throbber
LETTER
`
`doi:10.1038/nature24676
`
`IL-11 is a crucial determinant of cardiovascular
`fibrosis
`
`Sebastian Schafer1,2*, Sivakumar Viswanathan2*, Anissa A. Widjaja2*, Wei-Wen Lim1, Aida Moreno-Moral2,
`Daniel M. DeLaughter3, Benjamin Ng1, Giannino Patone4, Kingsley Chow1, Ester Khin2, Jessie Tan1, Sonia P. Chothani2,
`Lei Ye1, Owen J. L. Rackham2, Nicole S. J. Ko2, Norliza E. Sahib2, Chee Jian Pua1, Nicole T. G. Zhen1, Chen Xie1, Mao Wang2,
`Henrike Maatz4, Shiqi Lim1, Kathrin Saar4, Susanne Blachut4, Enrico Petretto2, Sabine Schmidt4, Tracy Putoczki5,6,
`Nuno Guimarães-Camboa7, Hiroko Wakimoto3, Sebastiaan van Heesch4, Kristmundur Sigmundsson2, See L. Lim1,
`Jia L. Soon1,2, Victor T. T. Chao1,2, Yeow L. Chua1, Teing E. Tan1, Sylvia M. Evans7,8,9, Yee J. Loh1,10, Muhammad H. Jamal1,
`Kim K. Ong1,10, Kim C. Chua1, Boon-Hean Ong1, Mathew J. Chakaramakkil1, Jonathan G. Seidman3, Christine E. Seidman3,11,12,
`Norbert Hubner4,13,14,15, Kenny Y. K. Sin1,2 & Stuart A. Cook1,2,16,17
`
`Fibrosis is a common pathology in cardiovascular disease1. In the
`heart, fibrosis causes mechanical and electrical dysfunction1,2 and
`in the kidney, it predicts the onset of renal failure3. Transforming
`growth factor β1 (TGFβ1) is the principal pro-fibrotic factor4,5, but
`its inhibition is associated with side effects due to its pleiotropic
`roles6,7. We hypothesized that downstream effectors of TGFβ1 in
`fibroblasts could be attractive therapeutic targets and lack upstream
`toxicity. Here we show, using integrated imaging–genomics analyses
`of primary human fibroblasts, that upregulation of interleukin-11
`(IL-11) is the dominant transcriptional response to TGFβ1 exposure
`and required for its pro-fibrotic effect. IL-11 and its receptor
`(IL11RA) are expressed specifically in fibroblasts, in which they
`drive non-canonical, ERK-dependent autocrine signalling that
`is required for fibrogenic protein synthesis. In mice, fibroblast-
`specific Il11 transgene expression or Il-11 injection causes heart
`and kidney fibrosis and organ failure, whereas genetic deletion
`of Il11ra1 protects against disease. Therefore, inhibition of IL-11
`prevents fibroblast activation across organs and species in response
`to a range of important pro-fibrotic stimuli. These results reveal a
`central role of IL-11 in fibrosis and we propose that inhibition of
`IL-11 is a potential therapeutic strategy to treat fibrotic diseases.
`Trans-differentiation of fibroblasts into activated myofibroblasts,
`which express α -smooth muscle actin (ACTA2) and secrete extracellular
`matrix (ECM) proteins, is a defining feature of fibrosis8. We automated
`the quantification of myofibroblasts and ECM production in primary
`human cardiac fibroblast cultures (n = 84; Extended Data Table 1
`and Supplementary Table 1), and performed RNA sequencing (RNA-
`seq) on paired unstimulated and TGFβ 1-stimulated samples (Extended
`Data Fig. 1). Genes were ranked on the basis of the magnitude and
`significance of their differential expression and their correlation with
`myofibroblasts. Typical fibrosis genes such as COMP and NOX49 were
`among the most-upregulated genes (Fig. 1a and Supplementary Table 2).
`Gene set enrichment analysis showed upregulation of genes important
`for protein secretion (Supplementary Table 3).
`Notably, IL11 expression showed the most positive correlation with
`myofibroblast numbers (r = 0.47, Padjusted = 6.44 × 10−6; Spearman’s
`correlation) (Fig. 1a) and upregulation of IL11 expression (8.5-fold,
`
`Padjusted = 6 × 10−218; DESeq210) defined the dominant transcriptional
`response of cardiac fibroblasts to TGFβ 1. IL11 expression is highly
`specific to fibroblasts, especially when stimulated, but is undetectable
`in most healthy human tissues and cells11,12 (Extended Data Fig. 2). To
`further explore the biological context of IL-11, we examined the expres-
`sion of its receptor (IL11RA) compared to the receptor of IL-6 (IL6R),
`a close family member, across 512 cell lines12. IL6R was present at high
`levels in immune cells, whereas IL11RA was most highly expressed in
`fibroblasts (Fig. 1b).
`To investigate Il11 expression in vivo, we performed single-cell
`RNA-seq of hearts from a PlnR9C/+ mouse, which has a cardiac fibrosis
`phenotype13, and a wild-type control mouse (Fig. 1c and Extended
`Data Fig. 3). Both Il11 (P = 5.6 × 10−8) and Il11ra1 (P = 2.2 × 10−16)
`were enriched in fibroblasts and Il11 was highly expressed in fibroblast
`subpopulations that had transcriptional features of TGFβ 1 activation
`or ECM production (Fig. 1d). Il11-expressing cells were most common
`in fibrotic PlnR9C/+ hearts and we confirmed Il-11 protein upregulation
`in this model (Extended Data Fig. 3).
