolism , Humans , Immunohistochemistry , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/metabolism , Primary Cell Culture , Real-Time Polymerase Chain Reaction
2.
Clin Biochem ; 47(10-11): 901-8, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24954692

ABSTRACT

OBJECTIVES: Annexin A3 (ANXA3) is a potential marker for prostate cancer (PCa). We aimed to develop robust immunoassays suitable for quantifying ANXA3 in urine samples obtained following digital rectal examination (DRE) in order to facilitate the diagnostic performance evaluation of this marker. DESIGN AND METHODS: Anti-ANXA3 monoclonal antibodies were generated and their epitopes mapped. Two different ANXA3 assay prototypes were established on the VIDAS® automated immunoanalyser and analytical validation was carried out using post-DRE urine samples obtained from patients with PCa (n=23) or benign prostate hyperplasia (n=31). RESULTS: The assays had the same capture antibody (TGC44) but different detection antibodies (13A12 or 5C5), recognizing novel distinct epitopes. Both had a lower limit of quantification <1ng/mL and were highly specific for ANXA3, not cross-reacting with other annexins. Interassay imprecision was ≤11% and ≤15% for 13A12 and 5C5 assays, respectively. Surprisingly, a total lack of correlation was observed between ANXA3 levels measured by these two assays in post-DRE urines, indicating detection of distinct antigenic variants. Two freeze-thaw cycles did not affect analyte stability in either assay, whereas a lack of stability of antigenic variants was observed when samples were stored at -80°C for 1month. CONCLUSIONS: Two different antigenic variants of ANXA3 are present in post-DRE urines and their clinical significance for diagnosis of prostate cancer should be further investigated. These variants are not stable over time in samples preserved at -80°C. Until this issue is resolved, ANXA3 should only be measured in freshly collected samples.


Subject(s)
Annexin A3/urine , Biomarkers, Tumor/urine , Digital Rectal Examination , Neoplasm Proteins/urine , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/urine , Animals , Antibodies, Monoclonal, Murine-Derived/chemistry , Antibodies, Neoplasm/chemistry , Cryopreservation , Enzyme-Linked Immunosorbent Assay , Epitopes/urine , Humans , Male , Mice , Mice, Inbred BALB C , Protein Stability
3.
Mutat Res ; 746(2): 113-23, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22405942

ABSTRACT

The vision of the toxicology in the 21st century movement is to overcome the currently used animal tests and identify molecular pathways of toxicity, using human in vitro systems with the aim to provide the most relevant mechanistic information for human risk assessment. It is expected to translate key surrogate biomarkers to novel types of toxicity-related high throughput screening of the many thousands of compounds which need to be tested during development phases of the pharmaceutical industry and with regard to the REACH legislation in Europe. Systems biology, an emerging and increasingly popular field of research, appears to be the discipline of choice to integrate results from transcriptomics, proteomics, epigenomics and metabonomics technologies used to analyze samples from toxicological models. The challenges, however, with respect to data generation, statistical treatment, bioinformatic integration and interpretation or in silico modeling remain formidable. One of the main difficulties is the fact that the sheer number of molecular species is inflated enormously in the course of translation from genes to proteins due to post-translational modifications. Moreover, at the level of proteins, time scales of cellular reactions to toxic insults can be very fast, ranging from milliseconds to seconds. Linear dynamic ranges of concentration differences between conditions can also differ by several orders of magnitude. So, the search for protein biomarkers of toxicity requires sophisticated strategies for time-resolved quantitative differential approaches. The statistical principles, normalization of primary data and principal component and cluster analysis have been well developed for genomics/transcriptomics and partly for proteomics, but have not been widely adapted to technologies like metabonomics. Also, the integration of functional data, in particular data from mass spectrometry, with the aim of modeling pathways of toxicity for human risk assessment, is still at an infant stage.


