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 234 毫秒
1.
The success of high-throughput proteomics hinges on the ability of computational methods to identify peptides from tandem mass spectra (MS/MS). However, a common limitation of most peptide identification approaches is the nearly ubiquitous assumption that each MS/MS spectrum is generated from a single peptide. We propose a new computational approach for the identification of mixture spectra generated from more than one peptide. Capitalizing on the growing availability of large libraries of single-peptide spectra (spectral libraries), our quantitative approach is able to identify up to 98% of all mixture spectra from equally abundant peptides and automatically adjust to varying abundance ratios of up to 10:1. Furthermore, we show how theoretical bounds on spectral similarity avoid the need to compare each experimental spectrum against all possible combinations of candidate peptides (achieving speedups of over five orders of magnitude) and demonstrate that mixture-spectra can be identified in a matter of seconds against proteome-scale spectral libraries. Although our approach was developed for and is demonstrated on peptide spectra, we argue that the generality of the methods allows for their direct application to other types of spectral libraries and mixture spectra.The success of tandem MS (MS/MS1) approaches to peptide identification is partly due to advances in computational techniques allowing for the reliable interpretation of MS/MS spectra. Mainstream computational techniques mainly fall into two categories: database search approaches that score each spectrum against peptides in a sequence database (14) or de novo techniques that directly reconstruct the peptide sequence from each spectrum (58). The combination of these methods with advances in high-throughput MS/MS have promoted the accelerated growth of spectral libraries, collections of peptide MS/MS spectra the identification of which were validated by accepted statistical methods (9, 10) and often also manually confirmed by mass spectrometry experts. The similar concept of spectral archives was also recently proposed to denote spectral libraries including “interesting” nonidentified spectra (11) (i.e. recurring spectra with good de novo reconstructions but no database match). The growing availability of these large collections of MS/MS spectra has reignited the development of alternative peptide identification approaches based on spectral matching (1214) and alignment (1517) algorithms.However, mainstream approaches were developed under the (often unstated) assumption that each MS/MS spectrum is generated from a single peptide. Although chromatographic procedures greatly contribute to making this a reasonable assumption, there are several situations where it is difficult or even impossible to separate pairs of peptides. Examples include certain permutations of the peptide sequence or post-translational modifications (see (18) for examples of co-eluting histone modification variants). In addition, innovative experimental setups have demonstrated the potential for increased throughput in peptide identification using mixture spectra; examples include data-independent acquisition (19) ion-mobility MS (20), and MSE strategies (21).To alleviate the algorithmic bottleneck in such scenarios, we describe a computational approach, M-SPLIT (mixture-spectrum partitioning using library of identified tandem mass spectra), that is able to reliably and efficiently identify peptides from mixture spectra, which are generated from a pair of peptides. In brief, a mixture spectrum is modeled as linear combination of two single-peptide spectra, and peptide identification is done by searching against a spectral library. We show that efficient filtration and accurate branch-and-bound strategies can be used to avoid the huge computational cost of searching all possible pairs. Thus equipped, our approach is able to identify the correct matches by considering only a minuscule fraction of all possible matches. Beyond potentially enhancing the identification capabilities of current MS/MS acquisition setups, we argue that the availability of methods to reliably identify MS/MS spectra from mixtures of peptides could enable the collection of MS/MS data using accelerated chromatography setups to obtain the same or better peptide identification results in a fraction of the experimental time currently required for exhaustive peptide separation.  相似文献   

2.
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   

3.
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   

4.
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   

5.
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   

6.
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   

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

8.
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   

9.
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   

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

13.
A Boolean network is a model used to study the interactions between different genes in genetic regulatory networks. In this paper, we present several algorithms using gene ordering and feedback vertex sets to identify singleton attractors and small attractors in Boolean networks. We analyze the average case time complexities of some of the proposed algorithms. For instance, it is shown that the outdegree-based ordering algorithm for finding singleton attractors works in time for , which is much faster than the naive time algorithm, where is the number of genes and is the maximum indegree. We performed extensive computational experiments on these algorithms, which resulted in good agreement with theoretical results. In contrast, we give a simple and complete proof for showing that finding an attractor with the shortest period is NP-hard.[1,2,3,4,5,6,7,8,9,10,11,12,13,14,15,16,17,18,19,20,21,22,23,24,25,26,27,28,29,30,31,32]  相似文献   

