Skip to main content

Abstract

Background:

DYRK1a (dual-specificity tyrosine phosphorylation-regulated kinase 1a) contributes to the control of cycling cells, including cardiomyocytes. However, the effects of inhibition of DYRK1a on cardiac function and cycling cardiomyocytes after myocardial infarction (MI) remain unknown.

Methods:

We investigated the impacts of pharmacological inhibition and conditional genetic ablation of DYRK1a on endogenous cardiomyocyte cycling and left ventricular systolic function in ischemia-reperfusion (I/R) MI using αMHC-MerDreMer-Ki67p-RoxedCre::Rox-Lox-tdTomato-eGFP (RLTG) (denoted αDKRC::RLTG) and αMHC-Cre::Fucci2aR::DYRK1aflox/flox mice.

Results:

We observed that harmine, an inhibitor of DYRK1a, improved left ventricular ejection fraction (39.5±1.6% and 29.1±1.6%, harmine versus placebo, respectively), 2 weeks after I/R MI. Harmine also increased cardiomyocyte cycling after I/R MI in αDKRC::RLTG mice, 10.8±1.5 versus 24.3±2.6 enhanced Green Fluorescent Protein (eGFP)+ cardiomyocytes, placebo versus harmine, respectively, P=1.0×103. The effects of harmine on left ventricular ejection fraction were attenuated in αDKRC::DTA mice that expressed an inducible diphtheria toxin in adult cycling cardiomyocytes. The conditional cardiomyocyte-specific genetic ablation of DYRK1a in αMHC-Cre::Fucci2aR::DYRK1aflox/flox (denoted DYRK1a k/o) mice caused cardiomyocyte hyperplasia at baseline (210±28 versus 126±5 cardiomyocytes per 40× field, DYRK1a k/o versus controls, respectively, P=1.7×102) without changes in cardiac function compared with controls, or compensatory changes in the expression of other DYRK isoforms. After I/R MI, DYRK1a k/o mice had improved left ventricular function (left ventricular ejection fraction 41.8±2.2% and 26.4±0.8%, DYRK1a k/o versus control, respectively, P=3.7×10−2). RNAseq of cardiomyocytes isolated from αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice after I/R MI or Sham surgeries identified enrichment in mitotic cell cycle genes in αMHC-Cre::Fucci2aR::DYRK1aflox/flox compared with αMHC-Cre::Fucci2aR.

Conclusions:

The pharmacological inhibition or cardiomyocyte-specific ablation of DYRK1a caused baseline hyperplasia and improved cardiac function after I/R MI, with an increase in cell cycle gene expression, suggesting the inhibition of DYRK1a may serve as a therapeutic target to treat MI.

Graphical Abstract

Novelty and Significance

What Is Known?

Adult mammalian cardiomyocytes have limited cycling after an injury.
Signaling nodes active in developing hearts and downregulated in adult hearts have been implicated as potential pathways controlling cardiomyocyte cycling.
DYRK1a (dual-specificity tyrosine phosphorylation-regulated kinase 1a) regulates cell growth in several tissues.

What New Information Does This Article Contribute?

Our findings show that the pharmacological inhibition of DYRK1a after ischemia-reperfusion myocardial infarction enhances adult cardiomyocyte cycling and improves left ventricular function.
The cardiomyocyte-specific ablation of DYRK1a causes hyperplasia and improved cardiac function after myocardial infarction.
Cardiomyocytes that lack DYRK1a increase the expression of cell cycle-related genes after myocardial infarction.

Summary

Enhancing the adult mammalian cardiomyocytes cycling after an injury is an attractive strategy to improve cardiac function after myocardial infarction. One approach to identifying pathways that enhance cardiomyocyte cycling is to compare gene expression during normal development when cells exit the cycle. Therefore, we analyzed gene expression of embryonic, neonatal, and adult hearts and hypothesized that DYRK1a might serve as one potential signal controlling cardiomyocyte cycling. Using our αDKRC (cardiomyocyte-specific αMHC-MerDreMer-Ki67p-RoxedCre) transgenic reporter mouse, we observed a chemical inhibitor of DYRK1a, harmine, increased cardiomyocyte cycling, and improved ventricular function after ischemia-reperfusion myocardial infarction. The ablation of cycling cardiomyocytes attenuated the beneficial effects of harmine. We also observed that the cardiomyocyte-specific ablation of DYRK1a caused hyperplasia, improved cardiac function, and increased the expression of cell cycle-related genes after ischemia-reperfusion myocardial infarction. The results suggest that enhancing cardiomyocyte cycling, possibly through the inhibition of DYRK1a, may serve as a strategy to improve cardiac function after MI.
Rate-limiting pathways that potentially enhance cardiomyocyte cycling may serve as targets to treat cardiovascular disease. Endogenously cycling adult cardiomyocytes are scarce; however, our previous findings suggest that their ablation worsens myocardial function after infarction.1 Conversely, enhancing cardiomyocyte proliferation is associated with improved left ventricular (LV) function after injury, suggesting signals that induce cycling are potential avenues to treat MI.2–5
Generally, cardiomyocyte cycling involves canonical cell cycle proteins and redundant brakes such that removing one or even several brakes will not be sufficient to generate enough cardiomyocytes to confer substantial overall functional benefit. However, upstream signaling nodes that induce global changes in cycling programs may circumvent several brakes simultaneously and increase endogenous cardiomyocyte cycling. Clearly, manipulating critical signaling nodes such as the Hippo/YAP axis induced proliferation and improvement in myocardial function after injury.2,3,5,6 Thus, manipulating upstream nodal signals that control G1/S and G2/M transitions, rather than canonical cell cycle proteins, may also induce cardiomyocyte cycling and improve cardiac function.
Many pathways regulate the cell cycle. In some cells, the Myb-MuvB/dimerization partner, RB-like, E2F, and multi-vulval class B (DREAM) multiprotein complex controls G1/S and G2/M checkpoints at the level of genes targeted by E2F and Myb.7–11 The MuvB/DREAM complex acts as a brake, preventing the expression of genes at the G1/S and G2/M transitions in several cell types.7,10 Growth signals release the brake at G1/S and change the MuvB/DREAM complex into an accelerator at G2/M, enhancing the exit from quiescence to active cell cycling (Figure S1). Cardiomyocyte cycling involves E2Fs12–17; however, the contribution of the MuvB/DREAM complex is unclear. The ablation of one or a few components of the MuvB/DREAM complex likely impacts cell growth.9,11,18 Therefore, we focused on upstream regulators that potentially regulate the expression of many genes controlled by the MuvB/DREAM complex. The DYRK1a (dual-specificity tyrosine phosphorylation-regulated kinase 1a) is a critical regulator of MuvB/DREAM and influences cycling in several cells.9 The inhibition of DYRK1a promotes cell cycling and proliferation in mouse and human pancreatic islet cells,19–24 and DYRK1a has been implicated in cardiomyocyte growth through cyclin D125. Given the importance of E2F signals in cardiomyocyte growth and the potential of DYRK1a as a regulator of genes expressed at the G1/S and G2/M boundaries in cycling cells, we investigated the effects of pharmacological inhibition and genetic ablation of DYRK1a on cardiomyocyte cycling and cardiac function after ischemia-reperfusion (I/R) MI.
Using our recently published reporter mouse, αDKRC::RLTG, that labels endogenously cycling adult cardiomyocytes,1 we observed the DYRK1a inhibitor, harmine, enhanced cardiomyocyte cycling, and improved myocardial function after I/R MI. The expression of Diphtheria toxin to ablate cardiomyocytes that cycle attenuated the beneficial effects of harmine in LV systolic function after MI. Next, we generated αMHC-Cre::Fucci2aR::DYRK1aflox/flox mice to label cardiomyocyte nuclei and ablated DYRK1a. At baseline, αMHC-Cre::Fucci2aR::DYRK1aflox/flox mice had cardiomyocyte hyperplasia; however, cardiac function was similar to control mice. The cardiomyocyte-specific ablation of DYRK1a improved cardiac function after I/R MI. To understand the pathways involved in DYRK1a ablation, we performed RNAseq of cardiomyocytes isolated from αMHC-Cre::Fucci2aR::DYRK1aflox/flox after I/R MI or sham surgeries and identified differential enrichment in mitotic cell cycle genes, including pathways controlled by the MuvB/DREAM complex. Collectively, our findings suggest the inhibition of DYRK1a is a potential therapeutic target to enhance cardiomyocyte cycling and improve heart function after MI.

Methods

Data Availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.
See Supplemental Material for detailed methods.

