N-terminal BET bromodomain inhibitors disrupt a BRD4-p65 interaction and reduce inducible nitric oxide synthase transcription in pancreatic β-cells

Chronic inflammation of pancreatic islets is a key driver of β-cell damage that can lead to autoreactivity and the eventual onset of autoimmune diabetes (T1D). In the islet, elevated levels of proinflammatory cytokines induce the transcription of the inducible nitric oxide synthase (iNOS) gene, NOS2...

Full description

Bibliographic Details
Main Authors: Joshua A. Nord, Sarah L. Wynia-Smith, Alyssa L. Gehant, Rachel A. Jones Lipinski, Aaron Naatz, Inmaculada Rioja, Rab K. Prinjha, John A. Corbett, Brian C. Smith
Format: Article
Language:English
Published: Frontiers Media S.A. 2022-09-01
Series:Frontiers in Endocrinology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fendo.2022.923925/full
_version_ 1811209630508908544
author Joshua A. Nord
Sarah L. Wynia-Smith
Alyssa L. Gehant
Rachel A. Jones Lipinski
Aaron Naatz
Inmaculada Rioja
Rab K. Prinjha
John A. Corbett
Brian C. Smith
author_facet Joshua A. Nord
Sarah L. Wynia-Smith
Alyssa L. Gehant
Rachel A. Jones Lipinski
Aaron Naatz
Inmaculada Rioja
Rab K. Prinjha
John A. Corbett
Brian C. Smith
author_sort Joshua A. Nord
collection DOAJ
description Chronic inflammation of pancreatic islets is a key driver of β-cell damage that can lead to autoreactivity and the eventual onset of autoimmune diabetes (T1D). In the islet, elevated levels of proinflammatory cytokines induce the transcription of the inducible nitric oxide synthase (iNOS) gene, NOS2, ultimately resulting in increased nitric oxide (NO). Excessive or prolonged exposure to NO causes β-cell dysfunction and failure associated with defects in mitochondrial respiration. Recent studies showed that inhibition of the bromodomain and extraterminal domain (BET) family of proteins, a druggable class of epigenetic reader proteins, prevents the onset and progression of T1D in the non-obese diabetic mouse model. We hypothesized that BET proteins co-activate transcription of cytokine-induced inflammatory gene targets in β-cells and that selective, chemotherapeutic inhibition of BET bromodomains could reduce such transcription. Here, we investigated the ability of BET bromodomain small molecule inhibitors to reduce the β-cell response to the proinflammatory cytokine interleukin 1 beta (IL-1β). BET bromodomain inhibition attenuated IL-1β-induced transcription of the inflammatory mediator NOS2 and consequent iNOS protein and NO production. Reduced NOS2 transcription is consistent with inhibition of NF-κB facilitated by disrupting the interaction of a single BET family member, BRD4, with the NF-κB subunit, p65. Using recently reported selective inhibitors of the first and second BET bromodomains, inhibition of only the first bromodomain was necessary to reduce the interaction of BRD4 with p65 in β-cells. Moreover, inhibition of the first bromodomain was sufficient to mitigate IL-1β-driven decreases in mitochondrial oxygen consumption rates and β-cell viability. By identifying a role for the interaction between BRD4 and p65 in controlling the response of β-cells to proinflammatory cytokines, we provide mechanistic information on how BET bromodomain inhibition can decrease inflammation. These studies also support the potential therapeutic application of more selective BET bromodomain inhibitors in attenuating β-cell inflammation.
first_indexed 2024-04-12T04:42:31Z
format Article
id doaj.art-cb05e51596ea4c4db54e368ed7161c17
institution Directory Open Access Journal
issn 1664-2392
language English
last_indexed 2024-04-12T04:42:31Z
publishDate 2022-09-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Endocrinology
spelling doaj.art-cb05e51596ea4c4db54e368ed7161c172022-12-22T03:47:35ZengFrontiers Media S.A.Frontiers in Endocrinology1664-23922022-09-011310.3389/fendo.2022.923925923925N-terminal BET bromodomain inhibitors disrupt a BRD4-p65 interaction and reduce inducible nitric oxide synthase transcription in pancreatic β-cellsJoshua A. Nord0Sarah L. Wynia-Smith1Alyssa L. Gehant2Rachel A. Jones Lipinski3Aaron Naatz4Inmaculada Rioja5Rab K. Prinjha6John A. Corbett7Brian C. Smith8Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United StatesDepartment of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United StatesDepartment of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United StatesDepartment of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United StatesDepartment of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United StatesImmuno-Epigenetics, Immunology Research Unit, GlaxoSmithKline Medicines Research Centre, Stevenage, United KingdomImmuno-Epigenetics, Immunology Research Unit, GlaxoSmithKline Medicines Research Centre, Stevenage, United KingdomDepartment of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United StatesDepartment of Biochemistry, Medical College of Wisconsin, Milwaukee, WI, United StatesChronic inflammation of pancreatic islets is a key driver of β-cell damage that can lead to