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 31 毫秒
1.
Quantifying the similarity of spectra is an important task in various areas of spectroscopy, for example, to identify a compound by comparing sample spectra to those of reference standards. In mass spectrometry based discovery proteomics, spectral comparisons are used to infer the amino acid sequence of peptides. In targeted proteomics by selected reaction monitoring (SRM) or SWATH MS, predetermined sets of fragment ion signals integrated over chromatographic time are used to identify target peptides in complex samples. In both cases, confidence in peptide identification is directly related to the quality of spectral matches. In this study, we used sets of simulated spectra of well-controlled dissimilarity to benchmark different spectral comparison measures and to develop a robust scoring scheme that quantifies the similarity of fragment ion spectra. We applied the normalized spectral contrast angle score to quantify the similarity of spectra to objectively assess fragment ion variability of tandem mass spectrometric datasets, to evaluate portability of peptide fragment ion spectra for targeted mass spectrometry across different types of mass spectrometers and to discriminate target assays from decoys in targeted proteomics. Altogether, this study validates the use of the normalized spectral contrast angle as a sensitive spectral similarity measure for targeted proteomics, and more generally provides a methodology to assess the performance of spectral comparisons and to support the rational selection of the most appropriate similarity measure. The algorithms used in this study are made publicly available as an open source toolset with a graphical user interface.In “bottom-up” proteomics, peptide sequences are identified by the information contained in their fragment ion spectra (1). Various methods have been developed to generate peptide fragment ion spectra and to match them to their corresponding peptide sequences. They can be broadly grouped into discovery and targeted methods. In the widely used discovery (also referred to as shotgun) proteomic approach, peptides are identified by establishing peptide to spectrum matches via a method referred to as database searching. Each acquired fragment ion spectrum is searched against theoretical peptide fragment ion spectra computed from the entries of a specified sequence database, whereby the database search space is constrained to a user defined precursor mass tolerance (2, 3). The quality of the match between experimental and theoretical spectra is typically expressed with multiple scores. These include the number of matching or nonmatching fragments, the number of consecutive fragment ion matches among others. With few exceptions (47) commonly used search engines do not use the relative intensities of the acquired fragment ion signals even though this information could be expected to strengthen the confidence of peptide identification because the relative fragment ion intensity pattern acquired under controlled fragmentation conditions can be considered as a unique “fingerprint” for a given precursor. Thanks to community efforts in acquiring and sharing large number of datasets, the proteomes of some species are now essentially mapped out and experimental fragment ion spectra covering entire proteomes are increasingly becoming accessible through spectral databases (816). This has catalyzed the emergence of new proteomics strategies that differ from classical database searching in that they use prior spectral information to identify peptides. Those comprise inclusion list sequencing (directed sequencing), spectral library matching, and targeted proteomics (17). These methods explicitly use the information contained in empirical fragment ion spectra, including the fragment ion signal intensity to identify the target peptide. For these methods, it is therefore of highest importance to accurately control and quantify the degree of reproducibility of the fragment ion spectra across experiments, instruments, labs, methods, and to quantitatively assess the similarity of spectra. To date, dot product (1824), its corresponding arccosine spectral contrast angle (2527) and (Pearson-like) spectral correlation (2831), and other geometrical distance measures (18, 32), have been used in the literature for assessing spectral similarity. These measures have been used in different contexts including shotgun spectra clustering (19, 26), spectral library searching (18, 20, 21, 24, 25, 2729), cross-instrument fragmentation comparisons (22, 30) and for scoring transitions in targeted proteomics analyses such as selected reaction monitoring (SRM)1 (23, 31). However, to our knowledge, those scores have never been objectively benchmarked for their performance in discriminating well-defined levels of dissimilarities between spectra. In particular, similarity scores obtained by different methods have not yet been compared for targeted proteomics applications, where the sensitive discrimination of highly similar spectra is critical for the confident identification of targeted peptides.In this study, we have developed a method to objectively assess the similarity of fragment ion spectra. We provide an open-source toolset that supports these analyses. Using a computationally generated benchmark spectral library with increasing levels of well-controlled spectral dissimilarity, we performed a comprehensive and unbiased comparison of the performance of the main scores used to assess spectral similarity in mass spectrometry.We then exemplify how this method, in conjunction with its corresponding benchmarked perturbation spectra set, can be applied to answer several relevant questions for MS-based proteomics. As a first application, we show that it can efficiently assess the absolute levels of peptide fragmentation variability inherent to any given mass spectrometer. By comparing the instrument''s intrinsic fragmentation conservation distribution to that of the benchmarked perturbation spectra set, nominal values of spectral similarity scores can indeed be translated into a more directly understandable percentage of variability inherent to the instrument fragmentation. As a second application, we show that the method can be used to derive an absolute measure to estimate the conservation of peptide fragmentation between instruments or across proteomics methods. This allowed us to quantitatively evaluate, for example, the transferability of fragment ion spectra acquired by data dependent analysis in a first instrument into a fragment/transition assay list used for targeted proteomics applications (e.g. SRM or targeted extraction of data independent acquisition SWATH MS (33)) on another instrument. Third, we used the method to probe the fragmentation patterns of peptides carrying a post-translation modification (e.g. phosphorylation) by comparing the spectra of modified peptide with those of their unmodified counterparts. Finally, we used the method to determine the overall level of fragmentation conservation that is required to support target-decoy discrimination and peptide identification in targeted proteomics approaches such as SRM and SWATH MS.  相似文献   

2.
Understanding how a small brain region, the suprachiasmatic nucleus (SCN), can synchronize the body''s circadian rhythms is an ongoing research area. This important time-keeping system requires a complex suite of peptide hormones and transmitters that remain incompletely characterized. Here, capillary liquid chromatography and FTMS have been coupled with tailored software for the analysis of endogenous peptides present in the SCN of the rat brain. After ex vivo processing of brain slices, peptide extraction, identification, and characterization from tandem FTMS data with <5-ppm mass accuracy produced a hyperconfident list of 102 endogenous peptides, including 33 previously unidentified peptides, and 12 peptides that were post-translationally modified with amidation, phosphorylation, pyroglutamylation, or acetylation. This characterization of endogenous peptides from the SCN will aid in understanding the molecular mechanisms that mediate rhythmic behaviors in mammals.Central nervous system neuropeptides function in cell-to-cell signaling and are involved in many physiological processes such as circadian rhythms, pain, hunger, feeding, and body weight regulation (14). Neuropeptides are produced from larger protein precursors by the selective action of endopeptidases, which cleave at mono- or dibasic sites and then remove the C-terminal basic residues (1, 2). Some neuropeptides undergo functionally important post-translational modifications (PTMs),1 including amidation, phosphorylation, pyroglutamylation, or acetylation. These aspects of peptide synthesis impact the properties of neuropeptides, further expanding their diverse physiological implications. Therefore, unveiling new peptides and unreported peptide properties is critical to advancing our understanding of nervous system function.Historically, the analysis of neuropeptides was performed by Edman degradation in which the N-terminal amino acid is sequentially removed. However, analysis by this method is slow and does not allow for sequencing of the peptides containing N-terminal PTMs (5). Immunological techniques, such as radioimmunoassay and immunohistochemistry, are used for measuring relative peptide levels and spatial localization, but these methods only detect peptide sequences with known structure (6). More direct, high throughput methods of analyzing brain regions can be used.Mass spectrometry, a rapid and sensitive method that has been used for the analysis of complex biological samples, can detect and identify the precise forms of neuropeptides without prior knowledge of peptide identity, with these approaches making up the field of peptidomics (712). The direct tissue and single neuron analysis by MALDI MS has enabled the discovery of hundreds of neuropeptides in the last decade, and the neuronal homogenate analysis by fractionation and subsequent ESI or MALDI MS has yielded an equivalent number of new brain peptides (5). Several recent peptidome studies, including the work by Dowell et al. (10), have used the specificity of FTMS for peptide discovery (10, 1315). Here, we combine the ability to fragment ions at ultrahigh mass accuracy (16) with a software pipeline designed for neuropeptide discovery. We use nanocapillary reversed-phase LC coupled to 12 Tesla FTMS for the analysis of peptides present in the suprachiasmatic nucleus (SCN) of rat brain.A relatively small, paired brain nucleus located at the base of the hypothalamus directly above the optic chiasm, the SCN contains a biological clock that generates circadian rhythms in behaviors and homeostatic functions (17, 18). The SCN comprises ∼10,000 cellular clocks that are integrated as a tissue level clock which, in turn, orchestrates circadian rhythms throughout the brain and body. It is sensitive to incoming signals from the light-sensing retina and other brain regions, which cause temporal adjustments that align the SCN appropriately with changes in environmental or behavioral state. Previous physiological studies have implicated peptides as critical synchronizers of normal SCN function as well as mediators of SCN inputs, internal signal processing, and outputs; however, only a small number of peptides have been identified and explored in the SCN, leaving unresolved many circadian mechanisms that may involve peptide function.Most peptide expression in the SCN has only been studied through indirect antibody-based techniques (1929), although we recently used MS approaches to characterize several peptides detected in SCN releasates (30). Previous studies indicate that the SCN expresses a rich diversity of peptides relative to other brain regions studied with the same techniques. Previously used immunohistochemical approaches are not only inadequate for comprehensively evaluating PTMs and alternate isoforms of known peptides but are also incapable of exhaustively examining the full peptide complement of this complex biological network of peptidergic inputs and intrinsic components. A comprehensive study of SCN peptidomics is required that utilizes high resolution strategies for directly analyzing the peptide content of the neuronal networks comprising the SCN.In our study, the SCN was obtained from ex vivo coronal brain slices via tissue punch and subjected to multistage peptide extraction. The SCN tissue extract was analyzed by FTMS/MS, and the high resolution MS and MS/MS data were processed using ProSightPC 2.0 (16), which allows the identification and characterization of peptides or proteins from high mass accuracy MS/MS data. In addition, the Sequence Gazer included in ProSightPC was used for manually determining PTMs (31, 32). As a result, a total of 102 endogenous peptides were identified, including 33 that were previously unidentified, and 12 PTMs (including amidation, phosphorylation, pyroglutamylation, and acetylation) were found. The present study is the first comprehensive peptidomics study for identifying peptides present within the mammalian SCN. In fact, this is one of the first peptidome studies to work with discrete brain nuclei as opposed to larger brain structures and follows up on our recent report using LC-ion trap for analysis of the peptides in the supraoptic nucleus (33); here, the use of FTMS allows a greater range of PTMs to be confirmed and allows higher confidence in the peptide assignments. This information on the peptides in the SCN will serve as a basis to more exhaustively explore the extent that previously unreported SCN neuropeptides may function in SCN regulation of mammalian circadian physiology.  相似文献   

3.