Results

DYRK1a Is a Potential Regulator of Postnatal Cardiomyocyte Growth

Analyses of gene expression during postnatal heart growth are complex but can implicate candidate signals controlling the exit of cardiomyocytes from the cell cycle. Therefore, we analyzed three publically available datasets of gene expression of cardiomyocytes from the adult, neonatal, and embryonic mouse hearts to identify pathways potentially controlling postnatal cardiomyocyte growth that may serve as targets to induce adult cardiomyocyte cycling after injury. First, we examined RNA expression data obtained from FACS-purified cardiomyocytes of embryonic E11-12, neonatal P3-4, and adult (10 weeks old) C57BL/6 mice described by Walsh et al26 (NCBI Gene Expression Omnibus [GEO Accession number GSE17020]). Genes clustered into 7 categories based on expression defined as (1) slow downregulation, (2) rapid downregulation, (3) moderately rapid downregulation, (4) transiently up-regulated in neonatal, (5) slow upregulation, (6) rapid upregulation, and (7) moderately rapid upregulation (Figure S2A; Table S1). Clusters 5 to 7 corresponding to the genes upregulated in hearts of adults and neonates compared with embryos were enriched for pathways associated with extracellular matrix organization and Interleukin-10 signaling (Figure S2B and S2C). To identify pathways that were downregulated during the exit of cardiomyocytes from the cell cycle, we analyzed Clusters 1 to 3 using Signatures Database (MSigDB) gene set enrichment analysis27,28 (Figure S2B and S2C). Consistent with the findings reported by Walsh et al, we identified enrichment in genes associated with E2F targets, G2/M checkpoint gene sets, and E2F1 signals, known to participate in the control of cell cycle and DNA replication.
Second, we analyzed data from Talman et al,29 who produced a detailed molecular atlas of postnatal mouse heart development at ages P1, P4, P9, and P23 using RNAseq, proteomics, and metabolomics approaches. Recognizing the importance of the MuvB/DREAM complex in the regulation of G1/S and G2/M checkpoints at the level of genes targeted by E2F and Myb, we examined the temporal change in 214 genes predicted to be regulated by the MuvB/DREAM complex, based on the presence of proximal E2F or CHR sites (Figure S2D).7,8 Peak gene expression of the 214 genes occurred at P4 during the postnatal heart growth, corresponding to the window when cardiomyocytes exit the mitotic cell cycle.30 The findings suggested a correlation between the expression of genes regulated by the MuvB/DREAM complex and postnatal cardiomyocyte growth and cycling.
Third, we analyzed data from a multicellular transcriptional analysis of mammalian heart regeneration.31 Consistent with the observations of E2Fs in normal postnatal cardiomyocyte growth, several transcription factors regulated by the MuvB-DREAM complex, including E2F1, E2F2, FoxM1, Mybl2, and Myb, were differentially expressed in cardiomyocytes in P1 hearts. Based on our analyses of the three published investigations,26,29,31 we hypothesized that a unifying theme was the MuvB-DREAM complex as a candidate regulator of cardiomyocyte cycling.
Next, we quantified the expression of seventeen genes encoding components of the MuvB-DREAM complex of hearts isolated from P1, P3, P5, P7, P10, P14, and P28 C57B6J mice (Figure S2E). E2F1, E2F3, E2F4, and E2F5 had peak expression between P5 and P7, the postnatal period when cardiomyocytes exit the cell cycle. Rbl1 (p107) and Rbl2 (p130) had reciprocal changes, consistent with changes in expression of these genes in proliferating and quiescent cells.14,15,32,33 Components of the DREAM complex, including Lin52, Lin9, Lin37, and Lin54, were differentially expressed in postnatal hearts.
Since the MuvB/DREAM complex is a multimeric protein structure, the ablation of one or a few complex proteins is likely detrimental to cell growth. Therefore, we focused our attention on upstream regulators of the MuvB/DREAM complex since this was predicted to influence the subsequent expression of a large number of cell cycle genes. DYRK1a is a regulator of MuvB/DREAM and implicated in cell cycling.9,10 For example, the inhibition of DYRK1a promotes cell cycling and proliferation in mouse and human pancreatic islet cells,19–24 and DYRK1a has been implicated in cardiomyocyte growth through cyclin D125. Given the importance of E2F signals in cardiomyocyte growth and the potential of DYRK1a as a regulator of genes expressed at the G1/S and G2/M boundaries in cycling cells, we investigated the effects of pharmacological inhibition and genetic ablation of DYRK1a inhibition on cardiomyocyte cycling and cardiac function after I/R MI.

Harmine, an Inhibitor of DYRK1a, Improved Left Ventricular Function and Increased Cardiomyocyte Cycling After IR MI

Recently, we described a new transgenic mouse, αMHC (alpha myosin heavy chain)-MerDreMer-Ki67p-RoxedCre::Rox-Lox-tdTomato-eGFP (denoted αDKRC::RLTG) that restricted Cre expression to cycling adult cardiomyocytes and uniquely integrated spatial and temporal adult cardiomyocyte cycling events.1 αDKRC::RLTG mice provide the unique opportunity to quantify the effects of inhibitors on endogenous cardiomyocyte cycling after MI, in vivo. Therefore, we investigated if harmine, an ATP-competitive inhibitor of DYRK1a shown to promote pancreatic islet cell proliferation,23,34 induced endogenous cardiomyocyte cycling and changes in myocardial function after I/R MI. Harmine was administered after I/R MI via osmotic minipump at a dose of 10 mg/kg per animal over 2-week interval, comparable to the dose of harmine reported in in vivo investigations. Adult αDKRC::RLTG mice underwent pretreatment with tamoxifen (1 mg/day intraperitoneal [IP] injection×5 days for 2 cycles) to activate the αDKRC reporter. Two weeks after recovery from tamoxifen, we performed baseline echocardiography and 60 minutes of LAD ligation-mediated ischemia followed by reperfusion (Figure 1A). After reperfusion, but before the conclusion of surgeries, mice underwent the insertion of subcutaneous osmotic minipumps containing harmine (10 mg/kg per mL) or saline. Echocardiography was repeated 2 weeks after surgery, and eGFP+ cardiomyocytes and infarct sizes were quantified using protocols previously described. Harmine improved left ventricular ejection fraction (LVEF; Figure 1B and 1C) and LV end-systolic volume (Figure 1D and 1E), but not LV end-diastolic volume (Figure 1F and 1G) compared with saline controls.
Figure 1. Harmine improved left ventricular function after ischemia-reperfusion myocardial infarction. A, Outline of the experimental protocol testing the effects of harmine and saline (Control) in adult αDKRC::RLTG mice after 60 min of LAD ligation-mediated ischemia and reperfusion (I/R) myocardial infarction (MI). B and C, The absolute values (%) (B) and percent changes (C) in left ventricular (LV) ejection fraction (EF) of mice treated with harmine minipumps (10 mg/kg per d) or saline after 60 min of I/R MI. Bars are the mean±SD at baseline and 2 wks after I/R MI in B. Open circles are measurements for individual animals. P=1.0×102 for baseline vs I/R MI in the harmine treatment group, P=1.0×104 for baseline vs I/R MI in the Saline treatment group, and P=1.0×104 for harmine vs saline treatment in the I/R MI groups. P were calculated by 2-way ANOVA with Bonferroni multiple comparison test using 6 comparisons. Values are means±SD. N=8 animals per group. C, P=3.0×104 for the percent change in left ventricular ejection fraction between harmine and saline treatments. P calculated by Student t test. D and E, The absolute values (ul) (D) and percent changes (E) in LV end-systolic volume (ESV) of mice treated with harmine minipumps (10 mg/kg per d) or saline after 60 min of I/R MI. Bars are the mean±SD at baseline and 2 wks after I/R MI in D. Open circles are measurements for individual animals. P=4.0×102 for LV ESV at baseline vs I/R MI in the harmine treatment group, P=1.0×104 for LV ESV baseline vs I/R MI in the saline treatment group and P=5.0×103 for LV ESV harmine vs saline treatment in the I/R MI groups. P were calculated by 2-way ANOVA with Bonferroni multiple comparison test using 6 comparisons. Values are means±SD. N=8 animals per group. E, P=1.0×10-2 for the percent change in LV ESV between harmine and saline treatments. P calculated by Student t test. F and G, The absolute values (%) (F) and percent changes (G) in LV end-diastolic volumes (EDV) of mice treated with harmine minipumps (10 mg/kg per d) or saline after 60 min of I/R MI. Bars are the mean±SD at baseline and 2 wks after I/R MI in F. Open circles are measurements for individual animals. P=1.4×101 for LV EDV at baseline vs I/R MI in the harmine treatment group, P=2.0×104 for LV EDV at baseline vs I/R MI in the saline treatment group, and P=9.0×102 for LV EDV harmine vs saline treatment in the I/R MI groups. P were calculated by 2-way ANOVA with Bonferroni multiple comparison test using 6 comparisons. Values are means±SD. N=8 animals per group. G, P=2.5×102 for the percent change in LV ESV between harmine and saline treatments. P calculated by Student t test.
The infarcts of mice treated with harmine were similar to saline controls (Figure 2A and 2B). Two weeks after I/R MI, eGFP+ cardiomyocytes representing adult cardiomyocytes that re-entered the cell cycle were readily identified in hearts of αDKRC::RLTG (Figure 2C through 2E; Figure S3). GFP+ cardiomyocytes increased in number after I/R MI in harmine-treated animals compared with saline controls (24.3±2.6 versus 10.8±1.5 cardiomyocytes per 10-micron section, harmine versus saline, P=1.0×10−3) with the majority of eGFP+ cardiomyocytes localized in the ischemic and border zones (Figure 2F). In addition, LVEF, EDD, and ESD were linearly related to the number of eGFP+ cardiomyocytes (Figure 2G through 2I), suggesting that an increase in cycling cardiomyocytes corresponded to improved LV function after MI despite similar infarct sizes (Figure 2A). Thus, the pharmacological inhibition of DYRK1a increased the number of cycled cardiomyocytes and improved LV systolic function after I/R MI.
Figure 2. Harmine enhanced cardiomyocyte cycling after ischemia-reperfusion myocardial infarction. A, Infarct sizes for each group corresponding to Figure 1. Open circles are values for individual animals. Bars are group means±SD. P=4.5×101 by Student t test. B, Representative Masson-Trichrome stained short-axis sections of hearts of mice treated with harmine or saline. Scale bars denote 1 mm. C, Representative short-axis immunohistochemical (IHC) images of a heart from αDKRC::RLTG mouse treated with saline 2 wk after ischemia-reperfusion (I/R) myocardial infarction (MI). White circles denote eGFP+ cardiomyocytes. Scale bar denotes 1 mm. D, Representative short-axis IHC images of a heart from αDKRC::RLTG mouse treated with harmine 2 wk after I/R MI. White circles denote eGFP+ cardiomyocytes. Scale bar denotes 1 mm. E, Representative Immunohistochemical image of eGFP+ cardiomyocytes in the border zone of a harmine-treated animal. Arrows denote eGFP+ cardiomyocytes. Scale bar denotes 500 microns. F, Quantification of eGFP+ cardiomyocytes per section from I/R MI hearts treated with saline or harmine and the infarct, border, and remote zones of I/R MI hearts. Bars are group means±SD. Open circles are individual animals. N=8 animals in the saline group and N=7 animals in the harmine group. P=1.0×103 for total eGFP+ cardiomyocytes of saline vs harmine-treated animals after I/R MI. P=6.0×102 for eGFP+ cardiomyocytes in the ischemia area of saline vs harmine-treated animals after I/R MI. P=2.0×102 for eGFP+ cardiomyocytes in the border area of saline vs harmine-treated animals after I/R MI. P=5.0×102 for eGFP+ cardiomyocytes in the remote area of saline vs harmine-treated animals after I/R MI. P calculated by Mann-Whitney U test. G–I, Relationship between echocardiographic parameters and total number of cycled cardiomyocytes per animal. Animals treated with saline (gray circles) and harmine (open circles) are shown for LVEF, r2=0.61 (G), EDV, r2=0.39 (H), and ESD, r2=0.52 (I). The Pearson coefficient (r2) calculated by linear regression.