`IL11 has been linked to haematopoiesis14 and tumorigenesis15,
`among other roles. In contrast to a previous cardiac study16, we found
`that recombinant human IL-11 (rhIL-11) is strongly pro-fibrotic in
`cardiac fibroblasts, increasing myofibroblasts and ECM production,
`motility, contraction and invasion (Fig. 1e and Extended Data Fig. 4).
`To better understand the apparent contradiction between our data
`and the previous work in which rhIL-11 was used in mouse models16,
`we tested whether rhIL-11 could activate mouse cardiac fibroblasts.
`rhIL-11 was mostly ineffective in mouse cardiac fibroblasts, whereas
`recombinant mouse Il-11 (rmIl-11) activated mouse cardiac and renal
`fibroblasts (Extended Data Fig. 4). rhIL-11 strongly activated human
`renal fibroblasts.
`Our findings implicate a pro-fibrotic role for IL-11 downstream
`of TGFβ 1. In the presence of neutralizing anti-IL-11 antibodies, the
`pro-fibrotic effects of TGFβ 1 were greatly diminished across a wide
`range of fibrosis assays (Fig. 1f–i and Extended Data Fig. 4). We made
`ligand traps by fusing IL11RA and gp130 (IL11RA:gp130) and these
`traps inhibited the pro-fibrotic effects of TGFβ 1 in a dose- dependent
`manner. The specificity of IL-11 inhibition was further confirmed
`
`1National Heart Centre Singapore, Singapore. 2Duke–National University of Singapore Medical School, Singapore. 3Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115,
`USA. 4Cardiovascular and Metabolic Sciences, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rossle Strasse 10, 13125 Berlin, Germany. 5Inflammation
`Division, Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia. 6Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3050, Australia.
`7Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, California, USA. 8Department of Medicine, University of California at San Diego, La Jolla,
`California 92093, USA. 9Department of Pharmacology, University of California at San Diego, La Jolla, California 92093, USA. 10Kandang Kerbau Women’s and Children’s Hospital, Singapore.
`11Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusettes 02115, USA. 12Howard Hughes Medical Institute, Chevy Chase, Maryland 20815, USA. 13DZHK
`(German Centre for Cardiovascular Research), partner site, Berlin, Germany. 14Charité-Universitätsmedizin, Berlin, Germany. 15Berlin Institute of Health (BIH), Berlin, Germany. 16National Heart
`and Lung Institute, Imperial College London, London, UK. 17MRC-London Institute of Medical Sciences, Hammersmith Hospital Campus, Du Cane Road, London, W12 0NN, UK.
`*These authors contributed equally to this work.
`
`1 1 0 | N A T U R E | V O L 5 5 2 | 7 D E C E M B E R 2 0 1 7
`
`© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
`
`Lassen - Exhibit 1066, p. 1
`
`

`

`Fibroblasts
`Osteoblasts
`Adipocytes
`Melanocytes
`Mesenchymal stem cells
`
`CD14+ monocytes
`CD14+CD16+ monocytes
`
`200
`IL6R expression (TPM)
`
`400
`
`TGF 1
`
`Proliferating
`
`b
`
`50
`
`25
`
`0
`0
`
`IL11RA expression (TPM)
`
`RASL11B
`
`XYLT1
`
`IL11
`
`COMP
`
`1
`2
`3
`ACTA2 correlation
`
`–0.5 0
`
`0.5
`
`4
`
`5
`
`6
`
`7
`
`8
`9
`10
`11
`Genomic position
`
`12
`
`14 15 16
`
`17
`
`19 20
`X
`–log2 (Padjusted)
`200
`400
`600
`
`ECM
`production
`
`20
`
`10
`
`0
`
`–10
`
`–20
`
`d
`
`tSNE2
`
`Endothelial
`cells
`
`Vessel
`endothelial
`
`Epicardial
`
`Smooth
`muscle
`
`Macrophages
`
`Proliferating
`(cid:31)broblasts
`
`Cardiomyocytes
`Red
`blood cells
`
`Fibroblasts
`
`10
`
`02468
`
`40
`
`20
`
`0
`
`–20
`
`a
`
`Fold change in RNA expression
`
`c
`
`tSNE2
`
`25
`
`Heart
`50
`
`–30
`
`IL-11-expressing cells
`–20
`–10
`
`Quiescent
`
`120
`
`110
`
`100
`
`90
`
`80
`
`g
`
`Gel contraction (mm2)
`
`i
`
`1.5
`
`1.0
`
`0.5
`
`scratch closure
`Fold change in
`
`TGF 1
`+
`IgG
`
`TGF 1
`+
`anti-IL-11
`
`f
`
`ACTA2
`
`Collagen I
`
`POSTN
`
`–40
`
`e
`
`Lymphocytes
`IL-11-expressing cells
`–25
`
`0
`tSNE1
`
`Control
`
`TGF 1
`
`IL-11
`
`ACTA2
`
`Collagen I
`
`POSTN
`
`Cardiac (cid:31)broblasts
`20
`
`10
`
`**
`
`0
`tSNE1
`h
`
`1.0
`
`0.5
`
`cell migration
`Fold change in
`
`*
`
`TGF 1
`+
`IgG
`
`TGF 1
`+
`anti-IL-11
`
`TGF 1
`+
`IgG
`
`TGF 1
`+
`anti-IL-11
`
`**
`
`200 m
`
`Figure 1 | Fibrosis target discovery platform identifies IL-11. a, RNA-
`seq of primary cardiac fibroblasts (n = 84 biologically independent
`samples) with or without TGFβ 1 treatment (5 ng ml−1, 24 h) and
`Spearman’s correlation of expression changes with fibroblast activation
`(Supplementary Table 2). DEseq210 fold change in expression and false-
`discovery rate (FDR)-adjusted P values are shown. b, RNA expression in
`transcripts per million (TPM) of IL11RA and IL6R across 512 cell lines
`from the FANTOM repository12. c, Single-cell resolution of cardiac
`Il11 expression (more than 0 reads per cell). t-distributed stochastic
`neighbour embedding (tSNE) analysis28 clusters cell types of the heart.