Subject(s)
Biomarkers/analysis , Proteins/analysis , Proteomics/methods , Toxicity Tests/methods , Animal Testing Alternatives/methods , Animals , Computational Biology , Developmental Biology/methods , Embryonic Stem Cells , Epigenesis, Genetic , Humans , Metabolomics , Neoplasms/chemistry , Proteomics/classification , Systems Biology , Toxicology/methods , Transcriptome , Validation Studies as Topic
4.
J Proteome Res ; 9(11): 5727-38, 2010 Nov 05.
Article in English | MEDLINE | ID: mdl-20919680

ABSTRACT

There are new challenges for hazard and risk assessment in the chemical industry with regard to REACH legislation in Europe and related activities in the U.S. and Japan, which require the development of novel in vitro models for the molecular characterization of drug- or chemical-related effects replacing conventional animal testing. In the frame of a European FP6 project on reproductive toxicology ( www.reprotect.eu ), we prepared protein samples from mouse embryonic stem cells differentiated into contracting cardiomyocytes according to the validated embryonic stem cell test (EST) protocol, which had been exposed to toxic substances selected by an expert committee from different in vivo categories of embryotoxicity. Lysates were used to carry out the following investigations: (i) identify optimal dose range conditions in the EST that are suitable for (ii) performing a differential quantitative proteomic study of underlying molecular pathways, (iii) define classes of substances with similar proteomic response patterns, (iv) relate these classes to the traditional in vivo categories of embryotoxicity with (v) the final goal to identify novel surrogate protein biomarker candidates for embryo toxicity. We found two distinct classes of toxic substances (Dinoseb, Ochratoxin-A, and Nitrofen vs ß-aminoproprionitril, Metoclopramide, Doxylamine succinate, and d-penicillamine) with clear pathway-related differences in their proteomic patterns. Most notably, different responses to cluster 1 and cluster 2 substances were observed for Heat shock protein ß-1, Ras-GTPase-activating protein SH3-domain binding protein, Ran binding protein 5, and Calreticulin, Dihydropyrimidinase-like 2 (Ulip2 protein). On the other hand, Heat shock protein 8 and Fscn1 protein were down-regulated by all compounds from both clusters.


Subject(s)
Animal Testing Alternatives/methods , Drug-Related Side Effects and Adverse Reactions , Embryonic Stem Cells/drug effects , Proteins/drug effects , Toxicity Tests , Animals , Biomarkers , Cell Differentiation , Cells, Cultured , Mice , Myocytes, Cardiac
5.
Reprod Toxicol ; 30(1): 121-30, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20493250

ABSTRACT

Novel molecular content for fast in vitro strategies in the context of safety tests concerning developmental toxicity has a potential to substantially reduce animal experiments according to the "3R" concept (Reduce/Refine/Replace). Here we present and discuss data from a differential proteomic profiling of samples generated using embryonic stem cell derived in vitro models treated with a set of model substances. Among substance-dependent proteomic changes, potential surrogate markers were some isoforms of heat shock proteins and a component of the Ras pathway, present in several redundant isoforms due to posttranslational modifications. Both proteins are implicated in cell migration, cell survival, growth and embryonic development. Using the examples of warfarin and lovastatin, two substances with entirely different primary targets, the surrogate marker signature nevertheless indicates a common embryotoxic mode of action. We discuss these findings observed in in vitro toxicity tests, in a context of clinical validation and evidence-based toxicology.


Subject(s)
Animal Testing Alternatives , Embryonic Stem Cells/drug effects , Lovastatin/toxicity , Teratogens/toxicity , Toxicity Tests/methods , Warfarin/toxicity , 3T3 Cells , Animals , Cell Differentiation/drug effects , Cell Survival/drug effects , Electrophoresis, Polyacrylamide Gel , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Endpoint Determination , Heat-Shock Proteins/biosynthesis , Inhibitory Concentration 50 , Mice , Myocytes, Cardiac/cytology , Myocytes, Cardiac/drug effects , Reproducibility of Results , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Toxicity Tests/standards , ras Proteins/biosynthesis
6.
J Urol ; 181(1): 343-53, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19012935

