You are on page 1of 10

Arteriosclerosis, Thrombosis, and Vascular Biology

ORIGINAL RESEARCH

Differential Roles of Endothelial Cell-Derived


and Smooth Muscle Cell-Derived Fibronectin
Containing Extra Domain A in Early and Late
Atherosclerosis
Prakash Doddapattar, Rishabh Dev, Manish Jain, Nirav Dhanesha, Anil K. Chauhan

OBJECTIVE: The extracellular matrix of atherosclerotic arteries contains abundant deposits of cellular Fn-EDA (fibronectin
containing extra domain A), suggesting a functional role in the pathophysiology of atherosclerosis. Fn-EDA is synthesized
by several cell types, including endothelial cells (ECs) and smooth muscle cells (SMCs), which are known to contribute
to different stages of atherosclerosis. Although previous studies using global Fn-EDA-deficient mice have demonstrated
that Fn-EDA is proatherogenic, the cell-specific role of EC versus SMC-derived-Fn-EDA in atherosclerosis has not been
investigated yet.

APPROACH AND RESULTS: To determine the relative contribution of different pools of Fn-EDA in atherosclerosis, we generated
mutant strains lacking Fn-EDA in the ECs (Fn-EDAEC-KO) or smooth muscle cells (Fn-EDASMC-KO) on apolipoprotein E-deficient
(Apoe−/−) background. The extent of atherosclerotic lesion progression was evaluated in whole aortae, and cross-sections
of the aortic sinus in male and female mice fed a high-fat Western diet for either 4 weeks (early atherosclerosis) or 14
weeks (late atherosclerosis). Irrespective of sex, Fn-EDAEC-KO, but not Fn-EDASMC-KO mice, exhibited significantly reduced
Downloaded from http://ahajournals.org by on May 31, 2020

early atherogenesis concomitant with decrease in inflammatory cells (neutrophil and macrophage) and VCAM (vascular cell
adhesion molecule)-1 expression levels within the plaques. In late atherosclerosis model, irrespective of sex, Fn-EDASMC-
KO
mice exhibited significantly reduced atherogenesis, but not Fn-EDAEC-KO mice, that was concomitant with decreased
macrophage content within plaques. Lesional SMCs, collagen content, and plasma inflammatory cytokines (TNF-α [tumor
necrosis factor α] and IL (interleukin)-1β), total cholesterol, and triglyceride levels were comparable among groups.

CONCLUSIONS: EC-derived Fn-EDA contributes to early atherosclerosis, whereas smooth muscle cell-derived Fn-EDA
contributes to late atherosclerosis.

Key Words: atherosclerosis ◼ cytokines ◼ endothelial cells ◼ fibronectin ◼ macrophages

A
therosclerotic lesion progression is a chronic inflam- atherosclerosis.1 In human and murine models, unlike
matory process characterized by fatty streaks, healthy arteries, the ECM of atherosclerotic arteries con-
leukocytes, smooth muscle cells (SMCs), calcifica- tains abundant deposits of Fn (fibronectin), suggesting a
tion, and extracellular matrix (ECM) remodeling. During functional role for Fn in the pathophysiology of athero-
chronic inflammation, ECM proteins or their fragments sclerosis.2,3 Fn is alternatively spliced that results in vari-
can modulate the migration of several cell types, includ- able inclusion of extra domain A (EDA), extra domain B
ing endothelial cells (ECs), SMCs, and monocyte/mac- (EDB), and the type III homologies connecting segment.
rophages, all known to contribute to different stages of The amino acid sequence and splicing patterns of the


Correspondence to: Prakash Doddapattar, Department of Internal Medicine, University of Iowa, 3120 Medical Labs, Iowa City, IA 52242, Email prakash-doddapattar@
uiowa.edu; or Anil K. Chauhan, Department of Internal Medicine, University of Iowa, 3120 Medical Labs, Iowa City, IA 52242, Email anil-chauhan@uiowa.edu
The Data Supplement is available with this article at https://www.ahajournals.org/doi/suppl/10.1161/ATVBAHA.120.314459.
For Sources of Funding and Disclosures, see page xxx.
© 2020 American Heart Association, Inc.
Arterioscler Thromb Vasc Biol is available at www.ahajournals.org/journal/atvb

Arterioscler Thromb Vasc Biol. 2020;40:00–00. DOI: 10.1161/ATVBAHA.120.314459 July 2020   1