`Il11 expression is highly enriched in fibroblasts. χ2 test (P = 5.7 × 10−8).
`d, tSNE analysis of fibroblasts alone shows highest Il-11 expression in
`ECM-secreting and TGFβ 1-activated fibroblasts. χ2 test (P = 0.033).
`
`200 m
`
`TGF 1
`TGF 1
`+
`+
`IgG
`anti-IL-11
`c, d, Cardiac cells were sequenced from n = 1 mouse, the experiment was
`repeated once with similar results. e, f, Representative images (chosen
`from 42 per condition) of cardiac fibroblasts immunostained for ACTA2,
`collagen I or periostin (POSTN) after a 24-h incubation without stimulus
`(control), TGFβ 1 or IL-11 (5 ng ml−1) (e) or with TGFβ 1 (5 ng ml−1) and
`an anti-IL-11 neutralizing antibody or an IgG control (2 μ g ml−1) (f).
`g, Cardiac fibroblasts were seeded in collagen gel and the area of
`contraction determined (n = 3 biologically independent samples) after
`72 h. h, Trans-well migration assay (colourimetrically quantified, n = 3
`biologically independent samples, 24 h). i, Scratch assay of wound closure
`in a monolayer of cardiac fibroblasts (n = 5 biologically independent
`samples) after 24 h. g–i, Two-tailed Student’s t-test; data are mean ± s.d.;
`* P < 0.05; * * P < 0.01.
`
`through inhibition of IL11RA using receptor-blocking antibodies or
`by short interfering RNA (siRNA), all of which attenuated the effects
`of TGFβ 1. By contrast, anti-IL-6 antibodies had no effect on TGFβ 1-
`induced cardiac fibroblast activation (Extended Data Fig. 4).
`We next investigated the consequences of IL-11 signalling in cardiac
`fibroblasts using RNA-seq. Surprisingly, the effect of IL-11 on the
` transcriptome was negligible, whereas TGFβ 1-driven transcriptional
`regulation in cardiac fibroblasts from the same patients remained
` profound (Fig. 2a, b). We repeated the experiment using cardiac fibro-
`blasts from multiple patients and consistently observed very little
`effect of IL-11 on mRNA levels but, in the cell culture supernatants of
`identical samples, we reproducibly documented pro-fibrotic protein
` secretion. Therefore, the effects of IL-11 on cardiac fibroblasts are
`mainly at the protein level and are unrelated to transcriptional changes
`(Extended Data Fig. 5). This differs from effects of IL-11 on cancer cells,
`in which JAK–STAT signalling is involved17.
`IL-11 can bind to free IL11RA, which is shed from cardiac fibro-
`blast membranes (Extended Data Fig. 5) and signal in trans in cells
`
`that express GP130 (also known as IL6STP1)18. We generated an
`IL11RA:IL-11 fusion protein (hyperIL-11), which mimics the trans-
`signalling complex19. Concentrations of hyperIL-11 as low as
`0.2 ng ml−1 activated cardiac fibroblasts (Extended Data Fig. 5),
` supporting a role for IL-11 trans-signalling in fibrosis.
`We found that both IL11 and IL11RA are expressed in fibro-
`blasts, which implies that an autocrine signalling loop exists. Using
`hyperIL-11, which is not detected in an IL-11 enzyme-linked immuno-
`sorbent assay (ELISA), we confirmed the existence of autocrine IL-11
`signalling in cardiac fibroblasts. This autocrine, feed-forward loop of
`hyperIL-11-induced IL-11 secretion is dependent on de novo IL-11
`protein synthesis and secretion (Fig. 2c and Extended Data Fig. 5), but
`is independent of IL11 RNA levels. In separate experiments, rhIL-11
`(not detected by IL-11 ELISA) cis-signalling also strongly induced
`endogenous IL-11 secretion, and this also occurred in the absence of
`changes in IL11 mRNA levels (Extended Data Fig. 5).
