Cargando…

Epitranscriptome profiling of spleen mRNA m(6)A methylation reveals pathways of host responses to malaria parasite infection

N(6) -Methyladenosine (m(6)A), the most abundant mammalian mRNA modification, has been reported to modulate various viral infections. Although it has been confirmed that RNA modifications can also modulate the replication and development of different parasites, the role of the RNA epitranscriptome i...

Descripción completa

Detalles Bibliográficos
Autores principales: Wang, Luoluo, Wu, Jian, Liu, Runzhou, Chen, Wenjun, Pang, Zhichang, Zhou, Fan, Xia, Lu, Huang, Jia, Pan, Tao, Su, Xin-zhuan, Wang, Xiaoyun
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9531237/
https://www.ncbi.nlm.nih.gov/pubmed/36203583
http://dx.doi.org/10.3389/fimmu.2022.998756
_version_ 1784801860933124096
author Wang, Luoluo
Wu, Jian
Liu, Runzhou
Chen, Wenjun
Pang, Zhichang
Zhou, Fan
Xia, Lu
Huang, Jia
Pan, Tao
Su, Xin-zhuan
Wang, Xiaoyun
author_facet Wang, Luoluo
Wu, Jian
Liu, Runzhou
Chen, Wenjun
Pang, Zhichang
Zhou, Fan
Xia, Lu
Huang, Jia
Pan, Tao
Su, Xin-zhuan
Wang, Xiaoyun
author_sort Wang, Luoluo
collection PubMed
description N(6) -Methyladenosine (m(6)A), the most abundant mammalian mRNA modification, has been reported to modulate various viral infections. Although it has been confirmed that RNA modifications can also modulate the replication and development of different parasites, the role of the RNA epitranscriptome in the regulation of host response post parasite infection remains to be elucidated. Here we report host spleen m(6)A epitranscriptome landscapes induced by different strains of the malaria parasite Plasmodium yoelii. We found that malaria parasite infection dramatically changes host spleen m(6)A mRNA modification and gene expression. Additionally, malaria parasite infection reprograms host immune response pathways by regulating the m(6)A modification enzymes. Collectively, our study is the first characterization of host spleen m(6)A methylome triggered by malaria parasite infections, and our data identify m(6)A modifications as significant transcriptome-wide marks during host-parasite interactions. We demonstrate that host mRNA methylation machinery can sense and respond to malaria parasite infections, and provide new insights into epitranscriptomic mechanisms underlying parasite-induced pathogenesis.
format Online
Article
Text
id pubmed-9531237
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-95312372022-10-05 Epitranscriptome profiling of spleen mRNA m(6)A methylation reveals pathways of host responses to malaria parasite infection Wang, Luoluo Wu, Jian Liu, Runzhou Chen, Wenjun Pang, Zhichang Zhou, Fan Xia, Lu Huang, Jia Pan, Tao Su, Xin-zhuan Wang, Xiaoyun Front Immunol Immunology N(6) -Methyladenosine (m(6)A), the most abundant mammalian mRNA modification, has been reported to modulate various viral infections. Although it has been confirmed that RNA modifications can also modulate the replication and development of different parasites, the role of the RNA epitranscriptome in the regulation of host response post parasite infection remains to be elucidated. Here we report host spleen m(6)A epitranscriptome landscapes induced by different strains of the malaria parasite Plasmodium yoelii. We found that malaria parasite infection dramatically changes host spleen m(6)A mRNA modification and gene expression. Additionally, malaria parasite infection reprograms host immune response pathways by regulating the m(6)A modification enzymes. Collectively, our study is the first characterization of host spleen m(6)A methylome triggered by malaria parasite infections, and our data identify m(6)A modifications as significant transcriptome-wide marks during host-parasite interactions. We demonstrate that host mRNA methylation machinery can sense and respond to malaria parasite infections, and provide new insights into epitranscriptomic mechanisms underlying parasite-induced pathogenesis. Frontiers Media S.A. 2022-09-20 /pmc/articles/PMC9531237/ /pubmed/36203583 http://dx.doi.org/10.3389/fimmu.2022.998756 Text en Copyright © 2022 Wang, Wu, Liu, Chen, Pang, Zhou, Xia, Huang, Pan, Su and Wang 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
Wang, Luoluo
Wu, Jian
Liu, Runzhou
Chen, Wenjun
Pang, Zhichang
Zhou, Fan
Xia, Lu
Huang, Jia
Pan, Tao
Su, Xin-zhuan
Wang, Xiaoyun
Epitranscriptome profiling of spleen mRNA m(6)A methylation reveals pathways of host responses to malaria parasite infection
title Epitranscriptome profiling of spleen mRNA m(6)A methylation reveals pathways of host responses to malaria parasite infection
title_full Epitranscriptome profiling of spleen mRNA m(6)A methylation reveals pathways of host responses to malaria parasite infection
title_fullStr Epitranscriptome profiling of spleen mRNA m(6)A methylation reveals pathways of host responses to malaria parasite infection
title_full_unstemmed Epitranscriptome profiling of spleen mRNA m(6)A methylation reveals pathways of host responses to malaria parasite infection
title_short Epitranscriptome profiling of spleen mRNA m(6)A methylation reveals pathways of host responses to malaria parasite infection
title_sort epitranscriptome profiling of spleen mrna m(6)a methylation reveals pathways of host responses to malaria parasite infection
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9531237/
https://www.ncbi.nlm.nih.gov/pubmed/36203583
http://dx.doi.org/10.3389/fimmu.2022.998756
work_keys_str_mv AT wangluoluo epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection
AT wujian epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection
AT liurunzhou epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection
AT chenwenjun epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection
AT pangzhichang epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection
AT zhoufan epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection
AT xialu epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection
AT huangjia epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection
AT pantao epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection
AT suxinzhuan epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection
AT wangxiaoyun epitranscriptomeprofilingofspleenmrnam6amethylationrevealspathwaysofhostresponsestomalariaparasiteinfection