An Important Role for DNMT3A-Mediated DNA Methylation in Cardiomyocyte Metabolism and Contractility

Alexandra Madsen, Grit Höppner, Julia Krause, Marc N Hirt, Sandra D Laufer, Michaela Schweizer, Wilson Lek Wen Tan, Diogo Mosqueira, Chukwuemeka George Anene-Nzelu, Ives Lim, Roger S Y Foo, Arne Hansen, Thomas Eschenhagen, Justus Stenzig, Alexandra Madsen, Grit Höppner, Julia Krause, Marc N Hirt, Sandra D Laufer, Michaela Schweizer, Wilson Lek Wen Tan, Diogo Mosqueira, Chukwuemeka George Anene-Nzelu, Ives Lim, Roger S Y Foo, Arne Hansen, Thomas Eschenhagen, Justus Stenzig

Abstract

Background: DNA methylation acts as a mechanism of gene transcription regulation. It has recently gained attention as a possible therapeutic target in cardiac hypertrophy and heart failure. However, its exact role in cardiomyocytes remains controversial. Thus, we knocked out the main de novo DNA methyltransferase in cardiomyocytes, DNMT3A, in human induced pluripotent stem cells. Functional consequences of DNA methylation-deficiency under control and stress conditions were then assessed in human engineered heart tissue from knockout human induced pluripotent stem cell-derived cardiomyocytes.

Methods: DNMT3A was knocked out in human induced pluripotent stem cells by CRISPR/Cas9gene editing. Fibrin-based engineered heart tissue was generated from knockout and control human induced pluripotent stem cell-derived cardiomyocytes. Development and baseline contractility were analyzed by video-optical recording. Engineered heart tissue was subjected to different stress protocols, including serum starvation, serum variation, and restrictive feeding. Molecular, histological, and ultrastructural analyses were performed afterward.

Results: Knockout of DNMT3A in human cardiomyocytes had three main consequences for cardiomyocyte morphology and function: (1) Gene expression changes of contractile proteins such as higher atrial gene expression and lower MYH7/MYH6 ratio correlated with different contraction kinetics in knockout versus wild-type; (2) Aberrant activation of the glucose/lipid metabolism regulator peroxisome proliferator-activated receptor gamma was associated with accumulation of lipid vacuoles within knockout cardiomyocytes; (3) Hypoxia-inducible factor 1α protein instability was associated with impaired glucose metabolism and lower glycolytic enzyme expression, rendering knockout-engineered heart tissue sensitive to metabolic stress such as serum withdrawal and restrictive feeding.

Conclusion: The results suggest an important role of DNA methylation in the normal homeostasis of cardiomyocytes and during cardiac stress, which could make it an interesting target for cardiac therapy.

Trial registration: ClinicalTrials.gov NCT02417311.

Keywords: cardiac hypertrophy; epigenetics; tissue engineering.

Conflict of interest statement

Drs Eschenhagen and Hirt are cofounders of EHT Technologies GmbH, Hamburg. The other authors report no conflicts.

