Abstract
Extracellular signal-regulated kinases 1 and 2 (ERK1/2) are central mediators of cardiac hypertrophy and are discussed as potential therapeutic targets. However, direct inhibition of ERK1/2 leads to exacerbated cardiomyocyte death and impaired heart function. We have previously identified ERKThr188 autophosphorylation as a regulatory phosphorylation of ERK1/2 that is a key factor in cardiac hypertrophy. Here, we investigated whether interference with ERKThr188 phosphorylation permits the impairment of ERK1/2-mediated cardiac hypertrophy without increasing cardiomyocyte death. The impact of ERKThr188 phosphorylation on cardiomyocyte hypertrophy and cell survival was analyzed in isolated cells and in mice using the mutant ERK2T188A, which is dominant-negative for ERKThr188 signaling. ERK2T188A efficiently attenuated cardiomyocyte hypertrophic responses to phenylephrine and to chronic pressure overload, but it affected neither antiapoptotic ERK1/2 signaling nor overall physiological cardiac function. In contrast to its inhibition of pathological hypertrophy, ERK2T188A did not interfere with physiological cardiac growth occurring with age or upon voluntary exercise. A preferential role of ERKThr188 phosphorylation in pathological types of hypertrophy was also seen in patients with aortic valve stenosis: ERKThr188 phosphorylation was increased 8.5 ± 1.3-fold in high-gradient, rapidly progressing cases (≥40 mmHg gradient), whereas in low-gradient, slowly progressing cases, the increase was not significant. Because interference with ERKThr188 phosphorylation (i) inhibits pathological hypertrophy and (ii) does not impair antiapoptotic ERK1/2 signaling and because ERKThr188 phosphorylation shows strong prevalence for aortic stenosis patients with rapidly progressing course, we conclude that interference with ERKThr188 phosphorylation offers the possibility to selectively address pathological types of cardiac hypertrophy.
Keywords: MAPK, apoptosis, GPCRs
Cardiac hypertrophy has been identified as an independent risk factor of diastolic dysfunction, heart failure, arrhythmias, and sudden death (1). Various triggers initiate cardiac hypertrophy, but the type of trigger has been identified as decisive for an adaptive vs. a maladaptive outcome of cardiac hypertrophy. Whereas cardiovascular diseases such as hypertension, aortic stenosis, or myocardial infarction generally induce a pathological type of hypertrophy, chronic physical exercise or postnatal cardiac growth entail a compensatory and physiological type of hypertrophy. In contrast to physiological hypertrophy, pathological hypertrophy is characterized by accumulation of interstitial collagen and cell death, which both have been shown to contribute to increased cardiovascular risk (2–5). Therefore, it would be of great therapeutic interest to prevent pathological hypertrophy; however, such prevention requires a fine balance between inhibition of maladaptive and preservation of compensatory adaptive hypertrophy.
Hypertrophic stimuli are mediated via several intracellular signaling cascades that ultimately affect cardiac gene transcription. A central signaling cascade, which has been shown to promote the development of cardiac hypertrophy, is the mitogen-activated protein (MAP) kinase cascade consisting of the kinases rapid activation of fibrosarcoma (Raf), MAP/ERK kinase (MEK1/2), and ERK1/2. However, inhibition of ERK1/2-mediated hypertrophy using dominant-negative mutants or MEK inhibitors in isolated cardiomyocytes and in mice was accompanied by increased cardiomyocyte death (6–13). Therefore, to date, it has not been possible to exploit the potential of the ERK1/2 cascade as a therapeutic target in cardiac hypertrophy.
Here, we describe a possibility to selectively impair hypertrophic ERK1/2-mediated functions while leaving the antiapoptotic effects of ERK1/2 intact. We have previously discovered an ERK2Thr188 autophosphorylation (ERK2Thr188 corresponds to Thr208 in ERK1; subsequently referred to as ERKThr188 phosphorylation) that appears to be essential for the induction of ERK1/2-mediated cardiac hypertrophy in response to various stimuli (14, 15). Because inhibition of ERK1/2 is an effective strategy to prevent cardiac hypertrophy (6, 8, 12), we aimed to investigate whether specific interference with ERKThr188 phosphorylation had an effect on hypertrophy and on apoptosis in vivo. We further investigated the occurrence of ERKThr188 phosphorylation in patients with different forms of aortic stenosis. Our data suggest a clinical relevance of this autophosphorylation and further indicate that interference with ERKThr188 phosphorylation may be a selective therapeutic strategy in pathological ERK1/2-mediated cardiac hypertrophy.
Results
Interference with ERKThr188 Phosphorylation Attenuates ERK-Mediated Cardiomyocyte Hypertrophy but Not Cell Survival.
First, we assessed whether interference with ERKThr188 phosphorylation may be a strategy to attenuate cardiomyocyte hypertrophy without a simultaneous exaggeration of apoptotic cell death. For this study, we used an ERK2 mutant that cannot be phosphorylated at threonine 188, ERK2T188A (T188A). Interestingly, overexpression of ERK2T188A did not affect catalytic ERK activity, as monitored by MEK1/2-mediated phosphorylation of ERK1/2 at the so-called TEY motif [pERK1/2(TEY); Fig. S1 A–C] (6, 7). To exclude an overexpression artifact, we compared ERK2T188A with similar expression levels of ERK2 wild-type (WT) [ERK2T188T (T188T)] (Fig. S1D).