A complete understanding of the biological functions of large signaling peptides (>4 kDa) requires comprehensive characterization of their amino acid sequences and post-translational modifications, which presents significant analytical challenges. In the past decade, there has been great success with mass spectrometry-based de novo sequencing of small neuropeptides. However, these approaches are less applicable to larger neuropeptides because of the inefficient fragmentation of peptides larger than 4 kDa and their lower endogenous abundance. The conventional proteomics approach focuses on large-scale determination of protein identities via database searching, lacking the ability for in-depth elucidation of individual amino acid residues. Here, we present a multifaceted MS approach for identification and characterization of large crustacean hyperglycemic hormone (CHH)-family neuropeptides, a class of peptide hormones that play central roles in the regulation of many important physiological processes of crustaceans. Six crustacean CHH-family neuropeptides (8–9.5 kDa), including two novel peptides with extensive disulfide linkages and PTMs, were fully sequenced without reference to genomic databases. High-definition de novo sequencing was achieved by a combination of bottom-up, off-line top-down, and on-line top-down tandem MS methods. Statistical evaluation indicated that these methods provided complementary information for sequence interpretation and increased the local identification confidence of each amino acid. Further investigations by MALDI imaging MS mapped the spatial distribution and colocalization patterns of various CHH-family neuropeptides in the neuroendocrine organs, revealing that two CHH-subfamilies are involved in distinct signaling pathways.Neuropeptides and hormones comprise a diverse class of signaling molecules involved in numerous essential physiological processes, including analgesia, reward, food intake, learning and memory (1). Disorders of the neurosecretory and neuroendocrine systems influence many pathological processes. For example, obesity results from failure of energy homeostasis in association with endocrine alterations (2, 3). Previous work from our lab used crustaceans as model organisms found that multiple neuropeptides were implicated in control of food intake, including RFamides, tachykinin related peptides, RYamides, and pyrokinins (46).Crustacean hyperglycemic hormone (CHH)1 family neuropeptides play a central role in energy homeostasis of crustaceans (717). Hyperglycemic response of the CHHs was first reported after injection of crude eyestalk extract in crustaceans. Based on their preprohormone organization, the CHH family can be grouped into two sub-families: subfamily-I containing CHH, and subfamily-II containing molt-inhibiting hormone (MIH) and mandibular organ-inhibiting hormone (MOIH). The preprohormones of the subfamily-I have a CHH precursor related peptide (CPRP) that is cleaved off during processing; and preprohormones of the subfamily-II lack the CPRP (9). Uncovering their physiological functions will provide new insights into neuroendocrine regulation of energy homeostasis.Characterization of CHH-family neuropeptides is challenging. They are comprised of more than 70 amino acids and often contain multiple post-translational modifications (PTMs) and complex disulfide bridge connections (7). In addition, physiological concentrations of these peptide hormones are typically below picomolar level, and most crustacean species do not have available genome and proteome databases to assist MS-based sequencing.MS-based neuropeptidomics provides a powerful tool for rapid discovery and analysis of a large number of endogenous peptides from the brain and the central nervous system. Our group and others have greatly expanded the peptidomes of many model organisms (3, 1833). For example, we have discovered more than 200 neuropeptides with several neuropeptide families consisting of as many as 20–40 members in a simple crustacean model system (5, 6, 2531, 34). However, a majority of these neuropeptides are small peptides with 5–15 amino acid residues long, leaving a gap of identifying larger signaling peptides from organisms without sequenced genome. The observed lack of larger size peptide hormones can be attributed to the lack of effective de novo sequencing strategies for neuropeptides larger than 4 kDa, which are inherently more difficult to fragment using conventional techniques (3437). Although classical proteomics studies examine larger proteins, these tools are limited to identification based on database searching with one or more peptides matching without complete amino acid sequence coverage (36, 38).Large populations of neuropeptides from 4–10 kDa exist in the nervous systems of both vertebrates and invertebrates (9, 39, 40). Understanding their functional roles requires sufficient molecular knowledge and a unique analytical approach. Therefore, developing effective and reliable methods for de novo sequencing of large neuropeptides at the individual amino acid residue level is an urgent gap to fill in neurobiology. In this study, we present a multifaceted MS strategy aimed at high-definition de novo sequencing and comprehensive characterization of the CHH-family neuropeptides in crustacean central nervous system. The high-definition de novo sequencing was achieved by a combination of three methods: (1) enzymatic digestion and LC-tandem mass spectrometry (MS/MS) bottom-up analysis to generate detailed sequences of proteolytic peptides; (2) off-line LC fractionation and subsequent top-down MS/MS to obtain high-quality fragmentation maps of intact peptides; and (3) on-line LC coupled to top-down MS/MS to allow rapid sequence analysis of low abundance peptides. Combining the three methods overcomes the limitations of each, and thus offers complementary and high-confidence determination of amino acid residues. We report the complete sequence analysis of six CHH-family neuropeptides including the discovery of two novel peptides. With the accurate molecular information, MALDI imaging and ion mobility MS were conducted for the first time to explore their anatomical distribution and biochemical properties.  相似文献   

4.
The growing use of selected reaction monitoring (SRM) mass spectrometry in proteomic analyses led us to investigate how to identify peptides by SRM using only a minimal number of fragment ions. By using a computational model of the SRM work flow we computed the potential interferences from other peptides in a given proteome. From these results, we selected the deterministic SRM addresses that contained sufficient information to confer peptide and protein identity that we termed unique ion signatures (UIS). We computationally showed that UIS comprised of only two transitions are diagnostic for >99% of Escherichia coli proteins and >96% of human proteins that possess a sequence-unique peptide. We demonstrated an example of experimental use of UIS using a modified SRM methodology to profile the E. coli tricarboxylic acid cycle from a single injection of cell lysate. In addition, we showed the potential of UIS to form the first functionally orthogonal approach to validate peptide assignments obtained from conventional analyses of MS/MS spectra. The UIS methodology is a novel deterministic peptide identification method for MS/MS spectra based on information content. These robust theoretical assays will have widespread use when integrated with previously collected MS/MS data and conventional proteomics technologies.Shotgun proteomic analyses using multidimensional LC/MS/MS show great capacity for rapid protein analysis. This is arguably the most prevalent work flow for high throughput comparative proteomics, utilizing information-dependent acquisition (IDA)1 to acquire MS/MS triggered by the signals generated from incoming peptides (13). Despite the utility and widespread use of this approach, there remain inherent problems including a relatively high level of ambiguous and false peptide assignments (∼5%) as well as high numbers of unassigned mass spectra (46). The reason for this level of ambiguity stems in part from the non-deterministic nature of the identification algorithms. Without the use of reference standards the only way to know a spectrum was generated by a given peptide with absolute certainty is for the spectrum to contain a fragment pattern that conclusively demonstrates the presence of each amino acid. Unfortunately this level of coverage is extremely rare in proteomics data.More recently, selected reaction monitoring (SRM) or multiple reaction monitoring (MRM) mass spectrometry methods have been deployed for proteomic analyses (720). This has occurred as proteomics has matured from a discovery-oriented discipline into a more targeted and quantitative field. The method is conventionally conducted using triple quadrupole mass spectrometers where two rounds of mass selection provide excellent fidelity and sensitivity to monitor one or more predetermined target peptides generally in the context of a complex sample such as a cell lysate. Using this approach the mass spectrometer continually monitors the selected precursor ion m/z (Q1) and a subsequent product ion m/z (Q3) from the target analyte. SRM experiments can be used to conduct several rounds of these scans targeting different product ions in an attempt to bolster the confidence that the Q1 → Q3 transitions monitor the intended analyte with fidelity. A key point of contrast with IDA experiments is the need to preselect target analytes for monitoring. This can be achieved by harvesting data from previous discovery-based experiments or by in silico predictions such as MRM-initiated detection and sequencing (MIDAS) (10, 12). Regardless the key underlying principle of SRM in proteomics applications is that the selected set of precursor and product ions contain sufficient information to proxy for the target peptide and thereby its protein of origin. Given that proteomics SRM experiments are conducted with a minimal set of transitions, one must accept that a degree of uncertainty resides in any such assay. To date, the magnitude of this uncertainty has not been studied. This remains a key point even with MS instruments capable of conducting subsequent full MS/MS scans triggered by SRM (e.g. QTrap) as these are lower sensitivity scans that may contain insufficient fragmentation data to conclusively confer peptide identity.The problem of interference is also present in SRM experiments. To achieve acceptable sensitivity a large Q1 m/z window (±0.3–1.0 m/z) is needed. This in turn allows other peptides with similar Q1 m/z and elution properties to interfere with detection of the desired target. The frequency of these interferences would likely increase as the complexity of the sample increases creating a greater likelihood of false positives. Clearly this is not an unexpected result as conventional peptide identification strategies utilizing tandem MS result in some false assignments. Therefore, it would be unreasonable to expect that SRM assays that typically utilize fewer product ions than MS/MS experiments would not also encounter similar interference (21).In this study we investigated the information content of SRM assays and in doing so exposed the potential redundancy. Computational simulations of the experiment enabled us to demonstrate that directed selection of SRM precursor and product ions can avoid the pitfalls of interference by selecting ion combinations that uniquely map to target peptides within the context of the simulation. We used these unique ion signatures (UIS) in a proof of concept study to direct SRM data acquisition for the exclusive detection of enzymes in the Escherichia coli tricarboxylic acid cycle. In addition, given that UIS have been calculated to uniquely define target peptides in the experimental context, we demonstrated the applicability of UIS as an orthogonal validation of peptide identity for traditional MS/MS experiments.  相似文献   

5.
