Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Nanomedicine ; 17: 266-275, 2019 04.
Article in English | MEDLINE | ID: mdl-30794962

ABSTRACT

The complement system plays an essential role in both innate and adaptive immunity. The traditional understanding of this system comes from studies investigating complement proteins produced by the liver and present in plasma to "complement" the immune cell-mediated response to invading pathogens. Recently, it has been reported that immune cells including, but not limited to, T-cells and monocytes, express complement proteins. This complement is referred to as intracellular (IC) and implicated in the regulation of T-cell activation. The mechanisms and the structure-activity relationship between nanomaterials and IC, however, are currently unknown. Herein, we describe a structure-activity relationship study demonstrating that under in vitro conditions, only polymeric materials with cationic surfaces activate IC in T-cells. The effect also depends on particle size and occurs through a mechanism involving membrane damage, thereby IC on the cell surface serves as a self-opsonization marker in response to the nanoparticle-triggered danger affecting the cell integrity.


Subject(s)
Complement Activation , Lymphocyte Activation , Nanoparticles/adverse effects , Polymers/adverse effects , T-Lymphocytes/immunology , Cations/adverse effects , Cations/chemistry , Cells, Cultured , Complement Activation/drug effects , Humans , Jurkat Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Nanoparticles/chemistry , Polymers/chemistry , T-Lymphocytes/drug effects
2.
Sci Rep ; 13(1): 14907, 2023 09 09.
Article in English | MEDLINE | ID: mdl-37689790

ABSTRACT

All-trans-retinoic acid (ATRA), the retinoic acid receptors (RARs) agonist, regulates cell growth, differentiation, immunity, and survival. We report that ATRA-treatment repressed cancer growth in syngeneic immunocompetent, but not immunodeficient mice. The tumor microenvironment was implicated: CD8+ T cell depletion antagonized ATRA's anti-tumorigenic effects in syngeneic mice. ATRA-treatment with checkpoint blockade did not cooperatively inhibit murine lung cancer growth. To augment ATRA's anti-tumorigenicity without promoting its pro-tumorigenic potential, an RARγ agonist (IRX4647) was used since it regulates T cell biology. Treating with IRX4647 in combination with an immune checkpoint (anti-PD-L1) inhibitor resulted in a statistically significant suppression of syngeneic 344SQ lung cancers in mice-a model known for its resistance to checkpoints and characterized by low basal T cell and PD-L1 expression. This combined treatment notably elevated CD4+ T-cell presence within the tumor microenvironment and increased IL-5 and IL-13 tumor levels, while simultaneously decreasing CD38 in the tumor stroma. IL-5 and/or IL-13 treatments increased CD4+ more than CD8+ T-cells in mice. IRX4647-treatment did not appreciably affect in vitro lung cancer growth, despite RARγ expression. Pharmacokinetic analysis found IRX4647 plasma half-life was 6 h in mice. Yet, RARα antagonist (IRX6696)-treatment with anti-PD-L1 did not repress syngeneic lung cancer growth. Together, these findings provide a rationale for a clinical trial investigating an RARγ agonist to augment check point blockade response in cancers.


Subject(s)
CD8-Positive T-Lymphocytes , Lung Neoplasms , Animals , Mice , Interleukin-13 , Interleukin-5 , Tumor Microenvironment , Receptors, Retinoic Acid , Lung Neoplasms/drug therapy , Tretinoin , Carcinogenesis
3.
Blood ; 116(10): 1755-60, 2010 Sep 09.
Article in English | MEDLINE | ID: mdl-20511543

