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 265 毫秒
1.
LC-MS/MS analysis on a linear ion trap LTQ mass spectrometer, combined with data processing, stringent, and sequence-similarity database searching tools, was employed in a layered manner to identify proteins in organisms with unsequenced genomes. Highly specific stringent searches (MASCOT) were applied as a first layer screen to identify either known (i.e. present in a database) proteins, or unknown proteins sharing identical peptides with related database sequences. Once the confidently matched spectra were removed, the remainder was filtered against a nonannotated library of background spectra that cleaned up the dataset from spectra of common protein and chemical contaminants. The rectified spectral dataset was further subjected to rapid batch de novo interpretation by PepNovo software, followed by the MS BLAST sequence-similarity search that used multiple redundant and partially accurate candidate peptide sequences. Importantly, a single dataset was acquired at the uncompromised sensitivity with no need of manual selection of MS/MS spectra for subsequent de novo interpretation. This approach enabled a completely automated identification of novel proteins that were, otherwise, missed by conventional database searches.  相似文献   

2.
De novo peptide sequencing via tandem mass spectrometry.   总被引:10,自引:0,他引:10  
Peptide sequencing via tandem mass spectrometry (MS/MS) is one of the most powerful tools in proteomics for identifying proteins. Because complete genome sequences are accumulating rapidly, the recent trend in interpretation of MS/MS spectra has been database search. However, de novo MS/MS spectral interpretation remains an open problem typically involving manual interpretation by expert mass spectrometrists. We have developed a new algorithm, SHERENGA, for de novo interpretation that automatically learns fragment ion types and intensity thresholds from a collection of test spectra generated from any type of mass spectrometer. The test data are used to construct optimal path scoring in the graph representations of MS/MS spectra. A ranked list of high scoring paths corresponds to potential peptide sequences. SHERENGA is most useful for interpreting sequences of peptides resulting from unknown proteins and for validating the results of database search algorithms in fully automated, high-throughput peptide sequencing.  相似文献   

3.
High-throughput proteomics is made possible by a combination of modern mass spectrometry instruments capable of generating many millions of tandem mass (MS(2)) spectra on a daily basis and the increasingly sophisticated associated software for their automated identification. Despite the growing accumulation of collections of identified spectra and the regular generation of MS(2) data from related peptides, the mainstream approach for peptide identification is still the nearly two decades old approach of matching one MS(2) spectrum at a time against a database of protein sequences. Moreover, database search tools overwhelmingly continue to require that users guess in advance a small set of 4-6 post-translational modifications that may be present in their data in order to avoid incurring substantial false positive and negative rates. The spectral networks paradigm for analysis of MS(2) spectra differs from the mainstream database search paradigm in three fundamental ways. First, spectral networks are based on matching spectra against other spectra instead of against protein sequences. Second, spectral networks find spectra from related peptides even before considering their possible identifications. Third, spectral networks determine consensus identifications from sets of spectra from related peptides instead of separately attempting to identify one spectrum at a time. Even though spectral networks algorithms are still in their infancy, they have already delivered the longest and most accurate de novo sequences to date, revealed a new route for the discovery of unexpected post-translational modifications and highly-modified peptides, enabled automated sequencing of cyclic non-ribosomal peptides with unknown amino acids and are now defining a novel approach for mapping the entire molecular output of biological systems that is suitable for analysis with tandem mass spectrometry. Here we review the current state of spectral networks algorithms and discuss possible future directions for automated interpretation of spectra from any class of molecules.  相似文献   

4.
LC-MS/MS has demonstrated potential for detecting plant pathogens. Unlike PCR or ELISA, LC-MS/MS does not require pathogen-specific reagents for the detection of pathogen-specific proteins and peptides. However, the MS/MS approach we and others have explored does require a protein sequence reference database and database-search software to interpret tandem mass spectra. To evaluate the limitations of database composition on pathogen identification, we analyzed proteins from cultured Ustilago maydis, Phytophthora sojae, Fusarium graminearum, and Rhizoctonia solani by LC-MS/MS. When the search database did not contain sequences for a target pathogen, or contained sequences to related pathogens, target pathogen spectra were reliably matched to protein sequences from nontarget organisms, giving an illusion that proteins from nontarget organisms were identified. Our analysis demonstrates that when database-search software is used as part of the identification process, a paradox exists whereby additional sequences needed to detect a wide variety of possible organisms may lead to more cross-species protein matches and misidentification of pathogens.  相似文献   

