3,322 research outputs found

    Identification of exonic regions in DNA sequences using cross-correlation and noise suppression by discrete wavelet transform

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>The identification of protein coding regions (exons) in DNA sequences using signal processing techniques is an important component of bioinformatics and biological signal processing. In this paper, a new method is presented for the identification of exonic regions in DNA sequences. This method is based on the cross-correlation technique that can identify periodic regions in DNA sequences.</p> <p>Results</p> <p>The method reduces the dependency of window length on identification accuracy. The proposed algorithm is applied to different eukaryotic datasets and the output results are compared with those of other established methods. The proposed method increased the accuracy of exon detection by 4% to 41% relative to the most common digital signal processing methods for exon prediction.</p> <p>Conclusions</p> <p>We demonstrated that periodic signals can be estimated using cross-correlation. In addition, discrete wavelet transform (DWT) can minimise noise while maintaining the signal. The proposed algorithm, which combines cross-correlation and DWT, significantly increases the accuracy of exonic region identification.</p

    Mutation of Arabidopsis SPLICEOSOMAL TIMEKEEPER LOCUS1 Causes Circadian Clock Defects

    Get PDF
    The circadian clock plays a crucial role in coordinating plant metabolic and physiological functions with predictable environmental variables, such as dusk and dawn, while also modulating responses to biotic and abiotic challenges. Much of the initial characterization of the circadian system has focused on transcriptional initiation, but it is now apparent that considerable regulation is exerted after this key regulatory step. Transcript processing, protein stability, and cofactor availability have all been reported to influence circadian rhythms in a variety of species. We used a genetic screen to identify a mutation within a putative RNA binding protein (SPLICEOSOMAL TIMEKEEPER LOCUS1 [STIPL1]) that induces a long circadian period phenotype under constant conditions. STIPL1 is a homolog of the spliceosomal proteins TFP11 (Homo sapiens) and Ntr1p (Saccharomyces cerevisiae) involved in spliceosome disassembly. Analysis of general and alternative splicing using a high-resolution RT-PCR system revealed that mutation of this protein causes less efficient splicing of most but not all of the introns analyzed. In particular, the altered accumulation of circadian-associated transcripts may contribute to the observed mutant phenotype. Interestingly, mutation of a close homolog of STIPL1, STIP-LIKE2, does not cause a circadian phenotype, which suggests divergence in function between these family members. Our work highlights the importance of posttranscriptional control within the clock mechanism. © 2012 American Society of Plant Biologists. All rights reserved

    Circadian rhythms and post-transcriptional regulation in higher plants

    Get PDF
    The circadian clock of plants allows them to cope with daily changes in their environment. This is accomplished by the rhythmic regulation of gene expression, in a process that involves many regulatory steps. One of the key steps involved at the RNA level is post-transcriptional regulation, which ensures a correct control on the different amounts and types of mRNA that will ultimately define the current physiological state of the plant cell. Recent advances in the study of the processes of regulation of pre-mRNA processing, RNA turn-over and surveillance, regulation of translation, function of lncRNAs, biogenesis and function of small RNAs, and the development of bioinformatics tools have helped to vastly expand our understanding of how this regulatory step performs its role. In this work we review the current progress in circadian regulation at the post-transcriptional level research in plants. It is the continuous interaction of all the information flow control post-transcriptional processes that allow a plant to precisely time and predict daily environmental changes.Fil: Romanowski, Andrés. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Investigaciones Bioquímicas de Buenos Aires. Fundación Instituto Leloir. Instituto de Investigaciones Bioquímicas de Buenos Aires; ArgentinaFil: Yanovsky, Marcelo Javier. Consejo Nacional de Investigaciones Científicas y Técnicas. Oficina de Coordinación Administrativa Parque Centenario. Instituto de Investigaciones Bioquímicas de Buenos Aires. Fundación Instituto Leloir. Instituto de Investigaciones Bioquímicas de Buenos Aires; Argentin

    Optimization of High Field Asymmetric Waveform Ion Mobility Spectrometry to enhance the comprehensiveness of mass spectrometry-based proteomic analyses

