Cargando…

Epigenome Editing Durability Varies Widely Across Cardiovascular Disease Target Genes

BACKGROUND: Hepatic knockdown of the proprotein convertase subtilisin/kexin type 9 (PCSK9) gene or the angiopoietin-like 3 (ANGPTL3) gene has been demonstrated to reduce blood low-density lipoprotein cholesterol (LDL-C) levels, and hepatic knockdown of the angiotensinogen (AGT) gene has been demonst...

Descripción completa

Detalles Bibliográficos
Autores principales: Whittaker, Madelynn N., Testa, Lauren C., Quigley, Aidan, Jindal, Ishaan, Cortez-Alvarado, Saúl V., Qu, Ping, Yang, Yifan, Alameh, Mohamad-Gabriel, Musunuru, Kiran, Wang, Xiao
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Cold Spring Harbor Laboratory 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10245770/
https://www.ncbi.nlm.nih.gov/pubmed/37292627
http://dx.doi.org/10.1101/2023.05.17.541156
_version_ 1785054922870358016
author Whittaker, Madelynn N.
Testa, Lauren C.
Quigley, Aidan
Jindal, Ishaan
Cortez-Alvarado, Saúl V.
Qu, Ping
Yang, Yifan
Alameh, Mohamad-Gabriel
Musunuru, Kiran
Wang, Xiao
author_facet Whittaker, Madelynn N.
Testa, Lauren C.
Quigley, Aidan
Jindal, Ishaan
Cortez-Alvarado, Saúl V.
Qu, Ping
Yang, Yifan
Alameh, Mohamad-Gabriel
Musunuru, Kiran
Wang, Xiao
author_sort Whittaker, Madelynn N.
collection PubMed
description BACKGROUND: Hepatic knockdown of the proprotein convertase subtilisin/kexin type 9 (PCSK9) gene or the angiopoietin-like 3 (ANGPTL3) gene has been demonstrated to reduce blood low-density lipoprotein cholesterol (LDL-C) levels, and hepatic knockdown of the angiotensinogen (AGT) gene has been demonstrated to reduce blood pressure. Genome editing can productively target each of these three genes in hepatocytes in the liver, offering the possibility of durable “one-and-done” therapies for hypercholesterolemia and hypertension. However, concerns around making permanent gene sequence changes via DNA strand breaks might hinder acceptance of these therapies. Epigenome editing offers an alternative approach to gene inactivation, via silencing of gene expression by methylation of the promoter region, but the long-term durability of epigenome editing remains to be established. METHODS: We assessed the ability of epigenome editing to durably reduce the expression of the human PCSK9, ANGPTL3, and AGT genes in HuH-7 hepatoma cells. Using the CRISPRoff epigenome editor, we identified guide RNAs that produced efficient gene knockdown immediately after transfection. We assessed the durability of gene expression and methylation changes through serial cell passages. RESULTS: Cells treated with CRISPRoff and PCSK9 guide RNAs were maintained for up to 124 cell doublings and demonstrated durable knockdown of gene expression and increased CpG dinucleotide methylation in the promoter, exon 1, and intron 1 regions. In contrast, cells treated with CRISPRoff and ANGPTL3 guide RNAs experienced only transient knockdown of gene expression. Cells treated with CRISPRoff and AGT guide RNAs also experienced transient knockdown of gene expression; although initially there was increased CpG methylation throughout the early part of the gene, this methylation was geographically heterogeneous—transient in the promoter, and stable in intron 1. CONCLUSIONS: This work demonstrates precise and durable gene regulation via methylation, supporting a new therapeutic approach for protection against cardiovascular disease via knockdown of genes such as PCSK9. However, the durability of knockdown with methylation changes is not generalizable across target genes, likely limiting the therapeutic potential of epigenome editing compared to other modalities.
format Online
Article
Text
id pubmed-10245770
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Cold Spring Harbor Laboratory
record_format MEDLINE/PubMed