ABSTRACT

PURPOSE: In prostate cancer cases the early diagnosis of tumors carrying a high risk of progression is of the utmost importance. There is an urgent clinical need to avoid unnecessary biopsies and subsequent overtreatment. We validated annexin A3 as a diagnostic marker for prostatic disease in typical clinical populations and relevant segments, such as patients with a negative digital rectal examination and low prostate specific antigen. MATERIALS AND METHODS: We performed a blinded clinical study (ClinicalTrials.gov Identifier NCT00400894) from September 2005 to January 2007 in 591 patients who were continuously recruited from 4 European urological clinics. Urine was obtained directly after digital rectal examination and the annexin A3 concentration in urine was quantified by Western blot. Statistical analysis included combinations of annexin A3 with total, percent free, complexed and percent complexed prostate specific antigen. RESULTS: Combined readouts of prostate specific antigen and urinary annexin A3 were superior to all others with an area under the ROC curve of 0.82 for a total prostate specific antigen range of 2 to 6 ng/ml, 0.83 for a total prostate specific antigen range of 4 to 10 ng/ml and 0.81 in all patients. The best performing prostate specific antigen derivative was percent free prostate specific antigen with an area under the ROC curve of 0.68 for a total prostate specific antigen range of 2 to 6 ng/ml, 0.72 for a total prostate specific antigen range of 4 to 10 ng/ml and 0.73 in all patients. Annexin A3 has an inverse relationship to cancer and, therefore, its specificity was much better than that of prostate specific antigen. CONCLUSIONS: Annexin A3 quantification in urine provides a novel noninvasive biomarker with high specificity. Annexin A3 is complementary to prostate specific antigen or to any other cancer marker. It has a huge potential to avoid unnecessary biopsies with a particular strength in the clinically relevant large group of patients who have a negative digital rectal examination and prostate specific antigen in the lower range of values (2 to 10 ng/ml).


Subject(s)
Annexin A3/urine , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/urine , Biomarkers/urine , Early Detection of Cancer , Humans , Male , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Sensitivity and Specificity
7.
Breast Cancer Res ; 10(5): R85, 2008.
Article in English | MEDLINE | ID: mdl-18922159

ABSTRACT

INTRODUCTION: Breast tumors lacking the estrogen receptor-alpha (ER-alpha) have increased incidence of resistance to therapy and poorer clinical prognosis. METHODS: Whole tissue sections from 16 cryopreserved breast cancer tumors that were either positive or negative for the ER (eight ER positive and eight ER negative) were differentially analyzed by multiplex imaging of two-dimensional PAGE gels using 54 cm isoelectric focusing. Differentially detected spots of Progesterone Receptor Membrane Component 1 (PGRMC1) were shown to differ in phosphorylation status by differential two dimensional polyacrylamide gel electrophoresis of phosphatase-treated tumor proteins. Site directed mutagenesis was used to create putative phosphorylation site point mutants in PGRMC1. Stable transfectants of these mutants in MCF7 cells were assayed for their survival after oxidative stress, and for AKT kinase phosphorylation. Immune fluorescence using anti-PGRMC1 monoclonal antibody 5G7 was performed on breast cancer tissue microarrays. RESULTS: Proteins significantly differentially abundant between estrogen receptor negative and estrogen receptor positive tumors at the 0.1% level were consistent with published profiles, suggesting an altered keratin pool, and increased inflammation and wound responses in estrogen receptor negative tumors. Two of three spots of PGRMC1 were more abundant in estrogen receptor negative tumors. Phosphatase treatment of breast tumor proteins indicated that the PGRMC1 isoforms differed in their phosphorylation status. Simultaneous mutation of PGRMC1 serine-56 and serine-180 [corrected] fully abrogated the sensitivity of stably transfected MCF7 breast cancer cells to peroxide-induced cell death. Immune fluorescence revealed that PGRMC1 was primarily expressed in ER-negative basal epithelial cells of mammary ductules. Even in advanced tumors, high levels of ER or PGRMC1 were almost mutually exclusive in individual cells. In five out of five examined ductal in situ breast cancers of comedo type, PGRMC1 was expressed in glucose transporter 1 negative or positive poorly oxygenated cells surrounding the necrotic core, surrounded by a more distal halo of ER-positive cells. CONCLUSIONS: PGRMC1 phosphorylation may be involved in the clinical differences that underpin breast tumors of differing ER status.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Lobular/metabolism , Estrogen Receptor alpha/metabolism , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Protein Processing, Post-Translational , Proteomics , Receptors, Progesterone/metabolism , Amino Acid Substitution , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/pathology , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Estrogens , Female , Humans , Membrane Proteins/genetics , Neoplasm Proteins/genetics , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Phosphorylation , Phosphoserine/metabolism , Protein Isoforms/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Progesterone/genetics , Recombinant Fusion Proteins/metabolism , Wound Healing/genetics
8.
J Proteome Res ; 7(6): 2262-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18465891

ABSTRACT

In a drug reprofiling attempt, we explored novel neuroprotective properties of 4-azasteroids by synthesizing chemical affinity tags capturing adenine nucleotide translocator-1, as a potential target. Dutasteride inhibits the mitochondrial transition pore and induces an increase of autophagosomal structures in human cell lines. In vivo, a surprising reduction of the beta-amyloid plaque load in a model for cerebral amyloidosis appears to connect release of neurotoxic peptides, mitochondrial apoptosis and autophagy.


