1,072
Views
1
CrossRef citations to date
0
Altmetric
Research Paper

IL-6/STAT3 signaling in mice with dysfunctional type-2 ryanodine receptor

, &
Article: e1158379 | Received 29 Jan 2016, Accepted 19 Feb 2016, Published online: 07 Mar 2016

ABSTRACT

Mice with genetically modified cardiac ryanodine receptor (Ryr2ADA/ADA mice) are impaired in regulation by calmodulin, develop severe cardiac hypertrophy and die about 2 weeks after birth. We hypothesized that the interleukin 6 (IL-6)/signal transducer and activator of transcription-3 (STAT3) signaling pathway has a role in the development of the Ryr2ADA/ADA cardiac hypertrophy phenotype, and determined cardiac function and protein levels of IL-6, phosphorylation levels of STAT3, and downstream targets c-Fos and c-Myc in wild-type and RyR2ADA/ADA mice, mice with a disrupted IL-6 gene, and mice treated with STAT3 inhibitor NSC74859. IL-6 protein levels were increased at postnatal day 1 but not day 10, whereas pSTAT3-Tyr705/STAT3 ratio and c-Fos and c-Myc protein levels increased in hearts of 10-day but not 1-day old Ryr2ADA/ADA mice compared with wild type. Both STAT3 and pSTAT3-Tyr705 accumulated in the nuclear fraction of 10-day old Ryr2ADA/ADA mice compared with wild type. Ryr2ADA /ADA /IL-6−/− mice lived 1.5 times longer, had decreased heart to body weight ratio, and reduced c-Fos and c-Myc protein levels. The STAT3 inhibitor NSC74859 prolonged life span by 1.3-fold, decreased heart to body weight ratio, increased cardiac performance, and decreased pSTAT-Tyr705/STAT3 ratio and IL-6, c-Fos and c-Myc protein levels of Ryr2ADA /ADA mice. The results suggest that upregulation of IL-6 and STAT3 signaling contributes to cardiac hypertrophy and early death of mice with a dysfunctional ryanodine receptor. They further suggest that STAT3 inhibitors may be clinically useful agents in patients with altered Ca2+ handling in the heart.

ABBREVIATIONS

CaM=

calmodulin

IL-6=

interleukin 6

mTOR=

inhibitor of mammalian target of rapamycin

RyR2=

cardiac ryanodine receptor

SR=

sarcoplasmic reticulum

STAT3=

signal transducer and activator of transcription-3

INTRODUCTION

Type-2 ryanodine receptors (RyR2s) release Ca2+ from the sarcoplasmic reticulum (SR) to cause cardiac muscle contraction. The RyR2s are composed of 4 560-kDa RyR2 and 4 12.6-kDa FK506 binding protein subunits, and regulated by Ca2+ and associated proteins such as calmodulin (CaM).Citation1-4 CaM is a 16.7-kDa Ca2+ binding protein that modulates proteins through CaM-dependent protein kinases and phosphatases or through direct binding.Citation5 In mouse cardiomyocytes, the majority of CaM is bound to RyR2Citation6 which reduces the release of Ca2+ from SR by inhibiting RyR2.Citation7-9

To determine the role of CaM in regulating RyR2 in vivo, a mouse model with 3 amino acid replacements in the CaM binding domain of RyR2 (RyR2-W3587A/L3591D/F3603A, RyR2ADA) was generated.Citation10 The triple mutation impaired inhibition of RyR2 by CaM at diastolic and systolic Ca2+, and resulted in severe cardiac hypertrophy and death of mice within about 2 weeks after birth. The mutations did not appear to introduce other major structural and functional changes in RyR2. The mutant mice had reduced RyR2 content and SERCA2a activity, lower Cav1.2 current density and showed irregularities in local and global Ca2+ transients.Citation10-11 Ultrastructural analysis indicated only minor structural changes in myofibrillar organization, however, a reduction in SR/T-tubule junctions and RyR2 content were observed in mutant hearts which had a 2–3-fold increase in size compared to wild type.Citation12 Additionally, several signaling molecules were implicated in cardiac hypertrophy of Ryr2ADA/ADA mice. Phosphorylation levels of extracellular signal-regulated kinases 1 and 2, p90 ribosomal S6 kinase and glycogen synthase kinases 3α and 3β increased in hearts of embryonic day 16.5 Ryr2ADA/ADA mice.Citation13 Class II histone deacetylase/myocyte enhancer factor-2 signaling and nuclear factor of activated T cell transcriptional activity were up-regulated in the hearts from 1-day old Ryr2ADA/ADA mice but not hearts from E16.5 mice.Citation10,13 Calcineurin A-β ablation decreased heart weight and improved cardiac contractility and extended the lifespan of Ryr2ADA/ADA mice by 2-fold, without suppressing the expression of genes associated with cardiac hypertrophy.Citation13 Treatment with rapamycin, an inhibitor of mammalian target of rapamycin (mTOR), inhibited phosphorylation of mTOR and downstream targets ribosomal S6 kinase and ribosomal protein S6, decreased heart size, and improved cardiac function but did not extend the lifespan of Ryr2ADA/ADA mice.Citation14 The results suggest that multiple signaling pathways are involved in the development of cardiac hypertrophy and heart failure in Ryr2ADA/ADA mice expressing cardiac RyR2 impaired in regulation by CaM.

The present study explores the role of interleukin-6 (IL-6) and signal transducer and activator of transcription 3 (STAT3) signaling in cardiac hypertrophy and heart failure in Ryr2ADA/ADA mice. In addition to its well-known role in inflammatory and immunological processes, increased circulating levels of IL-6 and related cytokines have been linked to heart failure in patients and animal models.Citation15 Binding of IL-6 to membrane-bound and soluble receptors results in a complex with glycoprotein (gp) 130.Citation15 In turn, formation of the IL-6/IL-6R/gp130 complex triggers 3 major downstream signaling pathways, janus kinase (JAK)/(STAT), mitogen activated protein kinase and phosphatidylinositol 3-kinase (PI3K)-AKT dependent pathways.Citation15,16 Seven structurally related but functionally distinct isoforms of STATCitation17 are expressed in cardiomyocytes.Citation18 Among these, STAT3 has been implicated in the development of cardiac hypertrophy.Citation15,19-24 Binding of IL-6 to its receptor and complex formation with gp130 causes phosphorylation of associated JAKs and recruitment and phosphorylation of STAT3. Following its phosphorylation, STAT3 translocates to the nucleus where it binds to response elements of c-Fos and c-Myc target genes.Citation25-27 Absence of a hypertrophic response in the absence of soluble IL-6 receptorCitation28 and dependence of STAT3 phosphorylation on the soluble receptorCitation29 suggested that Il-6 mediates its effect on STAT3 through the soluble receptor in cardiomyocytes.