The Ablation of Cycling Cardiomyocytes Attenuated the Beneficial Effects of Harmine on LV Function After MI

Next, we used αDKRC::RLTG/DTA mice to ablate cardiomyocytes that re-entered the cell cycle and investigated if the beneficial effects of harmine on LV function after I/R MI were in part attributable to cardiomyocyte cycling. Male and female αDKRC::RLTG/DTA and littermate controls (+::RLTG/DTA) were pretreated with Tamoxifen and underwent I/R MI followed by the immediate implantation of an osmotic pump to deliver harmine (10 mg/kg per mL; Figure 3A). Serial echocardiography showed that αDKRC::RLTG/DTA treated with harmine had progressively decreased LVEF (Figure 3B and 3C) and increased end systolic volume (Figure 3D and 3E), with no differences in end diastolic volume (Figure 3F and 3G) compared with littermate controls (+::RLTG/DTA) treated with harmine. The changes in LVEF were similar to prior observations using αDKRC::DTA mice (Figure S4).1 The changes in LV parameters suggested that the effects of harmine on LV remodeling after I/R MI required cycling cardiomyocytes.
Figure 3. The ablation of cycling cardiomyocytes attenuated the beneficial effects of harmine on left ventricular (LV) systolic function after ischemia-reperfusion (I/R) myocardial infarction (MI). A, Protocol using αDKRC (cardiomyocyte-specific αMHC-MerDreMer-Ki67p-RoxedCre)::RLTG (Rox-Lox-tdTomato-eGFP)/Rosa26-DTA (Diphtheria Toxin) (αDKRC::RLTG/DTA) and +::RLTG/DTA littermate controls treated with Tamoxifen (1 mg/kg IP [intraperitoneal] daily×5 d×2 cycles) followed by recovery period before undergoing 60 min of left anterior descending (LAD) artery-ligation I/R (MI) and implantation of an osmotic minipump to deliver harmine (10 mg/kg per d). Serial echocardiography was performed before and after MI. N=7 in the αDKRC::RLTG/DTA and N=6 in the +::RLTG/DTA group. B and C, The absolute values (%) (B) and percent changes (C) in left ventricular (LV) ejection fraction (EF) of αDKRC::RLTG/DTA and +::RLTG/DTA mice treated with harmine minipumps (10 mg/kg per d) after 60 min of I/R MI. Open and closed circles are means±SDs of +::RLTG/DTA and αDKRC::RLTG/DTA mice, respectively. * denotes P=1.0×104 and # P=1.0×104 for the indicated time. P were calculated by Student t test. N=7 in the αDKRC::RLTG/DTA and N=6 in the +::RLTG/DTA group. C, P=1.0×104 for the percent change in LVEF between +::RLTG/DTA and αDKRC::RLTG/DTA mice. P calculated by Student t test. D and E, The absolute values (uL) (D) and percent changes (E) in LV end-systolic volume (ESV) of αDKRC::RLTG/DTA and +::RLTG/DTA mice treated with harmine minipumps (10 mg/kg per d) after 60 min of I/R MI. Open and closed circles are means±SDs of +::RLTG/DTA and αDKRC::RLTG/DTA mice, respectively. * P=4.0×103 for the indicated time. P were calculated by Student t test. N=7 in the αDKRC::RLTG/DTA and N=6 in the +::RLTG/DTA group. D, P=8.0×102 for the percent change in LVEF between +::RLTG/DTA and αDKRC::RLTG/DTA mice. P calculated by Student t test. F and G, The absolute values (%) (F) and percent changes (G) in LV end-diastolic volumes (EDV) of αDKRC::RLTG/DTA and +::RLTG/DTA mice treated with harmine minipumps (10 mg/kg per d) after 60 min of I/R MI. Open and closed circles are means±SDs of +::RLTG/DTA and αDKRC::RLTG/DTA mice, respectively. N=7 in the αDKRC::RLTG/DTA and N=6 in the +::RLTG/DTA group. D, P=5.6×10-1 for the percent change in LVEF between +::RLTG/DTA and αDKRC::RLTG/DTA mice. P calculated by Student t test.

