首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 328 毫秒
1.
2.
Olfactory receptors (ORs) are expressed not only in the sensory neurons of the olfactory epithelium, where they detect volatile substances, but also in various other tissues where their potential functions are largely unknown. Here, we report the physiological characterization of human OR51E2, also named prostate-specific G-protein-coupled receptor (PSGR) due to its reported up-regulation in prostate cancer. We identified androstenone derivatives as ligands for the recombinant receptor. PSGR can also be activated with the odorant β-ionone. Activation of the endogenous receptor in prostate cancer cells by the identified ligands evoked an intracellular Ca2+ increase. Exposure to β-ionone resulted in the activation of members of the MAPK family and inhibition of cell proliferation. Our data give support to the hypothesis that because PSGR signaling could reduce growth of prostate cancer cells, specific receptor ligands might therefore be potential candidates for prostate cancer treatment.Excessive signaling by G-protein-coupled receptors (GPCRs)3 such as endothelin A receptor (1), bradykinin 1 receptor (2), follicle-stimulating hormone receptor (3), and thrombin receptor (4, 5) is known to occur in prostate cancers due to strong overexpression of the respective receptors. Activation of some of these GPCRs results in androgen-independent androgen receptor activation, thus promoting the transition of prostate cancer cells from an androgen-dependent to an androgen-independent state (6, 7).The prostate-specific G-protein-coupled receptor (PSGR) is a class A GPCR that was initially identified as a prostate-specific tumor biomarker (810). It is specifically expressed in prostate epithelial cells, and its expression increases significantly in human prostate intraepithelial neoplasia and prostate tumors, suggesting that PSGR may play an important role in early prostate cancer development and progression (9, 11). Although expression of the human PSGR was found to be prostate-specific (10, 12), mRNA can also be detected in the olfactory zone and the medulla oblongata of the human brain (12). Human PSGR shares 93% amino acid homology to the respective mouse and rat homologues, which are also expressed in the brain (12). Interestingly, PSGR has numerous sequence motifs in common with the large superfamily of olfactory receptors (ORs), which build the largest class of human GPCRs and allow the recognition of a wide range of structurally diverse molecules in the nasal epithelium (1315). Recently, also the steroid hormones androstenone and androstadienone were identified as OR ligands (16). In addition to their role in the sensory neurons of the nose, ORs have been found in different tissues throughout the body (17, 18). Their function(s) in these extranasal locations are questionable except for in a few cases where functional studies have been performed in spermatozoa (19, 20) and in enterochromaffin cells of the gastrointestinal tract (21).Here, we report the identification of steroid ligands of heterologously expressed PSGR and investigate the functional relevance of PSGR expression in prostate tissue. Steroid hormones elicited rapid Ca2+ responses in the LNCaP prostate cancer cell line and in primary human prostate epithelial cells. Moreover, activated PSGR causes phosphorylation of p38 and stress-activated protein kinase/c-Jun NH2-terminal kinase (SAPK/JNK) mitogen-activated protein kinases (MAPKs), resulting in reduced proliferation rates in prostate cancer cells.  相似文献   

3.
Glycosylation is one of the most common and important protein modifications in biological systems. Many glycoproteins naturally occur at low abundances, which makes comprehensive analysis extremely difficult. Additionally, glycans are highly heterogeneous, which further complicates analysis in complex samples. Lectin enrichment has been commonly used, but each lectin is inherently specific to one or several carbohydrates, and thus no single or collection of lectin(s) can bind to all glycans. Here we have employed a boronic acid-based chemical method to universally enrich glycopeptides. The reaction between boronic acids and sugars has been extensively investigated, and it is well known that the interaction between boronic acid and diols is one of the strongest reversible covalent bond interactions in an aqueous environment. This strong covalent interaction provides a great opportunity to catch glycopeptides and glycoproteins by boronic acid, whereas the reversible property allows their release without side effects. More importantly, the boronic acid-diol recognition is universal, which provides great capability and potential for comprehensively mapping glycosylation sites in complex biological samples. By combining boronic acid enrichment with PNGase F treatment in heavy-oxygen water and MS, we have identified 816 N-glycosylation sites in 332 yeast proteins, among which 675 sites were well-localized with greater than 99% confidence. The results demonstrated that the boronic acid-based chemical method can effectively enrich glycopeptides for comprehensive analysis of protein glycosylation. A general trend seen within the large data set was that there were fewer glycosylation sites toward the C termini of proteins. Of the 332 glycoproteins identified in yeast, 194 were membrane proteins. Many proteins get glycosylated in the high-mannose N-glycan biosynthetic and GPI anchor biosynthetic pathways. Compared with lectin enrichment, the current method is more cost-efficient, generic, and effective. This method can be extensively applied to different complex samples for the comprehensive analysis of protein glycosylation.Glycosylation is an extremely important protein modification that frequently regulates protein folding, trafficking, and stability. It is also involved in a wide range of cellular events (1) such as immune response (2, 3), cell proliferation (4), cell-cell interactions (5), and signal transduction (6). Aberrant protein glycosylation is believed to have a direct correlation with the development of several diseases, including diabetes, infectious diseases, and cancer (711). Secretory proteins frequently get glycosylated, including those in body fluids such as blood, saliva, and urine (12, 13). Samples containing these proteins can be easily obtained and used for diagnostic and therapeutic purposes. Several glycoproteins have previously been identified as biomarkers, including Her2/Neu in breast cancer (14), prostate-specific antigen (PSA) in prostate cancer (15), and CA125 in ovarian cancer (16, 17), which highlights the clinical importance of identifying glycoproteins as indicators or biomarkers of diseases. Therefore, effective methods for systematic analysis of protein glycosylation are essential to understand the mechanisms of glycobiology, identify drug targets and discover biomarkers.Approximately half of mammalian cell proteins are estimated to be glycosylated at any given time (18). There have been many reports regarding identification of protein glycosylation sites and elucidation of glycan structures (1930). Glycan structure analysis can lead to potential therapeutic and diagnostic applications (31, 32), but it is also critical to identify which proteins are glycosylated as well as the sites at which the modification occurs. Despite progress in recent years, the large-scale analysis of protein glycosylation sites using MS-based proteomics methods is still a challenge. Without an effective enrichment method, the low abundance of glycoproteins prohibits the identification of the majority of sites using the popular intensity-dependent MS sequence method.About a decade ago, a very beautiful and elegant method based on hydrazide chemistry was developed to enrich glycopeptides. Hydrazide conjugated beads reacted with aldehydes formed from the oxidation of cis-diols in glycans (33). This method has been extensively applied to many different types of biological samples (3441). Besides the hydrazide-based enrichment method, lectins have also been frequently used to enrich glycopeptides or glycoproteins before MS analysis (28, 29, 4246). However, there are many different types of lectins, and each is specific to certain glycans (47, 48). Therefore, no combination of lectins can bind to all glycosylated peptides or proteins, which prevents comprehensive analysis of protein glycosylation. Because of the complexity of biological samples, effective enrichment methods are critical for the comprehensive analysis of protein glycosylation before MS analysis.One common feature of all glycoproteins and glycopeptides is that they contain multiple hydroxyl groups in their glycans. From a chemistry point of view, this can be exploited to effectively enrich them. Ideally, chemical enrichment probes must have both strong and specific interactions with multiple hydroxyl groups. The reaction between boronic acids and 1,2- or 1,3-cis-diols in sugars has been extensively studied (4952) and applied for the small-scale analysis of glycoproteins (5355). Furthermore, boronate affinity chromatography has been employed for the analysis of nonenzymatically glycated peptides (56, 57). Boronic acid-based chemical enrichment methods are expected to have great potential for global analysis of glycopeptides when combined with modern MS-based proteomics techniques. However, the method has not yet been used for the comprehensive analysis of protein N-glycosylation in complex biological samples (58).Yeast is an excellent model biological system that has been extensively used in a wide range of experiments. Last year, two papers reported the large-scale analysis of protein N-glycosylation in yeast (59, 60). In one study, a new MS-based method was developed based on N-glycopeptide mass envelopes with a pattern via metabolic incorporation of a defined mixture of N-acetylglucosamine isotopologs into N-glycans. Peptides with the recoded envelopes were specifically targeted for fragmentation, facilitating high confidence site mapping (59). Using this method, 133 N-glycosylation sites were confidently identified in 58 yeast proteins. When combined with an effective enrichment method, this MS-based analysis will provide a more complete coverage of the N-glycoproteome. The other work combined lectin enrichment with digestion by two enzymes (Glu_c and trypsin) to increase the peptide coverage, and 516 well-localized N-glycosylation sites were identified in 214 yeast proteins by MS (60).Here we have comprehensively identified protein N-glycosylation sites in yeast by combining a boronic acid-based chemical enrichment method with MS-based proteomics techniques. Magnetic beads conjugated with boronic acid were systematically optimized to selectively enrich glycosylated peptides from yeast whole cell lysates. The enriched peptides were subsequently treated with Peptide-N4-(N-acetyl-beta-glucosaminyl)asparagine amidase (PNGase F)1 in heavy-oxygen water. Finally, peptides were analyzed by an on-line LC-MS system. Over 800 protein N-glycosylation sites were identified in the yeast proteome, which clearly demonstrates that the boronic acid-based chemical method is an effective enrichment method for large-scale analysis of protein glycosylation by MS.  相似文献   

4.
5.
Glycosylation is one of the most important and common forms of protein post-translational modification that is involved in many physiological functions and biological pathways. Altered glycosylation has been associated with a variety of diseases, including cancer, inflammatory and degenerative diseases. Glycoproteins are becoming important targets for the development of biomarkers for disease diagnosis, prognosis, and therapeutic response to drugs. The emerging technology of glycoproteomics, which focuses on glycoproteome analysis, is increasingly becoming an important tool for biomarker discovery. An in-depth, comprehensive identification of aberrant glycoproteins, and further, quantitative detection of specific glycosylation abnormalities in a complex environment require a concerted approach drawing from a variety of techniques. This report provides an overview of the recent advances in mass spectrometry based glycoproteomic methods and technology, in the context of biomarker discovery and clinical application.With recent advances in proteomics, analytical and computational technologies, glycoproteomics—the global analysis of glycoproteins—is rapidly emerging as a subfield of proteomics with high biological and clinical relevance. Glycoproteomics integrates glycoprotein enrichment and proteomics technologies to support the systematic identification and quantification of glycoproteins in a complex sample. The recent development of these techniques has stimulated great interest in applying the technology in clinical translational studies, in particular, protein biomarker research.While glycomics is the study of glycome (repertoire of glycans), glycoproteomics focuses on studying the profile of glycosylated proteins, i.e. the glycoproteome, in a biological system. Considerable work has been done to characterize the sequences and primary structure of the glycan moieties attached to proteins (13), and their structural alterations related to cancer (46). Recent reports have provided a comprehensive overview of the concept of glycomics and its prospective in biomarker research (710). In contrast, this review is focused on recent developments in glycoproteomic techniques and their unique application and technical challenge to biomarker discovery.