Subject(s)
Amyloidosis/drug therapy , Autophagy/drug effects , Azasteroids/pharmacology , Brain Diseases/drug therapy , Mitochondrial Membrane Transport Proteins/metabolism , Amyloid beta-Peptides/pharmacology , Amyloid beta-Protein Precursor/genetics , Amyloidosis/metabolism , Amyloidosis/pathology , Animals , Apoptosis/drug effects , Brain Diseases/metabolism , Brain Diseases/pathology , Disease Models, Animal , Dutasteride , Enzyme Inhibitors/pharmacology , Finasteride/pharmacology , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mitochondrial ADP, ATP Translocases/metabolism , Mitochondrial Permeability Transition Pore , N-Methylaspartate/pharmacology , Neocortex/drug effects , Neocortex/metabolism , Neocortex/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neuroprotective Agents/pharmacology , Plaque, Amyloid/drug effects , Plaque, Amyloid/metabolism , Plaque, Amyloid/pathology , Potassium Cyanide/pharmacology , Presenilins/genetics , Sex Factors , Testosterone/pharmacology
9.
Exp Gerontol ; 42(9): 887-98, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17689904

ABSTRACT

According to the 'free radical theory of ageing', the generation and accumulation of reactive oxygen species are key events during ageing of biological systems. Mitochondria are a major source of ROS and prominent targets for ROS-induced damage. Whereas mitochondrial DNA and membranes were shown to be oxidatively modified with ageing, mitochondrial protein oxidation is not well understood. The purpose of this study was an unbiased investigation of age-related changes in mitochondrial proteins and the molecular pathways by which ROS-induced protein oxidation may disturb cellular homeostasis. In a differential comparison of mitochondrial proteins from young and senescent strains of the fungal ageing model Podospora anserina, from brains of young (5 months) vs. older rats (17 and 31 months), and human cells, with normal and chemically accelerated in vitro ageing, we found certain redundant posttranslationally modified isoforms of subunits of ATP synthase affected across all three species. These appear to represent general susceptible hot spot targets for oxidative chemical changes of proteins accumulating during ageing, and potentially initiating various age-related pathologies and processes. This type of modification is discussed using the example of SAM-dependent O-methyltransferase from P. anserina (PaMTH1), which surprisingly was found to be enriched in mitochondrial preparations of senescent cultures.


Subject(s)
Aging/physiology , Mitochondria/chemistry , Mitochondrial Proton-Translocating ATPases/analysis , Protein Isoforms/analysis , Proteome , Animals , Cells, Cultured , Electrophoresis, Gel, Two-Dimensional , Fungal Proteins/analysis , Humans , Methyltransferases/analysis , Models, Biological , Oxidative Stress , Podospora/physiology , Protein Processing, Post-Translational , Rats , Reactive Oxygen Species/metabolism , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
10.
Proteomics ; 7(2): 313-22, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17205602

ABSTRACT

A differential quantitative protein expression study, comparing matched prostate cancerous and benign tissues from 31 patients, revealed proteins newly associated with prostate cancer. Average effects for 17 proteins whose abundance was significantly different (p<0.01) across patients ranged from 1.5- to 6.1-fold, and included a number of known cancer markers. The most differentially abundant proteins between cancer and benign samples were isopeptidase T, serum amyloid P (SAP), annexin A3 (ANXA3) and mitochondrial enoyl coenzyme-A hydratase. SAP is restricted to stroma in healthy tissue, and the lower abundance in tumours may be explained by the reduced stromal content. ANXA3 is present in healthy epithelial cells, exhibits strong staining in precancerous prostatic intraepithelial neoplasia, and is relatively less abundant in individual tumour cells of increasing Gleason pattern (GP), despite exhibiting higher overall tissue abundance in tumours. ANXA3 staining was predominantly cytoplasmic, yet nuclear localization was also observed. Strongly staining single cells, possibly phagocytes, were interspersed in highly dedifferentiated GP5 tumour areas among tumour cells without measurable ANXA3. Local recurrent androgen ablation therapy-resistant tumours exhibit heterogenous low levels of ANXA3 staining. Results are discussed focussing on the potential implications for tumour tissues.


