Cardiac pathologies in mouse loss of imprinting models are due to misexpression of H19 long noncoding RNA

Maternal loss of imprinting (LOI) at the H19/IGF2 locus results in biallelic IGF2 and reduced H19 expression and is associated with Beckwith–-Wiedemann syndrome (BWS). We use mouse models for LOI to understand the relative importance of Igf2 and H19 mis-expression in BWS phenotypes. Here we focus on...

Full description

Bibliographic Details
Main Authors: Ki-Sun Park, Beenish Rahat, Hyung Chul Lee, Zu-Xi Yu, Jacob Noeker, Apratim Mitra, Connor M Kean, Russell H Knutsen, Danielle Springer, Claudia M Gebert, Beth A Kozel, Karl Pfeifer
Format: Article
Language:English
Published: eLife Sciences Publications Ltd 2021-08-01
Series:eLife
Subjects:
Online Access:https://elifesciences.org/articles/67250
id doaj-9585b4d7825b4eb19926c26cb2be1f3c
record_format Article
spelling doaj-9585b4d7825b4eb19926c26cb2be1f3c2021-09-08T13:41:48ZengeLife Sciences Publications LtdeLife2050-084X2021-08-011010.7554/eLife.67250Cardiac pathologies in mouse loss of imprinting models are due to misexpression of H19 long noncoding RNAKi-Sun Park0https://orcid.org/0000-0003-1322-767XBeenish Rahat1https://orcid.org/0000-0003-0371-2356Hyung Chul Lee2Zu-Xi Yu3Jacob Noeker4https://orcid.org/0000-0002-4344-3114Apratim Mitra5Connor M Kean6Russell H Knutsen7https://orcid.org/0000-0001-6761-6502Danielle Springer8https://orcid.org/0000-0002-6261-9744Claudia M Gebert9https://orcid.org/0000-0002-1282-1602Beth A Kozel10https://orcid.org/0000-0002-9757-7118Karl Pfeifer11https://orcid.org/0000-0002-0254-682XDivision of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United StatesDivision of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United StatesDivision of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United StatesPathology Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, United StatesDivision of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United StatesDivision of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United StatesDivision of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United StatesLaboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, United StatesMurine Phenotyping Core, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, United StatesDivision of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United StatesLaboratory of Vascular and Matrix Genetics, National Heart Lung and Blood Institute, National Institutes of Health, Bethesda, United StatesDivision of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, United StatesMaternal loss of imprinting (LOI) at the H19/IGF2 locus results in biallelic IGF2 and reduced H19 expression and is associated with Beckwith–-Wiedemann syndrome (BWS). We use mouse models for LOI to understand the relative importance of Igf2 and H19 mis-expression in BWS phenotypes. Here we focus on cardiovascular phenotypes and show that neonatal cardiomegaly is exclusively dependent on increased Igf2. Circulating IGF2 binds cardiomyocyte receptors to hyperactivate mTOR signaling, resulting in cellular hyperplasia and hypertrophy. These Igf2-dependent phenotypes are transient: cardiac size returns to normal once Igf2 expression is suppressed postnatally. However, reduced H19 expression is sufficient to cause progressive heart pathologies including fibrosis and reduced ventricular function. In the heart, H19 expression is primarily in endothelial cells (ECs) and regulates EC differentiation both in vivo and in vitro. Finally, we establish novel mouse models to show that cardiac phenotypes depend on H19 lncRNA interactions with Mirlet7 microRNAs.https://elifesciences.org/articles/67250genomic imprintingH19 lncRNAIGF2beckwith wiedemann syndromeepigeneticsheart
collection DOAJ
language English
format Article
sources DOAJ
author Ki-Sun Park
Beenish Rahat
Hyung Chul Lee
Zu-Xi Yu
Jacob Noeker
Apratim Mitra
Connor M Kean
Russell H Knutsen
Danielle Springer
Claudia M Gebert
Beth A Kozel
Karl Pfeifer
spellingShingle Ki-Sun Park
Beenish Rahat
Hyung Chul Lee
Zu-Xi Yu
Jacob Noeker
Apratim Mitra
Connor M Kean
Russell H Knutsen
Danielle Springer
Claudia M Gebert
Beth A Kozel
Karl Pfeifer
Cardiac pathologies in mouse loss of imprinting models are due to misexpression of H19 long noncoding RNA
eLife
genomic imprinting
H19 lncRNA
IGF2
beckwith wiedemann syndrome
epigenetics
heart
author_facet Ki-Sun Park
Beenish Rahat
Hyung Chul Lee
Zu-Xi Yu
Jacob Noeker
Apratim Mitra
Connor M Kean
Russell H Knutsen
Danielle Springer
Claudia M Gebert
Beth A Kozel
Karl Pfeifer
author_sort Ki-Sun Park
title Cardiac pathologies in mouse loss of imprinting models are due to misexpression of H19 long noncoding RNA
title_short Cardiac pathologies in mouse loss of imprinting models are due to misexpression of H19 long noncoding RNA
title_full Cardiac pathologies in mouse loss of imprinting models are due to misexpression of H19 long noncoding RNA
title_fullStr Cardiac pathologies in mouse loss of imprinting models are due to misexpression of H19 long noncoding RNA
title_full_unstemmed Cardiac pathologies in mouse loss of imprinting models are due to misexpression of H19 long noncoding RNA
title_sort cardiac pathologies in mouse loss of imprinting models are due to misexpression of h19 long noncoding rna
publisher eLife Sciences Publications Ltd
series eLife
issn 2050-084X
publishDate 2021-08-01
description Maternal loss of imprinting (LOI) at the H19/IGF2 locus results in biallelic IGF2 and reduced H19 expression and is associated with Beckwith–-Wiedemann syndrome (BWS). We use mouse models for LOI to understand the relative importance of Igf2 and H19 mis-expression in BWS phenotypes. Here we focus on cardiovascular phenotypes and show that neonatal cardiomegaly is exclusively dependent on increased Igf2. Circulating IGF2 binds cardiomyocyte receptors to hyperactivate mTOR signaling, resulting in cellular hyperplasia and hypertrophy. These Igf2-dependent phenotypes are transient: cardiac size returns to normal once Igf2 expression is suppressed postnatally. However, reduced H19 expression is sufficient to cause progressive heart pathologies including fibrosis and reduced ventricular function. In the heart, H19 expression is primarily in endothelial cells (ECs) and regulates EC differentiation both in vivo and in vitro. Finally, we establish novel mouse models to show that cardiac phenotypes depend on H19 lncRNA interactions with Mirlet7 microRNAs.
topic genomic imprinting
H19 lncRNA
IGF2
beckwith wiedemann syndrome
epigenetics
heart
url https://elifesciences.org/articles/67250
work_keys_str_mv AT kisunpark cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT beenishrahat cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT hyungchullee cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT zuxiyu cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT jacobnoeker cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT apratimmitra cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT connormkean cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT russellhknutsen cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT daniellespringer cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT claudiamgebert cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT bethakozel cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
AT karlpfeifer cardiacpathologiesinmouselossofimprintingmodelsareduetomisexpressionofh19longnoncodingrna
_version_ 1717762390599139328