Here we show that IL-6 protein levels, STAT3 phosphorylation, and c-Fos and c-Myc protein levels are upregulated in Ryr2ADA/ADA mice. Crossing Ryr2ADA/ADA mice with Il-6−/− mice or treatment of Ryr2ADA/ADA mice with STAT3 inhibitor NSC74859 decreased pSTAT-Tyr705 phosphorylation and c-Fos and c-Myc protein levels, reduced heart size, prolonged life span, and improved cardiac performance of Ryr2ADA/ADA mice. Our findings suggest that IL-6/STAT3 signaling contributes to the development of cardiac hypertrophy in Ryr2ADAADA mice. A preliminary report of this work has been presented in abstract form.Citation30

MATERIALS AND METHODS

Materials

STAT3 mouse monoclonal antibody (catalog No. 9139), pSTAT3-Tyr705 rabbit monoclonal antibody (catalog No. 9145), pSTAT3-Ser-727 rabbit polyclonal antibody (catalog No. 9134), c-Fos rabbit monoclonal antibody (catalog No. 2250), and GAPDH rabbit monoclonal antibody (catalog No. 2118) were from Cell Signaling. c-Myc mouse monoclonal antibody (catalog No. sc-40) was from Santa Cruz, and IL-6 mouse monoclonal antibody (A3218) was from eBioscience. STAT3 inhibitor NSC74859 (also known as S3I-201) (catalog No. S1155) was from Selleck Chemicals. Primary antibodies were used at 1:1000 dilution, except GAPDH (1:5000). For the secondary antibodies, the dilution was 1:10,000. Protease (catalog No. 11697498001) and phosphatase (catalog No. P5726) inhibitors were from Sigma-Aldrich. Other chemical reagents were from Sigma-Aldrich.

Animals

Ryr2+/+ and Ryr2ADA/ADA mice were obtained by backcrossing Ryr2+/ADA mice at least 10 times to 129/Svev background.Citation10 To generate Ryr2+/+ and Ryr2ADA/ADA mice with disrupted IL-6 gene, 129/Svev Ryr2+/ADA mice were mated with B6.129S2-Il-6tm1Kopf/J mice (Jackson Laboratory). The subsequent litters were backcrossed 3–4 times with 129/Svev Ryr2+/ADA mice. Animal experiments were approved by the Institutional Animal Care and Use Committee of the University of North Carolina at Chapel Hill.

Primers for genotyping of mice were: Ryr2, sense (s) 5′-CAAGGCTTAGGA GCTACGAG-3′ and antisense (a) 5′-TTCACGTTGCCCCTCGTTAC-3′; IL-6, (common) 5′-TTCCATCCAGTTG CCTTCTTGG-3′, (wild type reverse) 5′-TTCTCATTTCC ACGATTTCCCAG-3′, (mutant reverse) 5′-CCGGAGAACCTGCGTG CAATCC-3′.

STAT3 inhibitor treatment

STAT3 inhibitor NSC74859 was dissolved (1.5 mg/ml) in 0.15 M NaCl and 3% DMSO.Citation31 One day old mice were separated randomly into 4 groups. NSC74859 (20 µg/g body weight) was injected intraperitoneally once every day. Vehicle injection served as the negative control. For mRNA analysis, animals were sacrificed and hearts collected 1 h after the last injection. For protein analysis, hearts were collected 4 h after the final injection.

2.3. Echocardiography

To determine left ventricular cardiac function in mice, transthoracic M-mode echocardiography was performed on restrained unanesthetized 10-day old mice, using a 770 high resolution imaging system (VisualSonics) with a 40-MHz probe. Mice were restrained by taping down gently to on a warmed mouse board (Indus Industries for VisualSonics).

Quantitative RT- PCR

Total RNA was extracted from left ventricles of 10-day old mice using Trizol (Invitrogen) and reverse transcribed into cDNA using SuperScript First-Strand Synthesis System for RT-PCR Kit (Invitrogen). Real-Time PCR was performed using the following primers: IL-6, (s) 5′-CCGGAGAGGAGACTTC ACAG-3′ and (a) 5′-TCCACGATTTCCCAGA GAAC-3′; STAT3, (s) 5′-GGAGGAG GCATTTGGAA AGT-3′ and (a) 5′-GGCAGGTCAATGGTATTGCT-3′; c-Fos, (s) 5′-GTCCGGTTCCTTCTA TGCAG −3′ and (a) 5′- TAAGTAGTG CAGCCCGGAGT −3′; c-Myc, (s) 5′-GCTGGAGAT GATGACCGAGT-3′ and (a) 5′-AACCGCTCCACATACAGTCC-3′; ANP, (s) 5′ ATCTGCCCTCTTGAAAAGCA-3′ and (a) 5′- AAGCTGTTGCAGCCTAGTCC-3′; BNP, (s) 5′- CAGCTCTTGAAGGACCAAGG-3′ and (a) 5′- CCGATCCGGTCTATCTTGTG-3′; 18S RNA, (s) 5′-CGTCTGCCCTAT CAAC TTTCG-3′ and (a) 5′- CCTTGGATGTGGTAGC CGTT-3′. Real -time PCR was performed using Maxima® SYBR Green /Rox qPCR master mix (Thermo Scientific) and 7900 HT Fast Real Time PCR machine (Applied Biosystems). Cycling conditions were 95°C for 10 min, and 40 cycles of 95°C for 15 sec and 60°C for 30 sec. Melting curve analysis of products was performed following each amplification to verify the specificity of the PCR products. 18S RNA was used as an internal control for normalization.