ABSTRACT

In chronic granulomatous disease (CGD), defective phagocytic nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity causes reduced superoxide anion (O(2)(·)) radical production leading to frequent infections as well as granulomas and impaired wound healing indicative of excessive inflammation. Based on recent mouse studies, the lack of O(2)(·)-dependent interferon γ (IFNγ)-induced synthesis of kynurenine (kyn), an anti-inflammatory tryptophan metabolite produced by indolamine 2,3 deoxygenase (IDO), was proposed as a cause of hyperinflammation in CGD and this pathway has been considered for clinical intervention. Here, we show that IFNγ induces normal levels of kynurenine in cultures of O(2)(·)-deficient monocytes, dendritic cells, and polymorphonuclear leukocytes from gp91(PHOX)- or p47(PHOX)-deficient human CGD donors. Kynurenine accumulation was dose- and time-dependent as was that of a downstream metabolite, anthranilic acid. Furthermore, urinary and serum levels of kynurenine and a variety of other tryptophan metabolites were elevated rather than suppressed in CGD donors. Although we did not specifically evaluate kyn metabolism in local tissue or inflamed sites in humans, our data demonstrates that O(2)(·) anion is dispensable for the rate-limiting step in tryptophan degradation, and CGD patients do not appear to have either hematopoietic cell or systemic deficits in the production of the anti-inflammatory kynurenine molecule.


Subject(s)
Kynurenine/metabolism , Leukocytes/metabolism , Superoxides/metabolism , Tryptophan/metabolism , Cells, Cultured , Chromatography, Liquid , Dose-Response Relationship, Drug , Granulomatous Disease, Chronic/blood , Granulomatous Disease, Chronic/genetics , Granulomatous Disease, Chronic/urine , Humans , Immunoblotting , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Interferon-gamma/pharmacology , Kinetics , Leukocytes/cytology , Leukocytes/drug effects , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/genetics , Monocytes/cytology , Monocytes/drug effects , Monocytes/metabolism , NADPH Oxidase 2 , NADPH Oxidases/deficiency , NADPH Oxidases/genetics , Neutrophils/cytology , Neutrophils/drug effects , Neutrophils/metabolism , Spectrometry, Mass, Electrospray Ionization , Time Factors , Tryptophan/urine
4.
Anal Chem ; 83(6): 2394-6, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21319742

ABSTRACT

Proteomics is the study of all proteins in a biological sample. High-pressure liquid chromatography coupled online with mass spectrometry (HPLC/MS) is currently the method of choice for proteomic analysis. Proteins are extracted, separated at the protein or peptide level (after enzymatic digestion), and fractions are analyzed by HPLC/MS. Detection during off-line fractionation is generally conducted using UV-vis, which is not sensitive enough to distinguish fractions having the largest concentration of proteins/peptides and should not be combined prior to HPLC/MS. To overcome this deficiency, we utilize fluorescence or UV-laser induced fluorescence (UV-LIF) detection for measuring proteins/peptides during the off-line fractionation. Fluorescence detection allows low-abundance proteins/peptides that contain aromatic amino acids to be measured. In this study, peptide/protein samples fractionated using ion-exchange chromatography were detected using UV absorbance, fluorescence, and UV-LIF. The results indicated that fluorescence and UV-LIF were able to detect the lower abundance proteins/peptides to give a more representative chromatogram, allowing the analyst to decide which fractions should be combined prior to HPLC/tandem mass spectrometry (MS/MS) analysis.


Subject(s)
Chromatography, High Pressure Liquid/methods , Lasers , Peptide Fragments/analysis , Proteins/analysis , Proteomics/methods , Spectrometry, Fluorescence/methods , Spectrophotometry, Ultraviolet/methods , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Proteins/chemistry , Proteins/metabolism , Tandem Mass Spectrometry , Trypsin/metabolism
5.
J Sep Sci ; 34(24): 3484-92, 2011 Dec.
Article in English | MEDLINE | ID: mdl-22102289

ABSTRACT

Chromatography and electrophoresis have been used for the last half-century to separate small and large molecules. Advances in MS instrumentation and techniques for sample introduction into the mass analyzer (i.e. matrix-assisted laser desorption/ionization and electrospray ionization), chromatography in all its formats and modes and two-dimensional gel electrophoresis, including two-dimensional difference gel electrophoresis, enabled the separation of complex biological mixtures, such as the proteome and the metabolome, in a biological sample. These advances have made it possible to identify compounds that can be used to discriminate between two samples taken from healthy and diseased individuals. The objective is to find proteins or metabolites that can be used as a clinical test for the early diagnosis, prognosis and monitoring of the disease and the outcome of therapy. In this manuscript, we present an overview of what has been achieved in the search for biomarkers, with emphasis on cancer, using separation science and MS.