Figures

Figure 1.
Figure 1.
Contraction kinetics of DNA methyltransferase(DNMT) 3A knockoutengineered heart tissue (EHT). A, Normalized average contraction peaks of EHT from wild-type and knockout lines at a stimulation frequency of 1.5 Hz. Mean±SEM. B, Relaxation times from peak to 80% relaxation at 1.5 Hz. n=7-12 EHTs per group. C, Ratio of myosin heavy chain (MHC) isoforms in wild-type and DNMT3A−/− EHTs. MYH7 and MYH6 abundance was measured by NanoString analysis, n=3. D, Ratio of atrial (MYL7, MLC2A) and ventricular myosin light chain (MYL2, MLC2V) isoforms, measured by RNA sequencing. n=3. E, Relative expression of atrial-specific marker genes. n=3, False discovery rate- (FDR-) adjusted P value. F, PITX2 promoter methylation, measured by reduced representation bisulfite sequencing. n=3–4. G, Pacing capture of wild-type and knockout EHT at frequencies from 0.7 Hz to 6 Hz, beating frequency of each EHT line plotted against the stimulation frequency. Each dot represents a single EHT measurement. ***P<0.001.
Figure 2.
Figure 2.
Contraction behavior in serum-free culture conditions. Force development (A) and RR scatter (B) as a surrogate parameter for beating arrhythmias, with R being the interval between 2 neighboring force peaks, of wild-type and (C–D) knockout engineered heart tissue (EHT) over culture time under both standard serum-containing (green plus) and serum-free culture conditions (red minus). The dotted line marks beginning of serum withdrawal. Mean±SEM, n=51–58 EHTs from 3 batches. Representative contraction peaks of wild-type (E) and knockout EHTs (F) under serum-free conditions.
Figure 3.
Figure 3.
Gene expression changes in homozygous DNA methyltransferase(DNMT) 3A knockout engineered heart tissue(EHT). Transcript abundance of (A) low-expression genes and (B) high-expression genes in DNMT3A−/− EHTs. All values normalized to wild-type (dashed line); n=3 per group, **P<0.001, ***P<0.001. C, Pathway mapping of significantly differentially expressed genes in DNMT3A−/− vs wild-type. Dotted line represents significance threshold of P<0.05. Red, cardiac-related; blue, signaling-related; yellow, metabolism-related pathways.
Figure 4.
Figure 4.
Morphology of DNA methyltransferase(DNMT) 3A knockout engineered heart tissue(EHT). Histological analysis of (A) wild-type and (B) DNMT3A−/− EHTs. Left, Hematoxylin and eosin staining (H&E); scale bar, 50 µm. Middle, MLC2V staining; scale bar, 50 µm. Right, Lipid staining with Oil Red O; scale bar, 500 µm, red dots mark lipid accumulations. Transmission electron microscopy of wild-type (C) and knockout (D) EHTs showing intact sarcomeric structures but degenerated mitochondria in knockout EHT. Yellow arrowheads, sarcomeres; black arrowheads, mitochondria. Scale bar, 500 nm.
Figure 5.
Figure 5.
Upregulation ofperoxisome proliferator-activated receptor gamma (PPARγ) is responsible for lipid accumulation inengineered heart tissue (EHT). A, Normalized PPARγ transcripts in wild-type and DNA methyltransferase (DNMT) 3A−/− EHTs. n=3 EHTs per group, ***P<0.001. B, PPARγ promoter methylation in wild-type and DNMT3A−/− EHTs measured by bisulfite sequencing. Black dot, methylated cytosine; white dot, unmethylated cytosine. C, Hematoxylin and eosin staining (H&E) staining of vehicle (left) and PPARγ-inhibitor GW9662–treated DNMT3A−/− EHTs (right); scale bar, 50 µm. D, H&E staining of DNMT3A−/− EHTs cultured for 4 weeks in serum-containing conditions; scale bar, 50 µm.
Figure 6.
Figure 6.
Effect of DNA methyltransferase(DNMT) 3A knockout on engineered heart tissue(EHT) glucose metabolism. Effect of restrictive feeding with lactate or glucose-only medium on frequency (A), force (B), and RR scatter, with R being the interval between 2 neighboring force peaks, (C) of wild-type and knockout EHTs. n=8, *P<0.05, ***P<0.001. D, Relative transcript abundance of glycolytic genes in DNMT3A−/− #2 EHTs after 1 week of serum-free culture. n=3, values normalized to wild-type (dashed line). E and F, Quantification of hypoxia-inducible factor 1α (HIF-1α) protein by Western blot of DNMT3A−/− #1 and isogenic control EHTs cultured either with lipoprotein-deficient serum (LDS), control serum, or dialyzed serum. Loading control = ERK1/2; n=2 to 3 EHTs per group.
Figure 7.
Figure 7.
Effect of growth factor treatment of engineered heart tissue(EHT) under serum-free conditions. A, Effect of growth factor treatment (GFs; 20 ng/mL transforming growth factor [TGF]-β1, 10 ng/mL basic fibroblast growth factor [bFGF], 20 ng/mL insulin-like growth factor 1 [IGF-1], 10 ng/mL endothelial growth factor [EGF]) on force under serum-free conditions. n=10–15 EHTs from 2 batches per group. B and C, Quantification of hypoxia-inducible factor 1α (HIF-1α) protein by Western blot of wild-type and DNA methyltransferase (DNMT) 3A−/− EHTs cultured either with serum-free medium or with serum-free medium containing GFs. Loading control = ERK1/2; n=3 EHTs per group. D, Relative transcript abundance of glycolytic genes in DNMT3A−/− EHTs after 1 week of serum-free culture ± GFs. n=3, values normalized to wild-type (dashed line). Effect of GF treatment on glucose consumption (E) and lactate generation (F) of wild-type and DNMT3A−/− EHTs. n=4, values normalized to wild-type (dashed line). ns indicates nonsignificant, *P<0.05, **P<0.01, ***P<0.001.
Figure 8.
Figure 8.
Overview key experiments and findings. Dashed boxes indicate key experimental findings; colored boxes indicate conclusions drawn from experimental findings.