Glycoproteomics in Biomarker Discovery and Clinical Study

Most secretory and membrane-bound proteins produced by mammalian cells contain covalently linked glycans with diverse structures (2). The glycosylation form of a glycoprotein is highly specific at each glycosylation site and generally stable for a given cell type and physiological state. However, the glycosylation form of a protein can be altered significantly because of changes in cellular pathways and processes resulting from diseases, such as cancer, inflammation, and neurodegeneration. Such disease-associated alterations in glycoproteins can happen in one or both of two ways: 1) protein glycosylation sites are either hypo, hyper, or newly glycosylated and/or; 2) the glycosylation form of the attached carbohydrate moiety is altered. In fact, altered glycosylation patterns have long been recognized as hallmarks in cancer progression, in which tumor-specific glycoproteins are actively involved in neoplastic progression and metastasis (5, 6, 11, 12). Sensitive detection of such disease-associated glycosylation changes and abnormalities can provide a unique avenue to develop glycoprotein biomarkers for diagnosis and prognosis. In addition, intervention in the glycosylation and carbohydrate-dependent cellular pathways represent a potential new modality for cancer therapies (6, 11, 13). 14, 15) that are glycosylated proteins or protein complexes.

Table I

Listing of some of the US Food and Drug Administration (FDA) approved cancer biomarkers
Protein targetGlycosylationDetectionSourceDiseaseClinical biomarker
α-FetoproteinYesGlycoproteinSerumNonseminomatous testicular cancerDiagnosis
Human chorionic gonadotropin-βYesGlycoproteinSerumTesticular cancerDiagnosis
CA19–9YesCarbohydrateSerumPancreatic cancerMonitoring
CA125YesGlycoproteinSerumOvarian cancerMonitoring
CEA (carcinoembryonic antigen)YesProteinSerumColon cancerMonitoring
Epidermal growth factor receptorYesProteinTissueColon cancerTherapy selection
KITYesProtein (IHC)TissueGastrointestinal (GIST) cancerDiagnosis/Therapy selection
ThyroglobulinYesProteinSerumThyroid cancerMonitoring
PSA-prostate-specific antigen (Kallikrein 3)YesProteinSerumProstate cancerScreening/Monitoring/Diagnosis
CA15–3YesGlycoproteinSerumBreast cancerMonitoring
CA27–29YesGlycoproteinSerumBreast cancerMonitoring
HER2/NEUYesProtein (IHC), ProteinTissue, SerumBreast cancerPrognosis/Therapy selection/Monitoring
Fibrin/FDP-fibrin degradation proteinYesProteinUrineBladder cancerMonitoring
BTA-bladder tumour-associated antigen (Complement factor H related protein)YesProteinUrineBladder cancerMonitoring
CEA and mucin (high molecular weight)YesProtein (Immunofluorescence)UrineBladder cancerMonitoring
Open in a separate windowProtein biomarker development is a complex and challenging task. The criteria and approach applied for developing each individual biomarker can vary, depending on the purpose of the biomarker and the performance requirement for its clinical application (16, 17). In general, it has been suggested that the preclinical exploratory phase of protein biomarker development can be technically defined into four stages (18), including initial discovery of differential proteins; testing and selection of qualified candidates; verification of a subset of candidates; assay development and pre-clinical validation of potential biomarkers. Thanks to recent technological advances, mass spectrometry based glycoproteomics is now playing a major role in the initial phase of discovering aberrant glycoproteins associated with a disease. Glycoprotein enrichment techniques, coupled with multidimensional chromatographic separation and high-resolution mass spectrometry have greatly enhanced the analytical dynamic range and limit of detection for glycoprotein profiling in complex samples such as plasma, serum, other bodily fluids, or tissue. In addition, candidate-based quantitative glycoproteomics platforms have been introduced recently, allowing targeted detection of glycoprotein candidates in complex samples in a multiplexed fashion, providing a complementary tool for glycoprotein biomarker verification in addition to antibody based approaches. It is clear that glycoproteomics is gaining momentum in biomarker research.

Glycoproteomics Approaches

Glycoproteomic analysis is complicated not only by the variety of carbohydrates, but also by the complex linkage of the glycan to the protein. Glycosylation can occur at several different amino acid residues in the protein sequence. The most common and widely studied forms are N-linked and O-linked glycosylation. O-linked glycans are linked to the hydroxyl group on serine or threonine residues. N-linked glycans are attached to the amide group of asparagine residues in a consensus Asn-X-Ser/Thr sequence (X can be any amino acid except proline) (19). Other known, but less well studied forms of glycosylation include glycosylphosphatidylinositol anchors attached to protein carboxyl terminus, C-glycosylation that occurs on tryptophan residues (20), and S-linked glycosylation through a sulfur atom on cysteine or methionine (21, 22). Our following discussion is focused on glycoproteomic analysis of the most common N-linked and O-linked glycoproteins.A comprehensive analysis of glycoproteins in a complex biological sample requires a concerted approach. Although the specific methods for sample preparation can be different for different types of samples (e.g. plasma, serum, tissue, and cell lysate), a glycoproteomics pipeline typically consists of glycoprotein or glycopeptide enrichment, multidimensional protein or peptide separation, tandem mass spectrometric analysis, and bioinformatic data interpretation. For glycoprotein-based enrichment methods, proteolytic digestion can be performed before or after glycan cleavage, depending on the specific workflow and enrichment methods used. For glycopeptide enrichment, proteolytic digestion is typically performed before the isolation step so that glycopeptides, instead of glycoproteins, can be captured. For quantitative glycoproteomics profiling, additional steps, such as differential stable isotope labeling of the sample and controls, are required. Fig. 1 illustrates the general strategy for an integrated glycoproteomics analysis.Open in a separate windowFig. 1.The strategies of mass spectrometry based glycoproteomic analysis.Glycoproteins or glycopeptides can be effectively enriched using a variety of techniques (see below). Following the enrichment step, the workflow then splits into two directions: glycan analysis and glycoprotein analysis. The strategies for glycan analysis have been discussed in several reviews and will not be covered in this report. For glycoprotein analysis, bottom-up workflows (“shotgun proteomics”—peptide based proteomics analysis) (23) are still most common, providing not only detailed information of a glycoprotein profile, but also the specific mapping of glycosylation sites. It is notable that the reliable analysis of mass spectrometric data in glycoproteomic studies largely relies on bioinformatic tools and glyco-related databases that are available. An increasing number of algorithms and databases for glycan analysis have been developed and well documented in several recent reviews (2426). For glycoprotein and glycopeptide sequence analysis, a large number of well-characterized and annotated glycoproteins can be found in the UniProt Knowledgebase. In addition, many glycopeptide mass spectra are now available in the continually expanding PeptideAtlas library (27), which stores millions of high-resolution peptide fragment ion mass spectra acquired from a variety of biological and clinical samples for peptide and protein identification. Ultimately, all the data obtained from different aspects of the workflow need to be merged and interpreted in an integrated fashion so that the full extent of glycosylation changes associated with a particular biological state can be better revealed. To the best of our knowledge, the complete glycoform analysis of any glycoprotein in a specific cell type under any specific condition has not yet been accomplished for any glycoprotein with multiple glycosylation sites. Current technology can define the glycan compliment and profile the glycoproteins, but is not capable of putting them together to define the molecular species present. To date, such integrated studies still remain highly challenging, even with advanced tandem mass spectrometry technologies and growing bioinformatic resources (26, 2831).

Enrichment of the Glycoproteome

Characterization of the glycoproteome in a complex biological sample such as plasma, serum, or tissue, is analytically challenging because of the enormous complexity of protein and glycan constituents and the vast dynamic range of protein concentration in the sample. The selective enrichment of the glycoproteome is one of the most efficient ways to simplify the enormous complexity of a biological sample to achieve an in-depth glycoprotein analysis. Two approaches for glycoprotein enrichment have been widely applied: lectin affinity based enrichment methods (3136) and hydrazide chemistry-based solid phase extraction methods (3742). Recent studies have demonstrated that the two methods are complementary and a very effective means for the enrichment of glycoproteins or glycopeptides from human plasma and other bodily fluids (38, 39, 43). In addition, glycoprotein and glycopeptide enrichment using boronic acid (44, 45), size-exclusion chromatography (46), hydrophilic interaction (47) and a graphite powder microcolumn (48) have been reported.Lectin affinity enrichment is based on the specific binding interaction between a lectin and a distinct glycan structure attached on a glycoprotein (49, 50). There are a variety of lectin species that can selectively bind to different oligosaccharide epitopes. For instance, concanavalin A (ConA) binds to mannosyl and glucosyl residues of glycoproteins (51); wheat germ agglutinin (WGA) binds to N-acetyl-glucosamine and sialic acid (52); and jacalin (JAC) specifically recognizes galactosyl (β-1,3) N acetylgalactosamine and O-linked glycoproteins (53). Lectin affinity enrichment has been designed to enrich glycoproteins with specific glycan attachment from plasma, serum, tissue, and other biological samples through affinity chromatography and other methods. Multiple lectin species can also be combined to isolate multiple types of glycoproteins in complex biological samples (5459). Concanavalin A and wheat germ agglutinin, as well as jacalin are often used together to achieve a more extensive glycproteome characterization (31, 34, 57, 59, 60). Several reports have demonstrated a multilectin column approach to achieve a global enrichment of glycoproteins with various glycan attachments from serum and plasma (31, 34, 59, 61, 62). A recent study has developed a “filter aided sample preparation (FASP)” based method, which allows highly efficient enrichment of glycopeptides using multi-lectins (63). To date, most of the work using lectin affinity for targeted glycoprotein enrichment has focused on N-glycosylation because the binding specificity of lectin for O-glycosylation is less satisfactory. To overcome such caveat, efforts have been made using serial lectin columns of concanavalin A and jacelin in tandem to isolate O-glycopeptides from human serum (35).A hydrazide chemistry-based method has been applied to isolate glycoproteins and glycopeptides through the formation of covalent bonding between the glycans and the hydrazide groups (37). The carbohydrates on glycoproteins are first oxidized to form aldehyde groups, which sequentially react with hydrazide groups that are immobilized on a solid surface. The chemical reaction conjugates the glycoproteins to the solid phase by forming the covalent hydrazone bond. Although, conceptually, the majority of the glycoproteins in a biological sample can be captured using this method, the further analysis of the captured glycoproteins is practically limited by the method that can cleave glycoproteins or glycopeptides from the solid phase. Because there is a lack of efficient enzymes or chemicals that can specifically deglycosylate and/or release O-linked glycoproteins or glycopeptides from the solid phase, most of the studies have applied this method solely for N-linked glycoprotein analysis. PNGase F is the enzyme that can specifically release an N-glycosylated proteins or peptides (except those carrying α1→3 linked core fucose (38)) from its corresponding oligosaccharide groups. The hydrazide chemistry method is not only highly efficient in enriching N-linked glycoproteins or glycopeptides from a complex environment, but also allows great flexibility in its applications, such as capturing extracellular N-glycoproteins on live cells to monitor their abundant changes because of cell activation, differentiation, or other cellular activities (64). This method can be readily automated for analyzing a large quantity of samples.Recent studies have compared glycoprotein isolation methods. One study assessed lectin-based protocols and hydrophilic interaction chromatography for their performance in enriching glycoproteins and glycopeptides from serum (65). Other studies compared lectin affinity and hydrazide chemistry methods for their efficiency in isolating glycoproteins and glycopeptides from a complex biological sample (39, 66, 67). The methods are complementary in enriching glycoproteins because of their different mechanisms of glycoprotein capturing. When both methods were applied, it significantly improves the coverage of the glycoproteome, resulting in an increased number of glycoproteins identified. The lectin affinity method can be tailored to target glycoproteins with specific glycan structure(s) for isolation using different lectins, thus, affording flexibility for its application in glycoproteomic studies. The application of hydrazide chemistry method has been widely used for N-linked glycosylation study. The hydrazide chemistry essentially reacts with all the proteins with carbonyl groups, which may include glycoproteins with oxidized glycans (37, 40) and other oxidized proteins that carry carbonyl groups (6870). The high specificity of this method may mainly result from the specificity of PNGase F, the enzyme cleaving N-glycosidic bonds to release N-glycoproteins and peptides from the solid phase. This method affords high efficiency and specificity in enriching N-linked glycoproteins or glycopeptides from a complex sample, and can be easily incorporated into a proteomics workflow for integrated analysis. In addition to the lectin and hydrazide chemistry-based methods, it has been suggested that boronic acid-based solid phase extraction may also be useful for an overall glycoproteome enrichment (44, 45), on the basis of the evidence that boronic acid can form diester bonds with most glycans, including both N-linked and O-linked glycosylation (71).