ERK2T188A overexpression in neonatal rat cardiomyocytes (NRCMs) significantly attenuated phenylephrine-induced cardiomyocyte hypertrophy, as assessed by incorporation of [3H]isoleucine (Fig. 1A) and by cell size analysis (representative cells in Fig. 1B and quantification of cell size in Fig. S1E) (16, 17). Strikingly, ERK2T188A did not enhance H2O2-induced apoptotic cell death, as determined by terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) (Fig. 1C) and procaspase-3 levels (Fig. S1F) (18–20). In contrast, overexpression of ERK2T188T affected neither the hypertrophic (Fig. S1G) nor apoptotic response of NRCMs (Fig. S1H). Thus, expression of ERK2T188A causes an efficient inhibition of cardiomyocyte hypertrophy but has no effect on apoptosis.
Fig. 1.
Interference with ERKThr188 phosphorylation does not abrogate antiapoptotic effects of ERK. (A–C) NRCMs were transduced with Flag-tagged ERK2T188T (T188T) or ERK2T188A (T188A) and stimulated with phenylephrine (PHE) (4 µM; 24 h) (A and B) or with hydrogen peroxide (H2O2) (100 µM; 1 h) (C) as indicated. (A) [3H]Isoleucine incorporation assay (n = 9). *P < 0.001 vs. all other conditions. (B) Phalloidin staining. (Scale bar: 10 μm.) (C) TUNEL assay (n = 6–8; at least 150 cells per experiment and group). *P < 0.01 vs. unstimulated controls. (D–F) NRCMs were pretreated with PD98059 (30 µM; 60 min) and stimulated with phenylephrine (4 µM; 24 h) (D and E) or with H2O2 (100 µM; 1 h) (F) as indicated. (D) [3H]Isoleucine incorporation assay (n = 6). *P < 0.001 vs. all other conditions; not significant (n.s.) vs. unstimulated controls. (E) Phalloidin staining. (Scale bar: 10 μm.) (F) TUNEL assay (n = 7; at least 150 cells per experiment and group). *P < 0.01 vs. control; #P < 0.01 vs. all other conditions. (G) Analyses of procaspase-3 levels in NRCMs with (siERK1/2) or without (CON and CON siRNA) knock down of endogenous ERK1 and ERK2 expression. NRCMs were transfected with siERK1/2 or CON siRNA and adenovirally transduced with Flag-tagged ERK2T188T (T188T), Flag-tagged ERK2T188A (T188A), or GFP (CON). (n = 5–6). *P < 0.05. Shown are representative immunoblots.
These results are particularly interesting because, as reported in the Introduction, inhibition of the catalytic ERK1/2 activity (e.g., by using the MEK1/2 inhibitor PD98059) attenuates cardiomyocyte hypertrophy in analogous assays (Fig. 1 D and E and Fig. S1I) (21). In contrast to ERK2T188A, PD98059 leads to enhanced H2O2-induced apoptotic cell death (Fig. 1F and Fig. S1J).
To ensure that ERK2T188A overexpression does indeed not affect apoptotic ERK1/2 signaling, we next analyzed its effect on apoptosis at endogenous expression levels. For these experiments, we used siRNAs to reduce endogenous ERK1/2 expression (Fig. 1G, middle blot). Like ERK1/2 inhibition with PD98059 (Fig. S1K), knock down of ERK1/2 activated the programmed cell death machinery, as evidenced by decreased procaspase-3 levels (Fig. 1G, upper blot and graph). Accordingly, subsequent reexpression of WT ERK2 (T188T) reversed the apoptotic effect of the ERK1/2 knockdown (i.e., normalized the procaspase-3 expression levels; Fig. 1G), confirming the antiapoptotic role of endogenously expressed ERK1/2. Interestingly, reexpression of ERKT188A normalized procaspase-3 expression levels as efficiently as did reexpression of ERKT188T (Fig. 1G).
Taken together, our experiments confirmed that ERK1/2, indeed, mediate both cardiomyocyte hypertrophy and survival in NRCMs. Our experiments further suggest that interference with ERKThr188 phosphorylation—in contrast to catalytic inhibition of ERK1/2—might be a strategy to specifically target ERK-mediated hypertrophy without impairing ERK-mediated cell survival.
Inhibition of Hypertrophy by ERK2T188A Does Not Exacerbate Apoptosis in Vivo.
We then sought to confirm these findings obtained in isolated NRCMs in intact animals. For this study, we used FVB/N mice with transgenic cardiac expression of ERK2T188A (T188A), and as controls, we used nontransgenic WT mice, as well as mice transgenic for ERK2 WT (T188T). ERK2 constructs were transgenically expressed at similar levels as the overall expression level of endogenous ERK1 and ERK2 (Fig. S2A). These mice were subjected to chronic pressure overload stress induced by transverse aortic constriction (TAC) to induce hypertrophy and apoptosis (22). Aortic pressure gradients were the same in the three groups: 70.7 ± 4.3 for WT, 72.5 ± 6.0 for ERK2T188T, and 71.9 ± 2.6 for ERK2T188A.
Transgenic cardiac expression of ERK2T188A significantly reduced the hypertrophic response to TAC compared with control mice (WT or T188T), as measured by heart weight-to-tibia length ratios (Fig. 2A), LV wall thickness (Table S1), and cardiomyocyte size (Fig. 2B and Fig. S2B). Furthermore, echocardiographic analyses showed normal cardiac function in ERK2T188A-expressing mice (Table S1). Sirius red staining of histological sections revealed that ERK2T188A-overexpressing mice developed even less interstitial fibrosis in response to LV pressure overload (Fig. 2 C and D). Interestingly, whereas ERK-mediated cardiac hypertrophy was efficiently inhibited in ERKT188A transgenic mice, the extent of apoptosis induced by TAC was comparable in all three genotypes (Fig. 2 E and F).
