ArticlePDF Available

A neutralizing IL-11 antibody reduces vessel hyperplasia in a mouse carotid artery wire injury model

Authors:

Abstract and Figures

Vascular restenosis remains a major problem in patients with coronary artery disease (CAD) and peripheral artery disease (PAD). Neointimal hyperplasia, defined by post-procedure proliferation and migration of vascular smooth muscle cells (VSMCs) is a key underlying pathology. Here we investigated the role of Interleukin 11 (IL-11) in a mouse model of injury-related plaque development. Apoe−/− mice were fed a hyperlipidaemic diet and subjected to carotid wire injury of the right carotid. Mice were injected with an anti-IL11 antibody (X203), IgG control antibody or buffer. We performed ultrasound analysis to assess vessel wall thickness and blood velocity. Using histology and immunofluorescence approaches, we determined the effects of IL-11 inhibition on VSMC and macrophages phenotypes and fibrosis. Treatment of mice with carotid wire injury using X203 significantly reduced post-endothelial injury vessel wall thickness, and injury-related plaque, when compared to control. Immunofluorescence staining of the injury-related plaque showed that X203 treatment did not reduce macrophage numbers, but reduced the number of VSMCs and lowered matrix metalloproteinase 2 (MMP2) levels and collagen content in comparison to control. X203 treatment was associated with a significant increase in smooth muscle protein 22α (SM22α) positive cells in injury-related plaque compared to control, suggesting preservation of the contractile VSMC phenotype. Interestingly, X203 also reduced the collagen content of uninjured carotid arteries as compared to IgG, showing an additional effect on hyperlipidemia-induced arterial remodeling in the absence of mechanical injury. Therapeutic inhibition of IL-11 reduced vessel wall thickness, attenuated neointimal hyperplasia, and has favorable effects on vascular remodeling following wire-induced endothelial injury. This suggests IL-11 inhibition as a potential novel therapeutic approach to reduce arterial stenosis following revascularization in CAD and PAD patients.
This content is subject to copyright. Terms and conditions apply.
1
Vol.:(0123456789)
Scientic Reports | (2021) 11:20674 | https://doi.org/10.1038/s41598-021-99880-y
www.nature.com/scientificreports
A neutralizing IL‑11 antibody
reduces vessel hyperplasia
in a mouse carotid artery wire
injury model
David Schumacher1,2,15, Elisa A. Liehn3,4,5,6,15, Pakhwan Nilcham2, David Castaño Mayan7,8,9,
Chutima Rattanasopa7, Kaviya Anand7, Gustavo E. Crespo‑Avilan6,10,11,
Sauri Hernandez‑Resendiz6,10, Roshni R. Singaraja7,8,9, Stuart A. Cook6,10,12,16 &
Derek J. Hausenloy6,8,10,13,14,16*
Vascular restenosis remains a major problem in patients with coronary artery disease (CAD) and
peripheral artery disease (PAD). Neointimal hyperplasia, dened by post‑procedure proliferation
and migration of vascular smooth muscle cells (VSMCs) is a key underlying pathology. Here we
investigated the role of Interleukin 11 (IL‑11) in a mouse model of injury‑related plaque development.
Apoe−/− mice were fed a hyperlipidaemic diet and subjected to carotid wire injury of the right
carotid. Mice were injected with an anti‑IL11 antibody (X203), IgG control antibody or buer. We
performed ultrasound analysis to assess vessel wall thickness and blood velocity. Using histology
and immunouorescence approaches, we determined the eects of IL‑11 inhibition on VSMC
and macrophages phenotypes and brosis. Treatment of mice with carotid wire injury using X203
signicantly reduced post‑endothelial injury vessel wall thickness, and injury‑related plaque, when
compared to control. Immunouorescence staining of the injury‑related plaque showed that X203
treatment did not reduce macrophage numbers, but reduced the number of VSMCs and lowered
matrix metalloproteinase 2 (MMP2) levels and collagen content in comparison to control. X203
treatment was associated with a signicant increase in smooth muscle protein 22α (SM22α) positive
cells in injury‑related plaque compared to control, suggesting preservation of the contractile VSMC
phenotype. Interestingly, X203 also reduced the collagen content of uninjured carotid arteries as
compared to IgG, showing an additional eect on hyperlipidemia‑induced arterial remodeling in the
absence of mechanical injury. Therapeutic inhibition of IL‑11 reduced vessel wall thickness, attenuated
neointimal hyperplasia, and has favorable eects on vascular remodeling following wire‑induced
endothelial injury. This suggests IL‑11 inhibition as a potential novel therapeutic approach to reduce
arterial stenosis following revascularization in CAD and PAD patients.
OPEN
1Institute of Experimental Medicine and Systems Biology, University Hospital, RWTH Aachen University,
Aachen, Germany. 2Department of Anesthesiology, University Hospital, RWTH Aachen University, Aachen,
Germany. 3Department of Cardiology, Angiology and Intensive Medicine, University Hospital Aachen, Aachen,
Germany. 4Victor Babes National Institute of Pathology, Bucharest, Romania. 5Department of Intensive Care
and Intermediate Care, University Hospital, RWTH Aachen University, Aachen, Germany. 6National Heart
Research Institute Singapore, National Heart Centre, Singapore 169609, Singapore. 7Translational Laboratories
in Genetic Medicine, Agency for Science, Research and Technology, Singapore 138648, Singapore. 8Yong Loo
Lin School of Medicine, National University Singapore, Singapore 169857, Singapore. 9Cardiovascular Research
Institute, National University Health System, Singapore 119228, Singapore. 10Cardiovascular and Metabolic
Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore 169857,
Singapore. 11Department of Biochemistry, Medical Faculty, Justus Liebig-University, Giessen, Germany. 12MRC
LMS, London W12 0NN, UK. 13The Hatter Cardiovascular Institute, University College London, London WC1E
6BT, UK. 14Cardiovascular Research Center, College of Medical and Health Sciences, Asia University, Taichung,
Taiwan. 15These authors contributed equally: David Schumacher and Elisa A. Liehn. 16These authors jointly
supervised this work: Stuart A. Cook and Derek J. Hausenloy. *email: derek.hausenloy@duke-nus.edu.sg
Content courtesy of Springer Nature, terms of use apply. Rights reserved
2
Vol:.(1234567890)
Scientic Reports | (2021) 11:20674 | https://doi.org/10.1038/s41598-021-99880-y
www.nature.com/scientificreports/
Abbreviations
IL-11 Interleukin-11
VSMC Vascular smooth muscle cells
TGF-β Transforming growth factor-beta
ANGII Angiotensin-II
SMA Smooth muscle actin
MMP2 Matrix metalloproteinase 2
SM22 α Smooth muscle protein 22α
Despite advances in stent design and revascularization therapies, vascular restenosis remains a major problem in
patients with coronary artery disease (CAD) and peripheral artery disease (PAD)14. In-stent restenosis can lead
to severe complications such as cardiac ischemia and chronic limb threatening ischemia, and new therapeutic
strategies are needed to prevent these complications. Vascular smooth muscle cells (VSMCs) switching from its
contractile phenotype to a synthetic phenotype is a major contributor to neointimal hyperplasia, the key pathol-
ogy underlying vascular restenosis2,3.
VSMCs are specialized cells found within the medial layer of the vasculature where their primary function
is to regulate vessel tone and blood pressure. In response to vascular injury, VSMCs proliferate, migrate into the
tunica intima and assume a synthetic phenotype which is an adaptive response but results in vessel wall thicken-
ing. e synthetic VSMC phenotype is characterized by secretion of extracellular matrix, leading to brosis and
inammation. e cellular transition to a synthetic phenotype is termed phenotypic switching and plays a key
role in arterial restenosis, aortic remodelling, and the development of atherosclerosis511.
Two key factors associated with VSMC phenotypic switching and vascular pathologies such as atherosclerosis
and arterial restenosis are transforming growth factor-beta (TGFβ) and angiotensin-II (ANGII)1214. Fibroblast-
to-myobroblast dierentiation and VSMC phenotypic switching share many similarities, including the secretion
of extracellular matrix, cell proliferation and migration, and both transitions can be triggered by the same stimuli.