5.
Peptide and protein identification remains challenging in organisms with poorly annotated or rapidly evolving genomes, as are commonly encountered in environmental or biofuels research. Such limitations render tandem mass spectrometry (MS/MS) database search algorithms ineffective as they lack corresponding sequences required for peptide-spectrum matching. We address this challenge with the spectral networks approach to (1) match spectra of orthologous peptides across multiple related species and then (2) propagate peptide annotations from identified to unidentified spectra. We here present algorithms to assess the statistical significance of spectral alignments (Align-GF), reduce the impurity in spectral networks, and accurately estimate the error rate in propagated identifications. Analyzing three related Cyanothece species, a model organism for biohydrogen production, spectral networks identified peptides from highly divergent sequences from networks with dozens of variant peptides, including thousands of peptides in species lacking a sequenced genome. Our analysis further detected the presence of many novel putative peptides even in genomically characterized species, thus suggesting the possibility of gaps in our understanding of their proteomic and genomic expression. A web-based pipeline for spectral networks analysis is available at http://proteomics.ucsd.edu/software.Microorganisms have evolved their cellular metabolism to generate energy for life in unusual environments (1), and their capabilities are of great interest in the production of renewable bioenergy and could contribute toward managing the world''s current energy and climate crisis (2). Genomics studies have increased the number of sequenced bioenergy-related microbial genomes and revealed the possible biological reactions involved in bioenergy production (3). Studies of photosynthetic microorganisms, for example, have yielded insights into how they harvest solar energy and use it to produce bioenergy products (4). Despite this importance of microorganisms, the characterization of diverse microbial phenotypes by proteomics tandem mass spectrometry (MS/MS) has been limited. The dominant approaches for MS/MS analysis heavily rely on the availability of completely annotated genomes (i.e. accurate protein databases) (57), yet most microorganisms populating the planet have unsequenced or poorly annotated genomes. Thus it remains challenging to identify proteins from environmental and unculturable organisms.One solution to protein identification in a species with no sequenced genome is to use the genomes of closely related species (8). This requires matching MS/MS data to slightly different peptides in amino acid sequences (polymorphic, orthologous peptides); but matching shifted masses of peptides and their fragment ions is computationally expensive and challenging. Moreover, different species-specific post-translational modifications (PTMs)1 can make the cross-species identification more complex. The common computational approach is tolerantly matching de novo sequences derived from MS/MS data to the database while allowing for amino acid mutations and modifications (911). However, this approach critically depends on good de novo interpretations, which are nearly always partially incorrect and yield high-quality subsequences only for a small fraction of all spectra. The blind database search approach, developed to identify peptides with unexpected modifications, can also be used to directly match MS/MS data from unknown species to a database of closely related species, but its utilization is limited because of its exceptionally large search space (1218). These spectrum-database matching approaches to cross-species identification pose significant challenges in its speed and sensitivity with a huge database, which leads to a much longer search time and more false positive identifications (19, 20).As a complementary approach to spectrum-database matching, spectral library searching is an emerging and promising approach (21). A spectral library is a large collection of identified MS/MS spectra, and an unknown query spectrum can then be identified by direct spectral matching to the library. The great advantage of this approach is the reduction of search space and the use of fragmentation patterns of peptides. The spectral networks approach expands this concept to the identification of modified peptides in MS/MS data sets (22, 23). Spectral networks do not directly search a database, but groups MS/MS spectra by computing the pairwise similarity between MS/MS spectra of peptide variants and then constructs networks where each spectrum defines a node and each significant spectral pair, highly correlated in the fragmentation pattern, defines an edge (Fig. 1). In spectral networks, identification of spectra belonging to the same subnetwork should be related and thus the peptide sequence for an identified spectrum can be propagated to neighboring unidentified spectra.Open in a separate windowFig. 1.Overview of multi-species spectral networks. Nodes represent individual spectra and edges between nodes represent significant pairwise alignment between spectra; edges are labeled with amino acid mutations (dotted edges) or parent mass differences (solid edges). In spectral networks, a peptide and its related variants are ideally grouped into a single subnetwork. If at least one spectrum in a subnetwork is annotated (filled node), all the neighboring spectra (unfilled nodes) can potentially become identified by propagating the annotation over network edges. For example, all spectra in the subnetwork of “peptide A” (top left, blue network) can be annotated via up to three iterative propagations, first from A to {A1, A2, A3}, second from {A2, A3} to {A4, A5}, and third from {A4, A5} to A6. This paradigm can be equally applied to cross-species data analysis, as “peptide L” identified in species 1 (top middle, olive-colored network) is propagated to a node unidentified in species 2, identifying its orthologous “peptide l”, with a serine to alanine polymorphism. Thus, spectral networks enable the detection of orthologous peptide pairs between different species.We recently reported that a vast number of polymorphic, orthologous peptides across species are present in MS/MS data sets (24). We propose a new approach in cross-species proteomics research that aggregates MS/MS of multiple related species followed by spectral networks analysis of the pooled data to capitalize on pairs of spectra from orthologous peptides, as shown in Fig. 1. This approach does not require advance knowledge of the genomes for all species, and enables the identification of novel, polymorphic peptides across species via interspecies propagation. Compared with previous approaches, cross-species spectral network analysis has two major advantages. First, by matching spectra to spectra instead of spectra to database sequences, spectral networks only consider the sequence variability of peptides present in the samples instead of considering all possible variability across the whole database of related species; thus the performance of spectral networks is independent of database size. Second, the analysis of the set of highly related spectra increases the reliability in identifying polymorphic peptides in that multiple different spectra can support the same novel identification. The utility of spectral networks can be also expanded to the proteomic analysis of microbial communities that often contain hundreds of distinct organisms (25, 26). But despite the success of spectral networks in low complexity data sets (22, 23), the analysis of large multi-species proteomics data requires significantly higher reliability in spectral similarity scores because the number of pairwise spectral comparisons grows quadratically with the number of spectra.In this work, we present algorithmic and statistical advances to spectral networks to improve its utility with large and diverse spectral data sets. To statistically assess the significance of spectral alignments in pairing millions of spectra, we propose Align-GF (generating function for spectral alignment) to compute rigorous p values of a spectral pair based on the complete score histogram of all possible alignments between two spectra. We show that Align-GF successfully addressed the reliability challenge in a large data set analysis and demonstrated its utility by leading to a 4-fold increase in the sensitivity of spectral pairs. Even with this dramatically improved accuracy, a very small number of incorrect pairs in a network can still complicate propagation of annotations. To further progress toward the ideal scenario where each subnetwork consists of only spectra from a single peptide family, we introduce new procedures to split mixed networks from different peptide families and show that these effectively eliminate many false spectral pairs. Finally, we propose the first approach to calculation of false discovery rate (FDR) for spectral networks propagation of identifications from unmodified to progressively more modified peptides. The proposed FDR estimation was conservative and was more rigorous for highly modified peptides, and thus now makes propagation results comparable to other peptide identification approaches.The cross-species spectral networks techniques proposed here enabled the proteomic analysis of three different Cyanothece species, including a strain where the genome sequence is not known. Cyanobacteria are one of the most diverse and widely distributed microorganisms and have received significant consideration as satisfying various demands required in bioenergy generation (27). We show that spectral networks can improve peptide identification by up to 38% compared with mainstream approaches, including many polymorphic and modified peptides. Spectral networks could identify peptides with highly divergent sequences (with 7 amino acid mutations) by leveraging networks of variant peptides, and one example subnetwork of species-specific variants of phycobilisome proteins reflects the diversity of photosynthetic light-harvesting strategies (28). Our approach thus demonstrates the potential gains in multi-species proteomics and sets the stage for related developments in higher-complexity metaproteomics samples. Finally, spectral networks revealed many unidentified subnetworks containing only unidentified spectra, thus strongly suggesting the presence of novel peptides that are missing from current protein databases. Although we illustrate the potential of our approach on a specific set of bioenergy-related species, we note that the proposed approach is generic and should be applicable to any other set of related species. The diversity of biologically important protein families could be studied by comparing closely and more remotely related species.  相似文献   