14.
Protein–protein interactions (PPIs) are fundamental to the structure and function of protein complexes. Resolving the physical contacts between proteins as they occur in cells is critical to uncovering the molecular details underlying various cellular activities. To advance the study of PPIs in living cells, we have developed a new in vivo cross-linking mass spectrometry platform that couples a novel membrane-permeable, enrichable, and MS-cleavable cross-linker with multistage tandem mass spectrometry. This strategy permits the effective capture, enrichment, and identification of in vivo cross-linked products from mammalian cells and thus enables the determination of protein interaction interfaces. The utility of the developed method has been demonstrated by profiling PPIs in mammalian cells at the proteome scale and the targeted protein complex level. Our work represents a general approach for studying in vivo PPIs and provides a solid foundation for future studies toward the complete mapping of PPI networks in living systems.Protein–protein interactions (PPIs)1 play a key role in defining protein functions in biological systems. Aberrant PPIs can have drastic effects on biochemical activities essential to cell homeostasis, growth, and proliferation, and thereby lead to various human diseases (1). Consequently, PPI interfaces have been recognized as a new paradigm for drug development. Therefore, mapping PPIs and their interaction interfaces in living cells is critical not only for a comprehensive understanding of protein function and regulation, but also for describing the molecular mechanisms underlying human pathologies and identifying potential targets for better therapeutics.Several strategies exist for identifying and mapping PPIs, including yeast two-hybrid, protein microarray, and affinity purification mass spectrometry (AP-MS) (25). Thanks to new developments in sample preparation strategies, mass spectrometry technologies, and bioinformatics tools, AP-MS has become a powerful and preferred method for studying PPIs at the systems level (69). Unlike other approaches, AP-MS experiments allow the capture of protein interactions directly from their natural cellular environment, thus better retaining native protein structures and biologically relevant interactions. In addition, a broader scope of PPI networks can be obtained with greater sensitivity, accuracy, versatility, and speed. Despite the success of this very promising technique, AP-MS experiments can lead to the loss of weak/transient interactions and/or the reorganization of protein interactions during biochemical manipulation under native purification conditions. To circumvent these problems, in vivo chemical cross-linking has been successfully employed to stabilize protein interactions in native cells or tissues prior to cell lysis (1016). The resulting covalent bonds formed between interacting partners allow affinity purification under stringent and fully denaturing conditions, consequently reducing nonspecific background while preserving stable and weak/transient interactions (1216). Subsequent mass spectrometric analysis can reveal not only the identities of interacting proteins, but also cross-linked amino acid residues. The latter provides direct molecular evidence describing the physical contacts between and within proteins (17). This information can be used for computational modeling to establish structural topologies of proteins and protein complexes (1722), as well as for generating experimentally derived protein interaction network topology maps (23, 24). Thus, cross-linking mass spectrometry (XL-MS) strategies represent a powerful and emergent technology that possesses unparalleled capabilities for studying PPIs.Despite their great potential, current XL-MS studies that have aimed to identify cross-linked peptides have been mostly limited to in vitro cross-linking experiments, with few successfully identifying protein interaction interfaces in living cells (24, 25). This is largely because XL-MS studies remain challenging due to the inherent difficulty in the effective MS detection and accurate identification of cross-linked peptides, as well as in unambiguous assignment of cross-linked residues. In general, cross-linked products are heterogeneous and low in abundance relative to non-cross-linked products. In addition, their MS fragmentation is too complex to be interpreted using conventional database searching tools (17, 26). It is noted that almost all of the current in vivo PPI studies utilize formaldehyde cross-linking because of its membrane permeability and fast kinetics (1016). However, in comparison to the most commonly used amine reactive NHS ester cross-linkers, identification of formaldehyde cross-linked peptides is even more challenging because of its promiscuous nonspecific reactivity and extremely short spacer length (27). Therefore, further developments in reagents and methods are urgently needed to enable simple MS detection and effective identification of in vivo cross-linked products, and thus allow the mapping of authentic protein contact sites as established in cells, especially for protein complexes.Various efforts have been made to address the limitations of XL-MS studies, resulting in new developments in bioinformatics tools for improved data interpretation (2832) and new designs of cross-linking reagents for enhanced MS analysis of cross-linked peptides (24, 3339). Among these approaches, the development of new cross-linking reagents holds great promise for mapping PPIs on the systems level. One class of cross-linking reagents containing an enrichment handle have been shown to allow selective isolation of cross-linked products from complex mixtures, boosting their detectability by MS (3335, 4042). A second class of cross-linkers containing MS-cleavable bonds have proven to be effective in facilitating the unambiguous identification of cross-linked peptides (3639, 43, 44), as the resulting cross-linked products can be identified based on their characteristic and simplified fragmentation behavior during MS analysis. Therefore, an ideal cross-linking reagent would possess the combined features of both classes of cross-linkers. To advance the study of in vivo PPIs, we have developed a new XL-MS platform based on a novel membrane-permeable, enrichable, and MS-cleavable cross-linker, Azide-A-DSBSO (azide-tagged, acid-cleavable disuccinimidyl bis-sulfoxide), and multistage tandem mass spectrometry (MSn). This new XL-MS strategy has been successfully employed to map in vivo PPIs from mammalian cells at both the proteome scale and the targeted protein complex level.  相似文献   