References

    1. Ponikowski P, Anker SD, Alhabib KF, Cowie MR, Force TL, Hu S, Joorsma T, Krum H, Rastogi V, Rohde LE, et al. . Heart failure: Preventing disease and death worldwide. ESC Heart Fail. 2014; 373:941–955. doi: 10.1002/ehf2.12005
    1. Takahashi T, Allen PD, Izumo S. Expression of A-, B-, and C-type natriuretic peptide genes in failing and developing human ventricles: correlation with expression of the Ca(2+)-ATPase gene. Circ Res. 1992; 71:9–17. doi: 10.1161/01.res.71.1.9
    1. Arai M, Alpert NR, MacLennan DH, Barton P, Periasamy M. Alterations in sarcoplasmic reticulum gene expression in human heart failure: a possible mechanism for alterations in systolic and diastolic properties of the failing myocardium. Circ Res. 1993; 72:463–469. doi: 10.1161/01.res.72.2.463
    1. Locher MR, Razumova MV, Stelzer JE, Norman HS, Moss RL. Effects of low-level & α-myosin heavy chain expression on contractile kinetics in porcine myocardium. Am J Physiol Heart Circ Physiol. 2011; 300:H869–H878. doi: 10.1152/ajpheart.00452.2010
    1. Meissner A, Mikkelsen TS, Gu H, Wernig M, Hanna J, Sivachenko A, Zhang X, Bernstein BE, Nusbaum C, Jaffe DB, et al. . Genome-scale DNA methylation maps of pluripotent and differentiated cells. Nature. 2008; 454:766–770. doi: 10.1038/nature07107
    1. Bashtrykov P, Jankevicius G, Jurkowska RZ, Ragozin S, Jeltsch A. The UHRF1 protein stimulates the activity and specificity of the maintenance DNA methyltransferase DNMT1 by an allosteric mechanism. J Biol Chem. 2014; 289:4106–4115. doi: 10.1074/jbc.M113.528893
    1. Okano M, Xie S, Li E. Cloning and characterization of a family of novel mammalian DNA (cytosine-5) methyltransferases. Nat Genet. 1998; 19:219–220. doi: 10.1038/890
    1. Klose RJ, Bird AP. Genomic DNA methylation: the mark and its mediators. Trends Biochem Sci. 2006; 31:89–97. doi: 10.1016/j.tibs.2005.12.008
    1. Zhang CL, McKinsey TA, Chang S, Antos CL, Hill JA, Olson EN. Class II histone deacetylases act as signal-responsive repressors of cardiac hypertrophy. Cell. 2002; 110:479–488. doi: 10.1016/s0092-8674(02)00861-9
    1. Trivedi CM, Luo Y, Yin Z, Zhang M, Zhu W, Wang T, Floss T, Goettlicher M, Noppinger PR, Wurst W, et al. . Hdac2 regulates the cardiac hypertrophic response by modulating Gsk3 beta activity. Nat Med. 2007; 13:324–331. doi: 10.1038/nm1552
    1. Wei JQ, Shehadeh LA, Mitrani JM, Pessanha M, Slepak TI, Webster KA, Bishopric NH. Quantitative control of adaptive cardiac hypertrophy by acetyltransferase p300. Circulation. 2008; 118:934–946. doi: 10.1161/CIRCULATIONAHA.107.760488
    1. Zhang QJ, Chen HZ, Wang L, Liu DP, Hill JA, Liu ZP. The histone trimethyllysine demethylase JMJD2A promotes cardiac hypertrophy in response to hypertrophic stimuli in mice. J Clin Invest. 2011; 121:2447–2456. doi: 10.1172/JCI46277