We have recently discovered that IL-11, a little studied cytokine of the IL-6 family, is important for broblast
activation downstream of both TGFβ1 and ANGII as well as for VSMC phenotypic switching, in response to
the same stimuli15,16. We hypothesized that IL-11 might play a role in vessel hyperplasia and investigated the
eects of the neutralizing IL-11 antibody (X203) or an isotype control IgG antibody in a carotid wire-induced
endothelial injury mouse model.
Material and methods
All experiments and methods were performed in accordance with relevant guidelines and regulations. All animal
experiments were performed in accordance with ARRIVE (Animal Research: Reporting of InVivo Experiments)
guidelines and approved by the Biomedical Sciences Institute Singapore Institutional Animal Care Committee
at A*STAR (161165). All methods are reported in accordance with ARRIVE guidelines.
Mouse husbandry. All experiments were approved by the Biomedical Sciences Institute Singapore Insti-
tutional Animal Care Committee at A*STAR (161165). Mice were maintained on a 12h dark–light cycle, with
adlibitum access to water and were fed with lipid-rich Western-Type Diet (D12079B, Research Diets, NJ), as
indicated. Plasma for all experiments was isolated from blood withdrawn from the orbital sinus in EDTA coated
capillary tubes. Plasma alanine aminotransferase (ALT), aspartate aminotransferase (AST), low-density lipopro-
tein (LDL-C), high-density lipoprotein (HDL-C), triglycerides (TG) and total cholesterol were measured using
Cobas c111 (Roche Diagnostics, Switzerland).
Carotid artery wire injury model of vascular restenosis. Male, 10 to 12week Apoe−/− mice (C57BL/6J
background, Charles River Laboratory, Italy) were fed lipid-rich Western-Type Diet17 for a total of 3weeks:
1week before and 2weeks aer wire injury. Only male mice were used for this study to avoid the interference
of estrogen eects on injury-related plaque with our target of interest IL-11. For the wire injury procedure, mice
were anesthetized (100mg/kg ketamine hydrochloride, 10mg/kg xylazine i.p.) and subjected to endothelial
denudation of the le common carotid artery using a 1cm insertion of a exible 0.36mm guide wire through
a transverse arteriotomy of the external carotid artery, as previously described17. Prior to surgery and for up to
2days post-surgery, we performed analgesia with subcutaneous injection of Buprenorphine (0.05–0.1mg/kg).
Mice were randomly divided in three treatment groups: (1) PBS, (2) IgG, and (3) anti-IL-11 antibody treatment.
Twenty mg/kg anti-IL-11 mouse monoclonal antibody (Clone 3C6; X203) or control IgG (Clone 11E10) were
administered via intra-peritoneal injections16, twice per week beginning on the day of wire injury and continu-
ing for 2weeks (Fig.1A).
X203 was generated in mice using a cDNA encoding amino acid 22–199 of human IL-11 cloned into expres-
sion plasmids (Aldevron Freiburg GmbH, Freiburg, Germany), as described previously18. Its ecacy has already
been demonstrated in arterial remodeling16, myocardial infarction15, liver brosis19 and pulmonary brosis18.
Ultrasound measurements of the carotid arteries and heart. Mice were anesthetized with 2% iso-
urane and monitored to maintain heart rate above 500beats/min during measurements. Measurements were
performed in B-Mode and M-Mode. Velocities were recorded and measured in B-Mode (2D-realtime) using
angle correction and vessel diameters (wall thickness) were recorded and analyzed in M-Mode using a 40MHz
transducer and a small-animal ultrasound imager (Vevo 3100, FUJIFILM Visualsonics, Toronto, Canada) as well
as the VevoLab Soware (FUJIFILM Visualsonics, Toronto, Canada).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
3
Vol.:(0123456789)
Scientic Reports | (2021) 11:20674 | https://doi.org/10.1038/s41598-021-99880-y
www.nature.com/scientificreports/
Histology and immunohistochemistry. Two weeks following wire injury, mice were anesthetized
(100mg/kg ketamine, 10mg/kg xylazine, i.p.) and carotid arteries were excised, xed in formalin and embed-
ded in paran. e carotid arteries were then cut in 5µm serial sections starting from the bifurcation until
500µm, for all the collected sections. Verhoe Van Gieson Elastic stain was performed as recommended by the
manufacturer (ab150667, Abcam, Cambridge, UK) in 10 serial sections (50µm apart, starting from the bifurca-
tion) for the le side and 4 serial sections (50µm apart, starting from the bifurcation for the right side). Injury-
related plaque areas were determined for all sections using Diskus soware (Hilgers, Königswinter, Germany),
as previously described17. e average of all 10 sections for the le side (4 for right side) was considered as nal
restenosis area for each vessel.
For further measurements and to minimize variability of arterial layers aer mechanical injury, we performed
the measurements in whole vessel wall and have referred to it as injury-related plaque. Serial sections (3 sections
per mouse, 100µm apart) were stained to analyze the injury-related plaque and vessel for early dierentiation
of VSMCs (SM22α ab14106, Abcam, Cambridge, UK) and mature VSMCs (smooth muscle actin, M 0851 clone
1A4, DAKO, Germany), macrophages (Mac2, CL8942AP, Cedarlane, Germany) and MMP2 (ab110186, Abcam,
Cambridge, UK). e sections were counterstained with DAPI for quantication of total cells. Positive-stained
cells were counted in the injury-related plaque in each section and expressed as cells per injury-related plaque,
percentage of all cells or percentage of positive area from the total injury-related plaque area. e results are
represented as average of the measurements of all 3 slides.
ree serial sections, 100µm apart, were stained with Gomori’s 1-step trichrome stain (ab150686, Abcam,
Cambridge, UK). Blue-stained collagen content was analyzed with Cell P Soware (Olympus, Hamburg, Ger-
many) and expressed as a percentage of the injury-related plaque area. Final results were represented as average
of the measurements of all 3 slides.
Figure1. e eect of anti-IL-11 antibody (X203) treatment on plasma lipids. (A) A schematic of the
experimental design. (B) Plasma concentration of AST, ALT, triglycerides, total cholesterol, LDL cholesterol and
HDL cholesterol (N = 12–14/group, One-way ANOVA, Tukey’s multiple comparison test, Values: mean ± SD).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
4
Vol:.(1234567890)
Scientic Reports | (2021) 11:20674 | https://doi.org/10.1038/s41598-021-99880-y
www.nature.com/scientificreports/
Statistical analysis. Data are presented as mean ± SD. Statistical analysis were performed with Prism 6.1
soware (GraphPad). For analyses between more than 2 groups we used 1-way ANOVA followed by Tukey’s
multiple comparison test. P values of < 0.05 were considered signicant.
Results
X203 treatment for 2 weeks did not aect blood lipids. To investigate the eect of IL-11 inhibi-
tion on neointimal hyperplasia, we performed wire injury in mice randomly assigned to receive either control
IgG, the anti-IL-11 antibody X203, or no treatment (Fig.1A). Since circulating lipids play an important role in
arteriosclerosis and neointimal hyperplasia, we rst assessed plasma lipids. With short term treatment, there
were no dierences in triglyceride, total cholesterol, LDL cholesterol or HDL cholesterol levels in mice across
experimental groups (Fig.1B). Furthermore, liver transaminases were similar in all treatment groups (Fig.1B)
suggesting that systemic anti-IL-11 antibody administration did not aect plasma lipids or liver function over
the experimental time course.
X203 treatment reduced post‑wire injury neointimal hyperplasia. To determine the eect of IL-11
inhibition on neointimal hyperplasia, we performed ultrasound analyses of injured carotid arteries. Wall thick-
ness was signicantly reduced in the X203 treated group compared to controls, whilst there were no dierences
in blood ow velocity (Fig.2A,B). Interestingly, the ultrasound measurements of right-side, uninjured carotid
artery show signicant thinning aer X203 treatment, while velocity showed no dierences (Fig.2C,D), dem-
onstrating the eect of the X203 treatment on arterial remodeling. Original acquired ultrasound images are now
presented in a supplementary gure (Suppl. Fig.1).