Subject(s)
Biomarkers, Tumor/analysis , Metabolomics , Proteomics , Biomarkers, Tumor/isolation & purification , Biomarkers, Tumor/metabolism , Chemical Fractionation , Chromatography, Gas , Electrophoresis , Humans , Mass Spectrometry
6.
Oncotarget ; 12(20): 2022-2038, 2021 Sep 28.
Article in English | MEDLINE | ID: mdl-34611477

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths in the USA and worldwide. Yet, about 95% of new drug candidates validated in preclinical phase eventually fail in clinical trials. Such a high attrition rate is attributed mostly to the inability of conventional two-dimensionally (2D) cultured cancer cells to mimic native three-dimensional (3D) growth of malignant cells in human tumors. To ascertain phenotypical differences between these two distinct culture conditions, we carried out a comparative proteomic analysis of a membrane fraction obtained from 3D- and 2D-cultured NSCLC model cell line NCI-H23. This analysis revealed a map of 1,166 (24%) protein species regulated in culture dependent manner, including differential regulation of a subset of cell surface-based CD molecules. We confirmed exclusive expression of CD99, CD146 and CD239 in 3D culture. Furthermore, label-free quantitation, targeting KRas proteoform-specific peptides, revealed upregulation of both wild type and monoallelic KRas4BG12C mutant at the surface of 3D cultured cells. In order to reduce the high attrition rate of new drug candidates, the results of this study strongly suggests exploiting base-line molecular profiling of a large number of patient-derived NSCLC cell lines grown in 2D and 3D culture, prior to actual drug candidate testing.

7.
Nat Commun ; 12(1): 7318, 2021 12 16.
Article in English | MEDLINE | ID: mdl-34916494

ABSTRACT

Proteasome substrate receptor hRpn13 is a promising anti-cancer target. By integrated in silico and biophysical screening, we identified a chemical scaffold that binds hRpn13 with non-covalent interactions that mimic the proteasome and a weak electrophile for Michael addition. hRpn13 Pru domain binds proteasomes and ubiquitin whereas its DEUBAD domain binds deubiquitinating enzyme UCHL5. NMR revealed lead compound XL5 to interdigitate into a hydrophobic pocket created by lateral movement of a Pru ß-hairpin with an exposed end for Proteolysis Targeting Chimeras (PROTACs). Implementing XL5-PROTACs as chemical probes identified a DEUBAD-lacking hRpn13 species (hRpn13Pru) present naturally with cell type-dependent abundance. XL5-PROTACs preferentially target hRpn13Pru, causing its ubiquitination. Gene-editing and rescue experiments established hRpn13 requirement for XL5-PROTAC-triggered apoptosis. These data establish hRpn13 as an anti-cancer target for multiple myeloma and introduce an hRpn13-targeting scaffold that can be optimized for preclinical trials against hRpn13Pru-producing cancer types.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , Multiple Myeloma/metabolism , Ubiquitination , Apoptosis , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Intracellular Signaling Peptides and Proteins/genetics , Multiple Myeloma/genetics , Proteasome Endopeptidase Complex/metabolism , Transcription Factors/metabolism , Transcriptional Activation , Ubiquitin/metabolism
8.
J Proteome Res ; 9(12): 6696-704, 2010 Dec 03.
Article in English | MEDLINE | ID: mdl-20968308

ABSTRACT

Affinity purification of protein complexes followed by identification using liquid chromatography/mass spectrometry (LC-MS/MS) is a robust method to study the fundamental process of protein interaction. Although affinity isolation reduces the complexity of the sample, fractionation prior to LC-MS/MS analysis is still necessary to maximize protein coverage. In this study, we compared the protein coverage obtained via LC-MS/MS analysis of protein complexes prefractionated using two commonly employed methods, SDS-PAGE and strong cation exchange chromatography (SCX). The two complexes analyzed focused on the nuclear proteins Bmi-1 and GATA3 that were expressed within the cells at low and high levels, respectively. Prefractionation of the complexes at the peptide level using SCX consistently resulted in the identification of approximately 3-fold more proteins compared to separation at the protein level using SDS-PAGE. The increase in the number of identified proteins was especially pronounced for the Bmi-1 complex, where the target protein was expressed at a low level. The data show that prefractionation of affinity isolated protein complexes using SCX prior to LC-MS/MS analysis significantly increases the number of identified proteins and individual protein coverage, particularly for target proteins expressed at low levels.