    Full text link
    La grande complexité des échantillons biologiques peut compliquer l'identification des protéines et compromettre la profondeur et la couverture des analyses protéomiques utilisant la spectrométrie de masse. Des techniques de séparation permettant d’améliorer l’efficacité et la sélectivité des analyses LC-MS/MS peuvent être employées pour surmonter ces limitations. La spectrométrie de mobilité ionique différentielle, utilisant un champ électrique élevé en forme d'onde asymétrique (FAIMS), a montré des avantages significatifs dans l’amélioration de la transmission d'ions peptidiques à charges multiples, et ce, en réduisant les ions interférents. Dans ce contexte, l'objectif de cette thèse était d'explorer les capacités analytiques de FAIMS afin d'élargir à la fois la gamme dynamique de détection des protéines/peptides et la précision des mesures en protéomique quantitative par spectrométrie de masse. Pour cela, nous avons systématiquement intégré FAIMS dans des approches classiques en protéomique afin de déterminer les changements dynamiques du protéome humain en réponse à l’hyperthermie. Nous avons d’abord étudié les avantages de FAIMS par rapport à la quantification par marquage isobare (tandem mass tag, TMT). Cette approche permet le marquage d'ions peptidiques avec différents groupements chimiques dont les masses nominales sont identiques mais différant par leur distribution respective d'isotopes stables. Les ions peptidiques marqués par TMT produisent des ions rapporteurs de masses distinctes une fois fragmentés en MS/MS. Malheureusement, la co-sélection d'ions précurseurs conduit souvent à des spectres MS/MS chimériques et une approche plus lente basée sur le MS3 est nécessaire pour une quantification précise. Comme FAIMS améliore l’efficacité de séparation en transmettant sélectivement des ions en fonction de leur voltage de compensation (CV), nous avons obtenu moins de co-sélection de peptides. FAIMS a amélioré la quantification des peptides TMT au niveau MS2 et a permis d’obtenir 68% plus de peptides quantifiés par rapport aux analyses LC-MS/MS classiques, fournissant ainsi un aperçu plus vaste des changements dynamiques du protéome humain en réponse au stress thermique. De plus, nous avons étudié le marquage métabolique par incorporation d’acides aminés marqués par des isotopes stables en culture cellulaire (SILAC). Si des interférences co-éluent avec les isotopes SILAC, la quantification devient imprécise et les contreparties de SILAC peuvent être assignées de manière erronée aux ions interférants du chromatogramme, faussant ainsi le rapport SILAC. Le fractionnement post-ionisation FAIMS pourrait filtrer les ions appartenant au bruit de fond qui pourraient autrement être attribués à une paire ou à un triplet SILAC pour la quantification. Dans ce projet, FAIMS a été particulièrement bénéfique pour les espèces peu abondantes et s’est montré plus performant que le fractionnement par échange de cations (SCX). En outre, FAIMS a permis la séparation des phosphoisomères fréquemment observés dans les extraits complexes de phosphoprotéomes. Le troisième objectif de ce travail de recherche était d'explorer la séparation de l'état de charge et la transmission améliorée de peptides fortement chargés avec FAIMS et son application à l'analyse de peptides SUMOylés. FAIMS pourrait ainsi améliorer la transmission des peptides SUMOylés triplement chargés par rapport aux peptides tryptiques usuels, lesquels sont principalement doublement chargés. Ceci permettait l'enrichissement en phase gazeuse des ions peptides SUMOylés. FAIMS est une approche alternative plus simple pour fractionner les peptides SUMOylés, ce qui réduit les pertes d’échantillon et permet de simplifier le traitement des échantillons, tout en augmentant l’efficacité de séparation de manière plus automatisée et en ajoutant un ordre de grandeur de sensibilité. Le dernier objectif de cette thèse était d’améliorer l’instrumentation de FAIMS en le jumelant aux instruments à la fine pointe de la technologie. Avec un nouveau dispositif FAIMS, développé par nos collaborateurs chez Thermo Fisher Scientific, nous avons montré une amélioration dans la robustesse et la transmission des ions pour la nouvelle interface. Dans des expériences simples en protéomique shotgun, FAIMS a étendu la gamme dynamique d'un ordre de grandeur pour une couverture protéomique plus profonde par rapport aux analyses LC-MS/MS classiques. En outre, le fractionnement en phase gazeuse de FAIMS a généré moins d’analyses chimériques en MS2, ce qui a permis d’obtenir plus d’identifications et une meilleure quantification. Pour ce faire, nous avons directement comparé le LC-FAIMS-MS/MS au LC-MS/MS/MS en utilisant la sélection de précurseur synchrone (SPS) avec et sans fractionnement en phase inverse