Preparations of heart fractions

Whole heart homogenates were prepared in 20 mM imidazole, pH 7.0, containing 0.3 M sucrose, 0.15 M NaCl, protease and phosphatase inhibitor cocktails, 25 mM β-glycerophosphate, 5 mM NaF, and 2.5 mM NaVO4 using Tekmar Tissumizer for 3 × 7 s at a setting of 13,500 rpm. Cytosolic and nuclear fractions were prepared using Pierce Biotechnology Ne-Per Nuclear and Cytoplasmic Extraction Reagents kit. Cellular fractions were stored in small aliquots at −80°C. Protein concentrations were determined using the bicinchoninic acid assay.

Immunoblot analysis

Homogenates (20 μg protein/lane) were separated by 10% SDS/PAGE, transferred to nitrocellulose membranes, and probed with monoclonal or polyclonal antibodies. Western blots were developed using enhanced chemiluminescence and quantified using ImageQuantTL Analysis Software. GAPDH was the loading control.

Statistical analysis

Statistical analysis was performed using Sigma-plot 11. T-test was used to compare 2 groups and ANOVA followed by Tukey's test to analyze the significance among multi groups. Statistical significance was taken at the p < 0.05 level.

RESULTS

Upregulation of IL-6 and pSTAT3-Tyr-705 in Ryr2ADA/ADA mouse hearts

We reported previously that mice with impaired CaM inhibition of the cardiac ryanodine receptor (Ryr2ADA/ADA mice) show signs of cardiac hypertrophy at postnatal day 1 and die within about 2 weeks after birth.Citation10 To determine the mechanism of cardiac hypertrophy induced by the RyR2 mutation, protein levels of IL-6 and STAT3, phosphorylation levels of pSTAT3-Tyr705 and pSTAT3-Tyr727, and protein levels of c-Fos and c-Myc downstream transcription factors were determined. An increase in IL-6 protein was observed in 1-day old but not 10-day old Ryr2ADA/ADA hearts, whereas STAT3 protein levels were similar in wild-type Ryr2+/+ hearts (). STAT3 is activated by phosphorylation at Tyr-705 following phosphorylation of janus kinase (JAK) in IL-6/IL-6R/p130-JAK complex, whereas phosphorylation at Ser-727 is mediated through the mitogen activated protein kinase and mTOR pathways.Citation15 The pSTAT3-Tyr705/STAT3 ratio increased at postnatal day 10, but not at postnatal day 1 () or days 3–4 (not shown). No change in the pSTAT3-Ser727/STAT3 ratio was observed at postnatal days 1 and 10. Consistent with the absence of an elevated pSTAT3-Tyr705/STAT3 ratio at postnatal days 1 and 3–4, c-Fos and c-Myc increased only during the later stages of cardiac hypertrophy compared to wild-type hearts (). The data suggested the IL-6/STAT3 signaling pathway may have a role in development of the Ryr2ADA/ADA cardiac hypertrophy phenotype.

FIGURE 1. IL-6 and downstream signaling molecule expression in hearts of Ryr2+/+ and Ryr2ADA/ADA mice. (A) Immunoblots of heart homogenates from 1-day old and 10-day old Ryr2+/+ (WT) and Ryr2ADA/ADA (ADA) mice. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was the loading control. (B) Relative protein and phosphorylation levels in 1-day old and 10-day old Ryr2ADA/ADA compared to Ryr2+/+ mice. Data are the mean ± SEM of 5–19 samples. *p < 0.05 compared to Ryr2+/+, #p < 0.05 compared to corresponding 1 day sample using one way ANOVA.

FIGURE 1. IL-6 and downstream signaling molecule expression in hearts of Ryr2+/+ and Ryr2ADA/ADA mice. (A) Immunoblots of heart homogenates from 1-day old and 10-day old Ryr2+/+ (WT) and Ryr2ADA/ADA (ADA) mice. Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was the loading control. (B) Relative protein and phosphorylation levels in 1-day old and 10-day old Ryr2ADA/ADA compared to Ryr2+/+ mice. Data are the mean ± SEM of 5–19 samples. *p < 0.05 compared to Ryr2+/+, #p < 0.05 compared to corresponding 1 day sample using one way ANOVA.

To determine whether STAT3 and phosphorylated pSTAT3-Tyr705 translocated to the nucleus in hearts of Ryr2ADA/ADA mice, cytosolic and nuclear fractions were prepared. GAPDH and histone 3 served as cytosolic and nuclear markers, respectively (). In agreement with the nuclear accumulation of unphosphorylated STAT3 in response to IL-6,Citation32 STAT3 was enriched in the nuclear fraction of 10-day old Ryr2ADA/ADA hearts. pSTAT-Tyr705 was predominantly localized in the nuclear fraction of Ryr2ADA/ADA mouse hearts compared to wild-type hearts.

FIGURE 2. Nuclear and cytosolic location of STAT3 and pSTAT3-Tyr705. (A) Immunoblots of nuclear (N) and cytosolic (C) fractions from hearts of 10-day old Ryr2+/+ (WT) and Ryr2ADA/ADA (ADA) mice. GAPDH and histone 3 (His3) were markers for cytosolic and nuclear fractions, respectively. (B) Protein and phosphorylation levels of Ryr2ADA/ADA mice were normalized to Ryr2+/+. Data are the mean ± SEM of 5–6 samples.*p < 0.05 compared to Ryr2+/+ using t-test.

FIGURE 2. Nuclear and cytosolic location of STAT3 and pSTAT3-Tyr705. (A) Immunoblots of nuclear (N) and cytosolic (C) fractions from hearts of 10-day old Ryr2+/+ (WT) and Ryr2ADA/ADA (ADA) mice. GAPDH and histone 3 (His3) were markers for cytosolic and nuclear fractions, respectively. (B) Protein and phosphorylation levels of Ryr2ADA/ADA mice were normalized to Ryr2+/+. Data are the mean ± SEM of 5–6 samples.*p < 0.05 compared to Ryr2+/+ using t-test.

