IL-33 and ST2 comprise a critical biomechanically induced and cardioprotective signaling system

Shoji Sanada, Daihiko Hakuno, Luke J Higgins, Eric R Schreiter, Andrew N J McKenzie, Richard T Lee, Shoji Sanada, Daihiko Hakuno, Luke J Higgins, Eric R Schreiter, Andrew N J McKenzie, Richard T Lee

Abstract

ST2 is an IL-1 receptor family member with transmembrane (ST2L) and soluble (sST2) isoforms. sST2 is a mechanically induced cardiomyocyte protein, and serum sST2 levels predict outcome in patients with acute myocardial infarction or chronic heart failure. Recently, IL-33 was identified as a functional ligand of ST2L, allowing exploration of the role of ST2 in myocardium. We found that IL-33 was a biomechanically induced protein predominantly synthesized by cardiac fibroblasts. IL-33 markedly antagonized angiotensin II- and phenylephrine-induced cardiomyocyte hypertrophy. Although IL-33 activated NF-kappaB, it inhibited angiotensin II- and phenylephrine-induced phosphorylation of inhibitor of NF-kappa B alpha (I kappa B alpha) and NF-kappaB nuclear binding activity. sST2 blocked antihypertrophic effects of IL-33, indicating that sST2 functions in myocardium as a soluble decoy receptor. Following pressure overload by transverse aortic constriction (TAC), ST2(-/-) mice had more left ventricular hypertrophy, more chamber dilation, reduced fractional shortening, more fibrosis, and impaired survival compared with WT littermates. Furthermore, recombinant IL-33 treatment reduced hypertrophy and fibrosis and improved survival after TAC in WT mice, but not in ST2(-/-) littermates. Thus, IL-33/ST2 signaling is a mechanically activated, cardioprotective fibroblast-cardiomyocyte paracrine system, which we believe to be novel. IL-33 may have therapeutic potential for beneficially regulating the myocardial response to overload.