Doddapattar et al Role of SMC vs Endothelial Fn-EDA in Atherogenesis
Original Research - VB

Nonstandard Abbreviations and Acronyms Highlights


α-SMA alpha smooth muscle cell actin • The extracellular matrix of atherosclerotic arteries
BMT bone marrow transplantation contains abundant deposits of Fn-EDA (fibronectin
EC endothelial cell containing extra domain A), suggesting a functional
role in the pathophysiology of atherosclerosis.
ECM extracellular matrix
• Endothelial cell-derived Fn-EDA, but not smooth
EDB extra domain B muscle cell-derived Fn-EDA, contributes to early
Fn-EDA fibronectin containing extra domain A atherosclerosis in mice.
IFN interferon • Smooth muscle cell-derived Fn-EDA, but not endo-
IL interleukin thelial cell-derived Fn-EDA, is the primary determi-
nant that contributes to late atherosclerosis.
LDL-R low-density lipoprotein receptor
MCP-1 monocyte chemoattractant protein 1
mTOR mammalian target of rapamycin EC-derived Fn-EDA contributes to early atherosclerosis,
NF-κB nuclear factor-κB whereas SMC-derived Fn-EDA contributes to advanced
PCR polymerase chain reaction atherosclerosis.
SMC smooth muscle cell
TGF-β transforming growth factor β
TLR-4 toll-like receptor 4 MATERIALS AND METHODS
TNF-α tumor necrosis factor α The data that support the findings of this study are available
from the corresponding author upon reasonable request. For
VCAM-1 vascular cell adhesion molecule 1
details of reagents and murine strains, please see the Major
Resources Table in the Data Supplement.
EDA and EDB are highly conserved (>90%) in the ver-
tebrates, with either total inclusion or exclusion, includ- Mice
ing human, mouse, rat, chicken, and zebrafish.4 Although Six-weeks-old male and female Fn-EDAfl/flApoe−/− mice on
the predominant form of Fn in plasma of healthy humans the C57BL/6J background were used in the present study.11
Fn-EDAfl/flApoe−/− mice were crossed with the Apoe−/− Tie2Cre+
Downloaded from http://ahajournals.org by on May 31, 2020

contains negligible amounts of EDA or EDB, inclusion of


these alternatively spliced domains has been observed in mice to generate Fn-EDAfl/flTie2Cre+Apoe−/− (Fn-EDAEC-KO, EC
animal models of vascular injuries,5,6 and patients at risk KO of Fn-EDA) as described.16 SM22αCre+ mice (004746, Tg
[Tagln-cre]1 Her/J; The Jackson Laboratory) were crossed with
for diabetes mellitus,7 and atherosclerosis.8,9
Apoe−/− mice to generate SM22αCre+Apoe−/− mice. Fn-EDAfl/
Several studies have demonstrated that global dele- fl
Apoe−/− mice were crossed to SM22αCre+Apoe−/− mice to
tion of Fn-EDA in apolipoprotein E-deficient (Apoe−/−) generate Fn-EDAfl/flSM22αCre+Apoe−/− mice (Fn-EDASMC-KO,
or LDL-R (low-density lipoprotein receptor)-deficient smooth muscle KO of Fn-EDA) as described.15 Mice were gen-
mice reduces atherosclerosis suggesting that Fn-EDA otyped by polymerase chain reaction (PCR) according to proto-
is proatherogenic.10–12 Fn-EDB accumulation has also cols from The Jackson Laboratory and as described before.15–17
been found in human atherosclerotic plaques13; however, The University of Iowa Animal Care and Use Committee
whether it is proatherogenic remains unclear. The Fn approved all protocols.
present in the atherosclerotic lesions could arise from
several sources, including the leak of plasma Fn (pFn) Mice Diet Feeding and Preparation of Tissues
from the circulation, Fn-derived from ECs, and Fn-derived Both male and female mice (Fn-EDAfl/flTie2Cre+Apoe−/−,
from SMCs. Studies using human aortic ECs showed Fn-EDAfl/fl Apoe−/−, EDAfl/flSM22αCre+Apoe−/−) were fed a high-
that EC-derived Fn regulates integrin α5β1-mediated fat Western diet (20% milk fat and 0.2% cholesterol, Harlan
adhesion formation, Fn fibrillogenesis, and inflammation Teklad) beginning at 6 weeks of age until they were euthanized
in response to oxidized LDL, suggesting EC-derived Fn at 5 months of age (ie, 14 weeks on high-fat Western diet to
may contribute to early atherogenesis.14 Other studies induce late atherosclerosis). Mice were fed a normal labora-
showed that Fn-EDA is expressed in the vicinity of SMCs tory diet (NIH-31 modified mouse diet: 7913) before high-fat
and was a characteristic feature of synthetic SMCs that Western diet. All mice were fed 4 weeks of high-fat Western
diet beginning at 6 weeks of age to study early atherosclerosis.
are known to contribute to different stages of athero-
For plasma isolation, blood samples were collected in heparin-
sclerosis.6,15 Despite these observations, there are no
ized tubes by retro-orbital plexus puncture after overnight fast-
studies that provide conclusive evidence on the role of ing. Before tissue isolation, mice were anesthetized with 100
EC-derived versus SMC-derived Fn-EDA in atherogen- mg/kg ketamine/10 mg/kg xylazine and perfused via the left
esis. Also unclear is whether Fn-EDA derived from these ventricle with 10 mL PBS, followed by 10 mL of 4% parafor-
cell types contributes to the early or late atherogenesis. maldehyde under physiological pressure. After perfusion, aor-
Herein, using mutant mice, we provide evidence that tae were isolated, dehydrated for 5 minutes in 60% isopropyl