Fig. 2.
Inhibition of hypertrophy by phosphorylation-deficient ERK2T188A does not exacerbate apoptosis in vivo. (A–F) WT mice and transgenic mice expressing ERK2T188T (T188T) or ERK2T188A (T188A) without (CON) or with surgically induced TAC (6 wk). Analysis of heart weight-to-tibia length ratios (A), histological H&E-stained sections (scale bar: 50 μm) (B), quantification (C) and representative images (D) of interstitial fibrosis in Sirius red-stained sections of ventricular myocardium (scale bar: 200μm), caspase-3 activity (E), and TUNEL-positive cell nuclei (F) (n = 5–14 mice per group). *P < 0.01; #P < 0.01 vs. unstimulated controls.
Thus, interference with ERKThr188 phosphorylation permits efficient inhibition of TAC-induced cardiac hypertrophy and cardiac remodeling, with no adverse effects on cell survival and cardiac function.
ERK2T188A Prevents Activation of Nuclear, Hypertrophic but Not Cytosolic Antiapoptotic ERK1/2 Signaling.
To understand the mechanism that enables ERK2T188A to distinguish between the different ERK functions (i.e., hypertrophy vs. apoptosis), we compared its effects on ERK1/2 target phosphorylation, looking at both cytosolic and nuclear targets. As ERK1/2 targets, which mediate the antiapoptotic effects of ERK1/2 and are, thus, primarily located within the cytosol, we investigated p90 ribosomal S6 kinase (p90RSK) and B-cell lymphoma 2-interacting mediator of cell death (BIM) (23). As a nuclear target contributing to the transcriptional hypertrophic effects of ERK1/2, we analyzed ERK1/2-mediated phosphorylation of the nuclear ETS (E twenty-six)-like 1 transcription factor (Elk1) (24). Both PD98059 and ERK2T188A expression inhibited phenylephrine-induced phosphorylation of nuclear Elk1, but only PD98059 interfered with phenylephrine- or H2O2-induced phosphorylation of cytosolic p90RSK and BIM (representative blots in Fig. 3 A and B; quantification in Fig. S3 A–F and G–L).
Fig. 3.
Interference with ERKThr188 phosphorylation specifically attenuates nuclear ERK-target activation. (A and B) Western blot analyses of NRCMs adenovirally transduced with Flag-tagged WT ERK2T188T (T188T) or Flag-tagged phosphorylation-deficient ERK2T188A (T188A) or pretreated with PD98059 (30 µM; 60 min) as indicated. Cells were stimulated without (CON) or with phenylephrine (PHE) (4 µM; 10 min) or H2O2 (100 µM or 400 µM; 1 h). (A and B) Analysis of nuclear (Elk1) and cytosolic (p90RSK and BIM) ERK1/2 targets using phospho-specific antibodies directed against ERK1/2 phosphorylation sites of these target proteins. (C) Representative confocal images of NRCMs transduced with YFP-tagged T188T or YFP-T188A stimulated without or with phenylephrine (4 µM; 10 min) or H2O2 (100 µM; 1 h). Nuclei were stained with DAPI. (Scale bar: 20 μm.) (D) Immunoblot analysis of ERKThr188 phosphorylation of mock-transfected NRCMs stimulated with phenylephrine (4 µM; 10 min) or H2O2 (100 µM; 60 min). All experiments were reproduced at least four times.
These experiments suggest that interference with ERKThr188 phosphorylation (in contrast to inhibition of ERK1/2 activity by PD98059) selectively inhibits nuclear ERK targets, which may be explained by a prominent role of ERKThr188 phosphorylation in nuclear accumulation of ERK1/2: (i) the ERK2T188A mutant did not—unlike WT ERK2—accumulate within the nucleus in response to a hypertrophic stimulus (Fig. 3C); and (ii) WT ERK2 did not accumulate in the nucleus in response to H2O2 (Fig. 3C), which activated ERK1/2 [i.e., pERK1/2(TEY)] but did not induce ERKThr188 phosphorylation (representative immunoblots in Fig. 3D and quantification in Fig. S3 M–P).
ERK2T188A Does Not Affect Physiological Growth of the Heart.
Even though cardiac hypertrophy is a risk factor for heart failure, therapeutic prevention of cardiac hypertrophy in patients should ideally impair neither the heart’s normal postnatal growth nor its adequate growth in response to exercise (3, 4). Therefore, we next investigated the influence of ERK2T188A expression on physiological growth of the heart. For this analysis, we assessed cardiac growth (i) in mice until the age of 9 mo and (ii) in mice that exercised voluntarily in a running wheel for a period of 3 wk.
Postnatal cardiac growth up to 9 mo, as well as exercise-driven cardiac growth, was indistinguishable between ERK2T188A-expressing and control mice (WT and T188T), as assessed by ventricular wall thicknesses (Fig. 4A), cardiomyocyte size (representative sections in Fig. 4B and quantification in Fig. 4C), and heart weight-to-tibia length or heart weight-to-body weight ratios, respectively (Fig. S4 A and B). Furthermore, echocardiographic analysis of heart function (Table S2) and histological analysis (representative sections in Fig. 4D and quantification in Fig. 4E) did not reveal any pathological changes in ERK2T188A-expressing mice at the age of 9 mo or after 3 wk of running-wheel exercise. Thus, interference with ERKThr188 phosphorylation does not appear to interfere with physiological and healthy growth of the heart. In line with these findings, ERKThr188 phosphorylation, which is necessary for ERK1/2-mediated cardiac hypertrophy (14), was elevated only in TAC-operated mice but not in exercised mouse hearts (Fig. 4F).
