throbber
Therapeutic Activation of Signal Transducer and Activator
`of Transcription 3 by Interleukin-11 Ameliorates Cardiac
`Fibrosis After Myocardial Infarction
`
`Masanori Obana, MS; Makiko Maeda, PhD; Koji Takeda, MD; Akiko Hayama, MS;
`Tomomi Mohri, PhD; Tomomi Yamashita, BS; Yoshikazu Nakaoka, MD, PhD;
`Issei Komuro, MD, PhD; Kiyoshi Takeda, MD, PhD; Goro Matsumiya, MD, PhD;
`Junichi Azuma, MD; Yasushi Fujio, MD, PhD
`
`Background—Glycoprotein 130 is the common receptor subunit for the interleukin (IL)-6 cytokine family. Previously, we
`reported that pretreatment of IL-11, an IL-6 family cytokine, activates the glycoprotein 130 signaling pathway in
`cardiomyocytes and prevents ischemia/reperfusion injury in vivo; however, its long-term effects on cardiac remodeling
`after myocardial infarction (MI) remain to be elucidated.
`Methods and Results—MI was generated by ligating the left coronary artery in C57BL/6 mice. Real-time reverse
`transcription polymerase chain reaction analyses showed that IL-11 mRNA was remarkably upregulated in the hearts
`exposed to MI. Intravenous injection of IL-11 activated signal transducer and activator of transcription 3 (STAT3), a
`downstream signaling molecule of glycoprotein 130, in cardiomyocytes in vivo, suggesting that cardiac myocytes are
`target cells of IL-11 in the hearts. Twenty-four hours after coronary ligation, IL-11 was administered intravenously,
`followed by consecutive administration every 24 hours for 4 days. IL-11 treatment reduced fibrosis area 14 days after
`MI, attenuating cardiac dysfunction. Consistent with a previous report that STAT3 exhibits antiapoptotic and angiogenic
`activity in the heart, IL-11 treatment prevented apoptotic cell death of the bordering myocardium adjacent to the infarct
`zone and increased capillary density at the border zone. Importantly, cardiac-specific ablation of STAT3 abrogated
`IL-11–mediated attenuation of fibrosis and was associated with left ventricular enlargement. Moreover, with the use of
`cardiac-specific transgenic mice expressing constitutively active STAT3, cardiac STAT3 activation was shown to be
`sufficient to prevent adverse cardiac remodeling.
`Conclusions—IL-11 attenuated cardiac fibrosis after MI through STAT3. Activation of the IL-11/glycoprotein 130/STAT3
`axis may be a novel therapeutic strategy against cardiovascular diseases. (Circulation. 2010;121:684-691.)
`
`Key Words: interleukins 䡲 myocardial infarction 䡲 remodeling 䡲 signal transduction
`
`A fter myocardial injury, various kinds of neurohumoral
`
`factors and cytokines modulate cardiac remodeling.
`Among them,
`leukemia inhibitory factor
`(LIF) and
`cardiotrophin-1, which belong to the interleukin (IL)-6 fam-
`ily, play important roles in cardioprotection.1,2 LIF and
`cardiotrophin-1 are secreted from cardiomyocytes in response
`to pathological stress.3–5 These cytokines bind and activate
`LIF receptor in cardiomyocytes.6 Activated LIF receptor
`makes a dimer with glycoprotein 130 (gp130), followed by
`activation of signal transducer and activator of transcription 3
`(STAT3).7 STAT3 activation promotes cardiomyocyte sur-
`vival and vascular formation in the heart.8 –10 Thus, cardiac
`activation of the gp130/STAT3 system may be a potential
`
`therapeutic strategy against cardiovascular diseases; however,
`therapies targeting gp130 have not been proposed.
`
`Clinical Perspective on p 691
`
`The difficulty in therapeutic activation of gp130 is derived
`from its receptor system. Gp130 is expressed ubiquitously as
`the common receptor subunit of IL-6 family cytokines.11 IL-6
`family cytokines bind their specific receptor ␣ subunits,
`followed by activation of a common gp130 receptor. Pleio-
`tropic effects of IL-6 family cytokines are explained by the
`differential expression of receptor ␣ subunits. Most members
`of the IL-6 family, whose receptor ␣ subunits are expressed
`abundantly in inflammatory cells, would evoke severe in-
`
`Downloaded from http://ahajournals.org by on October 29, 2019
`
`Received July 14, 2009; accepted December 4, 2009.
`From the Department of Clinical Pharmacology and Pharmacogenomics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
`(M.O., A.H., T.M., T.Y., J.A., Y.F.); Department of Clinical Pharmacogenomics, School of Pharmacy, Hyogo University of Health Sciences, Hyogo,
`Japan (M.M., J.A.); and Department of Cardiovascular Surgery (K.T., G.M.), Department of Cardiovascular Medicine (Y.N., I.K.), and Laboratory of
`Immune Regulation, Department of Microbiology and Immunology (K.T.), Graduate School of Medicine, Osaka University, Osaka, Japan.