In addition, injury-related plaque (Fig.3A), neointimal areas (Fig.3B) were signicantly reduced in X203-
treated mice compared to controls. ere were no dierences in tunica media area between the treatment groups
(Fig.3C) Representative images are shown in Fig.3D. Analyzing the right, uninjured carotid arteries, we found
no dierences in total vessel area (Fig.3E), intima (Fig.3F) or media (Fig.3G). Representative images are shown
in Fig.3H. One out of 12 control carotid arteries (Suppl. Fig.2A) and one out of 13 IgG-treated carotid arteries
developed native atherosclerotic plaques, whereas none of the 14 X203-treated carotid arteries developed native
atherosclerotic plaque.
X203 treatment had no eect on post‑endothelial injury macrophage injury‑related plaque
inltration. Inammation and macrophages play an important role in arteriosclerosis and vascular
restenosis20. erefore, we assessed the eect of IL-11 inhibition on the number of macrophages inltrating the
Figure2. e eect of anti-IL-11 antibody (X203) treatment on vessel wall thickness. (A) Velocity and wall
thickness of injured le carotid artery (N = 10/group, One-way ANOVA, Tukey’s multiple comparison test,
Values ± SD). (B) Representative M-Mode images of injured le carotid artery. Brackets show carotid artery size,
and arrows point out the measured wall thickness. (C) Velocity and wall thickness of uninjured right carotid
artery (N = 10/group, One-way ANOVA, Tukey’s multiple comparison test, Values ± SD). (D) Representative
M-Mode images of uninjured right carotid artery. Brackets show carotid artery size, and arrows point out the
measured wall thickness.
Content courtesy of Springer Nature, terms of use apply. Rights reserved
5
Vol.:(0123456789)
Scientic Reports | (2021) 11:20674 | https://doi.org/10.1038/s41598-021-99880-y
www.nature.com/scientificreports/
injury-related plaque area. Neither the proportion of macrophages nor the absolute number of macrophages
were dierent across the treatment groups (Fig.4A–C). e right carotid arteries showed no staining or isolated
subendothelial staining for macrophage marker Mac2 (Fig.4D), except the one carotid artery from the control
groups presenting with native atherosclerotic plaque, which showed predominately macrophages inltration
(Suppl. Fig.2B).
X203 treatment reduced post‑endothelial injury VSMC accumulation. Given the central role
of VSMC switching to a synthetic phenotype characterized by proliferation and migration in post-endothelial
injury neointimal hyperplasia, we investigated the eect of IL-11 inhibition on VSMC accumulation and pheno-
type. X203 treatment reduced the numbers of VSMCs in the injury-related plaque (Fig.5A,B,D), and increased
the numbers of VSMCs expressing SM22α, a contractile marker (Fig.5C,E), when compared to controls, sug-
Figure3. e eect of anti-IL-11 antibody (X203) treatment on neointimal hyperplasia. (A) Injury-related
plaque area (N = 11/group, One-way ANOVA, Tukey’s multiple comparison test, Values ± SD). (B) Intima area
(N = 11/group, One-way ANOVA, Tukey’s multiple comparison test, Values ± SD). (C) Media area (N = 11/group,
One-way ANOVA, Tukey’s multiple comparison test, Values ± SD). (D) Representative images of Verhoe Van
Gieson Elastic stain (scale bar 200μm). (E) Vessel area of control uninjured right carotid arteries (N = 12–14/
group, One-way ANOVA, Tukey’s multiple comparison test, Values ± SD). (F) Intima area (N = 12–14/group,
One-way ANOVA, Tukey’s multiple comparison test, Values ± SD). (G) Media area (N = 12–14/group, One-way
ANOVA, Tukey’s multiple comparison test, Values ± SD). (H) Representative images of Verhoe Van Gieson
Elastic stain (scale bar 200μm).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
6
Vol:.(1234567890)
Scientic Reports | (2021) 11:20674 | https://doi.org/10.1038/s41598-021-99880-y
www.nature.com/scientificreports/
gesting that most VSMCs in the injury-related plaques of the X203-treated mice were contractile in phenotype,
suggesting atheroprotection. e right uninjured carotid arteries showed mainly vascular smooth muscle cells
in the vessel wall (Fig.5F).
X203 treatment reduced post‑endothelial injury injury‑related plaque brosis. Several recent
studies have reported the pro-brotic properties of IL-1115,16. Given the critical role of VSMC switching to a
synthetic phenotype in post-endothelial injury neointimal hyperplasia, which is characterized by the secretion
of extracellular matrix, we investigated the eect of IL-11 inhibition on MMP2 expression and collagen content.
X203 treatment decreased MMP2 expression (Fig.6A,B) and reduced collagen content (Fig.6C,D) in the injury-
related plaque, compared to controls, again suggesting that inhibition of IL-11 is atheroprotective via reducing
VSMC phenotype switching. Interestingly, the uninjured right carotid arteries showed a signicant reduction of
collagen content aer X203 treatment (Fig.6E,F), demonstrating the anti-brotic role of X203 treatment and
protection against arterial remodeling. is may account for the right carotid arteries appearing thinner during
the ultrasound measurements.
Figure4. e eect of anti-IL-11 antibody (X203) treatment on macrophage inltration. (A) Macrophages
per injury-related plaque (N = 11/group, One-way ANOVA, Tukey’s multiple comparison test, Values ± SD).
(B) Macrophage proportion of all cells (N = 11/group, One-way ANOVA, Tukey’s multiple comparison test,
Values ± SD). (C) Representative images of Mac2 immunouorescence staining and corresponding DAPI
staining (insets) (scale bar 100µm). Transparent red-lines were traced to delineate the injury-related plaque area
used for quantications. (D) Representative images of Mac2 staining in the right uninjured carotid arteries and
corresponding DAPI staining (insets) (scale bar 100µm).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
7
Vol.:(0123456789)
Scientic Reports | (2021) 11:20674 | https://doi.org/10.1038/s41598-021-99880-y
www.nature.com/scientificreports/
Discussion
We demonstrate for the rst time that inhibition of IL-11 reduces vessel wall thickness and neointimal hyper-
plasia in a carotid wire-induced endothelial injury mouse model. ese ndings wereassociated with benecial
eects on post-endothelial injury injury-related plaque remodelling as evidenced by decreased accumulation
of VSMCs, increased proportion of contractile VSMCs, lower MMP2 expression, and reduced collagen content
in the injury-related plaque. Interestingly, inhibition of IL-11 reduced the collagen content of uninjured carotid
arteries as compared to either IgG or buer control mice, showing an additional eect on hyperlipidemia-induced
arterial remodeling in the absence of mechanical injury.
Figure5. e eect of anti-IL-11 antibody (X203) treatment on injury-related plaque VSMC accumulation.
(A) SMA+ VSMCs per injury-related plaque (N = 11/group, One-way ANOVA, Tukey’s multiple comparison
test, Values ± SD). (B) SMA+ VSMCs percent of all cells (N = 11/group, One-way ANOVA, Tukey’s multiple
comparison test, Values ± SD). (C) SM22α expression in VSMCs in the injury-related plaque (N = 11/group,
One-way ANOVA, Tukey’s multiple comparison test, Values ± SD). (D) Representative images of SMA
immunouorescence staining (red) and corresponding DAPI staining (blue, insets) in le, injured carotid
arteries (scale bar 100µm). Transparent yellow-lines were traced to delineate the injury-related plaque area
used for quantications. (E) Representative images of SM22α (green) and SMA (red) immunouorescence
co-staining (yellow, scale bar 50μm). Transparent blue-lines were traced to delineate the injury-related plaque
area used for quantications. (F) Representative images of SMA immunouorescence staining (red) and
corresponding DAPI staining (blue, insets) in right, uninjured carotid arteries (scale bar 100µm).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
8
Vol:.(1234567890)
Scientic Reports | (2021) 11:20674 | https://doi.org/10.1038/s41598-021-99880-y
www.nature.com/scientificreports/
Autocrine IL-11 signaling is a key downstream eector of TGFβ1 and ANGII in dierent cell types involved
in extracellular matrix (ECM) production15,16,18,19. In cardiac broblasts15, lung broblasts18 and hepatic stellate
cells19, IL-11 is required for ERK-dependent myobroblast activation. Interestingly, TGFβ1 induces IL-11 secre-
tion from aortic and coronary artery VSMCs21,22. Recently, we showed a role of IL-11 in phenotypic switching
of VSMCs and discovered the existence of an autocrine loop of IL-11 activity in VSMCs, which is required
downstream of both TGFβ1 and ANGII for phenotypic switching to occur in the context of aortic modelling16.
