Sustained and intermittent hypoxia differentially modulate primary monocyte immunothrombotic responses to IL-1β stimulation

Venous thromboembolism (VTE) is a leading cause of preventable deaths in hospitals, and its incidence is not decreasing despite extensive efforts in clinical and laboratory research. Venous thrombi are primarily formed in the valve pockets of deep veins, where activated monocytes play a crucial role...

Full description

Bibliographic Details
Main Authors: Casper J.E. Wahlund, Safak Çaglayan, Paulo Czarnewski, John-Bjarne Hansen, Omri Snir
Format: Article
Language:English
Published: Frontiers Media S.A. 2023-09-01
Series:Frontiers in Immunology
Subjects:
Online Access:https://www.frontiersin.org/articles/10.3389/fimmu.2023.1240597/full
_version_ 1797688827651817472
author Casper J.E. Wahlund
Safak Çaglayan
Paulo Czarnewski
Paulo Czarnewski
John-Bjarne Hansen
John-Bjarne Hansen
Omri Snir
Omri Snir
author_facet Casper J.E. Wahlund
Safak Çaglayan
Paulo Czarnewski
Paulo Czarnewski
John-Bjarne Hansen
John-Bjarne Hansen
Omri Snir
Omri Snir
author_sort Casper J.E. Wahlund
collection DOAJ
description Venous thromboembolism (VTE) is a leading cause of preventable deaths in hospitals, and its incidence is not decreasing despite extensive efforts in clinical and laboratory research. Venous thrombi are primarily formed in the valve pockets of deep veins, where activated monocytes play a crucial role in bridging innate immune activation and hemostatic pathways through the production of inflammatory cytokines, chemokines, and tissue factor (TF) – a principal initiator of coagulation. In the valve pocket inflammation and hypoxia (sustained/intermittent) coexist, however their combined effects on immunothrombotic processes are poorly understood. Inflammation is strongly associated with VTE, while the additional contribution of hypoxia remains largely unexplored. To investigate this, we modelled the intricate conditions of the venous valve pocket using a state-of-the-art hypoxia chamber with software-controlled oxygen cycling. We comprehensively studied the effects of sustained and intermittent hypoxia alone, and in combination with VTE-associated inflammatory stimuli on primary monocytes. TF expression and activity was measured in monocytes subjected to sustained and intermittent hypoxia alone, or in combination with IL-1β. Monocyte responses were further analyzed in detailed by RNA sequencing and validated by ELISA. Stimulation with IL-1β alone promoted both transcription and activity of TF. Interestingly, the stimulatory effect of IL-1β on TF was attenuated by sustained hypoxia, but not by intermittent hypoxia. Our transcriptome analysis further confirmed that sustained hypoxia limited the pro-inflammatory response induced by IL-1β, and triggered a metabolic shift in monocytes. Intermittent hypoxia alone had a modest effect on monocyte transcript. However, in combination with IL-1β intermittent hypoxia significantly altered the expression of 2207 genes and enhanced the IL-1β-stimulatory effects on several chemokine and interleukin genes (e.g., IL-19, IL-24, IL-32, MIF), as well as genes involved in coagulation (thrombomodulin) and fibrinolysis (VEGFA, MMP9, MMP14 and PAI-1). Increased production of CCL2, IL-6 and TNF following stimulation with intermittent hypoxia and IL-1β was confirmed by ELISA. Our findings provide valuable insights into how the different hypoxic profiles shape the immunothrombotic response of monocytes and shed new light on the early events in the pathogenesis of venous thrombosis.
first_indexed 2024-03-12T01:37:12Z
format Article
id doaj.art-762a9bb2fa2d401fbd3dd6ef392e402e
institution Directory Open Access Journal
issn 1664-3224
language English
last_indexed 2024-03-12T01:37:12Z
publishDate 2023-09-01
publisher Frontiers Media S.A.
record_format Article
series Frontiers in Immunology
spelling doaj.art-762a9bb2fa2d401fbd3dd6ef392e402e2023-09-11T05:57:08ZengFrontiers Media S.A.Frontiers in Immunology1664-32242023-09-011410.3389/fimmu.2023.12405971240597Sustained and intermittent hypoxia differentially modulate primary monocyte immunothrombotic responses to IL-1β stimulationCasper J.E. Wahlund0Safak Çaglayan1Paulo Czarnewski2Paulo Czarnewski3John-Bjarne Hansen4John-Bjarne Hansen5Omri Snir6Omri Snir7Thrombosis Research Group (TREC), Department of Clinical Medicine, UiT – The Arctic University of Norway, Tromsø, NorwayThrombosis Research Group (TREC), Department of Clinical Medicine, UiT – The Arctic University of Norway, Tromsø, NorwayScience for Life Laboratory, Department of Gene Technology, KTH Royal Institute of Technology, Stockholm, SwedenScience for Life Laboratory, Department of Biochemistry and Biophysics, National Bioinformatics Infrastructure Sweden, Stockholm University, Stockholm, SwedenThrombosis Research Group (TREC), Department of Clinical Medicine, UiT – The Arctic University of Norway, Tromsø, NorwayDivision of Internal Medicine, University Hospital of North Norway, Tromsø, NorwayThrombosis Research Group (TREC), Department of Clinical Medicine, UiT – The Arctic University