`Singapore Exhibit 2003
`The online-only Data Supplement is available with this article at http://circ.ahajournals.org/cgi/content/full/CIRCULATIONAHA.109.893677/DC1.
`Correspondence to Yasushi Fujio, MD, PhD, 1-6 Yamada-oka, Suita City, 565-0871, Osaka, Japan. E-mail fujio@phs.osaka-u.ac.jp
`Lassen v. Singapore et al.
`© 2010 American Heart Association, Inc.
`PGR2019-00053
`DOI: 10.1161/CIRCULATIONAHA.109.893677
`
`Circulation is available at http://circ.ahajournals.org
`
`684
`
`

`

`Therapeutic Activation of Signal Transducer and Activator
`of Transcription 3 by Interleukin-11 Ameliorates Cardiac
`Fibrosis After Myocardial Infarction
`
`Masanori Obana, MS; Makiko Maeda, PhD; Koji Takeda, MD; Akiko Hayama, MS;
`Tomomi Mohri, PhD; Tomomi Yamashita, BS; Yoshikazu Nakaoka, MD, PhD;
`Issei Komuro, MD, PhD; Kiyoshi Takeda, MD, PhD; Goro Matsumiya, MD, PhD;
`Junichi Azuma, MD; Yasushi Fujio, MD, PhD
`
`Background—Glycoprotein 130 is the common receptor subunit for the interleukin (IL)-6 cytokine family. Previously, we
`reported that pretreatment of IL-11, an IL-6 family cytokine, activates the glycoprotein 130 signaling pathway in
`cardiomyocytes and prevents ischemia/reperfusion injury in vivo; however, its long-term effects on cardiac remodeling
`after myocardial infarction (MI) remain to be elucidated.
`Methods and Results—MI was generated by ligating the left coronary artery in C57BL/6 mice. Real-time reverse
`transcription polymerase chain reaction analyses showed that IL-11 mRNA was remarkably upregulated in the hearts
`exposed to MI. Intravenous injection of IL-11 activated signal transducer and activator of transcription 3 (STAT3), a
`downstream signaling molecule of glycoprotein 130, in cardiomyocytes in vivo, suggesting that cardiac myocytes are
`target cells of IL-11 in the hearts. Twenty-four hours after coronary ligation, IL-11 was administered intravenously,
`followed by consecutive administration every 24 hours for 4 days. IL-11 treatment reduced fibrosis area 14 days after
`MI, attenuating cardiac dysfunction. Consistent with a previous report that STAT3 exhibits antiapoptotic and angiogenic
`activity in the heart, IL-11 treatment prevented apoptotic cell death of the bordering myocardium adjacent to the infarct
`zone and increased capillary density at the border zone. Importantly, cardiac-specific ablation of STAT3 abrogated
`IL-11–mediated attenuation of fibrosis and was associated with left ventricular enlargement. Moreover, with the use of
`cardiac-specific transgenic mice expressing constitutively active STAT3, cardiac STAT3 activation was shown to be
`sufficient to prevent adverse cardiac remodeling.
`Conclusions—IL-11 attenuated cardiac fibrosis after MI through STAT3. Activation of the IL-11/glycoprotein 130/STAT3
`axis may be a novel therapeutic strategy against cardiovascular diseases. (Circulation. 2010;121:684-691.)
`
`Key Words: interleukins 䡲 myocardial infarction 䡲 remodeling 䡲 signal transduction
`
`A fter myocardial injury, various kinds of neurohumoral
`
`factors and cytokines modulate cardiac remodeling.
`Among them,
`leukemia inhibitory factor
`(LIF) and
`cardiotrophin-1, which belong to the interleukin (IL)-6 fam-
`ily, play important roles in cardioprotection.1,2 LIF and
`cardiotrophin-1 are secreted from cardiomyocytes in response
`to pathological stress.3–5 These cytokines bind and activate
`LIF receptor in cardiomyocytes.6 Activated LIF receptor
`makes a dimer with glycoprotein 130 (gp130), followed by
`activation of signal transducer and activator of transcription 3
`(STAT3).7 STAT3 activation promotes cardiomyocyte sur-
`vival and vascular formation in the heart.8 –10 Thus, cardiac
`activation of the gp130/STAT3 system may be a potential
`
`therapeutic strategy against cardiovascular diseases; however,
`therapies targeting gp130 have not been proposed.