6.
Identification of fusion proteins has contributed significantly to our understanding of cancer progression, yielding important predictive markers and therapeutic targets. While fusion proteins can be potentially identified by mass spectrometry, all previously found fusion proteins were identified using genomic (rather than mass spectrometry) technologies. This lack of MS/MS applications in studies of fusion proteins is caused by the lack of computational tools that are able to interpret mass spectra from peptides covering unknown fusion breakpoints (fusion peptides). Indeed, the number of potential fusion peptides is so large that the existing MS/MS database search tools become impractical even in the case of small genomes. We explore computational approaches to identifying fusion peptides, propose an algorithm for solving the fusion peptide identification problem, and analyze the performance of this algorithm on simulated data. We further illustrate how this approach can be modified for human exons prediction.  相似文献   

7.
Hu Y  Li Y  Lam H 《Proteomics》2011,11(24):4702-4711
Spectral library searching is a promising alternative to sequence database searching in peptide identification from MS/MS spectra. The key advantage of spectral library searching is the utilization of more spectral features to improve score discrimination between good and bad matches, and hence sensitivity. However, the coverage of reference spectral library is limited by current experimental and computational methods. We developed a computational approach to expand the coverage of spectral libraries with semi-empirical spectra predicted from perturbing known spectra of similar sequences, such as those with single amino acid substitutions. We hypothesized that the peptide of similar sequences should produce similar fragmentation patterns, at least in most cases. Our results confirm our hypothesis and specify when this approach can be applied. In actual spectral searching of real data sets, the sensitivity advantage of spectral library searching over sequence database searching can be mostly retained even when all real spectra are replaced by semi-empirical ones. We demonstrated the applicability of this approach by detecting several known non-synonymous single-nucleotide polymorphisms in three large human data sets by spectral searching.  相似文献   

8.
Neuropeptidomics is used to characterize endogenous peptides in the brain of tree shrews (Tupaia belangeri). Tree shrews are small animals similar to rodents in size but close relatives of primates, and are excellent models for brain research. Currently, tree shrews have no complete proteome information available on which direct database search can be allowed for neuropeptide identification. To increase the capability in the identification of neuropeptides in tree shrews, we developed an integrated mass spectrometry (MS)-based approach that combines methods including data-dependent, directed, and targeted liquid chromatography (LC)-Fourier transform (FT)-tandem MS (MS/MS) analysis, database construction, de novo sequencing, precursor protein search, and homology analysis. Using this integrated approach, we identified 107 endogenous peptides that have sequences identical or similar to those from other mammalian species. High accuracy MS and tandem MS information, with BLAST analysis and chromatographic characteristics were used to confirm the sequences of all the identified peptides. Interestingly, further sequence homology analysis demonstrated that tree shrew peptides have a significantly higher degree of homology to equivalent sequences in humans than those in mice or rats, consistent with the close phylogenetic relationship between tree shrews and primates. Our results provide the first extensive characterization of the peptidome in tree shrews, which now permits characterization of their function in nervous and endocrine system. As the approach developed fully used the conservative properties of neuropeptides in evolution and the advantage of high accuracy MS, it can be portable for identification of neuropeptides in other species for which the fully sequenced genomes or proteomes are not available.  相似文献   

9.
10.
Clustering millions of tandem mass spectra   总被引:1,自引:0,他引:1  
Tandem mass spectrometry (MS/MS) experiments often generate redundant data sets containing multiple spectra of the same peptides. Clustering of MS/MS spectra takes advantage of this redundancy by identifying multiple spectra of the same peptide and replacing them with a single representative spectrum. Analyzing only representative spectra results in significant speed-up of MS/MS database searches. We present an efficient clustering approach for analyzing large MS/MS data sets (over 10 million spectra) with a capability to reduce the number of spectra submitted to further analysis by an order of magnitude. The MS/MS database search of clustered spectra results in fewer spurious hits to the database and increases number of peptide identifications as compared to regular nonclustered searches. Our open source software MS-Clustering is available for download at http://peptide.ucsd.edu or can be run online at http://proteomics.bioprojects.org/MassSpec.  相似文献   