    1. Kronlage M, Dewenter M, Grosso J, Fleming T, Oehl U, Lehmann LH, Falcão-Pires I, Leite-Moreira AF, Volk N, Gröne HJ, et al. . O-GlcNacylation of histone deacetylase 4 protects the diabetic heart from failure. Circulation. 2019; 140:580–594. doi: 10.1161/CIRCULATIONAHA.117.031942
    1. Movassagh M, Choy MK, Knowles DA, Cordeddu L, Haider S, Down T, Siggens L, Vujic A, Simeoni I, Penkett C, et al. . Distinct epigenomic features in end-stage failing human hearts. Circulation. 2011; 124:2411–2422. doi: 10.1161/CIRCULATIONAHA.111.040071
    1. Haas J, Frese KS, Park YJ, Keller A, Vogel B, Lindroth AM, Weichenhan D, Franke J, Fischer S, Bauer A, et al. . Alterations in cardiac DNA methylation in human dilated cardiomyopathy. EMBO Mol Med. 2013; 5:413–429. doi: 10.1002/emmm.201201553
    1. Gilsbach R, Preissl S, Grüning BA, Schnick T, Burger L, Benes V, Würch A, Bönisch U, Günther S, Backofen R, et al. . Dynamic DNA methylation orchestrates cardiomyocyte development, maturation and disease. Nat Commun. 2014; 5:5288. doi: 10.1038/ncomms6288
    1. Xiao D, Dasgupta C, Chen M, Zhang K, Buchholz J, Xu Z, Zhang L. Inhibition of DNA methylation reverses norepinephrine-induced cardiac hypertrophy in rats. Cardiovasc Res. 2014; 101:373–382. doi: 10.1093/cvr/cvt264
    1. Watson CJ, Horgan S, Neary R, Glezeva N, Tea I, Corrigan N, McDonald K, Ledwidge M, Baugh J. Epigenetic therapy for the treatment of hypertension-induced cardiac hypertrophy and fibrosis. J Cardiovasc Pharmacol Ther. 2016; 21:127–137. doi: 10.1177/1074248415591698
    1. Stenzig J, Hirt MN, Löser A, Bartholdt LM, Hensel JT, Werner TR, Riemenschneider M, Indenbirken D, Guenther T, Müller C, et al. . DNA methylation in an engineered heart tissue model of cardiac hypertrophy: common signatures and effects of DNA methylation inhibitors. Basic Res Cardiol. 2016; 111:9. doi: 10.1007/s00395-015-0528-z
    1. Stenzig J, Schneeberger Y, Löser A, Peters BS, Schaefer A, Zhao RR, Ng SL, Höppner G, Geertz B, Hirt MN, et al. . Pharmacological inhibition of DNA methylation attenuates pressure overload-induced cardiac hypertrophy in rats. J Mol Cell Cardiol. 2018; 120:53–63. doi: 10.1016/j.yjmcc.2018.05.012
    1. Vujic A, Robinson EL, Ito M, Haider S, Ackers-Johnson M, See K, Methner C, Figg N, Brien P, Roderick HL, et al. . Experimental heart failure modelled by the cardiomyocyte-specific loss of an epigenome modifier, DNMT3B. J Mol Cell Cardiol. 2015; 82:174–183. doi: 10.1016/j.yjmcc.2015.03.007
    1. Nührenberg TG, Hammann N, Schnick T, Preißl S, Witten A, Stoll M, Gilsbach R, Neumann FJ, Hein L. Cardiac myocyte de novo DNA methyltransferases 3a/3b are dispensable for cardiac function and remodeling after chronic pressure overload in mice. PLoS One. 2015; 10:e0131019. doi: 10.1371/journal.pone.0131019
    1. Mannhardt I, Breckwoldt K, Letuffe-Brenière D, Schaaf S, Schulz H, Neuber C, Benzin A, Werner T, Eder A, Schulze T, et al. . Human engineered heart tissue: analysis of contractile force. Stem Cell Reports. 2016; 7:29–42. doi: 10.1016/j.stemcr.2016.04.011
