Cargando…

Bone marrow deficiency of mRNA decaying protein Tristetraprolin increases inflammation and mitochondrial ROS but reduces hepatic lipoprotein production in LDLR knockout mice

Tristetraprolin (TTP), an mRNA binding and decaying protein, plays a significant role in controlling inflammation by decaying mRNAs encoding inflammatory cytokines such as TNFalpha. We aimed to test a hypothesis that TTP in bone marrow (BM) cells regulates atherogenesis by modulating inflammation an...

Descripción completa

Detalles Bibliográficos
Autores principales: Saaoud, Fatma, Wang, Junfeng, Iwanowycz, Stephen, Wang, Yuzhen, Altomare, Diego, Shao, Ying, Liu, Jianguo, Blackshear, Perry J., Lessner, Susan M., Murphy, E. Angela, Wang, Hong, Yang, Xiaofeng, Fan, Daping
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Elsevier 2020
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7767740/
https://www.ncbi.nlm.nih.gov/pubmed/32591281
http://dx.doi.org/10.1016/j.redox.2020.101609
_version_ 1783629027006742528
author Saaoud, Fatma
Wang, Junfeng
Iwanowycz, Stephen
Wang, Yuzhen
Altomare, Diego
Shao, Ying
Liu, Jianguo
Blackshear, Perry J.
Lessner, Susan M.
Murphy, E. Angela
Wang, Hong
Yang, Xiaofeng
Fan, Daping
author_facet Saaoud, Fatma
Wang, Junfeng
Iwanowycz, Stephen
Wang, Yuzhen
Altomare, Diego
Shao, Ying
Liu, Jianguo
Blackshear, Perry J.
Lessner, Susan M.
Murphy, E. Angela
Wang, Hong
Yang, Xiaofeng
Fan, Daping
author_sort Saaoud, Fatma
collection PubMed
description Tristetraprolin (TTP), an mRNA binding and decaying protein, plays a significant role in controlling inflammation by decaying mRNAs encoding inflammatory cytokines such as TNFalpha. We aimed to test a hypothesis that TTP in bone marrow (BM) cells regulates atherogenesis by modulating inflammation and lipid metabolism through the modulation of oxidative stress pathways by TTP target genes. In a BM transplantation study, lethally irradiated atherogenic LDLR(−/−) mice were reconstituted with BM cells from either wild type (TTP(+/+)) or TTP knockout (TTP(−/−)) mice, and fed a Western diet for 12 weeks. We made the following observations: (1) TTP(−/−) BM recipients display a significantly higher systemic and multi-organ inflammation than TTP(+/+) BM recipients; (2) BM TTP deficiency modulates hepatic expression of genes, detected by microarray, involved in lipid metabolism, inflammatory responses, and oxidative stress; (3) TTP(−/−) BM derived macrophages increase production of mitochondrial reactive oxygen species (mtROS); (4) BM-TTP(−/−) mice display a significant reduction in serum VLDL/LDL levels, and attenuated hepatic steatosis compared to controls; and (5) Reduction of serum VLDL/LDL levels offsets the increased inflammation, resulting in no changes in atherosclerosis. These findings provide a novel mechanistic insight into the roles of TTP-mediated mRNA decay in bone marrow-derived cells in regulating systemic inflammation, oxidative stress, and liver VLDL/LDL biogenesis.
format Online
Article
Text
id pubmed-7767740
institution National Center for Biotechnology Information
language English
publishDate 2020
publisher Elsevier
record_format MEDLINE/PubMed
spelling pubmed-77677402020-12-29 Bone marrow deficiency of mRNA decaying protein Tristetraprolin increases inflammation and mitochondrial ROS but reduces hepatic lipoprotein production in LDLR knockout mice Saaoud, Fatma Wang, Junfeng Iwanowycz, Stephen Wang, Yuzhen Altomare, Diego Shao, Ying Liu, Jianguo Blackshear, Perry J. Lessner, Susan M. Murphy, E. Angela Wang, Hong Yang, Xiaofeng Fan, Daping Redox Biol Articles from the Special Issue on Redox Signalling and Cardiovascular Disease; Edited by Christopher Kevil and Yabing Chen Tristetraprolin (TTP), an mRNA binding and decaying protein, plays a significant role in controlling inflammation by decaying mRNAs encoding inflammatory cytokines such as TNFalpha. We aimed to test a hypothesis that TTP in bone marrow (BM) cells regulates atherogenesis by modulating inflammation and lipid metabolism through the modulation of oxidative stress pathways by TTP target genes. In a BM transplantation study, lethally irradiated atherogenic LDLR(−/−) mice were reconstituted with BM cells from either wild type (TTP(+/+)) or TTP knockout (TTP(−/−)) mice, and fed a Western diet for 12 weeks. We made the following observations: (1) TTP(−/−) BM recipients display a significantly higher systemic and multi-organ inflammation than TTP(+/+) BM recipients; (2) BM TTP deficiency modulates hepatic expression of genes, detected by microarray, involved in lipid metabolism, inflammatory responses, and oxidative stress; (3) TTP(−/−) BM derived macrophages increase production of mitochondrial reactive oxygen species (mtROS); (4) BM-TTP(−/−) mice display a significant reduction in serum VLDL/LDL levels, and attenuated hepatic steatosis compared to controls; and (5) Reduction of serum VLDL/LDL levels offsets the increased inflammation, resulting in no changes in atherosclerosis. These findings provide a novel mechanistic insight into the roles of TTP-mediated mRNA decay in bone marrow-derived cells in regulating systemic inflammation, oxidative stress, and liver VLDL/LDL biogenesis. Elsevier 2020-06-17 /pmc/articles/PMC7767740/ /pubmed/32591281 http://dx.doi.org/10.1016/j.redox.2020.101609 Text en © 2020 The Authors. Published by Elsevier B.V. http://creativecommons.org/licenses/by-nc-nd/4.0/ This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
spellingShingle Articles from the Special Issue on Redox Signalling and Cardiovascular Disease; Edited by Christopher Kevil and Yabing Chen
Saaoud, Fatma
Wang, Junfeng
Iwanowycz, Stephen
Wang, Yuzhen
Altomare, Diego
Shao, Ying
Liu, Jianguo
Blackshear, Perry J.
Lessner, Susan M.
Murphy, E. Angela
Wang, Hong
Yang, Xiaofeng
Fan, Daping
Bone marrow deficiency of mRNA decaying protein Tristetraprolin increases inflammation and mitochondrial ROS but reduces hepatic lipoprotein production in LDLR knockout mice
title Bone marrow deficiency of mRNA decaying protein Tristetraprolin increases inflammation and mitochondrial ROS but reduces hepatic lipoprotein production in LDLR knockout mice
title_full Bone marrow deficiency of mRNA decaying protein Tristetraprolin increases inflammation and mitochondrial ROS but reduces hepatic lipoprotein production in LDLR knockout mice
title_fullStr Bone marrow deficiency of mRNA decaying protein Tristetraprolin increases inflammation and mitochondrial ROS but reduces hepatic lipoprotein production in LDLR knockout mice
title_full_unstemmed Bone marrow deficiency of mRNA decaying protein Tristetraprolin increases inflammation and mitochondrial ROS but reduces hepatic lipoprotein production in LDLR knockout mice
title_short Bone marrow deficiency of mRNA decaying protein Tristetraprolin increases inflammation and mitochondrial ROS but reduces hepatic lipoprotein production in LDLR knockout mice
title_sort bone marrow deficiency of mrna decaying protein tristetraprolin increases inflammation and mitochondrial ros but reduces hepatic lipoprotein production in ldlr knockout mice
topic Articles from the Special Issue on Redox Signalling and Cardiovascular Disease; Edited by Christopher Kevil and Yabing Chen
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7767740/
https://www.ncbi.nlm.nih.gov/pubmed/32591281
http://dx.doi.org/10.1016/j.redox.2020.101609
work_keys_str_mv AT saaoudfatma bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT wangjunfeng bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT iwanowyczstephen bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT wangyuzhen bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT altomarediego bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT shaoying bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT liujianguo bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT blackshearperryj bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT lessnersusanm bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT murphyeangela bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT wanghong bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT yangxiaofeng bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice
AT fandaping bonemarrowdeficiencyofmrnadecayingproteintristetraprolinincreasesinflammationandmitochondrialrosbutreduceshepaticlipoproteinproductioninldlrknockoutmice