`
`Clinical Perspective on p 691
`
`The difficulty in therapeutic activation of gp130 is derived
`from its receptor system. Gp130 is expressed ubiquitously as
`the common receptor subunit of IL-6 family cytokines.11 IL-6
`family cytokines bind their specific receptor ␣ subunits,
`followed by activation of a common gp130 receptor. Pleio-
`tropic effects of IL-6 family cytokines are explained by the
`differential expression of receptor ␣ subunits. Most members
`of the IL-6 family, whose receptor ␣ subunits are expressed
`abundantly in inflammatory cells, would evoke severe in-
`
`Downloaded from http://ahajournals.org by on October 29, 2019
`
`Received July 14, 2009; accepted December 4, 2009.
`From the Department of Clinical Pharmacology and Pharmacogenomics, Graduate School of Pharmaceutical Sciences, Osaka University, Osaka, Japan
`(M.O., A.H., T.M., T.Y., J.A., Y.F.); Department of Clinical Pharmacogenomics, School of Pharmacy, Hyogo University of Health Sciences, Hyogo,
`Japan (M.M., J.A.); and Department of Cardiovascular Surgery (K.T., G.M.), Department of Cardiovascular Medicine (Y.N., I.K.), and Laboratory of
`Immune Regulation, Department of Microbiology and Immunology (K.T.), Graduate School of Medicine, Osaka University, Osaka, Japan.
`The online-only Data Supplement is available with this article at http://circ.ahajournals.org/cgi/content/full/CIRCULATIONAHA.109.893677/DC1.
`Correspondence to Yasushi Fujio, MD, PhD, 1-6 Yamada-oka, Suita City, 565-0871, Osaka, Japan. E-mail fujio@phs.osaka-u.ac.jp
`© 2010 American Heart Association, Inc.
`
`Circulation is available at http://circ.ahajournals.org
`
`DOI: 10.1161/CIRCULATIONAHA.109.893677
`
`684
`
`

`

`Obana et al
`
`IL-11 as a Novel Cardioprotective Cytokine
`
`685
`
`if administered
`flammation12 as a serious adverse event
`systemically. Therefore, to achieve clinical use of IL-6 family
`cytokines, the cytokine that induces only a tolerable level of
`inflammation should be selected.
`IL-11 is a hematopoietic IL-6 family cytokine with pleiotropic
`effects. IL-11 exhibits thrombopoietic activity, and recombinant
`human IL-11 is used clinically for thrombocytopenia.13 In
`contrast to other IL-6 family members, IL-11 exhibits anti-
`inflammatory activity against chronic inflammatory diseases,
`such as Crohn disease.14 Moreover, recombinant human IL-11
`protects epithelial cells of the intestine from tissue damage,
`suggesting its cytoprotective property.15 Recently, we demon-
`strated that the IL-11 receptor is expressed in cardiomyocytes
`and that pretreatment of IL-11 confers resistance to ischemia/
`reperfusion injury in a murine model as a preconditioning
`effect.16 When the limited level of clinical adverse effects of
`recombinant human IL-11 is considered,13 IL-11 may be a
`candidate to be available clinically as cardiac gp130-targeting
`therapy against heart diseases.
`In this study, we investigated the long-term effects of IL-11
`treatment after myocardial infarction (MI). In addition, we
`report that IL-11 treatment prevents adverse cardiac remod-
`eling through the STAT3 pathway.
`
`Methods
`
`Animal Care
`The care of all animals was in compliance with the Osaka University
`animal care guidelines. The investigation conforms to the Guide for
`the Care and Use of Laboratory Animals published by the US
`National Institutes of Health (National Institutes of Health publica-
`tion No. 85-23, revised 1996).
`
`Coronary Artery Ligation and IL-11 Treatment
`MI was generated by coronary artery ligation according to the
`previous report1 with minor modifications. Briefly, C57BL/6 mice (8
`to 10 weeks old; Japan SLC) were anesthetized and ventilated with
`80% oxygen containing 1.5% isoflurane (Merck). After left-side
`thoracotomy, the left coronary artery was ligated with 7-0 silk
`sutures. Infarction was confirmed by discoloration of the ventricle
`and ST-T changes on ECG. The chest and skin were closed with 5-0
`silk sutures. In preliminary experiments, we confirmed that the
`operation reproducibly generates infarction with the initial area at
`risk 20% to 25% per left ventricular (LV) area, as analyzed by Evans
`blue exclusion assays.16 Sham-operated mice were subjected to
`similar surgery, except that no ligature was placed. Twenty-four
`hours after MI operation, mice were randomly assigned to 2 groups:
`the IL-11 group and control group. In the IL-11 group, recombinant
`human IL-11 (Peprotech) was administered intravenously for 5 days
`consecutively;
`the control group received the same volume of
`phosphate-buffered saline (PBS) during the same period.