`TGFβ 1 activation of non-canonical ERK signalling in fibroblasts
`is important for fibrosis4,5 and we observed that IL-11 also activated
`
`7 D E C E M B E R 2 0 1 7 | V O L 5 5 2 | N A T U R E | 1 1 1
`
`Letter reSeArCH
`
`© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
`
`Lassen - Exhibit 1066, p. 2
`
`

`

`IL11 stimulation
`COL1A1 RNA fc: 1.03
`COL1A2 RNA fc: 1.02
`COL3A1 RNA fc: 1.12
`
`2
`
`1 0
`
`–1
`
`–2
`
` log2(fold change in RNA)
`
`TGF 1 stimulation
`
`b
`
`2
`
`1 0
`
`–1
`
`–2
`
` log2(fold change in RNA)
`
`a
`
`POSTN RNA fc: 1.01
`ACTA2 RNA fc: 1.00
`MMP2 RNA fc: 1.01
`103
`105
`1
`10
`Normalized mean counts
`
`Unstim ulated
`
`
`
`
`
`AngII
`
`CTGFED N1bFGFIL-11IL-13
`
`O S M
`
`
`
`PD GFTGF 1
`
`p-ERK1/2
`p-P38
`p-STAT3
`p-SMAD2
`
`p-AKT
`
`p-mTOR
`
`GAPDH
`
`Stimulant
`
`Stimulant
`+
`anti-IL-11
`
`Stimulant
`+
`IgG
`
`g T
`
`GF 1
`
`1
`
`105
`103
`10
`Normalized mean counts
`
`– + –+
`– – + +
`
`BFA
`Hyper-IL-11
`
`Intracellular IL-11
`GAPDH
`
`25 kDa
`
`35 kDa
`
`d
`ACTA2+
`
`Collagen I
`
`MEK inhibitors
`
`e
`
`40 kDa
`40 kDa
`100 kDa
`
`70 kDa
`70 kDa
`
`260 kDa
`
`–
`+
`+
`–
`
`–
`+
`–
`+
`
`35 kDa
`
`1.5×
`
`Protein (P = 4 × 10-4)
`RNA (P = 0.90)
`
`0
`
`28
`1
`2
`24
`48
`After hyper-IL-11 treatment (h)
`
`72
`
`POSTN
`TGF 1
`+
`+
`–
`+
`–
`–
`+
`–
`IL-11
`–
`+
`–
`–
`U0126
`+
`–
`–
`–
`PD98059
`Fluorescent signal:
`1×
`
`IL-11 protein
`IL11 RNA
`
`****
`
`****
`
`****
`
`30
`
`20
`
`****
`
`****
`
`c
`
`80
`
`60
`
`40
`
`20
`
`Fold change in IL-11
`
`f
`
`PDGF
`
`AngII
`
`EDN1
`
`CTGF
`
`IL-13
`
`OSM
`
`IL11 RNA expressiont
`
`Fold change in
`
`10
`
`TGF 1
`****
`
`****
`
`****
`
`POSTN
`
`Collagen I
`
`ACTA2+
`
`POSTN
`
`Collagen I
`
`ACTA2+
`
`POSTN
`
`Collagen I
`
`ACTA2+
`
`NS
`
`bFGF
`
`NS
`
`NS
`
`Unstimulated
`
`Fluorescent signal:
`1×
`1.5×
`Il11ra1+/+
`Il11ra1–/–
`fibroblasts in response to various pro-fibrotic stimuli (see also Extended
`Data Fig. 5). f, ELISA (supernatant, n = 3 biologically independent
`samples) and RT–qPCR (n = 2 biologically independent samples) of
`IL-11 expression in cardiac fibroblasts after 24 h stimulation with AngII
`(100 nM), CTGF (50 ng ml−1), EDN1 (250 ng ml−1), bFGF (10 ng ml−1),
`IL-13 (100 ng ml−1), OSM (100 ng ml−1), PDGF (200 ng ml−1) and TGFβ 1
`(5 ng ml−1). Two-tailed Dunnett’s test; Data are mean ± s.d. g, Cardiac
`fibroblasts were incubated with pro-fibrotic cytokines (24 h) and cardiac
`fibroblast activation was reduced by anti-IL-11 antibodies (2 μ g ml−1;
`Extended Data Fig. 6). h, Pro-fibrotic proteins (Operetta assay n = 7
`measurements per n = 2 independent experiments) are not upregulated
`in cardiac fibroblasts from Il11ra1−/− mice in response to TGFβ 1, IL-11
`(5 ng ml−1) or AngII (100 nM, 24 h). Experiments were repeated twice
`with similar results. Two-tailed Dunnett’s test; box-and-whisker plots
`show median (middle line), 25th–75th percentiles (box) and 10th–90th
`percentiles (whiskers); * * * * P < 0.0001; NS, not significant.