Subject(s)
Chemical Fractionation/methods , Chromatography, Ion Exchange/methods , Electrophoresis, Polyacrylamide Gel/methods , Multiprotein Complexes/analysis , Cations , Cell Line, Tumor , Chromatography, Liquid , GATA3 Transcription Factor/analysis , GATA3 Transcription Factor/genetics , HEK293 Cells , Humans , Mass Spectrometry , Nuclear Proteins/analysis , Nuclear Proteins/genetics , Polycomb Repressive Complex 1 , Proto-Oncogene Proteins/analysis , Proto-Oncogene Proteins/genetics , Repressor Proteins/analysis , Repressor Proteins/genetics , Reproducibility of Results , Transfection
9.
Anal Chem ; 82(13): 5878-86, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20540505

ABSTRACT

Differential (18)O/(16)O stable isotope labeling of peptides that relies on enzyme-catalyzed oxygen exchange at their carboxyl termini in the presence of H(2)(18)O has been widely used for relative quantitation of peptides/proteins. The role of tryptic proteolysis in bottom-up shotgun proteomics and low reagent costs have made trypsin-catalyzed (18)O postdigestion exchange a convenient and affordable stable isotope labeling approach. However, it is known that trypsin-catalyzed (18)O exchange at the carboxyl terminus is in many instances inhomogeneous/incomplete. The extent of the (18)O exchange/incorporation fluctuates from peptide to peptide mostly due to variable enzyme-substrate affinity. Thus, accurate calculation and interpretation of peptide ratios are analytically complicated and in some regard deficient. Therefore, a computational approach capable of improved measurement of actual (18)O incorporation for each differentially labeled peptide pair is needed. In this regard, we have developed an algorithmic method that relies on the trapezoidal rule to integrate peak intensities of all detected isotopic species across a particular peptide ion over the retention time, which fits the isotopic manifold to Poisson distributions. Optimal values for manifold fitting were calculated and then (18)O/(16)O ratios derived via evolutionary programming. The algorithm is tested using trypsin-catalyzed (18)O postdigestion exchange to differentially label bovine serum albumin (BSA) at a priori determined ratios. Both accuracy and precision are improved utilizing this rigorous mathematical approach. We further demonstrate the effectiveness of this method to accurately calculate (18)O/(16)O ratios in a large scale proteomic quantitation of detergent resistant membrane microdomains (DRMMs) isolated from cells expressing wild-type HIV-1 Gag and its nonmyristylated mutant.


Subject(s)
Algorithms , Isotope Labeling/methods , Peptides/chemistry , Amino Acid Sequence , Animals , Cattle , HeLa Cells , Humans , Membrane Microdomains/metabolism , Molecular Sequence Data , Oxygen Isotopes/chemistry , Serum Albumin, Bovine/chemistry , Serum Albumin, Bovine/metabolism , Trypsin/metabolism , gag Gene Products, Human Immunodeficiency Virus/chemistry , gag Gene Products, Human Immunodeficiency Virus/metabolism
10.
Anal Chem ; 82(5): 1584-8, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-20121140