`
`Real-Time Reverse Transcription Polymerase
`Chain Reaction
`Real-time reverse transcription polymerase chain reaction (RT-PCR)
`was performed according to the manufacturer’s protocol. Total RNA
`was prepared from hearts at various time points after operation. In
`some experiments, the hearts were cut into 2 pieces: infarct area and
`remote area. The infarct area is the damaged or fibrotic region with
`its surrounding border zone, and the remote area is the portion
`separated from the infarct area by ⬎1 mm.
`Total RNA (1 ␮g) was subjected to first-strand cDNA synthesis
`with oligo(dT) primer. IL-11 was quantified by real-time RT-PCR
`with the use of the ABI-PRISM 7700 sequence detection system
`(Applied Biosystems Inc) with the SYBR green system (Applied
`
`Biosystems). As an internal control, the expression of GAPDH
`mRNA was estimated with the SYBR green system. The primers for
`IL-11 or GAPDH are as follows: IL-11 forward: 5⬘-CTGCCC-
`ACCTTGGCCATGAG-3⬘; IL-11 reverse: 5⬘-CCAGGCGAGACA-
`TCAAGAAAGA-3⬘; GAPDH forward: 5⬘-GCCGGTGCTGAGTAT-
`GTCGT-3⬘; GAPDH reverse: 5⬘-CCCTTTTGGCTCCACCCTT-3⬘.
`
`Immunoblot Analyses
`Immunoblot analyses were performed as described previously.17
`Heart homogenates were prepared in buffer containing 150 mmol/L
`NaCl, 10 mmol/L Tris-HCl (pH 7.5), 1 mmol/L EDTA, 1% Triton
`X-100, 1% deoxycholic acid, and 1 mmol/L dithiothreitol. Proteins
`were separated by SDS-PAGE and transferred onto the polyvinyli-
`dene difluoride membrane (Millipore). The membrane was immu-
`noblotted with anti-phospho-STAT3 (p-STAT3) (Cell Signaling
`Technology, Danvers, Mass), anti-Bcl-2 (BD Transduction Labora-
`tories), anti-survivin (Santa Cruz Biotechnology, Inc, Santa Cruz,
`Calif), or anti-cleaved caspase 3 (Cell Signaling Technology) anti-
`body. The membrane was reprobed with anti-STAT3 (Santa Cruz
`Biotechnology) or anti-GAPDH (Chemicon, Temecula, Calif) anti-
`body to show equal amount loading.
`
`Histological Estimation of Cardiac Fibrosis
`The frozen sections (5-␮m thick) were prepared from the portion
`⬇300 ␮m distal to the ligation point and stained with Masson’s
`trichrome. Photomicrographs were taken, and fibrotic circumference
`and area were measured with the use of Scion Image (Scion Corpora-
`tion) by a researcher who was blinded to the treatment. Fibrotic
`circumference and area were calculated as a percentage of LV circum-
`ference and area, respectively. Infarct wall thickness was measured
`perpendicular to the infarcted wall at 3 separate regions and averaged.
`
`Analysis of Cardiac Function
`Fourteen days after MI, mice were anesthetized (50 mg/kg pento-
`barbital) and heparinized (50 U) via intraperitoneal injection. The
`hearts were excised rapidly and placed in ice-cold modified Tyrode’s
`solution (140 mmol/L NaCl, 5.4 mmol/L KCl, 1.8 mmol/L CaCl2,
`0.45 mmol/L MgCl2, 0.33 mmol/L NaH2PO4, 5.5 mmol/L glucose,
`5 mmol/L HEPES [pH 7.4]). The aorta was cannulated and retro-
`gradely perfused at a constant pressure of 100 mm Hg with Tyrode’s
`solution bubbled with 80% oxygen at 37°C. The experiments were
`performed at 37°C by immersing the heart in Tyrode’s solution in a
`water-jacketed chamber. The hearts were paced at 420 bpm. The
`fluid-filled balloon was inserted into the LV to monitor cardiac
`function. The balloon was attached to a pressure transducer, which
`was coupled to a 4S PowerLab (AD Instruments). LV developed
`pressure and ⫾dP/dt were measured.
`
`Immunofluorescent Microscopic Analyses
`The hearts were harvested 15 minutes after intravenous injection of
`IL-11, and the frozen sections were prepared. The sections were
`stained with anti-p-STAT3 and anti-sarcomeric ␣-actinin (Sigma)
`antibodies. Alexa Fluor 488 – conjugated goat anti-rabbit IgG (Mo-
`lecular Probes) and Alexa Fluor 546 – conjugated goat anti-mouse
`IgG (Molecular Probes) were used as secondary antibodies. Nuclei
`were also stained with Hoechst 33258.
`Apoptotic cell death was detected by a terminal deoxynucleotide
`transferase–mediated dUTP nick-end labeling (TUNEL) staining
`with an in situ Apoptosis Detection Kit (TaKaRa). The section was
`costained with anti-sarcomeric ␣-actinin antibody to identify the
`cardiac myocytes. For quantitative analyses, apoptotic myocytes
`were counted in number by a researcher who was blinded to the
`assay conditions.