2   July 2020 Arterioscler Thromb Vasc Biol. 2020;40:00–00. DOI: 10.1161/ATVBAHA.120.314459
Doddapattar et al Role of SMC vs Endothelial Fn-EDA in Atherogenesis

alcohol and stained with Oil Red O. Hearts containing aortic µg/mL, Invitrogen, no. A11081), goat anti-rat 488 (4 µg/

Original Research - VB
roots were dissected and fixed overnight in 4% paraformalde- mL, Invitrogen, no. A-11006), goat anti-rabbit IgG 546 (4
hyde before embedding in paraffin. µg/mL, Invitrogen no. A11035), and goat anti-mouse 488
(Catalog no. A32723) were used. Nuclei were stained using
the Antifade mounting medium with DAPI (Vector laborato-
Extent and Composition of Atherosclerotic ries no. H:1200). For the chromogenic method, slides were
Lesions incubated with streptavidin-HRP for 40 minutes room tem-
The whole aortae were isolated and stained with Oil Red O, perature and washed thrice with PBS for 5 minutes and incu-
and the en face lesion area was measured by morphometry bated with DAB substrate (Vector laboratories no. SK400)
using NIH ImageJ software to measure atherosclerosis lesion for <2 minutes until color develops. Hematoxylin counter-
progression. Lesion areas in the aortic sinus were quantified by stained slides were dehydrated, mounted using permount,
using 5 µm thick serial cross-sections that were cut through and examined using an Olympus fluorescent microscope
the aorta beginning at the origin of the aortic valve leaflets and (BX51). Mean fluorescence intensity was quantified using
stained by VerHoeffs/Van Gieson method. The cross-sectional the ImageJ software as previously described (Jensen, 2013).
lesion area from each mouse was calculated by taking the Protein colocalization was analyzed using the Pearson colo-
mean value of 4 sections (each 80 µm apart, beginning at the calization coefficients (http://rsb.info.nih.gov/ij/plugins/
aortic valve leaflets and spanning 320 µm) as described previ- track/jacop.html). Colocalization was considered positive for
ously.18 For quantification, NIH ImageJ software was used. values ranging from 0.5 to 1. Measurements were obtained
from 2 different fields per murine section, 4 to 6 different
Bone Marrow Transplantation fields for human samples. For negative control, incubation
without primary antibodies and with isotype-matched immu-
Bone marrow transplantation (BMT) of either Fn-EDAfl/
noglobulins was used. A mean for each mouse was calcu-
Tie2Cre+ Apoe−/− or Fn-EDAfl/flApoe−/− mice were performed
fl
lated using the mean value of 4 sections (each 80 µm apart,
on 6-weeks-old mice. Mice were irradiated with 2 doses of
beginning at the aortic valve leaflets and spanning 320 µm).
6.5-Gy at an interval of 4 hours between the first and sec-
ond irradiations before BMT as described.18 Under sterile
conditions, bone marrow cells (1×107) were extracted from Determination of Plasma Total Cholesterol and
excised femurs and tibias of euthanized mice, suspended in Lipid Levels
sterile PBS and injected into the retro-orbital venous plexus Overnight fasted blood from each mouse was collected in hep-
of lethally irradiated recipient mice. Post-transplantation, mice arinized tubes by retro-orbital venous plexus puncture. Plasma
were maintained in sterile cages and fed autoclaved food and was separated and analyzed for total cholesterol and triglyceride
water ad libitum. Following BMT experiments were performed:
Downloaded from http://ahajournals.org by on May 31, 2020