autoreactivity and the eventual onset of autoimmune diabetes (T1D). In the islet, elevated levels of proinflammatory cytokines induce the transcription of the inducible nitric oxide synthase (iNOS) gene, NOS2, ultimately resulting in increased nitric oxide (NO). Excessive or prolonged exposure to NO causes β-cell dysfunction and failure associated with defects in mitochondrial respiration. Recent studies showed that inhibition of the bromodomain and extraterminal domain (BET) family of proteins, a druggable class of epigenetic reader proteins, prevents the onset and progression of T1D in the non-obese diabetic mouse model. We hypothesized that BET proteins co-activate transcription of cytokine-induced inflammatory gene targets in β-cells and that selective, chemotherapeutic inhibition of BET bromodomains could reduce such transcription. Here, we investigated the ability of BET bromodomain small molecule inhibitors to reduce the β-cell response to the proinflammatory cytokine interleukin 1 beta (IL-1β). BET bromodomain inhibition attenuated IL-1β-induced transcription of the inflammatory mediator NOS2 and consequent iNOS protein and NO production. Reduced NOS2 transcription is consistent with inhibition of NF-κB facilitated by disrupting the interaction of a single BET family member, BRD4, with the NF-κB subunit, p65. Using recently reported selective inhibitors of the first and second BET bromodomains, inhibition of only the first bromodomain was necessary to reduce the interaction of BRD4 with p65 in β-cells. Moreover, inhibition of the first bromodomain was sufficient to mitigate IL-1β-driven decreases in mitochondrial oxygen consumption rates and β-cell viability. By identifying a role for the interaction between BRD4 and p65 in controlling the response of β-cells to proinflammatory cytokines, we provide mechanistic information on how BET bromodomain inhibition can decrease inflammation. These studies also support the potential therapeutic application of more selective BET bromodomain inhibitors in attenuating β-cell inflammation.https://www.frontiersin.org/articles/10.3389/fendo.2022.923925/fullBET bromodomainBRD4diabetessmall molecule inhibitoriNOSnitric oxide
spellingShingle Joshua A. Nord
Sarah L. Wynia-Smith
Alyssa L. Gehant
Rachel A. Jones Lipinski
Aaron Naatz
Inmaculada Rioja
Rab K. Prinjha
John A. Corbett
Brian C. Smith
N-terminal BET bromodomain inhibitors disrupt a BRD4-p65 interaction and reduce inducible nitric oxide synthase transcription in pancreatic β-cells
Frontiers in Endocrinology
BET bromodomain
BRD4
diabetes
small molecule inhibitor
iNOS
nitric oxide
title N-terminal BET bromodomain inhibitors disrupt a BRD4-p65 interaction and reduce inducible nitric oxide synthase transcription in pancreatic β-cells
title_full N-terminal BET bromodomain inhibitors disrupt a BRD4-p65 interaction and reduce inducible nitric oxide synthase transcription in pancreatic β-cells
title_fullStr N-terminal BET bromodomain inhibitors disrupt a BRD4-p65 interaction and reduce inducible nitric oxide synthase transcription in pancreatic β-cells
title_full_unstemmed N-terminal BET bromodomain inhibitors disrupt a BRD4-p65 interaction and reduce inducible nitric oxide synthase transcription in pancreatic β-cells
title_short N-terminal BET bromodomain inhibitors disrupt a BRD4-p65 interaction and reduce inducible nitric oxide synthase transcription in pancreatic β-cells
title_sort n terminal bet bromodomain inhibitors disrupt a brd4 p65 interaction and reduce inducible nitric oxide synthase transcription in pancreatic β cells
topic BET bromodomain
BRD4
diabetes
small molecule inhibitor
iNOS
nitric oxide
url https://www.frontiersin.org/articles/10.3389/fendo.2022.923925/full
work_keys_str_mv AT joshuaanord nterminalbetbromodomaininhibitorsdisruptabrd4p65interactionandreduceinduciblenitricoxidesynthasetranscriptioninpancreaticbcells
AT sarahlwyniasmith nterminalbetbromodomaininhibitorsdisruptabrd4p65interactionandreduceinduciblenitricoxidesynthasetranscriptioninpancreaticbcells
AT alyssalgehant nterminalbetbromodomaininhibitorsdisruptabrd4p65interactionandreduceinduciblenitricoxidesynthasetranscriptioninpancreaticbcells
AT rachelajoneslipinski nterminalbetbromodomaininhibitorsdisruptabrd4p65interactionandreduceinduciblenitricoxidesynthasetranscriptioninpancreaticbcells
AT aaronnaatz nterminalbetbromodomaininhibitorsdisruptabrd4p65interactionandreduceinduciblenitricoxidesynthasetranscriptioninpancreaticbcells
AT inmaculadarioja nterminalbetbromodomaininhibitorsdisruptabrd4p65interactionandreduceinduciblenitricoxidesynthasetranscriptioninpancreaticbcells
AT rabkprinjha nterminalbetbromodomaininhibitorsdisruptabrd4p65interactionandreduceinduciblenitricoxidesynthasetranscriptioninpancreaticbcells
AT johnacorbett nterminalbetbromodomaininhibitorsdisruptabrd4p65interactionandreduceinduciblenitricoxidesynthasetranscriptioninpancreaticbcells
AT briancsmith nterminalbetbromodomaininhibitorsdisruptabrd4p65interactionandreduceinduciblenitricoxidesynthasetranscriptioninpancreaticbcells