`
`Immunohistochemical Analyses
`The frozen sections were prepared as described above. Capillary
`density was examined by immunohistochemical staining with the use
`of the Vectastain ABC kit (Vector Laboratories) with anti-CD31
`antibody (BD Biosciences, San Jose, Calif). To estimate capillary
`
`Downloaded from http://ahajournals.org by on October 29, 2019
`
`

`

`Figure 1. The transcript of IL-11 was elevated after MI, and IL-11
`activated STAT3 in cardiomyocytes in murine hearts. A, Expression
`of IL-11 in infarct area or remote area of postinfarct hearts was
`analyzed by real-time RT-PCR. Expression of IL-11 was normalized
`with that of GAPDH. Data are shown as mean⫾SD (n⫽4 mice for
`each condition). *P⬍0.05 vs non-MI (sham) by 1-way ANOVA fol-
`lowed by Bonferroni test. B, IL-11 (8 ␮g/kg) was administered in-
`travenously in mice for the indicated time. The lysates from hearts
`were immunoblotted with anti-p-STAT3 antibody. Blots were
`reprobed with anti-STAT3 antibody. Representative data are
`shown (left). Quantitative analyses of p-STAT3 are shown (right).
`Data are shown as mean⫾SD (n⫽4 mice for each condition).
`*P⬍0.05 vs 0 minutes by 1-way ANOVA followed by Bonferroni
`test. C, Fifteen minutes after injection of IL-11 (8 ␮g/kg) or PBS,
`frozen sections were prepared from the hearts. The sections were
`costained with anti-p-STAT3 and anti-sarcomeric ␣-actinin anti-
`bodies. Hoechst 33258 staining was also performed to identify the
`nuclei. The images are representative of 20 fields obtained from 4
`mice. Bar⫽50 ␮m.
`
`IL-11 Administration Attenuates Cardiac
`Remodeling After MI
`To examine the effects of IL-11 on adverse cardiac remod-
`eling, IL-11 was administered to the mice after MI operation,
`and cardiac fibrosis was histologically estimated at day 14
`after MI (Figure 2). Both fibrotic circumference and fibrotic
`area were reduced by IL-11 in a dose-dependent manner
`(Figures 2B and C). Treatment of IL-11 at 8 ␮g/kg achieved
`a submaximal reduction in fibrotic circumference by 28.9%
`(PBS, 58.6⫾9.6%; IL-11, 41.7⫾10.0%) and fibrotic area by
`33.1% (PBS, 39.8⫾9.3%; IL-11, 26.6⫾7.4%). Interestingly,
`IL-11–treated hearts showed an increase in infarct wall
`thickness compared with PBS-treated hearts (Figure 2D). To
`
`686
`
`Circulation
`
`February 9, 2010
`
`density quantitatively, the number of capillaries was counted by a
`researcher who was blinded to the assay conditions.
`
`Conditional Ablation of the STAT3 Gene in
`Cardiomyocytes of Adult Murine Heart
`The transgenic mice in which Cre recombinase fused to the mutated
`estrogen receptor domains (MerCreMer) were driven by the cardio-
`myocyte-specific ␣-myosin heavy chain (␣-MHC) promoter, desig-
`nated as ␣-MHC-MerCreMer mice, were a gift from Dr Molkentin.18
`We crossed the ␣-MHC-MerCreMer mice with mice that carried
`floxed STAT3 alleles (STAT3flox/flox)19 and produced ␣-MHC-
`MerCreMer/STAT3flox/flox mutant mice. To induce Cre-mediated
`recombination, mice were treated with 20 mg/kg tamoxifen (Sigma,
`St Louis, Mo) by intraperitoneal
`injection once per day for 5
`consecutive days. Five days after the last treatment, the level of
`STAT3 expression decreased dramatically, and the mutant mice
`underwent MI as described above.
`
`Cardiac-Specific Transgenic Mice Expressing
`Constitutively Active STAT3
`Generation of cardiac-specific transgenic mice expressing constitu-
`tively active STAT3 was described previously.9
`
`Statistical Analysis
`Data are presented as mean⫾SD. Comparisons between 2 groups
`were performed with the use of the unpaired t
`test. One-way
`ANOVA with the Bonferroni test was used for multiple compari-
`sons. Differences were considered statistically significant when the
`calculated (2-tailed) P value was ⬍0.05.