basique. Des mesures quantitatives comparables ont été obtenues pour toutes les méthodes, à l'exception du fait que FAIMS a parmi d’obtenir un nombre 2,5 fois plus grand de peptides quantifiables par rapport aux expériences sans FAIMS. Globalement, cette thèse met en évidence certains des avantages que FAIMS peut offrir aux expériences en protéomique en améliorant à la fois l'identification et la quantification des peptides.The high complexity of biological samples can confound protein identification and compromise the depth and coverage of mass spectrometry-based proteomic analyses. Separation techniques that provide improved peak capacity and selectivity of LC-MS/MS analyses are often sought to overcome these limitations. High-field asymmetric waveform ion mobility spectrometry (FAIMS), a differential ion mobility device, has shown significant advantages by enhancing the transmission of multiple-charged peptide ions by reducing singly-charged interferences. In this context, the goal of this thesis was to explore the analytical capabilities of FAIMS to extend both the dynamic range of proteins/peptides detection and the precision of quantitative proteomic measurements by mass spectrometry. For this, we systematically integrated FAIMS in standard workflows to monitor the dynamic changes of the human proteome in response to hyperthermia. We first studied the merits of FAIMS to aid isobaric labeling quantification with tandem mass tags (TMT). This approach allows the labeling of peptide ions with different chemical groups of identical nominal masses but differing in their respective distribution of stable isotopes. TMT-labeled peptide ions produce reporter ions of distinct masses once fragmented by MS/MS. Unfortunately, the co-selection of precursor ions often leads to chimeric MS/MS spectra, and a slower MS3 centric approach is needed for precise quantification. Since FAIMS improves peak capacity by selectively transmitting ions based on their compensation voltage (CV), we obtained less peptide co-selection. FAIMS improved TMT quantification at the MS2 level and achieved 68 % more quantified peptides compared to regular LC-MS/MS, providing a deeper insight into the dynamic changes of the human proteome in response to heat stress. Further, we investigated stable isotope labeling by amino acids in cell culture (SILAC) quantification. If interferences co-elute simultaneously with SILAC isotopomers, quantification becomes inaccurate and SILAC counterparts can be missassigned to interfering ions in the highly populated chromatogram, thus skewing the SILAC ratio. FAIMS post-ionization fractionation could filter out background ions that can otherwise be attributed to a SILAC pair/triplet for quantification. In this work, FAIMS was especially beneficial for low abundant species and outperformed the standard strong cation exchange (SCX) fractionation workflow. In addition, FAIMS allowed the separation of phosphoisomers that are frequently observed in complex phosphoproteome extracts. The third aim of this work explored the charge state separation and enhanced transmission of highly charged peptides with FAIMS and its application for SUMOylated peptide analysis. FAIMS could enhance the transmission of triply charged SUMOylated peptides over typical tryptic peptide that are predominantly doubly charged, by applying more negative CVs with FAIMS. This allowed for gas-phase enrichment of SUMOylated peptide ions. FAIMS is an alternate and more straightforward approach to fractionate SUMOylated peptides that reduced sample loss, avoided sample processing, while increasing peak capacity in a more automated manner and added one order of magnitude in sensitivity. The last aim of this thesis was to improve the FAIMS instrumentation by interfacing it to the latest state-of-the-art instruments. With a new FAIMS device developed by our collaborators at Thermo Fisher Scientific, we demonstrate the robustness and the improved ion transmission for the new interface. In simple shotgun proteomics, FAIMS extended the dynamic range by one order of magnitude for deeper proteome coverage compared to regular LC-MS/MS. Moreover, fewer MS2 chimeric scans were generated with FAIMS gas-phase fractionation, which garnered more identifications and better quantification. For this, we directly compared LC-FAIMS-MS/MS to LC-MS/MS/MS using synchronous precursor selection (SPS) with and without basic reverse phase fractionation. Comparable quantitative measurements were obtained for all methods, except that FAIMS provided a 2.5-fold increase in the number of quantifiable peptides compared with non-FAIMS experiments. Overall, this thesis highlights some of the advantages that FAIMS can provide for proteomics experiments by improving both peptide identification and quantification