The Cardiomyocyte-Specific Ablation of DYRK1a Caused Baseline Hyperplasia

Pharmacological studies are clinically more practical but inherently less specific in terms of the cell populations affected by drug treatment and potential off-target effects. Therefore, we investigated the effects of cardiomyocyte-specific genetic ablation of DYRK1a on cardiac function after I/R MI. We generated conditional knockouts, αMHC-Cre::Fucci2aR::DYRK1aflox/flox mice, using DYRK1aflox/floxmice created and validated by Thompson et al35 in the context of a Cre-inducible second-generation Fluorescence Ubiquitin Cell Cycle Indicator, Fucci2aR, to label cardiomyocyte nuclei (Figure 4A).36 αMHC-Cre::Fucci2aR mice served as controls. Of note, the αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice were bred >9 generations into a C57BL/6J genetic background.
Figure 4. The cardiomyocyte-specific ablation of DYRK1a (dual-specificity tyrosine phosphorylation-regulated kinase 1a) caused baseline hyperplasia. A, Overview of the αMHC-Cre::Fucci2aR::DYRK1aflox/flox (denoted DYRK1a k/o) mouse. B, Quantitative polymerase chain reaction (qPCR) of DYRK1a, DYRK1b, and DYRK2 expression of total RNA extracted from cardiomyocytes isolated from adult αMHC-Cre::Fucci2aR::DYRK1aflox/flox (DYRK1a k/o) and αMHC-Cre::Fucci2aR (Control) mice. The probe set for DYRK1a corresponded to Exons 5 and 6, the exons flanked by LoxP sites (A). P calculated by Mann-Whitney U test. Values are mean±SD. Open circles are values for individual animals. N=3 for αMHC-Cre::Fucci2aR::DYRK1aflox/flox (DYRK1a k/o) and N=5 for αMHC-Cre::Fucci2aR animals (Control). C, Number of cardiomyocytes per 40×field for αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR animals. Values are means±SD. Open circles are individual animals. N=5 animals per group, n=8 fields per animal. P=1.7×102 for number of cardiomyocytes per 40×field of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR animals. P by Student t test. D, Violin plots of the cross-sectional areas (CSAs) (um2) of cardiomyocytes of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR animals. N=3 animals per group, n=317 cells for αMHC-Cre::Fucci2aR and n=257 cells for αMHC-Cre::Fucci2aR::DYRK1aflox/flox. P=1.0×101 for cardiomyocyte CSA of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR animals. P by Mann-Whitney U test. The mean values for each animal was used to account for nonindependence. E and F, Representative immunofluorescence images of hearts of αMHC-Cre::Fucci2aR (E) and αMHC-Cre::Fucci2aR::DYRK1aflox/flox (F) mice. Red – mCherry labeling cardiomyocyte nuclei. Gray—wheat germ agglutinin. Scale bars=25 microns. G, LV end-diastolic volume (EDV) (uL) of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice. Values are mean±SD. Open circles are measurements for individual animals. N=17 animals for αMHC-Cre::Fucci2aR and N=13 animals for αMHC-Cre::Fucci2aR::DYRK1aflox/flox. P=8.0×102 for LV EDV of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR animals. P by Student t test. H, LV end-systolic volume (ESV) (uL) of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice. Values are mean±SD. Open circles are measurements for individual animals. N=17 animals for αMHC-Cre::Fucci2aR and N=animals for αMHC-Cre::Fucci2aR::DYRK1aflox/flox. P=1.1×101 for LV ESV of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR animals. P by Student t test. I, LV Ejection Fraction (EF) (%) of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice. Values are mean±SD. Open circles are measurements for individual animals. N=17 animals for αMHC-Cre::Fucci2aR and N=13 animals for αMHC-Cre::Fucci2aR::DYRK1aflox/flox. P=8.8×102 for LVEF of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR animals. P by Student t test. J and K, Gravimetric measurements of hearts from αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice expressed as heart weight (mg) to tibia length (mm) (J) and heart weight (mg) to body weight (g) (K). Values are mean±SD. Open circles are measurements for individual animals. N=8 animals per group. P by Student t test.
αMHC-Cre::Fucci2aR::DYRK1aflox/flox mice were viable and fertile. Adult cardiomyocytes isolated from αMHC-Cre::Fucci2aR::DYRK1aflox/flox mice had reduced expression of DYRK1a without compensatory changes in the expression of other DYRKs, compared with cardiomyocytes of αMHC-Cre::Fucci2aR controls (Figure 4B). At baseline, the hearts of αMHC-Cre::Fucci2aR::DYRK1aflox/flox mice had smaller cardiomyocyte cross-sectional areas and increased cardiomyocytes per area, consistent with hyperplasia (Figure 4C through 4F). Baseline left ventricular end-diastolic volume, end-systolic volume, and ejection fractions were also similar between αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice (Figure 4G through 4I). Heart weights were similar between αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice but with a trend toward slightly increased heart size in the DYRK1a k/o mice (Figure 4J and 4K). The results suggested that the ablation of DYRK1a caused cardiomyocyte hyperplasia.
Next, we performed 60-minute I/R MI and sham surgeries using αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice (Figure 5A). Four weeks after I/R MI, αMHC-Cre::Fucci2aR::DYRK1aflox/flox mice had improved LVEF (Figure 5B and 5C) and LV end-systolic volumes (Figure 5D and 5E) compared with αMHC-Cre::Fucci2aR. LV end-diastolic volumes were similar between αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice (Figure 5F and 5G). Next, we analyzed the infarct characteristic of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice (Figure 6A through 6E). The infarct sizes were quantified as the percentage infarct area (PIA) (Figure 6A), the percent infarct circumference of the short-axis (PIC) (Figure 6B), the ratio of the PIA to PIC (Figure 6C) at the level of the mid-ventricle that corresponds to infarct thickness in the infarcted area, and the ratio of PIC to PIA (Figure 6D) that corresponds to thickness of noninfarcted myocardium in the infarcted area. αMHC-Cre::Fucci2aR::DYRK1aflox/flox had similar PIAs compared with control αMHC-Cre::Fucci2aR mice. However, αMHC-Cre::Fucci2aR::DYRK1aflox/flox had smaller PICs compared with control αMHC-Cre::Fucci2aR mice, suggesting that the cardiomyocyte-specific ablation of DYRK1a influenced post-I/R MI LV remodeling to produce thicker scars with smaller circumferences.
Figure 5. The cardiomyocyte-specific ablation of DYRK1a (dual-specificity tyrosine phosphorylation-regulated kinase 1a) improved left ventricular function after ischemia-reperfusion (I/R) myocardial infarction (MI). A, Outline of the experimental protocol testing the effects of the cardiomyocyte-specific ablation of DYRK1a in αMHC-Cre::Fucci2aR::DYRK1aflox/flox (DYRK1a k/o) and αMHC-Cre::Fucci2aR (Control) mice after 60 min of LAD ligation-mediated I/R MI. B and C, The absolute values (%) (B) and percent changes (C) in left ventricular (LV) ejection fractions (EFs) of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice after 60 min of I/R MI. Bars are the means±SDs at baseline, 24 h after surgery, and 4 wks after I/R MI in B. Open circles are measurements for individual animals. P were calculated by a 2-way ANOVA with Tukey multiple comparison test. C, P=7.0×103 for the percent change in LVEF between sham and I/R MI at 4 wks compared with baseline in the αMHC-Cre::Fucci2aR group. P=1.2×102 for the percent change in LVEF between sham and I/R MI in the αMHC-Cre::Fucci2aR::DYRK1aflox/flox group. P were calculated by a Kruskal-Wallis test with Dunn multiple comparison test. Values are means±SD. N=11 animals in the αMHC-Cre::Fucci2aR::DYRK1aflox/flox sham group, N=9 animals in the αMHC-Cre::Fucci2aR::DYRK1aflox/flox I/R MI group, N=4 animals in the αMHC-Cre::Fucci2aR sham group, N=5 animals in the αMHC-Cre::Fucci2aR I/R MI group. D and E, The absolute values (%) (D) and percent changes (E) in left ventricular (LV) end-systolic volume (ESD) (uL) of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice after 60 min of I/R MI. Bars are the means±SDs at baseline, 24 h after surgery, and 4 wks after I/R MI in B. Open circles are measurements for individual animals. P were calculated by a 2-way ANOVA with Tukey multiple comparison test. E, the percent changes in LV ESD at 4 wks compared with baseline are shown. Values are means±SD. P=0.02 for the percent change in LV EDD at 4 wks compared with baseline between sham and I/R MI in the αMHC-Cre::Fucci2aR group. P=2.6×10-1 for the percent change in LV EDD between sham and I/R MI in the αMHC-Cre::Fucci2aR::DYRK1aflox/flox group. P were calculated by a Kruskal-Wallis test with Dunn multiple comparison test. N=11 animals in the αMHC-Cre::Fucci2aR::DYRK1aflox/flox sham group, N=9 animals in the αMHC-Cre::Fucci2aR::DYRK1aflox/flox I/R MI group, N=4 animals in the αMHC-Cre::Fucci2aR sham group, N=5 animals in the αMHC-Cre::Fucci2aR I/R MI group. F and G, The absolute values (%) (F) and percent changes (G) in left ventricular (LV) end-diastolic volume (EDD) (uL) of αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice after 60 min of I/R MI. Bars are the means±SDs at baseline, 24 h after surgery, and 4 wks after I/R MI in B. Open circles are measurements for individual animals. P were calculated by 2-way ANOVA with Tukey multiple comparison test. N=11 animals in the αMHC-Cre::Fucci2aR::DYRK1aflox/flox sham group, N=9 animals in the αMHC-Cre::Fucci2aR::DYRK1aflox/flox I/R MI group, N=4 animals in the αMHC-Cre::Fucci2aR sham group, N=5 animals in the αMHC-Cre::Fucci2aR I/R MI group.
Figure 6. The cardiomyocyte-specific ablation of DYRK1a (dual-specificity tyrosine phosphorylation-regulated kinase 1a) altered infarct parameters. A, Percent infarct area for αMHC-Cre::Fucci2aR (Control) and αMHC-Cre::Fucci2aR::DYRK1aflox/flox (DYRK1a k/o) animals corresponding to Figure 5. Open circles are values for individual animals. Bars are group means±SD. P=3.9×101 for the percent infarct area between αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox animals. P were calculated by Student t test. N=8 animals per group. B, Percent circumference of the infarct for αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox animals corresponding to Figure 5. Open circles are values for individual animals. Bars are group means±SD. P=5.2×102 for the percent circumference of the infarct between αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox animals. P were calculated by Student t test. N=8 animals per group. C, Ratio of the percent infarct area to percent circumference of the infarct for αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox animals corresponding to Figure 5. Open circles are values for individual animals. Bars are group means±SD. P=4.6×102 for percent infarct area to percent circumference of the infarct between αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox animals. P were calculated by Student t test. N=8 animals per group. D, Ratio of the percent circumference of the infarct to percent infarct area for αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox animals corresponding to Figure 5. Open circles are values for individual animals. Bars are group means±SD. P=4.6×102 for percent infarct area to percent circumference of the infarct between αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox animals. P were calculated by Student t test. N=8 animals per group. E, Representative short axis base to apex sections of hearts from αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox animals 4 wks after I/R MI. Hearts were stained with Masson trichrome. Infarcted areas are shown. F, Representations of the infarct area and circumference of αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox animals 4 wks after I/R MI.
Since the knockout of DYRK1a could potentially alter the degree of injury occurring with ischemia, we performed (2,3,5-Triphenyltetrazolium chloride (TTC) staining (Figure S5A through S5C). The area-at-risk and area-of-necrosis 24 hours after MI were similar between αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice (Figure S5B and S5C). The results suggested that the cardiomyocyte-specific genetic ablation of DYRK1a improved LV function after I/R MI without a change in the area at risk. In light of the hyperplasia observed in DYRK1a k/o mouse hearts, the changes in LV remodeling after I/R MI could be a result of a larger initial number of cardiac myocytes contributing to the remodeling response after injury. The change in infarct parameters may also contribute to improved LV function after I/R MI in DYRK1a k/o mouse hearts.