Mass Spectrometric Analysis of Glycoproteome

Mass spectrometry, because of its high sensitivity and selectivity, has been one of the most versatile and powerful tools in glycoprotein analysis, to identify the glycoproteins, evaluate glycosylation sites, and elucidate the oligosaccharide structures (56, 72, 73). The utility of a top-down approach (intact protein based proteomics analysis) (74) for glycoprotein characterization in a complex sample is still technically challenging with the current technology. The most versatile and widely used current glycoproteomics methods are based on characterizing glycopeptides generated by the digestion of glycoproteins, analyzing either deglycosylated glycopeptides or intact glycopeptides with glycan attachment, as illustrated in Fig. 1.The direct analysis of intact glycopeptides with carbohydrate attachments is complicated by the mixed information obtained from the fragment ion spectra, which may include fragment ions from the peptide backbone, the carbohydrate group and the combinations of both. Although it is technically challenging to comprehensively analyze intact glycopeptides in a global scale for a complex biological sample, complementary information regarding peptide backbone and glycan structure can likely be obtained in a single measurement. Early work using collision-induced dissociation (CID)1 has identified a few key features that are characteristics of the fragmentation of glycopeptides, providing the basis for intact glycopeptide identification (7579). The analysis of intact glycopeptides has been carried out using a variety of different instruments, including electrospray ionization (EST)-based ion trap (IT) (8084), quadrupole ion trap (QIT) (8587), Fourier transform ion cyclotron resonance (FTICR) (31, 57, 88, 89), ion trap/time-of-flight (IT/TOF) (90, 91), and quadrupole/time-of-flight (Q/TOF) (9297); matrix-assisted laser desorption/ionization (MALDI) based Q/TOF (98100), quadrupole ion trap/time-of-flight (QIT/TOF) (86, 101, 102), and tandem time-of-flight (TOF/TOF) (81, 82, 101, 103105) mass spectrometers. In general, the CID generated MS/MS spectrum of a glycopeptide is dominated by B- and Y-type glycosidic cleavage ions (carbohydrate fragments) (106), and b- and y-type peptide fragments from the peptide backbone. However, the MS/MS fragmentation data obtained from different instruments can have pronounced difference in providing structure information on glycan and peptide backbone, depending on the experimental setting and instrumentation used for mass analysis, including ionization methods, collision techniques and mass analyzers. Low energy CID with electrospray ionization-based ion trap, Fourier transform-ion cyclotron resonance, and Q/TOF instrument predominantly generates fragments of glycosidic bonds. The increase of collision energy using Fourier transform-ion cyclotron resonance, and Q/TOF instruments result in the more efficient fragmentation of b- and y- ions from the peptide backbone. MALDI ionization generates predominantly singly charged precursor ions, which are more stable and usually fragmented using higher energies via CID or post-source decay (PSD), generating fragments from both the peptide backbone and the glycan (98100, 103, 107110). Although Q/TOF instruments have been widely used for intact glycopeptide characterization, one unique feature of the ion trap instrument is that it allows repeated ion isolation/CID fragmentation cycles, which can provide a wealth of complementary information to interpret the structure of a glycan moiety and peptide backbone (56, 86, 111). Recently, fragmentation techniques using different mechanisms from CID have been introduced and applied for glycopeptide analysis, including infrared multiphoton dissociation (IRMPD) (112115), electon-capture dissociation (ECD) (112120) and electron-transfer disassociation (ETD) (85, 121123). The application of infrared multiphoton dissociation and electon-capture dissociation is largely performed with Fourier transform-ion cyclotron resonance instruments. Complementary to CID fragmentation, electon-capture dissociation and electron-transfer disassociation tend to cleave the peptide backbone with no loss of the glycan moiety, providing specific information on localizing the glycosidic modification. More details regarding mass spectrometric analysis of intact glycopeptides can be found in recent reviews (56, 124). Although great efforts have been made to apply a variety of mass spectrometry techniques to study both N-linked (32, 56, 86, 87, 112114, 125130) and O-linked (90, 116, 119, 120, 130140) glycopeptides, the interpretation of the fragment spectrum of an intact glycopeptide still requires intensive manual assignment and evaluation. A recent study has demonstrated the feasibility to develop an automated workflow for analyzing intact glycopeptides in mixtures (141). In general, however, a high throughput, large scale profiling of intact glycopeptides in a complex sample still remains a challenge with current technology.The analysis of deglycosylated peptides requires the removal of glycan attachments from glycopeptides. Fortunately, for N-linked glycopeptides, the N-glycosidic bond can be specifically cleaved using the enzyme PNGase F, providing deglycosylated peptides, which can then be analyzed directly using shotgun proteomics. The PNGase F-catalyzed deglycosylation results in the conversion of asparagine to aspartic acid in the glycopeptide sequence, which introduces a mass difference of 0.9840 Da. Such distinct mass differences can be used to precisely map the N-linked glycosylation sites using high resolution mass spectrometers. Stable isotope labeling introduced by enzymatic cleavage of glycans in H218O has also been used to enhance the precise identification of N-glycosylation sites (33, 142, 143). The removal of O-linked glycans is less straightforward, most assays rely on chemical deglycosylation methods, such as trifluoromethansulfonic acid (144), hydrazinolysis (145), β-elimination (146), and periodate oxidation (35, 147). The application of these methods suffers from a variety of limitations, such as low specificity for O-linked glycosylation, degradation of the peptide backbone, and modifications of the amino acid residues—all of which can complicate or compromise O-linked glycoproteomics analysis in a complex sample. Most of the large scale glycoproteomics studies using the deglycosylation approach have been focused on N-glycoproteins, which are prevalent in blood and a rich source for biomarker discovery. O-glycosylation lacks a common core, consensus sequence, and universal enzyme that can specifically remove the glycans from the peptide backbone, thus, is more challenging to analyze for large scale profiling.Following deglycosylation, the glycopeptides can be treated and analyzed as stripped peptides using a shotgun proteomics pipeline. MS/MS fragment spectra with b-ions and y-ions generated from CID are searched against protein databases using search algorithms, such as SEQUEST (148), MASCOT (149), and X!tandem (150), and subsequently validated via statistical analysis (151154), to provide peptide and protein identifications with known false discovery rate. The N-glycosylation sites can be precisely mapped using the consensus sequence of Asn-X-Ser/Thr, in which asparagine is converted to aspartic acid following enzyme cleavage introducing a mass difference of 0.9840 Dalton. A variety of mass spectrometers have been used to analyze glycoproteins, in particular N-linked glycoproteins, in complex biological and clinical samples using the deglycosylation approach. These studies include electrospray ionization-based ion trap (3739, 41, 67, 155157), Orbitrap (158), Q/TOF (33, 35, 142, 155), triple quadrupole (159), Fourier transform-ion cyclotron resonance (64, 160); and MALDI based TOF/TOF (41, 161) and Q/TOF (37). Recently, an attempt was made to apply ion mobility-mass spectrometry (IM-MS) to characterize deglycosylated glycopeptides and the corresponding carbohydrates simultaneously (162) in a single measurement. The approach of analyzing deglycosylated glycopeptides makes it possible to utilize available proteomics technology for large-scale glycoproteome profiling, especially N-linked glycoproteins, in a high-throughput fashion.