`
`Results
`
`IL-11 Is a Cardiac Cytokine That Activates
`STAT3 in Cardiomyocytes In Vivo
`We analyzed the expression of IL-11 mRNA in hearts at various
`time points after MI. Hearts were separated into infarct area and
`remote area, and the expression of IL-11 mRNA was measured
`by real-time RT-PCR (Figure 1A). The expression of IL-11
`transcript was elevated, with its peak at 1 day after MI, and was
`gradually reduced at both infarct and remote areas. In the infarct
`area, the enhanced expression of IL-11 was sustained for 14
`days. These data indicate that IL-11 is produced in the heart
`during cardiac remodeling after MI.
`Next, we examined whether intravenous administration of
`IL-11 stimulates STAT3 in hearts by immunoblot analysis
`with anti-p-STAT3 antibody (Figure 1B). STAT3 phosphor-
`ylation was induced rapidly and reduced to the basal level
`within 180 minutes after IL-11 injection. IL-11 activated
`STAT3 in the heart in a dose-dependent manner (Figure I in
`the online-only Data Supplement).
`To confirm that STAT3 activation occurred in cardiomyo-
`cytes, we performed immunohistological analyses to detect
`the nuclear localization of activated p-STAT3 (Figure 1C).
`Nuclear staining of p-STAT3 was detected in the IL-11–
`treated hearts but not in untreated hearts. Notably, ⬎90% of
`nuclei of sarcomeric ␣-actinin–positive cells were also posi-
`tively stained with anti-p-STAT3 antibody, indicating that
`IL-11 treatment results in STAT3 activation in cardiomyo-
`cytes in vivo. As is the case with noninfarcted mice,
`p-STAT3 was localized mainly in cardiomyocyte nuclei of
`postinfarct hearts, and IL-11 treatment increased the fre-
`quency of p-STAT3–positive cardiac myocytes (Figure II in
`the online-only Data Supplement).
`
`Downloaded from http://ahajournals.org by on October 29, 2019
`
`

`

`Obana et al
`
`IL-11 as a Novel Cardioprotective Cytokine
`
`687
`
`Figure 2. IL-11 attenuated cardiac fibro-
`sis after MI. Heart sections (3 sections
`from each mouse) were prepared 14
`days after MI and stained with Masson’s
`trichrome. A, Images are representative
`of 24 to 27 obtained from 8 to 9 mice.
`Bar⫽1 mm. B to D, Ratio of fibrotic cir-
`cumference to LV circumference (B),
`ratio of fibrotic area to LV area (C), and
`infarct wall thickness (D) were quantita-
`tively estimated. Data are shown as
`mean⫾SD (n⫽8 mice for PBS; n⫽9 mice
`for 3 ␮g/kg; n⫽9 mice for 8 ␮g/kg; n⫽8
`mice for 20 ␮g/kg). *P⬍0.05 vs PBS by
`1-way ANOVA followed by Bonferroni
`test.
`
`examine the effects of IL-11 on LV hypertrophy, we analyzed
`expression of ␣-skeletal muscle actin mRNA, a well-known
`marker of LV hypertrophy (Figure III in the online-only Data
`Supplement). IL-11 treatment showed a tendency to reduce
`␣-skeletal muscle actin expression, although its reduction was
`not statistically significant.
`To clarify whether IL-11 prevents cardiac dysfunction after
`MI, we measured LV developed pressure and ⫾dP/dt by a
`Langendorff apparatus. As acute myocardial damage, MI rapidly
`impaired LV developed pressure and ⫾dP/dt at day 1 (Table I in
`the online-only Data Supplement) before IL-11 administration
`was started. Because IL-11 treatment attenuated cardiac fibrosis
`that occurred during the following 2 weeks, we examined the
`effects of IL-11 on cardiac function 2 weeks after MI (Table).
`IL-11 treatment ameliorated chronic cardiac dysfunction in a
`dose-dependent manner compared with the PBS-treated group.
`Consistent with the attenuation of fibrosis, treatment of IL-11 at
`a dose of 8 ␮g/kg submaximally prevented cardiac dysfunction.
`Thus, further experiments were performed with the use of IL-11
`at a dose of 8 ␮g/kg.
`We also confirmed the inhibitory effects of IL-11 on
`adverse cardiac remodeling 28 days after MI. IL-11 treatment
`
`Table.
`
`Effects of IL-11 on Cardiac Function at Day 14 After MI
`
`IL-11, ␮g/kg
`
`Parameters
`
`PBS
`
`3
`
`8
`
`20
`
`LVDP, mm Hg
`
`32.4⫾4.4
`
`34.9⫾6.3
`
`45.3⫾6.3*
`
`44.3⫾13.5*
`
`⫹dP/dt,
`mm Hg/s
`
`⫺dP/dt,
`mm Hg/s
`
`884⫾113
`
`958⫾162
`
`1210⫾148*
`
`1243⫾324*
`
`⫺713⫾93 ⫺737⫾176 ⫺1022⫾210* ⫺1043⫾326*
`
`Data are mean⫾SD (n⫽8 mice for PBS; n⫽9 mice for 3 ␮g/kg; n⫽9 mice
`for 8 ␮g/kg; n⫽8 mice for 20 ␮g/kg). LVDP indicates LV developed pressure.
