Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
ACS Omega ; 8(33): 30177-30185, 2023 Aug 22.
Article in English | MEDLINE | ID: mdl-37636935

ABSTRACT

E3 ligases are enzymes that play a critical role in ubiquitin-mediated protein degradation and are involved in various cellular processes. Pharmacophore analysis is a useful approach for predicting E3 ligase binding selectivity, which involves identifying key chemical features necessary for a ligand to interact with a specific protein target cavity. While pharmacophore analysis is not always sufficient to accurately predict ligand binding affinity, it can be a valuable tool for filtering and/or designing focused libraries for screening campaigns. In this study, we present a fast and an inexpensive approach using a pharmacophore fingerprinting scheme known as ErG, which is used in a multi-class machine learning classification model. This model can assign the correct E3 ligase binder to its known E3 ligase and predict the probability of each molecule to bind to different E3 ligases. Practical applications of this approach are demonstrated on commercial libraries such as Asinex for the rational design of E3 ligase binders. The scripts and data associated with this study can be found on GitHub at https://github.com/Fraunhofer-ITMP/E3_binder_Model.

2.
Bioinform Adv ; 3(1): vbad045, 2023.
Article in English | MEDLINE | ID: mdl-37187795

ABSTRACT

Summary: The outbreak of Mpox virus (MPXV) infection in May 2022 is declared a global health emergency by WHO. A total of 84 330 cases have been confirmed as of 5 January 2023 and the numbers are on the rise. The MPXV pathophysiology and its underlying mechanisms are unfortunately not yet understood. Likewise, the knowledge of biochemicals and drugs used against MPXV and their downstream effects is sparse. In this work, using Knowledge Graph (KG) representations we have depicted chemical and biological aspects of MPXV. To achieve this, we have collected and rationally assembled several biological study results, assays, drug candidates and pre-clinical evidence to form a dynamic and comprehensive network. The KG is compliant with FAIR annotations allowing seamless transformation and integration to/with other formats and infrastructures. Availability and implementation: The programmatic scripts for Mpox KG are publicly available at https://github.com/Fraunhofer-ITMP/mpox-kg. It is hosted publicly at https://doi.org/10.18119/N9SG7D. Supplementary information: Supplementary data are available at Bioinformatics Advances online.

3.
Angew Chem Int Ed Engl ; 61(46): e202205858, 2022 11 14.
Article in English | MEDLINE | ID: mdl-36115062

ABSTRACT

SARS-CoV-2 (SCoV2) and its variants of concern pose serious challenges to the public health. The variants increased challenges to vaccines, thus necessitating for development of new intervention strategies including anti-virals. Within the international Covid19-NMR consortium, we have identified binders targeting the RNA genome of SCoV2. We established protocols for the production and NMR characterization of more than 80 % of all SCoV2 proteins. Here, we performed an NMR screening using a fragment library for binding to 25 SCoV2 proteins and identified hits also against previously unexplored SCoV2 proteins. Computational mapping was used to predict binding sites and identify functional moieties (chemotypes) of the ligands occupying these pockets. Striking consensus was observed between NMR-detected binding sites of the main protease and the computational procedure. Our investigation provides novel structural and chemical space for structure-based drug design against the SCoV2 proteome.


Subject(s)
COVID-19 Drug Treatment , SARS-CoV-2 , Humans , Proteome , Ligands , Drug Design
4.
Sci Rep ; 11(1): 11049, 2021 05 26.
Article in English | MEDLINE | ID: mdl-34040048

ABSTRACT

The SARS-CoV-2 pandemic has challenged researchers at a global scale. The scientific community's massive response has resulted in a flood of experiments, analyses, hypotheses, and publications, especially in the field of drug repurposing. However, many of the proposed therapeutic compounds obtained from SARS-CoV-2 specific assays are not in agreement and thus demonstrate the need for a singular source of COVID-19 related information from which a rational selection of drug repurposing candidates can be made. In this paper, we present the COVID-19 PHARMACOME, a comprehensive drug-target-mechanism graph generated from a compilation of 10 separate disease maps and sources of experimental data focused on SARS-CoV-2/COVID-19 pathophysiology. By applying our systematic approach, we were able to predict the synergistic effect of specific drug pairs, such as Remdesivir and Thioguanosine or Nelfinavir and Raloxifene, on SARS-CoV-2 infection. Experimental validation of our results demonstrate that our graph can be used to not only explore the involved mechanistic pathways, but also to identify novel combinations of drug repurposing candidates.


Subject(s)
Antiviral Agents/therapeutic use , COVID-19 Drug Treatment , Drug Repositioning/methods , SARS-CoV-2/physiology , Adenosine Monophosphate/analogs & derivatives , Adenosine Monophosphate/therapeutic use , Alanine/analogs & derivatives , Alanine/therapeutic use , Combined Modality Therapy , Computational Biology , Drug Synergism , Drug Therapy, Combination , GTP Phosphohydrolases/therapeutic use , Humans , Knowledge Bases , Nelfinavir/therapeutic use , Pandemics , Raloxifene Hydrochloride/therapeutic use
5.
J Alzheimers Dis ; 80(2): 831-840, 2021.
Article in English | MEDLINE | ID: mdl-33554913

ABSTRACT

BACKGROUND: Neuroimaging markers provide quantitative insight into brain structure and function in neurodegenerative diseases, such as Alzheimer's disease, where we lack mechanistic insights to explain pathophysiology. These mechanisms are often mediated by genes and genetic variations and are often studied through the lens of genome-wide association studies. Linking these two disparate layers (i.e., imaging and genetic variation) through causal relationships between biological entities involved in the disease's etiology would pave the way to large-scale mechanistic reasoning and interpretation. OBJECTIVE: We explore how genetic variants may lead to functional alterations of intermediate molecular traits, which can further impact neuroimaging hallmarks over a series of biological processes across multiple scales. METHODS: We present an approach in which knowledge pertaining to single nucleotide polymorphisms and imaging readouts is extracted from the literature, encoded in Biological Expression Language, and used in a novel workflow to assist in the functional interpretation of SNPs in a clinical context. RESULTS: We demonstrate our approach in a case scenario which proposes KANSL1 as a candidate gene that accounts for the clinically reported correlation between the incidence of the genetic variants and hippocampal atrophy. We find that the workflow prioritizes multiple mechanisms reported in the literature through which KANSL1 may have an impact on hippocampal atrophy such as through the dysregulation of cell proliferation, synaptic plasticity, and metabolic processes. CONCLUSION: We have presented an approach that enables pinpointing relevant genetic variants as well as investigating their functional role in biological processes spanning across several, diverse biological scales.


Subject(s)
Alzheimer Disease/genetics , Genetic Predisposition to Disease/genetics , Neuroimaging , Systems Biology , Alzheimer Disease/diagnostic imaging , Biomarkers/metabolism , Brain/metabolism , Brain/pathology , Genome-Wide Association Study/methods , Humans , Phenotype , Polymorphism, Single Nucleotide/genetics , Systems Biology/methods
6.
Bioinformatics ; 37(9): 1332-1334, 2021 06 09.
Article in English | MEDLINE | ID: mdl-32976572