Ryr2ADA/ADA/IL-6−/− mice. In addition to IL-6, other members of the cytokine family might have contributed to the Ryr2ADA/ADA cardiac hypertrophy phenotype.Citation15 To obtain evidence that IL-6 signaling was associated with cardiac hypertrophy, we generated Ryr2+/+ and Ryr2ADA/ADA mice carrying a disrupted IL-6 gene. 129/Svev Ryr2+/ADA mice were mated with B6.129S2-Il-6tm1Kopf/J mice. The subsequent litters were backcrossed 3–4 times with 129/Svev Ryr2+/ADA mice. Accordingly, littermates with a predominantly 129/Svev genetic background were obtained, out of which the 4 genotypes investigated were Ryr2+/+, Ryr2ADA/ADA, Ryr2+/+/Il6−/− and Ryr2ADA/ADA/Il6−/−. Ryr2+/+/IL-6−/− mice carrying an IL-6 gene disrupted in the first coding exon by insertion of neo cassetteCitation33 were viable without change in body to heart weight ratio compared with Ryr2+/+ mice (). However, loss of IL-6 significantly decreased heart weight to body weight ratio in Ryr2ADA/ADA /IL-6−/−mice compared to Ryr2ADA/ADA mice (). Ryr2ADA/ADA/Il-6−/− mice also lived longer compared to Ryr2ADA/ADA mice, with mean lifetimes of 25.1 ± 3.5 d and 16.9 ± 1.3 d (p < 0.05), respectively (). Echocardiography indicated that the IL-6 mutation did not significantly alter cardiac performance in Ryr2+/+ and Ryr2ADA/ADA mice ().

FIGURE 3. Ryr2ADA/ADA and Ryr2ADA/ADA/IL-6−/− mice survival. Mean lifetimes ± SEM of Ryr2ADA/ADA and Ryr2ADA/ADA/IL-6−/− mice of 16.9 ± 1.3 (n = 9) and 25.1 ± 3.5 (n = 7), respectively, were significantly different (p < 0.05).

FIGURE 3. Ryr2ADA/ADA and Ryr2ADA/ADA/IL-6−/− mice survival. Mean lifetimes ± SEM of Ryr2ADA/ADA and Ryr2ADA/ADA/IL-6−/− mice of 16.9 ± 1.3 (n = 9) and 25.1 ± 3.5 (n = 7), respectively, were significantly different (p < 0.05).

Table 1. Body and heart weights and echocardiography of 10-day old Ryr2+/+ and Ryr2ADA/ADA mice.

Co-expression of Ryr2ADA/ADA and IL-6 mutants did not alter STAT3 protein levels and pSTAT-Ser-727/STAT3 ratios among the 4 genotypes (). The elevated pSTAT3-Tyr705/STAT3 ratio in the hearts of Ryr2ADA/ADA mice was modestly (not significant) decreased in the mutant mice carrying the IL-6 mutation, whereas the IL-6 mutant significantly decreased c-Fos and c-Myc protein levels in Ryr2ADA/ADA mice. No significant change was observed in Ryr2+/+ /Il-6−/− mice compared with Ryr2+/+ mice (). Taken together, the results of and suggest that IL-6 had a role in decreasing lifespan, increasing heart size and upregulating c-Fos and c-Myc protein levels in Ryr2ADA/ADA mice. However, the data also suggest that other mechanisms exist that contributed to the Ryr2ADA/ADA phenotype.

FIGURE 4. Expression of IL-6 downstream signaling molecules in mice targeted for Ryr2ADA and IL-6−/−. (A) Immunoblots of heart homogenates from 10-day old Ryr2+/+ (WT) and Ryr2ADA/ADA (ADA) mice with and without IL-6. GAPDH was the loading control. (B) Protein levels and phosphorylation ratios of Ryr2ADA/ADA mice were normalized to Ryr2+/+. Data are the mean ± SEM of 6–14 determinations. Footnotep < 0.05 compared with Ryr2+/+, #p < 0.05 compared with Ryr2ADA/ADA, using one way ANOVA.

FIGURE 4. Expression of IL-6 downstream signaling molecules in mice targeted for Ryr2ADA and IL-6−/−. (A) Immunoblots of heart homogenates from 10-day old Ryr2+/+ (WT) and Ryr2ADA/ADA (ADA) mice with and without IL-6. GAPDH was the loading control. (B) Protein levels and phosphorylation ratios of Ryr2ADA/ADA mice were normalized to Ryr2+/+. Data are the mean ± SEM of 6–14 determinations. Footnotep < 0.05 compared with Ryr2+/+, #p < 0.05 compared with Ryr2ADA/ADA, using one way ANOVA.

Treatment with STAT3 inhibitor NSC74859

To obtain more direct evidence for STAT3 regulation in cardiac hypertrophy of Ryr2ADA/ADA mice, newborn mice were daily injected with specific STAT3 inhibitor NSC74859.Citation34 Treatment with NSC74859 significantly decreased the heart to body weight ratio in Ryr2ADA/ADA mice compared to no change in Ryr2+/+ mice. The lifespan of Ryr2ADA/ADA mice increased, with mean lifetimes of 18.4 ± 1.8 d and 13.9 ± 0.5 d with or without NSC74859 (p < 0.05), respectively (). Echocardiography showed that NSC74859 significantly improved left ventricular dimension at end systole (2.58 vs 3.16 without NSC), fractional shortening (17.2% vs 10.2% without NSC74859, as determined by t-test) and ejection fraction (36.7% vs 22.6% without NSC) in 10-day old Ryr2ADA/ADA mice (). Values obtained for wild type remained similar with and without NSC74859 treatment (0.42 vs 0.50, 73.0% vs 68.9%, and 96.4 % vs 95.4%, respectively).

FIGURE 5. Survival data from Ryr2ADA/ADA mice treated with STAT3 inhibitor NSC74859. Mean lifetimes ± SEM of Ryr2ADA/ADA mice treated without (Control) and with NSC74859 (NSC) of13.9 ± 0.5 (n = 13) and 18.4 ± 1.8 (n = 13), respectively, were significantly different (p < 0.05).