Figures

Figure 1. IL-33 is induced by mechanical…
Figure 1. IL-33 is induced by mechanical strain in cardiac fibroblasts.
(A and B) Quantitative analyses of gene expression of IL-33 by quantitative PCR (A) and sST2 by Northern analysis (B) in rat neonatal cardiomyocytes (white bars) and fibroblasts (black bars) are shown above with representative images from Northern analyses of cardiac fibroblast RNA. Cells were subjected to cyclic strain (8%, 1 Hz) for the indicated periods. Values are relative to β-tubulin expression and are expressed as percentage of control in cardiac fibroblasts. Data are from at least 3 sets of independent experiments. *P < 0.05, **P < 0.01 versus baseline. (C) Coomassie stain showed that the recombinant mature rat and human IL-33 with N-terminal His tag (10 and 3 μg protein, respectively, was loaded) were of high purity. (D) Pull-down assay of recombinant rat IL-33 with mouse ST2L-Fc protein. The recombinant protein exhibited specific binding to mouse ST2. (E) Western analysis of cardiomyocytes and cardiac fibroblasts subjected to cyclic strain (each 10 μg protein sample from whole cell lysate) for the indicated periods. For reference, 0.1 ng of recombinant IL-33 was applied in the right lane. (F) Representative immunofluorescence microscopy images of left ventricular samples 1 week after sham operation or TAC. Anti-vimentin (top panels) or anti–discoidin domain receptor–2 (DDR-2; bottom panels) antibody was used to detect fibroblasts (red) for dual staining with IL-33 (green). Pressure overload by TAC induced IL-33 expression, particularly in noncardiomyocyte interstitial cells. Scale bar: 10 μm.
Figure 2. IL-33 blocks prohypertrophic stimuli in…
Figure 2. IL-33 blocks prohypertrophic stimuli in cardiomyocytes and sST2 inhibits IL-33.
(A) IL-33 demonstrated a nonsignificant trend toward stimulating hypertrophy, but IL-33 blocked angiotensin II– and phenylephrine-induced (Phe) leucine uptake in a dose-dependent manner. (B) sST2 dose-dependently reversed the antihypertrophic effect of IL-33 under angiotensin II and phenylephrine. (C) Anti-ST2L monoclonal antibody, which blocks membrane-bound ST2L receptor–binding activity, blocked the antihypertrophic effect of IL-33, unlike control IgG. (D) Leucine uptake was not affected by sST2 compared with either baseline or control protein IL-1R–related protein 2 (IL-1Rrp2), but further enhanced hypertrophy under angiotensin II and phenylephrine. Data are from 3–5 sets of experiments. (E) Quantitative analysis of in vitro cell size measurements of cardiomyocytes was consistent with leucine incorporation assays (n = 200 each). *P < 0.05 versus baseline; #P < 0.05. (F) Induced secretion of both sST2 and IL-33 can reduce free IL-33. Cardiac fibroblasts were treated with indicated doses of PMA for 24 hours to induce sST2 and IL-33. In the top blots, conditioned media (20 μl) were analyzed by Western analysis. PMA dose-dependently increased secretion of both IL-33 and sST2. Below, media were preincubated in the presence or absence of 20 μg sST2-Fc protein and then incubated with presaturated beads for 2 hours. Samples preincubated with sST2-Fc had little IL-33, indicating that preincubation with sST2-Fc removed free IL-33. PMA dose-dependently decreased free IL-33 despite an increase in overall IL-33; these data suggest that induced sST2 can function as a decoy receptor, decreasing free IL-33.
Figure 3. IL-33 transiently activates NF-κB but…
Figure 3. IL-33 transiently activates NF-κB but blocks NF-κB activation by hypertrophic stimuli.
(A and B) NF-κB nuclear binding activity measured by EMSA in cardiomyocytes (A) and cardiac fibroblasts (B). (C and D) IκBα phosphorylation evaluated by Western analysis in cardiomyocytes (C) and cardiac fibroblasts (D). Values are relative to control density and are expressed as percent increase compared with control. Both angiotensin II and phenylephrine significantly activated NF-κB. IL-33 also activated NF-κB, but IL-33 markedly attenuated angiotensin II– and phenylephrine-induced NF-κB activation in cardiomyocytes, unlike in cardiac fibroblasts. IκBα phosphorylation was similarly affected by IL-33 treatment. (E) IL-33 (10 ng/ml) did not block IκBα phosphorylation (Western analysis) and NF-κB activity (EMSA) induced by PDGF-BB (10 ng/ml) or TNF-α (10 ng/ml), unlike angiotensin II and phenylephrine. (F) Western analysis for MAPKs and Akt in cardiomyocytes. IL-33 (10 ng/ml) activated all MAPKs, generally to a lesser extent than did IL-1β (10 ng/ml). IL-33 attenuated angiotensin II–induced phosphorylation of p38 MAPK and JNK, but not ERK or Akt. Data are from 4–5 sets of experiments. (G and H) GPCR agonist–induced ROS generation, as measured by 2,7-dichlorodihydrofluorecein diacetate, in cardiomyocytes (G) and cardiac fibroblasts (H). Both angiotensin II and phenylephrine significantly induced ROS generation, which was inhibited by IL-33, in cardiomyocytes. These data suggest that IL-33 can inhibit ROS-dependent hypertrophic signals. *P < 0.05 versus baseline; #P < 0.05 versus the same treatment group with IL-33.