`
`****
`
`****
`
`****
`
`2468
`
`change in expression
`
`IL-11 protein fold
`
`AngII
`
`CTGF
`
`EDN1
`
`IL-13
`
`OSM PDGF
`
`bFGF
`****
`
`3
`
`2
`
`1
`
`Fold change in collagen I
`
`protein expression
`
`NS
`
`NS
`
`NS
`
`****
`
`****
`
`****
`
`Unstimulated
`
`2
`
`1
`
`h
`
`ACTA2+ cells
`
`1234
`
`Fold change in POSTN
`
`protein expression
`
`****
`
`****
`
`NS
`
`NS
`
`NS
`
`Unstimulated
`
`TGF 1
`IL-11
`AngII
`Il11ra1+/+
`Il11ra1–/–
`Il11ra1+/+
`Il11ra1–/–
`Figure 2 | Non-canonical IL-11 signalling drives fibrogenic protein
`synthesis. a, b, RNA-seq of primary cardiac fibroblasts in response
`to TGFβ 1 (a) or IL-11 (b) (n = 6 biologically independent samples,
`5 ng ml−1, 24 h). Red indicates significantly differentially expressed
`genes (FDR ≤ 0.05, DEseq210). RNA expression of genes associated with
`fibrosis is not increased by IL-11 treatment. fc, fold change. c, ELISA and
`quantitative PCR with reverse transcription (RT–qPCR) assays of IL-11
`expression (n = 3 biologically independent samples) after hyperIL-11
`treatment (0.2 ng ml−1). Benjamini–Hochberg corrected one-way
`ANOVA; data are mean ± s.d. Inset, western blot of cardiac fibroblast
`lysates after hyperIL-11 stimulation and brefeldin A (BFA, 1 μ g ml−1)
`treatment indicates canonical secretion of IL-11. d, Cardiac fibroblasts
`were incubated with TGFβ 1, IL-11 (5 ng ml−1) and MEK inhibitors
`U0126 or PD98059 (10 μ M, 24 h). ACTA2+ cells and ECM production
`was assessed and normalized to non-stimulated cells. e, Western blots of
`phosphorylated protein (p-) expression of signalling pathways in cardiac
`
`ERK in cardiac fibroblasts and that both TGFβ 1 and IL-11 required
`ERK to induce pro-fibrotic phenotypes (Fig. 2d and Extended Data
`Fig. 5). Because the effects of IL-11 on pro-fibrotic gene expression are
`post-transcriptional, we suggest that this phenomenon may be driven,
`in part, by activation of ERK and its downstream substrates (Extended
`Data Fig. 5).
`We therefore investigated the activation of STAT, SMAD, ERK and
`kinases that are important for protein synthesis in cardiac fibroblasts
`in response to stimulation by a range of pro-fibrotic factors. These
`factors included established stimuli for cardiac fibrosis (endothelin-1
`(END1), angiotensin II (AngII) and PDGF) and other key pro-fibrotic
`cytokines (OSM, bFGF, CTGF and IL13)1,5. IL-11 activation of STAT in
`cardiac fibroblasts was negligible, consistent with its lack of transcrip-
`tional effects in this cell type. As expected, TGFβ 1 activated SMAD,
`but the only pathway that was consistently activated by all stimuli was
`the ERK pathway (Fig. 2e). All pro-fibrotic stimuli that were tested
`induced changes in IL-11 protein levels but not mRNA; only TGFβ 1
`increased SMAD-dependent IL11 transcription (Fig. 2f and Extended
`Data Fig. 5). Remarkably, as seen with TGFβ 1, fibroblast activation in
`
`response to all pro-fibrotic stimuli that were tested was dependent on
`IL-11 signalling (Fig. 2g and Extended Data Fig. 6).
`We studied the fibrotic response of cardiac fibroblasts from
`Il11ra1−/− mice20. TGFβ 1-induced transcriptional regulation in
`Il11ra1−/− cardiac fibroblasts was maintained and similar to that of
`Il11ra1+/+ (wild-type) cardiac fibroblasts (R2 = 0.94, P < 2.2 × 10−16,
`Spearman’s correlation; Extended Data Fig. 7). However, protein-based
`assays showed that cardiac fibroblasts from Il11ra1−/− mice did not
`increase synthesis of ECM proteins or become myofibroblasts upon
`stimulation (Fig. 2h and Extended Data Fig. 7), again demonstrating
`the effect of IL-11 at the protein level.
`We investigated the role of Il-11 in a mouse model of myocardial
`infarction, but rather than injecting rhIL-11, as had been done in the
`previous study16, we administered rmIl-11. We measured epicardial
`activation, a defining feature of active fibrosis in myocardial infarction21
`and found that rmIl-11 robustly stimulated fibroblasts in the epicardium
`(Fig. 3a–c) and caused ventricular impairment (Fig. 3d, e). We then
`tested the effects of rmIl-11 injection in healthy mice using regimens
`similar to those used for rhIL-11 in patients with cancer22. rmIL-11
`
`1 1 2 | N A T U R E | V O L 5 5 2 | 7 D E C E M B E R 2 0 1 7
`
`reSeArCH
`
`Letter
`
`© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
`
`Lassen - Exhibit 1066, p. 3
`
`

`

`***
`
`*
`
`**
`
`Sham
`
`MI
`+
`PBS
`
`MI
`+
`rmIl-11
`
`****