11.
Full-length de novo sequencing from tandem mass (MS/MS) spectra of unknown proteins such as antibodies or proteins from organisms with unsequenced genomes remains a challenging open problem. Conventional algorithms designed to individually sequence each MS/MS spectrum are limited by incomplete peptide fragmentation or low signal to noise ratios and tend to result in short de novo sequences at low sequencing accuracy. Our shotgun protein sequencing (SPS) approach was developed to ameliorate these limitations by first finding groups of unidentified spectra from the same peptides (contigs) and then deriving a consensus de novo sequence for each assembled set of spectra (contig sequences). But whereas SPS enables much more accurate reconstruction of de novo sequences longer than can be recovered from individual MS/MS spectra, it still requires error-tolerant matching to homologous proteins to group smaller contig sequences into full-length protein sequences, thus limiting its effectiveness on sequences from poorly annotated proteins. Using low and high resolution CID and high resolution HCD MS/MS spectra, we address this limitation with a Meta-SPS algorithm designed to overlap and further assemble SPS contigs into Meta-SPS de novo contig sequences extending as long as 100 amino acids at over 97% accuracy without requiring any knowledge of homologous protein sequences. We demonstrate Meta-SPS using distinct MS/MS data sets obtained with separate enzymatic digestions and discuss how the remaining de novo sequencing limitations relate to MS/MS acquisition settings.Database search tools, such as Sequest (3), Mascot (4), and InsPecT (5), are the most frequently used methods for reliable protein identification in tandem mass (MS/MS) spectrometry based proteomics. These operate by separately matching each MS/MS spectrum to peptide sequences from reference protein databases where all proteins of interest are presumably contained. But this assumption often does not hold true as many important proteins, such as monoclonal antibodies, are not contained in any database because mechanisms of antibody variation (including genetic recombination and somatic hyper-mutation (6)) constantly create new proteins with novel unique sequences. These mechanisms of variation are the foundation of adaptive immune systems and have enabled highly successful antibody-based therapeutic strategies (7, 8). Nevertheless, such variation also means that antibody MS/MS spectra are typically impossible to identify via standard database search techniques whenever the corresponding sequences are not known in advance. An inherent drawback of database search strategies is that they are only as good as the database(s) being searched and incomplete databases often result in proteins being misidentified or left unidentified (9).Despite the importance of novel protein identification, few high-throughput methods have been developed for de novo sequencing of unknown proteins. Low-throughput Edman degradation is a well-known de novo sequencing approach that can accurately call amino acid sequences in N/C-terminal regions of unknown proteins but has drawbacks that make it unsuitable for sequencing proteins longer than 50 amino acids or proteins with post-translational modifications (10, 11). Many have recognized the potential of tandem mass spectrometry for protein sequencing. For example, in 1987 Johnson and Biemann (12) manually sequenced a complete protein from rabbit bone marrow. Meanwhile, automated de novo sequencing methods that rely on interpretations of individual MS/MS spectra are limited in that they typically cannot reconstruct long (8+ AA) sequences without mis-predicting 1 in 5 AA on average for low accuracy collision-induced dissociation (CID) spectra (13, 14). Recent advances in de novo peptide sequencing have improved sequencing accuracy to over 95% for high resolution higher energy collisional dissociation (HCD)1 spectra (15), but at limited sequence coverage (Chi H et al. report only 55% sequence coverage of peptides identified by database search). In fact, all current per-spectrum de novo sequencing strategies face a significant tradeoff between sequencing accuracy and coverage as spectra exhibiting complete peptide fragmentation rarely cover entire target proteins, yet are required to accurately reconstruct full-length peptide sequences. An alternative approach to separately sequencing individual spectra is to simultaneously interpret multiple MS/MS spectra from overlapping peptides. This Shotgun Protein Sequencing (SPS) paradigm differs from traditional algorithms by deriving consensus sequences from contigs - sets of multiple MS/MS spectra from distinct peptides with overlapping sequences (1, 16). Because SPS aggregates multiple spectra from overlapping peptides, protein sequences extending beyond the length of enzymatically digested peptides can be extracted from spectra with incomplete peptide fragmentation. Furthermore, SPS has been found to generate sequences that frequently cover 90–95+% of the target protein sequence(s) whereas mis-predicting only 1 out of every 20 amino acids on high resolution MS/MS spectra (2). But a remaining limitation of SPS is that it still generates fragmented sequences that do not singularly cover large regions of the target protein sequences, much less complete proteins: SPS sequences have an average length of 10–15 amino acids (depending on input data) and the longest recovered SPS de novo sequence is less than 45 amino acids long (1).The considerable limitations of de novo sequencing strategies have typically been addressed by attempting to circumvent them using error-tolerant matching to known protein sequences. One such strategy (17) is to generate short de novo sequence tags and then match them exactly to protein databases without requiring matching the N/C-term flanking masses (to allow for unexpected polymorphisms or post-translational modifications). Short sequence tags are usually derived from parts of the spectrum with high signal-to-noise ratios and typically have higher sequencing accuracy than full-length de novo sequences (18). This approach was later extended in MS-Shotgun (19) and continues to be a popular technique for speeding up database search tools (5, 2022). Homology matching of full length de novo sequences was first explored in CIDentify (23) and later in MS-BLAST (24) by searching de novo sequences using FASTA and WU-BLAST2 (respectively) to find homologous matches to sequences of related proteins; FASTS (25) also approached the problem using a modified version of FASTA. However, common de novo sequencing errors tend to produce sequences that are heavily penalized in pure sequence homology searches. For example, missing peaks in MS/MS spectra may easily cause GA subsequences to be reconstructed as Q or AG (same-mass sequences), thus making subsequent BLAST searches unlikely to succeed. This issue was partially considered in CIDentify and more thoroughly addressed in SPIDER (26) by explicitly modeling de novo sequencing errors together with BLOSUM scores in MS/MS-based sequence homology searches. In addition, OpenSea (27) further explored database matching of de novo sequences for analysis of unexpected post-translational modifications (PTMs). Finally, Shen et al. (28) used short unique de novo sequence tags, called UStags, to discover protein-localized PTMs.Recent approaches to homology matching of de novo sequences have built on genome assembly and sequencing techniques to achieve database-assisted full-length sequencing of unknown proteins. Comparative Shotgun Protein Sequencing (cSPS) complemented SPS assembly techniques with usage of error tolerant matching of de novo sequences to find overlapping SPS de novo sequences that are then further assembled into full-length protein sequences (2). cSPS was designed to support the sequencing of highly divergent proteins that have regions close enough in homology to transfer matches from a reference. cSPS was shown to enable de novo sequencing of monoclonal antibodies at 95+% sequencing accuracy, while simultaneously tolerating and identifying unexpected PTMs (29). In difference from cSPS, Champs (30) de novo sequences individual spectra to obtain putative peptide sequences, which are then mapped to homologous proteins to correct sequencing errors and reconstruct protein sequences with 100% accuracy and 99% coverage. However, Champs is designed to only map peptides that differ from the reference sequence by one or two amino acids and does not handle PTMs. As such, its sequencing accuracy is not directly comparable to that of cSPS as Champs was not designed to sequence highly divergent proteins (such as monoclonal antibodies) with multiple PTMs, insertions, deletions, and/or recombinations. GenoMS (31) extended the approaches in cSPS/Champs by explicitly modeling protein splice variants as paths in splice graphs where nodes represent translated exon regions (32). MS/MS spectra are first searched for exact sequence matches against all possible protein isoforms. The remaining unidentified MS/MS spectra are then aligned to the matched peptides and de novo sequenced to extend the matched sequences into novel regions. Reported sequences are 97–99% accurate and cover 96–99% of target proteins depending on sequence similarity between the novel and reference sequences (31). However, GenoMS de novo sequences are usually extended less than 3 amino acids beyond matched peptides because sequencing accuracy degrades as sequences are extended, thus preventing the consistent extension of long (10+ AA) sequences. Altogether, the use of homology matching approaches for full-length de novo protein sequencing continues to be limited by 1) requiring the previous knowledge of closely related protein sequences and 2) the inherent difficulties in statistically significant homology-tolerant matching of error-prone short de novo sequences.The Meta-SPS approach proposed here seeks to de novo sequence complete proteins, or long protein regions, without any use of a database. Meta-SPS builds upon SPS by treating SPS de novo sequences (contig sequences) as input spectra and further assembling them into longer de novo sequences (meta-contig sequences). We show that Meta-SPS extends de novo sequences to lengths over 100 AA while boosting sequencing accuracy to only 1 mistake per 40 amino acid predictions, thus enabling database-free de novo sequencing of completely novel proteins while also allowing error-tolerant matching approaches to support higher-divergence homologies (by searching longer, more accurate de novo sequences). Meta-SPS algorithms are demonstrated on CID and HCD MS/MS spectra and its limitations are discussed in relation to the underlying limitations of bottom-up tandem mass spectrometry.  相似文献   