Subject(s)
Annexin A3/metabolism , Biomarkers, Tumor , Prostatic Hyperplasia/metabolism , Prostatic Neoplasms/metabolism , Radioisotopes , Adult , Aged , Diagnosis, Differential , Humans , Male , Middle Aged , Prostatic Hyperplasia/diagnosis , Prostatic Neoplasms/diagnosis
11.
J Proteome Res ; 5(12): 3453-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17137348

ABSTRACT

Serum and plasma are the major sources of human material for clinical molecular diagnostics and drug discovery. However, due to the high abundance of some proteins, of which serum albumin (SA) is most prominent, lower-abundance proteins often remain undetectable in proteomic analysis of these body fluids. We have used hexadecanedionic acid (HDA) immobilized to Sepharose 4B to develop an affinity resin that is effective in the removal of SA from plasma. Two-dimensional gel analysis of the SA-depleted samples shows a significant enhancement of the low-abundance proteins and highly specific capture of serum albumin. The HDA resin shows better performance in terms of specificity than dye-based resins.


Subject(s)
Affinity Labels/chemistry , Palmitic Acids , Sepharose , Serum Albumin/chemistry , Serum/chemistry , Computational Biology , Electrophoresis, Gel, Two-Dimensional , Humans , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
12.
J Chromatogr A ; 1123(2): 170-81, 2006 Aug 11.
Article in English | MEDLINE | ID: mdl-16822517

ABSTRACT

In this article we evaluate methods used to reveal the molecular complexity, which is generated in biological samples by posttranslational modifications (PTM) of proteins. We show how distinct molecular differences on the level of phosphorylation sites in a single protein (ovalbumin) can be resolved with different success using 1D and 2D gel-electrophoresis and reversed-phase liquid chromatography (LC) with monolithic polystyrol-divinylbenzol (PS-DVB) columns for protein separation, and matrix-assisted laser desorption ionisation-time of flight mass spectrometry (MALDI-TOF MS) for protein identification. Phosphorylation site analysis was performed using enzymatic dephosphorylation in combination with differential peptide mass mapping. Liquid chromatography-MALDI-TOF MS coupling with subsequent on-target tryptic protein digestion turned out to be the fastest method tested but yielded low resolution for the analysis of PTM, whereas 2D gel-electrophoresis, due to its unique capability of resolving highly complex isoform pattern, turned out to be the most suitable method for this purpose. The evaluated methods complement one another and in connection with efficient technologies for differential and quantitative analysis, these approaches have the potential to reveal novel molecular details of protein biomarkers.


Subject(s)
Biomarkers/analysis , Ovalbumin/isolation & purification , Protein Isoforms/isolation & purification , Protein Processing, Post-Translational , Proteome/analysis , Amino Acid Sequence , Chromatography, High Pressure Liquid/methods , Chromatography, Liquid/methods , Electrophoresis, Gel, Two-Dimensional/methods , Electrophoresis, Polyacrylamide Gel/methods , Isoelectric Focusing/methods , Molecular Sequence Data , Ovalbumin/metabolism , Phosphorylation , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods , Trypsin/metabolism
13.
Electrophoresis ; 27(9): 1840-52, 2006 May.
Article in English | MEDLINE | ID: mdl-16645950

