SwePub
Tyck till om SwePub Sök här!
Sök i SwePub databas

  Utökad sökning

Träfflista för sökning "L773:1467 5463 OR L773:1477 4054 "

Sökning: L773:1467 5463 OR L773:1477 4054

  • Resultat 1-10 av 41
Sortera/gruppera träfflistan
   
NumreringReferensOmslagsbildHitta
1.
  • Bonner, Stephen, et al. (författare)
  • A review of biomedical datasets relating to drug discovery: a knowledge graph perspective
  • 2022
  • Ingår i: Briefings in Bioinformatics. - : Oxford University Press (OUP). - 1467-5463 .- 1477-4054. ; In Press
  • Forskningsöversikt (refereegranskat)abstract
    • Drug discovery and development is a complex and costly process. Machine learning approaches are being investigated to help improve the effectiveness and speed of multiple stages of the drug discovery pipeline. Of these, those that use Knowledge Graphs (KG) have promise in many tasks, including drug repurposing, drug toxicity prediction and target gene-disease prioritization. In a drug discovery KG, crucial elements including genes, diseases and drugs are represented as entities, while relationships between them indicate an interaction. However, to construct high-quality KGs, suitable data are required. In this review, we detail publicly available sources suitable for use in constructing drug discovery focused KGs. We aim to help guide machine learning and KG practitioners who are interested in applying new techniques to the drug discovery field, but who may be unfamiliar with the relevant data sources. The datasets are selected via strict criteria, categorized according to the primary type of information contained within and are considered based upon what information could be extracted to build a KG. We then present a comparative analysis of existing public drug discovery KGs and an evaluation of selected motivating case studies from the literature. Additionally, we raise numerous and unique challenges and issues associated with the domain and its datasets, while also highlighting key future research directions. We hope this review will motivate KGs use in solving key and emerging questions in the drug discovery domain.
  •  
2.
  • Bonner, Stephen, et al. (författare)
  • Implications of topological imbalance for representation learning on biomedical knowledge graphs
  • 2022
  • Ingår i: Briefings in Bioinformatics. - : Oxford University Press (OUP). - 1467-5463 .- 1477-4054. ; In Press
  • Tidskriftsartikel (refereegranskat)abstract
    • Adoption of recently developed methods from machine learning has given rise to creation of drug-discovery knowledge graphs (KGs) that utilize the interconnected nature of the domain. Graph-based modelling of the data, combined with KG embedding (KGE) methods, are promising as they provide a more intuitive representation and are suitable for inference tasks such as predicting missing links. One common application is to produce ranked lists of genes for a given disease, where the rank is based on the perceived likelihood of association between the gene and the disease. It is thus critical that these predictions are not only pertinent but also biologically meaningful. However, KGs can be biased either directly due to the underlying data sources that are integrated or due to modelling choices in the construction of the graph, one consequence of which is that certain entities can get topologically overrepresented. We demonstrate the effect of these inherent structural imbalances, resulting in densely connected entities being highly ranked no matter the context. We provide support for this observation across different datasets, models as well as predictive tasks. Further, we present various graph perturbation experiments which yield more support to the observation that KGE models can be more influenced by the frequency of entities rather than any biological information encoded within the relations. Our results highlight the importance of data modelling choices, and emphasizes the need for practitioners to be mindful of these issues when interpreting model outputs and during KG composition.
  •  
3.
  • Chattopadhyay, Subhayan, et al. (författare)
  • Tracing the evolution of aneuploid cancers by multiregional sequencing with CRUST
  • 2021
  • Ingår i: Briefings in Bioinformatics. - : Oxford University Press (OUP). - 1477-4054 .- 1467-5463. ; 22:6
  • Tidskriftsartikel (refereegranskat)abstract
    • Clonal deconvolution of mutational landscapes is crucial to understand the evolutionary dynamics of cancer. Two limiting factors for clonal deconvolution that have remained unresolved are variation in purity and chromosomal copy number across different samples of the same tumor. We developed a semi-supervised algorithm that tracks variant calls through multi-sample spatiotemporal tumor data. While normalizing allele frequencies based on purity, it also adjusts for copy number changes at clonal deconvolution. Absent à priori copy number data, it renders in silico copy number estimations from bulk sequences. Using published and simulated tumor sequences, we reliably segregated clonal/subclonal variants even at a low sequencing depth (~50×). Given at least one pure tumor sample (>70% purity), we could normalize and deconvolve paired samples down to a purity of 40%. This renders a reliable clonal reconstruction well adapted to multi-regionally sampled solid tumors, which are often aneuploid and contaminated by non-cancer cells.
  •  
4.
  • Chen, Mingshuai, et al. (författare)
  • Fuzzy kernel evidence Random Forest for identifying pseudouridine sites
  • 2024
  • Ingår i: Briefings in Bioinformatics. - Oxford : Oxford University Press. - 1467-5463 .- 1477-4054. ; 25:3, s. 1-14
  • Tidskriftsartikel (refereegranskat)abstract
    • Pseudouridine is an RNA modification that is widely distributed in both prokaryotes and eukaryotes, and plays a critical role in numerous biological activities. Despite its importance, the precise identification of pseudouridine sites through experimental approaches poses significant challenges, requiring substantial time and resources.Therefore, there is a growing need for computational techniques that can reliably and quickly identify pseudouridine sites from vast amounts of RNA sequencing data. In this study, we propose fuzzy kernel evidence Random Forest (FKeERF) to identify pseudouridine sites. This method is called PseU-FKeERF, which demonstrates high accuracy in identifying pseudouridine sites from RNA sequencing data. The PseU-FKeERF model selected four RNA feature coding schemes with relatively good performance for feature combination, and then input them into the newly proposed FKeERF method for category prediction. FKeERF not only uses fuzzy logic to expand the original feature space, but also combines kernel methods that are easy to interpret in general for category prediction. Both cross-validation tests and independent tests on benchmark datasets have shown that PseU-FKeERF has better predictive performance than several state-of-the-art methods. This new method not only improves the accuracy of pseudouridine site identification, but also provides a certain reference for disease control and related drug development in the future. © The Author(s) 2024. Published by Oxford University Press.
  •  
5.
  • Cirenajwis, Helena, et al. (författare)
  • Performance of gene expression-based single sample predictors for assessment of clinicopathological subgroups and molecular subtypes in cancers : a case comparison study in non-small cell lung cancer
  • 2019
  • Ingår i: Briefings in Bioinformatics. - : Oxford University Press (OUP). - 1477-4054 .- 1467-5463. ; 21:2, s. 729-740
  • Tidskriftsartikel (refereegranskat)abstract
    • The development of multigene classifiers for cancer prognosis, treatment prediction, molecular subtypes or clinicopathological groups has been a cornerstone in transcriptomic analyses of human malignancies for nearly two decades. However, many reported classifiers are critically limited by different preprocessing needs like normalization and data centering. In response, a new breed of classifiers, single sample predictors (SSPs), has emerged. SSPs classify samples in an N-of-1 fashion, relying on, e.g. gene rules comparing expression values within a sample. To date, several methods have been reported, but there is a lack of head-to-head performance comparison for typical cancer classification problems, representing an unmet methodological need in cancer bioinformatics. To resolve this need, we performed an evaluation of two SSPs [k-top-scoring pair classifier (kTSP) and absolute intrinsic molecular subtyping (AIMS)] for two case examples of different magnitude of difficulty in non-small cell lung cancer: gene expression–based classification of (i) tumor histology and (ii) molecular subtype. Through the analysis of ~2000 lung cancer samples for each case example (n = 1918 and n = 2106, respectively), we compared the performance of the methods for different sample compositions, training data set sizes, gene expression platforms and gene rule selections. Three main conclusions are drawn from the comparisons: both methods are platform independent, they select largely overlapping gene rules associated with actual underlying tumor biology and, for large training data sets, they behave interchangeably performance-wise. While SSPs like AIMS and kTSP offer new possibilities to move gene expression signatures/predictors closer to a clinical context, they are still importantly limited by the difficultness of the classification problem at hand.
  •  
6.
  • Danielsson, Frida, et al. (författare)
  • Assessing the consistency of public human tissue RNA-seq data sets
  • 2015
  • Ingår i: Briefings in Bioinformatics. - : Oxford University Press. - 1467-5463 .- 1477-4054. ; 16:6, s. 941-949
  • Tidskriftsartikel (refereegranskat)abstract
    • Sequencing-based gene expression methods like RNA-sequencing (RNA-seq) have become increasingly common, but it is often claimed that results obtained in different studies are not comparable owing to the influence of laboratory batch effects, differences in RNA extraction and sequencing library preparation methods and bioinformatics processing pipelines. It would be unfortunate if different experiments were in fact incomparable, as there is great promise in data fusion and meta-analysis applied to sequencing data sets. We therefore compared reported gene expression measurements for ostensibly similar samples (specifically, human brain, heart and kidney samples) in several different RNA-seq studies to assess their overall consistency and to examine the factors contributing most to systematic differences. The same comparisons were also performed after preprocessing all data in a consistent way, eliminating potential bias from bioinformatics pipelines. We conclude that published human tissue RNA-seq expression measurements appear relatively consistent in the sense that samples cluster by tissue rather than laboratory of origin given simple preprocessing transformations. The article is supplemented by a detailed walkthrough with embedded R code and figures.
  •  
7.
  • Doran, S., et al. (författare)
  • Multi-omics approaches for revealing the complexity of cardiovascular disease
  • 2021
  • Ingår i: Briefings in bioinformatics. - : Oxford University Press (OUP). - 1467-5463 .- 1477-4054. ; 22:5
  • Tidskriftsartikel (refereegranskat)abstract
    • The development and progression of cardiovascular disease (CVD) can mainly be attributed to the narrowing of blood vessels caused by atherosclerosis and thrombosis, which induces organ damage that will result in end-organ dysfunction characterized by events such as myocardial infarction or stroke. It is also essential to consider other contributory factors to CVD, including cardiac remodelling caused by cardiomyopathies and co-morbidities with other diseases such as chronic kidney disease. Besides, there is a growing amount of evidence linking the gut microbiota to CVD through several metabolic pathways. Hence, it is of utmost importance to decipher the underlying molecular mechanisms associated with these disease states to elucidate the development and progression of CVD. A wide array of systems biology approaches incorporating multi-omics data have emerged as an invaluable tool in establishing alterations in specific cell types and identifying modifications in signalling events that promote disease development. Here, we review recent studies that apply multi-omics approaches to further understand the underlying causes of CVD and provide possible treatment strategies by identifying novel drug targets and biomarkers. We also discuss very recent advances in gut microbiota research with an emphasis on how diet and microbial composition can impact the development of CVD. Finally, we present various biological network analyses and other independent studies that have been employed for providing mechanistic explanation and developing treatment strategies for end-stage CVD, namely myocardial infarction and stroke.
  •  
8.
  • Emanuelsson, Olof (författare)
  • Predicting protein subcellular localisation from amino acid sequence information.
  • 2002
  • Ingår i: Briefings in Bioinformatics. - 1467-5463 .- 1477-4054. ; 3:4, s. 361-76
  • Tidskriftsartikel (refereegranskat)abstract
    • Predicting the subcellular localisation of proteins is an important part of the elucidation of their functions and interactions. Here, the amino acid sequence motifs that direct proteins to their proper subcellular compartment are surveyed, different methods for localisation prediction are discussed, and some benchmarks for the more commonly used predictors are presented.
  •  
9.
  • Fang, MY, et al. (författare)
  • VIPPID: a gene-specific single nucleotide variant pathogenicity prediction tool for primary immunodeficiency diseases
  • 2022
  • Ingår i: Briefings in bioinformatics. - : Oxford University Press (OUP). - 1477-4054 .- 1467-5463. ; 23:5
  • Tidskriftsartikel (refereegranskat)abstract
    • Distinguishing pathogenic variants from non-pathogenic ones remains a major challenge in clinical genetic testing of primary immunodeficiency (PID) patients. Most of the existing mutation pathogenicity prediction tools treat all mutations as homogeneous entities, ignoring the differences in characteristics of different genes, and use the same model for genes in different diseases. In this study, we developed a single nucleotide variant (SNV) pathogenicity prediction tool, Variant Impact Predictor for PIDs (VIPPID; https://mylab.shinyapps.io/VIPPID/), which was tailored for PIDs genes and used a specific model for each of the most prevalent PID known genes. It employed a Conditional Inference Forest model and utilized information of 85 features of SNVs and scores from 20 existing prediction tools. Evaluation of VIPPID showed that it had superior performance (area under the curve = 0.91) over non-specific conventional tools. In addition, we also showed that the gene-specific model outperformed the non-gene-specific models. Our study demonstrated that disease-specific and gene-specific models can improve SNV pathogenicity prediction performance. This observation supports the notion that each feature of mutations in the model can be potentially used, in a new algorithm, to investigate the characteristics and function of the encoded proteins.
  •  
10.
  • Flores, Samuel, et al. (författare)
  • Multiscale modeling of macromolecular biosystems
  • 2012
  • Ingår i: Briefings in Bioinformatics. - : Oxford University Press (OUP). - 1467-5463 .- 1477-4054. ; 13:4, s. 395-405
  • Tidskriftsartikel (refereegranskat)abstract
    • In this article, we review the recent progress in multiresolution modeling of structure and dynamics of protein, RNA and their complexes. Many approaches using both physics-based and knowledge-based potentials have been developed at multiple granularities to model both protein and RNA. Coarse graining can be achieved not only in the length, but also in the time domain using discrete time and discrete state kinetic network models. Models with different resolutions can be combined either in a sequential or parallel fashion. Similarly, the modeling of assemblies is also often achieved using multiple granularities.The progress shows that a multiresolution approach has considerable potential to continue extending the length and time scales of macromolecular modeling.
  •  
Skapa referenser, mejla, bekava och länka
  • Resultat 1-10 av 41
Typ av publikation
tidskriftsartikel (35)
forskningsöversikt (6)
Typ av innehåll
refereegranskat (39)
övrigt vetenskapligt/konstnärligt (2)
Författare/redaktör
aut (4)
Li, CX (3)
Nielsen, Jens B, 196 ... (2)
Uhlén, Mathias (2)
Sonnhammer, Erik L L (2)
Borén, Jan, 1963 (2)
visa fler...
Staaf, Johan (2)
Gao, J. (1)
Li, L. (1)
Li, Y. (1)
Li, X. (1)
Zhou, M. (1)
Mardinoglu, Adil (1)
Lambrix, Patrick (1)
Adcock, IM (1)
Arif, Muhammad (1)
Huss, Mikael (1)
Marabita, F (1)
Jörnsten, Rebecka, 1 ... (1)
Fang, MY (1)
Landegren, Ulf (1)
Tegner, J (1)
Kerkhoven, Eduard, 1 ... (1)
Alarcon-Riquelme, ME (1)
Zounemat-Kermani, N (1)
Mardinoglu, Adil, 19 ... (1)
Stamatopoulos, K (1)
Agathangelidis, A (1)
Psomopoulos, F (1)
Chatzidimitriou, A (1)
Wheelock, AM (1)
van Duijn, Cornelia ... (1)
Ganna, Andrea (1)
Ingelsson, Erik (1)
Davis, S (1)
Morgan, M (1)
Orešič, Matej, 1967- (1)
Makinen, Veli (1)
Lindgren, David (1)
Vallon-Christersson, ... (1)
Chen, Wei (1)
Emanuelsson, Olof (1)
James, Peter (1)
Nordlund, P (1)
Ramos, M (1)
Wichmann, H. Erich (1)
De Marinis, Yang (1)
Schlesner, Matthias (1)
Schmitt, Thomas (1)
Pawitan, Yudi (1)
visa färre...
Lärosäte
Karolinska Institutet (15)
Stockholms universitet (6)
Uppsala universitet (5)
Lunds universitet (5)
Chalmers tekniska högskola (5)
Kungliga Tekniska Högskolan (4)
visa fler...
Linköpings universitet (3)
Göteborgs universitet (2)
Örebro universitet (2)
Högskolan i Halmstad (1)
Jönköping University (1)
Högskolan i Skövde (1)
Sveriges Lantbruksuniversitet (1)
visa färre...
Språk
Engelska (41)
Forskningsämne (UKÄ/SCB)
Naturvetenskap (22)
Medicin och hälsovetenskap (6)
Teknik (1)
Lantbruksvetenskap (1)

År

Kungliga biblioteket hanterar dina personuppgifter i enlighet med EU:s dataskyddsförordning (2018), GDPR. Läs mer om hur det funkar här.
Så här hanterar KB dina uppgifter vid användning av denna tjänst.

 
pil uppåt Stäng

Kopiera och spara länken för att återkomma till aktuell vy