    Gene discovery in developmental neuropsychiatric disorders : clues from chromosomal rearrangements

    Get PDF

    Investigation of the structural and functional relationships of oneogene proteins

    Get PDF
    Proteins are the biomolecular workhorses driving the most biological processes in any living organism. These processes are based on selective interactions between particular proteins. So far, the rules governing the coding of the protein&#039;s biological function, i.e. its ability to selectively interact with other biomolecules, have not been elucidated. The resonant recognition model (RRM) is a novel physicomathematical approach established to analyze the interaction between a protein and its target. The RRM assumes that the specificities of protein interactions are based on the resonant electromagnetic energy transfer at the specific frequency for each interaction. One of the main applications of this model is to predict the location of a protein&#039;s biological active site(s) using digital signal processing. This paper incorporates the continuous wavelet transform (CWT) into the RRM to predict the active sites, for a chosen protein example. We have investigated the oncogene functional group using digital signal analysis methods, in particular Fourier transform and CWT; determined oncogenes&#039; characteristic frequency and functional active sites; and performed the design of the peptide analogous. The results obtained provide new insights into the structure-function relationships of the analyzed oncogene protein family

    12-h clock regulation of genetic information flow by XBP1s

    Get PDF
    © The Author(s), 2020. This article is distributed under the terms of the Creative Commons Attribution License. The definitive version was published in Pan, Y., Ballance, H., Meng, H., Gonzalez, N., Kim, S., Abdurehman, L., York, B., Chen, X., Schnytzer, Y., Levy, O., Dacso, C. C., McClung, C. A., O'Malley, B. W., Liu, S., & Zhu, B. 12-h clock regulation of genetic information flow by XBP1s. Plos Biology, 18(1), (2020): e3000580, doi:10.1371/journal.pbio.3000580.Our group recently characterized a cell-autonomous mammalian 12-h clock independent from the circadian clock, but its function and mechanism of regulation remain poorly understood. Here, we show that in mouse liver, transcriptional regulation significantly contributes to the establishment of 12-h rhythms of mRNA expression in a manner dependent on Spliced Form of X-box Binding Protein 1 (XBP1s). Mechanistically, the motif stringency of XBP1s promoter binding sites dictates XBP1s’s ability to drive 12-h rhythms of nascent mRNA transcription at dawn and dusk, which are enriched for basal transcription regulation, mRNA processing and export, ribosome biogenesis, translation initiation, and protein processing/sorting in the Endoplasmic Reticulum (ER)-Golgi in a temporal order consistent with the progressive molecular processing sequence described by the central dogma information flow (CEDIF). We further identified GA-binding proteins (GABPs) as putative novel transcriptional regulators driving 12-h rhythms of gene expression with more diverse phases. These 12-h rhythms of gene expression are cell autonomous and evolutionarily conserved in marine animals possessing a circatidal clock. Our results demonstrate an evolutionarily conserved, intricate network of transcriptional control of the mammalian 12-h clock that mediates diverse biological pathways. We speculate that the 12-h clock is coopted to accommodate elevated gene expression and processing in mammals at the two rush hours, with the particular genes processed at each rush hour regulated by the circadian and/or tissue-specific pathways.This study was supported by the American Diabetes Association junior faculty development award 1-18-JDF-025 to B.Z., by funding from National Institute of Health HD07879 and 1P01DK113954 to B.W.O, by funding from National Science Foundation award 1703170 to C.C.D. and B.Z., and by funding from Brockman Foundation to C.C.D and B.W.O. This work was further supported by the UPMC Genome Center with funding from UPMC’s Immunotherapy and Transplant Center. This research was supported in part by the University of Pittsburgh Center for Research Computing through the resources provided. Research reported in this publication was further supported by the National Institute of Diabetes And Digestive And Kidney Diseases of the National Institutes of Health under award number P30DK120531 to Pittsburgh Liver Research Center, in which both S.L. and B.Z. are members. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript
    • …
    corecore