However, the role of IL-11 in phenotypic switching and function of VSMCs in the setting of neointimal hyper-
plasia following endothelial injury has not been investigated. It is known that X203 treatment has a positive
eect on arterial remodeling in the context of hypertension16, on cardiac brosis and healing aer myocardial
infarction15, on liver brosis19 and in idiopathic pulmonary brosis18.
In this study, we showed that inhibiting IL-11 reduced neointimal proliferation and had favorable eects on
injury-related plaque remodelling following wire-induced endothelial injury in the mouse carotid artery. As
expected, carotid wire injury induced neointimal hyperplasia in control mice as evidenced by an increase in vessel
wall thickness and tunica intima area, and treatment with IgG had no eect on these parameters. Treatment with
the anti-IL-11 antibody X203 signicantly reduced vessel wall thickness (with no eects on carotid artery veloc-
ity) and decreased total injury-related plaque area (with a reduction in tunica intima area but no eect on media
area). e reduction in injury-related plaque area with X203 treatment was associated with decreased numbers of
VSMCs, with an increased expression of SM22α, a marker for VSMCs with a preserved contractile phenotype23.
ese ndings are consistent with our prior study showing that genetic or antibody-mediated inhibition of
IL-11 attenuated VSMC phenotypic switching16. Several studies have identied macrophages to be important
contributors to vascular restenosis24,25. However, we found no dierences in macrophage injury-related plaque
Figure6. e eect of anti-IL-11 antibody (X203) treatment on post-endothelial injury injury-related plaque
brosis. (A) MMP2 positive staining per injury-related plaque (N = 11/group, One-way ANOVA, Tukey’s
multiple comparison test, Values ± SD). (B) Representative images of MMP2 immunouorescence staining
(green, scale bar 100μm). Insets represent higher magnication of positive cells inside the restenosis plaque
(scale bar 50μm). Transparent red-lines were traced to delineate the injury-related plaque area used for
quantications. (C) Collagen content (blue) of the injury-related plaque (N = 11/group, One-way ANOVA,
Tukey’s multiple comparison test Values ± SD). (D) Representative images of Gomori stain (collagen in blue,
muscle in red, scale bar 50μm). (E) Collagen content (blue) of right, uninjured carotid arteries (N = 12–14/
group, One-way ANOVA, Tukey’s multiple comparison test Values ± SD). (F) Representative images of Gomori
stain (collagen in blue, muscle in red, scale bar 50μm).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
9
Vol.:(0123456789)
Scientic Reports | (2021) 11:20674 | https://doi.org/10.1038/s41598-021-99880-y
www.nature.com/scientificreports/
inltration with X203 treatment, suggesting that the benecial eects of inhibiting IL-11 on reducing neointimal
hyperplasia were independent of macrophage accumulation into the injury-related plaque at this timepoint.
In our previous study we showed that IL-11 induces phenotypic switching of VSMCs to a synthetic phenotype
characterized by secretion of collagen and extracellular matrix proteins, including MMP216, which is involved
with phenotype switching and migration26,27. Furthermore, expression of the VSMC contractile marker SMA22α
was also decreased in response to IL-11 antibodytreatment in the same study16. Consistent with a pathological
role for IL-11 on VSMC function, we show here that treatment with X203 decreased injury-related plaque MMP2
levels and increased injury-related plaque SMA22α levels, suggesting a favorable eect of IL-11 inhibition on
vascular remodelling following wire-induced endothelial injury.
We highlight that while IL-11 was discovered three decades ago, there is very little known of its eects in the
vasculature. Limited earlier studies have suggested IL-11 as anti-inammatory, anti-brotic and pro-regenera-
tive28 and in the vasculature, IL-11 has been thought to inhibit VSMC proliferation and plaque formation29, the
opposite of what we demonstrate here. One reason for the general misunderstanding of IL-11 function relates to
the repeated use of recombinant human IL-11 in mouse models of disease. Paradoxically, it was recently shown
that rhIL-11 is a competitive inhibitor of mouse IL-11 in mouse cells and thus much of the earlier literature may
need to be reconsidered30.
In conclusion, we show for the rst time that inhibition of IL-11 reduced neointimal hyperplasia following
endothelial injury and had favorable eects on vascular remodelling. ese ndings position the IL-11 antibody
(X203) as a novel therapeutic strategy for preventing post-angioplasty/stent restenosis and improving outcomes
in CAD and PAD patients undergoing revascularization.
Received: 11 May 2021; Accepted: 24 September 2021
References
1. Lee, M. S. & Banka, G. In-stent restenosis. Interv. Cardiol. Clin. 5, 211–220 (2016).
2. Osadnik, T. et al. e relationships between polymorphisms in genes encoding the growth factors TGF-beta1, PDGFB, EGF, bFGF
and VEGF-A and the restenosis process in patients with stable coronary artery disease treated with bare metal stent. PLoS ONE
11, e0150500 (2016).
3. Pallero, M. A. et al. Stainless steel ions stimulate increased thrombospondin-1-dependent TGF-beta activation by vascular smooth
muscle cells: implications for in-stent restenosis. J. Vasc. Res. 47, 309–322 (2010).
4. Nicolais, C. et al. erapeutic Options for In-Stent Restenosis. Curr. Cardiol. Rep. 20, 7 (2018).
5. van Varik, B. J. et al. Mechanisms of arterial remodeling: Lessons from genetic diseases. Front. Genet. 3, 290 (2012).
6. Alexander, M. R. & Owens, G. K. Epigenetic control of smooth muscle cell dierentiation and phenotypic switching in vascular
development and disease. Annu. Rev. Physiol. 74, 13–40 (2012).
7. Kuang, S. Q. et al. Aortic remodeling aer transverse aortic constriction in mice is attenuated with AT1 receptor blockade. Arte-
rioscler. romb. Vasc. Biol. 33, 2172–2179 (2013).
8. Elia, L. et al. UHRF1 epigenetically orchestrates smooth muscle cell plasticity in arterial disease. J. Clin. Invest. 128, 2473–2486
(2018).
9. Ren, X. S. et al. NLRP3 gene deletion attenuates angiotensin II-induced phenotypic transformation of vascular smooth muscle
cells and vascular remodeling. Cell Physiol. Biochem. 44, 2269–2280 (2017).
10. Zhang, Z. et al. Upregulation of TRPM7 channels by angiotensin II triggers phenotypic switching of vascular smooth muscle cells
of ascending aorta. Circ. Res. 111, 1137–1146 (2012).
11. Clement, M. et al. Vascular smooth muscle cell plasticity and autophagy in dissecting aortic aneurysms. Arterioscler. romb. Vasc.
Biol. 39, 1149–1159 (2019).
12. Gallo, E. M. et al. Angiotensin II-dependent TGF-beta signaling contributes to Loeys-Dietz syndrome vascular pathogenesis. J.
Clin. Invest. 124, 448–460 (2014).
13. Holm, T. M. et al. Noncanonical TGFbeta signaling contributes to aortic aneurysm progression in Marfan syndrome mice. Science
332, 358–361 (2011).
14. Ford, C. M., Li, S. & Pickering, J. G. Angiotensin II stimulates collagen synthesis in human vascular smooth muscle cells. Involve-
ment of the AT(1) receptor, transforming growth factor-beta, and tyrosine phosphorylation. Arterioscler. romb. Vasc. Biol. 19,
1843–1851 (1999).