6.
7.
Knowledge of elaborate structures of protein complexes is fundamental for understanding their functions and regulations. Although cross-linking coupled with mass spectrometry (MS) has been presented as a feasible strategy for structural elucidation of large multisubunit protein complexes, this method has proven challenging because of technical difficulties in unambiguous identification of cross-linked peptides and determination of cross-linked sites by MS analysis. In this work, we developed a novel cross-linking strategy using a newly designed MS-cleavable cross-linker, disuccinimidyl sulfoxide (DSSO). DSSO contains two symmetric collision-induced dissociation (CID)-cleavable sites that allow effective identification of DSSO-cross-linked peptides based on their distinct fragmentation patterns unique to cross-linking types (i.e. interlink, intralink, and dead end). The CID-induced separation of interlinked peptides in MS/MS permits MS3 analysis of single peptide chain fragment ions with defined modifications (due to DSSO remnants) for easy interpretation and unambiguous identification using existing database searching tools. Integration of data analyses from three generated data sets (MS, MS/MS, and MS3) allows high confidence identification of DSSO cross-linked peptides. The efficacy of the newly developed DSSO-based cross-linking strategy was demonstrated using model peptides and proteins. In addition, this method was successfully used for structural characterization of the yeast 20 S proteasome complex. In total, 13 non-redundant interlinked peptides of the 20 S proteasome were identified, representing the first application of an MS-cleavable cross-linker for the characterization of a multisubunit protein complex. Given its effectiveness and simplicity, this cross-linking strategy can find a broad range of applications in elucidating the structural topology of proteins and protein complexes.Proteins form stable and dynamic multisubunit complexes under different physiological conditions to maintain cell viability and normal cell homeostasis. Detailed knowledge of protein interactions and protein complex structures is fundamental to understanding how individual proteins function within a complex and how the complex functions as a whole. However, structural elucidation of large multisubunit protein complexes has been difficult because of a lack of technologies that can effectively handle their dynamic and heterogeneous nature. Traditional methods such as nuclear magnetic resonance (NMR) analysis and x-ray crystallography can yield detailed information on protein structures; however, NMR spectroscopy requires large quantities of pure protein in a specific solvent, whereas x-ray crystallography is often limited by the crystallization process.In recent years, chemical cross-linking coupled with mass spectrometry (MS) has become a powerful method for studying protein interactions (13). Chemical cross-linking stabilizes protein interactions through the formation of covalent bonds and allows the detection of stable, weak, and/or transient protein-protein interactions in native cells or tissues (49). In addition to capturing protein interacting partners, many studies have shown that chemical cross-linking can yield low resolution structural information about the constraints within a molecule (2, 3, 10) or protein complex (1113). The application of chemical cross-linking, enzymatic digestion, and subsequent mass spectrometric and computational analyses for the elucidation of three-dimensional protein structures offers distinct advantages over traditional methods because of its speed, sensitivity, and versatility. Identification of cross-linked peptides provides distance constraints that aid in constructing the structural topology of proteins and/or protein complexes. Although this approach has been successful, effective detection and accurate identification of cross-linked peptides as well as unambiguous assignment of cross-linked sites remain extremely challenging due to their low abundance and complicated fragmentation behavior in MS analysis (2, 3, 10, 14). Therefore, new reagents and methods are urgently needed to allow unambiguous identification of cross-linked products and to improve the speed and accuracy of data analysis to facilitate its application in structural elucidation of large protein complexes.A number of approaches have been developed to facilitate MS detection of low abundance cross-linked peptides from complex mixtures. These include selective enrichment using affinity purification with biotinylated cross-linkers (1517) and click chemistry with alkyne-tagged (18) or azide-tagged (19, 20) cross-linkers. In addition, Staudinger ligation has recently been shown to be effective for selective enrichment of azide-tagged cross-linked peptides (21). Apart from enrichment, detection of cross-linked peptides can be achieved by isotope-labeled (2224), fluorescently labeled (25), and mass tag-labeled cross-linking reagents (16, 26). These methods can identify cross-linked peptides with MS analysis, but interpretation of the data generated from interlinked peptides (two peptides connected with the cross-link) by automated database searching remains difficult. Several bioinformatics tools have thus been developed to interpret MS/MS data and determine interlinked peptide sequences from complex mixtures (12, 14, 2732). Although promising, further developments are still needed to make such data analyses as robust and reliable as analyzing MS/MS data of single peptide sequences using existing database searching tools (e.g. Protein Prospector, Mascot, or SEQUEST).Various types of cleavable cross-linkers with distinct chemical properties have been developed to facilitate MS identification and characterization of cross-linked peptides. These include UV photocleavable (33), chemical cleavable (19), isotopically coded cleavable (24), and MS-cleavable reagents (16, 26, 3438). MS-cleavable cross-linkers have received considerable attention because the resulting cross-linked products can be identified based on their characteristic fragmentation behavior observed during MS analysis. Gas-phase cleavage sites result in the detection of a “reporter” ion (26), single peptide chain fragment ions (3538), or both reporter and fragment ions (16, 34). In each case, further structural characterization of the peptide product ions generated during the cleavage reaction can be accomplished by subsequent MSn1 analysis. Among these linkers, the “fixed charge” sulfonium ion-containing cross-linker developed by Lu et al. (37) appears to be the most attractive as it allows specific and selective fragmentation of cross-linked peptides regardless of their charge and amino acid composition based on their studies with model peptides.Despite the availability of multiple types of cleavable cross-linkers, most of the applications have been limited to the study of model peptides and single proteins. Additionally, complicated synthesis and fragmentation patterns have impeded most of the known MS-cleavable cross-linkers from wide adaptation by the community. Here we describe the design and characterization of a novel and simple MS-cleavable cross-linker, DSSO, and its application to model peptides and proteins and the yeast 20 S proteasome complex. In combination with new software developed for data integration, we were able to identify DSSO-cross-linked peptides from complex peptide mixtures with speed and accuracy. Given its effectiveness and simplicity, we anticipate a broader application of this MS-cleavable cross-linker in the study of structural topology of other protein complexes using cross-linking and mass spectrometry.  相似文献   

8.
9.