Fig. 4.

ERKThr188 phosphorylation is not involved in physiological hypertrophy. (A–E) Echocardiographically determined interventricular wall thickness (IVS) (A), representative histological H&E-stained sections (scale bar: 50 μm) (B), quantification of cross-sectional cardiomyocyte areas (C), corresponding Sirius red-stained sections (scale bar: 200 μm) (D), and quantification of interstitial fibrosis of ventricular myocardium (E) of WT mice and transgenic mice with cardiac expression of wild-type ERK2T188T (T188T) or phosphorylation-deficient ERK2T188A (T188A) at the age of 8 wk (CON), at the age of 9 mo (9 MON), and after 3 wk of voluntary running-wheel exercise (RUN) (n = 7–15). *P < 0.05 vs. CON. (F) Representative immunoblots of ERK1/2 phosphorylations in heart lysates of 8-wk-old WT mice, heart lysates of mice after 3 wk of voluntary running-wheel exercise (RUN), or after 6 wk of pressure overload (TAC) of n = 9 experiments. Bar graph shows the quantification of pERK(Thr188) from n = 9 experiments. *P < 0.001 vs. all other conditions.
Differential ERKThr188 Phosphorylation in Patients with High- and Low-Gradient Aortic Stenosis.
To further characterize the involvement of ERKThr188 phosphorylation in cardiac hypertrophy, we analyzed septum samples of patients with aortic valve stenosis. These patients were subgrouped into high (≥40 mmHg) or low (<40 mmHg) transvalvular pressure gradients, which showed low or high degrees of interstitial fibrosis, respectively (Fig. 5A) (25). Despite different transvalvular pressure gradients, both groups had a similar extent of hypertrophy at the time of surgery, as determined by echocardiographic analysis of heart weight (Table S3), diastolic wall thickness (Table S3), and histological analysis of cardiomyocyte cell area (Fig. 5B). At the time of surgery, ejection fractions and aortic valve areas were also undistinguishable in both patient groups (Table S3). Furthermore, canonical ERK1/2 activation (pERK[TEY]; Fig. 5C) was similar in both patient groups. Interestingly, however, ERKThr188 phosphorylation was >eightfold increased in patients with high transvalvular pressure gradients but not significantly (<twofold) enhanced in patients with low gradients (Fig. 5C). Of these patients, 10/12 with high pressure gradients, but only 1/8 with low gradients, had a rapid course of disease (Fig. 5D and Table S3). This indicates a close correlation between pressure gradients, rapid progression, and ERKThr188 phosphorylation, even though the extent of ventricular hypertrophy at the time of surgery was similar in all patients (Table S3, Fig. 5B, and Fig. S5). Thus, in contrast to similar canonical ERK1/2 activation in all samples, increased ERKThr188 phosphorylation was seen specifically in the cases with rapid progression and high gradients (Fig. 5C). This indicates that ERKThr188 phosphorylation depends on the type of hypertrophic trigger and seems to be correlated with the rapidity of disease development (Fig. 5D), rather than with its final extent of hypertrophy (Fig. S5).
Fig. 5.
Differential ERKThr188 phosphorylation in patients with high- and low-gradient aortic stenosis. Analysis of septum samples of patients with aortic valve stenosis subgrouped in patients with high (≥40 mmHg; high-gradient) or low (<40 mmHg; low-gradient) transvalvular pressure gradients. (A) Masson–Goldner staining of interstitial fibrosis. (Scale bar: 200 μm.) (B) Analysis of cardiomyocyte cell area. (C) Immunoblot analysis of ERKThr188 phosphorylation (pERK[Thr188]) and ERK1/2 activation in patients with low and high gradients compared with nonfailing control myocardium, as monitored by canonical ERK1/2 phosphorylation (pERK[TEY]) (n = 5–12 samples per group). *P < 0.05. (D) Speed of disease progression in patients with high- or low-gradient aortic stenosis. Rapid progress is defined by an increase in peak aortic flow velocity ≥0.3 m⋅s−1y−1.
Taken together, the analyses of patients with aortic valve stenosis further substantiate the hypothesis that ERKThr188 phosphorylation is not increased in all types of cardiac hypertrophy but is dependent on the type of hypertrophic trigger and is, in particular, associated with high pressure load in mice and with rapidly progressing forms of aortic stenosis in patients.
Discussion
Pathological hypertrophy is a key risk factor for myocardial infarction, arrhythmia, and sudden death, whereas physiological growth of the heart in response to exercise is generally accepted to be protective, i.e., to preserve or even improve heart function (1, 4, 26). Although maladaptive hypertrophy involves cell death, collagen deposits, fibrosis, and ventricular stiffening, these hallmarks of pathological remodeling are absent in the “athlete’s” heart (1, 27). Although there is an ongoing debate whether heart failure-associated ventricular hypertrophy has a compensatory role in heart failure patients, evidence is increasing that inhibition of pathological hypertrophy may be beneficial (3, 14, 15, 28–30).
On the molecular level, pathological cardiac hypertrophy is mediated by various proteins including, Gαq, extracellular signal-regulated kinases, protein kinase C, Ca2+/calmodulin, and nuclear factor of activated T cells. Even though these proteins are associated with pathological hypertrophy, many of them also play vital roles in normal cardiomyocyte functions (4–7, 31, 32). Therefore, it is a challenging but critical task to identify features within pathological hypertrophic signaling pathways that allow to specifically target maladaptive functions of candidate proteins.