Figure 4. IL-33/ST2 signaling is cardioprotective in…
Figure 4. IL-33/ST2 signaling is cardioprotective in vivo.
(A) Representative H&E and Sirius red stains and (B) quantitative analyses of samples from each group. Computer-based image analysis was used for measurements. ST2–/– mice developed more cardiomyocyte hypertrophy and cardiac fibrosis after TAC than did WT mice. Furthermore, treatment with IL-33 (2 μg/d i.p.) significantly improved these changes in WT mice, but not in ST2–/– mice. C, nonoperated control. Scale bar: 10 μm. (C) Gross measurement of heart weight normalized to body weight was consistent with the histomorphometric analyses. *P < 0.05 versus nonoperated control (B) or sham-operated WT (C); ΧP < 0.05 versus the same treatment in WT; †P < 0.05 versus sham in the same group; #P < 0.05.
Figure 5. IL-33/ST2 signaling is cardioprotective in…
Figure 5. IL-33/ST2 signaling is cardioprotective in vivo.
(A) Echocardiographic analysis at 4 weeks after operation demonstrated increased left ventricular mass, left ventricular wall thickness, and reduced fractional shortening in ST2–/– mice. Treatment with IL-33 reduced hypertrophy only in WT mice. IL-33 caused no significant change under non-stress conditions in vivo. n = 10 (nonoperated control); 8 (WT sham); 10 (WT TAC); 8 (WT sham + IL-33); 10 (WT TAC + IL-33); 8 (ST2–/– sham); 12 (ST2–/– TAC); 8 (ST2–/– sham + IL-33); and 10 (ST2–/– TAC + IL-33). (B) Representative images and (C) quantitative analysis of mRNA expression of ANP and BNP relative to internal control (18S) in the left ventricle at 1 week after operation, as assessed by Northern analysis. White and black bars indicate sham-operated and TAC, respectively. (D) NF-κB activation from EMSA in vivo 1 week after operation. ANP and BNP expression and NF-κB activity increased in ST2–/– mice compared with WT mice; IL-33 reversed these changes only in WT mice. Positive and negative control mixtures as well as specific competition mixtures and supershift induce by p65 antibody are also shown. *P < 0.05 versus nonoperated control (A) or sham-operated WT (C); ΧP < 0.05 versus the same treatment in WT; †P < 0.05 versus sham in the same group; #P < 0.05.
Figure 6. IL-33 improves survival after TAC…
Figure 6. IL-33 improves survival after TAC and reduces TAC-induced macrophage infiltration, but does not inhibit apoptosis in vivo.
TAC was performed on WT and ST2–/– littermates. (A) Quantitative analysis of macrophages and TUNEL stain–positive nuclei. Computer-based image analysis was used. TAC increased macrophage infiltration after 1 week of operation. IL-33 alone did not induce macrophage infiltration in either WT or ST2–/– mice but co-treatment reduced macrophage infiltration after TAC only in WT mice. TAC approximately doubled the number of TUNEL-positive nuclei after 4 weeks in both WT and ST2–/– mice. IL-33 treatment did not affect TUNEL positivity. (B) Kaplan-Meier survival curve analysis revealed that the survival of ST2–/– mice under TAC was significantly reduced compared with that of WT mice. This experiment was blinded so that all procedures were performed without knowledge of mouse genotype. (C) Serial echocardiographic analysis of surviving mice revealed that ST2–/– mice had increased left ventricular mass and left ventricular wall thickness and reduced contractile function compared with WT mice. *P < 0.05 versus sham-operated WT (A) or baseline (C); ΧP < 0.05 versus the same treatment in WT; †P < 0.05 versus Sham in the same group; #P < 0.05.
Figure 7. Rat IL-33 has weaker potency…
Figure 7. Rat IL-33 has weaker potency than human IL-33 and causes focal pulmonary inflammation in mice.
(A) Representative specimens of lung (H&E stain; original magnification, ×100) from mice 1 week after sham operation with or without 7 days’ treatment with rat IL-33 (2 μg/d). In the pulmonary parenchyma of WT mice, IL-33 treatment led to mild focal infiltrations of inflammatory cells (arrows) within and adjacent to vessels. This was not seen in vehicle-treated mice. These changes were not observed in ST2–/– mice with or without IL-33 treatment. (B) Time- and dose-dependent activation of ERK, JNK, p38, and p65–NF-κB by mouse IL-1β and recombinant mature rat or human IL-33 are shown. Mature rat IL-33 showed relatively weaker activation of MAPKs and NF-κB than the other proteins.