    1. Breckwoldt K, Letuffe-Brenière D, Mannhardt I, Schulze T, Ulmer B, Werner T, Benzin A, Klampe B, Reinsch MC, Laufer S, et al. . Differentiation of cardiomyocytes and generation of human engineered heart tissue. Nat Protoc. 2017; 12:1177–1197. doi: 10.1038/nprot.2017.033
    1. Hirt MN, Sörensen NA, Bartholdt LM, Boeddinghaus J, Schaaf S, Eder A, Vollert I, Stöhr A, Schulze T, Witten A, et al. . Increased afterload induces pathological cardiac hypertrophy: a new in vitro model. Basic Res Cardiol. 2012; 107:307. doi: 10.1007/s00395-012-0307-z
    1. Eder A, Hansen A, Uebeler J, Schulze T, Neuber C, Schaaf S, Yuan L, Christ T, Vos MA, Eschenhagen T. Effects of proarrhythmic drugs on relaxation time and beating pattern in rat engineered heart tissue. Basic Res Cardiol. 2014; 109:436. doi: 10.1007/s00395-014-0436-7
    1. Eder A, Vollert I, Hansen A, Eschenhagen T. Human engineered heart tissue as a model system for drug testing. Adv Drug Deliv Rev. 2016; 96:214–224. doi: 10.1016/j.addr.2015.05.010
    1. Ran FA, Hsu PD, Wright J, Agarwala V, Scott DA, Zhang F. Genome engineering using the CRISPR-Cas9 system. Nat Protoc. 2013; 8:2281–2308. doi: 10.1038/nprot.2013.143
    1. Liao J, Karnik R, Gu H, Ziller MJ, Clement K, Tsankov AM, Akopian V, Gifford CA, Donaghey J, Galonska C, et al. . Targeted disruption of DNMT1, DNMT3A and DNMT3B in human embryonic stem cells. Nat Genet. 2015; 47:469–478. doi: 10.1038/ng.3258
    1. Hansen A, Eder A, Bönstrup M, Flato M, Mewe M, Schaaf S, Aksehirlioglu B, Schwoerer AP, Schwörer A, Uebeler J, et al. . Development of a drug screening platform based on engineered heart tissue. Circ Res. 2010; 107:35–44. doi: 10.1161/CIRCRESAHA.109.211458
    1. Hirt MN, Boeddinghaus J, Mitchell A, Schaaf S, Börnchen C, Müller C, Schulz H, Hubner N, Stenzig J, Stoehr A, et al. . Functional improvement and maturation of rat and human engineered heart tissue by chronic electrical stimulation. J Mol Cell Cardiol. 2014; 74:151–161. doi: 10.1016/j.yjmcc.2014.05.009
    1. Goldstein JL, Basu SK, Brown MS. Receptor-mediated endocytosis of low-density lipoprotein in cultured cells. Methods Enzymol. 1983; 98:241–260. doi: 10.1016/0076-6879(83)98152-1
    1. Schlegel A, Stainier DY. Microsomal triglyceride transfer protein is required for yolk lipid utilization and absorption of dietary lipids in zebrafish larvae. Biochemistry. 2006; 45:15179–15187. doi: 10.1021/bi0619268
    1. Fraher D, Ellis MK, Morrison S, McGee SL, Ward AC, Walder K, Gibert Y. Lipid abundance in zebrafish embryos is regulated by complementary actions of the endocannabinoid system and retinoic acid pathway. Endocrinology. 2015; 156:3596–3609. doi: 10.1210/EN.2015-1315
    1. Mosqueira D, Lis-Slimak K, Denning C. High-throughput phenotyping toolkit for characterizing cellular models of hypertrophic cardiomyopathy in vitro. Methods Protoc. 2019; 2:83. doi: 10.3390/mps2040083