FIGURE 5. Survival data from Ryr2ADA/ADA mice treated with STAT3 inhibitor NSC74859. Mean lifetimes ± SEM of Ryr2ADA/ADA mice treated without (Control) and with NSC74859 (NSC) of13.9 ± 0.5 (n = 13) and 18.4 ± 1.8 (n = 13), respectively, were significantly different (p < 0.05).

Table 2. Body and heart weights and echocardiography of 10-day old Ryr2+/+ and Ryr2ADA/ADA mice with and without NSC74859.

Treatment with NSC74859 significantly lowered IL-6 protein levels in both Ryr2+/+ and Ryr2ADA/ADA hearts (). The pSTAT3-Tyr705/STAT3 ratio and the protein levels of c-Fos and c-Myc in Ryr2ADA/ADA mice were significantly reduced by NSC74859 without a change in Ryr2+/+ mice. The results show that the Ryr2ADA/ADA phenotype is linked to elevated phosphorylation of pSTAT3 at Tyr-705 and protein levels of c-Myc and c-Fos.

FIGURE 6. IL-6 and downstream signaling molecule levels in 10-day old mice treated with or without STAT3 inhibitor NSC74859. (A) Immunoblots of heart homogenates from 10-day old Ryr2+/+ (WT) and Ryr2ADA/ADA (ADA) mice treated with or without NSC74859 (NSC). GAPDH was the loading control. (B) Protein levels and phosphorylation ratios of Ryr2ADA/ADA mice were normalized to Ryr2+/+ Control. Data are the mean ± SEM of 4–10 samples. *p < 0.05 compared to Ryr2+/+ mice without NSC74859, #p < 0.05 compared to Ryr2ADA/ADA mice without NSC74859, using one way ANOVA.

FIGURE 6. IL-6 and downstream signaling molecule levels in 10-day old mice treated with or without STAT3 inhibitor NSC74859. (A) Immunoblots of heart homogenates from 10-day old Ryr2+/+ (WT) and Ryr2ADA/ADA (ADA) mice treated with or without NSC74859 (NSC). GAPDH was the loading control. (B) Protein levels and phosphorylation ratios of Ryr2ADA/ADA mice were normalized to Ryr2+/+ Control. Data are the mean ± SEM of 4–10 samples. *p < 0.05 compared to Ryr2+/+ mice without NSC74859, #p < 0.05 compared to Ryr2ADA/ADA mice without NSC74859, using one way ANOVA.

Next we asked whether NSC74859 altered the mRNA levels of other cardiac hypertrophy associated genes. Quantitative RT-PCR showed that c-Fos and c-Myc mRNA levels increased in hearts from 10-day old Ryr2ADA/ADA mice compared with Ryr2+/+ mice. NSC74859 decreased 5-fold c-Fos mRNA levels in Ryr2ADA/ADA hearts without changing the levels in Ryr2+/+ mice (). c-Myc mRNA levels were not significantly reduced in the mutant mice. In agreement with our previous report,Citation10 brain (BNP) and atrial (ANP) natriuretic peptide mRNA levels were significantly increased in hearts from Ryr2ADA/ADA mice compared to Ryr2+/+ mice. Interestingly, mRNA levels of cardiac hypertrophy associated genes BNP and ANP were not reduced by NSC74859 in hearts from Ryr2ADA/ADA mice. Indeed, in the case of ANP, NSC74859 increased rather than decreased the mRNA level in Ryr2ADA/ADA mice. The results suggest that, in addition to IL-6/STAT3 signaling, other mechanisms are associated with the Ryr2ADA/ADA phenotype.

FIGURE 7. c-Fos, c-Myc, ANP and BNP mRNA levels in hearts of Ryr2+/+ and Ryr2ADA/ADA mice treated with or without STAT3 inhibitor NSC74859. mRNA levels were measured by quantitative RT-PCR and normalized to levels in hearts of Ryr2+/+ mice not treated with the inhibitor (Ryr2+/+ Control). Data are the mean ± SEM of 5–6 samples. *p < 0.05 compared to Ryr2+/+ mice without NSC74859, #p < 0.05 compared with Ryr2ADA/ADA mice without NSC74859, %p < 0.05 compared with Ryr2+/+ mice with NSC74859, using one way ANOVA.

FIGURE 7. c-Fos, c-Myc, ANP and BNP mRNA levels in hearts of Ryr2+/+ and Ryr2ADA/ADA mice treated with or without STAT3 inhibitor NSC74859. mRNA levels were measured by quantitative RT-PCR and normalized to levels in hearts of Ryr2+/+ mice not treated with the inhibitor (Ryr2+/+ Control). Data are the mean ± SEM of 5–6 samples. *p < 0.05 compared to Ryr2+/+ mice without NSC74859, #p < 0.05 compared with Ryr2ADA/ADA mice without NSC74859, %p < 0.05 compared with Ryr2+/+ mice with NSC74859, using one way ANOVA.

DISCUSSION

Our findings suggest that aberrant Ca2+ handling in Ryr2ADA/ADA mice increases IL-6 expression and activation of STAT3 through Tyr-705 phosphorylation. IL-6 is released by various tissues including heart and is synthesized in fibroblasts and cardiomyocytes.Citation35 Elevation of IL-6 by chronic infusion was associated with left ventricular hypertrophy, fibrosis, and dysfunction,Citation36 overexpression of both IL-6 and IL-6 receptor developed myocardial hypertrophy,Citation28 and cardiac hypertrophy induced by angiotensin II infusion was attenuated by deletion of IL-6.Citation37 On the other hand, deletion of IL-6 did not attenuate left ventricular remodeling or dysfunction in pressure-overloaded hearts.Citation38 We find that IL-6 protein levels increased in 1-day old but not in 10-day old Ryr2ADA/ADA mice. Ryr2+/+/IL-6−/− mice carrying a disrupted IL-6 gene lived longer and had a decreased heart size without significantly altered cardiac performance. In addition, loss of IL-6 decreased protein levels of c-Fos and c-Myc, 2 STAT3-responsive transcription factorsCitation25,27 that are rapidly induced in the ventricular myocardium by pressure overload.Citation39