`*P⬍0.05 vs PBS by 1-way ANOVA followed by Bonferroni test.
`
`prevented cardiac fibrosis and preserved cardiac function
`(Figure IV in the online-only Data Supplement).
`To examine whether IL-11 reduces infarct size, infarct size
`was measured by Evans blue exclusion 2 days after MI. There
`was no significant difference in infarct size (PBS, 23.0⫾7.1%;
`IL-11 [8 ␮g/kg], 22.0⫾4.3%; n⫽4 mice for each group).
`
`IL-11 Treatment Exhibits Antiapoptotic and
`Proangiogenic Activity in the Heart
`Granulocyte colony-stimulating factor (G-CSF) was reported
`to prevent cardiac remodeling after MI, accompanied by
`antiapoptotic and proangiogenic effects through STAT3.20
`Because IL-11 activated STAT3 in cardiomyocytes (Figure
`1), we examined the effects of IL-11 on cardiomyocyte
`survival and vascular formation in postinfarct myocardium.
`To evaluate the antiapoptotic effects of IL-11, TUNEL
`staining was performed (Figure 3A). TUNEL-positive cardio-
`myocytes were detected mainly at the border zone, adjacent
`to the ischemic area, at both day 2 and day 4 after MI.
`Importantly, IL-11 treatment significantly reduced the num-
`ber of apoptotic cardiomyocytes compared with the PBS
`group. To further confirm the cytoprotective effects,
`the
`cleavage of caspase 3 was examined by immunoblot analyses
`with cleaved caspase 3–specific antibody (Figure 3B). Com-
`pared with PBS treatment, IL-11 significantly reduced the
`band intensity for cleaved caspase 3. To address the antiapo-
`ptotic pathways downstream of the IL-11 signal, we exam-
`ined the expression of cell survival proteins (Figure 3C).
`Immunoblot analysis revealed that Bcl-2 and survivin pro-
`teins, both of which have been characterized as downstream
`targets of STAT3,21–23 were increased in the IL-11 group
`(Bcl-2, 1.9-fold; survivin, 1.5-fold).
`Because activation of STAT3 mediated vascular formation
`during cardiac remodeling,20,24 capillary density was immu-
`
`Downloaded from http://ahajournals.org by on October 29, 2019
`
`

`

`688
`
`Circulation
`
`February 9, 2010
`
`Figure 3. IL-11 treatment suppressed car-
`diomyocyte apoptosis after MI. A, The fre-
`quency of apoptotic cardiomyocytes was
`estimated by TUNEL staining 2 days and 4
`days after MI. The sections were costained
`with anti-sarcomeric ␣-actinin antibody and
`Hoechst 33258 dye. The images are repre-
`sentative of 60 to 100 obtained from 4 to 5
`mice (15 to 20 fields from each mouse) (left).
`Arrowheads show TUNEL-positive, apoptot-
`ic cardiomyocytes. Bar⫽50 ␮m. Quantifica-
`tion of the apoptotic cardiomyocytes is
`shown (right). Visual fields (15 to 20) were
`randomly selected. Data are shown as
`mean⫾SD (n⫽5 mice for PBS [2 days]; n⫽4
`mice for IL-11 [2 days]; n⫽5mice for PBS [4
`days]; n⫽5 mice for IL-11 [4 days]). *P⬍0.05
`vs PBS by unpaired t test. B, The lysates
`from hearts at day 2 after MI were immuno-
`blotted with anti-cleaved caspase 3 and
`anti-GAPDH antibodies. Arrows indicate the
`cleaved caspase 3 fragments with molecular
`weight 17 and 19 kDa (left). Quantitative
`analysis of cleaved caspase 3 fragments is
`shown (right). Data are shown as mean⫾SD
`(n⫽6 mice for PBS; n⫽5 mice for IL-11).
`*P⬍0.05 vs PBS by unpaired t test. C, Rep-
`resentative results of immunoblotting with
`anti-Bcl-2 and anti-survivin antibodies. Blots
`were reprobed with anti-GAPDH antibody to
`show equal amount loading. Experiments
`were repeated 5 times with similar results.