Finding robust biomarkers for Parkinson disease (PD) is currently hampered by inherent technical limitations associated with imaging or antibody-based protein assays. To circumvent the challenges, we adapted a staged pipeline, starting from our previous proteomic profiling followed by high-throughput targeted mass spectrometry (MS), to identify peptides in human cerebrospinal fluid (CSF) for PD diagnosis and disease severity correlation. In this multicenter study consisting of training and validation sets, a total of 178 subjects were randomly selected from a retrospective cohort, matching age and sex between PD patients, healthy controls, and neurological controls with Alzheimer disease (AD). From ∼14,000 unique peptides displaying differences between PD and healthy control in proteomic investigations, 126 peptides were selected based on relevance and observability in CSF using bioinformatic analysis and MS screening, and then quantified by highly accurate and sensitive selected reaction monitoring (SRM) in the CSF of 30 PD patients versus 30 healthy controls (training set), followed by diagnostic (receiver operating characteristics) and disease severity correlation analyses. The most promising candidates were further tested in an independent cohort of 40 PD patients, 38 AD patients, and 40 healthy controls (validation set). A panel of five peptides (derived from SPP1, LRP1, CSF1R, EPHA4, and TIMP1) was identified to provide an area under curve (AUC) of 0.873 (sensitivity = 76.7%, specificity = 80.0%) for PD versus healthy controls in the training set. The performance was essentially confirmed in the validation set (AUC = 0.853, sensitivity = 82.5%, specificity = 82.5%). Additionally, this panel could also differentiate the PD and AD groups (AUC = 0.990, sensitivity = 95.0%, specificity = 97.4%). Furthermore, a combination of two peptides belonging to proteins TIMP1 and APLP1 significantly correlated with disease severity as determined by the Unified Parkinson''s Disease Rating Scale motor scores in both the training (r = 0.381, p = 0.038)j and the validation (r = 0.339, p = 0.032) sets. The novel panel of CSF peptides, if validated in independent cohorts, could be used to assist in clinical diagnosis of PD and has the potential to help monitoring or predicting disease progression.Parkinson disease (PD)1, the second most common neurodegenerative disease after Alzheimer disease (AD), afflicts roughly 2% of persons over the age of 65 years (1, 2). Currently, PD diagnosis is mainly based on observation of the cardinal motor indicators of the disease, patient response to drug treatment, and medical history (3, 4). There is an appreciable misdiagnosis rate (4), particularly at early disease stages. Additionally, no objective measure of disease progression or treatment effects has been established. Thus, objective, reliable, and reproducible biomarkers are clearly needed to aid in the diagnosis of PD and tracking or predicting the disease progression.The most sensitive tests developed to date are based on imaging modalities, which can detect functional and structural abnormalities even prior to the onset of motor dysfunction (5, 6). However, the usefulness of neuroimaging techniques is limited by high cost, limited accessibility, difficulty in reliable differentiation of PD from other atypical parkinsonian disorders and subjection to confounding factors such as medication and compensatory responses (47). Biochemical and molecular markers in cerebrospinal fluid (CSF) and other body fluids have also been actively investigated (5, 812). The most extensively studied candidate in CSF is probably α-synuclein, the major protein component of Lewy bodies and Lewy neurites, the pathological hallmarks of PD (2). The current consensus is that CSF α-synuclein concentrations are generally lower in patients with PD compared with controls (5, 810); the sensitivity and specificity, however, appear to be only moderate, and no correlation with PD severity or progression has been observed (8, 9). Notably, all these CSF protein markers are measured using antibody-based assays, which are often associated with relatively high variability, particularly when different detection techniques (different antibodies, sample preparation, calibrators, etc.) are used, leading to discrepant results across laboratories (5). It should also be stressed that this high variability in immunoassays is not unique to PD, because similar difficulty is encountered in AD and other related disorders (13, 14).One strategy to avoid the inherent technical limitations associated with antibodies is to use alternative techniques in which unique peptides are selected and precisely quantified with mass spectrometry (MS) techniques, for example, accurate inclusion mass screening (AIMS) (15) and selected reaction monitoring (SRM) (1618). To this end, in the last few years, we and others have utilized proteomic technologies to identify novel proteins and peptides associated with different disease states and stages (5, 6, 1925). Using brain tissue or CSF, these unbiased proteomic profiling studies have revealed disease-related alterations in hundreds of peptides derived from many proteins (1925). However, there are no quantitative assays for the majority of these candidate proteins/peptides, and development of such assays is limited by the lack of antibodies available for many of them. Thus, although a large library of potential peptide biomarkers has been developed, the vast majority never reach the stage of validation and clinical testing, hampered by the difficulty of de novo development of immunoassays, a process that is time consuming, prohibitively expensive to develop and very difficult to multiplex.In this study, we aim to establish a PD biomarker identification and verification pipeline, with the goal of prioritizing candidates and swiftly developing reliable quantitative assays. We focused on identifying peptides by SRM and AIMS, because these targeted proteomic technologies have been proposed as the basis of a viable biomarker pipeline (16) and have become a powerful tool in biomarker discovery because of their high sensitivity, accuracy and specificity. SRM, in particular, has emerged as an alternative to immunoaffinity-based measurements of defined protein sets with excellent reproducibility across different laboratories and instrument platforms (17, 18). The staged pipeline in the current investigation (Fig. 1) includes: (1) data-dependent and bioinformatic prioritization of thousands of candidate biomarkers identified in our previous profiling studies, (2) de novo development of antibody-free multiplex SRM assays to reliably measure tens to hundreds of peptides simultaneously, and (3) multiplex biomarker verification studies allowing identification and validation of models or panels of candidates in independent sample sets, two of which were used in this study.Open in a separate windowFig. 1.Overview of the workflow used for CSF peptide biomarker discovery and validation. AD, Alzheimer disease; AIMS, accurate inclusion mass screening; CO, healthy controls; DDA, data-dependent acquisition; PD, Parkinson disease; SRM, selected reaction monitoring.  相似文献   

10.
Cross-linking/mass spectrometry resolves protein–protein interactions or protein folds by help of distance constraints. Cross-linkers with specific properties such as isotope-labeled or collision-induced dissociation (CID)-cleavable cross-linkers are in frequent use to simplify the identification of cross-linked peptides. Here, we analyzed the mass spectrometric behavior of 910 unique cross-linked peptides in high-resolution MS1 and MS2 from published data and validate the observation by a ninefold larger set from currently unpublished data to explore if detailed understanding of their fragmentation behavior would allow computational delivery of information that otherwise would be obtained via isotope labels or CID cleavage of cross-linkers. Isotope-labeled cross-linkers reveal cross-linked and linear fragments in fragmentation spectra. We show that fragment mass and charge alone provide this information, alleviating the need for isotope-labeling for this purpose. Isotope-labeled cross-linkers also indicate cross-linker-containing, albeit not specifically cross-linked, peptides in MS1. We observed that acquisition can be guided to better than twofold enrich cross-linked peptides with minimal losses based on peptide mass and charge alone. By help of CID-cleavable cross-linkers, individual spectra with only linear fragments can be recorded for each peptide in a cross-link. We show that cross-linked fragments of ordinary cross-linked peptides can be linearized computationally and that a simplified subspectrum can be extracted that is enriched in information on one of the two linked peptides. This allows identifying candidates for this peptide in a simplified database search as we propose in a search strategy here. We conclude that the specific behavior of cross-linked peptides in mass spectrometers can be exploited to relax the requirements on cross-linkers.Cross-linking/mass spectrometry extends the use of mass-spectrometry-based proteomics from identification (1, 2), quantification (3), and characterization of protein complexes (4) into resolving protein structures and protein–protein interactions (58). Chemical reagents (cross-linkers) covalently connect amino acid pairs that are within a cross-linker-specific distance range in the native three-dimensional structure of a protein or protein complex. A cross-linking/mass spectrometry experiment is typically conducted in four steps: (1) cross-linking of the target protein or complex, (2) protein digestion (usually with trypsin), (3) LC-MS analysis, and (4) database search. The digested peptide mixture consists of linear and cross-linked peptides, and the latter can be enriched by strong cation exchange (9) or size exclusion chromatography (10). Cross-linked peptides are of high value as they provide direct information on the structure and interactions of proteins.Cross-linked peptides fragment under collision-induced dissociation (CID) conditions primarily into b- and y-ions, as do their linear counterparts. An important difference regarding database searches between linear and cross-linked peptides stems from not knowing which peptides might be cross-linked. Therefore, one has to consider each single peptide and all pairwise combinations of peptides in the database. Having n peptides leads to (n2 + n)/2 possible pairwise combinations. This leads to two major challenges: With increasing size of the database, search time and the risk of identifying false positives increases. One way of circumventing these problems is to use MS2-cleavable cross-linkers (11, 12), at the cost of limited experimental design and choice of cross-linker.In a first database search approach (13), all pairwise combinations of peptides in a database were considered in a concatenated and linearized form. Thereby, all possible single bond fragments are considered in one of the two database entries per peptide pair, and the cross-link can be identified by a normal protein identification algorithm. Already, the second search approach split the peptides for the purpose of their identification (14). Linear fragments were used to retrieve candidate peptides from the database that are then matched based on the known mass of the cross-linked pair and scored as a pair against the spectrum. Isotope-labeled cross-linkers were used to sort the linear and cross-linked fragments apart. Many other search tools and approaches have been developed since (10, 1519); see (20) for a more detailed list, at least some of which follow the general idea of an open modification search (2124).As a general concept for open modification search of cross-linked peptides, cross-linked peptides represent two peptides, each with an unknown modification given by the mass of the other peptide and the cross-linker. One identifies both peptides individually and then matches them based on knowing the mass of cross-linked pair (14, 22, 24). Alternatively, one peptide is identified first and, using that peptide and the cross-linker as a modification mass, the second peptide is identified from the database (21, 23). An important element of the open modification search approach is that it essentially converts the quadratic search space of the cross-linked peptides into a linear search space of modified peptides. Still, many peptides and many modification positions have to be considered, especially when working with large databases or when using highly reactive cross-linkers with limited amino acid selectivity (25).We hypothesize that detailed knowledge of the fragmentation behavior of cross-linked peptides might reveal ways to improve the identification of cross-linked peptides. Detailed analyses of the fragmentation behavior of linear peptides exist (2628), and the analysis of the fragmentation behavior of cross-linked peptides has guided the design of scores (24, 29). Further, cross-link-specific ions have been observed from higher energy collision dissociation (HCD) data (30). Isotope-labeled cross-linkers are used to distinguish cross-linked from linear fragments, generally in low-resolution MS2 of cross-linked peptides (14).We compared the mass spectrometric behavior of cross-linked peptides to that of linear peptides, using 910 high-resolution fragment spectra matched to unique cross-linked peptides from multiple different public datasets at 5% peptide-spectrum match (PSM)1 false discovery rate (FDR). In addition, we repeated all experiments with a larger sample set that contains 8,301 spectra—also including data from ongoing studies from our lab (Supplemental material S9-S12). This paper presents the mass spectrometric signature of cross-linked peptides that we identified in our analysis and the resulting heuristics that are incorporated into an integrated strategy for the analysis and identification of cross-linked peptides. We present computational strategies that indicate the possibility of alleviating the need for mass-spectrometrically restricted cross-linker choice.  相似文献   

11.
Selected reaction monitoring mass spectrometry (SRM-MS) is playing an increasing role in quantitative proteomics and biomarker discovery studies as a method for high throughput candidate quantification and verification. Although SRM-MS offers advantages in sensitivity and quantification compared with other MS-based techniques, current SRM technologies are still challenged by detection and quantification of low abundance proteins (e.g. present at ∼10 ng/ml or lower levels in blood plasma). Here we report enhanced detection sensitivity and reproducibility for SRM-based targeted proteomics by coupling a nanospray ionization multicapillary inlet/dual electrodynamic ion funnel interface to a commercial triple quadrupole mass spectrometer. Because of the increased efficiency in ion transmission, significant enhancements in overall signal intensities and improved limits of detection were observed with the new interface compared with the original interface for SRM measurements of tryptic peptides from proteins spiked into non-depleted mouse plasma over a range of concentrations. Overall, average SRM peak intensities were increased by ∼70-fold. The average level of detection for peptides also improved by ∼10-fold with notably improved reproducibility of peptide measurements as indicated by the reduced coefficients of variance. The ability to detect proteins ranging from 40 to 80 ng/ml within mouse plasma was demonstrated for all spiked proteins without the application of front-end immunoaffinity depletion and fractionation. This significant improvement in detection sensitivity for low abundance proteins in complex matrices is expected to enhance a broad range of SRM-MS applications including targeted protein and metabolite validation.Although mass spectrometry (MS)-based proteomics is a promising high throughput technology for biomarker discovery and validation (15), only a handful of cancer biomarkers have been approved by the United States Food and Drug Administration for clinical use in the last decade (6, 7). Assuming that low abundance biomarkers do exist in the biofluids to be studied, the success of biomarker discovery efforts primarily depends on the sensitivity, accuracy, and robustness of the measurement technologies; the quality and size of patient cohorts and clinical samples and execution within the context of an overall difficult and expensive path to clinical application that encompasses discovery, verification, and validation stages (1, 5, 810). A multiplexed assay platform increasingly considered for biomarker verification is selected reaction monitoring (SRM)1 by tandem mass spectrometry using e.g. a triple quadrupole (QqQ) mass spectrometer to attain high throughput quantitative measurements of targeted proteins in complex matrices (1, 11, 12).SRM utilizes two stages of mass filtering by selecting a specific analyte ion of interest (precursor ion) in the first stage followed by a specific fragment ion derived from the precursor (fragment ion) filter in the second stage after collision-activated dissociation. Typically, several transitions (precursor/fragment ion pairs) are monitored for greater selectivity and confidence in a targeted peptide assay, and large numbers of peptides can be monitored during a single LC-MS/MS analysis. The two-stage mass selection by individual quadrupoles enables more rapid and continuous monitoring of specific ions derived from analytes of interest such as peptides and leads to significantly enhanced detection sensitivity and quantitative accuracy compared with broad (i.e. non-targeted) LC-MS or LC-MS/MS measurements (11, 12). Both the sensitivity and selectivity of SRM-MS make this technique well suited for the targeted detection and quantification of low abundance proteins in highly complex biofluids (1316). The precision and reproducibility of SRM-based measurements of proteins in plasma across different laboratories have recently been assessed (17).Despite its promise, present SRM measurements still do not provide sufficient sensitivity for reliable detection and quantification of low abundance proteins in biofluids (e.g. present in plasma at ∼10 ng/ml or lower levels) primarily because of factors related to high sample complexity and the large dynamic range of relative protein abundances (7, 18, 19). Given sufficient selectivity, the sensitivity achievable is generally related to the peptide MS and MS/MS signal intensities obtained. One of the key factors limiting peptide MS intensities is the significant ion losses encountered between the electrospray ionization (ESI) source and the interface to the mass spectrometer. In typical LC-ESI-MS interfaces, the mass spectrometer inlet (e.g. heated capillary followed by a skimmer) presently provides total ion utilization and ion transmission efficiencies on the order of ∼1% (20) due to a combination of limited ion sampling from the atmospheric pressure ion source into the inlet and inefficient transmission of ions entering the first reduced pressure stage of the mass spectrometer.The electrodynamic ion funnel (21), which has been developed to efficiently capture, focus, and transmit ions to the high vacuum region of the mass spectrometer, is expected to provide a large benefit to SRM analyses. The original ion funnel interfaces, which operated at a maximum of ∼5 torr, were able to enhance signal intensities for a variety of MS analyzers (2224) by replacing the inefficient skimmer interface. Although achieving near lossless ion transmission to high vacuum, losses at the atmospheric pressure interface went unmitigated. More recently, a high pressure ion funnel interface capable of operating at a pressure of ∼30 torr was introduced (25). The higher operating pressures accommodated greater gas loads and enabled more efficient ion sampling from atmospheric pressure through a multicapillary inlet. With a dual ion funnel interface comprising a high pressure ion funnel with a heated multicapillary inlet followed by a standard ion funnel operated at 1–2 torrs, highly efficient ion sampling from atmospheric pressure to high vacuum is readily achieved.In this study, we report the enhanced sensitivity and reproducibility of SRM-based targeted proteomics measurements achieved by implementing a dual stage electrodynamic ion funnel interface that incorporates a multicapillary inlet with a triple quadrupole mass spectrometer. A series of LC-SRM-MS measurements were made using mouse plasma samples spiked with various concentrations of tryptic peptides from five standard proteins to evaluate the improvements in detection sensitivity and reproducibility attained by this modified interface relative to a standard Thermo (single capillary inlet/skimmer) interface. A ∼10-fold improvement in the limit of detection (LOD) as well as improved measurement reproducibility was achieved.  相似文献   

12.
Comprehensive analysis of the complex nature of the Human Leukocyte Antigen (HLA) class II ligandome is of utmost importance to understand the basis for CD4+ T cell mediated immunity and tolerance. Here, we implemented important improvements in the analysis of the repertoire of HLA-DR-presented peptides, using hybrid mass spectrometry-based peptide fragmentation techniques on a ligandome sample isolated from matured human monocyte-derived dendritic cells (DC). The reported data set constitutes nearly 14 thousand unique high-confident peptides, i.e. the largest single inventory of human DC derived HLA-DR ligands to date. From a technical viewpoint the most prominent finding is that no single peptide fragmentation technique could elucidate the majority of HLA-DR ligands, because of the wide range of physical chemical properties displayed by the HLA-DR ligandome. Our in-depth profiling allowed us to reveal a strikingly poor correlation between the source proteins identified in the HLA class II ligandome and the DC cellular proteome. Important selective sieving from the sampled proteome to the ligandome was evidenced by specificity in the sequences of the core regions both at their N- and C- termini, hence not only reflecting binding motifs but also dominant protease activity associated to the endolysosomal compartments. Moreover, we demonstrate that the HLA-DR ligandome reflects a surface representation of cell-compartments specific for biological events linked to the maturation of monocytes into antigen presenting cells. Our results present new perspectives into the complex nature of the HLA class II system and will aid future immunological studies in characterizing the full breadth of potential CD4+ T cell epitopes relevant in health and disease.Human Leukocyte Antigen (HLA)1 class II molecules on professional antigen presenting cells such as dendritic cells (DC) expose peptide fragments derived from exogenous and endogenous proteins to be screened by CD4+ T cells (1, 2). The activation and recruitment of CD4+ T cells recognizing disease-related peptide antigens is critical for the development of efficient antipathogen or antitumor immunity. Furthermore, the presentation of self-peptides and their interaction with CD4+ T cells is essential to maintain immunological tolerance and homeostasis (3). Knowledge of the nature of HLA class II-presented peptides on DC is of great importance to understand the rules of antigen processing and peptide binding motifs (4), whereas the identity of disease-related antigens may provide new knowledge on immunogenicity and leads for the development of vaccines and immunotherapy (5, 6).Mass spectrometry (MS) has proven effective for the analysis HLA class II-presented peptides (4, 7, 8). MS-based ligandome studies have demonstrated that HLA class II molecules predominantly present peptides derived from exogenous proteins that entered the cells by endocytosis and endogenous proteins that are associated with the endo-lysosomal compartments (4). Yet proteins residing in the cytosol, nucleus or mitochondria can also be presented by HLA class II molecules, primarily through autophagy (911). Multiple studies have mapped the HLA class II ligandome of antigen presenting cells in the context of infectious pathogens (12), autoimmune diseases (1317) or cancer (14, 18, 19), or those that are essential for self-tolerance in the human thymus (3, 20). Notwithstanding these efforts, and certainly not in line with the extensive knowledge on the HLA class I ligandome (21), the nature of the HLA class II-presented peptide repertoire and particular its relationship to the cellular source proteome remains poorly understood.To advance our knowledge on the HLA-DR ligandome on activated DC without having to deal with limitations in cell yield from peripheral human blood (12, 21, 22) or tissue isolates (3), we explored the use of MUTZ-3 cells. This cell line has been used as a model of human monocyte-derived DCs. MUTZ-3 cells can be matured to act as antigen presenting cells and express then high levels of HLA class II molecules, and can be propagated in vitro to large cell densities (2325). We also evaluated the performance of complementary and hybrid MS fragmentation techniques electron-transfer dissociation (ETD), electron-transfer/higher-energy collision dissociation (EThcD) (26), and higher-energy collision dissociation (HCD) to sequence and identify the HLA class II ligandome. Together this workflow allowed for the identification of an unprecedented large set of about 14 thousand unique peptide sequences presented by DC derived HLA-DR molecules, providing an in-depth view of the complexity of the HLA class II ligandome, revealing underlying features of antigen processing and surface-presentation to CD4+ T cells.  相似文献   

13.