References

    1. Sadoshima J., Izumo S. The cellular and molecular response of cardiac myocytes to mechanical stress. Annu. Rev. Physiol. 1997;59:551–571.
    1. McKinsey T.A., Olson E.N. Toward transcriptional therapies for the failing heart: chemical screens to modulate genes. J. Clin. Invest. . 2005;115:538–546. doi: 10.1172/JCI200524144.
    1. Diez J., Gonzalez A., Lopez B., Querejeta R. Mechanisms of disease: pathologic structural remodeling is more than adaptive hypertrophy in hypertensive heart disease. Nat. Clin. Pract. Cardiovasc. Med. 2005;2:209–216.
    1. Baudino T., Carver W., Giles W.R., Borg T.K. Cardiac fibroblasts: friend or foe? Am. J. Physiol. Heart Circ. Physiol. 2006;291:H1015–H1026.
    1. Grossman W., Jones D., McLaurin L.P. Wall stress and patterns of hypertrophy in the human left ventricle. J. Clin. Invest. 1975;56:56–64.
    1. Weinberg E.O., et al. Expression and regulation of ST2, an interleukin-1 receptor family member, in cardiomyocytes and myocardial infarction. Circulation. 2002;106:2961–2966.
    1. Iwahana H., et al. Different promoter usage and multiple transcription initiation sites of the interleukin-1 receptor-related human ST2 gene in UT-7 and TM12 cells. Eur. J. Biochem. 1999;264:397–406.
    1. Weinberg E.O., et al. Identification of serum soluble ST2 receptor as a novel heart failure biomarker. Circulation. 2003;107:721–726.
    1. Shimpo M., et al. Serum levels of the interleukin-1 receptor family member ST2 predict mortality and clinical outcome in acute myocardial infarction. Circulation. 2004;109:2186–2190.
    1. Kumar S., Minnich M.D., Young P.R. ST2/T1 protein functionally binds to two secreted proteins from Balb/c 3T3 and human umbilical vein endothelial cells but does not bind interleukin 1. J. Biol. Chem. 1995;270:27905–27913.
    1. Gayle M.A., et al. Cloning of a putative ligand for the T1/ST2 receptor. J. Biol. Chem. 1996;271:5784–5789.
    1. Schmitz J., et al. IL-33, an interleukin-1-like cytokine that signals via the IL-1 receptor-related protein ST2 and induces T helper type 2-associated cytokines. Immunity. 2005;23:479–490.
    1. Manabe I., Shindo T., Nagai R. Gene expression in fibroblasts and fibrosis: involvement in cardiac hypertrophy. Circ. Res. 2002;91:1103–1113.
    1. Marian A.J. Pathogenesis of diverse clinical and pathological phenotypes in hypertrophic cardiomyopathy. Lancet. 2000;355:58–60.
    1. Goldsmith E.C., et al. Organization of fibroblasts in the heart. Dev. Dyn. 2004;230:787–794.
    1. Kawano S., et al. Blockade of NF-kappaB ameliorates myocardial hypertrophy in response to chronic infusion of angiotensin II. Cardiovasc. Res. 2005;67:689–698.
    1. Hirotani S., et al. Involvement of nuclear factor-kappaB and apoptosis signal-regulating kinase 1 in G-protein-coupled receptor agonist-induced cardiomyocyte hypertrophy. Circulation. 2002;105:509–515.
    1. Townsend M.J., Fallon P.G., Matthews D.J., Jolin H.E., McKenzie A.N. T1/ST2-deficient mice demonstrate the importance of T1/ST2 in developing primary T helper cell type 2 responses. J. Exp. Med. 2000;191:1069–1076.
    1. Kuster G.M., et al. Mineralocorticoid receptor inhibition ameliorates the transition to myocardial failure and decreases oxidative stress and inflammation in mice with chronic pressure overload. Circulation. . 2005;111:420–427.
    1. Shioi T., et al. Increased expression of interleukin-1 beta and monocyte chemotactic and activating factor/monocyte chemoattractant protein-1 in the hypertrophied and failing heart with pressure overload. Circ. Res. 1997;81:664–671.
    1. Okada K., 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;110:705–712.
    1. Yang G., et al. Cyclosporine reduces left ventricular mass with chronic aortic banding in mice, which could be due to apoptosis and fibrosis. J. Mol. Cell. Cardiol. 2001;33:1505–1514.
    1. Dunne A., O’Neill L.A. The interleukin-1 receptor/Toll-like receptor superfamily: signal transduction during inflammation and host defense. Sci. STKE. 2003;2003:re3.
    1. Matsuzawa A., et al. ROS-dependent activation of the TRAF6-ASK1-p38 pathway is selectively required for TLR4-mediated innate immunity. Nat. Immunol. 2005;6:587–592.
    1. Ninomiya-Tsuji J., et al. The kinase TAK1 can activate the NIK-I kappaB as well as the MAP kinase cascade in the IL-1 signalling pathway. Nature. 1999;398:252–256.
    1. Zhang D., et al. TAK1 is activated in the myocardium after pressure overload and is sufficient to provoke heart failure in transgenic mice. Nat. Med. . 2000;6:556–563.
    1. Purcell N.H., et al. Activation of NF-kappa B is required for hypertrophic growth of primary rat neonatal ventricular cardiomyocytes. Proc. Natl. Acad. Sci. U. S. A. 2001;98:6668–6673.
    1. Freund C., et al. Requirement of nuclear factor-kappaB in angiotensin II- and isoproterenol-induced cardiac hypertrophy in vivo. Circulation. 2005;111:2319–2325.
    1. Ha T., et al. Reduced cardiac hypertrophy in toll-like receptor 4-deficient mice following pressure overload. Cardiovasc. Res. . 2005;68:224–234.
    1. Hoffmann A., Baltimore D. Circuitry of nuclear factor kappaB signaling. Immunol. Rev. 2006;210:171–186.
    1. Nelson D.E., et al. Oscillations in NF-kappaB signaling control the dynamics of gene expression. Science. 2004;306:704–708.
    1. Rockman H.A., Wachhorst S.P., Mao L., Ross J.J. ANG II receptor blockade prevents ventricular hypertrophy and ANF gene expression with pressure overload in mice. Am. J. Physiol. . 1994;266:H2468–H2475.
    1. Rapacciuolo A., et al. Important role of endogenous norepinephrine and epinephrine in the development of in vivo pressure-overload cardiac hypertrophy. J. Am. Coll. Cardiol. 2001;38:876–882.
    1. Ha T., et al. Blockade of MyD88 attenuates cardiac hypertrophy and decreases cardiac myocyte apoptosis in pressure overload-induced cardiac hypertrophy in vivo. Am. J. Physiol. Heart Circ. Physiol. 2006;290:H985–H994.
    1. Hingtgen S.D., et al. Nox2-containing NADPH oxidase and Akt activation play a key role in angiotensin II-induced cardiomyocyte hypertrophy. Physiol. Genomics. 2006;26:180–191.
    1. Nishida M., et al. G alpha 12/13- and reactive oxygen species-dependent activation of c-Jun NH2-terminal kinase and p38 mitogen-activated protein kinase by angiotensin receptor stimulation in rat neonatal cardiomyocytes. J. Biol. Chem. . 2005;280:18434–18441.
    1. Li Q., et al. Nox2 and Rac1 regulate H2O2-dependent recruitment of TRAF6 to endosomal interleukin-1 receptor complexes. Mol. Cell. Biol. 2006;26:140–154.
    1. Sarkar S., Vellaichamy E., Young D., Sen S. Influence of cytokines and growth factors in ANG II-mediated collagen upregulation by fibroblasts in rats: role of myocytes. Am. J. Physiol. Heart Circ. Physiol. . 2004;287:H107–H117.
    1. Saccani S., Polentarutti N., Penton-Rol G., Sims J.E., Mantovani A. Divergent effects of LPS on expression of IL-1 receptor family members in mononuclear phagocytes in vitro and in vivo. Cytokine. 1998;10:773–780.
    1. May L.T., Ndubuisi M.I., Patel K., Garcia D. Interleukin-6 chaperones in blood. Ann. N. Y. Acad. Sci. 1995;762:120–128.
    1. Economides A.N., et al. Cytokine traps: multi-component, high-affinity blockers of cytokine action. Nat. Med. 2003;9:47–52.
    1. Hsieh P.C., Davis M.E., Gannon J., MacGillivray C., Lee R.T. Controlled delivery of PDGF-BB for myocardial protection using injectable self-assembling peptide nanofibers. J. Clin. Invest. 2006;116:237–248. doi: 10.1172/JCI25878.
    1. Yokoyama T., et al. 1999Angiotensin II and mechanical stretch induce production of tumor necrosis factor in cardiac fibroblasts . Am. J. Physiol. 276H1968–H1976.
    1. Yamamoto K., et al. Induction of tenascin-C in cardiac myocytes by mechanical deformation. Role of reactive oxygen species. J. Biol. Chem. 1999;274:21840–21846.
    1. Hakuno D., Takahashi T., Lammerding J., Lee R.T. Focal adhesion kinase signaling regulates cardiogenesis of embryonic stem cells. J. Biol. Chem. 2005;280:39534–39544.
    1. Yamamoto K., Ohki R., Lee R.T., Ikeda U., Shimada K. Peroxisome proliferator-activated receptor gamma activators inhibit cardiac hypertrophy in cardiac myocytes. Circulation. 2001;104:1670–1675.
    1. Frantz S., et al. Toll4 (TLR4) expression in cardiac myocytes in normal and failing myocardium. J. Clin. Invest. 1999;104:271–280.
    1. Schulze P.C., et al. Hyperglycemia promotes oxidative stress through inhibition of thioredoxin function by thioredoxin-interacting protein. J. Biol. Chem. 2004;279:30369–30374.
    1. Rockman H.A., et al. Segregation of atrial-specific and inducible expression of an atrial natriuretic factor transgene in an in vivo murine model of cardiac hypertrophy. Proc. Natl. Acad. Sci. U. S. A. 1991;88:8277–8281.
    1. Tanaka N., et al. Transthoracic echocardiography in models of cardiac disease in the mouse. Circulation. 1996;94:1109–1117.
    1. Collins K.A., et al. Accuracy of echocardiographic estimates of left ventricular mass in mice. Am. J. Physiol. Heart Circ. Physiol. . 2001;280:H1954–H1962.
    1. Takemoto M., et al. Important role of tissue angiotensin-converting enzyme activity in the pathogenesis of coronary vascular and myocardial structural changes induced by long-term blockade of nitric oxide synthesis in rats. J. Clin. Invest. 1997;99:278–287.

Source: PubMed

3
Prenumerera