SwePub
Sök i SwePub databas

  Extended search

Träfflista för sökning "WFRF:(Erdem Cemal) "

Search: WFRF:(Erdem Cemal)

  • Result 1-10 of 11
Sort/group result
   
EnumerationReferenceCoverFind
1.
  • Brain, Martin, et al. (author)
  • Event-sequence testing using answer-set programming
  • 2012
  • In: International Journal on Advances in Software. - 1942-2628. ; 5:3 & 4, s. 237-251
  • Journal article (peer-reviewed)abstract
    • In many applications, faults are triggered by events that occur in a particular order. In fact, many bugs are caused by the interaction of only a low number of such events. Based on this assumption, sequence covering arrays (SCAs) have recently been proposed as suitable designs for event sequence testing. In practice, directly applying SCAs for testing is often impaired by additional constraints, and SCAs have to be adapted to fit application-specific needs. Modifying precomputed SCAs to account for problem variations can be problematic, if not impossible, and developing dedicated algorithms is costly. In this article, we propose answer-set programming (ASP), a well-known knowledge-representation formalism from the area of artificial intelligence based on logic programming, as a declarative paradigm for computing SCAs. Our approach allows to concisely state complex coverage criteria in an elaboration tolerant way, i.e., small variations of a problem specification require only small modifications of the ASP representation. Employing ASP for computing SCAs is further justified by new complexity results related to event-sequence testing that are established in this work.
  •  
2.
  • Erdem, Cemal, et al. (author)
  • A scalable, open-source implementation of a large-scale mechanistic model for single cell proliferation and death signaling
  • 2022
  • In: Nature Communications. - : Nature Publishing Group. - 2041-1723. ; 13:1
  • Journal article (peer-reviewed)abstract
    • Abstract Mechanistic models of how single cells respond to different perturbations can help integrate disparate big data sets or predict response to varied drug combinations. However, the construction and simulation of such models have proved challenging. Here, we developed a python-based model creation and simulation pipeline that converts a few structured text files into an SBML standard and is high-performance- and cloud-computing ready. We applied this pipeline to our large-scale, mechanistic pan-cancer signaling model (named SPARCED) and demonstrate it by adding an IFNγ pathway submodel. We then investigated whether a putative crosstalk mechanism could be consistent with experimental observations from the LINCS MCF10A Data Cube that IFNγ acts as an anti-proliferative factor. The analyses suggested this observation can be explained by IFNγ-induced SOCS1 sequestering activated EGF receptors. This work forms a foundational recipe for increased mechanistic model-based data integration on a single-cell level, an important building block for clinically-predictive mechanistic models.
  •  
3.
  • Erdem, Cemal, et al. (author)
  • Inhibition of RPS6K reveals context-dependent Akt activity in luminal breast cancer cells
  • 2021
  • In: PloS Computational Biology. - : Public Library of Science (PLoS). - 1553-734X .- 1553-7358. ; 17:6
  • Journal article (peer-reviewed)abstract
    • Aberrant signaling through insulin (Ins) and insulin-like growth factor I (IGF1) receptors contribute to the risk and advancement of many cancer types by activating cell survival cascades. Similarities between these pathways have thus far prevented the development of pharmacological interventions that specifically target either Ins or IGF1 signaling. To identify differences in early Ins and IGF1 signaling mechanisms, we developed a dual receptor (IGF1R & InsR) computational response model. The model suggested that ribosomal protein S6 kinase (RPS6K) plays a critical role in regulating MAPK and Akt activation levels in response to Ins and IGF1 stimulation. As predicted, perturbing RPS6K kinase activity led to an increased Akt activation with Ins stimulation compared to IGF1 stimulation. Being able to discern differential downstream signaling, we can explore improved anti-IGF1R cancer therapies by eliminating the emergence of compensation mechanisms without disrupting InsR signaling.
  •  
4.
  • Erdem, Cemal, et al. (author)
  • Mathematical modeling of Behçet's disease : A dynamical systems approach
  • 2015
  • In: Journal of biological systems. - : World Scientific. - 0218-3390. ; 23:02, s. 231-257
  • Journal article (peer-reviewed)abstract