`
`nohistochemically estimated with anti-CD31 antibody (Fig-
`ure 4). In the border zone, capillary density was increased by
`IL-11 in a dose-dependent manner. Because the frequency of
`apoptosis was higher at day 2 than at day 4 (30.9⫾7.1 versus
`5.7⫾2.8 TUNEL-positive cardiomyocytes/mm2,
`respec-
`tively; P⬍0.01), cardiomyocyte loss occurs mainly at days 1
`to 3. On the basis of this finding, to clarify the relation
`between antiapoptosis and angiogenesis mediated by IL-
`11, we compared the effectiveness between cytokine treat-
`ment at days 1 to 3 and that at days 3 to 5 (Table II in the
`online-only Data Supplement). IL-11 treatment at days 1 to
`3 attenuated cardiac fibrosis, whereas that at days 3 to 5
`did not. Interestingly, treatment at days 1 to 3 increased
`capillary density, whereas that at days 3 to 5 did not.
`Therefore, IL-11–mediated increase in capillary density is
`likely to be closely associated with improved myocyte
`viability.
`
`Activation of STAT3 Is Necessary and Sufficient
`for IL-11–Mediated Prevention of
`Cardiac Remodeling
`To assess the importance of cardiac STAT3 activation in
`IL-11–mediated attenuation of adverse remodeling, we gen-
`erated cardiac-specific conditional STAT3-deficient mice
`(STAT3 CKO mice) by establishing ␣-MHC-MerCreMer
`mice on STAT3flox/flox background (Figure 5A). The ablation
`of the STAT3 gene did not
`induce notable histological
`alterations at day 14 after sham operation. IL-11 treatment
`ameliorated postinfarct fibrosis in ␣-MHC-MerCreMer mice
`on STAT3wild/wild background after MI (Figure V in the
`online-only Data Supplement), as in the case with nontrans-
`
`genic mice (Figure 2). Importantly, IL-11–mediated preven-
`tion against adverse cardiac remodeling was abrogated in
`STAT3 CKO mice (Figure 5C and 5D). Interestingly, en-
`largement of LVs was observed in STAT3 CKO mice
`compared with wild-type mice exposed to MI without IL-11
`treatment (18.02⫾3.20 mm [n⫽6] versus 15.08⫾1.81 mm
`[n⫽8]; P⬍0.05), probably because STAT3 is endogenously
`activated after MI, even without IL-11 treatment (Figures II
`and VI in the online-only Data Supplement), and contributes
`to the prevention of adverse cardiac remodeling. Consis-
`tently, we also confirmed that IL-11–mediated attenuation of
`cardiac dysfunction was canceled in STAT3 CKO mice
`(Figure VII in the online-only Data Supplement). Moreover,
`the increase in capillary density, which was observed in
`response to IL-11 in the mice with STAT3wild/wild back-
`ground, was abrogated by STAT3 gene ablation (Figure VIII
`in the online-only Data Supplement). These data indicate that
`STAT3 is required for IL-11–mediated amelioration of ad-
`verse cardiac remodeling after MI.
`To reinforce the importance of STAT3 in cardioprotection,
`transgenic hearts expressing constitutively active STAT3
`(caSTAT3) were exposed to MI, and cardiac fibrosis was
`estimated. In caSTAT3 transgenic hearts, cardiac fibrosis was
`reduced by 47% compared with nontransgenic littermates
`(wild-type) (Figure 6). Moreover, similar to IL-11–treated
`hearts, caSTAT3 transgenic hearts showed an increase in
`fibrotic wall thickness. Importantly, cardiac dysfunction was
`ameliorated in caSTAT3 transgenic mice compared with
`wild-type mice (Figure IX in the online-only Data Supple-
`ment). Therefore, activation of STAT3 in cardiomyocytes
`was sufficient to suppress cardiac remodeling after MI.
`
`Downloaded from http://ahajournals.org by on October 29, 2019
`
`

`

`Obana et al
`
`IL-11 as a Novel Cardioprotective Cytokine
`
`689
`
`Figure 5. STAT3 was required for IL-11–mediated amelioration
`of adverse remodeling after MI. A, Generation of STAT3 condi-
`tional knockout mice. B, After tamoxifen treatment, sham or MI
`operation was performed in ␣-MHC-MerCreMer/STAT3flox/flox
`(flox/flox; n⫽5 mice for sham; n⫽6 mice for MI) or ␣-MHC-Mer-
`CreMer/STAT3wild/wild (wild/wild; n⫽4 mice for sham; n⫽6 mice
`for MI) mice, followed by IL-11 administration. Heart sections (3
`sections from each mouse) were prepared 14 days after MI and
`stained with Masson’s trichrome. The images are representative
`of 12 to 18 obtained from 4 to 6 mice. Bar⫽1 mm. C, LV cir-
`cumference was quantitatively estimated. D, Ratio of fibrotic
`circumference to LV circumference was quantitatively estimated.
`Data are shown as mean⫾SD. *P⬍0.05, #P⬍0.01 vs STAT3wild/wild
`by unpaired t test.
`
`IL-11 belongs to the IL-6 cytokine family, which uses gp130
`as its common receptor. Interestingly, LIF, a member of the IL-6
`cytokine family, has been r

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket