Cargando…

Information-theoretic analysis of a model of CAR-4–1BB-mediated NFκB activation

Systems biology utilizes computational approaches to examine an array of biological processes, such as cell signaling, metabolomics and pharmacology. This includes mathematical modeling of CAR T cells, a modality of cancer therapy by which genetically engineered immune cells recognize and combat a c...

Descripción completa

Detalles Bibliográficos
Autores principales: Tserunyan, Vardges, Finley, Stacey
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Cold Spring Harbor Laboratory 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10274880/
https://www.ncbi.nlm.nih.gov/pubmed/37333129
http://dx.doi.org/10.1101/2023.06.09.544433
_version_ 1785059804851470336
author Tserunyan, Vardges
Finley, Stacey
author_facet Tserunyan, Vardges
Finley, Stacey
author_sort Tserunyan, Vardges
collection PubMed
description Systems biology utilizes computational approaches to examine an array of biological processes, such as cell signaling, metabolomics and pharmacology. This includes mathematical modeling of CAR T cells, a modality of cancer therapy by which genetically engineered immune cells recognize and combat a cancerous target. While successful against hematologic malignancies, CAR T cells have shown limited success against other cancer types. Thus, more research is needed to understand their mechanisms of action and leverage their full potential. In our work, we set out to apply information theory on a mathematical model of cell signaling of CAR-mediated activation following antigen encounter. First, we estimated channel capacity for CAR-4–1BB-mediated NFκB signal transduction. Next, we evaluated the pathway’s ability to distinguish contrasting “low” and “high” antigen concentration levels, depending on the amount of intrinsic noise. Finally, we assessed the fidelity by which NFκB activation reflects the encountered antigen concentration, depending on the prevalence of antigen-positive targets in tumor population. We found that in most scenarios, fold change in the nuclear concentration of NFκB carries a higher channel capacity for the pathway than NFκB’s absolute response. Additionally, we found that most errors in transducing the antigen signal through the pathway skew towards underestimating the concentration of encountered antigen. Finally, we found that disabling IKKβ deactivation could increase signaling fidelity against targets with antigen-negative cells. Our information-theoretic analysis of signal transduction can provide novel perspectives on biological signaling, as well as enable a more informed path to cell engineering.
format Online
Article
Text
id pubmed-10274880
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Cold Spring Harbor Laboratory
record_format MEDLINE/PubMed
spelling pubmed-102748802023-06-17 Information-theoretic analysis of a model of CAR-4–1BB-mediated NFκB activation Tserunyan, Vardges Finley, Stacey bioRxiv Article Systems biology utilizes computational approaches to examine an array of biological processes, such as cell signaling, metabolomics and pharmacology. This includes mathematical modeling of CAR T cells, a modality of cancer therapy by which genetically engineered immune cells recognize and combat a cancerous target. While successful against hematologic malignancies, CAR T cells have shown limited success against other cancer types. Thus, more research is needed to understand their mechanisms of action and leverage their full potential. In our work, we set out to apply information theory on a mathematical model of cell signaling of CAR-mediated activation following antigen encounter. First, we estimated channel capacity for CAR-4–1BB-mediated NFκB signal transduction. Next, we evaluated the pathway’s ability to distinguish contrasting “low” and “high” antigen concentration levels, depending on the amount of intrinsic noise. Finally, we assessed the fidelity by which NFκB activation reflects the encountered antigen concentration, depending on the prevalence of antigen-positive targets in tumor population. We found that in most scenarios, fold change in the nuclear concentration of NFκB carries a higher channel capacity for the pathway than NFκB’s absolute response. Additionally, we found that most errors in transducing the antigen signal through the pathway skew towards underestimating the concentration of encountered antigen. Finally, we found that disabling IKKβ deactivation could increase signaling fidelity against targets with antigen-negative cells. Our information-theoretic analysis of signal transduction can provide novel perspectives on biological signaling, as well as enable a more informed path to cell engineering. Cold Spring Harbor Laboratory 2023-06-10 /pmc/articles/PMC10274880/ /pubmed/37333129 http://dx.doi.org/10.1101/2023.06.09.544433 Text en https://creativecommons.org/licenses/by-nc/4.0/This work is licensed under a Creative Commons Attribution-NonCommercial 4.0 International License (https://creativecommons.org/licenses/by-nc/4.0/) , which allows reusers to distribute, remix, adapt, and build upon the material in any medium or format for noncommercial purposes only, and only so long as attribution is given to the creator.
spellingShingle Article
Tserunyan, Vardges
Finley, Stacey
Information-theoretic analysis of a model of CAR-4–1BB-mediated NFκB activation
title Information-theoretic analysis of a model of CAR-4–1BB-mediated NFκB activation
title_full Information-theoretic analysis of a model of CAR-4–1BB-mediated NFκB activation
title_fullStr Information-theoretic analysis of a model of CAR-4–1BB-mediated NFκB activation
title_full_unstemmed Information-theoretic analysis of a model of CAR-4–1BB-mediated NFκB activation
title_short Information-theoretic analysis of a model of CAR-4–1BB-mediated NFκB activation
title_sort information-theoretic analysis of a model of car-4–1bb-mediated nfκb activation
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10274880/
https://www.ncbi.nlm.nih.gov/pubmed/37333129
http://dx.doi.org/10.1101/2023.06.09.544433
work_keys_str_mv AT tserunyanvardges informationtheoreticanalysisofamodelofcar41bbmediatednfkbactivation
AT finleystacey informationtheoreticanalysisofamodelofcar41bbmediatednfkbactivation