(both from DiaSys no. 113009911923, no. 157109911923)


(1) irradiated Fn-EDAfl/flApoe−/− mice reconstituted with BM levels by using enzymatic colorimetric assays according to the
from Fn-EDAfl/fl Apoe−/−, (2) irradiated Fn-EDAfl/flApoe−/− mice manufacturer’s protocol.
reconstituted with BM from Fn-EDAfl/flTie2Cre+Apoe−/−, (3)
irradiated Fn-EDAfl/flTie2Cre+Apoe−/− mice reconstituted with
BM from Fn-EDAfl/fl Apoe−/− donors. Successful BMT was con- Picrosirius Red Staining for Collagen
firmed after 4 weeks by PCR to check the presence of the Quantification of interstitial collagen content of the aortic
genomic DNA of the respective donor mice in peripheral blood sinus was performed as described.11 Briefly, serial cross-
cells. Total blood cell counts were obtained using an automated sections of 5 µm were stained with Weigert’s hematoxylin
veterinary hematology analyzer (ADVIA) to ascertain that BMT and washed in running tap water for 10 minutes followed by
did not affect the number of BM-derived blood cells. incubation for 4 hours in a freshly prepared 0.1% solution
of Sirius Red F3B (Sigma-Aldrich, no. 365548) in saturated
aqueous picric acid (Sigma-Aldrich, no. P-6744). Sections
Immunohistochemistry of Murine Samples were rinsed twice in acidified (0.01 N HCl) distilled water,
Tissue preparation and histochemical staining were per- dehydrated, and mounted in Permount (Fisher Scientific, no.
formed as described.18 Briefly, antigen retrieval was per- SP15-500). Polarization microscopy was used to analyze
formed before immunohistochemical staining. Slides were picrosirius red staining. Total collagen, thick mature orange-
rehydrated, and sections were blocked with 5% serum in red fibers, and thin immature green fibers were quantified
Tris-buffered saline at room temperature from the species in as described19 using NIH ImageJ software with a defined
which the secondary antibody was raised. For chromogenic threshold (minimum 100 and maximum 200). A mean for
detection, endogenous peroxidase activity was quenched each mouse was calculated using the mean value of 4 sec-
with 0.1% hydrogen peroxide (Fisher Scientific, no. H325) in tions (each 80 µm apart, beginning at the aortic valve leaf-
methanol for 15 minutes. Sections were stained for Lys6G.2 lets and spanning 320 µm).
(clone, 1A8, Biorad no. MCA6077GA), Mac3 (1.5 µg/
mL BD Pharmingen no. 550292) and smooth muscle cell
actin (α-SMA, Sigma, no. A5228), fibronectin EDA (Clone ELISA Assay for TNF-α and IL-1β
Fn-3E2, Sigma, no. F6140), CD31 (ab28364), VCAM-1 Overnight fasted plasma samples were used for determination
(no. SC-13160). After overnight incubation at 4°C, slides of TNF (tumor necrosis factor)-α and IL (interleukin)-1β with
were washed thrice with PBS for 5 minutes and incubated commercially available mouse ELISA kits (TNF-α, no. MTA00B;
with biotinylated secondary antibody for 1 hour at room IL-1β, no. MLB00C; R&D Systems) according to the kit manu-
temperature. For immunofluorescence, goat anti-rat 546 (4 facturer’s instructions.

Arterioscler Thromb Vasc Biol. 2020;40:00–00. DOI: 10.1161/ATVBAHA.120.314459 July 2020   3


Doddapattar et al Role of SMC vs Endothelial Fn-EDA in Atherogenesis

Statistical Analysis (Figure V in the Data Supplement), and α-SMA (Fig-


Original Research - VB

Results are reported as the mean±SEM. For statistical analysis, ure 2D) within aortic lesions were comparable between
the Prism Graph software package was used. Shapiro-Wilk test Fn-EDAEC-KO and control mice. Inflammatory cytokines
was used to check normality, and the Bartlett test was used TNF-α and IL-1β in plasma (Figure VI in the Data Supple-
to check equal variance. The statistical significance of the dif- ment), total cholesterol, and triglyceride levels (Table I in
ference between means was assessed using the unpaired the Data Supplement) were comparable among groups.
Student t test (for normally distributed data) or Mann Whitney Together, these results suggest that EC-derived Fn-EDA
test (for not normally distributed data) and 1-way ANOVA fol- contributes to early atherosclerosis, but not advanced
lowed by Tukey multiple comparisons test. P<0.05 was consid- atherosclerosis, most likely by promoting neutrophil and
ered significant. macrophage recruitment via VCAM-1.