The Major histocompatibility complex (MHC) class I peptidome is thought to be generated mostly through proteasomal degradation of cellular proteins, a notion that is based on the alterations in presentation of selected peptides following proteasome inhibition. We evaluated the effects of proteasome inhibitors, epoxomicin and bortezomib, on human cultured cancer cells. Because the inhibitors did not reduce the level of presentation of the cell surface human leukocyte antigen (HLA) molecules, we followed their effects on the rates of synthesis of both HLA peptidome and proteome of the cells, using dynamic stable isotope labeling in tissue culture (dynamic-SILAC). The inhibitors reduced the rates of synthesis of most cellular proteins and HLA peptides, yet the synthesis rates of some of the proteins and HLA peptides was not decreased by the inhibitors and of some even increased. Therefore, we concluded that the inhibitors affected the production of the HLA peptidome in a complex manner, including modulation of the synthesis rates of the source proteins of the HLA peptides, in addition to their effect on their degradation. The collected data may suggest that the current reliance on proteasome inhibition may overestimate the centrality of the proteasome in the generation of the MHC peptidome. It is therefore suggested that the relative contribution of the proteasomal and nonproteasomal pathways to the production of the MHC peptidome should be revaluated in accordance with the inhibitors effects on the synthesis rates of the source proteins of the MHC peptides.The repertoires and levels of peptides, presented by the major histocompatibility complex (MHC)1 class I molecules at the cells'' surface, are modulated by multiple factors. These include the rates of synthesis and degradation of their source proteins, the transport efficacy of the peptides through the transporter associated with antigen processing (TAP) into the endoplasmic reticulum (ER), their subsequent processing and loading onto the MHC molecules within the ER, and the rates of transport of the MHC molecules with their peptide cargo to the cell surface. The off-rates of the presented peptides, the residence time of the MHC complexes at the cell surface, and their retrograde transport back into the cytoplasm, influence, as well, the presented peptidomes (reviewed in (1)). Even though significant portions of the MHC class I peptidomes are thought to be derived from newly synthesized proteins, including misfolded proteins, defective ribosome products (DRiPs), and short lived proteins (SLiPs), most of the MHC peptidome is assumed to originate from long-lived proteins, which completed their functional cellular roles or became defective (retirees), (reviewed in (2)).The main protease, supplying the MHC peptidome production pipeline, is thought to be the proteasome (3). It is also responsible for generation of the final carboxyl termini of the MHC peptides (4), (reviewed in (5)). The final trimming of the n-termini of the peptides, within the endoplasmic reticulum (ER), is thought to be performed by amino peptidases, such as ERAP1/ERAAP, which fit the peptides into their binding groove on the MHC molecules (6) (reviewed in (7)). Nonproteasomal proteolytic pathways were also suggested as possible contributors to the MHC peptidome, including proteolysis by the ER resident Signal peptide peptidase (8, 9), the cytoplasmic proteases Insulin degrading enzyme (10), Tripeptidyl peptidase (1113), and a number of proteases within the endolysosome pathway (reviewed recently in (1417)). In contrast to the mostly cytoplasmic and ER production of the MHC class I peptidome, the class II peptidome is produced in a special compartment, associated with the endolysosome pathway (1820). This pathway is also thought to participate in the cross presentation of class I peptides, derived from proteins up-taken by professional antigen presenting cells (21), (reviewed in (1517, 22)).The centrality of the proteasomes in the generation of the MHC peptidome was deduced mostly from the observed change in presentation levels of small numbers of selected peptides, following proteasome inhibition (3, 23). Even the location of some of the genes encoding the catalytic subunits of the immunoproteasome (LMP2 and LMP7) (24) within the MHC class II genomic locus, was suggested to support the involvement of the proteasome in the generation of the MHC class I peptidome (25). Similar conclusions were deduced from alterations in peptide presentation, following expression of the catalytic subunits of the immunoproteasome (26), (reviewed in (5)). Yet, although most of the reports indicated reductions in presentation of selected peptides by proteasome inhibition (3, 2729), others have observed only limited, and sometimes even opposite effects (23, 3032).The matter is further complicated by the indirect effects of proteasome inhibition used for such studies on the arrest of protein synthesis by the cells (3335), on the transport rates of the MHC molecules to the cell surface, and on their retrograde transport back to the vesicular system (36) (reviewed in (37)). Proteasome inhibition likely causes shortage of free ubiquitin, reduced supply of free amino acids, and induces an ER unfolded protein response (UPR), which signals the cells to block most (but not all) cellular protein synthesis (reviewed in (38)). Because a significant portion of the MHC peptidome originates from degradation of DRiPs and SLiPs (reviewed in (2)), arrest of new protein synthesis should influence the presentation of their derived MHC peptides. Taken together, these arguments may suggest that merely following the changes in the presentation levels of the MHC molecules, or even of specific MHC peptides, after proteasome inhibition, does not provide the full picture for deducing the relative contribution of the proteasomal pathway to the production of the MHC peptidome (reviewed in (7)).MHC peptidome analysis can be performed relatively easily by modern capillary chromatography combined with mass spectrometry (reviewed in (39)). The peptides are recovered from immunoaffinity purified MHC molecules after detergent solubilization of the cells (40, 41), from soluble MHC molecules secreted to the cells'' growth medium (42, 43) or from patients'' plasma (44). The purified peptides pools are resolved by capillary chromatography and the individual peptides are identified and quantified by tandem mass spectrometry (40), (reviewed in (4547)). In cultured cells, quantitative analysis can also be followed by metabolic incorporation of stable isotope labeled amino acids (SILAC) (48). Furthermore, the rates of de novo synthesis of both MHC peptides and their proteins of origin can be followed using the dynamic-SILAC proteomics approach (49) with its further adaptation to HLA peptidomics (5052).This study attempts to define the relative contribution of the proteasomes to the production of HLA class I peptidome by simultaneously following the effects of proteasome inhibitors, epoxomicin and bortezomib (Velcade), on the rates of de novo synthesis of both the HLA class I peptidome and the cellular proteome of the same MCF-7 human breast cancer cultured cells. The proteasome inhibitors did not reduce the levels of HLA presentations, yet affected the rates of production of both the HLA peptidome and cellular proteome, mostly decreasing, but also increasing some of the synthesis rates of the HLA peptides and cellular proteins. Thus, we suggest that the degree of contribution of the proteasomal pathway to the production of the HLA-I peptidome should be re-evaluated in accordance with their effects on the entire HLA class-I peptidome, while taking into consideration the inhibitors'' effects on the synthesis (and degradation) rates of the source proteins of each of the studied HLA peptides.  相似文献   

14.