The ERK1/2 cascade has been reported to be activated and to be involved in diverse instances of cardiac hypertrophy and heart failure including cardiomyopathies, both in rodents and humans, and has, thus, been considered as a putative therapeutic target in pathological hypertrophy (6, 33–35). However, many mouse models point toward a pivotal role of ERK1/2 in the prevention of apoptotic cell death: cell death is significantly increased in mice with cardiac overexpression of dominant-negative Raf1, in mice treated with MEK1/2-inhibitors, and in ERK1−/−ERK2+/− mice (11, 12, 36), whereas overexpression of constitutively active MEK1 increases cell survival (37, 38). Therefore, inhibition of ERK1/2 activity is detrimental for the heart and, thus, cannot be used to target cardiac hypertrophy.
Our present study identifies ERKThr188 phosphorylation as a tool to differentiate between these distinct functions of ERK1/2 in the heart: increased cell death as a maladaptive and reduced hypertrophy as a potentially beneficial consequence of ERK1/2 inhibition. With the help of ERK2T188A, the impact of ERKThr188 phosphorylation on cardiomyocyte hypertrophy in vitro and in vivo was demonstrated. Expression of ERK2T188A attenuated the hypertrophic responses of pathological stimuli such as phenylephrine, angiotensin II, isoprenaline, and chronic pressure overload (14, 15). However, protective, antiapoptotic ERK1/2 signaling in vitro and in vivo (Fig. 1C and Fig. 2 E and F), as well as cardiac function (Tables S1 and S2), were not disturbed by ERK2T188A expression. Therefore, ERK2T188A appears to display its dominant-negative effect on nuclear ERK1/2 signaling via dimerization with endogenous WT ERK1/2 and by retaining them within the cytosol. This hypothesis is supported by the following: (i) the ability of ERK2T188A to dimerize with ERK2 (ref. 14 and Fig. S6 A and B); (ii) the ability of ERK2 to heterodimerize with ERK1 (ref. 15 and Fig. S6 A and B); (iii) the cytosolic retention of YFP-tagged WT ERK2 in the presence of ERK2T188A (Fig. S6C); and (iv) earlier observations that both ERK dimerization and ERKThr188 phosphorylation are required for nuclear accumulation (15, 39).
Interestingly, ERKThr188 phosphorylation may be controlled by β-arrestins, interaction partners of ERK1/2. Overexpression of β-arrestin2 prevented the interaction of ERK2 with Gβγ subunits, an interaction that is necessary to induce ERK2Thr188 phosphorylation (Fig. S6 F–I) (40, 41). β-Arrestin2 may, thus, be an endogenous competitor of ERKThr188 signaling.
ERKThr188 phosphorylation seems to be a prerequisite for nuclear accumulation of ERK1/2, and, therefore, ERKThr188 phosphorylation opens up a possibility to interfere with ERK-mediated cardiac hypertrophy without inhibiting catalytic ERK1/2 activity. Targeting of ERKThr188 phosphorylation may, therefore, allow to address the Raf/MEK/ERK1/2 cascade as a promising therapeutic target in cardiac hypertrophy. Of interest in this context, interference with ERKThr188 phosphorylation by expression of ERK2T188A did not interfere with physiological (postnatal or exercise-driven) growth of the heart, and even long-term interference with ERKThr188 phosphorylation did not reveal any cardiac adverse effects (Fig. 4 and Table S2). However, because this mechanism appears to be of a general nature, it will be important to delineate it in more detail in both cardiac and noncardiac cells and tissues.
The analysis of septal samples of patients with aortic valve stenosis further confirms that ERKThr188 phosphorylation is a marker for pathological hypertrophy: ERKThr188 phosphorylation was significantly increased in patients with high pressure gradients and with a rapid course of the disease (Fig. 5 C and D).
Interestingly, in patients with aortic valve stenosis, we found a negative correlation for fibrosis and ERKThr188 phosphorylation. Low-gradient patients had a significantly higher degree of interstitial fibrosis than those with high-gradient stenosis (Fig. 5A), but ERKThr188 phosphorylation was only significantly up-regulated in high-gradient patients (Fig. 5C). This observation essentially excludes the possibility that up-regulation of ERKThr188 phosphorylation in myocardial biopsies may originate from increased numbers or activation of cardiac fibroblasts attributable to increased interstitial fibrosis (42). Further clinical studies should aim to delineate whether these differences in ERKThr188 phosphorylation are simply attributable to the mechanical differences (gradient) or whether the two forms are different clinical entities.
Future studies will have to establish how these effects of ERKT188A may be therapeutically exploited. Although gene therapy with ERKT188A may be a possibility, it would seem most attractive to search for small molecule inhibitors that specifically interfere with ERKThr188 phosphorylation. Because the antiapoptotic effects of ERK1/2 depend on their enzymatic/catalytic activity (Fig. 1F), mechanistic prerequisites for ERKThr188 phosphorylation may represent interesting strategies for specific interference with ERKThr188 phosphorylation. ERKThr188 phosphorylation is dependent on (i) dimerization of activated ERK1/2 and (ii) subsequent binding of Gβγ subunits to dimeric ERK1/2 (14, 15). Thus, inhibition of ERK1/2 dimerization or inhibition of the ERK/Gβγ interaction might represent possibilities to specifically interfere with ERKThr188 phosphorylation and, thus, with ERK1/2-mediated hypertrophy without affecting the antiapoptotic ERK1/2 signaling in the heart. To date, these approaches have been positively tested with overexpressed peptides and with mutant proteins in cardiomyocytes in vitro and/or in vivo and await molecular modeling of small molecule inhibitors (14, 15). Such experiments will have to consider that ERK1/2 mediate a panoply of functions within the body and that such targeting strategies may interfere with pivotal ERK1/2 functions other than pathological hypertrophy.