spelling pubmed-102457702023-06-08 Epigenome Editing Durability Varies Widely Across Cardiovascular Disease Target Genes Whittaker, Madelynn N. Testa, Lauren C. Quigley, Aidan Jindal, Ishaan Cortez-Alvarado, Saúl V. Qu, Ping Yang, Yifan Alameh, Mohamad-Gabriel Musunuru, Kiran Wang, Xiao bioRxiv Article BACKGROUND: Hepatic knockdown of the proprotein convertase subtilisin/kexin type 9 (PCSK9) gene or the angiopoietin-like 3 (ANGPTL3) gene has been demonstrated to reduce blood low-density lipoprotein cholesterol (LDL-C) levels, and hepatic knockdown of the angiotensinogen (AGT) gene has been demonstrated to reduce blood pressure. Genome editing can productively target each of these three genes in hepatocytes in the liver, offering the possibility of durable “one-and-done” therapies for hypercholesterolemia and hypertension. However, concerns around making permanent gene sequence changes via DNA strand breaks might hinder acceptance of these therapies. Epigenome editing offers an alternative approach to gene inactivation, via silencing of gene expression by methylation of the promoter region, but the long-term durability of epigenome editing remains to be established. METHODS: We assessed the ability of epigenome editing to durably reduce the expression of the human PCSK9, ANGPTL3, and AGT genes in HuH-7 hepatoma cells. Using the CRISPRoff epigenome editor, we identified guide RNAs that produced efficient gene knockdown immediately after transfection. We assessed the durability of gene expression and methylation changes through serial cell passages. RESULTS: Cells treated with CRISPRoff and PCSK9 guide RNAs were maintained for up to 124 cell doublings and demonstrated durable knockdown of gene expression and increased CpG dinucleotide methylation in the promoter, exon 1, and intron 1 regions. In contrast, cells treated with CRISPRoff and ANGPTL3 guide RNAs experienced only transient knockdown of gene expression. Cells treated with CRISPRoff and AGT guide RNAs also experienced transient knockdown of gene expression; although initially there was increased CpG methylation throughout the early part of the gene, this methylation was geographically heterogeneous—transient in the promoter, and stable in intron 1. CONCLUSIONS: This work demonstrates precise and durable gene regulation via methylation, supporting a new therapeutic approach for protection against cardiovascular disease via knockdown of genes such as PCSK9. However, the durability of knockdown with methylation changes is not generalizable across target genes, likely limiting the therapeutic potential of epigenome editing compared to other modalities. Cold Spring Harbor Laboratory 2023-05-17 /pmc/articles/PMC10245770/ /pubmed/37292627 http://dx.doi.org/10.1101/2023.05.17.541156 Text en https://creativecommons.org/licenses/by-nc-nd/4.0/This work is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License (https://creativecommons.org/licenses/by-nc-nd/4.0/) , which allows reusers to copy and distribute the material in any medium or format in unadapted form only, for noncommercial purposes only, and only so long as attribution is given to the creator.
spellingShingle Article
Whittaker, Madelynn N.
Testa, Lauren C.
Quigley, Aidan
Jindal, Ishaan
Cortez-Alvarado, Saúl V.
Qu, Ping
Yang, Yifan
Alameh, Mohamad-Gabriel
Musunuru, Kiran
Wang, Xiao
Epigenome Editing Durability Varies Widely Across Cardiovascular Disease Target Genes
title Epigenome Editing Durability Varies Widely Across Cardiovascular Disease Target Genes
title_full Epigenome Editing Durability Varies Widely Across Cardiovascular Disease Target Genes
title_fullStr Epigenome Editing Durability Varies Widely Across Cardiovascular Disease Target Genes
title_full_unstemmed Epigenome Editing Durability Varies Widely Across Cardiovascular Disease Target Genes
title_short Epigenome Editing Durability Varies Widely Across Cardiovascular Disease Target Genes
title_sort epigenome editing durability varies widely across cardiovascular disease target genes
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10245770/
https://www.ncbi.nlm.nih.gov/pubmed/37292627
http://dx.doi.org/10.1101/2023.05.17.541156
work_keys_str_mv AT whittakermadelynnn epigenomeeditingdurabilityvarieswidelyacrosscardiovasculardiseasetargetgenes
AT testalaurenc epigenomeeditingdurabilityvarieswidelyacrosscardiovasculardiseasetargetgenes
AT quigleyaidan epigenomeeditingdurabilityvarieswidelyacrosscardiovasculardiseasetargetgenes
AT jindalishaan epigenomeeditingdurabilityvarieswidelyacrosscardiovasculardiseasetargetgenes
AT cortezalvaradosaulv epigenomeeditingdurabilityvarieswidelyacrosscardiovasculardiseasetargetgenes
AT quping epigenomeeditingdurabilityvarieswidelyacrosscardiovasculardiseasetargetgenes
AT yangyifan epigenomeeditingdurabilityvarieswidelyacrosscardiovasculardiseasetargetgenes
AT alamehmohamadgabriel epigenomeeditingdurabilityvarieswidelyacrosscardiovasculardiseasetargetgenes
AT musunurukiran epigenomeeditingdurabilityvarieswidelyacrosscardiovasculardiseasetargetgenes
AT wangxiao epigenomeeditingdurabilityvarieswidelyacrosscardiovasculardiseasetargetgenes