ABSTRACT

A method that relies on subtractive tissue-directed shot-gun proteomics to identify tumor proteins in the blood of a patient newly diagnosed with cancer is described. To avoid analytical and statistical biases caused by physiologic variability of protein expression in the human population, this method was applied on clinical specimens obtained from a single patient diagnosed with nonmetastatic renal cell carcinoma (RCC). The proteomes extracted from tumor, normal adjacent tissue and preoperative plasma were analyzed using 2D-liquid chromatography-mass spectrometry (LC-MS). The lists of identified proteins were filtered to discover proteins that (i) were found in the tumor but not normal tissue, (ii) were identified in matching plasma, and (iii) whose spectral count was higher in tumor tissue than plasma. These filtering criteria resulted in identification of eight tumor proteins in the blood. Subsequent Western-blot analysis confirmed the presence of cadherin-5, cadherin-11, DEAD-box protein-23, and pyruvate kinase in the blood of the patient in the study as well as in the blood of four other patients diagnosed with RCC. These results demonstrate the utility of a combined blood/tissue analysis strategy that permits the detection of tumor proteins in the blood of a patient diagnosed with RCC.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Renal Cell/blood , Kidney Neoplasms/blood , Biomarkers, Tumor/analysis , Carcinoma, Renal Cell/diagnosis , Chromatography, Liquid , Humans , Kidney Neoplasms/diagnosis , Mass Spectrometry
11.
Methods Mol Biol ; 1647: 71-90, 2017.
Article in English | MEDLINE | ID: mdl-28808996

ABSTRACT

The discovery of novel drug targets and biomarkers via mass spectrometry (MS)-based proteomic analysis of clinical specimens has proven to be challenging. The wide dynamic range of protein concentration in clinical specimens and the high background/noise originating from highly abundant proteins in tissue homogenates and serum/plasma encompass two major analytical obstacles. Immunoaffinity depletion of highly abundant blood-derived proteins from serum/plasma is a well-established approach adopted by numerous researchers; however, the utilization of this technique for immunodepletion of tissue homogenates obtained from fresh frozen clinical specimens is lacking. We first developed immunoaffinity depletion of highly abundant blood-derived proteins from tissue homogenates, using renal cell carcinoma as a model disease, and followed this study by applying it to different tissue types. Tissue homogenate immunoaffinity depletion of highly abundant proteins may be equally important as is the recognized need for depletion of serum/plasma, enabling more sensitive MS-based discovery of novel drug targets, and/or clinical biomarkers from complex clinical samples. Provided is a detailed protocol designed to guide the researcher through the preparation and immunoaffinity depletion of fresh frozen tissue homogenates for two-dimensional liquid chromatography, tandem mass spectrometry (2D-LC-MS/MS)-based molecular profiling of tissue specimens in the context of drug target and/or biomarker discovery.


Subject(s)
Analytic Sample Preparation Methods , Biomarkers, Pharmacological/analysis , Blood Proteins/isolation & purification , Molecular Targeted Therapy , Proteomics , Specimen Handling/methods , Tandem Mass Spectrometry/methods , Carcinoma, Renal Cell/metabolism , Chromatography, Liquid/methods , Humans , Kidney Neoplasms/metabolism
12.
Cell Chem Biol ; 24(2): 231-242, 2017 Feb 16.
Article in English | MEDLINE | ID: mdl-28163016

ABSTRACT

Non-enzymatic protein modification driven by thioester reactivity is thought to play a major role in the establishment of cellular lysine acylation. However, the specific protein targets of this process are largely unknown. Here we report an experimental strategy to investigate non-enzymatic acylation in cells. Specifically, we develop a chemoproteomic method that separates thioester reactivity from enzymatic utilization, allowing selective enrichment of non-enzymatic acylation targets. Applying this method to cancer cell lines identifies numerous candidate targets of non-enzymatic acylation, including several enzymes in lower glycolysis. Functional studies highlight malonyl-CoA as a reactive thioester metabolite that can modify and inhibit glycolytic enzyme activity. Finally, we show that synthetic thioesters can be used as novel reagents to probe non-enzymatic acylation in living cells. Our studies provide new insights into the targets and drivers of non-enzymatic acylation, and demonstrate the utility of reactivity-based methods to experimentally investigate this phenomenon in biology and disease.