Taken together, our results show that specific interference with ERKThr188 phosphorylation permits differential targeting of distinct ERK1/2 effects (hypertrophy vs. cell survival), as well as of distinct types of hypertrophy (pathological vs. physiological). Selective occurrence of ERKThr188 phosphorylation in patients with rapidly progressing, high-gradient aortic valve stenosis further suggests ERKThr188 phosphorylation as a favorable target in pathological cardiac hypertrophy.
Materials and Methods
An expanded section on materials and methods is available in SI Materials and Methods.
Mice, Rats, and Cardiomyocytes.
The generation of transgenic mice overexpressing ERK2T188A (T188A) and ERK2T188T (T188T) under the control of the mouse α-myosin heavy chain promoter was described previously (14). For all experiments, male mice with an FVB/N background were used and as control isogenic WT FVB/N mice. Pregnant Sprague–Dawley rats were purchased from Janvier. NRCMs were prepared from left ventricles of 1- to 2-d-old Sprague–Dawley rats as described previously (14, 15, 43). Care of the animals was performed in accordance with the Committee on Animal Research of the regional government (Regierung von Unterfranken, Würzburg, Germany), who reviewed and approved all experimental protocols (Az.54-2531.01-62/06 and Az.55.2-2531.01-20/10) according to the corresponding national legislation.
Analysis of Patients with Aortic Valve Stenosis.
Twenty patients were regularly followed from moderate (aortic valve area, 1.0–1.5 cm2) to severe symptomatic aortic valve stenosis (aortic valve area, <1.0 cm2) and underwent transthoracic echocardiography at least 1 y before and within 3 wk before aortic valve replacement. Patients were grouped in high-gradient (≥40 mmHg; n = 12) and low transvalvular pressure gradient (<40 mmHg; n = 8) (25). Rapid progress of the stenosis was defined as an increase in peak aortic flow velocity ≥0.3 m⋅s−1⋅y−1 (44). Echocardiography was performed using the Vivid7 System (GE Vingmed Ultrasound) with a 3.5-NHz transducer. Biopsy samples of the basal septum were taken during aortic valve replacement. Informed consent was obtained from all patients. The study was reviewed and approved by the Ethics Committee of the Medical Faculty of the University of Würzburg and was conducted according to the principles outlined in the Declaration of Helsinki.
Statistical Analysis.
Statistical significance was evaluated by one-way ANOVA using GraphPad Prism software. Bonferroni’s test was used as post hoc test, with P < 0.05 regarded as significant. All data are shown as means ± SEM or as indicated. Individual experiments were reproduced at least three times. All histological and echocardiographic evaluations were performed in a blinded fashion.
Supplementary Material
Acknowledgments
We thank Martina Fischer, Nadine Yurdagül-Hemmrich, Marianne Babl, and Julia Becker for excellent technical assistance; Ulrike Zabel and Carsten Hoffmann for helpful discussions; and Daniel Becker and Julia Klose for help with the voluntary running-wheel experiments. This work was supported by Deutsche Forschungsgemeinschaft (Sonderforschungsbereich SFB688, TPA17) (to K.L. and M.J.L.) and by Bundesministerium für Bildung und Forschung (Comprehensive Heart Failure Center Würzburg; Project A2) (to K.L. and M.J.L.).
Footnotes
Conflict of interest statement: The University of Würzburg holds a patent on ERKThr188 antibodies.
*This Direct Submission article had a prearranged editor.
This article contains supporting information online at www.pnas.org/lookup/suppl/doi:10.1073/pnas.1221999110/-/DCSupplemental.
References
- 1.Levy D, Garrison RJ, Savage DD, Kannel WB, Castelli WP. Prognostic implications of echocardiographically determined left ventricular mass in the Framingham Heart Study. N Engl J Med. 1990;322(22):1561–1566. doi: 10.1056/NEJM199005313222203. [DOI] [PubMed] [Google Scholar]
- 2.Kang YJ. Cardiac hypertrophy: A risk factor for QT-prolongation and cardiac sudden death. Toxicol Pathol. 2006;34(1):58–66. doi: 10.1080/01926230500419421. [DOI] [PubMed] [Google Scholar]
- 3.Diwan A, Dorn GW., 2nd Decompensation of cardiac hypertrophy: Cellular mechanisms and novel therapeutic targets. Physiology (Bethesda) 2007;22:56–64. doi: 10.1152/physiol.00033.2006. [DOI] [PubMed] [Google Scholar]
- 4.Bernardo BC, Weeks KL, Pretorius L, McMullen JR. Molecular distinction between physiological and pathological cardiac hypertrophy: Experimental findings and therapeutic strategies. Pharmacol Ther. 2010;128(1):191–227. doi: 10.1016/j.pharmthera.2010.04.005. [DOI] [PubMed] [Google Scholar]