15. Schafer, S. et al. IL-11 is a crucial determinant of cardiovascular brosis. Nature 552, 110–115 (2017).
16. Lim, W. W. et al. Interleukin-11 is important for vascular smooth muscle phenotypic switching and aortic inammation, brosis
and remodeling in mouse models. Sci. Rep. 10, 17853 (2020).
17. Curaj, A., Zhoujun, W., Staudt, M. & Liehn, E. A. Induction of accelerated atherosclerosis in mice: e “wire-injury” model. J. Vis.
Exp. 162, e54571 (2020).
18. Ng, B. et al. Interleukin-11 is a therapeutic target in idiopathic pulmonary brosis. Sci. Transl. Med. 11, eaaw1237 (2019).
19. Widjaja, A. A. et al. Inhibiting interleukin 11 signaling reduces hepatocyte death and liver brosis, inammation, and steatosis in
mouse models of nonalcoholic steatohepatitis. Gastroenterology 157, 777–792 (2019).
20. Frodermann, V. & Nahrendorf, M. Macrophages and cardiovascular health. Physiol. Rev. 98, 2523–2569 (2018).
21. Lebastchi, A. H. et al. Activation of human vascular cells decreases their expression of transforming growth factor-beta. Athero-
sclerosis 219, 417–424 (2011).
22. Taki, H. et al. Monokine stimulation of interleukin-11 production by human vascular smooth muscle cells invitro. Atherosclerosis
144, 375–380 (1999).
23. Halayko, A. J. et al. Divergent dierentiation paths in airway smooth muscle culture: Induction of functionally contractile myocytes.
Am. J. Physiol. 276, 197–206 (1999).
24. Zhang, M. et al. In-stent restenosis is associated with neointimal angiogenesis and macrophage inltrates. Pathol. Res. Pract. 210,
1026–1030 (2014).
25. Yahagi, K. et al. Pathophysiology of native coronary, vein gra, and in-stent atherosclerosis. Nat. Rev. Cardiol. 13, 79–98 (2016).
26. Vigetti, D. et al. Matrix metalloproteinase 2 and tissue inhibitors of metalloproteinases regulate human aortic smooth muscle cell
migration during invitro aging. FASEB 20, 1118–1130 (2006).
27. Risinger, G. M., Hunt, T. S., Updike, D. L., Bullen, E. C. & Howard, E. W. Matrix metalloproteinase-2 expression by vascular smooth
muscle cells is mediated by both stimulatory and inhibitory signals in response to growth factors. J. Biol. Chem. 281, 25915–25925
(2006).
Content courtesy of Springer Nature, terms of use apply. Rights reserved
10
Vol:.(1234567890)
Scientic Reports | (2021) 11:20674 | https://doi.org/10.1038/s41598-021-99880-y
www.nature.com/scientificreports/
28. Cook, S. A. & Sebastian, S. S. Hiding in plain sight: Interleukin-11 emerges as a master regulator of brosis, tissue integrity, and
stromal inammation. Annu. Rev. Med. 27(71), 263–276 (2020).
29. Zimmerman, M. A. et al. Interleukin-11 attenuates human vascular smooth muscle cell proliferation. Am. J. Physiol. Heart Circ.
Physiol. 283(1), H175–H180 (2002).
30. Widjaja, A. A. et al. Redening IL11 as a regeneration-limiting hepatotoxin and therapeutic target in acetaminophen-induced
liver injury. Sci. Transl. Med. 13(597), eaba8146 (2021).
Acknowledgements
We thank Roya Soltan and Melanie Garbe for excellent technical assistance.
Author contributions
Conceptualization: D.J.H., S.A.C.; Methodology: E.A.L., D.S., R.R.S., S.A.C., D.J.H.; Formal analysis and inves-
tigation: D.S., E.A.L., P.N., D.C.M., C.R., K.A., G.C., S.H.R., R.R.S.; Writing—original dra preparation: D.S.,
E.A.L.; Writing—review and editing: R.R.S., D.J.H., S.A.C.; Funding acquisition: D.J.H.; Resources: D.J.H., E.A.L.,
S.A.C.; Supervision: D.J.H., S.A.C. All authors commented on previous versions of the manuscript and all authors
read and approved the nal manuscript.
Funding
is study was supported by the Interdisciplinary Centre for Clinical Research IZKF Aachen (junior research
group to E.A.L.). DS is supported by the Clinician Scientist program of the Faculty of Medicine of the RWTH
Aachen University. RRS was supported by the Agency for Science, Research and Technology (A*STAR) and
the National University of Singapore (NUS). SHR is supported by the Singapore Ministry of Health’s National
Medical Research Council under its Open Fund-Young Individual Research Grant (OF-YIRG)–(NMRC/
OFYIRG/0078/2018). SAC is supported by NMRC STaR award and the Tanoto Foundation. DJH is supported
by the Duke-NUS Signature Research Programme funded by the Ministry of Health, Singapore Ministry of
Healths National Medical Research Council under its Clinician Scientist-Senior Investigator scheme (NMRC/
CSA-SI/0011/2017), Centre Grant (CGAug16M006), and Collaborative Centre Grant scheme (NMRC/
CGAug16C006). is article is based upon work from COST Action EU-CARDIOPROTECTION CA16225
supported by COST (European Cooperation in Science and Technology).
Competing interests
SAC is an inventor on the patent applications: WO/2017/103108 (TREATMENT OF FIBROSIS),
WO/2018/109174 (IL-11 ANTIBODIES), WO/2018/109170 (IL-11RA ANTIBODIES). S.A.C. is a shareholder
of Enleofen Bio PTE LTD. e remaining authors (DS, EAL PN, DCM, CR, KA, GEC, SHR, RRS and DJH)
declare no competing interests.
Additional information
Supplementary Information e online version contains supplementary material available at https:// doi. org/
10. 1038/ s41598- 021- 99880-y.
Correspondence and requests for materials should be addressed to D.J.H.
Reprints and permissions information is available at www.nature.com/reprints.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and
institutional aliations.
Open Access is article is licensed under a Creative Commons Attribution 4.0 International
License, which permits use, sharing, adaptation, distribution and reproduction in any medium or
format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the
Creative Commons licence, and indicate if changes were made. e images or other third party material in this
article are included in the articles Creative Commons licence, unless indicated otherwise in a credit line to the
material. If material is not included in the article’s Creative Commons licence and your intended use is not
permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from
the copyright holder. To view a copy of this licence, visit http:// creat iveco mmons. org/ licen ses/ by/4. 0/.
© e Author(s) 2021
Content courtesy of Springer Nature, terms of use apply. Rights reserved
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com
... Prepared using the SYRCLE tool [31]. Green (low risk of bias); 36 red (high risk of bias); yellow (uncertain risk of bias) [5,15,18,[25][26][27]32,33]. 37 38 For the studies evaluating the relationship between IL-11 and cardiac fibrosis, 39 Masson's trichrome or picrosirius red were the stains used (five studies). ...
... Corroborating the cardiac findings, in a model of nonalcoholic steatohepatitis, IL-11 inhibition by X203 or X209 decreased liver fibrosis by suppressing ERK activation and consequently MMP2 and TIMP1 [35]. Similar results were observed in arterial remodeling [36], Marfan syndrome [28], and pulmonary fibrosis, which shows the influence of IL-11 on fibrosis [37]. ...
Article
Full-text available
Fibrosis is one of the main factors that impair the function of many organs. In the heart, fibrosis leads to contractile dysfunction and arrhythmias, which are important in the development of heart failure. Interleukin (IL)-11 is regulated in various heart diseases and has recently been reported to be an important cytokine in fibrosis in this organ. However, this topic has been little explored, and many questions persist. Thus, this systematic review aimed to report on possible IL-11 therapies evaluated in rodent model-induced cardiac fibrosis. Inclusion criteria were experimental in vivo studies that used different rodent models for cardiac fibrosis associated with IL-11 interventions, without year and language restrictions. The search in PubMed, Web of Science, and Embase databases was performed in October 2022. The risk of bias assessment of the studies was based on the guidelines of the SYRCLE tool, and data from the selected articles were also presented in a table as a narrative description. This review was based on eight studies in which five different interventions were used: recombinant human IL-11 (rhIL-11), anti-IL11 (X203), recombinant mouse IL-11 (rmIL-11), lentivirus (LV)-IL-11 + lutein, and anti-IL11RA (X209). Based on the included studies, the results were variable, with IL-11 overexpression inducing cardiac fibrosis, while inhibition protected against this process, preserving the function of this organ. Therefore, IL-11 stands out as a promising therapeutic target for cardiac fibrosis. However, further studies are needed to understand the mechanisms triggered by each treatment, as well as its safety and immunogenicity.
