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 312 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.
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   

3.
In large-scale proteomic experiments, multiple peptide precursors are often cofragmented simultaneously in the same mixture tandem mass (MS/MS) spectrum. These spectra tend to elude current computational tools because of the ubiquitous assumption that each spectrum is generated from only one peptide. Therefore, tools that consider multiple peptide matches to each MS/MS spectrum can potentially improve the relatively low spectrum identification rate often observed in proteomics experiments. More importantly, data independent acquisition protocols promoting the cofragmentation of multiple precursors are emerging as alternative methods that can greatly improve the throughput of peptide identifications but their success also depends on the availability of algorithms to identify multiple peptides from each MS/MS spectrum. Here we address a fundamental question in the identification of mixture MS/MS spectra: determining the statistical significance of multiple peptides matched to a given MS/MS spectrum. We propose the MixGF generating function model to rigorously compute the statistical significance of peptide identifications for mixture spectra and show that this approach improves the sensitivity of current mixture spectra database search tools by a ≈30–390%. Analysis of multiple data sets with MixGF reveals that in complex biological samples the number of identified mixture spectra can be as high as 20% of all the identified spectra and the number of unique peptides identified only in mixture spectra can be up to 35.4% of those identified in single-peptide spectra.The advancement of technology and instrumentation has made tandem mass (MS/MS)1 spectrometry the leading high-throughput method to analyze proteins (1, 2, 3). In typical experiments, tens of thousands to millions of MS/MS spectra are generated and enable researchers to probe various aspects of the proteome on a large scale. Part of this success hinges on the availability of computational methods that can analyze the large amount of data generated from these experiments. The classical question in computational proteomics asks: given an MS/MS spectrum, what is the peptide that generated the spectrum? However, it is increasingly being recognized that this assumption that each MS/MS spectrum comes from only one peptide is often not valid. Several recent analyses show that as many as 50% of the MS/MS spectra collected in typical proteomics experiments come from more than one peptide precursor (4, 5). The presence of multiple peptides in mixture spectra can decrease their identification rate to as low as one half of that for MS/MS spectra generated from only one peptide (6, 7, 8). In addition, there have been numerous developments in data independent acquisition (DIA) technologies where multiple peptide precursors are intentionally selected to cofragment in each MS/MS spectrum (9, 10, 11, 12, 13, 14, 15). These emerging technologies can address some of the enduring disadvantages of traditional data-dependent acquisition (DDA) methods (e.g. low reproducibility (16)) and potentially increase the throughput of peptide identification 5–10 fold (4, 17). However, despite the growing importance of mixture spectra in various contexts, there are still only a few computational tools that can analyze mixture spectra from more than one peptide (18, 19, 20, 21, 8, 22). Our recent analysis indicated that current database search methods for mixture spectra still have relatively low sensitivity compared with their single-peptide counterpart and the main bottleneck is their limited ability to separate true matches from false positive matches (8). Traditionally problem of peptide identification from MS/MS spectra involves two sub-problems: 1) define a Peptide-Spectrum-Match (PSM) scoring function that assigns each MS/MS spectrum to the peptide sequence that most likely generated the spectrum; and 2) given a set of top-scoring PSMs, select a subset that corresponds to statistical significance PSMs. Here we focus on the second problem, which is still an ongoing research question even for the case of single-peptide spectra (23, 24, 25, 26). Intuitively the second problem is difficult because one needs to consider spectra across the whole data set (instead of comparing different peptide candidates against one spectrum as in the first problem) and PSM scoring functions are often not well-calibrated across different spectra (i.e. a PSM score of 50 may be good for one spectrum but poor for a different spectrum). Ideally, a scoring function will give high scores to all true PSMs and low scores to false PSMs regardless of the peptide or spectrum being considered. However, in practice, some spectra may receive higher scores than others simply because they have more peaks or their precursor mass results in more peptide candidates being considered from the sequence database (27, 28). Therefore, a scoring function that accounts for spectrum or peptide-specific effects can make the scores more comparable and thus help assess the confidence of identifications across different spectra. The MS-GF solution to this problem is to compute the per-spectrum statistical significance of each top-scoring PSM, which can be defined as the probability that a random peptide (out of all possible peptide within parent mass tolerance) will match to the spectrum with a score at least as high as that of the top-scoring PSM. This measures how good the current best match is in relation to all possible peptides matching to the same spectrum, normalizing any spectrum effect from the scoring function. Intuitively, our proposed MixGF approach extends the MS-GF approach to now calculate the statistical significance of the top pair of peptides matched from the database to a given mixture spectrum M (i.e. the significance of the top peptide–peptide spectrum match (PPSM)). As such, MixGF determines the probability that a random pair of peptides (out of all possible peptides within parent mass tolerance) will match a given mixture spectrum with a score at least as high as that of the top-scoring PPSM.Despite the theoretical attractiveness of computing statistical significance, it is generally prohibitive for any database search methods to score all possible peptides against a spectrum. Therefore, earlier works in this direction focus on approximating this probability by assuming the score distribution of all PSMs follows certain analytical form such as the normal, Poisson or hypergeometric distributions (29, 30, 31). In practice, because score distributions are highly data-dependent and spectrum-specific, these model assumptions do not always hold. Other approaches tried to learn the score distribution empirically from the data (29, 27). However, one is most interested in the region of the score distribution where only a small fraction of false positives are allowed (typically at 1% FDR). This usually corresponds to the extreme tail of the distribution where p values are on the order of 10−9 or lower and thus there is typically lack of sufficient data points to accurately model the tail of the score distribution (32). More recently, Kim et al. (24) and Alves et al. (33), in parallel, proposed a generating function approach to compute the exact score distribution of random peptide matches for any spectra without explicitly matching all peptides to a spectrum. Because it is an exact computation, no assumption is made about the form of score distribution and the tail of the distribution can be computed very accurately. As a result, this approach substantially improved the ability to separate true matches from false positive ones and lead to a significant increase in sensitivity of peptide identification over state-of-the-art database search tools in single-peptide spectra (24).For mixture spectra, it is expected that the scores for the top-scoring match will be even less comparable across different spectra because now more than one peptide and different numbers of peptides can be matched to each spectrum at the same time. We extend the generating function approach (24) to rigorously compute the statistical significance of multiple-Peptide-Spectrum Matches (mPSMs) and demonstrate its utility toward addressing the peptide identification problem in mixture spectra. In particular, we show how to extend the generating approach for mixture from two peptides. We focus on this relatively simple case of mixture spectra because it accounts for a large fraction of mixture spectra presented in traditional DDA workflows (5). This allows us to test and develop algorithmic concepts using readily-available DDA data because data with more complex mixture spectra such as those from DIA workflows (11) is still not widely available in public repositories.  相似文献   