12.
BACKGROUND: The annotation of genomes from next-generation sequencing platforms needs to be rapid, high-throughput, and fully integrated and automated. Although a few Web-based annotation services have recently become available, they may not be the best solution for researchers that need to annotate a large number of genomes, possibly including proprietary data, and store them locally for further analysis. To address this need, we developed a standalone software application, the Annotation of microbial Genome Sequences (AGeS) system, which incorporates publicly available and in-house-developed bioinformatics tools and databases, many of which are parallelized for high-throughput performance. METHODOLOGY: The AGeS system supports three main capabilities. The first is the storage of input contig sequences and the resulting annotation data in a central, customized database. The second is the annotation of microbial genomes using an integrated software pipeline, which first analyzes contigs from high-throughput sequencing by locating genomic regions that code for proteins, RNA, and other genomic elements through the Do-It-Yourself Annotation (DIYA) framework. The identified protein-coding regions are then functionally annotated using the in-house-developed Pipeline for Protein Annotation (PIPA). The third capability is the visualization of annotated sequences using GBrowse. To date, we have implemented these capabilities for bacterial genomes. AGeS was evaluated by comparing its genome annotations with those provided by three other methods. Our results indicate that the software tools integrated into AGeS provide annotations that are in general agreement with those provided by the compared methods. This is demonstrated by a >94% overlap in the number of identified genes, a significant number of identical annotated features, and a >90% agreement in enzyme function predictions.  相似文献   