Subject(s)
Esters/metabolism , Sulfhydryl Compounds/metabolism , Acyl Coenzyme A/chemistry , Acyl Coenzyme A/metabolism , Acylation , Esters/chemistry , Humans , Models, Molecular , Molecular Structure , Proteomics , Sulfhydryl Compounds/chemistry , Tumor Cells, Cultured
13.
Oncotarget ; 7(52): 86948-86971, 2016 Dec 27.
Article in English | MEDLINE | ID: mdl-27894102

ABSTRACT

Oncogenic Ras mutants play a major role in the etiology of most aggressive and deadly carcinomas in humans. In spite of continuous efforts, effective pharmacological treatments targeting oncogenic Ras isoforms have not been developed. Cell-surface proteins represent top therapeutic targets primarily due to their accessibility and susceptibility to different modes of cancer therapy. To expand the treatment options of cancers driven by oncogenic Ras, new targets need to be identified and characterized at the surface of cancer cells expressing oncogenic Ras mutants. Here, we describe a mass spectrometry-based method for molecular profiling of the cell surface using KRasG12V transfected MCF10A (MCF10A-KRasG12V) as a model cell line of constitutively activated KRas and native MCF10A cells transduced with an empty vector (EV) as control. An extensive molecular map of the KRas surface was achieved by applying, in parallel, targeted hydrazide-based cell-surface capturing technology and global shotgun membrane proteomics to identify the proteins on the KRasG12V surface. This method allowed for integrated proteomic analysis that identified more than 500 cell-surface proteins found unique or upregulated on the surface of MCF10A-KRasG12V cells. Multistep bioinformatic processing was employed to elucidate and prioritize targets for cross-validation. Scanning electron microscopy and phenotypic cancer cell assays revealed changes at the cell surface consistent with malignant epithelial-to-mesenchymal transformation secondary to KRasG12V activation. Taken together, this dataset significantly expands the map of the KRasG12V surface and uncovers potential targets involved primarily in cell motility, cellular protrusion formation, and metastasis.


Subject(s)
Membrane Proteins/analysis , Mutant Proteins/analysis , Proteomics/methods , Proto-Oncogene Proteins p21(ras)/analysis , Antigens, CD/analysis , Antigens, Neoplasm , Basigin/analysis , Cell Adhesion Molecules/analysis , Cell Line, Tumor , Cell Movement , Computational Biology , Epithelial-Mesenchymal Transition , Glycoproteins/classification , Glycoproteins/physiology , Humans , Mass Spectrometry , Microscopy, Electron, Scanning , Neoplasm Proteins/analysis
14.
J Am Soc Mass Spectrom ; 16(8): 1221-30, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15979327

ABSTRACT

With advancements in the analytical technologies and methodologies in proteomics, there is great interest in biomarker discovery in biofluids such as serum and plasma. Current hypotheses suggest that the low molecular weight (LMW) serum proteome possesses an archive of clipped and cleaved protein fragments that may provide insight into disease development. Though these biofluids represent attractive samples from which new and more accurate disease biomarkers may be found, the intrinsic person-to-person variability in these samples complicates their discovery. Mice are one of the most extensively used animal models for studying human disease because they represent a highly controllable experimental model system. In this study, the LMW serum proteome was compared between xenografted tumor-bearing mice and control mice by differential labeling utilizing trypsin-mediated incorporation of the stable isotope of oxygen, 18O. The digestates were combined, fractionated by strong cation exchange chromatography, and analyzed by nanoflow reversed-phase liquid chromatography coupled online with tandem mass spectrometry, resulting in the identification of 6003 proteins identified by at least a single, fully tryptic peptide. Almost 1650 proteins were identified and quantitated by two or more fully tryptic peptides. The methodology adopted in this work provides the means for future quantitative measurements in comparative animal models of disease and in human disease cohorts.