RESULTS SMC-Derived Fn-EDA Contributes to Late


EC-Derived Fn-EDA Contributes to Early Atherosclerosis But Not Early Atherosclerosis
Atherosclerosis But Not Late Atherosclerosis We generated Fn-EDAfl/flSM22αCre+Apoe−/− to deter-
mine the role of SMC-derived Fn-EDA in atherosclerosis.
We generated Fn-EDAfl/flTie2Cre+Apoe−/− mice to deter-
Genomic PCR confirmed the presence of the SM22αCre
mine the role of EC-derived Fn-EDA in atherosclerosis.
gene in Fn-EDAfl/flSM22αCre+Apoe−/− (Figure VIIA in the
Because Tie2 is also expressed by hematopoietic cells
Data Supplement). RT-PCR confirmed the absence of
in addition to ECs, Fn-EDAfl/flTie2Cre+Apoe−/− mice were
Fn-EDA mRNA in SMCs but not in other cells of Fn-
transplanted with bone marrow from Fn-EDAfl/flApoe−/−
EDAfl/flSM22αCre+Apoe−/− mutant mice (Figure VIIB
mice, which constitutively express Fn-EDA in all cells
in the Data Supplement). Male and female mice were
and tissues. This strategy resulted in mutant mice that first fed a normal laboratory diet for 6 weeks, and then
are deficient for Fn-EDA in ECs (Fn-EDAEC-KO). Controls a high-fat Western diet for 4 or 14 weeks. In the early
were irradiated littermates Fn-EDAfl/flApoe−/− mice that atherosclerosis model, irrespective of sex, we observed
received bone marrow from Fn-EDAfl/flApoe−/− mice or that atherosclerosis lesion progression in the aorta and
Fn-EDAfl/flTie2Cre+Apoe−/− mice. Male and female mice aortic sinus was comparable between Fn-EDASMC-KO and
were examined separately to determine sex-based dif- controls (Figure 3A and 3B and Figure VIIIA and VIIIB
ferences. Mice were fed a normal laboratory diet for
Downloaded from http://ahajournals.org by on May 31, 2020

in the Data Supplement). Lesion neutrophil (Figure 3C),


10 weeks until the complete reconstitution of newly macrophage influx (Figure IX in the Data Supplement),
transplanted bone marrow and then a high-fat Western and VCAM-1 expression (Figure 3D) were comparable
diet for 4 or 14 weeks. Genomic PCR confirmed the among groups. In late atherosclerosis model, irrespec-
presence of the Tie2Cre gene in Fn-EDAfl/flTie2Cre+A tive of sex, we found that the mean lesion areas in the
poe−/− mice (Figure IA in the Data Supplement). RT-PCR aortae and aortic sinus were significantly smaller in Fn-
confirmed the absence of Fn-EDA mRNA in the ECs but EDASMC-KO mice when compared with controls (P<0.05,
not in other cells of Fn-EDAfl/flTie2Cre+Apoe−/− mutant Figure 4A and 4B and Figure VIIIC and VIIID in the
mice (Figure IB in the Data Supplement). Data Supplement). The decrease in lesion size in the
Next, we compared the extent of atherosclerosis in Fn-EDASMC-KO mice was associated with reduced mac-
whole aortae (by staining with Oil Red O and quantify- rophage infiltration (Figure 4C) with unaltered collagen
ing en face lesion area) and aortic sinus (by staining (Figure X in the Data Supplement and α-SMA content
with VerHoeffs/Van Gieson and quantifying the cross- (Figure 4D). Inflammatory cytokines TNF-α and IL-1β,
sectional area). In the early atherosclerosis model, irre- total cholesterol, and triglycerides levels in the plasma
spective of sex, Fn-EDAEC-KO mice exhibited reduced and leukocyte counts were comparable among groups
atherosclerosis lesion progression in aortae and aortic (Figure XI and Tables II and III in the Data Supplement).
sinus when compared with controls (P<0.05, Figure 1A Next, using immunostaining, we determined the per-
and 1B, and Figure IIA and IIB in the Data Supplement). centage of Fn-EDA associated with SMCs (α-SMA-
The decrease in lesion size in the Fn-EDAEC-KO mice positive) and ECs (CD31-positive) in autopsy samples
was associated with reduced neutrophil (Figure 1C) and from patients with coronary artery disease. We found a
macrophage content (Figure III in the Data Supplement) marked expression of Fn-EDA within advanced lesions
and decreased VCAM-1 expression (Figure 1D) within from diseased coronary arteries, whereas staining was
lesions suggesting reduced endothelial activation. Rep- virtually absent in arteries from healthy controls (Figure
resentative negative controls are shown (Figure IV in the XII in the Data Supplement). Double-label immunos-
Data Supplement). In late atherosclerosis model, lesion taining revealed a 3-fold increase in Fn-EDA staining
area in the aortae and aortic sinus (Figure 2A and 2B), associated with SMCs (Fn-EDA/α-SMA-positive) when
macrophage content (Figure 2C), total collagen content compared with ECs (Fn-EDA/CD31-positive; Figure XII
including both thick mature and thin immature fibers in the Data Supplement). Together, these results suggest