4.
5.
Current analytical strategies for collecting proteomic data using data-dependent acquisition (DDA) are limited by the low analytical reproducibility of the method. Proteomic discovery efforts that exploit the benefits of DDA, such as providing peptide sequence information, but that enable improved analytical reproducibility, represent an ideal scenario for maximizing measureable peptide identifications in “shotgun”-type proteomic studies. Therefore, we propose an analytical workflow combining DDA with retention time aligned extracted ion chromatogram (XIC) areas obtained from high mass accuracy MS1 data acquired in parallel. We applied this workflow to the analyses of sample matrixes prepared from mouse blood plasma and brain tissues and observed increases in peptide detection of up to 30.5% due to the comparison of peptide MS1 XIC areas following retention time alignment of co-identified peptides. Furthermore, we show that the approach is quantitative using peptide standards diluted into a complex matrix. These data revealed that peptide MS1 XIC areas provide linear response of over three orders of magnitude down to low femtomole (fmol) levels. These findings argue that augmenting “shotgun” proteomic workflows with retention time alignment of peptide identifications and comparative analyses of corresponding peptide MS1 XIC areas improve the analytical performance of global proteomic discovery methods using DDA.Label-free methods in mass spectrometry-based proteomics, such as those used in common “shotgun” proteomic studies, provide peptide sequence information as well as relative measurements of peptide abundance (13). A common data acquisition strategy is based on data-dependent acquisition (DDA)1 where the most abundant precursor ions are selected for tandem mass spectrometry (MS/MS) analysis (12). DDA attempts to minimize redundant peptide precursor selection and maximize the depth of proteome coverage (2). However, the analytical reproducibility of peptide identifications obtained using DDA-based methods result in <75% overlap between technical replicates (34). Comparisons of peptide identifications between replicate analyses have shown that the rate of new peptide identifications increases sharply following two replicate sample injections and gradually tapers off after approximately five replicate injections (4). This phenomenon is due, in part, to the semirandom sampling of peptides in a DDA experiment (5).Alternate label-free methods focused on measuring the abundance of intact peptide ions, such as the accurate mass and time tag (AMT) approach (68, 42), are aimed at differential analyses of extracted ion chromatogram (XIC) areas integrated from high mass accuracy peptide precursor mass spectra (MS1 spectra) exhibiting discrete chromatographic elution times. This method is particularly powerful for the analysis of post-translationally modified (PTM) peptides as pairing the low abundance of PTM candidates with the variable nature of DDA complicates comparisons between samples (9, 43). However, label-free strategies focused on the analysis of peptide MS1 XIC areas are dependent on a priori knowledge of peptide ions and retention times (210). Thus, prospective analyses of samples are needed to assess peptides and their respective retention times. This prospective analysis may not be possible for reagent-limited samples. Further, the usage of previously established peptide features in the analysis of different sample types can be confounded by unknown matrix effects that can produce variable retention time characteristics and peptide ion suppression (2). Therefore, proteomic strategies that make use of DDA, to provide peptide sequence information and identify features within the sample, but that also use MS1 data for comparisons between samples, represent an ideal combination for maximizing measureable peptide identification events in “shotgun” proteomic discovery analyses.Here we describe an analytical workflow that combines traditional DDA methods with the analysis of retention time aligned XIC areas extracted from high mass accuracy peptide precursor MS1 spectra. This method resulted in a 25.1% (±6.6%) increase in measureable peptide identification events across samples of diverse composition because of the inferential extraction of peptide MS1 XIC areas in sample sets lacking corresponding MS/MS events. These findings were observed in measurements of peptide MS1 XIC abundances using sample types ranging from tryptic digests of olfactory bulb tissues dissected from Homer2 knockout and wild-type mice to mouse blood plasma exhibiting differential levels of hemolysis. We further establish that this method is quantitative using a dilution series of known bovine standard peptide concentrations spiked into mouse blood plasma. These data show that comparative analysis between samples should be performed using peptide MS1 data as opposed to semirandomly sampled peptide MS/MS data. This approach maximizes the number of peptides that can be compared between samples.  相似文献   

6.
Based on conventional data-dependent acquisition strategy of shotgun proteomics, we present a new workflow DeMix, which significantly increases the efficiency of peptide identification for in-depth shotgun analysis of complex proteomes. Capitalizing on the high resolution and mass accuracy of Orbitrap-based tandem mass spectrometry, we developed a simple deconvolution method of “cloning” chimeric tandem spectra for cofragmented peptides. Additional to a database search, a simple rescoring scheme utilizes mass accuracy and converts the unwanted cofragmenting events into a surprising advantage of multiplexing. With the combination of cloning and rescoring, we obtained on average nine peptide-spectrum matches per second on a Q-Exactive workbench, whereas the actual MS/MS acquisition rate was close to seven spectra per second. This efficiency boost to 1.24 identified peptides per MS/MS spectrum enabled analysis of over 5000 human proteins in single-dimensional LC-MS/MS shotgun experiments with an only two-hour gradient. These findings suggest a change in the dominant “one MS/MS spectrum - one peptide” paradigm for data acquisition and analysis in shotgun data-dependent proteomics. DeMix also demonstrated higher robustness than conventional approaches in terms of lower variation among the results of consecutive LC-MS/MS runs.Shotgun proteomics analysis based on a combination of high performance liquid chromatography and tandem mass spectrometry (MS/MS) (1) has achieved remarkable speed and efficiency (27). In a single four-hour long high performance liquid chromatography-MS/MS run, over 40,000 peptides and 5000 proteins can be identified using a high-resolution Orbitrap mass spectrometer with data-dependent acquisition (DDA)1 (2, 3). However, in a typical LC-MS analysis of unfractionated human cell lysate, over 100,000 individual peptide isotopic patterns can be detected (4), which corresponds to simultaneous elution of hundreds of peptides. With this complexity, a mass spectrometer needs to achieve ≥25 Hz MS/MS acquisition rate to fully sample all the detectable peptides, and ≥17 Hz to cover reasonably abundant ones (4). Although this acquisition rate is reachable by modern time-of-flight (TOF) instruments, the reported DDA identification results do not encompass all expected peptides. Recently, the next-generation Orbitrap instrument, working at 20 Hz MS/MS acquisition rate, demonstrated nearly full profiling of yeast proteome using an 80 min gradient, which opened the way for comprehensive analysis of human proteome in a time efficient manner (5).During the high performance liquid chromatography-MS/MS DDA analysis of complex samples, high density of co-eluting peptides results in a high probability for two or more peptides to overlap within an MS/MS isolation window. With the commonly used ±1.0–2.0 Th isolation windows, most MS/MS spectra are chimeric (4, 810), with cofragmenting precursors being naturally multiplexed. However, as has been discussed previously (9, 10), the cofragmentation events are currently ignored in most of the conventional analysis workflows. According to the prevailing assumption of “one MS/MS spectrum–one peptide,” chimeric MS/MS spectra are generally unwelcome in DDA, because the product ions from different precursors may interfere with the assignment of MS/MS fragment identities, increasing the rate of false discoveries in database search (8, 9). In some studies, the precursor isolation width was set as narrow as ±0.35 Th to prevent unwanted ions from being coselected, fragmented or detected (4, 5).On the contrary, multiplexing by cofragmentation is considered to be one of the solid advantages in data-independent acquisition (DIA) (1013). In several commonly used DIA methods, the precursor ion selection windows are set much wider than in DDA: from 25 Th as in SWATH (12), to extremely broad range as in AIF (13). In order to use the benefit of MS/MS multiplexing in DDA, several approaches have been proposed to deconvolute chimeric MS/MS spectra. In “alternative peptide identification” method implemented in Percolator (14), a machine learning algorithm reranks and rescores peptide-spectrum matches (PSMs) obtained from one or more MS/MS search engines. But the deconvolution in Percolator is limited to cofragmented peptides with masses differing from the target peptide by the tolerance of the database search, which can be as narrow as a few ppm. The “active demultiplexing” method proposed by Ledvina et al. (15) actively separates MS/MS data from several precursors using masses of complementary fragments. However, higher-energy collisional dissociation often produces MS/MS spectra with too few complementary pairs for reliable peptide identification. The “MixDB” method introduces a sophisticated new search engine, also with a machine learning algorithm (9). And the “second peptide identification” method implemented in Andromeda/MaxQuant workflow (16) submits the same dataset to the search engine several times based on the list of chromatographic peptide features, subtracting assigned MS/MS peaks after each identification round. This approach is similar to the ProbIDTree search engine that also performed iterative identification while removing assigned peaks after each round of identification (17).One important factor for spectral deconvolution that has not been fully utilized in most conventional workflows is the excellent mass accuracy achievable with modern high-resolution mass spectrometry (18). An Orbitrap Fourier-transform mass spectrometer can provide mass accuracy in the range of hundreds of ppb (parts per billion) for mass peaks with high signal-to-noise (S/N) ratio (19). However, the mass error of peaks with lower S/N ratios can be significantly higher and exceed 1 ppm. Despite this dependence of the mass accuracy from the S/N level, most MS and MS/MS search engines only allow users to set hard cut-off values for the mass error tolerances. Moreover, some search engines do not provide the option of choosing a relative error tolerance for MS/MS fragments. Such negligent treatment of mass accuracy reduces the analytical power of high accuracy experiments (18).Identification results coming from different MS/MS search engines are sometimes not consistent because of different statistical assumptions used in scoring PSMs. Introduction of tools integrating the results of different search engines (14, 20, 21) makes the data interpretation even more complex and opaque for the user. The opposite trend—simplification of MS/MS data interpretation—is therefore a welcome development. For example, an extremely straightforward algorithm recently proposed by Wenger et al. (22) demonstrated a surprisingly high performance in peptide identification, even though it is only marginally more complex than simply counting the number of matches of theoretical fragment peaks in high resolution MS/MS, without any a priori statistical assumption.In order to take advantage of natural multiplexing of MS/MS spectra in DDA, as well as properly utilize high accuracy of Orbitrap-based mass spectrometry, we developed a simple and robust data analysis workflow DeMix. It is presented in Fig. 1 as an expansion of the conventional workflow. Principles of some of the processes used by the workflow are borrowed from other approaches, including the custom-made mass peak centroiding (20), chromatographic feature detection (19, 20), and two-pass database search with the first limited pass to provide a “software lock mass” for mass scale recalibration (23).Open in a separate windowFig. 1.An overview of the DeMix workflow that expands the conventional workflow, shown by the dashed line. Processes are colored in purple for TOPP, red for search engine (Morpheus/Mascot/MS-GF+), and blue for in-house programs.In DeMix workflow, the deconvolution of chimeric MS/MS spectra consists of simply “cloning” an MS/MS spectrum if a potential cofragmented peptide is detected. The list of candidate peptide precursors is generated from chromatographic feature detection, as in the MaxQuant/Andromeda workflow (16, 19), but using The OpenMS Proteomics Pipeline (TOPP) (20, 24). During the cloning, the precursor is replaced by the new candidate, but no changes in the MS/MS fragment list are made, and therefore the cloned MS/MS spectra remain chimeric. Processing such spectra requires a search engine tolerant to the presence of unassigned peaks, as such peaks are always expected when multiple precursors cofragment. Thus, we chose Morpheus (22) as a search engine. Based on the original search algorithm, we implement a reformed scoring scheme: Morpheus-AS (advanced scoring). It inherits all the basic principles from Morpheus but deeper utilizes the high mass accuracy of the data. This kind of database search removes the necessity of spectral processing for physical separation of MS/MS data into multiple subspectra (15), or consecutive subtraction of peaks (16, 17).Despite the fact that DeMix workflow is largely a combination of known approaches, it provides remarkable improvement compared with the state-of-the-art. On our Orbitrap Q-Exactive workbench, testing on a benchmark dataset of two-hour single-dimension LC-MS/MS experiments from HeLa cell lysate, we identified on average 1.24 peptide per MS/MS spectrum, breaking the “one MS/MS spectrum–one peptide” paradigm on the level of whole data set. At 1% false discovery rate (FDR), we obtained on average nine PSMs per second (at the actual acquisition rate of ca. seven MS/MS spectra per second), and detected 40 human proteins per minute.  相似文献   