15.
Top-down mass spectrometry (MS)-based proteomics is arguably a disruptive technology for the comprehensive analysis of all proteoforms arising from genetic variation, alternative splicing, and posttranslational modifications (PTMs). However, the complexity of top-down high-resolution mass spectra presents a significant challenge for data analysis. In contrast to the well-developed software packages available for data analysis in bottom-up proteomics, the data analysis tools in top-down proteomics remain underdeveloped. Moreover, despite recent efforts to develop algorithms and tools for the deconvolution of top-down high-resolution mass spectra and the identification of proteins from complex mixtures, a multifunctional software platform, which allows for the identification, quantitation, and characterization of proteoforms with visual validation, is still lacking. Herein, we have developed MASH Suite Pro, a comprehensive software tool for top-down proteomics with multifaceted functionality. MASH Suite Pro is capable of processing high-resolution MS and tandem MS (MS/MS) data using two deconvolution algorithms to optimize protein identification results. In addition, MASH Suite Pro allows for the characterization of PTMs and sequence variations, as well as the relative quantitation of multiple proteoforms in different experimental conditions. The program also provides visualization components for validation and correction of the computational outputs. Furthermore, MASH Suite Pro facilitates data reporting and presentation via direct output of the graphics. Thus, MASH Suite Pro significantly simplifies and speeds up the interpretation of high-resolution top-down proteomics data by integrating tools for protein identification, quantitation, characterization, and visual validation into a customizable and user-friendly interface. We envision that MASH Suite Pro will play an integral role in advancing the burgeoning field of top-down proteomics.With well-developed algorithms and computational tools for mass spectrometry (MS)1 data analysis, peptide-based bottom-up proteomics has gained considerable popularity in the field of systems biology (19). Nevertheless, the bottom-up approach is suboptimal for the analysis of protein posttranslational modifications (PTMs) and sequence variants as a result of protein digestion (10). Alternatively, the protein-based top-down proteomics approach analyzes intact proteins, which provides a “bird''s eye” view of all proteoforms (11), including those arising from sequence variations, alternative splicing, and diverse PTMs, making it a disruptive technology for the comprehensive analysis of proteoforms (1224). However, the complexity of top-down high-resolution mass spectra presents a significant challenge for data analysis. In contrast to the well-developed software packages available for processing data from bottom-up proteomics experiments, the data analysis tools in top-down proteomics remain underdeveloped.The initial step in the analysis of top-down proteomics data is deconvolution of high-resolution mass and tandem mass spectra. Thorough high-resolution analysis of spectra by horn (THRASH), which was the first algorithm developed for the deconvolution of high-resolution mass spectra (25), is still widely used. THRASH automatically detects and evaluates individual isotopomer envelopes by comparing the experimental isotopomer envelope with a theoretical envelope and reporting those that score higher than a user-defined threshold. Another commonly used algorithm, MS-Deconv, utilizes a combinatorial approach to address the difficulty of grouping MS peaks from overlapping isotopomer envelopes (26). Recently, UniDec, which employs a Bayesian approach to separate mass and charge dimensions (27), can also be applied to the deconvolution of high-resolution spectra. Although these algorithms assist in data processing, unfortunately, the deconvolution results often contain a considerable amount of misassigned peaks as a consequence of the complexity of the high-resolution MS and MS/MS data generated in top-down proteomics experiments. Errors such as these can undermine the accuracy of protein identification and PTM localization and, thus, necessitate the implementation of visual components that allow for the validation and manual correction of the computational outputs.Following spectral deconvolution, a typical top-down proteomics workflow incorporates identification, quantitation, and characterization of proteoforms; however, most of the recently developed