ABSTRACT

SUMMARY: The COVID-19 crisis has elicited a global response by the scientific community that has led to a burst of publications on the pathophysiology of the virus. However, without coordinated efforts to organize this knowledge, it can remain hidden away from individual research groups. By extracting and formalizing this knowledge in a structured and computable form, as in the form of a knowledge graph, researchers can readily reason and analyze this information on a much larger scale. Here, we present the COVID-19 Knowledge Graph, an expansive cause-and-effect network constructed from scientific literature on the new coronavirus that aims to provide a comprehensive view of its pathophysiology. To make this resource available to the research community and facilitate its exploration and analysis, we also implemented a web application and released the KG in multiple standard formats. AVAILABILITY AND IMPLEMENTATION: The COVID-19 Knowledge Graph is publicly available under CC-0 license at https://github.com/covid19kg and https://bikmi.covid19-knowledgespace.de. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
COVID-19 , Software , Humans , Pattern Recognition, Automated , Publications , SARS-CoV-2
7.
J Alzheimers Dis ; 78(1): 87-95, 2020.
Article in English | MEDLINE | ID: mdl-32925069

ABSTRACT

BACKGROUND: Recent studies have suggested comorbid association between Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) through identification of shared molecular mechanisms. However, the inference is pre-dominantly literature-based and lacks interpretation of pre-disposed genomic variants and transcriptomic measurables. OBJECTIVE: In this study, we aim to identify shared genetic variants and dysregulated genes in AD and T2DM and explore their functional roles in the comorbidity between the diseases. METHODS: The genetic variants for AD and T2DM were retrieved from GWAS catalog, GWAS central, dbSNP, and DisGeNet and subjected to linkage disequilibrium analysis. Next, shared variants were prioritized using RegulomeDB and Polyphen-2. Afterwards, a knowledge assembly embedding prioritized variants and their corresponding genes was created by mining relevant literature using Biological Expression Language. Finally, coherently perturbed genes from gene expression meta-analysis were mapped to the knowledge assembly to pinpoint biological entities and processes and depict a mechanistic link between AD and T2DM. RESULTS: Our analysis identified four genes (i.e., ABCG1, COMT, MMP9, and SOD2) that could have dual roles in both AD and T2DM. Using cartoon representation, we have illustrated a set of causal events surrounding these genes which are associated to biological processes such as oxidative stress, insulin resistance, apoptosis and cognition. CONCLUSION: Our approach of using data as the driving force for unraveling disease etiologies eliminates literature bias and enables identification of novel entities that serve as the bridge between comorbid conditions.


Subject(s)
Alzheimer Disease/genetics , Diabetes Mellitus, Type 2/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Models, Biological
8.
Sci Rep ; 10(1): 10971, 2020 07 03.
Article in English | MEDLINE | ID: mdl-32620927

ABSTRACT

Translational research of many disease areas requires a longitudinal understanding of disease development and progression across all biologically relevant scales. Several corresponding studies are now available. However, to compile a comprehensive picture of a specific disease, multiple studies need to be analyzed and compared. A large number of clinical studies is nowadays conducted in the context of drug development in pharmaceutical research. However, legal and ethical constraints typically do not allow for sharing sensitive patient data. In consequence there exist data "silos", which slow down the overall scientific progress in translational research. In this paper, we suggest the idea of a virtual cohort (VC) to address this limitation. Our key idea is to describe a longitudinal patient cohort with the help of a generative statistical model, namely a modular Bayesian Network, in which individual modules are represented as sparse autoencoder networks. We show that with the help of such a model we can simulate subjects that are highly similar to real ones. Our approach allows for incorporating arbitrary multi-scale, multi-modal data without making specific distribution assumptions. Moreover, we demonstrate the possibility to simulate interventions (e.g. via a treatment) in the VC. Overall, our proposed approach opens the possibility to build sufficiently realistic VCs for multiple disease areas in the future.