Paneth cells are a secretory epithelial lineage that release dense core granules rich in host defense peptides and proteins from the base of small intestinal crypts. Enteric α-defensins, termed cryptdins (Crps) in mice, are highly abundant in Paneth cell secretions and inherently resistant to proteolysis. Accordingly, we tested the hypothesis that enteric α-defensins of Paneth cell origin persist in a functional state in the mouse large bowel lumen. To test this idea, putative Crps purified from mouse distal colonic lumen were characterized biochemically and assayed in vitro for bactericidal peptide activities. The peptides comigrated with cryptdin control peptides in acid-urea-PAGE and SDS-PAGE, providing identification as putative Crps. Matrix-assisted laser desorption ionization time-of-flight mass spectrometry experiments showed that the molecular masses of the putative α-defensins matched those of the six most abundant known Crps, as well as N-terminally truncated forms of each, and that the peptides contain six Cys residues, consistent with identities as α-defensins. N-terminal sequencing definitively revealed peptides with N termini corresponding to full-length, (des-Leu)-truncated, and (des-Leu-Arg)-truncated N termini of Crps 1–4 and 6. Crps from mouse large bowel lumen were bactericidal in the low micromolar range. Thus, Paneth cell α-defensins secreted into the small intestinal lumen persist as intact and functional forms throughout the intestinal tract, suggesting that the peptides may mediate enteric innate immunity in the colonic lumen, far from their upstream point of secretion in small intestinal crypts.Antimicrobial peptides (AMPs)2 are released by epithelial cells onto mucosal surfaces as effectors of innate immunity (15). In mammals, most AMPs derive from two major families, the cathelicidins and defensins (6). The defensins comprise the α-, β-, and θ-defensin subfamilies, which are defined by the presence of six cysteine residues paired in characteristic tridisulfide arrays (7). α-Defensins are highly abundant in two primary cell lineages: phagocytic leukocytes, primarily neutrophils, of myeloid origin and Paneth cells, which are secretory epithelial cells located at the base of the crypts of Lieberkühn in the small intestine (810). Neutrophil α-defensins are stored in azurophilic granules and contribute to non-oxidative microbial cell killing in phagolysosomes (11, 12), except in mice whose neutrophils lack defensins (13). In the small bowel, α-defensins and other host defense proteins (1418) are released apically as components of Paneth cell secretory granules in response to cholinergic stimulation and after exposure to bacterial antigens (19). Therefore, the release of Paneth cell products into the crypt lumen is inferred to protect mitotically active crypt cells from colonization by potential pathogens and confer protection against enteric infection (7, 20, 21).Under normal, homeostatic conditions, Paneth cells are not found outside the small bowel, although they may appear ectopically in response to local inflammation throughout the gastrointestinal tract (22, 23). Paneth cell numbers increase progressively throughout the small intestine, occurring at highest numbers in the distal ileum (24). Mouse Paneth cells express numerous α-defensin isoforms, termed cryptdins (Crps) (25), that have broad spectrum antimicrobial activities (6, 26). Collectively, α-defensins constitute approximately seventy percent of the bactericidal peptide activity in mouse Paneth cell secretions (19), selectively killing bacteria by membrane-disruptive mechanisms (2730). The role of Paneth cell α-defensins in gastrointestinal mucosal immunity is evident from studies of mice transgenic for human enteric α-defensin-5, HD-5, which are immune to infection by orally administered Salmonella enterica sv. typhimurium (S. typhimurium) (31).The biosynthesis of mature, bactericidal α-defensins from their inactive precursors requires activation by lineage-specific proteolytic convertases. In mouse Paneth cells, inactive ∼8.4-kDa Crp precursors are processed intracellularly into microbicidal ∼4-kDa Crps by specific cleavage events mediated by matrix metalloproteinase-7 (MMP-7) (32, 33). MMP-7 null mice exhibit increased susceptibility to systemic S. typhimurium infection and decreased clearance of orally administered non-invasive Escherichia coli (19, 32). Although the α-defensin proregions are sensitive to proteolysis, the mature, disulfide-stabilized peptides resist digestion by their converting enzymes in vitro, whether the convertase is MMP-7 (32), trypsin (34), or neutrophil serine proteinases (35). Because α-defensins resist proteolysis in vitro, we hypothesized that Paneth cell α-defensins resist degradation and remain in a functional state in the large bowel, a complex, hostile environment containing varied proteases of both host and microbial origin.Here, we report on the isolation and characterization of a population of enteric α-defensins from the mouse colonic lumen. Full-length and N-terminally truncated Paneth cell α-defensins were identified and are abundant in the distal large bowel lumen.  相似文献   

15.
16.
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   

17.
Comprehensive proteomic profiling of biological specimens usually requires multidimensional chromatographic peptide fractionation prior to mass spectrometry. However, this approach can suffer from poor reproducibility because of the lack of standardization and automation of the entire workflow, thus compromising performance of quantitative proteomic investigations. To address these variables we developed an online peptide fractionation system comprising a multiphasic liquid chromatography (LC) chip that integrates reversed phase and strong cation exchange chromatography upstream of the mass spectrometer (MS). We showed superiority of this system for standardizing discovery and targeted proteomic workflows using cancer cell lysates and nondepleted human plasma. Five-step multiphase chip LC MS/MS acquisition showed clear advantages over analyses of unfractionated samples by identifying more peptides, consuming less sample and often improving the lower limits of quantitation, all in highly reproducible, automated, online configuration. We further showed that multiphase chip LC fractionation provided a facile means to detect many N- and C-terminal peptides (including acetylated N terminus) that are challenging to identify in complex tryptic peptide matrices because of less favorable ionization characteristics. Given as much as 95% of peptides were detected in only a single salt fraction from cell lysates we exploited this high reproducibility and coupled it with multiple reaction monitoring on a high-resolution MS instrument (MRM-HR). This approach increased target analyte peak area and improved lower limits of quantitation without negatively influencing variance or bias. Further, we showed a strategy to use multiphase LC chip fractionation LC-MS/MS for ion library generation to integrate with SWATHTM data-independent acquisition quantitative workflows. All MS data are available via ProteomeXchange with identifier PXD001464.Mass spectrometry based proteomic quantitation is an essential technique used for contemporary, integrative biological studies. Whether used in discovery experiments or for targeted biomarker applications, quantitative proteomic studies require high reproducibility at many levels. It requires reproducible run-to-run peptide detection, reproducible peptide quantitation, reproducible depth of proteome coverage, and ideally, a high degree of cross-laboratory analytical reproducibility. Mass spectrometry centered proteomics has evolved steadily over the past decade, now mature enough to derive extensive draft maps of the human proteome (1, 2). Nonetheless, a key requirement yet to be realized is to ensure that quantitative proteomics can be carried out in a timely manner while satisfying the aforementioned challenges associated with reproducibility. This is especially important for recent developments using data independent MS quantitation and multiple reaction monitoring on high-resolution MS (MRM-HR)1 as they are both highly dependent on LC peptide retention time reproducibility and precursor detectability, while attempting to maximize proteome coverage (3). Strategies usually employed to increase the depth of proteome coverage utilize various sample fractionation methods including gel-based separation, affinity enrichment or depletion, protein or peptide chemical modification-based enrichment, and various peptide chromatography methods, particularly ion exchange chromatography (410). In comparison to an unfractionated “naive” sample, the trade-off in using these enrichments/fractionation approaches are higher risk of sample losses, introduction of undesired chemical modifications (e.g. oxidation, deamidation, N-terminal lactam formation), and the potential for result skewing and bias, as well as numerous time and human resources required to perform the sample preparation tasks. Online-coupled approaches aim to minimize those risks and address resource constraints. A widely practiced example of the benefits of online sample fractionation has been the decade long use of combining strong cation exchange chromatography (SCX) with C18 reversed-phase (RP) for peptide fractionation (known as MudPIT – multidimensional protein identification technology), where SCX and RP is performed under the same buffer conditions and the SCX elution performed with volatile organic cations compatible with reversed phase separation (11). This approach greatly increases analyte detection while avoiding sample handling losses. The MudPIT approach has been widely used for discovery proteomics (1214), and we have previously shown that multiphasic separations also have utility for targeted proteomics when configured for selected reaction monitoring MS (SRM-MS). We showed substantial advantages of MudPIT-SRM-MS with reduced ion suppression, increased peak areas and lower limits of detection (LLOD) compared with conventional RP-SRM-MS (15).To improve the reproducibility of proteomic workflows, increase throughput and minimize sample loss, numerous microfluidic devices have been developed and integrated for proteomic applications (16, 17). These devices can broadly be classified into two groups: (1) microfluidic chips for peptide separation (1825) and; (2) proteome reactors that combine enzymatic processing with peptide based fractionation (2630). Because of the small dimension of these devices, they are readily able to integrate into nanoLC workflows. Various applications have been described including increasing proteome coverage (22, 27, 28) and targeting of phosphopeptides (24, 31, 32), glycopeptides and released glycans (29, 33, 34).In this work, we set out to take advantage of the benefits of multiphasic peptide separations and address the reproducibility needs required for high-throughput comparative proteomics using a variety of workflows. We integrated a multiphasic SCX and RP column in a “plug-and-play” microfluidic chip format for online fractionation, eliminating the need for users to make minimal dead volume connections between traps and columns. We show the flexibility of this format to provide robust peptide separation and reproducibility using conventional and topical mass spectrometry workflows. This was undertaken by coupling the multiphase liquid chromatography (LC) chip to a fast scanning Q-ToF mass spectrometer for data dependent MS/MS, data independent MS (SWATH) and for targeted proteomics using MRM-HR, showing clear advantages for repeatable analyses compared with conventional proteomic workflows.  相似文献   

18.