    1. Prondzynski M, Krämer E, Laufer SD, Shibamiya A, Pless O, Flenner F, Müller OJ, Münch J, Redwood C, Hansen A, et al. . Evaluation of MYBPC3 trans-splicing and gene replacement as therapeutic options in human iPSC-derived cardiomyocytes. Mol Ther Nucleic Acids. 2017; 7:475–486. doi: 10.1016/j.omtn.2017.05.008
    1. Huang da W, Sherman BT, Lempicki RA. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat Protoc. 2009; 4:44–57. doi: 10.1038/nprot.2008.211
    1. Huang da W, Sherman BT, Lempicki RA. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 2009; 37:1–13. doi: 10.1093/nar/gkn923
    1. Kosicki M, Tomberg K, Bradley A. Repair of double-strand breaks induced by CRISPR-Cas9 leads to large deletions and complex rearrangements. Nat Biotechnol. 2018; 36:765–771. doi: 10.1038/nbt.4192
    1. Närvä E, Autio R, Rahkonen N, Kong L, Harrison N, Kitsberg D, Borghese L, Itskovitz-Eldor J, Rasool O, Dvorak P, et al. . High-resolution DNA analysis of human embryonic stem cell lines reveals culture-induced copy number changes and loss of heterozygosity. Nat Biotechnol. 2010; 28:371–377. doi: 10.1038/nbt.1615
    1. Taapken SM, Nisler BS, Newton MA, Sampsell-Barron TL, Leonhard KA, McIntire EM, Montgomery KD. Karotypic abnormalities in human induced pluripotent stem cells and embryonic stem cells. Nat Biotechnol. 2011; 29:313–314. doi: 10.1038/nbt.1835
    1. Han P, Li W, Yang J, Shang C, Lin C-H, Cheng W, Hang CT, Cheng H-L, Chen C-H, Wong J, et al. . Epigenetic response to environmental stress: assembly of BRG1–G9a/GLP–DNMT3 repressive chromatin complex on Myh6 promoter in pathologically stressed hearts. Biochim Biophys Acta. 20161–10. doi: 10.1016/j.bbamcr.2016.03.002
    1. Hnisz D, Abraham BJ, Lee TI, Lau A, Saint-André V, Sigova AA, Hoke HA, Young RA. Super-enhancers in the control of cell identity and disease. Cell. 2013; 155:934–947. doi: 10.1016/j.cell.2013.09.053
    1. Jermann P, Hoerner L, Burger L, Schübeler D. Short sequences can efficiently recruit histone H3 lysine 27 trimethylation in the absence of enhancer activity and DNA methylation. Proc Natl Acad Sci U S A. 2014; 111:E3415–E3421. doi: 10.1073/pnas.1400672111
    1. Greenberg MVC, Bourc’his D. The diverse roles of DNA methylation in mammalian development and disease. Nat Rev Mol Cell Biol. 2019; 20:590–607. doi: 10.1038/s41580-019-0159-6
    1. He A, Shen X, Ma Q, Cao J, von Gise A, Zhou P, Wang G, Marquez VE, Orkin SH, Pu WT. PRC2 directly methylates GATA4 and represses its transcriptional activity. Genes Dev. 2012; 26:37–42. doi: 10.1101/gad.173930.111
    1. Tessari A, Pietrobon M, Notte A, Cifelli G, Gage PJ, Schneider MD, Lembo G, Campione M. Myocardial Pitx2 differentially regulates the left atrial identity and ventricular asymmetric remodeling programs. Circ Res. 2008; 102:813–822. doi: 10.1161/CIRCRESAHA.107.163188