4   July 2020 Arterioscler Thromb Vasc Biol. 2020;40:00–00. DOI: 10.1161/ATVBAHA.120.314459
Doddapattar et al Role of SMC vs Endothelial Fn-EDA in Atherogenesis

Original Research - VB
Downloaded from http://ahajournals.org by on May 31, 2020

Figure 1. Fn-EDAEC-KO mice exhibit reduced early atherosclerosis.


All the mice were females and fed a high-fat Western diet for 4 wk. A, Representative photomicrographs and quantification of Oil red O-stained
en face lesion area in the whole aortae. B, Representative photomicrographs of VerHoeffs/Van Geison-staining and quantification of cross-
sectional lesion area in aortic sinuses. C, Representative photomicrographs and quantification of neutrophil-stained (Lys6G.2-positive cells)
in aortic sinuses. Scale bar=100 μm. D, Representative photomicrographs and quantification of VCAM-1 (vascular cell adhesion molecule
1-stained positive cells) area in aortic sinuses. Scale bar=100 μm. Values are expressed as mean±SEM. Each dot represents a single mouse
(n=10–13/group). Statistical analysis: 1-way ANOVA followed by Tukey multiple comparisons test.

that SMC-derived Fn-EDA contributes to advanced ath- previous evidence suggesting that Fn-EDA is proath-
erosclerosis, but not early atherosclerosis. erogenic, the role of EC-derived versus SMC-derived
Fn-EDA in atherogenesis remains unclear. Using Fn-
EDAEC-KO and Fn-EDASMC-KO mice, we provide evidence
DISCUSSION that EC-derived Fn-EDA contributes to early atheroscle-
Understanding the mechanisms that contribute to the rosis, whereas SMC-derived Fn-EDA contributes to late
initiation and progression of atherosclerosis is vital in atherosclerosis, suggesting cell and stage-specific role
developing novel strategies to limit the lesion progres- of Fn-EDA during atherosclerosis.
sion before reaching the clinical manifestations. Abun- Previous studies have shown that Fn-EDA was
dant deposits of Fn-EDA have been found in the ECM of associated with ECs during early lesion progression
both human and murine atherosclerotic plaques. Despite but absent from adjacent uninvolved endothelium.12 A

Arterioscler Thromb Vasc Biol. 2020;40:00–00. DOI: 10.1161/ATVBAHA.120.314459 July 2020   5


Doddapattar et al Role of SMC vs Endothelial Fn-EDA in Atherogenesis
Original Research - VB
Downloaded from http://ahajournals.org by on May 31, 2020

Figure 2. Endothelial-derived Fn-EDA (fibronectin containing extra domain A) does not contribute to late atherosclerosis.
All the mice were females and fed a high-fat Western diet for 14 wk. A, Representative photomicrographs and quantification of Oil red O-
stained en face lesion area in the whole aortae. B, Representative photomicrographs of VerHoeffs/Van Geison-staining and quantification
of cross-sectional lesion area in aortic sinuses. Scale bar=500 μm. C, Representative photomicrographs and quantification of macrophage
(mac3-positive cells) in aortic sinuses. Scale bar=500 μm. D, Representative photomicrographs and quantification of smooth muscle cell-
stained (⍺-SMA-positive) area in aortic sinuses. Scale bar=100 μm Values are expressed as mean±SEM. Each dot represents a single mouse
(n=10–13/group). Statistical analysis: 1-way ANOVA followed by Tukey multiple comparisons test.