... In recent years, IL11RA has emerged as a significant player in various cancers, being implicated in gastric tumorigenesis, colorectal and MMP2/MMP9 levels contribute to ERK-mediated thoracic aortic pathology. 28 In a mouse model of injury-induced plaque development, therapeutic inhibition of IL-11 using X203 reduced vessel thickness, neointimal hyperplasia, and MMP2 levels, preserving the contractile VSMC phenotype, suggesting a potential therapy for arterial stenosis post-revascularization. 29 Binding of ligands to IL-11RA induces the phosphorylation of associated JAK kinases, leading to the recruitment of STAT family transcription factors, particularly STAT3. 30 ...
Article
Full-text available
Objective This study aimed to investigate the role of Interleukin‐11 receptor alpha (IL11RA) in skin cutaneous melanoma (SKCM) metastasis to the liver. Methods Human SKCM cell lines (A375, A375‐MA2, SK‐MEL‐28, RPMI‐7951) and primary dermal fibroblasts (HDFa) were utilized to assess IL11RA expression. IL11RA siRNA was transfected into RPMI‐7951 and A375‐MA2 cells for Wound healing and Transwell invasion assays. Il11ra knockout (KO) mice and wild‐type (WT) mice were injected with B16‐F10 cells into the spleen to evaluate hepatic melanoma metastasis. Correlation between IL11RA and MMP family genes was explored using online databases, including LinkedOmics, TIMER (Tumor Immune Estimation Resource), and GEPIA (Gene Expression Profiling Interactive Analysis). RT‐qPCR and Western blotting were performed for expression analysis of Mmp2 and Mmp9 in liver tissues of mice. The impact of IL11RA on the STAT3 pathway was investigated in vitro and in vivo. Results Elevated expression of IL11RA was observed in SKCM cell lines compared to normal cells. IL11RA downregulation significantly inhibited migratory and invasive capabilities of A375‐MA2 and RPMI‐7951 in vitro. Il11ra gene knockout in mice demonstrated a substantial reduction in hepatic melanoma metastasis. Correlation analyses revealed associations between IL11RA and MMP2/MMP8. Il11ra gene knockout significantly decreased Mmp2 expression while increasing Mmp8 in liver tissues. IL11RA correlated positively with STAT3, and its inhibition led to a suppressed STAT3 pathway in SKCM cells and mouse liver tissue. Conclusion IL11RA plays a crucial role in SKCM metastasis, affecting migratory and invasive abilities. Targeting IL11RA may offer a promising avenue for therapeutic interventions in cutaneous melanoma progression.
... Inhibition of IL-11 signalling has been shown to provide therapeutic benefit in models of arthritis, multiple sclerosis, neointimal hyperplasia, multiple fibrotic diseases, and gastrointestinal cancers [9][10][11]13,17,20,21,24,41,42 . Current IL-11 signalling inhibitors include IL-11 mutants 43,44 and antibodies against either IL-11 9,20,45 or IL-11Rα 11,[46][47][48] . ...
Article
Full-text available
Interleukin (IL-)11, an IL-6 family cytokine, has pivotal roles in autoimmune diseases, fibrotic complications, and solid cancers. Despite intense therapeutic targeting efforts, structural understanding of IL-11 signalling and mechanistic insights into current inhibitors are lacking. Here we present cryo-EM and crystal structures of the human IL-11 signalling complex, including the complex containing the complete extracellular domains of the shared IL-6 family β-receptor, gp130. We show that complex formation requires conformational reorganisation of IL-11 and that the membrane-proximal domains of gp130 are dynamic. We demonstrate that the cytokine mutant, IL-11 Mutein, competitively inhibits signalling in human cell lines. Structural shifts in IL-11 Mutein underlie inhibition by altering cytokine binding interactions at all three receptor-engaging sites and abrogating the final gp130 binding step. Our results reveal the structural basis of IL-11 signalling, define the molecular mechanisms of an inhibitor, and advance understanding of gp130-containing receptor complexes, with potential applications in therapeutic development.
... Additionally, an anti-IL-11 antibody has been evaluated in preclinical studies for liver and renal fibrosis among other fibrotic and inflammatory conditions. 25,44,45 Antibodies benefit from avidity effects due to bivalency but can also induce receptor clustering and reduce diffusion due to their larger molecular weight. Engineered cytokines such as enIL-11 ...
Article
Full-text available
The cytokine interleukin (IL)‐11 has been shown to play a role in promoting fibrosis and cancer, including lung adenocarcinoma, garnering interest as an attractive target for therapeutic intervention. We used combinatorial methods to engineer an IL‐11 variant that binds with higher affinity to the IL‐11 receptor and stimulates enhanced receptor‐mediated cell signaling. Introduction of two additional point mutations ablates IL‐11 ligand/receptor association with the gp130 coreceptor signaling complex, resulting in a high‐affinity receptor antagonist. Unlike wild‐type IL‐11, this engineered variant potently blocks IL‐11‐mediated cell signaling and slows tumor growth in a mouse model of lung cancer. Our approach highlights a strategy where native ligands can be engineered and exploited to create potent receptor antagonists.
Preprint
Full-text available
Appropriate cellular recognition of viruses is essential for the generation of effective innate and adaptive antiviral immunity. Viral sensors and their signalling components thus provide a crucial first line of host defence. Many exhibit subcellular relocalisation upon activation, triggering expression of interferon and antiviral genes. To identify novel signalling factors we analysed protein relocalisation on a global scale during viral infection. CREB Regulated Transcription Coactivators-2 and 3 (CRTC2/3) exhibited early cytoplasmic-to-nuclear translocation upon a diversity of viral stimuli, in diverse cell types. This movement was depended on Mitochondrial Antiviral Signalling Protein (MAVS), cyclo-oxygenase proteins and protein kinase A. We identify a key effect of transcription stimulated by CRTC2/3 translocation as production of the pro-fibrogenic cytokine interleukin-11. This may be important clinically in viral infections associated with fibrosis, including SARS-CoV-2.
Article
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality. An important task in cardiology is the search and study of new biomarkers. Scientific interest is actively focused on the study of interleukin-11 (IL-11). The purpose of the literature review was to analyze experimental and clinical studies devoted to the study of IL-11 as a diagnostic and prognostic marker in CVD. Material and methods. The article provides an overview of current publications. An analysis of literature sources was carried out, including all relevant publications in the databases PubMed, RSCI, MedLine, Google Scholar, Science Direct. Results. In endothelial cells, IL-11 primarily promotes angiogenesis, thereby exerting a beneficial effect on coronary heart disease. IL-11 also promotes vascular remodeling by inducing smooth muscle cell transformation and fibroblast activation. IL-11 may be involved in the onset and progression of pulmonary hypertension through the JAK/STAT3 pathway and aortic dissection processes through the non-classical ERK pathway. Conclusion. The presented literature review indicates the potentially important diagnostic and prognostic value of IL-11 assessment. Regulating the concentration and expression of IL-11 may be a promising strategy for the treatment of CVD.
Article
Full-text available
Vascular fibrosis is a widespread pathologic condition that arises during vascular remodeling in cardiovascular dysfunctions. According to previous studies, vascular fibrosis is characterized by endothelial matrix deposition and vascular wall thickening. The RAAS and TGF-β/Smad signaling pathways have been frequently highlighted. It is, however, far from explicit in terms of understanding the cause and progression of vascular fibrosis. In this review, we collected and categorized a large number of molecules which influence the fibrosing process, in order to acquire a better understanding of vascular fibrosis, particularly of pathologic dysfunction. Furthermore, several mediators that prevent vascular fibrosis are discussed in depth in this review, with the aim that this will contribute to the future prevention and treatment of related conditions.
