Deletion of Tet proteins results in quantitative disparities during ESC differentiation partially attributable to alterations in gene expression

Abstract Background The Tet protein family (Tet1, Tet2, and Tet3) regulate DNA methylation through conversion of 5-methylcytosine to 5-hydroxymethylcytosine which can ultimately result in DNA demethylation and play a critical role during early mammalian development and pluripotency. While multiple g...

Full description

Bibliographic Details
Main Authors: Michael Reimer, Kirthi Pulakanti, Linzheng Shi, Alex Abel, Mingyu Liang, Subramaniam Malarkannan, Sridhar Rao
Format: Article
Language:English
Published: BMC 2019-07-01
Series:BMC Developmental Biology
Subjects:
Online Access:http://link.springer.com/article/10.1186/s12861-019-0196-6
id doaj-e0b977ce8e7d486481c96765d6040621
record_format Article
spelling doaj-e0b977ce8e7d486481c96765d60406212020-11-25T03:32:34ZengBMCBMC Developmental Biology1471-213X2019-07-0119111310.1186/s12861-019-0196-6Deletion of Tet proteins results in quantitative disparities during ESC differentiation partially attributable to alterations in gene expressionMichael Reimer0Kirthi Pulakanti1Linzheng Shi2Alex Abel3Mingyu Liang4Subramaniam Malarkannan5Sridhar Rao6Blood Research Institute, VersitiBlood Research Institute, VersitiBlood Research Institute, VersitiBlood Research Institute, VersitiDepartment of Physiology, Center of Systems Molecular Medicine, Medical College of WisconsinBlood Research Institute, VersitiBlood Research Institute, VersitiAbstract Background The Tet protein family (Tet1, Tet2, and Tet3) regulate DNA methylation through conversion of 5-methylcytosine to 5-hydroxymethylcytosine which can ultimately result in DNA demethylation and play a critical role during early mammalian development and pluripotency. While multiple groups have generated knockouts combining loss of different Tet proteins in murine embryonic stem cells (ESCs), differences in genetic background and approaches has made it difficult to directly compare results and discern the direct mechanism by which Tet proteins regulate the transcriptome. To address this concern, we utilized genomic editing in an isogenic pluripotent background which permitted a quantitative, flow-cytometry based measurement of pluripotency in combination with genome-wide assessment of gene expression and DNA methylation changes. Our ultimate goal was to generate a resource of large-scale datasets to permit hypothesis-generating experiments. Results We demonstrate a quantitative disparity in the differentiation ability among Tet protein deletions, with Tet2 single knockout exhibiting the most severe defect, while loss of Tet1 alone or combinations of Tet genes showed a quantitatively intermediate phenotype. Using a combination of transcriptomic and epigenomic approaches we demonstrate an increase in DNA hypermethylation and a divergence of transcriptional profiles in pluripotency among Tet deletions, with loss of Tet2 having the most profound effect in undifferentiated ESCs. Conclusions We conclude that loss of Tet2 has the most dramatic effect both on the phenotype of ESCs and the transcriptome compared to other genotypes. While loss of Tet proteins increased DNA hypermethylation, especially in gene promoters, these changes in DNA methylation did not correlate with gene expression changes. Thus, while loss of different Tet proteins alters DNA methylation, this change does not appear to be directly responsible for transcriptome changes. Thus, loss of Tet proteins likely regulates the transcriptome epigenetically both through altering 5mC but also through additional mechanisms. Nonetheless, the transcriptome changes in pluripotent Tet2 −/− ESCs compared to wild-type implies that the disparities in differentiation can be partially attributed to baseline alterations in gene expression.http://link.springer.com/article/10.1186/s12861-019-0196-6Embryonic stem cellsDNA methylationTen eleven translocation (TET proteins)Differentiation
collection DOAJ
language English
format Article
sources DOAJ
author Michael Reimer
Kirthi Pulakanti
Linzheng Shi
Alex Abel
Mingyu Liang
Subramaniam Malarkannan
Sridhar Rao
spellingShingle Michael Reimer
Kirthi Pulakanti
Linzheng Shi
Alex Abel
Mingyu Liang
Subramaniam Malarkannan
Sridhar Rao
Deletion of Tet proteins results in quantitative disparities during ESC differentiation partially attributable to alterations in gene expression
BMC Developmental Biology
Embryonic stem cells
DNA methylation
Ten eleven translocation (TET proteins)
Differentiation
author_facet Michael Reimer
Kirthi Pulakanti
Linzheng Shi
Alex Abel
Mingyu Liang