The Ablation of DYRK1a Promotes the Expression of Cell Cycle Genes After I/R MI

Next, we investigated gene expression of cardiomyocytes to identify potential mechanisms responsible for the of DYRK1a ablation. Using Langendorff methods to retro-aortically perfuse collagenase ex vivo, we isolated cardiomyocytes from αMHC-Cre::Fucci2aR::DYRK1aflox/flox and control αMHC-Cre::Fucci2aR mice after I/R MI or Sham surgeries 7 days after I/R MI, corresponding to a time when cardiomyocyte cycling occurs after injury, and performed RNAseq (Figure 7A). Principal component analysis to reduce dimensionality of the datasets identified the clustering of samples into 4 distinct groups with 44% and 32% of the variance in gene expression attributed to MI compared with sham, and αMHC-Cre::Fucci2aR::DYRK1aflox/flox to αMHC-Cre::Fucci2aR, respectively (Figure 7B). Volcano plot of differential gene expression of sham control and sham DYRK1a k/o hearts showed an increase in expression of DREAM target genes and cardiac transcription factors (Nkx 2.5, Txb5, Gata4, and Gata6) in DYRK1a k/o cardiomyocyte preparations (Figure 7C). Volcano plots of cardiomyocyte gene expression comparing Sham and I/R MI for αMHC-Cre::Fucci2aR (Figure 8A and 8B) and αMHC-Cre::Fucci2aR::DYRK1aflox/flox identified that more genes had increased than decreased expression after I/R MI. To refine the analyses of genes that had increased expression between αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox after MI, we generated a Venn diagram of genes that had an adjusted P value of <0.05 and a 4-fold or greater expression (Figure 8C). One hundred seventy-six genes and 179 genes increased expression after I/R MI compared with Sham in cardiomyocytes isolated from αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox hearts, respectively (Table S2). Eighty-nine genes were uniquely expressed in αMHC-Cre::Fucci2aR::DYRK1aflox/flox cardiomyocytes after I/R MI. Eighty-six genes were uniquely expressed in αMHC-Cre::Fucci2aR cardiomyocytes after I/R MI. Ninety genes had shared expression in αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR cardiomyocytes. Using the Reactome database, we identified enrichment of pathways associated with mitotic cell cycle and cell cycle checkpoints in genes uniquely expressed in αMHC-Cre::Fucci2aR::DYRK1aflox/flox cardiomyocytes after I/R MI (Figure 8D and Figure S6). These genes included Anillin (Anln), Aurora Kinase B (Aurkb), Cyclin B1 (Ccnb1), Cyclin B2 (Ccnb2), Cyclin Dependent Kinase 1 (Cdk1), Cell Division Cycle 20 (Cdc20), Forkhead Box M1 (FOXM1), Kinetochore Localized Astrin (SPAG5) Binding Protein (KNSTRN), MYB Proto-Oncogene Like 2 (MYBL2), Polo Like Kinase 1 (PLK1), and Protein Regulator Of Cytokinesis 1 (PCR1), among others. Interleukin-10 and RUNX signaling were expressed in αMHC-Cre::Fucci2aR cardiomyocytes after I/R MI. Extracellular matrix formation genes has shared expression in αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR cardiomyocytes (Figure 8D). MsigDB gene set enrichment of the 89 genes uniquely expressed in αMHC-Cre::Fucci2aR::DYRK1aflox/flox cardiomyocytes after I/R MI identified G2/M checkpoint and E2F targets as the top hits, similar to the findings identified in the initial analyses of Cluster 1 from the gene expression of embryonic and postnatal hearts (Figure S1B). Thirty-five of the 89 genes (39%) were targets of the DREAM complex (Figure S7). Collectively, the results suggest that after I/R MI, the loss of DYRK1a in cardiomyocytes was associated with the expression of cell cycle genes.
Figure 7. The ablation of DYRK1a (dual-specificity tyrosine phosphorylation-regulated kinase 1a) promotes the expression of DREAM target genes. A, Overview of the experimental protocol of ischemia-reperfusion (I/R) myocardial infarction (MI) and sham surgeries in αMHC-Cre::Fucci2aR::DYRK1aflox/flox (DYRK1a k/o) and αMHC-Cre::Fucci2aR (Control) followed by cardiomyocyte isolation, and RNA and sequencing. B, Principal components analysis (PCA) plot of the 4 groups: αMHC-Cre::Fucci2aR::DYRK1aflox/flox (DYRK1a k/o) Sham, αMHC-Cre::Fucci2aR::DYRK1aflox/flox (DYRK1a k/o) I/R MI, αMHC-Cre::Fucci2aR (Control) Sham, and αMHC-Cre::Fucci2aR (Control) I/R MI. C, Volcano plot of gene expression of αMHC-Cre::Fucci2aR and αMHC-Cre::Fucci2aR::DYRK1aflox/flox cardiomyocytes after Sham surgeries. Green dots denote genes predicted to be targets of the DREAM complex, red dots denote genes encoding sarcomere proteins, and blue dots denote cardiac transcription factors.
Figure 8. The ablation of DYRK1a (dual-specificity tyrosine phosphorylation-regulated kinase 1a) promotes the expression of cell cycle genes after ischemia-reperfusion (I/R) myocardial infarction (MI). A, Volcano plot of gene expression of αMHC-Cre::Fucci2aR cardiomyocytes after I/R MI or Sham surgeries. Red dots represent genes with a >2-fold increase in expression and an adjusted P>0.05. Blue dots represent genes with <2-fold increase in expression and an adjusted P>0.05. Black dots represent genes with an adjusted P>0.05. Vertical dashed lines denote 4-fold levels of gene expression. The horizontal dashed line denotes an adjusted P=0.05. B, Volcano plot of gene expression of αMHC-Cre::Fucci2aR::DYRK1aflox/flox cardiomyocytes after Sham or I/R MI. Red dots represent genes with >2-fold increase in expression and an adjusted P>0.05. Blue dots represent genes with <2-fold increase in expression and an adjusted P>0.05. Black dots represent genes with an adjusted P>0.05. Vertical dashed lines denote 4-fold levels of gene expression. The horizontal dashed line denotes an adjusted P=0.05. C, Venn diagram of genes from C and D that had >2-fold increase in expression and an adjusted P>0.05. Eighty-nine genes were uniquely expressed in αMHC-Cre::Fucci2aR::DYRK1aflox/flox cardiomyocytes after I/R MI. Eighty-six genes were uniquely expressed in αMHC-Cre::Fucci2aR cardiomyocytes after I/R MI. Ninety genes had shared expression in αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR cardiomyocytes. D, Pathway enrichment or the genes in C based on Reactome database. Genes unique to the αMHC-Cre::Fucci2aR::DYRK1aflox/flox cardiomyocytes after I/R MI were enriched in mitotic cell cycle and cell cycle checkpoints. Genes that were unique to the αMHC-Cre::Fucci2aR cardiomyocytes after I/R MI were enriched in interleukin-10 and RUNX signaling. Genes in common between αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR cardiomyocytes after I/R MI were enriched in collagen formation and extracellular matrix organization. The tables list the Reactome pathway name, the number of genes in D found in the Reactome pathway, the number of total genes in the Reactome pathway, the ratio of genes found to total genes in the pathway, the P associated with over-represented genes within the Reactome pathway, and the false discovery rate (FDR).

Discussion

Cardiomyocyte cycling, like other cell types, involves canonical signaling proteins and redundant brakes to prevent aberrant growth. The removal of one or even several brakes is likely insufficient to generate enough functional cardiomyocytes to confer substantial overall benefit to cardiac function after injury, assuming the benefits are restricted to contractile function. Therefore, we sought to identify candidate upstream signals that potentially regulate genes expressed at the G1/S and G2/M boundaries to induce adult cardiomyocyte cycling after myocardial injury. We postulated that investigating gene expression during the normal transition when cardiomyocytes exit the cell cycle during neonatal heart growth would identify potential nodal signals for manipulation in adult cardiomyocytes.
Publically available gene expression data of embryonic, neonatal, and adult cardiomyocytes and hearts identified enrichments in pathways of E2Fs and G2/M checkpoints, and temporal changes in the expression of genes containing proximal E2F or CHR DNA-binding sites, known to be controlled by the DREAM complex. In addition, genes encoding components of the DREAM complex and E2Fs had peak expression in postnatal hearts during the window when cardiomyocytes exit the cell cycle. These observations suggested that the MuvB-DREAM complex represented a potential mechanism to induce cardiomyocyte cycling, and the inhibition of DYRK1a by pharmacological inhibition and genetic ablation may enhance cardiac function in part through enhanced cardiomyocyte cycling.
The administration of harmine at the time of reperfusion after MI improved systolic function and increased cardiomyocyte cycling in αDKRC::RLTG transgenic mice, without changes in infarct sizes. The harmine-induced cycled cardiomyocytes were predominantly in the ischemic and border zones. The observation that an increase in cycling cardiomyocytes corresponded to improved LV function after MI despite similar infarct sizes is particularly interesting in light of our recent findings that the ablation of cycling cardiomyocytes worsens cardiac function after MI. The number of cycled cells in response to harmine was sparse, and the fact that harmine did not induce uniform cardiomyocyte cycling suggests that harmine may activate cycling of the subpopulation of cardiomyocytes prone to reenter the cell cycle, particularly in the ischemic and border zones. Importantly, the I/R MI model did not produce a transmural infarct as seen with permanent LAD ligation MI models and therefore cardiomyocytes were present in the infarcted area. Moreover, the I/R MI model more closely recapitulates the clinical scenario of patients presenting with ST-elevation MI who undergo percutaneous revascularization within 90 minutes, a performance quality metric in the treatment of MI.37 Although harmine was started after reperfusion and continued for 2 weeks after MI, shorter dosing intervals of DYRK1a inhibitors after MI may provide similar beneficial results without the potential untoward sequelae of prolonged administration of pro-proliferative compounds that can theoretically induce cancer.