minimal Fn-EDA staining was also found around macro- in plasma total cholesterol and triglyceride levels. Despite
phages or early foam cells during early lesion progres- the fact that the atherogenic process initiates with the
sion.12 Another study found that cFn deposition by α5β1 deposition of LDL, several studies suggest that chronic
integrin promotes early atherosclerosis.14 Although these inflammation contributes to atherosclerotic lesion pro-
studies suggest that EC-derived Fn-EDA may contrib- gression, and lesion size does not always correlate with
ute to early atherosclerosis,12 there was no definitive evi- an increase in plasma cholesterol and LDL levels.20–22 It
dence that has examined how the lack of Fn-EDA from is known that ECM within the lesions can trap and retain
different specific cell type affects early atherogenesis. lipids.23 There could be several possibilities by which
Using Fn-EDAEC-KO and Fn-EDASMC-KO mice, we found EC-derived Fn-EDA may contribute to early atheroscle-
that Fn-EDA derived from ECs, but not from SMCs, con- rosis. First, by promoting early atherogenic inflammation
tributes to early atherosclerosis, independent of changes via integrin α5β1 as suggested previously.14 Second,

6   July 2020 Arterioscler Thromb Vasc Biol. 2020;40:00–00. DOI: 10.1161/ATVBAHA.120.314459
Doddapattar et al Role of SMC vs Endothelial Fn-EDA in Atherogenesis

Original Research - VB
Downloaded from http://ahajournals.org by on May 31, 2020

Figure 3. Smooth muscle cell-derived Fn-EDA (fibronectin containing extra domain A) does not contribute to early
atherosclerosis.
All the mice were females and fed a high-fat Western diet for 4 wk. A, Representative photomicrographs and quantification of Oil red O-stained
en face lesion area in the whole aortae. B, Representative photomicrographs of VerHoeffs/Van Geison-staining and quantification of cross-
sectional lesion area in aortic sinuses. Scale bar=500 μm. Lower: C, Representative photomicrographs and quantification of neutrophil-stained
(Lys6G.2-positive cells) in aortic sinuses. Scale bar=100 μm. D, Representative photomicrographs and quantification of VCAM-1 (vascular
cell adhesion molecule 1-stained positive cells) in aortic sinuses. Scale bar=100 μm. Each dot represents a single mouse (n=8–14/group).
Statistical analysis: unpaired Student t test.

by enhancing oxidized LDL-mediated NF-κB (nuclear neutrophils and known to contribute to adhesion, and
factor-κB) p65 activation and VCAM-1 expression.14,24 transmigration.25 Neutrophil depletion has been shown to
In agreement with these studies, we observed signifi- improve early atherosclerosis in mice.26 Studies suggest
cantly decreased VCAM-1 expression in Fn-EDAEC-KO that neutrophils contribute to early atherosclerosis via
mice. Third, by potentiating neutrophil interactions with multiple mechanisms including neutrophil extracellular
the vessel wall. We found that reduced lesion size in Fn- trap formation and interacting with monocytes to prime
EDAEC-KO mice was associated with decreased neutrophil them for inflammatory response.27,28 Fourth, by promoting
and macrophage content within lesions. The EDA of Fn NF-κB p65-mediated inflammation in both neutrophils
contains additional binding sites for integrin α9β1 and and monocytes/macrophages via innate immune recep-
α4β1. Integrin α9β1 is highly expressed on activated tor TLR-4 (toll-like receptor 4). Fn-EDA is an endogenous

Arterioscler Thromb Vasc Biol. 2020;40:00–00. DOI: 10.1161/ATVBAHA.120.314459 July 2020   7


Doddapattar et al Role of SMC vs Endothelial Fn-EDA in Atherogenesis
Original Research - VB
Downloaded from http://ahajournals.org by on May 31, 2020

Figure 4. Fn-EDASMC-KO mice exhibit reduced late atherosclerosis.