Subramaniam Malarkannan
Sridhar Rao
author_sort Michael Reimer
title Deletion of Tet proteins results in quantitative disparities during ESC differentiation partially attributable to alterations in gene expression
title_short Deletion of Tet proteins results in quantitative disparities during ESC differentiation partially attributable to alterations in gene expression
title_full Deletion of Tet proteins results in quantitative disparities during ESC differentiation partially attributable to alterations in gene expression
title_fullStr Deletion of Tet proteins results in quantitative disparities during ESC differentiation partially attributable to alterations in gene expression
title_full_unstemmed Deletion of Tet proteins results in quantitative disparities during ESC differentiation partially attributable to alterations in gene expression
title_sort deletion of tet proteins results in quantitative disparities during esc differentiation partially attributable to alterations in gene expression
publisher BMC
series BMC Developmental Biology
issn 1471-213X
publishDate 2019-07-01
description Abstract Background The Tet protein family (Tet1, Tet2, and Tet3) regulate DNA methylation through conversion of 5-methylcytosine to 5-hydroxymethylcytosine which can ultimately result in DNA demethylation and play a critical role during early mammalian development and pluripotency. While multiple groups have generated knockouts combining loss of different Tet proteins in murine embryonic stem cells (ESCs), differences in genetic background and approaches has made it difficult to directly compare results and discern the direct mechanism by which Tet proteins regulate the transcriptome. To address this concern, we utilized genomic editing in an isogenic pluripotent background which permitted a quantitative, flow-cytometry based measurement of pluripotency in combination with genome-wide assessment of gene expression and DNA methylation changes. Our ultimate goal was to generate a resource of large-scale datasets to permit hypothesis-generating experiments. Results We demonstrate a quantitative disparity in the differentiation ability among Tet protein deletions, with Tet2 single knockout exhibiting the most severe defect, while loss of Tet1 alone or combinations of Tet genes showed a quantitatively intermediate phenotype. Using a combination of transcriptomic and epigenomic approaches we demonstrate an increase in DNA hypermethylation and a divergence of transcriptional profiles in pluripotency among Tet deletions, with loss of Tet2 having the most profound effect in undifferentiated ESCs. Conclusions We conclude that loss of Tet2 has the most dramatic effect both on the phenotype of ESCs and the transcriptome compared to other genotypes. While loss of Tet proteins increased DNA hypermethylation, especially in gene promoters, these changes in DNA methylation did not correlate with gene expression changes. Thus, while loss of different Tet proteins alters DNA methylation, this change does not appear to be directly responsible for transcriptome changes. Thus, loss of Tet proteins likely regulates the transcriptome epigenetically both through altering 5mC but also through additional mechanisms. Nonetheless, the transcriptome changes in pluripotent Tet2 −/− ESCs compared to wild-type implies that the disparities in differentiation can be partially attributed to baseline alterations in gene expression.
topic Embryonic stem cells
DNA methylation
Ten eleven translocation (TET proteins)
Differentiation
url http://link.springer.com/article/10.1186/s12861-019-0196-6
work_keys_str_mv AT michaelreimer deletionoftetproteinsresultsinquantitativedisparitiesduringescdifferentiationpartiallyattributabletoalterationsingeneexpression
AT kirthipulakanti deletionoftetproteinsresultsinquantitativedisparitiesduringescdifferentiationpartiallyattributabletoalterationsingeneexpression
AT linzhengshi deletionoftetproteinsresultsinquantitativedisparitiesduringescdifferentiationpartiallyattributabletoalterationsingeneexpression
AT alexabel deletionoftetproteinsresultsinquantitativedisparitiesduringescdifferentiationpartiallyattributabletoalterationsingeneexpression
AT mingyuliang deletionoftetproteinsresultsinquantitativedisparitiesduringescdifferentiationpartiallyattributabletoalterationsingeneexpression
AT subramaniammalarkannan deletionoftetproteinsresultsinquantitativedisparitiesduringescdifferentiationpartiallyattributabletoalterationsingeneexpression
AT sridharrao deletionoftetproteinsresultsinquantitativedisparitiesduringescdifferentiationpartiallyattributabletoalterationsingeneexpression
_version_ 1724567469903839232