SORA

Advancing, promoting and sharing knowledge of health through excellence in teaching, clinical practice and research into the prevention and treatment of illness

Allele-specific control of rodent and human lncRNA KMT2E-AS1 promotes hypoxic endothelial pathology in pulmonary hypertension

Tai, Y-Y; Yu, Q; Tang, Y; Sun, W; Kelly, NJ; Okawa, S; Zhao, J; Schwantes-An, T-H; Lacoux, C; Torrino, S; et al. Tai, Y-Y; Yu, Q; Tang, Y; Sun, W; Kelly, NJ; Okawa, S; Zhao, J; Schwantes-An, T-H; Lacoux, C; Torrino, S; Al Aaraj, Y; El Khoury, W; Negi, V; Liu, M; Corey, CG; Belmonte, F; Vargas, SO; Schwartz, B; Bhat, B; Chau, BN; Karnes, JH; Satoh, T; Barndt, RJ; Wu, H; Parikh, VN; Wang, J; Zhang, Y; McNamara, D; Li, G; Speyer, G; Wang, B; Shiva, S; Kaufman, B; Kim, S; Gomez, D; Mari, B; Cho, MH; Boueiz, A; Pauciulo, MW; Southgate, L; Trembath, RC; Sitbon, O; Humbert, M; Graf, S; Morrell, NW; Rhodes, CJ; Wilkins, MR; Nouraie, M; Nichols, WC; Desai, AA; Bertero, T; Chan, SY (2024) Allele-specific control of rodent and human lncRNA KMT2E-AS1 promotes hypoxic endothelial pathology in pulmonary hypertension. Science Translational Medicine, 16 (729). ISSN 1946-6234 https://doi.org/10.1126/scitranslmed.add2029
SGUL Authors: Southgate, Laura

[img]
Preview
PDF Accepted Version
Available under License ["licenses_description_publisher" not defined].

Download (18MB) | Preview

Abstract

Hypoxic reprogramming of vasculature relies on genetic, epigenetic, and metabolic circuitry, but the control points are unknown. In pulmonary arterial hypertension (PAH), a disease driven by hypoxia inducible factor (HIF)–dependent vascular dysfunction, HIF-2α promoted expression of neighboring genes, long noncoding RNA (lncRNA) histone lysine N-methyltransferase 2E-antisense 1 (KMT2E-AS1) and histone lysine N-methyltransferase 2E (KMT2E). KMT2E-AS1 stabilized KMT2E protein to increase epigenetic histone 3 lysine 4 trimethylation (H3K4me3), driving HIF-2α–dependent metabolic and pathogenic endothelial activity. This lncRNA axis also increased HIF-2α expression across epigenetic, transcriptional, and posttranscriptional contexts, thus promoting a positive feedback loop to further augment HIF-2α activity. We identified a genetic association between rs73184087, a single-nucleotide variant (SNV) within a KMT2E intron, and disease risk in PAH discovery and replication patient cohorts and in a global meta-analysis. This SNV displayed allele (G)–specific association with HIF-2α, engaged in long-range chromatin interactions, and induced the lncRNA-KMT2E tandem in hypoxic (G/G) cells. In vivo, KMT2E-AS1 deficiency protected against PAH in mice, as did pharmacologic inhibition of histone methylation in rats. Conversely, forced lncRNA expression promoted more severe PH. Thus, the KMT2E-AS1/KMT2E pair orchestrates across convergent multi-ome landscapes to mediate HIF-2α pathobiology and represents a key clinical target in pulmonary hypertension.

Item Type: Article
Additional Information: This is the author’s version of the work. It is posted here by permission of the AAAS for personal use, not for redistribution. The definitive version was published in Science Translational Medicine on Vol 16, 10/01/2024, DOI: 10.1126/scitranslmed.add2029.
Keywords: 06 Biological Sciences, 11 Medical and Health Sciences
SGUL Research Institute / Research Centre: Academic Structure > Molecular and Clinical Sciences Research Institute (MCS)
Journal or Publication Title: Science Translational Medicine
ISSN: 1946-6234
Language: en
Dates:
DateEvent
10 January 2024Published
12 December 2023Accepted
Publisher License: Publisher's own licence
Projects:
Project IDFunderFunder ID
R01 HL124021National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
HL 122596National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
R00 HL133473National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
HL105333National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
HL137927National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
HL147148National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
HL089856National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
HL141601National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
HL143185National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
HL129964National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
18EIA33900027American Heart Associationhttp://dx.doi.org/10.13039/100000968
18CN045Plan Cancer 2018UNSPECIFIED
ANR-18-CE14-0025French National Research AgencyUNSPECIFIED
ANR-21-CE44-0036French National Research AgencyUNSPECIFIED
ANR-20-CE14-0006French National Research AgencyUNSPECIFIED
INCA-PLBIO 21-094French National Cancer InstituteUNSPECIFIED
FS/15/59/31839British Heart Foundationhttp://dx.doi.org/10.13039/501100000274
SBF004\1095Academy of Medical Scienceshttp://dx.doi.org/10.13039/501100000691
DBI-1942143NSFUNSPECIFIED
SP/12/12/29836British Heart Foundationhttp://dx.doi.org/10.13039/501100000274
MR/K020919/1Medical Research Councilhttp://dx.doi.org/10.13039/501100000265
S10OD023684National Institutes of Healthhttp://dx.doi.org/10.13039/100000002
URI: https://openaccess.sgul.ac.uk/id/eprint/116001
Publisher's version: https://doi.org/10.1126/scitranslmed.add2029

Actions (login required)

Edit Item Edit Item