Cargando…

DICER1: A Key Player in Rheumatoid Arthritis, at the Crossroads of Cellular Stress, Innate Immunity, and Chronic Inflammation in Aging

Loss-of-function or knockout mouse models have established a fundamental role for the RNAse III enzyme DICER1 in development and tissue morphogenesis and/or homeostasis. These functions are currently assumed to result mainly from the DICER1-dependent biogenesis of microRNAs which exhibit important g...

Descripción completa

Detalles Bibliográficos
Autores principales: De Cauwer, Aurore, Mariotte, Alexandre, Sibilia, Jean, Bahram, Seiamak, Georgel, Philippe
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6066587/
https://www.ncbi.nlm.nih.gov/pubmed/30087677
http://dx.doi.org/10.3389/fimmu.2018.01647
_version_ 1783342989329825792
author De Cauwer, Aurore
Mariotte, Alexandre
Sibilia, Jean
Bahram, Seiamak
Georgel, Philippe
author_facet De Cauwer, Aurore
Mariotte, Alexandre
Sibilia, Jean
Bahram, Seiamak
Georgel, Philippe
author_sort De Cauwer, Aurore
collection PubMed
description Loss-of-function or knockout mouse models have established a fundamental role for the RNAse III enzyme DICER1 in development and tissue morphogenesis and/or homeostasis. These functions are currently assumed to result mainly from the DICER1-dependent biogenesis of microRNAs which exhibit important gene expression regulatory properties. However, non-canonical DICER1 functions have recently emerged. These include interaction with the DNA damage response (DDR) pathway and the processing of cytotoxic non-coding RNAs, suggesting that DICER1 might also participate in the regulation of major cellular processes through miRNA-independent mechanisms. Recent findings indicated that reduced Dicer1 expression, which correlates with worsened symptoms in mouse models of joint inflammation, is also noted in fibroblast-like synoviocytes (FLS) harvested from rheumatoid arthritis (RA) patients, as opposed to FLS cultured from biopsies of osteoarthritic patients. In addition, low DICER1 levels are associated with the establishment of cellular stress and its associated responses, such as cellular senescence. Senescent and/or stressed cells are associated with an inflammatory secretome (cytokines and chemokines), as well as with “find-me” and “eat-me” signals which will attract and activate the innate immune compartment (NK cells, macrophages, and neutrophils) to be eliminated. Failure of this immunosurveillance mechanism and improper restauration of homeostasis could lead to the establishment of a systemic and chronic inflammatory state. In this review, we suggest that reduced DICER1 expression contributes to a vicious cycle during which accumulating inflammation and premature senescence, combined to inadequate innate immunity responses, creates the appropriate conditions for the initiation and/or progression of autoimmune-autoinflammatory diseases, such as RA.
format Online
Article
Text
id pubmed-6066587
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-60665872018-08-07 DICER1: A Key Player in Rheumatoid Arthritis, at the Crossroads of Cellular Stress, Innate Immunity, and Chronic Inflammation in Aging De Cauwer, Aurore Mariotte, Alexandre Sibilia, Jean Bahram, Seiamak Georgel, Philippe Front Immunol Immunology Loss-of-function or knockout mouse models have established a fundamental role for the RNAse III enzyme DICER1 in development and tissue morphogenesis and/or homeostasis. These functions are currently assumed to result mainly from the DICER1-dependent biogenesis of microRNAs which exhibit important gene expression regulatory properties. However, non-canonical DICER1 functions have recently emerged. These include interaction with the DNA damage response (DDR) pathway and the processing of cytotoxic non-coding RNAs, suggesting that DICER1 might also participate in the regulation of major cellular processes through miRNA-independent mechanisms. Recent findings indicated that reduced Dicer1 expression, which correlates with worsened symptoms in mouse models of joint inflammation, is also noted in fibroblast-like synoviocytes (FLS) harvested from rheumatoid arthritis (RA) patients, as opposed to FLS cultured from biopsies of osteoarthritic patients. In addition, low DICER1 levels are associated with the establishment of cellular stress and its associated responses, such as cellular senescence. Senescent and/or stressed cells are associated with an inflammatory secretome (cytokines and chemokines), as well as with “find-me” and “eat-me” signals which will attract and activate the innate immune compartment (NK cells, macrophages, and neutrophils) to be eliminated. Failure of this immunosurveillance mechanism and improper restauration of homeostasis could lead to the establishment of a systemic and chronic inflammatory state. In this review, we suggest that reduced DICER1 expression contributes to a vicious cycle during which accumulating inflammation and premature senescence, combined to inadequate innate immunity responses, creates the appropriate conditions for the initiation and/or progression of autoimmune-autoinflammatory diseases, such as RA. Frontiers Media S.A. 2018-07-24 /pmc/articles/PMC6066587/ /pubmed/30087677 http://dx.doi.org/10.3389/fimmu.2018.01647 Text en Copyright © 2018 De Cauwer, Mariotte, Sibilia, Bahram and Georgel. https://creativecommons.org/licenses/by/4.0/ This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
De Cauwer, Aurore
Mariotte, Alexandre
Sibilia, Jean
Bahram, Seiamak
Georgel, Philippe
DICER1: A Key Player in Rheumatoid Arthritis, at the Crossroads of Cellular Stress, Innate Immunity, and Chronic Inflammation in Aging
title DICER1: A Key Player in Rheumatoid Arthritis, at the Crossroads of Cellular Stress, Innate Immunity, and Chronic Inflammation in Aging
title_full DICER1: A Key Player in Rheumatoid Arthritis, at the Crossroads of Cellular Stress, Innate Immunity, and Chronic Inflammation in Aging
title_fullStr DICER1: A Key Player in Rheumatoid Arthritis, at the Crossroads of Cellular Stress, Innate Immunity, and Chronic Inflammation in Aging
title_full_unstemmed DICER1: A Key Player in Rheumatoid Arthritis, at the Crossroads of Cellular Stress, Innate Immunity, and Chronic Inflammation in Aging
title_short DICER1: A Key Player in Rheumatoid Arthritis, at the Crossroads of Cellular Stress, Innate Immunity, and Chronic Inflammation in Aging
title_sort dicer1: a key player in rheumatoid arthritis, at the crossroads of cellular stress, innate immunity, and chronic inflammation in aging
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6066587/
https://www.ncbi.nlm.nih.gov/pubmed/30087677
http://dx.doi.org/10.3389/fimmu.2018.01647
work_keys_str_mv AT decauweraurore dicer1akeyplayerinrheumatoidarthritisatthecrossroadsofcellularstressinnateimmunityandchronicinflammationinaging
AT mariottealexandre dicer1akeyplayerinrheumatoidarthritisatthecrossroadsofcellularstressinnateimmunityandchronicinflammationinaging
AT sibiliajean dicer1akeyplayerinrheumatoidarthritisatthecrossroadsofcellularstressinnateimmunityandchronicinflammationinaging
AT bahramseiamak dicer1akeyplayerinrheumatoidarthritisatthecrossroadsofcellularstressinnateimmunityandchronicinflammationinaging
AT georgelphilippe dicer1akeyplayerinrheumatoidarthritisatthecrossroadsofcellularstressinnateimmunityandchronicinflammationinaging