13.
A quality control algorithm for DNA sequencing projects.   总被引:2,自引:0,他引:2       下载免费PDF全文
Heterologous DNA sequences from rearrangements with the genomes of host cells, genomic fragments from hybrid cells, or impure tissue sources can threaten the purity of libraries that are derived from RNA or DNA. Hybridization methods can only detect contaminants from known or suspected heterologous sources, and whole library screening is technically very difficult. Detection of contaminating heterologous clones by sequence alignment is only possible when related sequences are present in a known database. We have developed a statistical test to identify heterologous sequences that is based on the differences in hexamer composition of DNA from different organisms. This test does not require that sequences similar to potential heterologous contaminants are present in the database, and can in principle detect contamination by previously unknown organisms. We have applied this test to the major public expressed sequence tag (EST) data sets to evaluate its utility as a quality control measure and a peer evaluation tool. There is detectable heterogeneity in most human and C.elegans EST data sets but it is not apparently associated with cross-species contamination. However, there is direct evidence for both yeast and bacterial sequence contamination in some public database sequences annotated as human. Results obtained with the hexamer test have been confirmed with similarity searches using sequences from the relevant data sets.  相似文献   

14.
We present an in silico approach for the reconstruction of complete mitochondrial genomes of non-model organisms directly from next-generation sequencing (NGS) data—mitochondrial baiting and iterative mapping (MITObim). The method is straightforward even if only (i) distantly related mitochondrial genomes or (ii) mitochondrial barcode sequences are available as starting-reference sequences or seeds, respectively. We demonstrate the efficiency of the approach in case studies using real NGS data sets of the two monogenean ectoparasites species Gyrodactylus thymalli and Gyrodactylus derjavinoides including their respective teleost hosts European grayling (Thymallus thymallus) and Rainbow trout (Oncorhynchus mykiss). MITObim appeared superior to existing tools in terms of accuracy, runtime and memory requirements and fully automatically recovered mitochondrial genomes exceeding 99.5% accuracy from total genomic DNA derived NGS data sets in <24 h using a standard desktop computer. The approach overcomes the limitations of traditional strategies for obtaining mitochondrial genomes for species with little or no mitochondrial sequence information at hand and represents a fast and highly efficient in silico alternative to laborious conventional strategies relying on initial long-range PCR. We furthermore demonstrate the applicability of MITObim for metagenomic/pooled data sets using simulated data. MITObim is an easy to use tool even for biologists with modest bioinformatics experience. The software is made available as open source pipeline under the MIT license at https://github.com/chrishah/MITObim.  相似文献   

15.
Timely classification and identification of bacteria is of vital importance in many areas of public health. We present a mass spectrometry (MS)-based proteomics approach for bacterial classification. In this method, a bacterial proteome database is derived from all potential protein coding open reading frames (ORFs) found in 170 fully sequenced bacterial genomes. Amino acid sequences of tryptic peptides obtained by LC-ESI MS/MS analysis of the digest of bacterial cell extracts are assigned to individual bacterial proteomes in the database. Phylogenetic profiles of these peptides are used to create a matrix of sequence-to-bacterium assignments. These matrixes, viewed as specific assignment bitmaps, are analyzed using statistical tools to reveal the relatedness between a test bacterial sample and the microorganism database. It is shown that, if a sufficient amount of sequence information is obtained from the MS/MS experiments, a bacterial sample can be classified to a strain level by using this proteomics method, leading to its positive identification.  相似文献   