Glycoproteomics Analysis in Blood and Other Bodily Fluids

An important target for blood-based diagnostic assays involves the detection and quantification of glycosylated proteins. Glycosylated proteins, especially N-linked glycoproteins, are ubiquitous among the proteins destined for extracellular environments (163), such as plasma or serum. A systematic and in-depth global profiling of the blood glycoproteome can provide fundamental knowledge for blood biomarker development, and is now possible with the development of glycoproteomics technologies. In the past few years, several large scale proteomics studies on profiling the glycoproteome of human plasma and serum have been reported (34, 35, 37, 38, 43, 61, 65, 164166), adding significant numbers of glycoproteins into the blood glycoproteome database. In one study (38), immunoaffinity subtraction and hydrazide chemistry were applied to enrich N-glycoproteins from human plasma. The captured plasma glycoproteins were subjected to two-dimensional liquid chromatography separation followed by tandem mass spectrometric analysis. A total of 2053 different N-glycopeptides were identified, covering 303 nonredundant glycoproteins, including many glycoproteins with low abundance in blood (38). In a different study, hydrazide chemistry-based solid phase extraction method was applied to enhance the detection of tissue-derived proteins in human plasma (167). Other studies have applied lectin affinity-based approaches to characterize the serum and plasma glycoproteome (34, 43, 166). These studies provide detailed identification regarding the individual N-glycosylation sites using high-resolution mass spectrometry. The efforts made in global profiling of glycoproteins in plasma and serum have not only greatly enhanced our understanding of the blood glycoproteome, but also have facilitated the development of new technologies that can be used for glycoprotein biomarker discovery. A variety of experimental designs and strategies for blood glycoprotein profiling have been applied for clinical disease studies, including prostate cancer (168), hepatocellular carcinoma (164, 168170), lung adenocarcinoma (61, 171), breast cancer (58, 165, 172), atopic dermatitis (169), ovarian cancer (173, 174), congenital disorders of glycosylation (175), and pancreatic cancer (156, 176). Most of these studies focused on the early stages of glycoprotein biomarker discovery and many of them exploited multilectin affinity techniques to isolate glycoproteins from serum or plasma.Glycoproteomics techniques have also been applied to study the glycoproteome of other bodily fluids. The complementary application of hydrazide chemistry-based solid phase extraction and lectin affinity method have led to the identification of 216 glycoproteins in human cerebrospinal fluid (CSF), including many low abundant ones (39). A hydrazide chemistry based study on human saliva has characterized 84 N-glycosylated peptides in 45 glycoproteins (177). The study on tear fluid identified 43 N-linked glycoproteins, including 19 proteins that have not been discovered in tear fluid previously (178). Other glycoproteomics studies on bodily fluids include N-glycoprotein profiling of lung adenocarcinoma pleural effusions (179), urine glycoprotein profiling (180), and urine glycoprotein signature identification for bladder cancer (181). In the urine glycoprotein profiling study, 150 annotated glycoproteins in addition to 43 predicted glycoproteins were identified (180). In our own study, 48 glycoproteins have so far been identified in pancreatic juice (unpublished data), adding complementary information to the pancreatic juice protein database (182184).

Glycoproteomics Analysis of Tissue and Cell Lysates

Protein glycosylation has been increasingly recognized as one of the prominent alterations involved in tumorigenesis, inflammation, and other disease states. The study of glycoproteins in cell and tissue carries great promise for defining biomarkers for diagnotic and therapeutic targets. The glycoproteomics studies in liver tissue (185, 186) and cell lines (187) have provided a fundamental understanding of the liver glycoproteome and identified protein candidates that are associated with highly metastatic liver cancer cells. In one of the studies, hydrazide chemistry and multiple enzyme digestion provided a complementary identification of 939 N-glycosylation sites covering 523 nonredundant glycoproteins in human liver tissue (185). Studies on ovarian cancer have focused on discovering putative glycoprotein biomarkers for improving diagnosis (173, 174) and therapeutic treatment (188). Glycoproteomics studies have also been carried out to study hepatocelluar carcinoma. Magnetic nanoparticle immobilized Concanavalin A was used to selectively enrich N-glycoproteins in a hepatocelluar carcinoma cell line leading to the identification of 184 glycosylation sites corresponding to 101 glycoproteins (189). In a different study, complementary methods of hydrophilic affinity and hydrazide chemistry were applied to investigate the secreted glycoproteins from a hepatocelluar carcinoma cell line, in which 300 different glycosylation sites within 194 glycoproteins were identified (190). While many of these studies focused on N-glycoproteins, mucin-type O-linked glycoproteins are the predominant forms of O-linked glycosylation and are difficult to analyze. A metabolic labeling method was developed to facilitate their identification in complex cell lysates using proteomic strategies (191).Cell surface and membrane proteins are particularly appealing for biomarker discovery, and many of them are glycosylated proteins. Both hydrazide chemistry- and lectin affinity-based approaches have been applied to specifically study cell surface and membrane N-glycoproteins that are associated with diseases, including colon carcinoma (192), breast cancer (158), and thyroid cancer (157). One study applied hydrazide chemistry to covalently label extracellular glycan moieties on live cells, providing highly specific and selective identification of cell surface N-glycoproteins (64). A complementary application of hydrazide chemistry and lectin affinity methods was demonstrated to profile cell membrane glycoproteins, significantly enhancing the glycoprotein identification (67).

Quantitative Glycoprotein Profiling

One of the major goals of clinical proteomics is to effectively identify dysregulated proteins that are specifically associated with a biological state, such as a disease. In the past decade, different quantitative proteomics techniques have been introduced and applied to study a wide variety of disease settings. These techniques are based on different mechanisms to facilitate mass spectrometric-based quantitative analysis, including stable isotopic or isobaric labeling using chemical reactions (e.g. ICAT and iTRAQ) (193195), metabolic incorporation (e.g. SILAC) (196) and enzymatic reactions (e.g. 18O labeling) (197, 198); as well as less quantitatively accurate label-free approaches (199, 200). The overview and comparison of these quantitative techniques can be found in several reports in the literature and are not discussed in this review. Most of these isotopic labeling techniques can be adapted and utilized for glycoproteomics analysis to quantitatively compare the glycoproteome of a diseased sample to a control, thus revealing the glycosylation occupancy of individual glycosylation sites that may be involved in a disease. In addition to the well-established labeling methods cited above, several more experimental labeling strategies have been described in the field of glycoproteomics. One study demonstrated the feasibility of using stable isotope labeled succinic anhydride for quantitative analysis of glycoproteins isolated from serum via hydrazide chemistry (37). In a different report, the heavy and light version of N-acetoxy-succinimide combining with lectin affinity selection was used to quantitatively profile serum glycopeptides in canine lymphoma and transitional cell carcinoma (201). Stable isotope labeled 2-nitrobenzenesulfenyl was also used for chemical labeling in a quantitative glycoprotein profiling study on the sera from patients with lung adenocarcinoma (202). O-Linked N-acetylglucosamine (O-GlcNAc) is an intracellular, reversible form of glycosylation that shares many features with phosphorylation (203). Studies have suggested that O-GlcNAc may play an important role in many biological processes (204). A quantitative study on O-GlcNAc glycosylation has been reported, in which a method termed quantitative isotopic and chemoenzymatic tagging (QUIC-Tag) was described using a biotin-avidin affinity strategy for O-GlcNAc glycopeptide enrichment and stable isotope-labeled formaldehyde for mass spectrometric quantification (205). Recently, the isobaric tag for relative and absolute quantitation (iTRAQ) technique, combined with different glycoprotein enrichment approaches, has been utilized in several quantitative glycoproteomics studies. In the study of hepatocellular carcinoma, N-linked glycoproteins were enriched from hepatocellular carcinoma patients and controls using multilectin column and then quantitatively compared using iTRAQ to reveal the differential proteins associated with hepatocellular carcinoma (206). In a different study, the approach of using narrow selectivity lectin affinity chromatography followed by iTRAQ labeling was demonstrated to selectively identify differential glycoproteins in plasma samples from breast cancer patients (165). Another study utilized hydrazide chemistry-based solid phase extraction and iTRAQ to investigate the tear fluid of patients with climatic droplet keratopathy in comparison of normal controls, identifying multiple N-glycosylation sites with differential occupancy associated with climatic droplet keratopathy (178).In addition to using chemical reactions to incorporate stable isotope tag for quantitative mass spectrometric analysis, 18O can be introduced into N-glycopeptides during enzymatic reactions, such as tryptic digestion (incorporation of two 18O into the peptide carboxyl-terminal) and PNGase F mediated hydrolysis (incorporation of one 18O into the asparagine of N-glycosylation sites (33)). Attempts have been made to apply this approach to identify differentially expressed N-glycosylation associated with ovarian cancer in serum (207). In a different approach, the SILAC technique allows incorporation of stable isotope-labeled amino acids into proteins during cell culturing process (196), and was applied to investigate the difference in cell surface N-glycoproteins among different cell types (64). A label-free approach has also been used for glycoproteomics profiling, including a method developed to profile intact glycopeptides in a complex sample (208) and a study that compares the plasma glycoproteome between psoriasis patients and healthy controls (209).

Targeted Glycoproteomics Analysis

Mass spectrometry based targeted proteomics has recently emerged as a multiplexed quantitative technique that affords highly specific and candidate-based detection of targeted peptides and proteins in a complex biological sample (18, 210214). The technique is based on the concept of stable isotope dilution utilizing stable isotope-labeled synthetic reference peptides, which precisely mimic their endogenous counterparts, to achieve targeted quantification (214). Such techniques can be applied to target specific glycoproteins or glycopeptides, to precisely quantify the status of candidate glycosylation sites and assess the glycosylation occupancy at the molecular level. However, it is technically impractical to use synthetic peptides to precisely mimic a large number of natural glycopeptides with intact a glycan moiety as internal standards because of the structure complexity and variation of the sugar chain. To overcome these technical obstacles, an alternative approach was proposed for targeted analysis of N-glycosylation occupancy, in which stable isotope-labeled peptides were synthesized to mimic the deglycosylated form of candidate glycopeptides as internal references (161). It is known that the deglycosylation step using PNGase F results in a conversion of asparagine to aspartic acid in the peptide sequence, introducing a mass difference of 0.9840 Da. This phenomenon was utilized to design a synthetic peptide to mimic the endogenous N-linked glycopeptide in its deglycosylation form with exact amino acid sequence of its endogenous counterpart and with 13C and 15N labeling on one of its amino acids (161). Therefore, each matched pair of reference and endogenous candidate glycopeptides should share the same chromatographic and mass spectrometric characteristics, and can only be distinguished by their mass difference and isotopic pattern because of isotopic labeling. This design conceptually ensures that the synthetic internal standard of a candidate glycopeptide will be detected simultaneously with its endogenous form under the same analytical conditions, thus, minimizing the systematic variation and providing reliable quantification (214). The strategy for targeted glycoproteomics analysis is schematically illustrated in Fig. 2.Open in a separate windowFig. 2.Targeted analysis of N-glycopeptides.The targeted glycoproteomics technique was first demonstrated to analyze N-glycopeptides that were extracted from human serum using an integrated pipeline combining a hydrazide chemistry-based solid phase extraction method and a data-driven liquid chromatography MALDI TOF/TOF mass spectrometric analysis to quantify 21 N-glycopeptides in human serum (161). A similar mass spectrometric platform was then applied in a different study to assess a subset of glycoprotein biomarker candidates in the sera from prostate cancer patients (215). The targeted glycoproteomics analysis has also been demonstrated using a triple Q/linear ion trap instrument with the selected reaction monitoring (also referred to as multiple reaction monitoring) technique for highly sensitive targeted detection of N-glycoproteins in plasma (159). The technique was applied to detect tissue inhibitor of metalloproteinase 1 (TIMP1), an aberrant glycoprotein associated with colorectal cancer, in the sera of colorectal cancer patients (216) using a tandem enrichment strategy, combing lectin glycoprotein enrichment followed by the method of stable isotope standards and capture by antipeptide antibodies (SISCAPA), to enhance the detection of tissue inhibitor of metalloproteinase 1 (216). These studies demonstrate an integrated pipeline for candidate-based glycoproteomics analysis with precise mapping of targeted N-linked motifs and absolute quantification of the glycoprotein targets in a complex biological sample. Such targeted glycoproteomics can reach a detection sensitivity at the nanogram per milliliter level for serum and plasma detection (159, 214216).