data analysis tools for top-down proteomics, including ProSightPC (28, 29), Mascot Top Down (also known as Big-Mascot) (30), MS-TopDown (31), and MS-Align+ (32), focus almost exclusively on protein identification. ProSightPC was the first software tool specifically developed for top-down protein identification. This software utilizes “shotgun annotated” databases (33) that include all possible proteoforms containing user-defined modifications. Consequently, ProSightPC is not optimized for identifying PTMs that are not defined by the user(s). Additionally, the inclusion of all possible modified forms within the database dramatically increases the size of the database and, thus, limits the search speed (32). Mascot Top Down (30) is based on standard Mascot but enables database searching using a higher mass limit for the precursor ions (up to 110 kDa), which allows for the identification of intact proteins. Protein identification using Mascot Top Down is fundamentally similar to that used in bottom-up proteomics (34), and, therefore, it is somewhat limited in terms of identifying unexpected PTMs. MS-TopDown (31) employs the spectral alignment algorithm (35), which matches the top-down tandem mass spectra to proteins in the database without prior knowledge of the PTMs. Nevertheless, MS-TopDown lacks statistical evaluation of the search results and performs slowly when searching against large databases. MS-Align+ also utilizes spectral alignment for top-down protein identification (32). It is capable of identifying unexpected PTMs and allows for efficient filtering of candidate proteins when the top-down spectra are searched against a large protein database. MS-Align+ also provides statistical evaluation for the selection of proteoform spectrum match (PrSM) with high confidence. More recently, Top-Down Mass Spectrometry Based Proteoform Identification and Characterization (TopPIC) was developed (http://proteomics.informatics.iupui.edu/software/toppic/index.html). TopPIC is an updated version of MS-Align+ with increased spectral alignment speed and reduced computing requirements. In addition, MSPathFinder, developed by Kim et al., also allows for the rapid identification of proteins from top-down tandem mass spectra (http://omics.pnl.gov/software/mspathfinder) using spectral alignment. Although software tools employing spectral alignment, such as MS-Align+ and MSPathFinder, are particularly useful for top-down protein identification, these programs operate using command line, making them difficult to use for those with limited knowledge of command syntax.Recently, new software tools have been developed for proteoform characterization (36, 37). Our group previously developed MASH Suite, a user-friendly interface for the processing, visualization, and validation of high-resolution MS and MS/MS data (36). Another software tool, ProSight Lite, developed recently by the Kelleher group (37), also allows characterization of protein PTMs. However, both of these software tools require prior knowledge of the protein sequence for the effective localization of PTMs. In addition, both software tools cannot process data from liquid chromatography (LC)-MS and LC-MS/MS experiments, which limits their usefulness in large-scale top-down proteomics. Thus, despite these recent efforts, a multifunctional software platform enabling identification, quantitation, and characterization of proteins from top-down spectra, as well as visual validation and data correction, is still lacking.Herein, we report the development of MASH Suite Pro, an integrated software platform, designed to incorporate tools for protein identification, quantitation, and characterization into a single comprehensive package for the analysis of top-down proteomics data. This program contains a user-friendly customizable interface similar to the previously developed MASH Suite (36) but also has a number of new capabilities, including the ability to handle complex proteomics datasets from LC-MS and LC-MS/MS experiments, as well as the ability to identify unknown proteins and PTMs using MS-Align+ (32). Importantly, MASH Suite Pro also provides visualization components for the validation and correction of the computational outputs, which ensures accurate and reliable deconvolution of the spectra and localization of PTMs and sequence variations.  相似文献   

16.
17.
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   

18.
19.
20.
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   

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