    • Behçet's Disease (BD) is a multi-systemic, auto-inflammatory disorder that is characterized by recurrent episodes of inflammatory manifestations affecting skin, mucosa, eyes, blood vessels, joints and several other organs. BD is classified as a multifactorial disease with an important contribution of genetics. Genetic studies suggest that there is a strong association of BD with a Class I major histocompatibility complex antigen, named HLA-B*51, along with several other weaker associations with genes encoding proteins involved in inflammation. However, pathogenic mechanisms associated with these genetic variations and their interactions with the environment have not been elucidated yet. In this paper, we present a mathematical model for BD based on a dynamical systems perspective that captures especially the relapsing nature of the disease. We propose a disease progression mechanism and construct a model, in the form of coupled ordinary differential equations (ODEs), which reveals the occurrence pattern of the disease in the population. According to our model, the disease has three distinct modes describing different phenotypes of people carrying HLA-B*51 tissue antigen, namely, the Healthy Carrier, the Potential Patient and the Active Patient. We herein present an exemplary mathematical model for BD, for the first time in the literature, that concisely captures the actions of many cell types together with genetic and environmental effects. The proposed model provides insight into this complex inflammatory disease which may lead to identification of new tools for its treatment and prevention.
  •  
5.
  • Erdem, Cemal, et al. (author)
  • MEMMAL : A tool for expanding large-scale mechanistic models with machine learned associations and big datasets
  • 2023
  • In: Frontiers in Systems Biology. - : Frontiers Media S.A.. - 2674-0702. ; 3
  • Journal article (peer-reviewed)abstract
    • Computational models that can explain and predict complex sub-cellular, cellular, and tissue-level drug response mechanisms could speed drug discovery and prioritize patient-specific treatments (i.e., precision medicine). Some models are mechanistic with detailed equations describing known (or supposed) physicochemical processes, while some are statistical or machine learning-based approaches, that explain datasets but have no mechanistic or causal guarantees. These two types of modeling are rarely combined, missing the opportunity to explore possibly causal but data-driven new knowledge while explaining what is already known. Here, we explore combining machine learned associations with mechanistic models to develop computational models that could more fully represent cellular behavior. In this proposed MEMMAL (MEchanistic Modeling with MAchine Learning) framework, machine learning/statistical models built using omics datasets provide predictions for new interactions between genes and proteins where there is physicochemical uncertainty. These interactions are used as a basis for new reactions in mechanistic models. As a test case, we focused on incorporating novel IFNγ/PD-L1 related associations into a large-scale mechanistic model for cell proliferation and death to better recapitulate the recently released NIH LINCS Consortium MCF10A dataset and enable description of the cellular response to checkpoint inhibitor immunotherapies. This work is a template for combining big-data-inferred interactions with mechanistic models, which could be more broadly applicable for building multi-scale precision medicine and whole cell models.
  •  
6.
  • Erdem, Cemal, et al. (author)
  • MOBILE pipeline enables identification of context-specific networks and regulatory mechanisms
  • 2023
  • In: Nature Communications. - : Springer Nature. - 2041-1723. ; 14:1
  • Journal article (peer-reviewed)abstract
    • Robust identification of context-specific network features that control cellular phenotypes remains a challenge. We here introduce MOBILE (Multi-Omics Binary Integration via Lasso Ensembles) to nominate molecular features associated with cellular phenotypes and pathways. First, we use MOBILE to nominate mechanisms of interferon-γ (IFNγ) regulated PD-L1 expression. Our analyses suggest that IFNγ-controlled PD-L1 expression involves BST2 , CLIC2 , FAM83D , ACSL5 , and HIST2H2AA3 genes, which were supported by prior literature. We also compare networks activated by related family members transforming growth factor-beta 1 (TGFβ1) and bone morphogenetic protein 2 (BMP2) and find that differences in ligand-induced changes in cell size and clustering properties are related to differences in laminin/collagen pathway activity. Finally, we demonstrate the broad applicability and adaptability of MOBILE by analyzing publicly available molecular datasets to investigate breast cancer subtype specific networks. Given the ever-growing availability of multi-omics datasets, we envision that MOBILE will be broadly useful for identification of context-specific molecular features and pathways.
  •  
7.
  • Erdem, Cemal, et al. (author)
  • Proteomic screening and lasso regression reveal differential signaling in insulin and insulin-like growth factor I (IGF1) pathways
  • 2016
  • In: Molecular & Cellular Proteomics. - : Elsevier. - 1535-9476 .- 1535-9484. ; 15:9, s. 3045-3057
  • Journal article (peer-reviewed)abstract