Cycled cardiomyocytes and cardiac function (LVEF) after I/R MI were linearly related. Previously, we observed that the ablation of cycled cardiomyocytes worsened cardiac function in animals that had similar infarct sizes after I/R MI.1 Moreover, the ablation of cycling cardiomyocytes attenuated the beneficial effects of harmine on LV function after I/R MI. One interpretation is that cycling cardiomyocytes express paracrine factors that contribute to myocardial function after injury, consistent with the observation that cycled cardiomyocytes are predominantly individual cells and not paired or clustered as would be expected of proliferation. Future investigations will characterize the gene expression profiles of cycling and noncycling cardiomyocytes in our αDKRC::RLTG transgenic mice to address this possible mechanism.
Since harmine likely has effects on multiple cell types and organs, we used a transgenic approach to examine the effects of the cardiomyocyte-specific ablation of DYRK1a after MI. At baseline, the ablation of DYRK1a in αMHC-Cre::Fucci2aR::DYRK1aflox/flox caused cardiomyocyte hyperplasia; however, the systolic function was similar to controls. Similarly, the ablation of Salvador or activated YAP expression caused cardiomyocyte hypertrophy with normal baseline systolic function.2,5,6 The genetic ablation of DYRK1a improved LV systolic function after MI and reduced infarct sizes. αMHC-Cre::Fucci2aR::DYRK1aflox/flox mice had area-at-risk and area-of-necrosis similar to αMHC-Cre::Fucci2aR controls, suggesting that DYRK1a ablation did not reduce the susceptibility to I/R injury. We then quantified infarct characteristics 4 weeks after I/R MI as the PIA, the percent infarct circumference of the short-axis (PIC), the ratio of the PIA to PIC at the level of the mid-ventricle that corresponds to infarct thickness in the infarcted area, and the ratio of PIC to PIA that corresponds to thickness of noninfarcted myocardium in the infarcted area. These measurements provide an estimation of scar thickness and size. The cardiomyocyte-specific knockout of DYRK1a produced thicker scars with smaller circumferences and these changes in remodeling may improve LV systolic function after injury.38 Future investigations using chronic ischemic cardiomyopathy models will potentially address the long-term impact of DYRK1a ablation in post-MI LV remodeling.
To explore potential mechanisms responsible for the beneficial effects of DYRK1a ablation in cardiomyocytes, we performed RNAseq on cardiomyocytes isolated from αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR mice 7 days after I/R MI or sham surgeries. Interestingly, the loss of DYRK1a was associated with the upregulation of cell cycle genes and downregulation of genes encoding contractile proteins after MI, compared with controls. One interpretation of the results is that the loss of DYRK1a promotes genetic programs associated with dedifferentiation and proliferation, consistent with observations of proliferating caridomyocytes.39–43 The gene signatures upregulated in cardiomyocytes of αMHC-Cre::Fucci2aR::DYRK1aflox/flox after I/R MI were similar differentially expressed in embryonic and neonatal cardiomyocytes described by Walsh et al.26 (Clusters 1-2 in Figures S2A, S2B, and S8). Conversely, the gene signatures upregulated in cardiomyocytes of αMHC-Cre::Fucci2aR control mice after I/R MI were similar to those differentially expressed in adult cardiomyocytes compared with embryonic and neonates (Clusters 5-6 in Figure S2A and S2C). Together, the results demonstrate the utility of gene expression data of developing hearts to identify candidate signaling nodes and potentially enhance adult cardiomyocyte cycling. DYRK1a inhibition represents one potential nodal regulator and the genetic ablation of DYRK1a in cardiomyocytes after myocardial injury recapitulates aspects of gene expression of developing myocardium.
Despite the unanticipated findings, there were limitations. First, the ablation of DYRK1a in αMHC-Cre::Fucci2aR::DYRK1aflox/flox mice occurred during cardiac development and, therefore, the adult phenotypes reflected the absence of DYRK1a during normal embryonic and post-natal heart growth. The hearts of αMHC-Cre::Fucci2aR::DYRK1aflox/flox had cardiomyocyte hyperplasia without changes in baseline echocardiographic parameters. In addition, the Fucci2aR signal of αMHC-Cre::Fucci2aR::DYRK1aflox/flox was mCherry+/mVenus-, similar to αMHC-Cre::Fucci2aR control mice, indicative of noncycling cardiomyocytes. Future investigations using tamoxifen-induced deletion of DYRK1a in cardiomyocytes harboring a mosaic analysis with double markers (MADM) reporter of cell proliferation44,45 (cTNTMerCreMer::MADM::DYRK1afl/fl) will address the potential of postdevelopmental ablation of DYRK1a to enhance cardiomyocyte cycling in the context of a reporter of mitosis. Importantly, cells that reenter the cell cycle can proliferate to generate new daughter cells or undergo endoreplication to multinucleate and increase polyploidy, processes that have important implications.46 Future investigations will quantify polyploidy in the context of DYRK1a.
Second, harmine, an ATP-competitive inhibitor for DYRK1a, could have off-target effects.34,47,48 Additional kinases may contribute to the observed effects in vivo despite the observations that cardiomyocyte-specific ablation of DYRK1a recapitulates harmine’s effects after MI. Investigation of structurally diverse DYRK1a inhibitors will be informative, particularly to the development of therapeutics to enhance cardiomyocyte cycling in the setting of myocardial infarction and percutaneous catheter-based reperfusion strategies.
Third, DYRK1a has many substrates in addition to the MuvB-DREAM complex that likely contribute to the overall effects observed with cell cycling.9–11,49 For example, DYRKs phosphorylate NFATs,50 Cyclins,25,35 p27/p21,51 and Lin52 of the DREAM complex.9 A genome-wide Drosophila RNAi screen identified DYRK-family kinases as regulators of NFAT.52 DYRK1a phosphorylates mammalian NFATc1, interfering with degradation and leading to NFAT1c stability.53 In cultured rat cardiomyocytes, adenovirus-mediated overexpression of DYRK1A antagonized calcineurin-mediated nuclear NFAT translocation and phenylephrine-induced hypertrophic growth response.50 However, in a mouse model of postdevelopmental DYRK1a expression, myocyte diameter, heart weight/body weight ratio, and echocardiographic measurements showed that myocardial expression of DYRK1A failed to reduce hypertrophy induced after transverse aortic banding.50 The results suggested insufficient long-term inhibition of NFAT by DYRK1a in the mouse model or additional pathways influenced by DYRK1a activity in vivo.
DYRK1a regulates the cardiomyocyte cell cycle through Cyclin D-dependent Rb/E2f-signaling.25,35 Transgenic mice with cardiac-specific overexpression of DYRK1a had increased phosphorylation of Ccnd2 with subsequent proteasomal degradation and hypo-phosphorylated Rb1, suppression of Rb/E2F-signaling, and reduced expression of E2F-target genes, which ultimately results in impaired cell cycle progression and lead to dilated cardiomyopathy.25
DYRK1a is located on chromosome 21 and has a naturally occurring gene dosage increase in trisomy 21.54 Dyrk1a mediates a dose-dependent increase in the duration and variability of the G1 phase and has been shown to shift cells within a p21-cyclin D1 signaling map to control the decision to enter the cell cycle.51 Additionally, DYRK1A is recruited preferentially to promoters of genes actively transcribed by RNA polymerase II, functionally associated with translation, RNA processing, and the cell cycle.55 Further investigations will address other DYRK1a-mediated signals in cycling cardiomyocytes.
Fourth, the RNAseq experiments have the limitation of contamination by noncardiomyocytes. All samples were prepared similarly and gene expression attributed to noncardiomyocytes should be filtered from the differential gene expression analyses. However, noncardiomyocyte cell populations could differ between αMHC-Cre::Fucci2aR::DYRK1aflox/flox and αMHC-Cre::Fucci2aR hearts after MI. For example, genes associated with interleukin-10 signals are enriched in αMHC-Cre::Fucci2aR hearts after MI. The improvement in cardiac function observed in αMHC-Cre::Fucci2aR::DYRK1aflox/flox could represent more favorable inflammatory responses mediated by changes in signals emanating from cardiomyocytes lacking DYRK1a.35 Future investigations of the temporal changes in inflammatory infiltrate after MI in hearts where cardiomyocytes lacking DYRK1a will be informative. Nonetheless, cardiomyocytes lacking DYRK1a the enrichment of genes associated with cycling after MI and recapitulated gene signatures associated with post-natal heart growth. Identifying the exact pathways mediating the effects of DYRK1a ablation on cycling programs in cardiomyocytes will improve our knowledge of targets to manipulate to improve myocardial function after MI.
Collectively, our findings suggest the inhibition of DYRK1a is a potential therapeutic target to enhance cardiomyocyte cycling and improve heart function after MI. Moreover, the MuvB-DREAM complex may serve as a regulator of cardiomyocyte cycling and merits further investigation.