To obtain evidence that IL-6 mediated its effects through phosphorylation of STAT3 at Tyr-705, Ryr2ADA/ADA mice were treated with the STAT3 inhibitor NSC74859 (S3I-201). NSC74859 interacted with the STAT3 SH2 domain and preferentially inhibited growth in cells that contained STAT3 activated by phosphorylation at Tyr-705 by inhibiting STAT3 dimer formation and STAT3 DNA-binding and transcriptional activities.Citation34 NSC74859 was effective in inhibiting angiotensin-II induced oxidative stress, endothelial dysfunction and hypertension in 4–5 month old miceCitation40 and in decreasing heart size, improving cardiac function and inhibiting cardiac hypertrophy markers such as ANP, c-Fos and c-Myc in a renal artery ligated rat model.Citation26 In the present study, in agreement with the results from the rat model,Citation26 daily injection of NSC74859 significantly increased lifespan and decreased c-Fos and c-Myc protein levels in Ryr2ADA/ADA/Il-6−/− mice. However, at variance with ANP expression at the protein level in the renal artery ligated rat model,Citation26 ANP mRNA expression was increased in Ryr2ADA/ADA by NSC74859 without a change in Ryr2+/+ mice. Thus the pathological effects of the RyR2ADA mutation appear to be mediated in part by additional mechanisms that may include an upregulation of calcineurin A-βCitation13 and mTORCitation14 signaling. Another possibility is that nuclear accumulation of unphosphorylated STAT3 contributed to the Ryr2ADA/ADA phenotype. NSC74859 was less effective in reducing growth and viability in normal fibroblast NIH3T3 cells than transformed NIH3T3 cells that harbored activated pSTAT3-Tyr705,Citation34 which suggests that nuclear STAT3 may promote the activation of genes that do not respond directly to phosphorylated STAT3.Citation41

An intriguing result from our studies was evidence that STAT3 inhibition by NSC74859 reduced the IL-6 protein level in hearts of Ryr2+/+ and Ryr2ADA/ADA mice. This could suggest at first sight that the effects of NSC74859 are due to its direct effect on IL-6 expression. However, this appears unlikely, as there is no evidence of a direct interaction with Il-6. More likely, NSC74859 inhibited Il-6 expression by inhibiting STAT3 transcriptional activity.Citation34 Myocardial IL-6 expression is regulated by multiple factors. Hypoxia-induced activation of IL-6 was mediated by transcriptional factors NF-IL-6 and NF-kB in primary cultures of myocytes prepared from 1-day old rat hearts.Citation42 In skeletal muscle, activator protein-1, NF-kB and inositol trisphosphate release of Ca2+ were involved in upregulation of IL-6 transcription.Citation43 In 8-week old mouse hearts, STAT1 promoted the expression of IL-6 and countered the suppressive role of p53.Citation44 In adult rat ventricular myocytes, IL-1 and β-adrenergic receptor synergistically increased IL-6 expression and secretion.Citation29 Further, coordinate regulation of STAT3 and microRNAs suggests that IL-6 expression was regulated by microRNAs.Citation45 Future studies will be required to clarify the relationship between the effects of NSC74859 on STAT3 signaling and suppression of IL-6 expression in the hearts of young Ryr2ADA/ADA mice.

In summary, our results indicate that impaired inhibition of RyR2 by CaM resulted in increased IL-6 expression, phosphorylation of STAT3 at Tyr-705, and nuclear accumulation of both phosphorylated and unphosphorylated STAT3. Generation of double targeted Ryr2ADA/ADA/IL-6−/− mice and treatment of Ryr2ADA/ADA mice with STAT3 inhibitor NSC74859 prolonged life span, decreased heart size, and decreased the expression levels of proteins associated with IL-6/STAT3 signaling. The results suggest that upregulation of IL-6 and STAT3 signaling contribute to the rapid development of cardiac hypertrophy and early death of Ryr2ADA/ADA mice.

DISCLOSURE OF POTENTIAL CONFLICTS OF INTEREST

No potential conflicts of interest were disclosed.