Concluding Remarks

The major challenge for a comprehensive glycoproteomics analysis arises not only from the enormous complexity and nonlinear dynamic range in protein constituent in a clinical sample, but also the profound biological intricacy within the molecule of a glycoprotein, involving the flexibility in glycan structures and the complex linkage with the corresponding protein. In the past decade, significant efforts have been made to structurally or quantitatively characterize the glycoproteome of a variety of biological samples, and to investigate the significant glycoproteins in a wide assortment of diseases. Shotgun proteomics-based techniques are still the most effective and versatile approach in glycoproteomics analysis, allowing high throughput and detailed analysis on individual glycosylation sites. Although glycoproteomics is quickly emerging as an important technique for clinical proteomics study and biomarker discovery, a comprehensive, quantitative glycoproteomics analysis in a complex biological sample still remains challenging. It is anticipated that with the continued evolution in mass spectrometry, separation technology, and bioinformatics many of the technical limitations associated with current glycoproteomics may be transient. There is no doubt that glycoproteomics is playing an increasingly important role in biomarker discovery and clinical study.  相似文献   

6.
7.
8.
9.
Virulence of the gastric pathogen Helicobacter pylori (Hp) is directly linked to the pathogen''s ability to glycosylate proteins; for example, Hp flagellin proteins are heavily glycosylated with the unusual nine-carbon sugar pseudaminic acid, and this modification is absolutely essential for Hp to synthesize functional flagella and colonize the host''s stomach. Although Hp''s glycans are linked to pathogenesis, Hp''s glycome remains poorly understood; only the two flagellin glycoproteins have been firmly characterized in Hp. Evidence from our laboratory suggests that Hp synthesizes a large number of as-yet unidentified glycoproteins. Here we set out to discover Hp''s glycoproteins by coupling glycan metabolic labeling with mass spectrometry analysis. An assessment of the subcellular distribution of azide-labeled proteins by Western blot analysis indicated that glycoproteins are present throughout Hp and may therefore serve diverse functions. To identify these species, Hp''s azide-labeled glycoproteins were tagged via Staudinger ligation, enriched by tandem affinity chromatography, and analyzed by multidimensional protein identification technology. Direct comparison of enriched azide-labeled glycoproteins with a mock-enriched control by both SDS-PAGE and mass spectrometry-based analyses confirmed the selective enrichment of azide-labeled glycoproteins. We identified 125 candidate glycoproteins with diverse biological functions, including those linked with pathogenesis. Mass spectrometry analyses of enriched azide-labeled glycoproteins before and after cleavage of O-linked glycans revealed the presence of Staudinger ligation-glycan adducts in samples only after beta-elimination, confirming the synthesis of O-linked glycoproteins in Hp. Finally, the secreted colonization factors urease alpha and urease beta were biochemically validated as glycosylated proteins via Western blot analysis as well as by mass spectrometry analysis of cleaved glycan products. These data set the stage for the development of glycosylation-based therapeutic strategies, such as new vaccines based on natively glycosylated Hp proteins, to eradicate Hp infection. Broadly, this report validates metabolic labeling as an effective and efficient approach for the identification of bacterial glycoproteins.Helicobacter pylori (Hp)1 infection poses a significant health risk to humans worldwide. The Gram-negative, pathogenic bacterium Hp colonizes the gastric tract of more than 50% of humans (1). Approximately 15% of infected individuals develop duodenal ulcers and 1% of infected individuals develop gastric cancer (2). Current treatment to clear infection requires “triple therapy” (3), a combination of multiple antibiotics that is often associated with negative side effects (4). Because of poor patient compliance and the evolution of antibiotic resistance, existing antibiotics are no longer effective at eradicating Hp infection (4). New treatment methods are needed to eliminate Hp from the human gastric tract.Recent work has focused on gaining insights into the pathogenesis of Hp to aid the development of new treatments. The most recent findings in this area have conclusively revealed that glycosylation of proteins in Hp is required for pathogenesis. Hp use complex flagella, comprised of flagellin proteins, to navigate the host''s gastric mucosa (5, 6). The flagellin proteins are heavily glycosylated with the unusual nine-carbon sugar pseudaminic acid, found exclusively in mucosal-associated pathogens (Hp (7), Campylobacter jejuni (8) and Pseudomonas aeruginosa (9)). This modification is absolutely essential for the formation of functional flagella on Hp (7, 10). Deletion of any one of the enzymes in the pseudaminic acid biosynthetic pathway results in Hp that lack flagella, are nonmotile, and are unable to colonize the host''s stomach (7). Although pseudaminic acid is critical for Hp virulence, it is absent from humans (11, 12). Therefore, insights into Hp''s pathogenesis have revealed that Hp''s glycan pseudaminic acid is a bona fide target of therapeutic intervention. This is one of a number of examples linking protein glycosylation to virulence in medically significant bacterial pathogens (13, 14).Despite these findings, Hp''s glycome remains poorly understood overall. Only the two flagellin glycoproteins have been firmly characterized in Hp (7) to date. Nine other candidate glycoproteins have been identified in Hp, but their glycosylation status has not been biochemically confirmed (15). The relative paucity of information regarding Hp''s glycoproteins is due in part to the previously held belief that protein glycosylation could not occur in bacteria (13, 16, 17). However, even after Szymanski (18, 19), Koomey (20), Guerry (21), Logan (7), Comstock and others (13, 16, 17) disproved this belief by firmly establishing the synthesis of glycoproteins in bacteria, the study of bacterial glycoproteins has presented unique challenges for analytical study (14, 22). For example, the unusual structures of bacterial glycans, which often contain amino- and deoxy-carbohydrates exclusively found in bacteria (12, 2325), hampers their identification using existing tools. Though methods such as the use of glycan-binding reagents (20, 24, 26, 27) and periodic acid/hydrazide glycan labeling (15) have successfully detected glycoproteins in a range of bacteria, they present limitations. Glycan binding-based methods are often limited because of the unavailability of lectins or antibodies with binding specificity for glycosylated proteins in the bacteria of interest (14, 22). Periodic acid/hydrazide-based labeling is plagued by a lack of specificity for glycosylated proteins (15). Thus, an efficient and robust approach to discover Hp''s glycoproteins is needed.In previous work, we established that the chemical technique known as metabolic oligosaccharide engineering (MOE), which was developed by Bertozzi (28, 29), Reutter (30), and others for the study of mammalian glycoproteins, is a powerful approach to label and detect Hp''s glycoproteins (31). Briefly, Hp metabolically processes the unnatural, azide-containing sugar peracetylated N-azidoacetylglucosamine (Ac4GlcNAz) (32), an analog of the common metabolic precursor N-acetylglucosamine (GlcNAc), into cellular glycoproteins (Fig. 1). Elaboration of azide-labeled glycoproteins via Staudinger ligation (33) with a phosphine probe conjugated to a FLAG peptide (Phos-FLAG) (34) followed by visualization with an anti-FLAG antibody (Fig. 1) revealed a glycoprotein fingerprint containing a large number of as-yet unidentified Hp glycoproteins that merit further investigation (31).Open in a separate windowFig. 1.Metabolic oligosaccharide engineering facilitates labeling and detection of Hp''s glycoproteins. Supplementation of Hp with Ac4GlcNAz leads to metabolic labeling of Hp''s N-linked and O-linked glycoproteins with azides. Azide-modified glycoproteins covalently labeled with Phos-FLAG can be detected via Western blot analysis with anti-FLAG antibody to yield Hp''s glycoprotein fingerprint, which contains a large number of as-yet unidentified glycoproteins.Here we describe a glycoproteomic identification strategy for the selective detection, isolation, and discovery of Hp''s glycoproteins. In particular, we demonstrate that glycan metabolic labeling coupled with mass spectrometry analysis is an efficient and robust chemical approach to identify novel glycoproteins in Hp. This work characterizes glycosylated virulence factors in Hp, thus opening the door to new vaccination and antibiotic therapies to eradicate Hp infection. Broadly, this work validates metabolic oligosaccharide engineering as a complementary method to discover bacterial glycoproteins.  相似文献   