ABSTRACT

The presence of progesterone receptor (PR) in estrogen receptor (ER)-positive breast cancer is associated with a good prognosis, and indicates that tumors are likely to respond to tamoxifen. However, ER+/PR- tumors respond less well. To reveal the potential molecular mechanism of this phenomenon, we sought to identify differential protein abundances between invasive ductal carcinoma cells from cryopreserved ER+/PR+ and ER+/PR- mammary tumor specimens. Because current proteomics methods are hampered in the examination of most primary human tumor samples by the extreme tissue heterogeneity, we used laser capture microdissection (LCM) to isolate tumor cells and developed a sample pooling strategy to analyze small sample protein lysates. Proteins from LCM-harvested tumors were pooled into four sub-pools from each condition of three tumors/sub-pool, and proteins from respective paired sub-pools were co-electrophoresed by 2-DE using 54-cm IEF over pH 4-9. Abundance ratios were accurately quantified by a differential multiplex radioactive ProteoTope method at low attomole levels ( approximately 3.6 microg protein per labeling reaction, <180 ng per multiplex protein sample per 54-cm gel). Applying this approach, differentially displayed proteins were identified by MS using comigrating non-radioactively labeled tumor proteins. They include decreased cytochrome b5 and transgelin, and more abundant CRABP-II, cyclophilin A, Neudesin, and hemoglobin in ER+/PR+ tumors versus ER+/PR- providing a possible explanation for differential susceptibility against tamoxifen as a result of deregulated cytochrome b5-dependent metabolism. This study demonstrates the potential of ProteoTope and LCM to enable extremely sensitive and precise differential analyses from well-defined primary clinical specimen.


Subject(s)
Breast Neoplasms/chemistry , Electrophoresis, Gel, Two-Dimensional/methods , Lasers , Microdissection/methods , Neoplasm Proteins/analysis , Proteomics/methods , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Cell Extracts/chemistry , Cryopreservation , Drug Resistance, Neoplasm , Female , Humans , Hydrogen-Ion Concentration , Iodine Radioisotopes/analysis , Mutation , Receptors, Estrogen/genetics , Receptors, Progesterone/genetics , Tamoxifen/therapeutic use
14.
J Proteome Res ; 5(3): 625-33, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16512678

ABSTRACT

The activity of mitochondria induces, as a byproduct, a variety of post-translational modifications in associated proteins, which have functional downstream consequences for processes such as apoptosis, autophagy, and plasticity; e.g., reactive oxygen species (ROS), which induce N-formyl-kynurenine from oxidized tryptophans in certain mitochondrial proteins which are localized in close spatial proximity to their source. This type of fast molecular changes has profound influence on cell death and survival with implications in a number of pathologies. The quantitative and differential analysis of bovine heart mitochondria by four 2D-PAGE methods, including 2D-PAGE with high-resolution IEF as first dimension, revealed that due to limited resolution, those methods employing blue native-, tricine-urea-, and 16-BAC-PAGE as the first dimension are less applicable for the differential quantitative analysis of redundant protein spots which might give insight into post-translational modifications that are relevant in age- and stress-related changes. Moreover, 2D-PAGE with high resolution IEF was able to resolve a surprisingly large number of membrane proteins from mitochondrial preparations. For aconitase-2, an enzyme playing an important role in mitochondrial aging, a more thorough molecular analysis of all separable isoforms was performed, leading to the identification of two particular N-formylkynurenine modifications. Next to protein redundancy, native protein-protein interactions, with the potential of relating certain post-translational modification patterns to distinct oligomeric states, e.g., oxidative phosphorylation super complexes, might provide novel and (patho-) physiologically relevant information. Among proteins identified, 14 new proteins (GenBank entries), previously not associated with mitochondria, were found.


Subject(s)
Aconitate Hydratase/biosynthesis , Aconitate Hydratase/chemistry , Kynurenine/analogs & derivatives , Mitochondria, Heart/enzymology , Reactive Oxygen Species/metabolism , Aconitate Hydratase/genetics , Animals , Biomarkers/metabolism , Cattle , Gene Expression Profiling , Humans , Isoenzymes/chemistry , Isoenzymes/genetics , Isoenzymes/metabolism , Kynurenine/chemistry
15.
J Proteome Res ; 4(6): 2117-25, 2005.
Article in English | MEDLINE | ID: mdl-16335957

ABSTRACT

We present a proof of principle study, using laser microdissection and pressure catapulting (LMPC) of two clinical tissue samples, each containing approximately 3.8 microg renal cell carcinoma protein and 3.8 microg normal kidney protein respectively from one patient. The study involved separate radio-iodination of each sample with both (125)I and (131)I, dual inverse replicate sample loading to high resolution 54 cm "daisy chain" serial immobilized pH gradient isoelectric focusing (IPG-IEF) 2D-PAGE gels, co-electrophoretic separation of cross-labeled proteins from different samples, and precision multiplex differential radioactive imaging to obtain signals specific for each sample coelectrophoresed within single gels but labeled with different isotopes of iodine, providing extremely precise intra-gel estimates of the abundance ratio for protein spots from both samples. Twelve multiplexed analytical radioactive SDS-gels from 4 serial IPG-IEF gels provided 24 individual radioactive images for a comprehensive analytical protein multiplex quantification study. A further 12 SDS gels containing (125)I-labeled sample were coelectrophoresed with preparative protein amounts obtained from whole tissue sections for the mass spectrometric identification of comigrating proteins. This consumed <40% of the (125)I-labeled sample, and <20% of the (131)I-labeled sample from the respective original 3.8 microg samples. Twenty-nine proteins were identified by mass spectrometry with PMF scores >70 that were >2-fold differentially abundant between the samples and t-test probabilities <0.05. We conclude that this combination of technologies provides excellent quality protein multiplex data for the differential abundance analysis of large numbers of proteins from extremely small samples, and is applicable to a broad range of clinical and related applications.