7.
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   

8.
9.
Quantitative analysis of discovery-based proteomic workflows now relies on high-throughput large-scale methods for identification and quantitation of proteins and post-translational modifications. Advancements in label-free quantitative techniques, using either data-dependent or data-independent mass spectrometric acquisitions, have coincided with improved instrumentation featuring greater precision, increased mass accuracy, and faster scan speeds. We recently reported on a new quantitative method called MS1 Filtering (Schilling et al. (2012) Mol. Cell. Proteomics 11, 202–214) for processing data-independent MS1 ion intensity chromatograms from peptide analytes using the Skyline software platform. In contrast, data-independent acquisitions from MS2 scans, or SWATH, can quantify all fragment ion intensities when reference spectra are available. As each SWATH acquisition cycle typically contains an MS1 scan, these two independent label-free quantitative approaches can be acquired in a single experiment. Here, we have expanded the capability of Skyline to extract both MS1 and MS2 ion intensity chromatograms from a single SWATH data-independent acquisition in an Integrated Dual Scan Analysis approach. The performance of both MS1 and MS2 data was examined in simple and complex samples using standard concentration curves. Cases of interferences in MS1 and MS2 ion intensity data were assessed, as were the differentiation and quantitation of phosphopeptide isomers in MS2 scan data. In addition, we demonstrated an approach for optimization of SWATH m/z window sizes to reduce interferences using MS1 scans as a guide. Finally, a correlation analysis was performed on both MS1 and MS2 ion intensity data obtained from SWATH acquisitions on a complex mixture using a linear model that automatically removes signals containing interferences. This work demonstrates the practical advantages of properly acquiring and processing MS1 precursor data in addition to MS2 fragment ion intensity data in a data-independent acquisition (SWATH), and provides an approach to simultaneously obtain independent measurements of relative peptide abundance from a single experiment.Mass spectrometry is the leading technology for large-scale identification and quantitation of proteins and post-translational modifications (PTMs)1 in biological systems (1, 2). Although several types of experimental designs are employed in such workflows, most large-scale applications use data-dependent acquisitions (DDA) where peptide precursors are first identified in the MS1 scan and one or more peaks are then selected for subsequent fragmentation to generate their corresponding MS2 spectra. In experiments using DDA, one can employ either chemical/metabolic labeling or label-free strategies for relative quantitation of peptides (and proteins) (3, 4). Depending on the type of labeling approach employed, i.e. metabolic labeling with SILAC or postmetabolic labeling with ICAT or isobaric tags such as iTRAQ or TMT, the relative quantitation of these peptides are made using either MS1 or MS2 ion intensity data (47). Label-free quantitative techniques have until recently been based entirely on integrated ion intensity measurements of precursors in the MS1 scan, or in the case of spectral counting the number of assigned MS2 spectra (3, 8, 9).Label-free approaches have recently generated more widespread interest (1012), in part because of their adaptability to a wide range of proteomic workflows, including human samples that are not amenable to most metabolic labeling techniques, or where chemical labeling may be cost prohibitive and/or interfere with subsequent enrichment steps (11, 13). However the use of DDA for label-free quantitation is also susceptible to several limitations including insufficient reproducibility because of under-sampling, digestion efficiency, as well as misidentifications (14, 15). Moreover, low ion abundance may prohibit peptide selection, especially in complex samples (14). These limitations often present challenges in data analysis when making comparisons across samples, or when a peptide is sampled in only one of the study conditions.To address the challenges in obtaining more comprehensive sampling in MS1 space, Purvine et al. first demonstrated the ability to obtain sequence information from peptides fragmented across the entire m/z range using “shotgun or parallel collision-induced dissociation (CID)” on an orthogonal time of flight instrument (16). Shortly thereafter Venable et al. reported on a data independent acquisition methodology to limit the complexity of the MS2 scan by using a segmented approach for the sequential isolation and fragmentation of all peptides in a defined precursor window (e.g. 10 m/z) using an ion trap mass spectrometer (17). However, the proper implementation of this DIA technique suffered from technical limitations of instruments available at that time, including slow acquisition rates and low MS2 resolution that made systematic product ion extraction problematic. To alleviate the challenge of long duty cycles in DIAs, researchers at the Waters Corporation adopted an alternative approach by rapidly switching between low (MS1) and high energy (MS2) scans and then using proprietary software to align peptide precursor and fragment ion information to determine peptide sequences (18, 19). Recent mass spectrometry innovations in efficient high-speed scanning capabilities, together with high-resolution data acquisition of both MS1 and MS2 scans, and multiplexing of scan windows have overcome many of these limitations (10, 20, 21). Moreover, the simultaneous development of novel software solutions for extracting ion intensity chromatograms based on spectral libraries has enabled the use of DIA for large-scale label free quantitation of multiple peptide analytes (21, 22). In addition to targeting specific peptides from a previously generated peptide spectral library, the data can also be reexamined (i.e. post-acquisition) for additional peptides of interest as new reference data emerges. On the SCIEX TripleTOF 5600, a quadrupole orthogonal time-of-flight mass spectrometer, this technique has been optimized and extended to what is called ‘SWATH MS2′ based on a combination of new technical and software improvements (10, 22).In a DIA experiment a MS1 survey scan is carried out across the mass range followed by a SWATH MS2 acquisition series, however the cycle time of the MS1 scan is dramatically shortened compared with DDA type experiments. The Q1 quadrupole is set to transmit a wider window, typically Δ25 m/z, to the collision cell in incremental steps over the full mass range. Therefore the MS/MS spectra produced during a SWATH MS2 acquisition are of much greater complexity as the MS/MS spectra are a composite of all fragment ions produced from peptide analytes with molecular ions within the selected MS1 m/z window. The cycle of data independent MS1 survey scans and SWATH MS2 scans is repeated throughout the entire LC-MS acquisition. Fragment ion information contained in these SWATH MS2 spectra can be used to uniquely identify specific peptides by comparisons to reference spectra or spectral libraries. Moreover, ion intensities of these fragment ions can also be used for quantitation. Although MS2 typically increases selectivity and reduces the chemical noise often observed in MS1 scans, quantifying peptides from SWATH MS2 scans can be problematic because of the presence of interferences in one or more fragment ions or decreased ion intensity of MS2 scans as compared with the MS1 precursor ion abundance.To partially alleviate some of these limitations in SWATH MS2 scan quantitation it is potentially advantageous to exploit MS1 ion intensity data, which is acquired independently as part of each SWATH scan cycle. Recently, our laboratories and others have developed label free quantitation tools for data dependent acquisitions (11, 12, 23) using MS1 ion intensity data. For example, the MS1 Filtering algorithm uses expanded features in the open source software application Skyline (11, 24). Skyline MS1 Filtering processes precursor ion intensity chromatograms of peptide analytes from full scan mass spectral data acquired during data dependent acquisitions by LC MS/MS. New graphical tools were developed within Skyline to enable visual inspection and manual interrogation and integration of extracted ion chromatograms across multiple acquisitions. MS1 Filtering was subsequently shown to have excellent linear response across several orders of magnitude with limits of detection in the low attomole range (11). We, and others, have demonstrated the utility of this method for carrying out large-scale quantitation of peptide analytes across a range of applications (2528). However, quantifying peptides based on MS1 precursor ion intensities can be compromised by a low signal-to-noise ratio. This is particularly the case when quantifying low abundance peptides in a complex sample where the MS1 ion “background” signal is high, or when chromatograms contain interferences, or partial overlap of multiple target precursor ions.Currently MS1 scans are underutilized or even deemphasized by some vendors during DIA workflows. However, we believe an opportunity exists that would improve data-independent acquisitions (DIA) experiments by including MS1 ion intensity data in the final data processing of LC-MS/MS acquisitions. Therefore, to address this possibility, we have adapted Skyline to efficiently extract and process both precursor and product ion chromatograms for label free quantitation across multiple samples. The graphical tools and features originally developed for SRM and MS1 Filtering experiments have been expanded to process DIA data sets from multiple vendors including SCIEX, Thermo, Waters, Bruker, and Agilent. These expanded features provide a single platform for data mining of targeted proteomics using both the MS1 and MS2 scans that we call Integrated Dual Scan Analysis, or IDSA. As a test of this approach, a series of SWATH MS2 acquisitions of simple and complex mixtures was analyzed on an SCIEX TripleTOF 5600 mass spectrometer. We also investigated the use of MS2 scans for differentiating a case of phosphopeptide isomers that are indistinguishable at the MS1 level. In addition, we investigated whether smaller SWATH m/z windows would provide more reliable quantitative data in these cases by reducing the number of potential interferences. Lastly, we performed a statistical assessment of the accuracy and reproducibility of the estimated (log) fold change of mitochondrial lysates from mouse liver at different concentration levels to better assess the overall value of acquiring MS1 and MS2 data in combination and as independent measurements during DIA experiments.  相似文献   

10.
The use of ultraviolet photodissociation (UVPD) for the activation and dissociation of peptide anions is evaluated for broader coverage of the proteome. To facilitate interpretation and assignment of the resulting UVPD mass spectra of peptide anions, the MassMatrix database search algorithm was modified to allow automated analysis of negative polarity MS/MS spectra. The new UVPD algorithms were developed based on the MassMatrix database search engine by adding specific fragmentation pathways for UVPD. The new UVPD fragmentation pathways in MassMatrix were rigorously and statistically optimized using two large data sets with high mass accuracy and high mass resolution for both MS1 and MS2 data acquired on an Orbitrap mass spectrometer for complex Halobacterium and HeLa proteome samples. Negative mode UVPD led to the identification of 3663 and 2350 peptides for the Halo and HeLa tryptic digests, respectively, corresponding to 655 and 645 peptides that were unique when compared with electron transfer dissociation (ETD), higher energy collision-induced dissociation, and collision-induced dissociation results for the same digests analyzed in the positive mode. In sum, 805 and 619 proteins were identified via UVPD for the Halobacterium and HeLa samples, respectively, with 49 and 50 unique proteins identified in contrast to the more conventional MS/MS methods. The algorithm also features automated charge determination for low mass accuracy data, precursor filtering (including intact charge-reduced peaks), and the ability to combine both positive and negative MS/MS spectra into a single search, and it is freely open to the public. The accuracy and specificity of the MassMatrix UVPD search algorithm was also assessed for low resolution, low mass accuracy data on a linear ion trap. Analysis of a known mixture of three mitogen-activated kinases yielded similar sequence coverage percentages for UVPD of peptide anions versus conventional collision-induced dissociation of peptide cations, and when these methods were combined into a single search, an increase of up to 13% sequence coverage was observed for the kinases. The ability to sequence peptide anions and cations in alternating scans in the same chromatographic run was also demonstrated. Because ETD has a significant bias toward identifying highly basic peptides, negative UVPD was used to improve the identification of the more acidic peptides in conjunction with positive ETD for the more basic species. In this case, tryptic peptides from the cytosolic section of HeLa cells were analyzed by polarity switching nanoLC-MS/MS utilizing ETD for cation sequencing and UVPD for anion sequencing. Relative to searching using ETD alone, positive/negative polarity switching significantly improved sequence coverages across identified proteins, resulting in a 33% increase in unique peptide identifications and more than twice the number of peptide spectral matches.The advent of new high-performance tandem mass spectrometers equipped with the most versatile collision- and electron-based activation methods and ever more powerful database search algorithms has catalyzed tremendous progress in the field of proteomics (14). Despite these advances in instrumentation and methodologies, there are few methods that fully exploit the information available from the acidic proteome or acidic regions of proteins. Typical high-throughput, bottom-up workflows consist of the chromatographic separation of complex mixtures of digested proteins followed by online mass spectrometry (MS) and MSn analysis. This bottom-up approach remains the most popular strategy for protein identification, biomarker discovery, quantitative proteomics, and elucidation of post-translational modifications. To date, proteome characterization via mass spectrometry has overwhelmingly focused on the analysis of peptide cations (5), resulting in an inherent bias toward basic peptides that easily ionize under acidic mobile phase conditions and positive polarity MS settings. Given that ∼50% of peptides/proteins are naturally acidic (6) and that many of the most important post-translational modifications (e.g. phosphorylation, acetylation, sulfonation, etc.) significantly decrease the isoelectric points of peptides (7, 8), there is a compelling need for better analytical methodologies for characterization of the acidic proteome.A principal reason for the shortage of methods for peptide anion characterization is the lack of MS/MS techniques suitable for the efficient and predictable dissociation of peptide anions. Although there are a growing array of new ion activation methods for the dissociation of peptides, most have been developed for the analysis of positively charged peptides. Collision-induced dissociation (CID)1 of peptide anions, for example, often yields unpredictable or uninformative fragmentation behavior, with spectra dominated by neutral losses from both precursor and product ions (9), resulting in insufficient peptide sequence information. The two most promising new electron-based methods, electron-capture dissociation and electron-transfer dissociation (ETD), are applicable only to positively charged ions, not to anions (1013). Because of the known inadequacy of CID and the lack of feasibility of electron-capture dissociation and ETD for peptide anion sequencing, several alternative MSn methods have been developed recently. Electron detachment dissociation using high-energy electrons to induce backbone cleavages was developed for peptide anions (14, 15). Another new technique, negative ETD, entails reactions of radical cation reagents with peptide anions to promote electron transfer from the peptide to the reagent that causes radical-directed dissociation (16, 17). Activated-electron photodetachment dissociation, an MS3 technique, uses UV irradiation to produce intact peptide radical anions, which are then collisionally activated (18, 19). Although they represent inroads in the characterization of peptide anions, these methods also suffer from several significant shortcomings. Electron detachment dissociation and activated-electron photodetachment dissociation are both low-efficiency methods that require long averaging cycles and activation times that range from half a second to multiple seconds, impeding the integration of these methods with chromatographic timescales (1419). In addition, the fragmentation patterns frequently yield many high-abundance neutral losses from product ions, which clutter the spectra (1417), and few sequence ions (14, 18, 19). Recently, we reported the use of 193-nm photons (ultraviolet photodissociation (UVPD)) for peptide anion activation, which was shown to yield rich and predictable fragmentation patterns with high sequence coverage on a fast liquid chromatographic timeline (20). This method showed promise for a range of peptide charge states (i.e. from 3- to 1-), as well as for both unmodified and phosphorylated species.Several widely used or commercial database searching techniques are available for automated “bottom-up” analysis of peptide cations; SEQUEST (21), MASCOT (22), OMSSA (23), X! Tandem (24), and MASPIC (25) are all popular choices and yield comparable results (26). MassMatrix (27), a recently introduced searching algorithm, uses a mass accuracy sensitive probability-based scoring scheme for both the total number of matched product ions and the total abundance of matched products. This searching method also utilizes LC retention times to filter false positive peptide matches (28) and has been shown to yield results comparable to or better than those obtained with SEQUEST, MASCOT, OMSSA, and X! Tandem (29). Despite the ongoing innovation in automated peptide cation analysis, there is a lack of publically available methods for automated peptide anion analysis.In this work, we have modified the mass accuracy sensitive probabilistic MassMatrix algorithms to allow database searching of negative polarity MS/MS spectra. The algorithm is specific to the fragmentation behavior generated from 193-nm UVPD of peptide anions. The UVPD pathways in MassMatrix were rigorously and statistically optimized using two large data sets with high mass accuracy and high mass resolution for both MS1 and MS2 data acquired on an Orbitrap mass spectrometer for complex HeLa and Halo proteome samples. For low mass accuracy/low mass resolution data, we also incorporated a charge-state-filtering algorithm that identifies the charge state of each MS/MS spectrum based on the fragmentation patterns prior to searching. MassMatrix not only can analyze both positive and negative polarity LC-MS/MS files separately, but also can combine files from different polarities and different dissociation methods into a single search, thus maximizing the information content for a given proteomics experiment. The explicit incorporation of mass accuracy in the scores for the UVPD MS/MS spectra of peptide anions increases peptide assignments and identifications. Finally, we showcase the utility of integrating MassMatrix searching with positive/negative polarity MS/MS switching (i.e. data-dependent positive ETD and negative UVPD during a single proteomic LC-MS/MS run). MassMatrix is available to the public as a free search engine online.  相似文献   

11.
The combination of chemical cross-linking and mass spectrometry has recently been shown to constitute a powerful tool for studying protein–protein interactions and elucidating the structure of large protein complexes. However, computational methods for interpreting the complex MS/MS spectra from linked peptides are still in their infancy, making the high-throughput application of this approach largely impractical. Because of the lack of large annotated datasets, most current approaches do not capture the specific fragmentation patterns of linked peptides and therefore are not optimal for the identification of cross-linked peptides. Here we propose a generic approach to address this problem and demonstrate it using disulfide-bridged peptide libraries to (i) efficiently generate large mass spectral reference data for linked peptides at a low cost and (ii) automatically train an algorithm that can efficiently and accurately identify linked peptides from MS/MS spectra. We show that using this approach we were able to identify thousands of MS/MS spectra from disulfide-bridged peptides through comparison with proteome-scale sequence databases and significantly improve the sensitivity of cross-linked peptide identification. This allowed us to identify 60% more direct pairwise interactions between the protein subunits in the 20S proteasome complex than existing tools on cross-linking studies of the proteasome complexes. The basic framework of this approach and the MS/MS reference dataset generated should be valuable resources for the future development of new tools for the identification of linked peptides.The study of protein–protein interactions is crucial to understanding how cellular systems function because proteins act in concert through a highly organized set of interactions. Most cellular processes are carried out by large macromolecular assemblies and regulated through complex cascades of transient protein–protein interactions (1). In the past several years numerous high-throughput studies have pioneered the systematic characterization of protein–protein interactions in model organisms (24). Such studies mainly utilize two techniques: the yeast two-hybrid system, which aims at identifying binary interactions (5), and affinity purification combined with tandem mass spectrometry analysis for the identification of multi-protein assemblies (68). Together these led to a rapid expansion of known protein–protein interactions in human and other model organisms. Patche and Aloy recently estimated that there are more than one million interactions catalogued to date (9).But despite rapid progress, most current techniques allow one to determine only whether proteins interact, which is only the first step toward understanding how proteins interact. A more complete picture comes from characterizing the three-dimensional structures of protein complexes, which provide mechanistic insights that govern how interactions occur and the high specificity observed inside the cell. Traditionally the gold-standard methods used to solve protein structures are x-ray crystallography and NMR, and there have been several efforts similar to structural genomics (10) aiming to comprehensively solve the structures of protein complexes (11, 12). Although there has been accelerated growth of structures for protein monomers in the Protein Data Bank in recent years (11), the growth of structures for protein complexes has remained relatively small (9). Many factors, including their large size, transient nature, and dynamics of interactions, have prevented many complexes from being solved via traditional approaches in structural biology. Thus, the development of complementary analytical techniques with which to probe the structure of large protein complexes continues to evolve (1318).Recent developments have advanced the analysis of protein structures and interaction by combining cross-linking and tandem mass spectrometry (17, 1924). The basic idea behind this technique is to capture and identify pairs of amino acid residues that are spatially close to each other. When these linked pairs of residues are from the same protein (intraprotein cross-links), they provide distance constraints that help one infer the possible conformations of protein structures. Conversely, when pairs of residues come from different proteins (interprotein cross-links), they provide information about how proteins interact with one another. Although cross-linking strategies date back almost a decade (25, 26), difficulty in analyzing the complex MS/MS spectrum generated from linked peptides made this approach challenging, and therefore it was not widely used. With recent advances in mass spectrometry instrumentation, there has been renewed interest in employing this strategy to determine protein structures and identify protein–protein interactions. However, most studies thus far have been focused on purified protein complexes. With today''s mass spectrometers being capable of analyzing tens of thousands of spectra in a single experiment, it is now potentially feasible to extend this approach to the analysis of complex biological samples. Researchers have tried to realize this goal using both experimental and computational approaches. Indeed, a plethora of chemical cross-linking reagents are now available for stabilizing these complexes, and some are designed to allow for easier peptide identification when employed in concert with MS analysis (20, 27, 28). There have also been several recent efforts to develop computational methods for the automatic identification of linked peptides from MS/MS spectra (2936). However, because of the lack of large annotated training data, most approaches to date either borrow fragmentation models learned from unlinked, linear peptides or learn the fragmentation statistics from training data of limited size (30, 37), which might not generalize well across different samples. In some cases it is possible to generate relatively large training data, but it is often very labor intensive and involves hundreds of separate LC-MS/MS runs (36). Here, employing disulfide-bridged peptides as an example, we propose a novel method that uses a combinatorial peptide library to (a) efficiently generate a large mass spectral reference dataset for linked peptides and (b) use these data to automatically train our new algorithm, MXDB, which can efficiently and accurately identify linked peptides from MS/MS spectra.  相似文献   