Article
Full-text available
Interleukin 11 (IL11) is an elusive member of the IL6 family of cytokines. While initially thought to be a haematopoietic and cytoprotective factor, more recent data show instead that IL11 is redundant for haematopoiesis and toxic. In this review, the reasons that led to the original misunderstandings of IL11 biology, which are now understandable, are explained with particular attention on the use of recombinant human IL11 in mice and humans. Following tissue injury, as part of an evolutionary ancient homeostatic response, IL11 is secreted from damaged mammalian cells to signal via JAK/STAT3, ERK/P90RSK, LKB1/mTOR and GSK3β/SNAI1 in autocrine and paracrine. This activates a program of mesenchymal transition of epithelial, stromal, and endothelial cells to cause inflammation, fibrosis, and stalled endogenous tissue repair, leading to organ failure. The role of IL11 signalling in cell- and organ-specific pathobiology is described, the large unknowns about IL11 biology are discussed and the promise of targeting IL11 signalling as a therapeutic approach is reviewed.
Article
Vascular inflammation is an early manifestation and common pathophysiological basis of numerous cardiovascular and cerebrovascular diseases; However, effective surveillance methods are lacking. In this study, we designed sulfur hexafluoride (SF 6 ) loaded polylactic acid‐co‐glycolic acid (PLGA) nanobubbles (NBs) with a surface assembly of cyclodextrin (CD) and sphingosine‐1‐phosphate (S1P) (S1P@CD‐PLGA NBs). The characterization results showed that S1P@CD‐PLGA NBs with diameters of approximately 200 nm had good stability, biosafety, and ultrasound imaging‐enhancement effects. When interacting with inflammatory vascular endothelial cells, S1P molecules encapsulated in cyclodextrin cavities exhibit a rapid, excellent, and stable targeting effect owing to their specific interaction with the highly expressed S1P receptor 1 (S1PR1) on the inflammatory vascular endothelial cells. Particularly, the S1P–S1PR1 interaction further activates the downstream signaling pathway of S1PR1 to reduce the expression of tumor necrosis factor‐α (TNF‐α) to protect endothelial cells. Furthermore, mouse models of carotid endothelial injuries and mesenteric thrombosis demonstrated that S1P@CD‐PLGA NBs have excellent capabilities for in vivo targeting imaging. In summary, this study proposes a new strategy of using S1P to target inflammatory vascular endothelial cells while reducing the expression of TNF‐α, which has the potential to be utilized in the targeted surveillance and treatment of vascular inflammatory diseases. This article is protected by copyright. All rights reserved
Article
Full-text available
Transforming growth factor beta-1 (TGFβ1) is a major driver of vascular smooth muscle cell (VSMC) phenotypic switching, an important pathobiology in arterial disease. We performed RNA-sequencing of TGFβ1-stimulated human aortic or arterial VSMCs which revealed large and consistent upregulation of Interleukin 11 (IL11). IL11 has an unknown function in VSMCs, which highly express the IL11 receptor alpha, suggestive of an autocrine loop. In vitro, IL11 activated ERK signaling, but inhibited STAT3 activity, and caused VSMC phenotypic switching to a similar extent as TGFβ1 or angiotensin II (ANGII) stimulation. Genetic or therapeutic inhibition of IL11 signaling reduced TGFβ1- or ANGII-induced VSMC phenotypic switching, placing IL11 activity downstream of these factors. Aortas of mice with Myh11-driven IL11 expression were remodeled and had reduced contractile but increased matrix and inflammatory genes expression. In two models of arterial pressure loading, IL11 was upregulated in the aorta and neutralizing IL11 antibodies reduced remodeling along with matrix and pro-inflammatory gene expression. These data show that IL11 plays an important role in VSMC phenotype switching, vascular inflammation and aortic pathobiology.
Article
Full-text available
Atherosclerosis is a proliferative fibro-inflammatory disease developing in the arterial wall, inducing a deficient blood flow or a lack of blood flow. Moreover, by rupture of the defective vascular wall, atherosclerosis induces occlusive thrombus formation, which represents the main cause of myocardial infarction or stroke and the most frequent cause of death. Despite the advances in the cardiovascular field, many questions remain unanswered, and additional basic research is essential to improve our understanding of the molecular mechanisms during atherosclerosis and its effects. Due to limited clinical studies, there is a need for representative animal models recreating atherosclerotic conditions such as neointima formation after stent implantation, balloon angioplasty, or endarterectomy. Since the mouse presents many advantages and is the most frequently used model for studying molecular processes, the current study proposes an invasive procedure of endothelial denudation, also known as the wire-injury model, which is representative of the human condition of neointima formation in arteries after revascularization procedures.
Article
Full-text available
Interleukin (IL)-11 is upregulated in a wide variety of fibro-inflammatory diseases such as systemic sclerosis, rheumatoid arthritis, pulmonary fibrosis, inflammatory bowel disease, kidney disease, drug-induced liver injury, and nonalcoholic steatohepatitis. IL-11 is a member of the IL-6 cytokine family and has several distinct properties that define its unique and nonredundant roles in disease. The IL-11 receptor is highly expressed on stromal, epithelial and polarized cells, where noncanonical IL-11 signaling drives the three pathologies common to all fibro-inflammatory diseases—myofibroblast activation, parenchymal cell dysfunction, and inflammation—while also inhibiting tissue regeneration. This cytokine has been little studied, and publications on IL-11 peaked in the early 1990s, when it was largely misunderstood. Here we describe recent advances in our understanding of IL-11 biology, outline how misconceptions as to its function came about, and highlight the large potential of therapies targeting IL-11 signaling for treating human disease.
Article
Full-text available
Background & aims: We studied the role of interleukin 11 (IL11) signaling in the pathogenesis of nonalcoholic steatohepatitis using hepatic stellate cells (HSCs), hepatocytes, and mouse models of non-alcoholic steatohepatitis (NASH). Methods: We stimulated mouse and human fibroblasts, HSCs, or hepatocytes with IL11 and other cytokines and analyzed them by imaging, immunoblot, and functional assays and ELISAs. Mice were given injections of IL11. Mice with disruption of the interleukin 11 receptor subunit alpha gene (Il11ra1-/-) mice and Il11ra1+/+ mice were fed a high-fat methionine- and choline-deficient diet (HFMCD) or a Western diet with liquid fructose (WDF) to induce steatohepatitis; control mice were fed normal chow. db/db mice were fed with MCD for 12 weeks and C57BL/6 NTac were fed with HFMCD for 10 weeks or WDF for 16 weeks. Some mice were given intraperitoneal injections of anti-IL11 (X203), anti-IL11RA (X209), or a control antibody at different timepoints on the diets. Livers and blood were collected; blood samples were analyzed by biochemistry and liver tissues were analyzed by histology, RNA-seq, immunoblots, immunohistochemistry, hydroxyproline, and mass cytometry time of flight assays. Results: HSCs incubated with cytokines produced IL11, resulting in activation (phosphorylation) of ERK and expression of markers of fibrosis. Livers of mice given injections of IL11 became damaged, with increased markers of fibrosis and inflammation. Following the HFMCD or WDF, livers from Il11ra1-/- mice had reduced steatosis, fibrosis, expression of markers of inflammation and steatohepatitis, and lower levels of serum lipids and glucose compared to and Il11ra1+/+ mice on the same diets. Depending on the time of administration of anti-IL11 or anti-IL11RA to wild-type mice on the HFMCD or WDF, or to db/db mice on the MCD, the antibodies prevented, stopped, or reversed development of fibrosis and steatosis. Blood samples from Il11ra1+/+ mice fed the HFMCD or WDF and given injections of anti-IL11 or anti-IL11RA, as well as from Il11ra1-/- mice fed the diets, had lower serum levels of lipids and glucose than mice not injected with antibody or with disruption of Il11ra1. Conclusions: Neutralizing antibodies that block IL11 signaling reduce fibrosis, steatosis, hepatocyte death, inflammation and hyperglycemia in mice with diet-induced steatohepatitis. These antibodies improve the cardiometabolic profile of mice and might be developed for treatment of NASH.