- 5.Rohini A, Agrawal N, Koyani CN, Singh R. Molecular targets and regulators of cardiac hypertrophy. Pharmacol Res. 2010;61(4):269–280. doi: 10.1016/j.phrs.2009.11.012. [DOI] [PubMed] [Google Scholar]
- 6.Lorenz K, Schmitt JP, Vidal M, Lohse MJ. Cardiac hypertrophy: Targeting Raf/MEK/ERK1/2-signaling. Int J Biochem Cell Biol. 2009;41(12):2351–2355. doi: 10.1016/j.biocel.2009.08.002. [DOI] [PubMed] [Google Scholar]
- 7.Heineke J, Molkentin JD. Regulation of cardiac hypertrophy by intracellular signalling pathways. Nat Rev Mol Cell Biol. 2006;7(8):589–600. doi: 10.1038/nrm1983. [DOI] [PubMed] [Google Scholar]
- 8.van Berlo JH, Elrod JW, Aronow BJ, Pu WT, Molkentin JD. Serine 105 phosphorylation of transcription factor GATA4 is necessary for stress-induced cardiac hypertrophy in vivo. Proc Natl Acad Sci USA. 2011;108(30):12331–12336. doi: 10.1073/pnas.1104499108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sheikh F, et al. An FHL1-containing complex within the cardiomyocyte sarcomere mediates hypertrophic biomechanical stress responses in mice. J Clin Invest. 2008;118(12):3870–3880. doi: 10.1172/JCI34472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Cheng H, et al. A novel preclinical strategy for identifying cardiotoxic kinase inhibitors and mechanisms of cardiotoxicity. Circ Res. 2011;109(12):1401–1409. doi: 10.1161/CIRCRESAHA.111.255695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Purcell NH, et al. Genetic inhibition of cardiac ERK1/2 promotes stress-induced apoptosis and heart failure but has no effect on hypertrophy in vivo. Proc Natl Acad Sci USA. 2007;104(35):14074–14079. doi: 10.1073/pnas.0610906104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Harris IS, et al. Raf-1 kinase is required for cardiac hypertrophy and cardiomyocyte survival in response to pressure overload. Circulation. 2004;110(6):718–723. doi: 10.1161/01.CIR.0000138190.50127.6A. [DOI] [PubMed] [Google Scholar]
- 13.Punn A, Mockridge JW, Farooqui S, Marber MS, Heads RJ. Sustained activation of p42/p44 mitogen-activated protein kinase during recovery from simulated ischaemia mediates adaptive cytoprotection in cardiomyocytes. Biochem J. 2000;350(Pt 3):891–899. [PMC free article] [PubMed] [Google Scholar]
- 14.Lorenz K, Schmitt JP, Schmitteckert EM, Lohse MJ. A new type of ERK1/2 autophosphorylation causes cardiac hypertrophy. Nat Med. 2009;15(1):75–83. doi: 10.1038/nm.1893. [DOI] [PubMed] [Google Scholar]
- 15.Vidal M, Wieland T, Lohse MJ, Lorenz K. β-Adrenergic receptor stimulation causes cardiac hypertrophy via a Gβγ/Erk-dependent pathway. Cardiovasc Res. 2012;96(2):255–264. doi: 10.1093/cvr/cvs249. [DOI] [PubMed] [Google Scholar]
- 16.Gan XT, et al. Inhibition of phenylephrine-induced cardiomyocyte hypertrophy by activation of multiple adenosine receptor subtypes. J Pharmacol Exp Ther. 2005;312(1):27–34. doi: 10.1124/jpet.104.073122. [DOI] [PubMed] [Google Scholar]
- 17.Glennon PE, et al. Depletion of mitogen-activated protein kinase using an antisense oligodeoxynucleotide approach downregulates the phenylephrine-induced hypertrophic response in rat cardiac myocytes. Circ Res. 1996;78(6):954–961. doi: 10.1161/01.res.78.6.954. [DOI] [PubMed] [Google Scholar]
- 18.Clerk A, Kemp TJ, Zoumpoulidou G, Sugden PH. Cardiac myocyte gene expression profiling during H2O2-induced apoptosis. Physiol Genomics. 2007;29(2):118–127. doi: 10.1152/physiolgenomics.00168.2006. [DOI] [PubMed] [Google Scholar]
- 19.Aikawa R, et al. Oxidative stress activates extracellular signal-regulated kinases through Src and Ras in cultured cardiac myocytes of neonatal rats. J Clin Invest. 1997;100(7):1813–1821. doi: 10.1172/JCI119709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Kepp O, Galluzzi L, Lipinski M, Yuan J, Kroemer G. Cell death assays for drug discovery. Nat Rev Drug Discov. 2011;10(3):221–237. doi: 10.1038/nrd3373. [DOI] [PubMed] [Google Scholar]
- 21.Alessi DR, Cuenda A, Cohen P, Dudley DT, Saltiel AR. PD 098059 is a specific inhibitor of the activation of mitogen-activated protein kinase kinase in vitro and in vivo. J Biol Chem. 1995;270(46):27489–27494. doi: 10.1074/jbc.270.46.27489. [DOI] [PubMed] [Google Scholar]
- 22.Okada KI, et al. Prolonged endoplasmic reticulum stress in hypertrophic and failing heart after aortic constriction: Possible contribution of endoplasmic reticulum stress to cardiac myocyte apoptosis. Circulation. 2004;110(6):705–712. doi: 10.1161/01.CIR.0000137836.95625.D4. [DOI] [PubMed] [Google Scholar]
- 23.Baines CP, Molkentin JD. STRESS signaling pathways that modulate cardiac myocyte apoptosis. J Mol Cell Cardiol. 2005;38(1):47–62. doi: 10.1016/j.yjmcc.2004.11.004. [DOI] [PubMed] [Google Scholar]