10.
11.
The opportunistic human pathogen Acinetobacter baumannii is a concern to health care systems worldwide because of its persistence in clinical settings and the growing frequency of multiple drug resistant infections. To combat this threat, it is necessary to understand factors associated with disease and environmental persistence of A. baumannii. Recently, it was shown that a single biosynthetic pathway was responsible for the generation of capsule polysaccharide and O-linked protein glycosylation. Because of the requirement of these carbohydrates for virulence and the non-template driven nature of glycan biogenesis we investigated the composition, diversity, and properties of the Acinetobacter glycoproteome. Utilizing global and targeted mass spectrometry methods, we examined 15 strains and found extensive glycan diversity in the O-linked glycoproteome of Acinetobacter. Comparison of the 26 glycoproteins identified revealed that different A. baumannii strains target similar protein substrates, both in characteristics of the sites of O-glycosylation and protein identity. Surprisingly, glycan micro-heterogeneity was also observed within nearly all isolates examined demonstrating glycan heterogeneity is a widespread phenomena in Acinetobacter O-linked glycosylation. By comparing the 11 main glycoforms and over 20 alternative glycoforms characterized within the 15 strains, trends within the glycan utilized for O-linked glycosylation could be observed. These trends reveal Acinetobacter O-linked glycosylation favors short (three to five residue) glycans with limited branching containing negatively charged sugars such as GlcNAc3NAcA4OAc or legionaminic/pseudaminic acid derivatives. These observations suggest that although highly diverse, the capsule/O-linked glycan biosynthetic pathways generate glycans with similar characteristics across all A. baumannii.Acinetobacter baumannii is an emerging opportunistic pathogen of increasing significance to health care institutions worldwide (13). The growing number of identified multiple drug resistant (MDR)1 strains (24), the ability of isolates to rapidly acquire resistance (3, 4), and the propensity of this agent to survive harsh environmental conditions (5) account for the increasing number of outbreaks in intensive care, burn, or high dependence health care units since the 1970s (25). The burden on the global health care system of MDR A. baumannii is further exacerbated by standard infection control measures often being insufficient to quell the spread of A. baumannii to high risk individuals and generally failing to remove A. baumannii from health care institutions (5). Because of these concerns, there is an urgent need to identify strategies to control A. baumannii as well as understand the mechanisms that enable its persistence in health care environments.Surface glycans have been identified as key virulence factors related to persistence and virulence within the clinical setting (68). Acinetobacter surface carbohydrates were first identified and studied in A. venetianus strain RAG-1, leading to the identification of a gene locus required for synthesis and export of the surface carbohydrates (9, 10). These carbohydrate synthesis loci are variable yet ubiquitous in A. baumannii (11, 12). Comparison of 12 known capsule structures from A. baumannii with the sequences of their carbohydrate synthesis loci has provided strong evidence that these loci are responsible for capsule synthesis with as many as 77 distinct serotypes identified by molecular serotyping (11). Because of the non-template driven nature of glycan synthesis, the identification and characterization of the glycans themselves are required to confirm the true diversity. This diversity has widespread implications for Acinetobacter biology as the resulting carbohydrate structures are not solely used for capsule biosynthesis but can be incorporated and utilized by other ubiquitous systems, such as O-linked protein glycosylation (13, 14).Although originally thought to be restricted to species such as Campylobacter jejuni (15, 16) and Neisseria meningitidis (17), bacterial protein glycosylation is now recognized as a common phenomenon within numerous pathogens and commensal bacteria (18, 19). Unlike eukaryotic glycosylation where robust and high-throughput technologies now exist to enrich (2022) and characterize both the glycan and peptide component of glycopeptides (2325), the diversity (glycan composition and linkage) within bacterial glycosylation systems makes few technologies broadly applicable to all bacterial glycoproteins. Because of this challenge a deeper understanding of the glycan diversity and substrates of glycosylation has been largely unachievable for the majority of known bacterial glycosylation systems. The recent implementation of selective glycopeptide enrichment methods (26, 27) and the use of multiple fragmentation approaches (28, 29) has facilitated identification of an increasing number of glycosylation substrates independent of prior knowledge of the glycan structure (3033). These developments have facilitated the undertaking of comparative glycosylation studies, revealing glycosylation is widespread in diverse genera and far more diverse then initially thought. For example, Nothaft et al. were able to show N-linked glycosylation was widespread in the Campylobacter genus and that two broad groupings of the N-glycans existed (34).During the initial characterization of A. baumannii O-linked glycosylation the use of selective enrichment of glycopeptides followed by mass spectrometry analysis with multiple fragmentation technologies was found to be an effective means to identify multiple glycosylated substrates in the strain ATCC 17978 (14). Interestingly in this strain, the glycan utilized for protein modification was identical to a single subunit of the capsule (13) and the loss of either protein glycosylation or glycan synthesis lead to decreases in biofilm formation and virulence (13, 14). Because of the diversity in the capsule carbohydrate synthesis loci and the ubiquitous distribution of the PglL O-oligosaccharyltransferase required for protein glycosylation, we hypothesized that the glycan variability might be also extended to O-linked glycosylation. This diversity, although common in surface carbohydrates such as the lipopolysaccharide of numerous Gram-negative pathogens (35), has only recently been observed within bacterial proteins glycosylation system that are typically conserved within species (36) and loosely across genus (34, 37).In this study, we explored the diversity within the O-linked protein glycosylation systems of Acinetobacter species. Our analysis complements the recent in silico studies of A. baumannii showing extensive glycan diversity exists in the carbohydrate synthesis loci (11, 12). Employing global strategies for the analysis of glycosylation, we experimentally demonstrate that the variation in O-glycan structure extends beyond the genetic diversity predicted by the carbohydrate loci alone and targets proteins of similar properties and identity. Using this knowledge, we developed a targeted approach for the detection of protein glycosylation, enabling streamlined analysis of glycosylation within a range of genetic backgrounds. We determined that; O-linked glycosylation is widespread in clinically relevant Acinetobacter species; inter- and intra-strain heterogeneity exist within glycan structures; glycan diversity, although extensive results in the generation of glycans with similar properties and that the utilization of a single glycan for capsule and O-linked glycosylation is a general feature of A. baumannii but may not be a general characteristic of all Acinetobacter species such as A. baylyi.  相似文献   

12.
13.
A decoding algorithm is tested that mechanistically models the progressive alignments that arise as the mRNA moves past the rRNA tail during translation elongation. Each of these alignments provides an opportunity for hybridization between the single-stranded, -terminal nucleotides of the 16S rRNA and the spatially accessible window of mRNA sequence, from which a free energy value can be calculated. Using this algorithm we show that a periodic, energetic pattern of frequency 1/3 is revealed. This periodic signal exists in the majority of coding regions of eubacterial genes, but not in the non-coding regions encoding the 16S and 23S rRNAs. Signal analysis reveals that the population of coding regions of each bacterial species has a mean phase that is correlated in a statistically significant way with species () content. These results suggest that the periodic signal could function as a synchronization signal for the maintenance of reading frame and that codon usage provides a mechanism for manipulation of signal phase.[1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32]  相似文献   

14.
Human CD36 is a class B scavenger receptor expressed in a variety of cell types such as macrophage and adipocytes. This plasma membrane glycoprotein has a wide range of ligands including oxidized low density lipoprotein and long chain fatty acids which involves the receptor in diseases such as atherosclerosis and insulin resistance. CD36 is heavily modified post-translationally by N-linked glycosylation, and 10 putative glycosylation sites situated in the large extracellular loop of the protein have been identified; however, their utilization and role in the folding and function of the protein have not been characterized. Using mass spectrometry on purified and peptide N-glycosidase F-deglycosylated CD36 and also by comparing the electrophoretic mobility of different glycosylation site mutants, we have determined that 9 of the 10 sites can be modified by glycosylation. Flow cytometric analysis of the different glycosylation mutants expressed in mammalian cells established that glycosylation is necessary for trafficking to the plasma membrane. Minimally glycosylated mutants that supported trafficking were identified and indicated the importance of carboxyl-terminal sites Asn-247, Asn-321, and Asn-417. However, unlike SRBI, no individual site was found to be essential for proper trafficking of CD36. Surprisingly, these minimally glycosylated mutants appear to be predominantly core-glycosylated, indicating that mature glycosylation is not necessary for surface expression in mammalian cells. The data also show that neither the nature nor the pattern of glycosylation is relevant to binding of modified low density lipoprotein.Human CD36, originally identified in platelets as glycoprotein IV (1), is a class B scavenger receptor localized to the plasma membrane. It is not expressed ubiquitously but is present in a variety of different cells and tissue types including epithelial cells (2), macrophages (3), endothelial cells of the microvasculature (4), and smooth muscle (5). Its function is complex, and its involvement in different disease scenarios, such as cancer (6), atherosclerosis (3, 7, 8), malaria (9), and insulin resistance (10), most likely reflects the interaction of the receptor with a particular ligand in a specific cell type. For example, CD36 expressed in monocytic macrophages functions as a scavenger receptor for the uptake of oxidized LDL2 (3, 11). Under certain physiological conditions, this results in the lipid loading of macrophages at the site of tissue damage in the arterial wall, leading to foam cell formation and plaque development, a key early stage in the pathogenesis of atherosclerosis (8, 12). In fat and muscle cells, CD36 plays an essential role in lipid homeostasis by uptake of long chain fatty acids (13). In this case CD36 deficiency has been linked to disorders in lipid metabolism, giving rise to increased incidences of insulin resistance and cardiomyopathies (11, 14, 15).Although much is known about the function of CD36, less is known about its structure. CD36 has no bacterial homologues but is a member of a protein family that also includes the mammalian proteins LIMPII (16), CLA-1 (17), SRBI (18), and the Drosophila proteins Croquemort (19) and emp (20). The sequence of 471 amino acids has two short hydrophobic regions at the carboxyl and amino termini separated by a large hydrophilic region (21); however, the topology of the protein is unclear with both ditopic (22) and type I (23) topological models proposed. Both are consistent in predicting that the large hydrophilic region is extracellular, which is clearly supported by epitope mapping studies (24). The protein is heavily modified post-translationally. The six extracellular cysteines, which are highly conserved within the orthologous CD36 subfamily, have been shown to be linked by disulfide bonds in bovine Cd36 (25), and the remaining four cysteines, two at each terminus, are palmitoylated (26), lending credence to the ditopic topological model.CD36 is also modified by N-linked glycosylation, which accounts for the observation that the protein migrates with an apparent molecular mass of 78–94 kDa on SDS-PAGE (4, 27) despite a theoretical mass for the polypeptide of 53 kDa. N-Linked glycosylation is a common modification of extracellular and secreted proteins, and defects in the glycosylation pathways lead to a wide range of serious diseases known collectively as congenital disorders of glycosylation (28). Glycosylation can be important for correct folding of proteins (29, 30) either by directly inducing and/or stabilizing the tertiary fold of the polypeptide (31) or via an affinity for lectin chaperones such as calnexin or calreticulin (32). Glycosylation has also been shown to be important for the trafficking of certain glycoproteins through affinity for lectin transport machinery (33). The glycosylation status of bovine Cd36 has already been determined with all eight putative sites shown to be glycosylated (34). Human and bovine CD36 are 83% identical (93% when similar residues are included) and share 7 glycosylation sites (human has 10 putative glycosylation sites). In the related mouse SRBI, which is 33% identical (54% similar) to human CD36, there are 11 putative N-linked glycosylation sites, only 3 of which are shared with the human protein. Site-directed mutagenesis of each of the 11 sites independently in SRBI in an otherwise wild type protein indicates that all are glycosylated, with two (Asn-108 and Asn-173) important for either trafficking or folding. Mutagenesis of either of these two residues resulted in very little cell surface expression of the protein (35); however, neither site is conserved in human CD36.To gain further understanding of the role of glycosylation of CD36, we used mutagenesis and biophysical analysis (mass spectrometry and gel electrophoresis) to identify unequivocally which glycosylation sites are occupied in human CD36. Antibody and ligand binding studies with these mutant proteins also provided insights into the role of glycosylation and site occupancy in the trafficking and function of the protein.  相似文献   

15.
Decomposing a biological sequence into its functional regions is an important prerequisite to understand the molecule. Using the multiple alignments of the sequences, we evaluate a segmentation based on the type of statistical variation pattern from each of the aligned sites. To describe such a more general pattern, we introduce multipattern consensus regions as segmented regions based on conserved as well as interdependent patterns. Thus the proposed consensus region considers patterns that are statistically significant and extends a local neighborhood. To show its relevance in protein sequence analysis, a cancer suppressor gene called p53 is examined. The results show significant associations between the detected regions and tendency of mutations, location on the 3D structure, and cancer hereditable factors that can be inferred from human twin studies.[1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27]  相似文献   