16.
A novel software tool named PTM-Explorer has been applied to LC-MS/MS datasets acquired within the Human Proteome Organisation (HUPO) Brain Proteome Project (BPP). PTM-Explorer enables automatic identification of peptide MS/MS spectra that were not explained in typical sequence database searches. The main focus was detection of PTMs, but PTM-Explorer detects also unspecific peptide cleavage, mass measurement errors, experimental modifications, amino acid substitutions, transpeptidation products and unknown mass shifts. To avoid a combinatorial problem the search is restricted to a set of selected protein sequences, which stem from previous protein identifications using a common sequence database search. Prior to application to the HUPO BPP data, PTM-Explorer was evaluated on excellently manually characterized and evaluated LC-MS/MS data sets from Alpha-A-Crystallin gel spots obtained from mouse eye lens. Besides various PTMs including phosphorylation, a wealth of experimental modifications and unspecific cleavage products were successfully detected, completing the primary structure information of the measured proteins. Our results indicate that a large amount of MS/MS spectra that currently remain unidentified in standard database searches contain valuable information that can only be elucidated using suitable software tools.  相似文献   

17.
Coding information is the main source of heterogeneity (non-randomness) in the sequences of microbial genomes. The heterogeneity corresponds to a cluster structure in triplet distributions of relatively short genomic fragments (200-400 bp). We found a universal 7-cluster structure in microbial genomic sequences and explained its properties. We show that codon usage of bacterial genomes is a multi-linear function of their genomic G+C-content with high accuracy. Based on the analysis of 143 completely sequenced bacterial genomes available in Genbank in August 2004, we show that there are four "pure" types of the 7-cluster structure observed. All 143 cluster animated 3D-scatters are collected in a database which is made available on our web-site (http://www.ihes.fr/~zinovyev/7clusters). The findings can be readily introduced into software for gene prediction, sequence alignment or microbial genomes classification.  相似文献   

18.
Archak S  Nagaraju J 《Fly》2007,1(5):279-281
Microsatellites show tremendous variation between genomes in terms of their occurrence and composition. Availability of whole genome sequences allows us to study microsatellite characteristics of fully sequenced insect genomes to understand the evolution and biological significance of microsatellites. InSatDb is an insect microsatellite database that provides an interactive interface to query information on microsatellites annotated with size (in base pairs and repeat units), genomic location (exon, intron, up-stream or transposon), nature (perfect or imperfect), and sequence composition (repeat motif and GC%). Here we present a snapshot of the distribution and composition of microsatellites in introns and exons of insect genomes. The data present interesting observations regarding the microsatellite life-cycle and genome flux.  相似文献   

19.
《Fly》2013,7(5):279-281
Microsatellites show tremendous variation between genomes in terms of their occurrence and composition. Availability of whole genome sequences allows us to study microsatellite characteristics of fully sequenced insect genomes to understand the evolution and biological significance of microsatellites. InSatDb is an insect microsatellite database that provides an interactive interface to query information on microsatellites annotated with size (in base pairs and repeat units); genomic location (exon, intron, up-stream or transposon); nature (perfect or imperfect); and sequence composition (repeat motif and GC%). Here, we present a snap shot of the distribution and composition of microsatellites in introns and exons of insect genomes. The data present interesting observations regarding the microsatellite life-cycle and genome flux.  相似文献   

20.
We demonstrate an approach for global quantitative analysis of protein mixtures using differential stable isotopic labeling of the enzyme-digested peptides combined with microbore liquid chromatography (LC) matrix-assisted laser desorption ionization (MALDI) mass spectrometry (MS). Microbore LC provides higher sample loading, compared to capillary LC, which facilitates the quantification of low abundance proteins in protein mixtures. In this work, microbore LC is combined with MALDI MS via a heated droplet interface. The compatibilities of two global peptide labeling methods (i.e., esterification to carboxylic groups and dimethylation to amine groups of peptides) with this LC-MALDI technique are evaluated. Using a quadrupole-time-of-flight mass spectrometer, MALDI spectra of the peptides in individual sample spots are obtained to determine the abundance ratio among pairs of differential isotopically labeled peptides. MS/MS spectra are subsequently obtained from the peptide pairs showing significant abundance differences to determine the sequences of selected peptides for protein identification. The peptide sequences determined from MS/MS database search are confirmed by using the overlaid fragment ion spectra generated from a pair of differentially labeled peptides. The effectiveness of this microbore LC-MALDI approach is demonstrated in the quantification and identification of peptides from a mixture of standard proteins as well as E. coli whole cell extract of known relative concentrations. It is shown that this approach provides a facile and economical means of comparing relative protein abundances from two proteome samples.  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