`***
`
`**
`
`80
`
`60
`
`40
`
`20
`
`60
`
`40
`
`20
`
`d
`
`Ejection fraction (%)
`
`e
`
`ESV (µl)
`
`Distal
`
`*
`
`**
`
`246
`
`b
`
`epicardial thickness
`
`Fold change in
`
`Sham
`
`MI
`+
`PBS
`
`MI
`+
`rmIl-11
`
`Proximal
`
`c
`
`*
`
`**
`
`epicardial thickness
`
`Fold change in
`
`10
`
`5
`
`a
`
`Epicardium
`
`Sham
`
`MI + PBS
`
`MI + rmIl-11
`
`Masson’s trichrome
`
`ACTA2
`
`50 m
`
`Col1a1–GFP epicardium
`
`Col1a1–GFP renal cortex
`
`f
`
`Control
`
`Sham
`
`MI
`+
`PBS
`
`MI
`+
`rmIl-11
`
`Sham
`
`MI
`+
`PBS
`
`MI
`+
`rmIl-11
`
`g
`Control
`
`Fold change in renal function markers
`Urea
`Creatinine
`
`rmIl-11
`injection
`
`****
`**
`
`2.0
`
`0.5
`
`1.0
`
`1.5
`
`**
`
`***
`
`Ventricle
`
`****
`
`2
`
`1
`
`123
`
`i
`
`HPA collagen ( g ml–1)
`
`k
`
`HPA collagen ( g ml–1)
`
`*
`
`Control rmIl-11
`Injection
`
`80
`
`70
`
`60
`
`50
`
`h
`
`Ejection fraction (%)
`
`DAPI GFP
`
`GFP
`
`DAPI GFP
`
`GFP
`
`50 µm
`
`50 µm
`
`Epicardium
`
`Renal cortex
`
`Control
`rmIl-11
`injection
`Kidney
`
`****
`
`Control
`Il-11-Tg
`Kidney
`
`1234
`
`6
`
`4
`
`2
`
`rmIl-11
`injection
`
`j
`
`Control
`
`Il-11-Tg
`
`Ventricle
`
`*
`
`80
`
`70
`
`60
`
`50
`
`l
`
`Ejection fraction (%)
`
`Masson’s trichrome
`
`ACTA2
`
`Masson’s trichrome
`
`ACTA2
`
`50 µm
`
`50 µm
`
`Figure 3 | Il-11 causes cardiovascular fibrosis and organ failure.
`a, Representative histological images of the epicardium with Masson’s
`trichrome staining and ACTA2 immunostaining in mice with
`myocardial infarction (MI) treated with rmIL-11 or PBS. b, c, Epicardial
`thickness of regions distal (b) or proximal (c) to the region of myocardial
`infarction. a–c, Sham, n = 5; myocardial infarction, n = 6; myocardial
`infarction + rmIl-11, n = 8 biologically independent samples.
`d, e, Echocardiography show a decrease in ejection fraction (d) and
`increase in end-systolic volume (ESV) (e) after myocardial infarction
`and rmIl-11 treatment (sham, n = 3; myocardial infarction, myocardial
`infarction + rmIl-11, n = 5 biologically independent samples). b–e, two-
`tailed, Holm–Sidak-corrected Student’s t-test; Data are mean ± s.d.
`f, Representative histological images (chosen from control, n = 3;
`rmIl-11 injection, n = 4 biologically independent samples) of tissues
`from a Col1a1–GFP-reporter mouse after rmIL-11 injection (100 μ g kg−1
`per day, three weeks). g, Serum urea and creatinine levels after rmIL-11
`injection in Col1a1–GFP reporter mice23 resulted in specific activa-
`tion of fibroblasts in the epicardium and renal interstitium (Fig. 3f).
`rmIl-11-treated mice had high circulating levels of TGFβ 1, but not
`of other pro-inflammatory factors and features of cardiac and renal
`impairment along with cardiovascular fibrosis (Fig. 3g–i and Extended
`Data Fig. 7).
`To investigate the effects of autocrine Il-11 signalling in fibroblasts
`in vivo, we generated rmIl-11-transgenic mice, which we crossed
`with inducible Col1a2–Cre mice (Il-11-Tg; see Methods). Il-11 was
`
`Control
`Il-11-Tg
`injection (control urea, n = 8; control creatinine, n = 7; rmIl-11, n = 12
`biologically independent samples). h, Reduced ejection fraction
`(echocardiography) in rmIl-11-treated mice. i, Hydroxyproline assay
`(HPA) quantifies cardiac and renal collagen content after rmIL-11
`treatment. h, i, Control, n = 8; rmIl-11, n = 11 biologically independent
`samples. j, Representative histological images of Masson’s trichrome
`staining and ACTA2 immunostaining in the epicardium Il-11-Tg mice.
`k, HPA indicates cardiac and renal collagen content in Il-11-Tg mice.
`j, k, Control, n = 12; Il-11-Tg, n = 4 biologically independent samples.
`i, k, Two-tailed Student’s t-test; box-and-whisker plots show median
`(middle line), 25th–75th percentiles (box) and 10th–90th percentiles
`(whiskers). l, Reduction in ejection fraction (echocardiography) in
`Il-11-Tg mice (control, n = 6; Il-11-Tg, n = 4 biologically independent
`samples). g, h, l, Two-tailed Student’s t-test; data are mean ± s.d.;
`* P < 0.05; * * P < 0.01; * * * P < 0.001; * * * * P < 0.0001.
`
`expressed after induction with tamoxifen and within two weeks, there
`was widespread activation of cardiac and renal fibroblasts and accu-
`mulation of collagen (Fig. 3j, k). This was accompanied by a reduction
`in cardiac function (Fig. 3l), increased serum TGFβ 1 and an increase
`in serum urea and creatinine, which are biomarkers of renal failure
`(Extended Data Fig. 7).
`Il-11 expression was found to be increased in three preclinical models
`of cardiovascular fibrosis (Fig. 4a, e, i). Therefore, using knockout and
`wild-type mice we determined whether inhibition of Il-11 could reduce
`
`7 D E C E M B E R 2 0 1 7 | V O L 5 5 2 | N A T U R E | 1 1 3
`
`Letter reSeArCH
`
`© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
`
`Lassen - Exhibit 1066, p. 4
`
`

`

`Heart
`
`Control
`
`AngII
`
`Il-11
`
`GAPDH
`
`P = 1 × 10–4
`
`P = 0.0262
`
`NS
`
`b
`
`Il11ra1+/+
`
`Il11ra1–/–
`
`Control
`
`AngII
`
`Control
`
`AngII
`
`100µm
`
`Il11ra1+/+
`AngII
`Control
`
`Il11ra1–/–
`AngII
`Control
`
`p-ERK1/2
`
`ERK1/2
`
`Il11ra1+/+
`
`Il11ra1–/–
`
`d
`
`40 kDa
`
`40 kDa
`
`Control
`AngII
`
`f
`
`Il-11
`
`GAPDH
`
`Il11ra1+/+
`
`Il11ra1–/–
`
`Heart
`
`Sham
`
`TAC
`
`P < 1 × 10–4
`P = 5 × 10–3
`
`Sham
`
`TAC
`
`Sham
`
`TAC
`
`a
`
`25 kDa
`
`35 kDa
`
`1.5
`
`1.0
`
`0.5
`
`c
`
`Fold change in HPA collagen
`
`e
`
`25 kDa
`
`35 kDa
`g
`
`4
`
`P = 2 × 10–3
`
`Il11ra1+/+
`
`Il11ra1–/–
`
`Kidney
`Control
`
`Folate
`
`P < 1 × 10–4
`
`P = 0.0022
`
`NS
`
`Sham
`TAC
`
`j
`
`Il-11
`
`GAPDH
`
`Control
`Folate
`
`Fold change in HPA collagen
`
`2
`
`i
`
`25 kDa
`
`35 kDa
`
`3
`
`2
`
`1
`
`k
`
`Fold change in HPA collagen
`
`Il11ra1+/+
`Il11ra1–/–
`Figure 4 | Inhibition of Il-11 reduces cardiovascular fibrosis.
`a, Western blot of cardiac Il-11 after AngIl infusion (2 mg kg−1 per day for
`28 days). b, c, Representative histological images (b; Masson’s trichrome
`staining) and collagen content (c) in the atrium of Il11ra1+/+ (control,
`n = 12; AngII, n = 9 biologically independent samples) and Il11ra1−/−
`(control, n = 5; AngII, n = 7 biologically independent samples) mice.
`d, Western blot of cardiac ERK activation after AngII infusion. e, Western
`blot of cardiac Il-11 after transverse aortic constriction (TAC).
`f, g, Representative histological images (f; Masson’s trichrome staining) and
`collagen content. g; HPA of hearts from Il11ra1+/+ (control, n = 4; TAC:
`
`fibrosis in these models. After either AngII infusion or transverse aortic
`constriction, less fibrosis occurred in the hearts of knockout mice com-
`pared to wild-type mice (Fig. 4b, c, f, g). This effect was independent
`of loading conditions (Extended Data Fig. 8). Similarly, after folate-
`induced kidney damage, knockout mice had reduced renal fibrosis
`(Fig. 4j, k). Deletion of Il11ra1 signalling resulted in reduced ERK
`signalling across all models tested, whereas p38 signalling was unaf-
`fected (Fig. 4d, h, l and Extended Data Fig. 9).
`IL-11 was discovered owing to its ability to sustain an IL-6-dependent
`haematopoetic cell line when secreted from fibroblastic cells24, but was
`later found to be redundant for haematopoiesis20. Here we show that
`IL11 is a crucial fibrosis gene acting downstream of TGFβ 1 and many
`other pro-fibrotic factors. We believe that the importance of IL-11 in
`fibroblasts may have gone unnoticed because its effects are apparent only
`at the post-transcriptional level (Extended Data Fig. 9). We highlight
`that IL11RA is not only expressed in fibroblasts but also in other cells
`and that IL-11 signalling may therefore be important in other cell types.
`
`1 1 4 | N A T U R E | V O L 5 5 2 | 7 D E C E M B E R 2 0 1 7
`
`n = 6 biologically independent samples) and Il11ra1−/− (control, n = 6;
`TAC, n = 6 biologically independent samples) mice after TAC or sham
`operations. h, Western blot of cardiac ERK activation after TAC. i, Western
`blot of renal Il-11 after folate treatment (180 mg kg−1). j, k, Representative
`histological images (j; Masson’s trichrome staining) and collagen content
`(k; HPA assay) of kidneys from Il11ra1+/+ (control, n = 5; folate, n = 9
`biologically independent samples) and Il11ra1−/− (control, n = 6;
`folate, n = 5 biologically independent samples) mice. c, g, k, Two-tailed,
`Sidak-corrected Student’s t-test; data are mean ± s.d. NS, not significant.
`l, Western blot of renal ERK activation after folate treatment.