Article Information

Author Contributions

All animal care and surgeries were in accordance with UVA ACUC Policy on Rodent Surgery and Perioperative Care under ACUC-approved animal protocol (UVA ACUC Wolf Laboratory protocol No. 4080). L.A. Bradley, A. Young, H.O. Billcheck, and M.J. Wolf designed experiments, generated reagents, and wrote the article. L.A. Bradley, A. Young, H.O. Billcheck, E. Farrar, and M.J. Wolf performed experiments. L.A. Bradley, A. Young, S. Tkachenko, J.J. Saucerman, S. Bekiranov, and M.J. Wolf analyzed the data. A. Young performed rodent surgeries. M.J. Wolf secured funding for the project.

Supplemental Material

Figures S1–S8
Tables S1–S3
Expanded Materials and Methods

Footnote

Nonstandard Abbreviations and Acronyms

DYRK1a
dual-specificity tyrosine phosphorylation-regulated kinase 1a
I/R
ischemia-reperfusion
PIA
percentage infarct area
MI
myocardial infarction

Supplemental Material

File (circres_circres-2021-320005_supp1.pdf)
File (circres_circres-2021-320005_supp2.pdf)
File (circres_circres-2021-320005_supp3.pdf)

References

1.
Bradley LA, Young A, Li H, Billcheck HO, Wolf MJ. Loss of endogenously cycling adult cardiomyocytes worsens myocardial function. Circ Res. 2021;128:155–168. doi: 10.1161/CIRCRESAHA.120.318277
2.
Heallen T, Zhang M, Wang J, Bonilla-Claudio M, Klysik E, Johnson RL, Martin JF. Hippo pathway inhibits Wnt signaling to restrain cardiomyocyte proliferation and heart size. Science. 2011;332:458–461. doi: 10.1126/science.1199010
3.
Leach JP, Heallen T, Zhang M, Rahmani M, Morikawa Y, Hill MC, Segura A, Willerson JT, Martin JF. Hippo pathway deficiency reverses systolic heart failure after infarction. Nature. 2017;550:260–264. doi: 10.1038/nature24045
4.
Mohamed TM, Stone NR, Berry EC, Radzinsky E, Huang Y, Pratt K, Ang YS, Yu P, Wang H, Tang S, et al. Chemical enhancement of in vitro and in vivo direct cardiac reprogramming. Circulation. 2017;135:978–995. doi: 10.1161/CIRCULATIONAHA.116.024692
5.
Xin M, Kim Y, Sutherland LB, Murakami M, Qi X, McAnally J, Porrello ER, Mahmoud AI, Tan W, Shelton JM, et al. Hippo pathway effector Yap promotes cardiac regeneration. Proc Natl Acad Sci USA. 2013;110:13839–13844. doi: 10.1073/pnas.1313192110
6.
von Gise A, Lin Z, Schlegelmilch K, Honor LB, Pan GM, Buck JN, Ma Q, Ishiwata T, Zhou B, Camargo FD, et al. YAP1, the nuclear target of Hippo signaling, stimulates heart growth through cardiomyocyte proliferation but not hypertrophy. Proc Natl Acad Sci USA. 2012;109:2394–2399. doi: 10.1073/pnas.1116136109
7.
Engeland K. Cell cycle arrest through indirect transcriptional repression by p53: I have a DREAM. Cell Death Differ. 2018;25:114–132. doi: 10.1038/cdd.2017.172
8.
Fischer M, Grossmann P, Padi M, DeCaprio JA. Integration of TP53, DREAM, MMB-FOXM1 and RB-E2F target gene analyses identifies cell cycle gene regulatory networks. Nucleic Acids Res. 2016;44:6070–6086. doi: 10.1093/nar/gkw523
9.
Litovchick L, Florens LA, Swanson SK, Washburn MP, DeCaprio JA. DYRK1A protein kinase promotes quiescence and senescence through DREAM complex assembly. Genes Dev. 2011;25:801–813. doi: 10.1101/gad.2034211
10.
Sadasivam S, DeCaprio JA. The DREAM complex: master coordinator of cell cycle-dependent gene expression. Nat Rev Cancer. 2013;13:585–595. doi: 10.1038/nrc3556
11.
Sadasivam S, Duan S, DeCaprio JA. The MuvB complex sequentially recruits B-Myb and FoxM1 to promote mitotic gene expression. Genes Dev. 2012;26:474–489. doi: 10.1101/gad.181933.111
12.
Angelis E, Zhao P, Zhang R, Goldhaber JI, Maclellan WR. The role of E2F-1 and downstream target genes in mediating ischemia/reperfusion injury in vivo. J Mol Cell Cardiol. 2011;51:919–926. doi: 10.1016/j.yjmcc.2011.09.012
13.
Kirshenbaum LA, Abdellatif M, Chakraborty S, Schneider MD. Human E2F-1 reactivates cell cycle progression in ventricular myocytes and represses cardiac gene transcription. Dev Biol. 1996;179:402–411. doi: 10.1006/dbio.1996.0270
14.
MacLellan WR, Garcia A, Oh H, Frenkel P, Jordan MC, Roos KP, Schneider MD. Overlapping roles of pocket proteins in the myocardium are unmasked by germ line deletion of p130 plus heart-specific deletion of Rb. Mol Cell Biol. 2005;25:2486–2497. doi: 10.1128/MCB.25.6.2486-2497.2005
15.
MacLellan WR, Xiao G, Abdellatif M, Schneider MD. A novel Rb- and p300-binding protein inhibits transactivation by MyoD. Mol Cell Biol. 2000;20:8903–8915. doi: 10.1128/MCB.20.23.8903-8915.2000
16.
Park DS, Tompkins RO, Liu F, Zhang J, Phoon CK, Zavadil J, Fishman GI. Pocket proteins critically regulate cell cycle exit of the trabecular myocardium and the ventricular conduction system. Biol Open. 2013;2:968–978. doi: 10.1242/bio.20135785
17.
Sdek P, Zhao P, Wang Y, Huang CJ, Ko CY, Butler PC, Weiss JN, Maclellan WR. Rb and p130 control cell cycle gene silencing to maintain the postmitotic phenotype in cardiac myocytes. J Cell Biol. 2011;194:407–423. doi: 10.1083/jcb.201012049
18.
Litovchick L, Sadasivam S, Florens L, Zhu X, Swanson SK, Velmurugan S, Chen R, Washburn MP, Liu XS, DeCaprio JA. Evolutionarily conserved multisubunit RBL2/p130 and E2F4 protein complex represses human cell cycle-dependent genes in quiescence. Mol Cell. 2007;26:539–551. doi: 10.1016/j.molcel.2007.04.015
19.
Ackeifi C, Swartz E, Kumar K, Liu H, Chalada S, Karakose E, Scott DK, Garcia-Ocaña A, Sanchez R, DeVita RJ, et al. Pharmacologic and genetic approaches define human pancreatic β cell mitogenic targets of DYRK1A inhibitors. JCI Insight. 2020;5:132594. doi: 10.1172/jci.insight.132594
20.
Dirice E, Walpita D, Vetere A, Meier BC, Kahraman S, Hu J, Dančík V, Burns SM, Gilbert TJ, Olson DE, et al. Inhibition of DYRK1A stimulates human β-Cell proliferation. Diabetes. 2016;65:1660–1671. doi: 10.2337/db15-1127
21.
Rachdi L, Kariyawasam D, Aïello V, Herault Y, Janel N, Delabar JM, Polak M, Scharfmann R. Dyrk1A induces pancreatic β cell mass expansion and improves glucose tolerance. Cell Cycle. 2014;13:2221–2229. doi: 10.4161/cc.29250
22.
Rachdi L, Kariyawasam D, Guez F, Aïello V, Arbonés ML, Janel N, Delabar JM, Polak M, Scharfmann R. Dyrk1a haploinsufficiency induces diabetes in mice through decreased pancreatic beta cell mass. Diabetologia. 2014;57:960–969. doi: 10.1007/s00125-014-3174-3
23.
Wang P, Alvarez-Perez JC, Felsenfeld DP, Liu H, Sivendran S, Bender A, Kumar A, Sanchez R, Scott DK, Garcia-Ocaña A, et al. A high-throughput chemical screen reveals that harmine-mediated inhibition of DYRK1A increases human pancreatic beta cell replication. Nat Med. 2015;21:383–388. doi: 10.1038/nm.3820
24.
Wang P, Karakose E, Liu H, Swartz E, Ackeifi C, Zlatanic V, Wilson J, González BJ, Bender A, Takane KK, et al. Combined inhibition of DYRK1A, SMAD, and trithorax pathways synergizes to induce robust replication in adult human beta cells. Cell Metab. 2019;29:638–652.e5. doi: 10.1016/j.cmet.2018.12.005
25.
Hille S, Dierck F, Kühl C, Sosna J, Adam-Klages S, Adam D, Lüllmann-Rauch R, Frey N, Kuhn C. Dyrk1a regulates the cardiomyocyte cell cycle via D-cyclin-dependent Rb/E2f-signalling. Cardiovasc Res. 2016;110:381–394. doi: 10.1093/cvr/cvw074
26.
Walsh S, Pontén A, Fleischmann BK, Jovinge S. Cardiomyocyte cell cycle control and growth estimation in vivo–an analysis based on cardiomyocyte nuclei. Cardiovasc Res. 2010;86:365–373. doi: 10.1093/cvr/cvq005
27.
Liberzon A, Birger C, Thorvaldsdóttir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Syst. 2015;1:417–425. doi: 10.1016/j.cels.2015.12.004
28.
Subramanian A, Tamayo P, Mootha VK, Mukherjee S, Ebert BL, Gillette MA, Paulovich A, Pomeroy SL, Golub TR, Lander ES, et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc Natl Acad Sci USA. 2005;102:15545–15550. doi: 10.1073/pnas.0506580102
29.
Talman V, Teppo J, Pöhö P, Movahedi P, Vaikkinen A, Karhu ST, Trošt K, Suvitaival T, Heikkonen J, Pahikkala T, et al. molecular atlas of postnatal mouse heart development. J Am Heart Assoc. 2018;7:e010378. doi: 10.1161/JAHA.118.010378
30.
Alkass K, Panula J, Westman M, Wu TD, Guerquin-Kern JL, Bergmann O. No evidence for cardiomyocyte number expansion in preadolescent mice. Cell. 2015;163:1026–1036. doi: 10.1016/j.cell.2015.10.035
31.
Quaife-Ryan GA, Sim CB, Ziemann M, Kaspi A, Rafehi H, Ramialison M, El-Osta A, Hudson JE, Porrello ER. Multicellular transcriptional analysis of mammalian heart regeneration. Circulation. 2017;136:1123–1139. doi: 10.1161/CIRCULATIONAHA.117.028252
32.
Cui M, Wang Z, Bassel-Duby R, Olson EN. Genetic and epigenetic regulation of cardiomyocytes in development, regeneration and disease. Development. 2018;145:dev171983. doi: 10.1242/dev.171983
33.
Henley SA, Dick FA. The retinoblastoma family of proteins and their regulatory functions in the mammalian cell division cycle. Cell Div. 2012;7:10. doi: 10.1186/1747-1028-7-10
34.
Kumar K, Wang P, Sanchez R, Swartz EA, Stewart AF, DeVita RJ. Development of Kinase-Selective, harmine-based DYRK1A inhibitors that induce pancreatic human β-Cell proliferation. J Med Chem. 2018;61:7687–7699. doi: 10.1021/acs.jmedchem.8b00658
35.
Thompson BJ, Bhansali R, Diebold L, Cook DE, Stolzenburg L, Casagrande AS, Besson T, Leblond B, Désiré L, Malinge S, et al. DYRK1A controls the transition from proliferation to quiescence during lymphoid development by destabilizing Cyclin D3. J Exp Med. 2015;212:953–970. doi: 10.1084/jem.20150002
36.
Mort RL, Ford MJ, Sakaue-Sawano A, Lindstrom NO, Casadio A, Douglas AT, Keighren MA, Hohenstein P, Miyawaki A, Jackson IJ. Fucci2a: a bicistronic cell cycle reporter that allows Cre mediated tissue specific expression in mice. Cell Cycle. 2014;13:2681–2696. doi: 10.4161/15384101.2015.945381
37.
O’Gara PT, Kushner FG, Ascheim DD, Casey DE, Chung MK, de Lemos JA, Ettinger SM, Fang JC, Fesmire FM, Franklin BA, et al. 2013 ACCF/AHA guideline for the management of ST-elevation myocardial infarction: executive summary: a report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J Am Coll Cardiol. 2013;61:485–510. doi: 10.1016/j.jacc.2012.11.018
38.
Richardson WJ, Clarke SA, Quinn TA, Holmes JW. Physiological Implications of Myocardial Scar Structure. Compr Physiol. 2015;5:1877–1909. doi: 10.1002/cphy.c140067
39.
D’Uva G, Aharonov A, Lauriola M, Kain D, Yahalom-Ronen Y, Carvalho S, Weisinger K, Bassat E, Rajchman D, Yifa O, et al. ERBB2 triggers mammalian heart regeneration by promoting cardiomyocyte dedifferentiation and proliferation. Nat Cell Biol. 2015;17:627–638. doi: 10.1038/ncb3149
40.
Jopling C, Sleep E, Raya M, Martí M, Raya A, Izpisúa Belmonte JC. Zebrafish heart regeneration occurs by cardiomyocyte dedifferentiation and proliferation. Nature. 2010;464:606–609. doi: 10.1038/nature08899
41.
Senyo SE, Lee RT, Kühn B. Cardiac regeneration based on mechanisms of cardiomyocyte proliferation and differentiation. Stem Cell Res. 2014;13:532–541. doi: 10.1016/j.scr.2014.09.003
42.
Wang WE, Li L, Xia X, Fu W, Liao Q, Lan C, Yang D, Chen H, Yue R, Zeng C, et al. Dedifferentiation, proliferation, and redifferentiation of adult mammalian cardiomyocytes after ischemic injury. Circulation. 2017;136:834–848. doi: 10.1161/CIRCULATIONAHA.116.024307
43.
Zhang Y, Li TS, Lee ST, Wawrowsky KA, Cheng K, Galang G, Malliaras K, Abraham MR, Wang C, Marbán E. Dedifferentiation and proliferation of mammalian cardiomyocytes. PLoS One. 2010;5:e12559. doi: 10.1371/journal.pone.0012559
44.
Tasic B, Miyamichi K, Hippenmeyer S, Dani VS, Zeng H, Joo W, Zong H, Chen-Tsai Y, Luo L. Extensions of MADM (mosaic analysis with double markers) in mice. PLoS One. 2012;7:e33332. doi: 10.1371/journal.pone.0033332
45.
Zong H, Espinosa JS, Su HH, Muzumdar MD, Luo L. Mosaic analysis with double markers in mice. Cell. 2005;121:479–492. doi: 10.1016/j.cell.2005.02.012
46.
Derks W, Bergmann O. Polyploidy in cardiomyocytes: roadblock to heart regeneration? Circ Res. 2020;126:552–565. doi: 10.1161/CIRCRESAHA.119.315408
47.
Frost D, Meechoovet B, Wang T, Gately S, Giorgetti M, Shcherbakova I, Dunckley T. β-carboline compounds, including harmine, inhibit DYRK1A and tau phosphorylation at multiple Alzheimer’s disease-related sites. PLoS One. 2011;6:e19264. doi: 10.1371/journal.pone.0019264
48.
Patel K, Gadewar M, Tripathi R, Prasad SK, Patel DK. A review on medicinal importance, pharmacological activity and bioanalytical aspects of beta-carboline alkaloid “Harmine”. Asian Pac J Trop Biomed. 2012;2:660–664. doi: 10.1016/S2221-1691(12)60116-6
49.
Boni J, Rubio-Perez C, López-Bigas N, Fillat C, de la Luna S. The DYRK family of kinases in cancer: molecular functions and therapeutic opportunities. Cancers (Basel). 2020;12:E2106. doi: 10.3390/cancers12082106
50.
Grebe C, Klingebiel TM, Grau SP, Toischer K, Didié M, Jacobshagen C, Dullin C, Hasenfuss G, Seidler T. Enhanced expression of DYRK1A in cardiomyocytes inhibits acute NFAT activation but does not prevent hypertrophy in vivo. Cardiovasc Res. 2011;90:521–528. doi: 10.1093/cvr/cvr023
51.
Chen JY, Lin JR, Tsai FC, Meyer T. Dosage of Dyrk1a shifts cells within a p21-cyclin D1 signaling map to control the decision to enter the cell cycle. Mol Cell. 2013;52:87–100. doi: 10.1016/j.molcel.2013.09.009
52.
Gwack Y, Sharma S, Nardone J, Tanasa B, Iuga A, Srikanth S, Okamura H, Bolton D, Feske S, Hogan PG, et al. A genome-wide Drosophila RNAi screen identifies DYRK-family kinases as regulators of NFAT. Nature. 2006;441:646–650. doi: 10.1038/nature04631
53.
Liu H, Wang K, Chen S, Sun Q, Zhang Y, Chen L, Sun X. NFATc1 phosphorylation by DYRK1A increases its protein stability. PLoS One. 2017;12:e0172985. doi: 10.1371/journal.pone.0172985
54.
Duchon A, Herault Y. DYRK1A, a dosage-sensitive gene involved in neurodevelopmental disorders, is a target for drug development in down syndrome. Front Behav Neurosci. 2016;10:104. doi: 10.3389/fnbeh.2016.00104
55.
Di Vona C, Bezdan D, Islam AB, Salichs E, López-Bigas N, Ossowski S, de la Luna S. Chromatin-wide profiling of DYRK1A reveals a role as a gene-specific RNA polymerase II CTD kinase. Mol Cell. 2015;57:506–520. doi: 10.1016/j.molcel.2014.12.026