Subject(s)
Blood Proteins/analysis , Lung Neoplasms/physiopathology , Mass Spectrometry/methods , Oxygen Isotopes , Proteomics/methods , Animals , Disease Models, Animal , Humans , Lung Neoplasms/blood , Mass Spectrometry/instrumentation , Mice , Molecular Weight , Neoplasm Transplantation , Proteomics/instrumentation , Transplantation, Heterologous
15.
Article in English | MEDLINE | ID: mdl-15680787

ABSTRACT

Current solution based proteomic analysis methods are generally based on enzymatic digestion of a protein mixture followed by separation using multidimensional liquid chromatography and/or electrophoresis where peptide identification is typically accomplished by tandem mass spectrometry (MS/MS). It is generally accepted that no single chromatographic or electrophoretic procedure is capable of resolving the complex mixture of peptides that results from a global proteolytic digest of a proteome. Therefore, combining two or more orthogonal (multimodal) separation procedures dramatically improves the overall resolution and results in a larger number of peptides being identified from complex proteome digests. Separation of a proteome digest is a particularly challenging analytical problem due to the large number of peptides and the wide concentration dynamic range. While it has been demonstrated that increasing the number of dimensions of separation prior to MS analysis increases the number of peptides that may be identified, a balance between the time invested and the overall results obtained must be carefully considered. This manuscript provides a review of two- and three-dimensional peptide separation strategies combined with MS for the analysis of complex peptide mixtures.


Subject(s)
Chromatography, Liquid/methods , Peptides/isolation & purification , Proteomics
16.
J Invest Dermatol ; 123(4): 691-9, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15373774

ABSTRACT

Membrane proteins are responsible for many critical cellular functions and identifying cell surface proteins on different keratinocyte populations by proteomic approaches would improve our understanding of their biological function. The ability to characterize membrane proteins, however, has lagged behind that of soluble proteins both in terms of throughput and protein coverage. In this study, a membrane proteomic investigation of keratinocytes using a two-dimensional liquid chromatography (LC) tandem-mass spectrometry (MS/MS) approach that relies on a buffered methanol-based solubilization, and tryptic digestion of purified plasma membrane is described. A highly enriched plasma membrane fraction was prepared from newborn foreskins using sucrose gradient centrifugation, followed by a single-tube solubilization and tryptic digestion of membrane proteins. This digestate was fractionated by strong cation-exchange chromatography and analyzed using microcapillary reversed-phase LC-MS/MS. In a set of 1306 identified proteins, 866 had a gene ontology (GO) annotation for cellular component, and 496 of these annotated proteins (57.3%) were assigned as known integral membrane proteins or membrane-associated proteins. Included in the identification of a large number of aqueous insoluble integral membrane proteins were many known intercellular adhesion proteins and gap junction proteins. Furthermore, 121 proteins from cholesterol-rich plasma membrane domains (caveolar and lipid rafts) were identified.


Subject(s)
Epidermis/chemistry , Keratinocytes/chemistry , Mass Spectrometry/methods , Proteomics , Caveolae/chemistry , Cell Adhesion Molecules/analysis , Cell Membrane/chemistry , Epidermal Cells , Humans , Infant, Newborn , Intercellular Junctions/chemistry , Membrane Microdomains/chemistry
17.
J Chromatogr A ; 979(1-2): 81-9, 2002 Dec 06.
Article in English | MEDLINE | ID: mdl-12498235

ABSTRACT

A two-dimensional liquid chromatography/capillary electrophoresis technique was developed for rapid and comprehensive mapping of cell extracts. The cell extracts were first separated by reversed-phase HPLC based on hydrophobicity. Fractions of the effluent from the HPLC system were collected into 96-well microtiter plates and dried under vacuum. The fractions were reconstituted with deionized water, separated by capillary array electrophoresis based on charge-to-size ratio, and detected by UV absorption at 214 nm. Prior to analysis by multiplexed capillary electrophoresis, the reconstituted fractions were concentrated on-column using large volume sample stacking with polarity switching. In this way, high-resolution analysis of even the minor components in the complicated mixture was possible.