- 24.Kehat I, Molkentin JD. Extracellular signal-regulated kinase 1/2 (ERK1/2) signaling in cardiac hypertrophy. Ann N Y Acad Sci. 2010;1188:96–102. doi: 10.1111/j.1749-6632.2009.05088.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Herrmann S, et al. Low-gradient aortic valve stenosis myocardial fibrosis and its influence on function and outcome. J Am Coll Cardiol. 2011;58(4):402–412. doi: 10.1016/j.jacc.2011.02.059. [DOI] [PubMed] [Google Scholar]
- 26.Maron BJ, Pelliccia A. The heart of trained athletes: Cardiac remodeling and the risks of sports, including sudden death. Circulation. 2006;114(15):1633–1644. doi: 10.1161/CIRCULATIONAHA.106.613562. [DOI] [PubMed] [Google Scholar]
- 27.Deschamps AM, Spinale FG. Matrix modulation and heart failure: New concepts question old beliefs. Curr Opin Cardiol. 2005;20(3):211–216. doi: 10.1097/01.hco.0000162397.44843.83. [DOI] [PubMed] [Google Scholar]
- 28.Esposito G, et al. Genetic alterations that inhibit in vivo pressure-overload hypertrophy prevent cardiac dysfunction despite increased wall stress. Circulation. 2002;105(1):85–92. doi: 10.1161/hc0102.101365. [DOI] [PubMed] [Google Scholar]
- 29.Buitrago M, et al. The transcriptional repressor Nab1 is a specific regulator of pathological cardiac hypertrophy. Nat Med. 2005;11(8):837–844. doi: 10.1038/nm1272. [DOI] [PubMed] [Google Scholar]
- 30.Heineke J, et al. CIB1 is a regulator of pathological cardiac hypertrophy. Nat Med. 2010;16(8):872–879. doi: 10.1038/nm.2181. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Hino A, et al. Enhanced binding of calmodulin to the ryanodine receptor corrects contractile dysfunction in failing hearts. Cardiovasc Res. 2012;96(3):433–443. doi: 10.1093/cvr/cvs271. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.George CH, Parthimos D, Silvester NC. 2012. A network-oriented perspective on cardiac calcium signaling. Am J Physiol Cell Physiol 303(9):C897–C910 10.1152/ajpcell.00388.2011. [DOI] [PMC free article] [PubMed]
- 33.Pandit B, et al. Gain-of-function RAF1 mutations cause Noonan and LEOPARD syndromes with hypertrophic cardiomyopathy. Nat Genet. 2007;39(8):1007–1012. doi: 10.1038/ng2073. [DOI] [PubMed] [Google Scholar]
- 34.Muslin AJ. MAPK signalling in cardiovascular health and disease: Molecular mechanisms and therapeutic targets. Clin Sci (Lond) 2008;115(7):203–218. doi: 10.1042/CS20070430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Barry SP, Davidson SM, Townsend PA. Molecular regulation of cardiac hypertrophy. Int J Biochem Cell Biol. 2008;40(10):2023–2039. doi: 10.1016/j.biocel.2008.02.020. [DOI] [PubMed] [Google Scholar]
- 36.Yue TL, et al. Inhibition of extracellular signal-regulated kinase enhances Ischemia/Reoxygenation-induced apoptosis in cultured cardiac myocytes and exaggerates reperfusion injury in isolated perfused heart. Circ Res. 2000;86(6):692–699. doi: 10.1161/01.res.86.6.692. [DOI] [PubMed] [Google Scholar]
- 37.Bueno OF, et al. The MEK1-ERK1/2 signaling pathway promotes compensated cardiac hypertrophy in transgenic mice. EMBO J. 2000;19(23):6341–6350. doi: 10.1093/emboj/19.23.6341. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lips DJ, et al. MEK1-ERK2 signaling pathway protects myocardium from ischemic injury in vivo. Circulation. 2004;109(16):1938–1941. doi: 10.1161/01.CIR.0000127126.73759.23. [DOI] [PubMed] [Google Scholar]
- 39.Khokhlatchev AV, et al. Phosphorylation of the MAP kinase ERK2 promotes its homodimerization and nuclear translocation. Cell. 1998;93(4):605–615. doi: 10.1016/s0092-8674(00)81189-7. [DOI] [PubMed] [Google Scholar]
- 40.Scott MG, et al. Differential nucleocytoplasmic shuttling of beta-arrestins. Characterization of a leucine-rich nuclear export signal in beta-arrestin2. J Biol Chem. 2002;277(40):37693–37701. doi: 10.1074/jbc.M207552200. [DOI] [PubMed] [Google Scholar]
- 41.Tohgo A, et al. The stability of the G protein-coupled receptor-beta-arrestin interaction determines the mechanism and functional consequence of ERK activation. J Biol Chem. 2003;278(8):6258–6267. doi: 10.1074/jbc.M212231200. [DOI] [PubMed] [Google Scholar]
- 42.Manabe I, Shindo T, Nagai R. Gene expression in fibroblasts and fibrosis: Involvement in cardiac hypertrophy. Circ Res. 2002;91(12):1103–1113. doi: 10.1161/01.res.0000046452.67724.b8. [DOI] [PubMed] [Google Scholar]
- 43.Lorenz K, Lohse MJ, Quitterer U. Protein kinase C switches the Raf kinase inhibitor from Raf-1 to GRK-2. Nature. 2003;426(6966):574–579. doi: 10.1038/nature02158. [DOI] [PubMed] [Google Scholar]
- 44.Rosenhek R, et al. Predictors of outcome in severe, asymptomatic aortic stenosis. N Engl J Med. 2000;343(9):611–617. doi: 10.1056/NEJM200008313430903. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.