`
`We note that rhIL-11 has been given to patients with myocardial
` infarction25 and it is possible that the use of rhIL-11 in cancer patients22
`causes fibrosis-related side effects. We therefore suggest that the use of
`rhIL-11 in humans should be reviewed. IL11 is highly upregulated in
`fibroblasts from patients with idiopathic pulmonary fibrosis or systemic
`sclerosis, by a 100-fold and 30-fold, respectively26; this suggests a role
`for IL-11 in fibrotic human disease beyond the cardiovascular system.
`IL-11 inhibitors may be particularly effective in treating fibrosis, because
`they would target a nodal point of pro-fibrotic signalling. Because of these
`results, and target safety data from human27 and mouse knockouts20, we
`propose that IL-11 is a potential therapeutic target.
`
`Online Content Methods, along with any additional Extended Data display items and
`Source Data, are available in the online version of the paper; references unique to
`these sections appear only in the online paper.
`
`received 22 March; accepted 2 November 2017.
`Published online 13 November 2017.
`
`h
`
`40 kDa
`
`40 kDa
`
`Il11ra1+/+
`TAC
`Sham
`
`Il11ra1–/–
`TAC
`Sham
`
`p-ERK1/2
`
`ERK1/2
`
`Il11ra1+/+
`
`Il11ra1–/–
`
`Control
`
`Folate
`
`Control
`
`Folate
`
`1mm
`
`Il11ra1+/+
`Folate
`Control
`
`Il11ra1–/–
`Folate
`Control
`
`l
`
`40 kDa
`
`40 kDa
`
`p-ERK1/2
`
`ERK1/2
`
`reSeArCH
`
`Letter
`
`© 2017 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
`
`Lassen - Exhibit 1066, p. 5
`
`

`

`1. Rockey, D. C., Bell, P. D. & Hill, J. A. Fibrosis—a common pathway to organ
`injury and failure. N. Engl. J. Med. 372, 1138–1149 (2015).
`2. Burstein, B. & Nattel, S. Atrial fibrosis: mechanisms and clinical relevance in
`atrial fibrillation. J. Am. Coll. Cardiol. 51, 802–809 (2008).
`3. Leaf, I. A. & Duffield, J. S. What can target kidney fibrosis? Nephrol. Dial.
`Transplant. 32 (suppl. 1), i89–i97 (2017).
`4. Davis, J. & Molkentin, J. D. Myofibroblasts: trust your heart and let fate decide.
`J. Mol. Cell. Cardiol. 70, 9–18 (2014).
`5. Akhurst, R. J. & Hata, A. Targeting the TGFβ signalling pathway in disease.
`Nat. Rev. Drug Discov. 11, 790–811 (2012).
`6. Bierie, B. et al. Abrogation of TGF-β signaling enhances chemokine production
`and correlates with prognosis in human breast cancer. J. Clin. Invest. 119,
`1571–1582 (2009).
`7. Shull, M. M. et al. Targeted disruption of the mouse transforming growth
`factor-β 1 gene results in multifocal inflammatory disease. Nature 359,
`693–699 (1992).
`8. Wynn, T. A. Cellular and molecular mechanisms of fibrosis. J. Pathol. 214,
`199–210 (2008).
`9. Cucoranu, I. et al. NAD(P)H oxidase 4 mediates transforming growth factor-β 1-
`induced differentiation of cardiac fibroblasts into myofibroblasts. Circ. Res. 97,
`900–907 (2005).
`10. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and
`dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
`11. GTEx Consortium. The Genotype-Tissue Expression (GTEx) project. Nat. Genet.
`45, 580–585 (2013).
`12. FANTOM Consortium and the RIKEN PMI and CLST (DGT). A promoter-level
`mammalian expression atlas. Nature 507, 462–470 (2014).
`13. Schmitt, J. P. et al. Dilated cardiomyopathy and heart failure caused by a
`mutation in phospholamban. Science 299, 1410–1413 (2003).
`14. Du, X. X., Neben, T., Goldman, S. & Williams, D. A. Effects of recombinant
`human interleukin-11 on hematopoietic reconstitution in transplant mice:
`acceleration of recovery of peripheral blood neutrophils and platelets. Blood
`81, 27–34 (1993).
`15. Putoczki, T. L. et al. Interleukin-11 is the dominant IL-6 family cytokine during
`gastrointestinal tumorigenesis and can be targeted therapeutically. Cancer Cell
`24, 257–271 (2013).
`16. Obana, M. et al. Therapeutic activation of signal transducer and activator of
`transcription 3 by interleukin-11 ameliorates cardiac fibrosis after myocardial
`infarction. Circulation 121, 684–691 (2010).
`17. Ernst, M. & Putoczki, T. L. Molecular pathways: IL11 as a tumor-promoting
`cytokine—translational implications for cancers. Clin. Cancer Res. 20,
`5579–5588 (2014).
`18. Lokau, J. et al. Proteolytic cleavage governs interleukin-11 trans-signaling.
`Cell Reports 14, 1761–1773 (2016).
`19. Dams-Kozlowska, H. et al. A designer hyper interleukin 11 (H11) is a
`biologically active cytokine. BMC Biotechnol. 12, 8 (2012).
`20. Nandurkar, H. H. et al. Adult mice with targeted mutation of the interleukin-11
`receptor (IL11Ra) display normal hematopoiesis. Blood 90, 2148–2159
`(1997).
`21. Duan, J. et al. Wnt1/β catenin injury response a

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