12.
13.
Isobaric labeling techniques coupled with high-resolution mass spectrometry have been widely employed in proteomic workflows requiring relative quantification. For each high-resolution tandem mass spectrum (MS/MS), isobaric labeling techniques can be used not only to quantify the peptide from different samples by reporter ions, but also to identify the peptide it is derived from. Because the ions related to isobaric labeling may act as noise in database searching, the MS/MS spectrum should be preprocessed before peptide or protein identification. In this article, we demonstrate that there are a lot of high-frequency, high-abundance isobaric related ions in the MS/MS spectrum, and removing isobaric related ions combined with deisotoping and deconvolution in MS/MS preprocessing procedures significantly improves the peptide/protein identification sensitivity. The user-friendly software package TurboRaw2MGF (v2.0) has been implemented for converting raw TIC data files to mascot generic format files and can be downloaded for free from https://github.com/shengqh/RCPA.Tools/releases as part of the software suite ProteomicsTools. The data have been deposited to the ProteomeXchange with identifier PXD000994.Mass spectrometry-based proteomics has been widely applied to investigate protein mixtures derived from tissue, cell lysates, or from body fluids (1, 2). Liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS)1 is the most popular strategy for protein/peptide mixtures analysis in shotgun proteomics (3). Large-scale protein/peptide mixtures are separated by liquid chromatography followed by online detection by tandem mass spectrometry. The capabilities of proteomics rely greatly on the performance of the mass spectrometer. With the improvement of MS technology, proteomics has benefited significantly from the high-resolution and excellent mass accuracy (4). In recent years, based on the higher efficiency of higher energy collision dissociation (HCD), a new “high–high” strategy (high-resolution MS as well as MS/MS(tandem MS)) has been applied instead of the “high–low” strategy (high-resolution MS, i.e. in Orbitrap, and low-resolution MS/MS, i.e. in ion trap) to obtain high quality tandem MS/MS data as well as full MS in shotgun proteomics. Both full MS scans and MS/MS scans can be performed, and the whole cycle time of MS detection is very compatible with the chromatographic time scale (5).High-resolution measurement is one of the most important features in mass spectrometric application. In this high–high strategy, high-resolution and accurate spectra will be achieved in tandem MS/MS scans as well as full MS scans, which makes isotopic peaks distinguishable from one another, thus enabling the easy calculation of precise charge states and monoisotopic mass. During an LC-MS/MS experiment, a multiply charged precursor ion (peptide) is usually isolated and fragmented, and then the multiple charge states of the fragment ions are generated and collected. After full extraction of peak lists from original tandem mass spectra, the commonly used search engines (i.e. Mascot (6), Sequest (7)) have no capability to distinguish isotopic peaks and recognize charge states, so all of the product ions are considered as all charge state hypotheses during the database search for protein identification. These multiple charge states of fragment ions and their isotopic cluster peaks can be incorrectly assigned by the search engine, which can cause false peptide identification. To overcome this issue, data preprocessing of the high-resolution MS/MS spectra is required before submitting them for identification. There are usually two major preprocessing steps used for high-resolution MS/MS data: deisotoping and deconvolution (8, 9). Deisotoping of spectra removes all isotopic peaks except monoisotopic peaks from multi-isotopic peaks. Deconvolution of spectra translates multiply charged ions to singly charged ions and also accumulates the intensity of fragment ions by summing up all the intensities from their multiply charged states. After performing these two data-preprocessing steps, the resulting spectra is simpler and cleaner and allows more precise database searching and accurate bioinformatics analysis.With the capacity to analyze multiple samples simultaneously, stable isotope labeling approaches have been widely used in quantitative proteomics. Stable isotope labeling approaches are categorized as metabolic labeling (SILAC, stable isotope labeling by amino acids in cell culture) and chemical labeling (10, 11). The peptides labeled by the SILAC approach are quantified by precursor ions in full MS spectra, whereas peptides that have been isobarically labeled using chemical means are quantified by reporter ions in MS/MS spectra. There are two similar isobaric chemical labeling methods: (1) isobaric tag for relative and absolute quantification (iTRAQ), and (2) tandem mass tag (TMT) (12, 13). These reagents contain an amino-reactive group that specifically reacts with N-terminal amino groups and epilson-amino groups of lysine residues to label digested peptides in a typical shotgun proteomics experiment. There are four different channels of isobaric tags: TMT two-plex, iTRAQ four-plex, TMT six-plex, and iTRAQ eight-plex (1216). The number before “plex” denotes the number of samples that can be analyzed by the mass spectrum simultaneously. Peptides labeled with different isotopic variants of the tag show identical or similar mass and appear as a single peak in full scans. This single peak may be selected for subsequent MS/MS analysis. In an MS/MS scan, the mass of reporter ions (114 to 117 for iTRAQ four-plex, 113 to 121 for iTRAQ eight-plex, and 126 to 131for TMT six-plex upon CID or HCD activation) are associated with corresponding samples, and the intensities represent the relative abundances of the labeled peptides. Meanwhile, the other ions from the MS/MS spectra can be used for peptide identification. Because of the multiplexing capability, isobaric labeling methods combined with bottom-up proteomics have been widely applied for accurate quantification of proteins on a global scale (14, 1719). Although mostly associated with peptide labeling, these isobaric labeling methods have also been applied at protein level (2023).For the proteomic analysis of isobarically labeled peptides/proteins in “high–high” MS strategy, the common consensus is that accurate reporter ions can contribute to more accurate quantification. However, there is no evidence to show how the ions related to isobaric labeling affect the peptide/protein identification and what preprocessing steps should be taken for high-resolution isobarically labeled MS/MS. To demonstrate the effectiveness and importance of preprocessing, we examined how the combination of preprocessing steps improved peptide/protein sensitivity in database searching. Several combinatorial ways of data-preprocessing were applied for high-throughput data analysis including deisotoping to keep simple monoisotopic mass peaks, deconvolution of ions with multiple charge states, and preservation of top 10 peaks in every 100 Dalton mass range. After systematic analysis of high-resolution isobarically labeled spectra, we further processed the spectra and removed interferential ions that were not related to the peptide. Our results suggested that the preprocessing of isobarically labeled high-resolution tandem mass spectra significantly improved the peptide/protein identification sensitivity.  相似文献   

14.
Peptide and protein identification remains challenging in organisms with poorly annotated or rapidly evolving genomes, as are commonly encountered in environmental or biofuels research. Such limitations render tandem mass spectrometry (MS/MS) database search algorithms ineffective as they lack corresponding sequences required for peptide-spectrum matching. We address this challenge with the spectral networks approach to (1) match spectra of orthologous peptides across multiple related species and then (2) propagate peptide annotations from identified to unidentified spectra. We here present algorithms to assess the statistical significance of spectral alignments (Align-GF), reduce the impurity in spectral networks, and accurately estimate the error rate in propagated identifications. Analyzing three related Cyanothece species, a model organism for biohydrogen production, spectral networks identified peptides from highly divergent sequences from networks with dozens of variant peptides, including thousands of peptides in species lacking a sequenced genome. Our analysis further detected the presence of many novel putative peptides even in genomically characterized species, thus suggesting the possibility of gaps in our understanding of their proteomic and genomic expression. A web-based pipeline for spectral networks analysis is available at http://proteomics.ucsd.edu/software.Microorganisms have evolved their cellular metabolism to generate energy for life in unusual environments (1), and their capabilities are of great interest in the production of renewable bioenergy and could contribute toward managing the world''s current energy and climate crisis (2). Genomics studies have increased the number of sequenced bioenergy-related microbial genomes and revealed the possible biological reactions involved in bioenergy production (3). Studies of photosynthetic microorganisms, for example, have yielded insights into how they harvest solar energy and use it to produce bioenergy products (4). Despite this importance of microorganisms, the characterization of diverse microbial phenotypes by proteomics tandem mass spectrometry (MS/MS) has been limited. The dominant approaches for MS/MS analysis heavily rely on the availability of completely annotated genomes (i.e. accurate protein databases) (57), yet most microorganisms populating the planet have unsequenced or poorly annotated genomes. Thus it remains challenging to identify proteins from environmental and unculturable organisms.One solution to protein identification in a species with no sequenced genome is to use the genomes of closely related species (8). This requires matching MS/MS data to slightly different peptides in amino acid sequences (polymorphic, orthologous peptides); but matching shifted masses of peptides and their fragment ions is computationally expensive and challenging. Moreover, different species-specific post-translational modifications (PTMs)1 can make the cross-species identification more complex. The common computational approach is tolerantly matching de novo sequences derived from MS/MS data to the database while allowing for amino acid mutations and modifications (911). However, this approach critically depends on good de novo interpretations, which are nearly always partially incorrect and yield high-quality subsequences only for a small fraction of all spectra. The blind database search approach, developed to identify peptides with unexpected modifications, can also be used to directly match MS/MS data from unknown species to a database of closely related species, but its utilization is limited because of its exceptionally large search space (1218). These spectrum-database matching approaches to cross-species identification pose significant challenges in its speed and sensitivity with a huge database, which leads to a much longer search time and more false positive identifications (19, 20).As a complementary approach to spectrum-database matching, spectral library searching is an emerging and promising approach (21). A spectral library is a large collection of identified MS/MS spectra, and an unknown query spectrum can then be identified by direct spectral matching to the library. The great advantage of this approach is the reduction of search space and the use of fragmentation patterns of peptides. The spectral networks approach expands this concept to the identification of modified peptides in MS/MS data sets (22, 23). Spectral networks do not directly search a database, but groups MS/MS spectra by computing the pairwise similarity between MS/MS spectra of peptide variants and then constructs networks where each spectrum defines a node and each significant spectral pair, highly correlated in the fragmentation pattern, defines an edge (Fig. 1). In spectral networks, identification of spectra belonging to the same subnetwork should be related and thus the peptide sequence for an identified spectrum can be propagated to neighboring unidentified spectra.Open in a separate windowFig. 1.Overview of multi-species spectral networks. Nodes represent individual spectra and edges between nodes represent significant pairwise alignment between spectra; edges are labeled with amino acid mutations (dotted edges) or parent mass differences (solid edges). In spectral networks, a peptide and its related variants are ideally grouped into a single subnetwork. If at least one spectrum in a subnetwork is annotated (filled node), all the neighboring spectra (unfilled nodes) can potentially become identified by propagating the annotation over network edges. For example, all spectra in the subnetwork of “peptide A” (top left, blue network) can be annotated via up to three iterative propagations, first from A to {A1, A2, A3}, second from {A2, A3} to {A4, A5}, and third from {A4, A5} to A6. This paradigm can be equally applied to cross-species data analysis, as “peptide L” identified in species 1 (top middle, olive-colored network) is propagated to a node unidentified in species 2, identifying its orthologous “peptide l”, with a serine to alanine polymorphism. Thus, spectral networks enable the detection of orthologous peptide pairs between different species.We recently reported that a vast number of polymorphic, orthologous peptides across species are present in MS/MS data sets (24). We propose a new approach in cross-species proteomics research that aggregates MS/MS of multiple related species followed by spectral networks analysis of the pooled data to capitalize on pairs of spectra from orthologous peptides, as shown in Fig. 1. This approach does not require advance knowledge of the genomes for all species, and enables the identification of novel, polymorphic peptides across species via interspecies propagation. Compared with previous approaches, cross-species spectral network analysis has two major advantages. First, by matching spectra to spectra instead of spectra to database sequences, spectral networks only consider the sequence variability of peptides present in the samples instead of considering all possible variability across the whole database of related species; thus the performance of spectral networks is independent of database size. Second, the analysis of the set of highly related spectra increases the reliability in identifying polymorphic peptides in that multiple different spectra can support the same novel identification. The utility of spectral networks can be also expanded to the proteomic analysis of microbial communities that often contain hundreds of distinct organisms (25, 26). But despite the success of spectral networks in low complexity data sets (22, 23), the analysis of large multi-species proteomics data requires significantly higher reliability in spectral similarity scores because the number of pairwise spectral comparisons grows quadratically with the number of spectra.In this work, we present algorithmic and statistical advances to spectral networks to improve its utility with large and diverse spectral data sets. To statistically assess the significance of spectral alignments in pairing millions of spectra, we propose Align-GF (generating function for spectral alignment) to compute rigorous p values of a spectral pair based on the complete score histogram of all possible alignments between two spectra. We show that Align-GF successfully addressed the reliability challenge in a large data set analysis and demonstrated its utility by leading to a 4-fold increase in the sensitivity of spectral pairs. Even with this dramatically improved accuracy, a very small number of incorrect pairs in a network can still complicate propagation of annotations. To further progress toward the ideal scenario where each subnetwork consists of only spectra from a single peptide family, we introduce new procedures to split mixed networks from different peptide families and show that these effectively eliminate many false spectral pairs. Finally, we propose the first approach to calculation of false discovery rate (FDR) for spectral networks propagation of identifications from unmodified to progressively more modified peptides. The proposed FDR estimation was conservative and was more rigorous for highly modified peptides, and thus now makes propagation results comparable to other peptide identification approaches.The cross-species spectral networks techniques proposed here enabled the proteomic analysis of three different Cyanothece species, including a strain where the genome sequence is not known. Cyanobacteria are one of the most diverse and widely distributed microorganisms and have received significant consideration as satisfying various demands required in bioenergy generation (27). We show that spectral networks can improve peptide identification by up to 38% compared with mainstream approaches, including many polymorphic and modified peptides. Spectral networks could identify peptides with highly divergent sequences (with 7 amino acid mutations) by leveraging networks of variant peptides, and one example subnetwork of species-specific variants of phycobilisome proteins reflects the diversity of photosynthetic light-harvesting strategies (28). Our approach thus demonstrates the potential gains in multi-species proteomics and sets the stage for related developments in higher-complexity metaproteomics samples. Finally, spectral networks revealed many unidentified subnetworks containing only unidentified spectra, thus strongly suggesting the presence of novel peptides that are missing from current protein databases. Although we illustrate the potential of our approach on a specific set of bioenergy-related species, we note that the proposed approach is generic and should be applicable to any other set of related species. The diversity of biologically important protein families could be studied by comparing closely and more remotely related species.  相似文献   

15.
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   

16.
The past 15 years have seen significant progress in LC-MS/MS peptide sequencing, including the advent of successful de novo and database search methods; however, analysis of glycopeptide and, more generally, glycoconjugate spectra remains a much more open problem, and much annotation is still performed manually. This is partly because glycans, unlike peptides, need not be linear chains and are instead described by trees. In this study, we introduce SweetSEQer, an extremely simple open source tool for identifying potential glycopeptide MS/MS spectra. We evaluate SweetSEQer on manually curated glycoconjugate spectra and on negative controls, and we demonstrate high quality filtering that can be easily improved for specific applications. We also demonstrate a high overlap between peaks annotated by experts and peaks annotated by SweetSEQer, as well as demonstrate inferred glycan graphs consistent with canonical glycan tree motifs. This study presents a novel tool for annotating spectra and producing glycan graphs from LC-MS/MS spectra. The tool is evaluated and shown to perform similarly to an expert on manually curated data.Protein glycosylation is a common modification, affecting ∼50% of all expressed proteins (1). Glycosylation affects critical biological functions, including cell-cell recognition, circulating half-life, substrate binding, immunogenicity, and others (2). Regrettably, determining the exact role glycosylation plays in different biological contexts is slowed by a dearth of analytical methods and of appropriate software. Such software is crucial for performing and aiding experts in data analysis complex glycosylation.Glycopeptides are highly heterogeneous in regard to glycan composition, glycan structure, and linkage stereochemistry in addition to the tens of thousands of possible peptides. The analysis of protein glycosylation is often segmented into three distinct types of mass spectrometry experiments, which together help to resolve this complexity. The first analyzes enzymatically or chemically released glycans (which may or may not be chemically modified), and the second determines glycosylation sites after release of glycans from peptides (the resulting mass spectra allow detection of glycosylation sites and the glycans on those sites simultaneously). The third determines the glycosylation sites and the glycans on those sites simultaneously, by MS of intact glycopeptides. Frequently, researchers will perform all three types of analysis, with the first two types providing information about possible combinations of glycan structures and peptides that could be found in the third experiment. Using this MS1 information, the problem is reduced to matching masses observed with a combinatorial pool of all possible glycans and all possible glycosylated peptides within a sample; however, this combinatorial approach alone is insufficient (3), and tandem mass spectrometry can provide copious additional information to help resolve the glycopeptide content from complex samples.The similar problem of inferring peptide sequences from MS/MS spectra has received considerably more attention. Peptide inference is more constrained than glycan inference, because the chain of MS/MS peaks corresponds to a linear peptide sequence; given an MS/MS spectrum, the linear peptide sequence can be inferred through brute force or dynamic programming via de novo methods (46) as described in Ref. 7. Additionally, the possible search space of peptides can be dramatically lowered by using database searching (821) as described in Ref. 7, which compares the MS/MS spectrum to the predicted spectra from only those peptides resulting from a protein database or translated open reading frames (ORFs) of a genomic database.The possible search space of glycans is larger than the search space of peptides because, in contrast to linear peptide chains, glycans may form branching trees. Identifying glycans using database search methodologies is impractical, as it is impractical to define the database when the detailed activities of the set of glycosyltransferases are not defined. Generating an overly large database would artificially inflate the set of incompletely characterized spectra, and too small of a search space would lead to inaccurate results. Furthermore, as glycosylation is not a template-driven process, no clear choice for a database matching approach is available, and de novo sequencing is therefore a more appropriate approach.As a result, few desirable software options are available for the high throughput analysis of tandem mass spectrometry data from intact glycopeptides (as noted in a recent review (22)). In fact, manual annotation of spectra is still commonplace, despite being slow and despite the potential for disagreement between different experts. Some available software requires user-defined lists of glycan and/or peptide masses as input, which is suboptimal from a sample consumption and throughput perspective (23, 24). These lists must typically be generated by parallel experiments or simply hypothesized a priori, meaning omissions in either list may affect the results. Furthermore, some software does not work on batched input files, meaning each spectrum must be analyzed separately (23, 2528). Moreover, there is an even greater lack of open source software for glycoproteomics, so modifying the existing software for the researchers individual applications is not easily achieved. The one open source tool that we know of (GlypID) is applicable only to the analysis of glycopeptide spectra acquired from a very specialized workflow, which requires MS1, CID, and higher-energy C-trap type dissociation (HCD) spectra (29). With that approach, oxonium ions from HCD spectra are necessary to predict the glycan class; potential peptide lists are queried by precursor m/z values (requiring accurate a priori knowledge of all modifications), and possible theoretical “N-linked” precursor m/z values are used to select candidate spectra (using templates, unlike de novo characterization). As a result, the tool is specialized and limited to analysis of “N-linked” glycopeptide spectra from very specific experimental setups.Free, open-source glycoproteomic software capable of batch analysis of general tandem mass spectrometry spectra of glycoconjugates is sorely needed. In this work, we present SweetSEQer, a tool for de novo analysis of tandem mass spectra of glycoconjugates (the most general class of spectra containing fragmentation involving sugars). Furthermore, because SweetSEQer is so general and simple, and because it does not require specific experimental setup, it is widely applicable to the analysis of general glycoconjugate spectra (e.g. it is already applicable to “O-linked” glycopeptide and glycoconjugate spectra). Moreover, because it is an open source and does not use external software, it not only eschews solving problems like MS1 deisotoping, it can also be easily customized and even used to augment and complement existing tools like GlypID (and, because we do not use a “copyleft” software license, our algorithm and code can even be added to non-open source and proprietary variants).SweetSEQer''s performance was tested on a validated, manually annotated set of glycoconjugate identifications from a urinary glycoproteomics study. Specificity was demonstrated by showing a low identification rate on negative control spectra from Escherichia coli. Annotated structures are shown to be consistent by a human expert by demonstrating a high overlap in identified glycan fragment ions, as well as a consistency between SweetSEQer''s predicted glycan graph and glycan chains produced by an expert. Our simple object-oriented python implementation is freely available (Apache 2.0 license) on line.  相似文献   