Subject(s)
Cell Extracts/chemistry , Chromatography, High Pressure Liquid/methods , Electrophoresis, Capillary/methods , Ovarian Neoplasms/chemistry , Spectrophotometry, Ultraviolet/methods , Female , Humans
18.
J Chromatogr A ; 1053(1-2): 37-42, 2004 Oct 22.
Article in English | MEDLINE | ID: mdl-15543970

ABSTRACT

A review of sheathless interfaces for capillary electrophoresis (CE)-mass spectrometry (MS) is presented. The review discusses the on-line CE-MS system requirements, advantages and weaknesses of current sheathless interface designs for CE-electrospray ionization MS, and comparison between sheath flow and sheathless interfaces. The advantages and limitations of three sheathless designs are discussed and commented upon, these include single-capillary, two-capillary and three-piece designs.


Subject(s)
Electrophoresis, Capillary/methods , Spectrometry, Mass, Electrospray Ionization/methods
19.
Biomark Med ; 8(2): 269-86, 2014.
Article in English | MEDLINE | ID: mdl-24521024

ABSTRACT

The discovery of clinically relevant cancer biomarkers using mass spectrometry (MS)-based proteomics has proven difficult, primarily because of the enormous dynamic range of blood-derived protein concentrations and the fact that the 22 most abundant blood-derived proteins constitute approximately 99% of the total plasma protein mass. Immunodepletion of clinical body fluid specimens (e.g., serum/plasma) for the removal of highly abundant proteins is a reasonable and reproducible solution. Often overlooked, clinical tissue specimens also contain a formidable amount of highly abundant blood-derived proteins present in tissue-embedded networks of blood/lymph capillaries and interstitial fluid. Hence, the dynamic range impediment to biomarker discovery remains a formidable obstacle, regardless of clinical sample type (solid tissue and/or body fluid). Thus, we optimized and applied simultaneous immunodepletion of blood-derived proteins from solid tissue and peripheral blood, using clear cell renal cell carcinoma as a model disease. Integrative analysis of data from this approach and genomic data obtained from the same type of tumor revealed concordant key pathways and protein targets germane to clear cell renal cell carcinoma. This includes the activation of the lipogenic pathway characterized by increased expression of adipophilin (PLIN2) along with 'cadherin switching', a phenomenon indicative of transcriptional reprogramming linked to renal epithelial dedifferentiation. We also applied immunodepletion of abundant blood-derived proteins to various tissue types (e.g., adipose tissue and breast tissue) showing unambiguously that the removal of abundant blood-derived proteins represents a powerful tool for the reproducible profiling of tissue proteomes. Herein, we show that the removal of abundant blood-derived proteins from solid tissue specimens is of equal importance to depletion of body fluids and recommend its routine use in the context of biological discovery and/or cancer biomarker research. Finally, this perspective presents the background, rationale and strategy for using tissue-directed high-resolution/accuracy MS-based shotgun proteomics to detect genuine tumor proteins in the peripheral blood of a patient diagnosed with nonmetastatic cancer, employing concurrent liquid chromatography-MS analysis of immunodepleted clinical tissue and blood specimens.


Subject(s)
Biomarkers, Tumor/analysis , Mass Spectrometry , Antibodies/immunology , Biomarkers, Tumor/blood , Blood Proteins/immunology , Blood Proteins/isolation & purification , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/pathology , Chromatography, High Pressure Liquid , Gene Expression Profiling , Humans , Kidney Neoplasms/blood , Kidney Neoplasms/metabolism , Kidney Neoplasms/pathology , Proteomics
20.
Methods Mol Biol ; 1002: 311-5, 2013.
Article in English | MEDLINE | ID: mdl-23625412

ABSTRACT

Chromatography in all its formats plays an important role in proteomics research. The search for protein biomarkers for different diseases has been an active area of research. The dynamic concentration range and the large number of proteins in a proteome require the development of multidimensional separation strategies to allow for the identification of the largest number of proteins. Strong cation-exchange chromatography (SCX) has been used extensively for the fractionation of proteins and peptides. This chapter provides a detailed description for the SCX fractionation of complex proteome samples.


Subject(s)
Chromatography, Ion Exchange/methods , Peptides/analysis , Proteomics/methods , Biomarkers/analysis , Cation Exchange Resins , Chemical Fractionation , Humans , Mass Spectrometry , Peptides/chemistry , Proteome/analysis
SELECTION OF CITATIONS
SEARCH DETAIL