Subject(s)
Bayes Theorem , Deep Learning , Translational Research, Biomedical/methods , Alzheimer Disease/diagnostic imaging , Alzheimer Disease/genetics , Brain/diagnostic imaging , Cohort Studies , Computer Simulation , Databases, Factual/statistics & numerical data , Disease Progression , Humans , Longitudinal Studies , Models, Statistical , Parkinson Disease/diagnosis , Polymorphism, Single Nucleotide , Translational Research, Biomedical/statistics & numerical data , User-Computer Interface
9.
Gigascience ; 8(11)2019 11 01.
Article in English | MEDLINE | ID: mdl-31730697

ABSTRACT

BACKGROUND: Precision medicine requires a stratification of patients by disease presentation that is sufficiently informative to allow for selecting treatments on a per-patient basis. For many diseases, such as neurological disorders, this stratification problem translates into a complex problem of clustering multivariate and relatively short time series because (i) these diseases are multifactorial and not well described by single clinical outcome variables and (ii) disease progression needs to be monitored over time. Additionally, clinical data often additionally are hindered by the presence of many missing values, further complicating any clustering attempts. FINDINGS: The problem of clustering multivariate short time series with many missing values is generally not well addressed in the literature. In this work, we propose a deep learning-based method to address this issue, variational deep embedding with recurrence (VaDER). VaDER relies on a Gaussian mixture variational autoencoder framework, which is further extended to (i) model multivariate time series and (ii) directly deal with missing values. We validated VaDER by accurately recovering clusters from simulated and benchmark data with known ground truth clustering, while varying the degree of missingness. We then used VaDER to successfully stratify patients with Alzheimer disease and patients with Parkinson disease into subgroups characterized by clinically divergent disease progression profiles. Additional analyses demonstrated that these clinical differences reflected known underlying aspects of Alzheimer disease and Parkinson disease. CONCLUSIONS: We believe our results show that VaDER can be of great value for future efforts in patient stratification, and multivariate time-series clustering in general.


Subject(s)
Alzheimer Disease/physiopathology , Databases, Factual , Deep Learning , Disease Progression , Models, Neurological , Parkinson Disease/physiopathology , Precision Medicine , Female , Humans , Male
10.
BMC Bioinformatics ; 20(1): 494, 2019 Oct 11.
Article in English | MEDLINE | ID: mdl-31604427

ABSTRACT

BACKGROUND: Literature derived knowledge assemblies have been used as an effective way of representing biological phenomenon and understanding disease etiology in systems biology. These include canonical pathway databases such as KEGG, Reactome and WikiPathways and disease specific network inventories such as causal biological networks database, PD map and NeuroMMSig. The represented knowledge in these resources delineates qualitative information focusing mainly on the causal relationships between biological entities. Genes, the major constituents of knowledge representations, tend to express differentially in different conditions such as cell types, brain regions and disease stages. A classical approach of interpreting a knowledge assembly is to explore gene expression patterns of the individual genes. However, an approach that enables quantification of the overall impact of differentially expressed genes in the corresponding network is still lacking. RESULTS: Using the concept of heat diffusion, we have devised an algorithm that is able to calculate the magnitude of regulation of a biological network using expression datasets. We have demonstrated that molecular mechanisms specific to Alzheimer (AD) and Parkinson Disease (PD) regulate with different intensities across spatial and temporal resolutions. Our approach depicts that the mitochondrial dysfunction in PD is severe in cortex and advanced stages of PD patients. Similarly, we have shown that the intensity of aggregation of neurofibrillary tangles (NFTs) in AD increases as the disease progresses. This finding is in concordance with previous studies that explain the burden of NFTs in stages of AD. CONCLUSIONS: This study is one of the first attempts that enable quantification of mechanisms represented as biological networks. We have been able to quantify the magnitude of regulation of a biological network and illustrate that the magnitudes are different across spatial and temporal resolution.