Subject(s)
Carcinoma, Renal Cell/metabolism , Gene Expression Regulation, Neoplastic , Isoelectric Focusing/methods , Liver Neoplasms/metabolism , Proteomics/methods , Cell Line, Tumor , Electrophoresis, Gel, Two-Dimensional , Electrophoresis, Polyacrylamide Gel , Gels , Humans , Hydrogen-Ion Concentration , Lasers , Mass Spectrometry , Odds Ratio , Proteins/chemistry , Proteome , Proton-Motive Force , Radioisotopes/therapeutic use
16.
Electrophoresis ; 26(16): 3185-90, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16041705

ABSTRACT

We previously demonstrated the separation of proteins by isoelectric focusing (IEF) over pH 4-8 immobilized pH gradients (IPGs) over 54 cm (Poland et al., Electrophoresis 2003, 24, 1271). Here we show that similar results can be conveniently achieved using commercially available IPGs of appropriate pH ranges positioned end-on-end in series during electrophoresis, which we term "daisy chain IEF". Proteins efficiently electrophorese from one IPG to another during IEF by traversing buffer-filled porous bridges between the serial IPGs. A variety of materials can function as bridges, including paper, polyacrylamide gels or even IPGs. The quality of two-dimensional (2-D) protein patterns is not apparently worse than that generated by conventional IEF using the same individual IPGs. A major advantage of this method is that sample is consumed efficiently, without the requirement for preliminary steps, such as chamber IEF. This advantage is pronounced when working with extremely limited sources of samples, such as with clinical biopsies or cellular subfractions. The present study was limited by the commercial availability of suitable pH gradients. Proteomics analyses could be further improved if commercial vendors would manufacture IPGs with suitable pH ranges to achieve high resolution (approximately 100 cm) IEF separation of proteins in one electrophoretic step over the pH range 2-12.


Subject(s)
Electrophoresis, Gel, Two-Dimensional/methods , Isoelectric Focusing/instrumentation , Isoelectric Focusing/methods , Proteins/isolation & purification , Proteomics/methods , Hydrogen-Ion Concentration , Miniaturization
17.
J Proteome Res ; 4(3): 900-8, 2005.
Article in English | MEDLINE | ID: mdl-15952737

ABSTRACT

Glutamate is the major excitatory neurotransmitter in the mammalian brain and is related to memory by calcium-conducting receptors. Neuregulins have emerged as long-term modulating molecules of synaptic signaling by glutamate receptors, playing a role in some cognition/memory-related disorders and moreover being part of transient functional microdomains, called lipid rafts. Here we characterize one specific isoform of neuregulin as a central biomarker for glutamate-related signaling, integrating results from in vitro and in vivo models by a differential functional and proteomic approach.


Subject(s)
Neuregulin-1/analysis , Proteomics/methods , Alzheimer Disease/pathology , Animals , Biomarkers , Calcium/metabolism , Cells, Cultured , Female , Glutamic Acid , Hippocampus/cytology , Humans , Learning , Nerve Tissue Proteins/analysis , Protein Isoforms , Rats , Rats, Sprague-Dawley , Signal Transduction
18.
Rapid Commun Mass Spectrom ; 17(12): 1283-1290, 2003.
Article in English | MEDLINE | ID: mdl-12811751

ABSTRACT

The new method of analysis of relative isotopologue abundances (ARIA) applied here is based on the evaluation of total isotope patterns of tryptic protein fragments measured by matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry (MALDI-TOFMS) to calculate the mixing ratios of composites consisting of stable isotope labelled and isotopically natural (unlabelled) proteins, as described in an accompanying paper in this issue. Recently, Sechi (Rapid Commun. Mass Spectrom. 2002; 16: 1416-1424) and Gehanne et al. (Rapid Commun. Mass Spectrom. 2002; 16: 1692-1698) introduced the use of differential quantitative mass analysis by MALDI-TOFMS using mixtures of standard proteins alkylated prior to two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) with either acrylamide (AA) or deuterium-labelled [2,3,3'-D(3)]-acrylamide (D3AA). In the present study we validate the AA/D3AA system, firstly by measuring the yield of proteins alkylated with AA, and secondly by using differential radioactive labels ((125)I and (131)I) to quantitatively establish that non-comigration in 2D-PAGE is negligible. ARIA is then applied to quantitatively estimate the relative proportions of peptides labelled with AA or D3AA in the validated system, using typical silver-stained 2D-PAGE protein spots from 2D gels loaded with 150 microg of total liver protein. The precision and limitations of ARIA quantification of peptides differentially alkylated with isotopomeric reagents are discussed.


The new method of analysis of relative isotopologue abundances (ARIA) applied here is based on the evaluation of total isotope patterns of tryptic protein fragments measured by matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry (MALDI-TOFMS) to calculate the mixing ratios of composites consisting of stable isotope labelled and isotopically natural (unlabelled) proteins, as described in an accompanying paper in this issue. Recently, Sechi (Rapid Commun. Mass Spectrom. 2002; 16: 1416-1424) and Gehanne et al. (Rapid Commun. Mass Spectrom. 2002; 16: 1692-1698) introduced the use of differential quantitative mass analysis by MALDI-TOFMS using mixtures of standard proteins alkylated prior to two-dimensional polyacrylamide gel electrophoresis (2D-PAGE) with either acrylamide (AA) or deuterium-labelled [2,3,3'-D3 ]-acrylamide (D3AA). In the present study we validate the AA/D3AA system, firstly by measuring the yield of proteins alkylated with AA, and secondly by using differential radioactive labels (125 I and 131 I) to quantitatively establish that non-comigration in 2D-PAGE is negligible. ARIA is then applied to quantitatively estimate the relative proportions of peptides labelled with AA or D3AA in the validated system, using typical silver-stained 2D-PAGE protein spots from 2D gels loaded with 150 µg of total liver protein. The precision and limitations of ARIA quantification of peptides differentially alkylated with isotopomeric reagents are discussed.


Subject(s)
Acrylamide/chemistry , Isotope Labeling , Proteins/analysis , Alkylation , Amino Acid Sequence , Animals , Electrophoresis, Gel, Two-Dimensional , Iodoacetamide , Isotopes/analysis , Liver/chemistry , Molecular Sequence Data , Proteins/chemistry , Swine
19.
Biochem J ; 365(Pt 3): 889-95, 2002 Aug 01.
Article in English | MEDLINE | ID: mdl-11939909

ABSTRACT

gamma-Tubulin is necessary for nucleation and polar orientation of microtubules in vivo. The molecular mechanism of microtubule nucleation by gamma-tubulin and the regulation of this process are not fully understood. Here we show that there are two gamma-tubulin forms in the brain that are present in complexes of various sizes. Large complexes tend to dissociate in the presence of a high salt concentration. Both gamma-tubulins co-polymerized with tubulin dimers, and multiple gamma-tubulin bands were identified in microtubule protein preparations under conditions of non-denaturing electrophoresis. Immunoprecipitation experiments with monoclonal antibodies against gamma-tubulin and alpha-tubulin revealed interactions of both gamma-tubulin forms with tubulin dimers, irrespective of the size of complexes. We suggest that, besides small and large gamma-tubulin complexes, other molecular gamma-tubulin form(s) exist in brain extracts. Two-dimensional electrophoresis revealed multiple charge variants of gamma-tubulin in both brain extracts and microtubule protein preparations. Post-translational modification(s) of gamma-tubulins might therefore have an important role in the regulation of microtubule nucleation in neuronal cells.


Subject(s)
Brain Chemistry , Tubulin/chemistry , Tubulin/metabolism , Animals , Cell Fractionation , Dimerization , Electrophoresis, Polyacrylamide Gel , Protein Isoforms , Swine , Tissue Extracts/chemistry , Tissue Extracts/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...