    1. Lemme M, Ulmer BM, Lemoine MD, Zech ATL, Flenner F, Ravens U, Reichenspurner H, Rol-Garcia M, Smith G, Hansen A, et al. . Atrial-like engineered heart tissue: an in vitro model of the human atrium. Stem Cell Reports. 2018; 11:1378–1390. doi: 10.1016/j.stemcr.2018.10.008
    1. Krause J, Löser A, Lemoine MD, Christ T, Scherschel K, Meyer C, Blankenberg S, Zeller T, Eschenhagen T, Stenzig J. Rat atrial engineered heart tissue: a new in vitro model to study atrial biology. Basic Res Cardiol. 2018; 113:41. doi: 10.1007/s00395-018-0701-2
    1. Vinarskaja A, Schulz WA, Ingenwerth M, Hader C, Arsov C. Association of PITX2 mRNA down-regulation in prostate cancer with promoter hypermethylation and poor prognosis. Urol Oncol. 2013; 31:622–627. doi: 10.1016/j.urolonc.2011.04.010
    1. Marfella R, Di Filippo C, Portoghese M, Barbieri M, Ferraraccio F, Siniscalchi M, Cacciapuoti F, Rossi F, D’Amico M, Paolisso G. Myocardial lipid accumulation in patients with pressure-overloaded heart and metabolic syndrome. J Lipid Res. 2009; 50:2314–2323. doi: 10.1194/jlr.P900032-JLR200
    1. Madrazo JA, Kelly DP. The PPAR trio: regulators of myocardial energy metabolism in health and disease. J Mol Cell Cardiol. 2008; 44:968–975. doi: 10.1016/j.yjmcc.2008.03.021
    1. Gilde AJ, van der Lee KA, Willemsen PH, Chinetti G, van der Leij FR, van der Vusse GJ, Staels B, van Bilsen M. Peroxisome proliferator-activated receptor (PPAR) alpha and PPARbeta/delta, but not PPARgamma, modulate the expression of genes involved in cardiac lipid metabolism. Circ Res. 2003; 92:518–524. doi: 10.1161/01.RES.0000060700.55247.7C
    1. Son NH, Park TS, Yamashita H, Yokoyama M, Huggins LA, Okajima K, Homma S, Szabolcs MJ, Huang LS, Goldberg IJ. Cardiomyocyte expression of PPARgamma leads to cardiac dysfunction in mice. J Clin Invest. 2007; 117:2791–2801. doi: 10.1172/JCI30335
    1. Rinaldi L, Avgustinova A, Martín M, Datta D, Solanas G, Neus P, Benitah SA. Loss of Dnmt3a and Dnmt3b does not affect epidermal homeostasis but promotes squamous transformation through PPARg. ELife. 2017; 6:1–25. doi: 10.7554/eLife.21697
    1. Dou X, Boyd-Kirkup JD, McDermott J, Zhang X, Li F, Rong B, Zhang R, Miao B, Chen P, Cheng H, et al. . The strand-biased mitochondrial DNA methylome and its regulation by DNMT3A. Genome Res. 2019; 29:1622–1634. doi: 10.1101/gr.234021.117
    1. Li L, Tao G, Hill MC, Zhang M, Morikawa Y, Martin JF. Pitx2 maintains mitochondrial function during regeneration to prevent myocardial fat deposition. Development. 2018; 145:dev168609. doi: 10.1242/dev.168609
    1. Fang X, Poulsen RR, Wang-Hu J, Shi O, Calvo NS, Simmons CS, Rivkees SA, Wendler CC. Knockdown of DNA methyltransferase 3a alters gene expression and inhibits function of embryonic cardiomyocytes. FASEB J. 2016; 30:3238–3255. doi: 10.1096/fj.201600346R
    1. Ohh M, Park CW, Ivan M, Hoffman MA, Kim TY, Huang LE, Pavletich N, Chau V, Kaelin WG. Ubiquitination of hypoxia-inducible factor requires direct binding to the beta-domain of the von Hippel-Lindau protein. Nat Cell Biol. 2000; 2:423–427. doi: 10.1038/35017054
    1. Appelhoff RJ, Tian YM, Raval RR, Turley H, Harris AL, Pugh CW, Ratcliffe PJ, Gleadle JM. Differential function of the prolyl hydroxylases PHD1, PHD2, and PHD3 in the regulation of hypoxia-inducible factor. J Biol Chem. 2004; 279:38458–38465. doi: 10.1074/jbc.M406026200
    1. Huang LE, Gu J, Schau M, Bunn HF. Regulation of hypoxia-inducible factor 1 is mediated by an O2-dependent degradation domain via the ubiquitin-proteasome pathway. Proc Natl Acad Sci U S A. 1998; 95:7987–7992. doi: 10.1073/pnas.95.14.7987
    1. Majmundar AJ, Wong WJ, Simon MC. Hypoxia-inducible factors and the response to hypoxic stress. Mol Cell. 2010; 40:294–309. doi: 10.1016/j.molcel.2010.09.022
    1. Laughner E, Taghavi P, Chiles K, Mahon PC, Semenza GL. HER2 (neu) signaling increases the rate of hypoxia-inducible factor 1alpha (HIF-1alpha) synthesis: novel mechanism for HIF-1-mediated vascular endothelial growth factor expression. Mol Cell Biol. 2001; 21:3995–4004. doi: 10.1128/MCB.21.12.3995-4004.2001
    1. Salnikow K, Donald SP, Bruick RK, Zhitkovich A, Phang JM, Kasprzak KS. Depletion of intracellular ascorbate by the carcinogenic metals nickel and cobalt results in the induction of hypoxic stress. J Biol Chem. 2004; 279:40337–40344. doi: 10.1074/jbc.M403057200
    1. Richard DE, Berra E, Gothié E, Roux D, Pouysségur J. p42/p44 mitogen-activated protein kinases phosphorylate hypoxia-inducible factor 1α (HIF-1α) and enhance the transcriptional activity of HIF-1. J Biol Chem. 1999; 274:32631–32637. doi: 10.1074/jbc.274.46.32631
    1. Lehmann LH, Jebessa ZH, Kreusser MM, Horsch A, He T, Kronlage M, Dewenter M, Sramek V, Oehl U, Krebs-Haupenthal J, et al. . A proteolytic fragment of histone deacetylase 4 protects the heart from failure by regulating the hexosamine biosynthetic pathway. Nat Med. 2018; 24:62–72. doi: 10.1038/nm.4452
    1. Ehrlich M. Expression of various genes is controlled by DNA methylation during mammalian development. J Cell Biochem. 2003; 88:899–910. doi: 10.1002/jcb.10464
    1. Kou CY-C, Lau SL-Y, Au K-W, Leung P-Y, Chim SS-C, Fung K-P, Waye MM-Y, Tsui SK-W. Epigenetic regulation of neonatal cardiomyocytes differentiation. Biochem Biophys Res Commun. 2010; 400:278–283. doi: 10.1016/j.bbrc.2010.08.064
    1. Okano M, Bell DW, Haber DA, Li E. DNA methyltransferases Dnmt3a and Dnmt3b are essential for de novo methylation and mammalian development. Cell. 1999; 99:247–257. doi: 10.1016/s0092-8674(00)81656-6
    1. Jaiswal S, Natarajan P, Silver AJ, Gibson CJ, Bick AG, Shvartz E, McConkey M, Gupta N, Gabriel S, Ardissino D, et al. . Clonal hematopoiesis and risk of atherosclerotic cardiovascular disease. N Engl J Med. 2017; 377:111–121. doi: 10.1056/NEJMoa1701719

Source: PubMed

3
Prenumerera