REFERENCES

  • Fill M, Copello JA. Ryanodine receptor calcium release channels. Physiol Rev 2002; 82:893-922; PMID:12270947; http://dx.doi.org/10.1152/physrev.00013.2002
  • Meissner G. Regulation of mammalian ryanodine receptors. Front Biosci 2002; 7:d2072-d2080; PMID:12438018; http://dx.doi.org/10.2741/meissner
  • Wehrens XH, Lehnart SE, Marks AR. Intracellular calcium release and cardiac disease. Annu Rev Physiol 2005; 67:69-98; PMID:15709953; http://dx.doi.org/10.1146/annurev.physiol.67.040403.114521
  • Zalk R, Lehnart SE, Marks AR. Modulation of the ryanodine receptor and intracellular calcium. Anu Rev Bioch 2007; 76:367-85; http://dx.doi.org/10.1146/annurev.biochem.76.053105.094237
  • Rhoads AR, Friedberg F. Sequence motifs for calmodulin recognition. FASEB J 1997; 11:331-40; PMID:9141499
  • Yang Y, Guo T, Oda T, Chakraborty A, Chen L, Uchinoumi H, Knowlton AA, Fruen BR, Cornea RL, Meissner G, et al. Cardiac myocyte Z-line calmodulin is mainly RyR2-bound, and reduction is arrhythmogenic and occurs in heart failure. Circ Res 2014; 114:295-306; PMID:24186966; http://dx.doi.org/10.1161/CIRCRESAHA.114.302857
  • Meissner G, Henderson JS. Rapid calcium release from cardiac sarcoplasmic reticulum vesicles is dependent on Ca2+ and is modulated by Mg2+, adenine nucleotide, and calmodulin. J Biol Chem 1987; 262:3065-73; PMID:2434495.
  • Yamaguchi N, Xu L, Pasek DA, Evans KE, Meissner G. Molecular basis of calmodulin binding to cardiac muscle Ca2+ release channel (ryanodine receptor). J Biol Chem 2003; 278:23480-6; PMID:12707260; http://dx.doi.org/10.1074/jbc.M301125200
  • Fruen BR, Bardy JM, Byrem TM, Strasburg GM, Louis CF. Differential Ca2+ sensitivity of skeletal and cardiac muscle ryanodine receptors in the presence of calmodulin. Am J Physiol 2000; 279:C724-33.
  • Yamaguchi N, Takahashi N, Xu L, Smithies O, Meissner G. Early cardiac hypertrophy in mice with impaired calmodulin regulation of cardiac muscle Ca2+ release channel. J Clin Invest 2007; 117:1344-53.; PMID:17431507; http://dx.doi.org/10.1172/JCI29515
  • Arnaiz-Cot JJ, Damon BJ, Zhang XH, Cleemann L, Yamaguchi N, Meissner GW, Morad M. Cardiac calcium signaling pathologies associated with defective calmodulin regulation of type 2 ryanodine receptor. J Physiol 2013; 591:4287-99; PMID:23836685; http://dx.doi.org/10.1113/jphysiol.2013.256123
  • Lavorato M, Huang TQ, Iyer VR, Perni S, Meissner G, Franzini-Armstrong C. Dyad content is reduced in cardiac myocytes of mice with impaired calmodulin regulation of RyR2. J Muscle Res Cell Motility 2015; 36:205-14; http://dx.doi.org/10.1007/s10974-015-9405-5
  • Yamaguchi N, Chakraborty A, Pasek DA, Molkentin JD, Meissner G. Dysfunctional ryanodine receptor and cardiac hypertrophy: role of signaling molecules. Am J Physiol 2011; 300:H2187-95; http://dx.doi.org/10.1152/ajpcell.00370.2010
  • Huang TQ, Zou MX, Pasek DA, Meissner G. mTOR signaling in mice with dysfunctional cardiac ryanodine receptor ion channel. J Receptor Ligand Channel Res 2015; 8:43-51; PMID:26312014
  • Fischer P, Hilfiker-Kleiner D. Survival pathways in hypertrophy and heart failure: The gp130-STAT3 axis. Basic Res Cardiol 2007; 102:393-411; PMID:17918316; http://dx.doi.org/10.1007/s00395-007-0674-z
  • Fahmi A, Smart N, Punn A, Jabr R, Marber M, Heads R. p42/p44-MAPK and PI3K are sufficient for IL-6 family cytokines/gp130 to signal to hypertrophy and survival in cardiomyocytes in the absence of JAK/STAT activation. Cell Signal 2013; 25:898-909; PMID:23268184; http://dx.doi.org/10.1016/j.cellsig.2012.12.008
  • Levy DE, Lee CK. What does Stat3 do? J Clin Invest 2002; 109:1143-8; PMID:11994402; http://dx.doi.org/10.1172/JCI0215650
  • Hilfiker-Kleiner D, Hilfiker A, Drexler H. Many good reasons to have STAT3 in the heart. Pharmacol Ther 2005; 107:131-7; PMID:15963355; http://dx.doi.org/10.1016/j.pharmthera.2005.02.003
  • Kunisada K, Tone E, Fujio Y, Matsui H, Yamauchi-Takihara K, Kishimoto T. Activation of gp130 transduces hypertrophic signals via STAT3 in cardiac myocytes. Circulation 1998; 98:346-52; PMID:9711940; http://dx.doi.org/10.1161/01.CIR.98.4.346
  • Yasukawa H, Hoshijima M, Gu Y, Nakamura T, Pradervand S, Hanada T, Hanakawa Y, Yoshimura A, Ross J, Jr., Chien KR. Suppressor of cytokine signaling-3 is a biomechanical stress-inducible gene that suppresses gp130-mediated cardiac myocyte hypertrophy and survival pathways. J Clin Invest 2001; 108:1459-67; PMID:11714737; http://dx.doi.org/10.1172/JCI13939
  • Haghikia A, Stapel B, Hoch M, Hilfiker-Kleiner D. STAT3 and cardiac remodeling. Heart Fail Rev 2011; 16:35-47; PMID:20407820; http://dx.doi.org/10.1007/s10741-010-9170-x
  • Knight R, Scarabelli T, Spephanou A. STAT transcription in the ischemic heart. JAK-STAT 2012; 1:111-7; PMID:24058759; http://dx.doi.org/10.4161/jkst.20078
  • Uozumi H, Hiroi Y, Zou Y, Takimoto E, Toko H, Niu P, Shimoyama M, Yazaki Y, Nagai R, Komuro I. gp130 plays a critical role in pressure overload-induced cardiac hypertrophy. J Biol Chem 2001; 276:23115-9; PMID:11262406; http://dx.doi.org/10.1074/jbc.M100814200
  • Yamauchi-Takihara K, Kishimoto T. A novel role for STAT3 in cardiac remodeling. Trends Cardiovasc Med 2000; 10:298-303; PMID:11343970; http://dx.doi.org/10.1016/S1050-1738(01)00066-4
  • Kiuchi N, Nakajima K, Ichiba M, Fukada T, Narimatsu M, Mizuno K, Hibi M, Hirano T. STAT3 is required for the gp130-mediated full activation of the c-myc gene. J Exp Med 1999; 189:63-73; PMID:9874564; http://dx.doi.org/10.1084/jem.189.1.63