17.
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   

18.
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   

19.
A crucial component of the analysis of shotgun proteomics datasets is the search engine, an algorithm that attempts to identify the peptide sequence from the parent molecular ion that produced each fragment ion spectrum in the dataset. There are many different search engines, both commercial and open source, each employing a somewhat different technique for spectrum identification. The set of high-scoring peptide-spectrum matches for a defined set of input spectra differs markedly among the various search engine results; individual engines each provide unique correct identifications among a core set of correlative identifications. This has led to the approach of combining the results from multiple search engines to achieve improved analysis of each dataset. Here we review the techniques and available software for combining the results of multiple search engines and briefly compare the relative performance of these techniques.The most commonly used proteomics approach, shotgun proteomics, has become an invaluable tool for the high-throughput characterization of proteins in biological samples (1). This workflow relies on the combination of protein digestion, liquid chromatography (LC)1 separation, tandem mass spectrometry (MS/MS), and sophisticated data analysis in its aim to derive an accurate and complete set of peptides and their inferred proteins that are present in the sample being studied. Although many variations are possible, the typical workflow begins with the digestion of proteins into peptides with a protease, typically trypsin. The resulting peptide mixture is first separated via LC and then subjected to mass spectrometry (MS) analysis. The MS instrument acquires fragment ion spectra on a subset of the peptide precursor ions that it measures. From the MS/MS spectra that measure the abundance and mass of the peptide ion fragments, peptides present in the mixture are identified and proteins are inferred by means of downstream computational analysis.The informatics component of the shotgun proteomics workflow is crucial for proper data analysis (2), and a wide variety of tools have emerged for this purpose (3). The typical informatics workflow can be summarized in a few steps: conversion from vendor proprietary formats to an open format, high-throughput interpretation of the MS/MS spectra with a search engine, and statistical validation of the results with estimation of the false discovery rate at a selected score threshold. Various tools for measuring relative peptide abundances may be applied, dependent on the type of quantitation technique applied in the experiment. Finally, the proteins present, and their abundance in the sample, are inferred based on the peptide identifications.One of the most computationally intensive and diverse steps in the computational analysis workflow is the use of a search engine to interpret the MS/MS spectra in order to determine the best matching peptide ion identifications (4), termed peptide-spectrum matches (PSMs). There are three main types of engines: sequence search engines such as X!Tandem (5), Mascot (6), SEQUEST (7), MyriMatch (8), MS-GFDB (9), and OMSSA (10), which attempt to match acquired spectra with theoretical spectra generated from possible peptide sequences contained in a protein sequence list; spectral library search engines such as SpectraST (11), X!Hunter (12), and Bibliospec (13), which attempt to match spectra with a library of previously observed and identified spectra; and de novo search engines such as PEAKS (14), PepNovo (15), and Lutefisk (16), which attempt to derive peptide identifications based on the MS/MS spectrum peak patterns alone, without reference sequences or previous spectra (17). Additionally, elements of de novo sequencing (short sequence tag extraction) and database searching have been combined to create hybrid search engines such as InSpecT (18) and PEAKS-DB (19).The goal of this review is to evaluate the potential improvement made possible by combining the search results of multiple search engines. On their own, most of the common search engines perform well on typical datasets, with the results having significant overlap between the algorithms (20); and yet, the degree to which there is divergence in the results of different search engines remains quite high. Disagreement between search engines, where multiple different peptide sequences are identified with high confidence, is quite rare. It is much more common to observe different engines being in agreement on the correct identification, yet with neither of the identifications having a probability high enough to allow it to pass the selected error criterion when analyzed independently. When the results are analyzed together, the agreement on the identification might propel the PSM to pass the same error criterion. In cases when only one engine scores the PSM highly enough that it passes an acceptance threshold, these identifications are reported within the acceptable error rate. Also, some engines use unique methods to consider peptides or modifications not considered by other engines. Even if the experimenter is careful to choose similar search parameters when running multiple tools, different search engines will allow one to set non-identical search parameters, which contributes to reduced overlap between the search results. Spectral library search engines tend to be far more sensitive and specific than sequence search engines, but only for peptide ions for which there is a spectrum in the library.Given that different search engines excel at identifying different subsets of PSMs, it seems natural to combine the power of multiple search engines to achieve a single, better result. Many algorithms and software tools have emerged that combine search results (2128), each demonstrating an improved final result over any individual search engine alone. Such improved results come at the cost of the increased complexity of managing multiple searches in an analysis pipeline, as well as a several-fold increase in computational time in what is already the most computationally expensive step. However, with the ever-growing availability of fast computers, computing clusters, and cloud computing resources, researchers now have within reach the ability to quickly search their MS/MS data using several of the still-growing number of search engine algorithms. In some cases the open-source search engines are quite similar to their commercial alternatives; for example, Comet (29) is very similar to SEQUEST. Given the significant amount of time the average researcher takes to design an experiment, process the samples, and acquire the data, it is natural that a researcher would wish to maximize the number and confidence of peptide and protein identifications in each dataset with a rigorous computational analysis. Furthermore, when using label-free spectral counting for abundance analysis, maximizing the number of PSMs increases the dynamic range and accuracy of the quantitative approach (23). Therefore, the demand to use multiple search engines and integrate their results with the goal of maximizing the amount of information gleaned from each dataset is expected to continue growing.Also emerging are software tools that use several iterative database searching passes of the same data, combining multiple database search tools, searches with different post-translational modifications, and searches against different databases in an attempt to use each specific tool under ideal conditions, utilizing each for its specific strengths and integrating the results (30). Some relevant aspects of such strategies are discussed by Tharakan et al. (31).In the following sections, we review the various approaches and software programs available to assist with the merging of results from different search engines. We also provide a performance comparison of the various approaches described here on a test dataset to assess the expected performance gains from the various described methods.  相似文献   

20.
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