eLetters(0)

eLetters should relate to an article recently published in the journal and are not a forum for providing unpublished data. Comments are reviewed for appropriate use of tone and language. Comments are not peer-reviewed. Acceptable comments are posted to the journal website only. Comments are not published in an issue and are not indexed in PubMed. Comments should be no longer than 500 words and will only be posted online. References are limited to 10. Authors of the article cited in the comment will be invited to reply, as appropriate.

Comments and feedback on AHA/ASA Scientific Statements and Guidelines should be directed to the AHA/ASA Manuscript Oversight Committee via its Correspondence page.

Information & Authors

Information

Published In

Go to Circulation Research
Go to Circulation Research
Circulation Research
Pages: 1345 - 1361
PubMed: 35369706

Versions

You are viewing the most recent version of this article.

History

Received: 5 August 2021
Revision received: 18 March 2022
Accepted: 22 March 2022
Published online: 4 April 2022
Published in print: 29 April 2022

Permissions

Request permissions for this article.

Keywords

  1. harmine
  2. hyperplasia
  3. myocardial infarction
  4. phosphorylation
  5. tyrosine

Subjects

Authors

Affiliations

Alexander Young
Department of Medicine (A.Y., L.A.B., E.F., H.O.B., M.J.W.), University of Virginia, Charlottesville.
Robert M. Berne Cardiovascular Research Center (A.Y., L.A.B., H.O.B., M.J.W.), University of Virginia, Charlottesville.
Leigh A. Bradley
Department of Medicine (A.Y., L.A.B., E.F., H.O.B., M.J.W.), University of Virginia, Charlottesville.
Robert M. Berne Cardiovascular Research Center (A.Y., L.A.B., H.O.B., M.J.W.), University of Virginia, Charlottesville.
Department of Medicine (A.Y., L.A.B., E.F., H.O.B., M.J.W.), University of Virginia, Charlottesville.
Helen O. Bilcheck
Department of Medicine (A.Y., L.A.B., E.F., H.O.B., M.J.W.), University of Virginia, Charlottesville.
Robert M. Berne Cardiovascular Research Center (A.Y., L.A.B., H.O.B., M.J.W.), University of Virginia, Charlottesville.
Departments of Biomedical Engineering (S.T., J.J.S.), University of Virginia, Charlottesville.
Jeffrey J. Saucerman
Departments of Biomedical Engineering (S.T., J.J.S.), University of Virginia, Charlottesville.
Biochemistry and Molecular Genetics (S.B.), University of Virginia, Charlottesville.
Department of Medicine (A.Y., L.A.B., E.F., H.O.B., M.J.W.), University of Virginia, Charlottesville.
Robert M. Berne Cardiovascular Research Center (A.Y., L.A.B., H.O.B., M.J.W.), University of Virginia, Charlottesville.

Notes

Supplemental Material is available at Supplemental Material
For Sources of Funding and Disclosures, see page 1359.
Correspondence to: Matthew J. Wolf, MD, PhD, Division of Cardiology, University of Virginia, Medical Research Building 5 (MR5), Rm G213, 415 Lane Rd, Charlottesville, VA 22908. Email [email protected]

Disclosures

Disclosures None.

Sources of Funding

Research reported in this publication was supported by the University of Virginia Center of Excellence in Cardiovascular Genetics and the National Heart, Lung, And Blood Institute of the National Institutes of Health under Award Number R01HL158718 (M.J. Wolf.). The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.”

Metrics & Citations

Metrics

Citations

Download Citations

If you have the appropriate software installed, you can download article citation data to the citation manager of your choice. Select your manager software from the list below and click Download.

  1. Recent Insights into Endogenous Mammalian Cardiac Regeneration Post-Myocardial Infarction, International Journal of Molecular Sciences, 25, 21, (11747), (2024).https://doi.org/10.3390/ijms252111747
    Crossref
  2. Increased Myocardial MAO-A, Atrogin-1, and IL-1β Expression in Transgenic Mice with Pancreatic Carcinoma—Benefit of MAO-A Inhibition for Cardiac Cachexia, Biomedicines, 12, 9, (2009), (2024).https://doi.org/10.3390/biomedicines12092009
    Crossref
  3. Leucettinib-21, a DYRK1A Kinase Inhibitor as Clinical Drug Candidate for Alzheimer’s Disease and Down Syndrome, Journal of Alzheimer's Disease, 101, s1, (S95-S113), (2024).https://doi.org/10.3233/JAD-240078
    Crossref
  4. Application of New Lineage Tracing Techniques in Cardiovascular Development and Physiology, Circulation Research, 134, 4, (445-458), (2024)./doi/10.1161/CIRCRESAHA.123.323179
    Abstract
  5. Increased dosage of DYRK1A leads to congenital heart defects in a mouse model of Down syndrome, Science Translational Medicine, 16, 731, (2024).https://doi.org/10.1126/scitranslmed.add6883
    Crossref
  6. The de novo purine synthesis enzyme Adssl1 promotes cardiomyocyte proliferation and cardiac regeneration, Science Signaling, 17, 860, (2024).https://doi.org/10.1126/scisignal.adn3285
    Crossref
  7. Targeting the DYRK1A kinase prevents cancer progression and metastasis and promotes cancer cells response to G1/S targeting chemotherapy drugs, npj Precision Oncology, 8, 1, (2024).https://doi.org/10.1038/s41698-024-00614-w
    Crossref
  8. Discovery and Functional Characterization of a Potent, Selective, and Metabolically Stable PROTAC of the Protein Kinases DYRK1A and DYRK1B, Journal of Medicinal Chemistry, 67, 19, (17259-17289), (2024).https://doi.org/10.1021/acs.jmedchem.4c01130
    Crossref
  9. The role of serine/threonine protein kinases in cardiovascular disease and potential therapeutic methods, Biomedicine & Pharmacotherapy, 177, (117093), (2024).https://doi.org/10.1016/j.biopha.2024.117093
    Crossref
  10. Insights from the protein interaction Universe of the multifunctional “Goldilocks” kinase DYRK1A, Frontiers in Cell and Developmental Biology, 11, (2023).https://doi.org/10.3389/fcell.2023.1277537
    Crossref
  11. See more
Loading...

View Options

View options

PDF and All Supplements

Download PDF and All Supplements

PDF/EPUB

View PDF/EPUB
Login options

Check if you have access through your login credentials or your institution to get full access on this article.

Personal login Institutional Login
Purchase Options

Purchase this article to access the full text.

Purchase access to this article for 24 hours

Inhibition of DYRK1a Enhances Cardiomyocyte Cycling After Myocardial Infarction
Circulation Research
  • Vol. 130
  • No. 9

Purchase access to this journal for 24 hours

Circulation Research
  • Vol. 130
  • No. 9
Restore your content access

Enter your email address to restore your content access:

Note: This functionality works only for purchases done as a guest. If you already have an account, log in to access the content to which you are entitled.

Figures

Tables

Media

Share

Share

Share article link

Share

Comment Response