  • Mir SA, Chatterjee A, Mitra A, Pathak K, Mahata SK, Sarkar S. Inhibition of signal transducer and activator of transcription 3 (STAT3) attenuates interleukin-6 (IL-6)-induced collagen synthesis and resultant hypertrophy in rat heart. J Biol Chem 2012; 287:2666-77; PMID:22157761; http://dx.doi.org/10.1074/jbc.M111.246173
  • Yang E, Lerner L, Besser DJ Jr. Independent and cooperative activation of chromosomal c-fos promoter by STAT3. J Biol Chem 2003; 278:15794-9; PMID:12600988; http://dx.doi.org/10.1074/jbc.M213073200
  • Hirota H, Yoshida K, Kishimoto T, Taga T. Continuous activation of gp130, a signal-transducing receptor component for interleukin 6-related cytokines, causes myocardial hypertrophy in mice. Proc Natl Acad Sci U S A 1995; 92:4862-6; PMID:7539136; http://dx.doi.org/10.1073/pnas.92.11.4862
  • Szabo-Fresnais N, Lefebvre F, Germain A, Fischmeister R, Pomerance M. A new regulation of IL-6 production in adult cardiomyocytes by β-adrenergic and IL-1 β receptors and induction of cellular hypertrophy by IL-6 trans-signalling. Cell Signal 2010; 22:1143-52; PMID:20227492; http://dx.doi.org/10.1016/j.cellsig.2010.03.009
  • Huang TQ, Lee JH, Zou MX, Pasek DA, Willis MS, Meissner G. STAT3 activatrion in cardiac hypertrophy induced by ryanodine receptor 2 mutation. FASEB J 2013; 27:386.5.
  • Chen W, Shen X, Xia X, Xu G, Ma T, Bai X, Liang T. NSC 74859-mediated inhibition of STAT3 enhances the anti-proliferative activity of cetuximab in hepatocellular carcinoma. Liver Int 2012; 32:70-7; PMID:22098470; http://dx.doi.org/10.1111/j.1478-3231.2011.02631.x
  • Yang J, Liao X, Agarwal MK, Barnes L, Auron PE, Stark GR. Unphosphorylated STAT3 accumulates in response to IL-6 and activates transcription by binding to NFkappaB. Genes & Devel 2007; 21:1396-408; http://dx.doi.org/10.1101/gad.1553707
  • Kopf M, Baumann H, Freer G, Freudenberg M, Lamers M, Kishimoto T, Zinkernagel R, Bluethmann H, Kohler G. Impaired immune and acute-phase responses in interleukin-6-deficient mice. Nature 1994; 368:339-42; PMID:8127368; http://dx.doi.org/10.1038/368339a0
  • Siddiquee K, Zhang S, Guida WC, Blaskovich MA, Greedy B, Lawrence HR, Yip MLR, Jove R, McLaughlin MM, Lawrence NJ, Sebti, SM, Turkson, J. Selective chemical probe inhibitor of Stat3, identified through structure-based virtual screening, induces antitumor activity. Proc Natl Acad Sci U S A 2007; 104:7391-6; PMID:17463090; http://dx.doi.org/10.1073/pnas.0609757104
  • Ancey C, Corbi P, Froger J, Delwail A, Wijdenes J, Gascan H, Potreau D, Lecron J. Secretion of IL-6, IL-11 and LIF by human cardiomyocytes in primary culture. Cytokine 2002; 18:199-205; PMID:12126642; http://dx.doi.org/10.1006/cyto.2002.1033
  • Melendez GC, McLarty JL, Levick SP, Du Y, Janicki JS, Brower GL. Interleukin 6 mediates myocardial fibrosis, concentric hypertrophy, and diastolic dysfunction in rats. Hypertension 2010; 56:225-31; PMID:20606113; http://dx.doi.org/10.1161/HYPERTENSIONAHA.109.148635
  • Coles B, Fielding CA, Rose-John S, Scheller J, Jones SA, O'Donnell VB. Classic interleukin-6 receptor signaling and interleukin-6 trans-signaling differentially control angiotensin II-dependent hypertension, cardiac signal transducer and activator of transcription-3 activation, and vascular hypertrophy in vivo. Am J Pathol 2007; 171:315-25; PMID:17591976; http://dx.doi.org/10.2353/ajpath.2007.061078
  • Lai NC, Gao MH, Tang E, Tang R, Guo T, Dalton ND, Deng A, Tang T. Pressure overload-induced cardiac remodeling and dysfunction in the absence of interleukin 6 in mice. Lab Invest 2012; 92:1518-26; PMID:22825686; http://dx.doi.org/10.1038/labinvest.2012.97
  • Izumo S, Nadal-Ginard B, Mahdavi V. Protooncogene induction and reprogramming of cardiac gene expression produced by pressure overload. Proc Natl Acad Sci USA 1988; 85:339-43; PMID:2963328; http://dx.doi.org/10.1073/pnas.85.2.339
  • Johnson AW, Kinzenbaw DA, Modrick ML, Faraci FM. Small-molecule inhibitors of signal transducer and activator of transcription 3 protect against angiotensin II-induced vascular dysfunction and hypertension. Hypertension 2013; 61:437-42; PMID:23266544; http://dx.doi.org/10.1161/HYPERTENSIONAHA.111.00299
  • Yang J, Stark G. Roles of unphoshorylated STATs in signaling. Cell Res 2008; 18:443-51; PMID:18364677; http://dx.doi.org/10.1038/cr.2008.41
  • Matsui H, Ihara Y, Fujio Y, Kunisada K, Akira S, Kishimoto T, Yamauchi-Takihara K. Induction of interleukin (IL)-6 by hypoxia is mediated by nuclear factor (NF)-κB and NF-IL6 in cardiac myocytes. Cardiovasc Res 1999; 42:104-12; PMID:10435001; http://dx.doi.org/10.1016/S0008-6363(98)00285-5
  • Juretić N, García-Huidobro P, Iturrieta JA, Jaimovich E, Riveros N. Depolarization-induced slow Ca2+ transients stimulate transcription of IL-6 gene in skeletal muscle cells. Am J Physiol 2006; 290:C1428-36; http://dx.doi.org/10.1152/ajpcell.00449.2005
  • Carroll CJ, Sayan BS, Bailey SG, McCormick J, Stephanou A, Latchman DS, Townsend PA. Regulation of myocardial interleukin-6 expression by p53 and STAT1. J Interferon Cytokine Res 2013; 33:542-8; PMID:23675777; http://dx.doi.org/10.1089/jir.2012.0165
  • Haghikia A, Hoch M, Stapel B, Hilfiker-Kleiner D. STAT3 regulation of and by microRNAs in development and disease. JAK-STAT 2012; 1:143-50; PMID:24058763; http://dx.doi.org/10.4161/jkst.19573