16.
Small cell lung cancer (SCLC) is an aggressive type of lung cancer, and the detection of SCLCs at an early stage is necessary for successful therapy and for improving cancer survival rates. Fucosylation is one of the most common glycosylation-based modifications. Increased levels of fucosylation have been reported in a number of pathological conditions, including cancers. In this study, we aimed to identify and validate the aberrant and selective fucosylated glycoproteins in the sera of patients with SCLC. Fucosylated glycoproteins were enriched by the Aleuria aurantia lectin column after serum albumin and IgG depletion. In a narrowed down and comparative data analysis of both label-free proteomics and isobaric peptide-tagging chemistry iTRAQ approaches, the fucosylated glycoproteins were identified as up- or down-regulated in the sera of limited disease and extensive disease stage patients with SCLC. Verification was performed by multiple reaction monitoring-mass spectrometry to select reliable markers. Four fucosylated proteins, APCS, C9, SERPINA4, and PON1, were selected and subsequently validated by hybrid A. aurantia lectin ELISA (HLE) and Western blotting. Compared with Western blotting, the HLE analysis of these four proteins produced more optimal diagnostic values for SCLC. The PON1 protein levels were significantly reduced in the sera of patients with SCLC, whereas the fucosylation levels of PON1 were significantly increased. Fucosylated PON1 exhibited an area under curve of 0.91 for the extensive disease stage by HLE, whereas the PON1 protein levels produced an area under curve of 0.82 by Western blot. The glycan structural analysis of PON1 by MS/MS identified a biantennary fucosylated glycan modification consisting of a core + 2HexNAc + 1Fuc at increased levels in the sera of patients with SCLC. In addition, the PON1 levels were decreased in the sera of the Lewis lung carcinoma lung cancer mouse model that we examined. Our data suggest that fucosylated protein biomarkers, such as PON1, and their fucosylation levels and patterns can serve as diagnostic and prognostic serological markers for SCLC.Lung cancer is the most common cause of cancer death worldwide, with over one million cases annually (World Health Organization, economics of tobacco control). Lung cancer can be divided into four major histological subtypes as follows: adenocarcinoma, squamous cell carcinoma, large cell carcinoma, and small cell carcinoma (1). Small cell lung cancer (SCLC)1 is distinguishable from the other three histological subtypes (non-SCLC) by its distinct histological appearance (1, 2). SCLC is a neuroendocrine lung cancer subtype, which accounts for ∼20% of all lung cancer cases (3). SCLC is an aggressive malignancy that exhibits early metastatic spread and a high relapse rate. Thus, despite current advanced therapeutic strategies, including chemotherapy and radiation therapy, the 5-year survival rates for SCLC remain between 5 and 10% (4, 5). As SCLC is generally metastasized by the time of diagnosis (6), current diagnostic and prognostic workups for SCLC, such as chest x-rays and computed tomography, are far from efficient in terms of early detection. Therefore, the development of novel diagnostic techniques to identify SCLC is important for the earlier diagnosis of primary or recurring cancers to facilitate more effective treatments and improved prognosis (6). The identification of novel serum biomarkers is a prominent solution for the convenient and easy diagnosis and early detection of cancer, and a combination of markers is expected to enhance sensitivity and specificity.Glycosylation is one of the major post-translational modifications of proteins for cellular function. Approximately 50% of human serum proteins, including secretory and membrane-bound proteins, are suggested to exhibit various N-linked glycosylation patterns (7). Such glycan structures of proteins during carcinogenesis can affect various aspects of the biological behaviors of tumor cells (811). In fact, altered glycosylation patterns have long been recognized as hallmarks of cancer progression, in which tumor-specific glycoproteins are actively involved in neoplastic progression and metastasis (1214). The sensitive detection of such disease-associated glycosylation changes and abnormalities can become a unique landmark to develop diagnostic and prognostic glycoprotein biomarkers.Among the carbohydrate structures on glycoproteins that are altered during carcinogenesis, much interest has been focused on the terminal monosaccharides, fucose and sialic acid (15, 16). Such terminal structures of monosaccharides are known to be involved in biological functions, including cellular recognition, fertilization, and development (17). Fucose is a constituent of the terminal structure of glycan chains and is associated with cancer and inflammation (10). The altered fucosylation of N-linked glycans has been reported in various cancers such as prostate cancer, breast cancer, liver cancer, ovarian cancer, and pancreatic cancer (18). The biological role of fucosylation in cancer is associated with cancer metastasis, tumor immune surveillance, modification of growth factor receptors, and cancer cell interaction with adhesion molecules (19). Carbohydrate antigen 19-9 and α-fetoprotein (AFP)-L3 fraction, which are fucosylated glycoproteins, have been used as tumor markers in the sera of patients with pancreatic and liver cancers, respectively (20). The fucosylated form of AFP (AFP-L3) has demonstrated more robust diagnostic power than AFP itself (21). Therefore, the analysis of specific glycosylated serum glycoproteins represents an important approach for the development of cancer diagnostic biomarkers.Glycoproteomic technologies, such as mass spectrometry-based approaches, have significantly improved biomarker discovery. Thus, in this study, we performed a proteomic screening of fucosylated glycoproteins associated with SCLC. We found that altered fucosylated glycoproteins, specifically the altered fucosylated glycan patterns of PON1, can serve as potential diagnostic and prognostic biomarkers for SCLC.  相似文献   

17.
18.
Endogenous regeneration and repair mechanisms are responsible for replacing dead and damaged cells to maintain or enhance tissue and organ function, and one of the best examples of endogenous repair mechanisms involves skeletal muscle. Although the molecular mechanisms that regulate the differentiation of satellite cells and myoblasts toward myofibers are not fully understood, cell surface proteins that sense and respond to their environment play an important role. The cell surface capturing technology was used here to uncover the cell surface N-linked glycoprotein subproteome of myoblasts and to identify potential markers of myoblast differentiation. 128 bona fide cell surface-exposed N-linked glycoproteins, including 117 transmembrane, four glycosylphosphatidylinositol-anchored, five extracellular matrix, and two membrane-associated proteins were identified from mouse C2C12 myoblasts. The data set revealed 36 cluster of differentiation-annotated proteins and confirmed the occupancy for 235 N-linked glycosylation sites. The identification of the N-glycosylation sites on the extracellular domain of the proteins allowed for the determination of the orientation of the identified proteins within the plasma membrane. One glycoprotein transmembrane orientation was found to be inconsistent with Swiss-Prot annotations, whereas ambiguous annotations for 14 other proteins were resolved. Several of the identified N-linked glycoproteins, including aquaporin-1 and β-sarcoglycan, were found in validation experiments to change in overall abundance as the myoblasts differentiate toward myotubes. Therefore, the strategy and data presented shed new light on the complexity of the myoblast cell surface subproteome and reveal new targets for the clinically important characterization of cell intermediates during myoblast differentiation into myotubes.Endogenous regeneration and repair mechanisms are responsible for replacing dead and damaged cells to maintain or enhance tissue and organ function. One of the best examples of endogenous repair mechanisms involves skeletal muscle, which has innate regenerative capacity (for reviews, see Refs. 14). Skeletal muscle repair begins with satellite cells, a heterogeneous population of mitotically quiescent cells located in the basal lamina that surrounds adult skeletal myofibers (5, 6), that, when activated, rapidly proliferate (7). The progeny of activated satellite cells, known as myogenic precursor cells or myoblasts, undergo several rounds of division prior to withdrawal from the cell cycle. This is followed by fusion to form terminally differentiated multinucleated myotubes and skeletal myofibers (7, 8). These cells effectively repair or replace damaged cells or contribute to an increase in skeletal muscle mass.The molecular mechanisms that regulate differentiation of satellite cells and myoblasts toward myofibers are not fully understood, although it is known that the cell surface proteome plays an important biological role in skeletal muscle differentiation. Examples include how cell surface proteins modulate myoblast elongation, orientation, and fusion (for a review, see Ref. 8). The organization and fusion of myoblasts is mediated, in part, by cadherins (for reviews, see Refs. 9 and 10), which enhance skeletal muscle differentiation and are implicated in myoblast fusion (11). Neogenin, another cell surface protein, is also a likely regulator of myotube formation via the netrin ligand signal transduction pathway (12, 13), and the family of sphingosine 1-phosphate receptors (Edg receptors) are known key signal transduction molecules involved in regulating myogenic differentiation (1417). Given the important role of these proteins, identifying and characterizing the cell surface proteins present on myoblasts in a more comprehensive approach could provide insights into the molecular mechanisms involved in skeletal muscle development and repair. The identification of naturally occurring cell surface proteins (i.e. markers) could also foster the enrichment and/or characterization of cell intermediates during differentiation that could be useful therapeutically.Although it is possible to use techniques such as flow cytometry, antibody arrays, and microscopy to probe for known proteins on the cell surface in discrete populations, these methods rely on a priori knowledge of the proteins present on the cell surface and the availability/specificity of an antibody. Proteomics approaches coupled with mass spectrometry offer an alternative approach that is antibody-independent and allows for the de novo discovery of proteins on the surface. One approach, which was used in the current study, exploits the fact that a majority of the cell surface proteins are glycosylated (18). The method uses hydrazide chemistry (19) to immobilize and enrich for glycoproteins/glycopeptides, and previous studies using this chemistry have successfully identified soluble glycoproteins (2024) as well as cell surface glycoproteins (2528). A recently optimized hydrazide chemistry strategy by Wollscheid et al. (29) termed cell surface capturing (CSC)1 technology, reports the ability to identify cell surface (plasma membrane) proteins specifically with little (<15%) contamination from non-cell surface proteins. The specificity stems from the fact that the oligosaccharide structure is labeled using membrane-impermeable reagents while the cells are intact rather than after cell lysis. Consequently, only extracellular oligosaccharides are labeled and subsequently captured. Utilizing information regarding the glycosylation site then allows for a rapid elimination of nonspecifically captured proteins (i.e. non-cell surface proteins) during the data analysis process, a feature that makes this approach unique to methods where no label or tag is used. Additionally, the CSC technology provides information about glycosylation site occupancy (i.e. whether a potential N-linked glycosylation site is actually glycosylated), which is important for determining the protein orientation within the membrane and, therefore, antigen selection and antibody design.To uncover information about the cell surface of myoblasts and to identify potential markers of myoblast differentiation, we used the CSC technology on the mouse myoblast C2C12 cell line model system (30, 31). This adherent cell line derived from satellite cells has routinely been used as a model for skeletal muscle development (e.g. Refs. 1, 32, and 33), skeletal muscle differentiation (e.g. Refs. 3436), and studying muscular dystrophy (e.g. Refs. 3739). Additionally, these cells have been used in cell-based therapies (e.g. Refs. 4042). Using the CSC technology, 128 cell surface N-linked glycoproteins were identified, including several that were found to change in overall abundance as the myoblasts differentiate toward myotubes. The current data also confirmed the occupancy of 235 N-linked glycosites of which 226 were previously unconfirmed. The new information provided by the current study is expected to facilitate the development of useful tools for studying the differentiation of myoblasts toward myotubes.  相似文献   