of Norway, Tromsø, NorwayDivision of Internal Medicine, University Hospital of North Norway, Tromsø, NorwayVenous thromboembolism (VTE) is a leading cause of preventable deaths in hospitals, and its incidence is not decreasing despite extensive efforts in clinical and laboratory research. Venous thrombi are primarily formed in the valve pockets of deep veins, where activated monocytes play a crucial role in bridging innate immune activation and hemostatic pathways through the production of inflammatory cytokines, chemokines, and tissue factor (TF) – a principal initiator of coagulation. In the valve pocket inflammation and hypoxia (sustained/intermittent) coexist, however their combined effects on immunothrombotic processes are poorly understood. Inflammation is strongly associated with VTE, while the additional contribution of hypoxia remains largely unexplored. To investigate this, we modelled the intricate conditions of the venous valve pocket using a state-of-the-art hypoxia chamber with software-controlled oxygen cycling. We comprehensively studied the effects of sustained and intermittent hypoxia alone, and in combination with VTE-associated inflammatory stimuli on primary monocytes. TF expression and activity was measured in monocytes subjected to sustained and intermittent hypoxia alone, or in combination with IL-1β. Monocyte responses were further analyzed in detailed by RNA sequencing and validated by ELISA. Stimulation with IL-1β alone promoted both transcription and activity of TF. Interestingly, the stimulatory effect of IL-1β on TF was attenuated by sustained hypoxia, but not by intermittent hypoxia. Our transcriptome analysis further confirmed that sustained hypoxia limited the pro-inflammatory response induced by IL-1β, and triggered a metabolic shift in monocytes. Intermittent hypoxia alone had a modest effect on monocyte transcript. However, in combination with IL-1β intermittent hypoxia significantly altered the expression of 2207 genes and enhanced the IL-1β-stimulatory effects on several chemokine and interleukin genes (e.g., IL-19, IL-24, IL-32, MIF), as well as genes involved in coagulation (thrombomodulin) and fibrinolysis (VEGFA, MMP9, MMP14 and PAI-1). Increased production of CCL2, IL-6 and TNF following stimulation with intermittent hypoxia and IL-1β was confirmed by ELISA. Our findings provide valuable insights into how the different hypoxic profiles shape the immunothrombotic response of monocytes and shed new light on the early events in the pathogenesis of venous thrombosis.https://www.frontiersin.org/articles/10.3389/fimmu.2023.1240597/fullmonocytesvenous thromboembolismimmunothrombosistissue factorSPP1deep vein thrombosis
spellingShingle Casper J.E. Wahlund
Safak Çaglayan
Paulo Czarnewski
Paulo Czarnewski
John-Bjarne Hansen
John-Bjarne Hansen
Omri Snir
Omri Snir
Sustained and intermittent hypoxia differentially modulate primary monocyte immunothrombotic responses to IL-1β stimulation
Frontiers in Immunology
monocytes
venous thromboembolism
immunothrombosis
tissue factor
SPP1
deep vein thrombosis
title Sustained and intermittent hypoxia differentially modulate primary monocyte immunothrombotic responses to IL-1β stimulation
title_full Sustained and intermittent hypoxia differentially modulate primary monocyte immunothrombotic responses to IL-1β stimulation
title_fullStr Sustained and intermittent hypoxia differentially modulate primary monocyte immunothrombotic responses to IL-1β stimulation
title_full_unstemmed Sustained and intermittent hypoxia differentially modulate primary monocyte immunothrombotic responses to IL-1β stimulation
title_short Sustained and intermittent hypoxia differentially modulate primary monocyte immunothrombotic responses to IL-1β stimulation
title_sort sustained and intermittent hypoxia differentially modulate primary monocyte immunothrombotic responses to il 1β stimulation
topic monocytes
venous thromboembolism
immunothrombosis
tissue factor
SPP1
deep vein thrombosis
url https://www.frontiersin.org/articles/10.3389/fimmu.2023.1240597/full
work_keys_str_mv AT casperjewahlund sustainedandintermittenthypoxiadifferentiallymodulateprimarymonocyteimmunothromboticresponsestoil1bstimulation
AT safakcaglayan sustainedandintermittenthypoxiadifferentiallymodulateprimarymonocyteimmunothromboticresponsestoil1bstimulation
AT pauloczarnewski sustainedandintermittenthypoxiadifferentiallymodulateprimarymonocyteimmunothromboticresponsestoil1bstimulation
AT pauloczarnewski sustainedandintermittenthypoxiadifferentiallymodulateprimarymonocyteimmunothromboticresponsestoil1bstimulation
AT johnbjarnehansen sustainedandintermittenthypoxiadifferentiallymodulateprimarymonocyteimmunothromboticresponsestoil1bstimulation
AT johnbjarnehansen sustainedandintermittenthypoxiadifferentiallymodulateprimarymonocyteimmunothromboticresponsestoil1bstimulation
AT omrisnir sustainedandintermittenthypoxiadifferentiallymodulateprimarymonocyteimmunothromboticresponsestoil1bstimulation
AT omrisnir sustainedandintermittenthypoxiadifferentiallymodulateprimarymonocyteimmunothromboticresponsestoil1bstimulation