Database search programs are essential tools for identifying peptides via mass spectrometry (MS) in shotgun proteomics. Simultaneously achieving high sensitivity and high specificity during a database search is crucial for improving proteome coverage. Here we present JUMP, a new hybrid database search program that generates amino acid tags and ranks peptide spectrum matches (PSMs) by an integrated score from the tags and pattern matching. In a typical run of liquid chromatography coupled with high-resolution tandem MS, more than 95% of MS/MS spectra can generate at least one tag, whereas the remaining spectra are usually too poor to derive genuine PSMs. To enhance search sensitivity, the JUMP program enables the use of tags as short as one amino acid. Using a target-decoy strategy, we compared JUMP with other programs (e.g. SEQUEST, Mascot, PEAKS DB, and InsPecT) in the analysis of multiple datasets and found that JUMP outperformed these preexisting programs. JUMP also permitted the analysis of multiple co-fragmented peptides from “mixture spectra” to further increase PSMs. In addition, JUMP-derived tags allowed partial de novo sequencing and facilitated the unambiguous assignment of modified residues. In summary, JUMP is an effective database search algorithm complementary to current search programs.Peptide identification by tandem mass spectra is a critical step in mass spectrometry (MS)-based1 proteomics (1). Numerous computational algorithms and software tools have been developed for this purpose (26). These algorithms can be classified into three categories: (i) pattern-based database search, (ii) de novo sequencing, and (iii) hybrid search that combines database search and de novo sequencing. With the continuous development of high-performance liquid chromatography and high-resolution mass spectrometers, it is now possible to analyze almost all protein components in mammalian cells (7). In contrast to rapid data collection, it remains a challenge to extract accurate information from the raw data to identify peptides with low false positive rates (specificity) and minimal false negatives (sensitivity) (8).Database search methods usually assign peptide sequences by comparing MS/MS spectra to theoretical peptide spectra predicted from a protein database, as exemplified in SEQUEST (9), Mascot (10), OMSSA (11), X!Tandem (12), Spectrum Mill (13), ProteinProspector (14), MyriMatch (15), Crux (16), MS-GFDB (17), Andromeda (18), BaMS2 (19), and Morpheus (20). Some other programs, such as SpectraST (21) and Pepitome (22), utilize a spectral library composed of experimentally identified and validated MS/MS spectra. These methods use a variety of scoring algorithms to rank potential peptide spectrum matches (PSMs) and select the top hit as a putative PSM. However, not all PSMs are correctly assigned. For example, false peptides may be assigned to MS/MS spectra with numerous noisy peaks and poor fragmentation patterns. If the samples contain unknown protein modifications, mutations, and contaminants, the related MS/MS spectra also result in false positives, as their corresponding peptides are not in the database. Other false positives may be generated simply by random matches. Therefore, it is of importance to remove these false PSMs to improve dataset quality. One common approach is to filter putative PSMs to achieve a final list with a predefined false discovery rate (FDR) via a target-decoy strategy, in which decoy proteins are merged with target proteins in the same database for estimating false PSMs (2326). However, the true and false PSMs are not always distinguishable based on matching scores. It is a problem to set up an appropriate score threshold to achieve maximal sensitivity and high specificity (13, 27, 28).De novo methods, including Lutefisk (29), PEAKS (30), NovoHMM (31), PepNovo (32), pNovo (33), Vonovo (34), and UniNovo (35), identify peptide sequences directly from MS/MS spectra. These methods can be used to derive novel peptides and post-translational modifications without a database, which is useful, especially when the related genome is not sequenced. High-resolution MS/MS spectra greatly facilitate the generation of peptide sequences in these de novo methods. However, because MS/MS fragmentation cannot always produce all predicted product ions, only a portion of collected MS/MS spectra have sufficient quality to extract partial or full peptide sequences, leading to lower sensitivity than achieved with the database search methods.To improve the sensitivity of the de novo methods, a hybrid approach has been proposed to integrate peptide sequence tags into PSM scoring during database searches (36). Numerous software packages have been developed, such as GutenTag (37), InsPecT (38), Byonic (39), DirecTag (40), and PEAKS DB (41). These methods use peptide tag sequences to filter a protein database, followed by error-tolerant database searching. One restriction in most of these algorithms is the requirement of a minimum tag length of three amino acids for matching protein sequences in the database. This restriction reduces the sensitivity of the database search, because it filters out some high-quality spectra in which consecutive tags cannot be generated.In this paper, we describe JUMP, a novel tag-based hybrid algorithm for peptide identification. The program is optimized to balance sensitivity and specificity during tag derivation and MS/MS pattern matching. JUMP can use all potential sequence tags, including tags consisting of only one amino acid. When we compared its performance to that of two widely used search algorithms, SEQUEST and Mascot, JUMP identified ∼30% more PSMs at the same FDR threshold. In addition, the program provides two additional features: (i) using tag sequences to improve modification site assignment, and (ii) analyzing co-fragmented peptides from mixture MS/MS spectra.  相似文献   

19.
20.
Allelic polymorphism of the apolipoprotein E (ApoE) gene (ApoE ε2, ApoE ε3 and ApoE ε4 alleles) gives rise to three protein isoforms (ApoE2, ApoE3 and ApoE4) that differ by 1 or 2 amino acids. Inheritance of the ApoE ε4 allele is a risk factor for developing Alzheimer''s disease (AD). The potential diagnostic value of ApoE protein levels in biological fluids (i.e. cerebrospinal fluid, plasma and serum) for distinguishing between AD patients and healthy elderly subjects is subject to great controversy. Although a recent study reported subnormal total ApoE and ApoE4 levels in the plasma of AD patients, other studies have found normal or even elevated protein levels (versus controls). Because all previously reported assays were based on immunoenzymatic techniques, we decided to develop an orthogonal assay based on targeted mass spectrometry by tracking (i) a proteotypic peptide common to all ApoE isoforms and (ii) a peptide that is specific for the ε4 allele. After trypsin digestion, the ApoE4-specific peptide contains an oxidation-prone methionine residue. The endogenous methionine oxidation level was evaluated in a small cohort (n = 68) of heterozygous ε3ε4 carriers containing both healthy controls and AD patients. As expected, the proportion of oxidized residues varied from 0 to 10%, with an average of 5%. We therefore developed a standardized strategy for the unbiased, absolute quantification of ApoE4, based on performic acid oxidization of methionine. Once the sample workflow had been thoroughly validated, it was applied to the concomitant quantification of total ApoE and ApoE4 isoform in a large case-control study (n = 669). The final measurements were consistent with most previously reported ApoE concentration values and confirm the influence of the different alleles on the protein expression level. Our results illustrate (i) the reliability of selected reaction monitoring-based assays and (ii) the value of the oxidization step for unbiased monitoring of methionine-containing proteotypic peptides. Furthermore, a statistical analysis indicated that neither total ApoE and ApoE4 levels nor the ApoE/ApoE4 ratio correlated with the diagnosis of AD. These findings reinforce the conclusions of previous studies in which plasma ApoE levels had no obvious clinical significance.Apolipoprotein E (ApoE) is a 299-amino acid protein associated with lipoproteins in the plasma and the cerebrospinal fluid (1). The three major genetic variants of ApoE in the general population isoforms (E2, E3, and E4, encoded by the ε2, ε3 and ε4 alleles, respectively) differ by a single amino acid: E2 (cys112, cys158), E3 (cys112, arg158), and E4 (arg112, arg158). Among the different susceptibility genes associated with the risk of late-onset Alzheimer''s disease (AD)1 and other neurological conditions, ApoE has been identified as a strong genetic determinant (1). Moreover, the risk of AD is strongly correlated with ApoE4 allele because the presence of one or two copies of the allele increases risk of late-onset AD by about three or 12 times, respectively. Furthermore, the presence of one or two copies of ApoE4 allele correlates with an earlier age of onset by about 10–20 years with regard to noncarriers in patients with late-onset disease (24).The diagnostic value of an ApoE assay in AD is however subject to debate. Indeed, supranormal (5), subnormal (6), and nondiscriminating (710) plasma concentrations of ApoE in AD cohorts have been reported in many studies based on immunoassay techniques. We wondered whether this ambiguity might be resolved by applying quantitative mass spectrometry (MS). Indeed, the combination of standardized, stable isotope dilution with targeted MS in selected reaction monitoring (SRM) mode is now widely accepted as a valuable alternative to immunoassays for accurate protein quantification (1117) and may constitute the eagerly awaited bridge between the discovery and verification phases for candidate biomarker panels (1822). A range of pilot studies have evaluated the sensitivity that can be achieved by SRM monitoring (23, 24); when tracking protein in a whole plasma hydrolysate, limits of quantification (LOQs) in the low μg/ml range are frequently reported (12). However, LOQs in the low nanogram/ml range or the detection of proteins expressed with a low copy number per cell require either immuno-enrichment of the target protein or fractionation of proteotypic peptides by cation exchange (24, 25) or off-gel electrophoresis (26). A generic strategy based on stable isotope standards and capture by antipeptide antibodies (referred to as SISCAPA) is a promising alternative to more traditional enrichment strategies. Numerous reports have already demonstrated the ability of single or multiplexed immuno-SRM to achieve limits of quantification of 1 nanogram/ml or below when starting from less than 100 μl of biofluid (27). Precision, reproducibility and robustness are also key parameters in the development of SRM-based assay platforms dedicated to the clinical verification phase. Indeed, accuracy may even be a secondary consideration at this point, because the main objective is to discriminate between specific and nondiscriminating biomarker candidates within the clinical panel. Various pilot studies of the intra- and interclass performances of SRM monitoring have demonstrated acceptable coefficients of variation and levels of imprecision (below 20%) and, importantly, have pinpointed the main pitfalls that can distort assay results (2833). Indeed, performance levels and reliability of an assay can be drastically compromised by intraclass variance within the proteotypic peptide fraction, which is caused by chemical modifications of certain amino acid side chains. In particular, peptides containing methionine residues are usually not recommended for SRM-based assays, because the sulfur atom is prone to oxidation (3436) both in vivo (37, 38) and during sample handling or storage (giving rise to methionine sulfoxide). Unfortunately, peptides containing one or more methionine residues are sometimes the only ones available. By way of an example, 17% of the 625 proteins identified by only one peptide in Human Plasma PeptideAtlas database contain at least one methionine. Hence, quantification of this type of proteotypic peptide is challenging because the ratio between nonoxidized and mono-oxidized sulfoxide forms of heavy-labeled standards also changes during storage. To circumvent this limitation, complete oxidization of the methionine-containing peptide population with chemical oxidant has been attempted but was not sufficiently robust or reproducible over several weeks in the context of large clinical study (39, 40).Here, we report on the absolute quantification of total ApoE and ApoE4 isoform peptides by liquid chromatography (LC)-SRM targeted MS in a case-control study (n = 669). Because ApoE4-specific trypsin peptide possesses a single methionine residue (LGADMEDVR), oxidation was performed immediately after enzymatic digestion. The completeness of was carefully checked. We continuously assessed the overall performance of the assay by introducing quality control samples throughout the clinical cohort study, in order to ensure the validity of statistical comparisons between healthy controls and groups of AD patients.  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