19.
Disruption of Golgi α-mannosidase II activity can result in type II congenital dyserythropoietic anemia and induce lupus-like autoimmunity in mice. Here, we isolated a mutant human embryonic kidney (HEK) 293T cell line called Lec36, which displays sensitivity to ricin that lies between the parental HEK 293T cells, in which the secreted and membrane-expressed proteins are dominated by complex-type glycosylation, and 293S Lec1 cells, which produce only oligomannose-type N-linked glycans. Stem cell marker 19A was transiently expressed in the HEK 293T Lec36 cells and in parental HEK 293T cells with and without the potent Golgi α-mannosidase II inhibitor, swainsonine. Negative ion nano-electrospray ionization mass spectra of the 19A N-linked glycans from HEK 293T Lec36 and swainsonine-treated HEK 293T cells were qualitatively indistinguishable and, as shown by collision-induced dissociation spectra, were dominated by hybrid-type glycosylation. Nucleotide sequencing revealed mutations in each allele of MAN2A1, the gene encoding Golgi α-mannosidase II: a point mutation that mapped to the active site was found in one allele, and an in-frame deletion of 12 nucleotides was found in the other allele. Expression of the wild type but not the mutant MAN2A1 alleles in Lec36 cells restored processing of the 19A reporter glycoprotein to complex-type glycosylation. The Lec36 cell line will be useful for expressing therapeutic glycoproteins with hybrid-type glycans and as a sensitive host for detecting mutations in human MAN2A1 causing type II congenital dyserythropoietic anemia.Mammalian N-linked glycosylation is characterized by significant chemical heterogeneity generated by an array of competing glycosidases and glycosyltransferases (1). The structural analysis of recombinant glycoproteins, such as human erythropoietin (2, 3), has illustrated the capacity of mammalian expression systems for generating diverse N-linked glycans.Heterogeneity develops during egress of a glycoprotein through the secretory system (1). N-linked glycosylation is initiated in the rough endoplasmic reticulum (ER)4 by the co-translational transfer of Glc3Man9GlcNAc2 to the asparagine residues of the glycosylation sequon. In the absence of protein misfolding, hydrolysis by ER α-mannosidase I plus α-glucosidase I and II results in the transfer of glycoproteins dominated by the D1,D3 isomer of Man8GlcNAc2 glycans to the Golgi apparatus (4). Further processing by Golgi α-mannosidases IA–C generates Man5GlcNAc2 (57), the principle substrate for UDP-N-acetyl-d-glucosamine:α-3-d-mannoside β1,2-N-acetylglucosaminyltransferase I (GnT I). The action of this enzyme yields classic hybrid-type glycans with mannosyl 6-antennae and processed 3-antennae (1). In the absence of the GnT III-mediated addition of bisecting GlcNAc, the two terminal α-mannose residues of the 6-antenna of hybrid-type glycans are cleaved by Golgi α-mannosidase II, forming mono-antennary complex-type glycans. These may then be processed by N-acetylglucosaminyltransferases, generating multiantennary complex-type glycans of enormous potential heterogeneity following the sequential transfer of monosaccharides such as galactose, N-acetylgalactosamine, fucose, and N-acetylneuraminic acid (8).The importance of this carbohydrate diversity in metazoan biology is illustrated by the disease phenotypes that manifest when the biosynthesis of particular glycoforms is disrupted. In humans, about 12 congenital disorders of glycosylation (CDG) have been identified with defects in the biosynthesis of N-linked glycans (9). One disorder characterized by changes in glycosylation is congenital dyserythropoietic anemia type II (hereditary erythroblastic multinuclearity with a positive acidified serum lysis test (HEMPAS)) (10, 11). HEMPAS is a heterogenous autosomal recessive disorder that renders erythrocytes prone to lysis. Although the precise molecular basis of HEMPAS remains to be determined, it is characterized by either a reduction in β1→4-galactosyltransferase, GnT II, or, in some patients, Golgi α-mannosidase II activity (11, 12). Interestingly, the increase in cell surface terminal mannose in mice deficient in Golgi α-mannosidase II leads to autoimmunity through chronic activation of the innate immune system (13, 14).Lectin-resistant (Lec) cell lines harboring loss- or gain-of-function mutations affecting the biosynthesis of N-glycans have emerged as powerful tools for the investigation of these disorders (15). For example, genetic complementation using Lec2, containing a mutation in the cytosine monophosphate sialic acid transporter, was used to identify a novel CDG, type IIf (16). Lectin-resistant cell lines can also be used as hosts to study naturally occurring mutations, as in the case of CHO Lec23 cells used to screen α-glucosidase I mutations in CDG, type IIb (17). Other applications of lectin-resistant cell lines include the expression of specific glycoforms of therapeutic glycoproteins. Manipulating the structure of their carbohydrate moieties modulates the pharmacological properties of glycoproteins by altering their bioactivity, serum half-life, and/or tissue tropism (18). For example, β-glucocerebrosidase expressed in CHO Lec1 cells (deficient in GnT I activity) exhibits mannosylation and improved macrophage uptake for the treatment of Gaucher disease (19). Lectin-resistant CHO cell lines have also been used to improve the crystallizability of glycoproteins for structural determination by x-ray crystallography (2024).The expression of therapeutic glycoproteins as one or more defined “glycoforms” is essential for their optimization and may even be necessary to obtain regulatory approval (25). To this end, eukaryotic expression systems have been developed that allow glycosylation to be controlled. Recently, Pichia pastoris-based strains with human glycosyltransferases have been established, allowing the expression of glycoforms with oligomannose-, hybrid-, and some complex-type glycans (26, 27) and even sialylated complex-type structures (28). However, mammalian expression remains the dominant technology in industrial settings, presumably because of its reliability for the expression of human secreted glycoproteins.Although the majority of lectin-resistant cell lines have been generated using CHO cells, no Golgi α-mannosidase II-deficient CHO cell line has been generated thus far (15). Furthermore, only one human lectin-resistant cell line, i.e. GnT I-deficient (Lec1) HEK 293S cells (29), has been produced. Hybrid-type glycosylation has been reported to accumulate in ricin-resistant baby hamster kidney cells (30, 31); however, these cells contain a reduced but detectable level of cell-surface complex-type glycans, consistent with an incomplete ablation of Golgi α-mannosidase II activity (31). Moreover, the hybrids from one of these lines contain a trimannosyl rather than pentamannosyl core and appear to be heavily influenced by GnT II deficiency, resulting in the formation of what are now commonly called monoantennary complex-type glycans (3234). We now describe the isolation of an HEK 293T cell line mutated at the MAN2A1 locus and deficient in Golgi α-mannosidase II activity via selection with ricin.  相似文献   

20.
Entamoeba histolytica, the protist that causes amebic dysentery and liver abscess, has a truncated Asn-linked glycan (N-glycan) precursor composed of seven sugars (Man5GlcNAc2). Here, we show that glycoproteins with unmodified N-glycans are aggregated and capped on the surface of E. histolytica trophozoites by the antiretroviral lectin cyanovirin-N and then replenished from large intracellular pools. Cyanovirin-N cocaps the Gal/GalNAc adherence lectin, as well as glycoproteins containing O-phosphodiester-linked glycans recognized by an anti-proteophosphoglycan monoclonal antibody. Cyanovirin-N inhibits phagocytosis by E. histolytica trophozoites of mucin-coated beads, a surrogate assay for amebic virulence. For technical reasons, we used the plant lectin concanavalin A rather than cyanovirin-N to enrich secreted and membrane proteins for mass spectrometric identification. E. histolytica glycoproteins with occupied N-glycan sites include Gal/GalNAc lectins, proteases, and 17 previously hypothetical proteins. The latter glycoproteins, as well as 50 previously hypothetical proteins enriched by concanavalin A, may be vaccine targets as they are abundant and unique. In summary, the antiretroviral lectin cyanovirin-N binds to well-known and novel targets on the surface of E. histolytica that are rapidly replenished from large intracellular pools.Entamoeba histolytica causes amebic dysentery and liver abscess in the developing world (10, 20, 29). We are interested in E. histolytica glycoproteins containing Asn-linked glycans (N-glycans) for numerous reasons. E. histolytica makes an N-glycan precursor that contains 7 sugars (Man5GlcNAc2-PP-dolichol) rather than 14 sugars (Glc3Man9GlcNAc2-PP-dolichol) made by most animals, plants, and fungi (21, 31, 44). E. histolytica N-glycans are used for quality control of glycoprotein folding in the endoplasmic reticulum (ER) lumen, and there is positive selection for sites of N-linked glycosylation in secreted and membrane proteins of E. histolytica (5, 11, 53).Unprocessed Man5GlcNAc2, by far the most abundant E. histolytica N-glycan, is present on the plasma membrane and vesicular membranes (31). The antiretroviral lectin cyanovirin-N, which is specific for α-1,2-linked mannose present on unprocessed N-glycans, binds E. histolytica N-glycans and forms aggregates or caps on the surface of E. histolytica trophozoites (1, 25, 31, 44, 45). E. histolytica glycoproteins are also capped by the plant lectin concanavalin A (ConA), which has a broader carbohydrate specificity (mannose and glucose) than cyanovirin-N (3, 16, 18, 19). Heavy subunits of the Gal/GalNAc lectin, the most important E. histolytica vaccine candidate, have 7 to 10 potential sites for N-linked glycosylation (32, 39, 43). Inhibition of N-glycan synthesis results in Gal/GalNAc lectins that are unable to bind to sugars on host epithelial cells.Carbohydrates appear to be an important target on the surface of E. histolytica as anti-proteophosphoglycan (PPG) monoclonal antibodies bind to O-phosphodiester-linked glycans and protect animal models from amebic infection (6, 33, 35, 40, 48). Lectin affinity columns are a powerful method for enriching unique parasite glycoproteins that may be identified by mass spectrometry (MS) of tryptic fragments (17, 55). For example, we recently used the plant lectin wheat germ agglutinin to dramatically enrich glycoproteins with short N-glycans of Giardia (42).The goal of the present studies was to explore further the interaction of the antiretroviral lectin cyanovirin-N with E. histolytica trophozoites in vitro. Questions asked included the following: Are E. histolytica glycoproteins with N-glycans replenished on the plasma membrane after capping with cyanovirin-N? What is the effect of cyanovirin-N capping on other amebic virulence factors and/or vaccine candidates (e.g., the Gal/GalNAc lectin and PPG)? Is capping by cyanovirin-N mediated by actin, as described for capping by the Gal/GalNAc lectin and ConA? What is the effect of the cyanovirin-N on amebic phagocytosis of mucin-coated beads, a surrogate assay for virulence? Which trophozoite glycoproteins are potential targets of cyanovirin-N (identified by mass spectrometry of lectin-enriched E. histolytica proteins)? Are any of them potential vaccine candidates?  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号