Subject(s)
Algorithms , Brain/metabolism , Neurodegenerative Diseases/metabolism , Systems Biology/methods , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Gene Expression Regulation , Humans , Metabolic Networks and Pathways , Mitochondria/metabolism , Mitochondria/physiology , Neurodegenerative Diseases/genetics , Neurodegenerative Diseases/physiopathology , Parkinson Disease/genetics , Parkinson Disease/metabolism , Parkinson Disease/physiopathology , Protein Interaction Maps , Signal Transduction
11.
J Alzheimers Dis ; 60(2): 721-731, 2017.
Article in English | MEDLINE | ID: mdl-28922161

ABSTRACT

BACKGROUND: Various studies suggest a comorbid association between Alzheimer's disease (AD) and type 2 diabetes mellitus (T2DM) indicating that there could be shared underlying pathophysiological mechanisms. OBJECTIVE: This study aims to systematically model relevant knowledge at the molecular level to find a mechanistic rationale explaining the existing comorbid association between AD and T2DM. METHOD: We have used a knowledge-based modeling approach to build two network models for AD and T2DM using Biological Expression Language (BEL), which is capable of capturing and representing causal and correlative relationships at both molecular and clinical levels from various knowledge resources. RESULTS: Using comparative analysis, we have identified several putative "shared pathways". We demonstrate, at a mechanistic level, how the insulin signaling pathway is related to other significant AD pathways such as the neurotrophin signaling pathway, PI3K/AKT signaling, MTOR signaling, and MAPK signaling and how these pathways do cross-talk with each other both in AD and T2DM. In addition, we present a mechanistic hypothesis that explains both favorable and adverse effects of the anti-diabetic drug metformin in AD. CONCLUSION: The two computable models introduced here provide a powerful framework to identify plausible mechanistic links shared between AD and T2DM and thereby identify targeted pathways for new therapeutics. Our approach can also be used to provide mechanistic answers to the question of why some T2DM treatments seem to increase the risk of AD.


Subject(s)
Alzheimer Disease/epidemiology , Diabetes Mellitus, Type 2/epidemiology , Diabetes Mellitus, Type 2/physiopathology , Insulin/metabolism , Signal Transduction/physiology , Alzheimer Disease/chemically induced , Alzheimer Disease/metabolism , Comorbidity , Computer Simulation , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Female , Humans , Hypoglycemic Agents/adverse effects , Male , Metabolic Networks and Pathways , Metformin/adverse effects , Models, Biological
12.
J Alzheimers Dis ; 59(3): 1045-1055, 2017.
Article in English | MEDLINE | ID: mdl-28731442

ABSTRACT

Perturbance in inflammatory pathways have been identified as one of the major factors which leads to neurodegenerative diseases (NDD). Owing to the limited access of human brain tissues and the immense complexity of the brain, animal models, specifically mouse models, play a key role in advancing the NDD field. However, many of these mouse models fail to reproduce the clinical manifestations and end points of the disease. NDD drugs, which passed the efficacy test in mice, were repeatedly not successful in clinical trials. There are numerous studies which are supporting and opposing the applicability of mouse models in neuroinflammation and NDD. In this paper, we assessed to what extend a mouse can mimic the cellular and molecular interactions in humans at a mechanism level. Based on our mechanistic modeling approach, we investigate the failure of a neuroinflammation targeted drug in the late phases of clinical trials based on the comparative analyses between the two species.


Subject(s)
Cytokines/metabolism , Disease Models, Animal , Encephalitis/etiology , Neurodegenerative Diseases/complications , Signal Transduction/physiology , Animals , Brain/metabolism , Brain/pathology , Encephalitis/genetics , Humans , Mice , Neurodegenerative Diseases/genetics , Signal Transduction/genetics , Species Specificity
13.
Bioinformatics ; 33(22): 3679-3681, 2017 Nov 15.
Article in English | MEDLINE | ID: mdl-28651363

ABSTRACT

MOTIVATION: The concept of a 'mechanism-based taxonomy of human disease' is currently replacing the outdated paradigm of diseases classified by clinical appearance. We have tackled the paradigm of mechanism-based patient subgroup identification in the challenging area of research on neurodegenerative diseases. RESULTS: We have developed a knowledge base representing essential pathophysiology mechanisms of neurodegenerative diseases. Together with dedicated algorithms, this knowledge base forms the basis for a 'mechanism-enrichment server' that supports the mechanistic interpretation of multiscale, multimodal clinical data. AVAILABILITY AND IMPLEMENTATION: NeuroMMSig is available at http://neurommsig.scai.fraunhofer.de/. CONTACT: martin.hofmann-apitius@scai.fraunhofer.de. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Algorithms , Knowledge Bases , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Humans , Internet , Models, Biological , Neurodegenerative Diseases/genetics , Software
14.
J Alzheimers Dis ; 56(2): 677-686, 2017.
Article in English | MEDLINE | ID: mdl-28035920

ABSTRACT

Neurodegenerative diseases including Alzheimer's disease are complex to tackle because of the complexity of the brain, both in structure and function. Such complexity is reflected by the involvement of various brain regions and multiple pathways in the etiology of neurodegenerative diseases that render single drug target approaches ineffective. Particularly in the area of neurodegeneration, attention has been drawn to repurposing existing drugs with proven efficacy and safety profiles. However, there is a lack of systematic analysis of the brain chemical space to predict the feasibility of repurposing strategies. Using a mechanism-based, drug-target interaction modeling approach, we have identified promising drug candidates for repositioning. Mechanistic cause-and-effect models consolidate relevant prior knowledge on drugs, targets, and pathways from the scientific literature and integrate insights derived from experimental data. We demonstrate the power of this approach by predicting two repositioning candidates for Alzheimer's disease and one for amyotrophic lateral sclerosis.


Subject(s)
Alzheimer Disease/drug therapy , Amyotrophic Lateral Sclerosis/drug therapy , Drug Repositioning , Neuroprotective Agents/pharmacology , Neuroprotective Agents/therapeutic use , Alzheimer Disease/metabolism , Amyotrophic Lateral Sclerosis/metabolism , Computational Biology , Computer Simulation , Cyclosporine/pharmacology , Cyclosporine/therapeutic use , Donepezil , Drug Repositioning/methods , Humans , Indans/pharmacology , Indans/therapeutic use , Molecular Probes , Piperidines/pharmacology , Piperidines/therapeutic use , Riluzole/pharmacology , Riluzole/therapeutic use , Structure-Activity Relationship
15.
Article in English | MEDLINE | ID: mdl-27554092

ABSTRACT

Success in extracting biological relationships is mainly dependent on the complexity of the task as well as the availability of high-quality training data. Here, we describe the new corpora in the systems biology modeling language BEL for training and testing biological relationship extraction systems that we prepared for the BioCreative V BEL track. BEL was designed to capture relationships not only between proteins or chemicals, but also complex events such as biological processes or disease states. A BEL nanopub is the smallest unit of information and represents a biological relationship with its provenance. In BEL relationships (called BEL statements), the entities are normalized to defined namespaces mainly derived from public repositories, such as sequence databases, MeSH or publicly available ontologies. In the BEL nanopubs, the BEL statements are associated with citation information and supportive evidence such as a text excerpt. To enable the training of extraction tools, we prepared BEL resources and made them available to the community. We selected a subset of these resources focusing on a reduced set of namespaces, namely, human and mouse genes, ChEBI chemicals, MeSH diseases and GO biological processes, as well as relationship types 'increases' and 'decreases'. The published training corpus contains 11 000 BEL statements from over 6000 supportive text excerpts. For method evaluation, we selected and re-annotated two smaller subcorpora containing 100 text excerpts. For this re-annotation, the inter-annotator agreement was measured by the BEL track evaluation environment and resulted in a maximal F-score of 91.18% for full statement agreement. In addition, for a set of 100 BEL statements, we do not only provide the gold standard expert annotations, but also text excerpts pre-selected by two automated systems. Those text excerpts were evaluated and manually annotated as true or false supportive in the course of the BioCreative V BEL track task.Database URL: http://wiki.openbel.org/display/BIOC/Datasets.


Subject(s)
Data Curation/methods , Data Mining/methods , Natural Language Processing , Animals , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...