Article
Full-text available
Objective— Recent studies suggested the occurrence of phenotypic switching of vascular smooth muscle cells (VSMCs) during the development of aortic aneurysm (AA). However, lineage-tracing studies are still lacking, and the behavior of VSMCs during the formation of dissecting AA is poorly understood. Approach and Results— We used multicolor lineage tracing of VSMCs to track their fate after injury in murine models of Ang II (angiotensin II)–induced dissecting AA. We also addressed the direct impact of autophagy on the response of VSMCs to AA dissection. Finally, we studied the relevance of these processes to human AAs. Here, we show that a subset of medial VSMCs undergoes clonal expansion and that VSMC outgrowths are observed in the adventitia and borders of the false channel during Ang II–induced development of dissecting AA. The clonally expanded VSMCs undergo phenotypic switching with downregulation of VSMC differentiation markers and upregulation of phagocytic markers, indicative of functional changes. In particular, autophagy and endoplasmic reticulum stress responses are activated in the injured VSMCs. Loss of autophagy in VSMCs through deletion of autophagy protein 5 gene ( Atg5 ) increases the susceptibility of VSMCs to death, enhances endoplasmic reticulum stress activation, and promotes IRE (inositol-requiring enzyme) 1α-dependent VSMC inflammation. These alterations culminate in increased severity of aortic disease and higher incidence of fatal AA dissection in mice with VSMC-restricted deletion of Atg5 . We also report increased expression of autophagy and endoplasmic reticulum stress markers in VSMCs of human dissecting AAs. Conclusions— VSMCs undergo clonal expansion and phenotypic switching in Ang II–induced dissecting AAs in mice. We also identify a critical role for autophagy in regulating VSMC death and endoplasmic reticulum stress–dependent inflammation with important consequences for aortic wall homeostasis and repair.
Article
Full-text available
Research during the last decade has generated numerous insights on the presence, phenotype, and function of myeloid cells in cardiovascular organs. Newer tools with improved detection sensitivities revealed sizable populations of tissue-resident macrophages in all major healthy tissues. The heart and blood vessels contain robust numbers of these cells; for instance, 8% of noncardiomyocytes in the heart are macrophages. This number and the cell's phenotype change dramatically in disease conditions. While steady-state macrophages are mostly monocyte independent, macrophages residing in the inflamed vascular wall and the diseased heart derive from hematopoietic organs. In this review, we will highlight signals that regulate macrophage supply and function, imaging applications that can detect changes in cell numbers and phenotype, and opportunities to modulate cardiovascular inflammation by targeting macrophage biology. We strive to provide a systems-wide picture, i.e., to focus not only on cardiovascular organs but also on tissues involved in regulating cell supply and phenotype, as well as comorbidities that promote cardiovascular disease. We will summarize current developments at the intersection of immunology, detection technology, and cardiovascular health.
Article
Full-text available
Adult vascular smooth muscle cells (VSMCs) possess the peculiar ability to de-differentiate in response to extracellular cues, such as vascular damage and inflammation. De-differentiated VSMCs are proliferative, migratory, and have decreased contractile capacity. VSMC dedifferentiation contributes not only to vascular repair, but also to cardiovascular pathologies, such as intimal hyperplasia/restenosis in coronary artery or peripheral vascular diseases and arterial aneurysm. We here demonstrate the role of ubiquitin-like, containing PHD and RING finger domains, 1 (UHRF1) as an epigenetic master regulator of VSMC plasticity. The expression of UHRF1 correlates with the development of a wide array of vascular pathologies associated also with modulation of non-coding RNAs, such as microRNAs. Importantly, miR-145, a pivotal gene regulating VSMC plasticity, which is reduced in vascular diseases, was found to control Uhrf1 mRNA translation. In turn, UHRF1 triggers VSMC proliferation by directly repressing the promoters of cell cycle inhibitor genes, such as p21 and p27, and of key pro-differentiation genes via the methylation of DNA and histones. Local vascular viral delivery of Uhrf1 shRNAs or Uhrf1 VSMC-specific deletion prevented intimal hyperplasia in mouse carotid artery and decreased vessel damage in a mouse model of aortic aneurysm.Our study demonstrates the fundamental role of Uhrf1 in regulating VSMC phenotype by promoting proliferation and de-differentiation. UHRF1 targeting may hold therapeutic potential in vascular pathologies, modulating also the VSMC component.
Article
Full-text available
Purpose of review: In-stent restenosis (ISR) is a complex disease process that became apparent shortly after the introduction of stents into clinical practice. This review seeks to define in-stent restenosis (ISR) as well as to summarize the major treatment options that have been developed and studied over the past two decades. Recent findings: Recent developments in drug-coated balloons and bioresorbable vascular scaffolds have added new potential treatments for ISR. Two recent network meta-analyses performed a head-to-head comparison of all the various treatment modalities in order to identify the best approach to management of ISR. Current data suggests that repeat stenting with second-generation drug-eluting stents is most likely to lead to the best angiographic and clinical outcomes. In situations where repeat stenting is not preferable, drug-coated balloon therapy seems to be a reasonably effective alternative.
Article
Acetaminophen ( N -acetyl- p -aminophenol; APAP) toxicity is a common cause of liver damage. In the mouse model of APAP-induced liver injury (AILI), interleukin 11 (IL11) is highly up-regulated and administration of recombinant human IL11 (rhIL11) has been shown to be protective. Here, we demonstrate that the beneficial effect of rhIL11 in the mouse model of AILI is due to its inhibition of endogenous mouse IL11 activity. Our results show that species-matched IL11 behaves like a hepatotoxin. IL11 secreted from APAP-damaged human and mouse hepatocytes triggered an autocrine loop of NADPH oxidase 4 (NOX4)–dependent cell death, which occurred downstream of APAP-initiated mitochondrial dysfunction. Hepatocyte-specific deletion of Il11 receptor subunit alpha chain 1 ( Il11ra1 ) in adult mice protected against AILI despite normal APAP metabolism and glutathione (GSH) depletion. Mice with germline deletion of Il11 were also protected from AILI, and deletion of Il1ra1 or Il11 was associated with reduced c-Jun N-terminal kinase (JNK) and extracellular signal–regulated kinase (ERK) activation and quickly restored GSH concentrations. Administration of a neutralizing IL11RA antibody reduced AILI in mice across genetic backgrounds and promoted survival when administered up to 10 hours after APAP. Inhibition of IL11 signaling was associated with the up-regulation of markers of liver regenerations: cyclins and proliferating cell nuclear antigen (PCNA) as well as with phosphorylation of retinoblastoma protein (RB) 24 hours after AILI. Our data suggest that species-matched IL11 is a hepatotoxin and that IL11 signaling might be an effective therapeutic target for APAP-induced liver damage.
Article
Idiopathic pulmonary fibrosis (IPF) is a progressive fibrotic lung disease where invasive pulmonary myofibroblasts secrete collagen and destroy lung integrity. Here, we show that interleukin-11 ( IL11 ) is up-regulated in the lung of patients with IPF, associated with disease severity, and IL-11 is secreted from IPF fibroblasts. In vitro, IL-11 stimulates lung fibroblasts to become invasive actin alpha 2, smooth muscle–positive (ACTA2 ⁺ ), collagen-secreting myofibroblasts in an extracellular signal–regulated kinase (ERK)–dependent, posttranscriptional manner. In mice, fibroblast-specific transgenic expression or administration of murine IL-11 induces lung myofibroblasts and causes lung fibrosis. IL-11 receptor subunit alpha-1 ( Il11ra1 )–deleted mice, whose lung fibroblasts are unresponsive to profibrotic stimulation, are protected from fibrosis in the bleomycin mouse model of pulmonary fibrosis. We generated an IL-11–neutralizing antibody that blocks lung fibroblast activation downstream of multiple stimuli and reverses myofibroblast activation. In therapeutic studies, anti–IL-11 treatment diminished lung inflammation and reversed lung fibrosis while inhibiting ERK and SMAD activation in mice. These data prioritize IL-11 as a drug target for lung fibrosis and IPF.