    • Insulin and insulin-like growth factor I (IGF1) influence cancer risk and progression through poorly understood mechanisms. To better understand the roles of insulin and IGF1 signaling in breast cancer, we combined proteomic screening with computational network inference to uncover differences in IGF1 and insulin induced signaling. Using reverse phase protein array, we measured the levels of 134 proteins in 21 breast cancer cell lines stimulated with IGF1 or insulin for up to 48 h. We then constructed directed protein expression networks using three separate methods: (i) lasso regression, (ii) conventional matrix inversion, and (iii) entropy maximization. These networks, named here as the time translation models, were analyzed and the inferred interactions were ranked by differential magnitude to identify pathway differences. The two top candidates, chosen for experimental validation, were shown to regulate IGF1/insulin induced phosphorylation events. First, acetyl-CoA carboxylase (ACC) knock-down was shown to increase the level of mitogen-activated protein kinase (MAPK) phosphorylation. Second, stable knock-down of E-Cadherin increased the phospho-Akt protein levels. Both of the knock-down perturbations incurred phosphorylation responses stronger in IGF1 stimulated cells compared with insulin. Overall, the time-translation modeling coupled to wet-lab experiments has proven to be powerful in inferring differential interactions downstream of IGF1 and insulin signaling, in vitro.
  •  
8.
  • Erdem, Esra, et al. (author)
  • Answer-set programming as a new approach to event-sequence testing
  • 2011
  • In: VALID 2011 - 3rd International Conference on Advances in System Testing and Validation Lifecycle. - 9781612081687 ; , s. 25-34
  • Conference paper (peer-reviewed)abstract
    • In many applications, faults are triggered by events that occur in a particular order. Based on the assumption that most bugs are caused by the interaction of a low number of events, Kuhn et al. recently introduced sequence covering arrays (SCAs) as suitable designs for event sequence testing. In practice, directly applying SCAs for testing is often impaired by additional constraints, and SCAs have to be adapted to fit application-specific needs. Modifying precomputed SCAs to account for problem variations can be problematic, if not impossible, and developing dedicated algorithms is costly. In this paper, we propose answer-set programming (ASP), a well-known knowledge-representation formalism from the area of artificial intelligence based on logic programming, as a declarative paradigm for computing SCAs. Our approach allows to concisely state complex coverage criteria in an elaboration tolerant way, i.e., small variations of a problem specification require only small modifications of the ASP representation.
  •  
9.
  • Gross, Sean M., et al. (author)
  • A multi-omic analysis of MCF10A cells provides a resource for integrative assessment of ligand-mediated molecular and phenotypic responses
  • 2022
  • In: Communications Biology. - : Springer Nature. - 2399-3642. ; 5:1
  • Journal article (peer-reviewed)abstract
    • The phenotype of a cell and its underlying molecular state is strongly influenced by extracellular signals, including growth factors, hormones, and extracellular matrix proteins. While these signals are normally tightly controlled, their dysregulation leads to phenotypic and molecular states associated with diverse diseases. To develop a detailed understanding of the linkage between molecular and phenotypic changes, we generated a comprehensive dataset that catalogs the transcriptional, proteomic, epigenomic and phenotypic responses of MCF10A mammary epithelial cells after exposure to the ligands EGF, HGF, OSM, IFNG, TGFB and BMP2. Systematic assessment of the molecular and cellular phenotypes induced by these ligands comprise the LINCS Microenvironment (ME) perturbation dataset, which has been curated and made publicly available for community-wide analysis and development of novel computational methods ( synapse.org/LINCS_MCF10A ). In illustrative analyses, we demonstrate how this dataset can be used to discover functionally related molecular features linked to specific cellular phenotypes. Beyond these analyses, this dataset will serve as a resource for the broader scientific community to mine for biological insights, to compare signals carried across distinct molecular modalities, and to develop new computational methods for integrative data analysis.
  •  
10.
  • Mutsuddy, Arnab, et al. (author)
  • Computational speed-up of large-scale, single-cell model simulations via a fully integrated SBML-based format
  • 2023
  • In: Bioinformatics Advances. - : Oxford University Press. - 2635-0041. ; 3:1
  • Journal article (peer-reviewed)abstract
    • Summary: Large-scale and whole-cell modeling has multiple challenges, including scalable model building and module communication bottlenecks (e.g. between metabolism, gene expression, signaling, etc.). We previously developed an open-source, scalable format for a large-scale mechanistic model of proliferation and death signaling dynamics, but communication bottlenecks between gene expression and protein biochemistry modules remained. Here, we developed two solutions to communication bottlenecks that speed-up simulation by ∼4-fold for hybrid stochastic-deterministic simulations and by over 100-fold for fully deterministic simulations. Fully deterministic speed-up facilitates model initialization, parameter estimation and sensitivity analysis tasks.Availability and implementation: Source code is freely available at https://github.com/birtwistlelab/SPARCED/releases/tag/v1.3.0 implemented in python, and supported on Linux, Windows and MacOS (via Docker).
  •  
Skapa referenser, mejla, bekava och länka
  • Result 1-10 of 11

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 Close

Copy and save the link in order to return to this view