All the mice were females and fed a high-fat Western diet for 14 wk. A, Representative photomicrographs and quantification of Oil red
O-stained en face lesion area in the whole aortae. B, Representative photomicrographs of VerHoeffs/Van Geison-staining and quantification of
cross-sectional lesion area in aortic sinuses. Scale bar=500 μm. C, Representative photomicrographs and quantification of macrophage (mac3-
positive cells) area in aortic sinuses. Scale bar=500 μm. D, Representative photomicrographs and quantification of smooth muscle cell-stained
(⍺-SMA-positive) area in aortic sinuses. Scale bar=100 μm. Each dot represents a single mouse (n=8–14/group). Statistical analysis: unpaired
Student t test.

ligand for TLR-4. Previously, we have shown that cFn lesions.12 Consistent with these studies, we provide evi-
potentiates NF-κB p65-mediated inflammation in bone- dence that SMC-derived Fn-EDA, but not EC-derived
marrow-derived neutrophils and macrophages via TLR- Fn-EDA, contributes to late atherosclerosis by recruit-
4.11 In line with our observations, a recent study found ment of monocytes/macrophages without altering SMCs
that EC-derived Fn-EDA was increased by disturbed flow and collagen content. It is possible that the leak of circu-
and interacted with TLR-4 to promote inflammation.29 lating Fn-EDA into the vessel wall over time may mask
Herein, we found that in human diseased coronary the effectiveness of Fn-EDA deletion in Fn-EDAEC-KO
lesions, a majority of Fn-EDA staining was colocalized mice on late atherogenesis. Such a possibility is mini-
with SMCs, whereas Fn-EDA staining colocalized with mal because lack of Fn-EDA, specifically, in the plasma
ECs was less when compared with SMCs. Our results does not affect atherogenesis, suggesting that the Fn-
are in agreement with the previous study, where less Fn- EDA produced by cells present in atherosclerotic plaque
EDA staining was associated with ECs in the advanced contributes to atherogenesis.10 There could be several

8   July 2020 Arterioscler Thromb Vasc Biol. 2020;40:00–00. DOI: 10.1161/ATVBAHA.120.314459
Doddapattar et al Role of SMC vs Endothelial Fn-EDA in Atherogenesis

possibilities by which SMC-derived Fn-EDA may con- 6. Glukhova MA, Frid MG, Shekhonin BV, Vasilevskaya TD, Grunwald J,

Original Research - VB
Saginati M, Koteliansky VE. Expression of extra domain A fibronectin
tribute to late atherosclerosis. First, by modulating SMCs sequence in vascular smooth muscle cells is phenotype dependent. J Cell
from contractile to synthetic phenotype via integrins.6,15 Biol. 1989;109:357–366. doi: 10.1083/jcb.109.1.357
SMC-derived Fn-EDA contributes to synthetic pheno- 7. Kanters SD, Banga JD, Algra A, Frijns RC, Beutler JJ, Fijnheer R. Plasma
levels of cellular fibronectin in diabetes. Diabetes Care. 2001;24:323–327.
type via Akt1/mTOR (mammalian target of rapamycin) doi: 10.2337/diacare.24.2.323
signaling.15 In addition to ECM components, synthetic 8. Lemańska-Perek A, Krzyżanowska-Gołąb D, Pupek M, Klimeczek P,
SMC produces many cytokines such as PDGF, TGF Witkiewicz W, Kątnik-Prastowska I. Analysis of soluble molecular fibro-
nectin-fibrin complexes and EDA-fibronectin concentration in plasma of
(transforming growth factor)-β, IFN (interferon)-γ, and patients with atherosclerosis. Inflammation. 2016;39:1059–1068. doi:
MCP-1 (monocyte chemoattractant protein 1) that may 10.1007/s10753-016-0336-0
promote monocyte/macrophage recruitment.30 Second, 9. Fiechter M, Frey K, Fugmann T, Kaufmann PA, Neri D. Comparative in
vivo analysis of the atherosclerotic plaque targeting properties of eight
by potentiating TLR-4 signaling in monocytes/macro- human monoclonal antibodies. Atherosclerosis. 2011;214:325–330. doi:
phages. In addition to integrins including α5β1, α3β1, 10.1016/j.atherosclerosis.2010.11.027
αvβ1, αvβ3, Fn-EDA is an endogenous ligand for TLR-4. 10. Pulakazhi Venu VK, Uboldi P, Dhyani A, Patrini A, Baetta R, Ferri N,
Corsini A, Muro AF, Catapano AL, Norata GD. Fibronectin extra domain
Previously, using double-deficient mice, we have shown A stabilises atherosclerotic plaques in apolipoprotein E and in LDL-
that TLR-4 partially contributes to Fn-EDA-mediated receptor-deficient mice. Thromb Haemost. 2015;114:186–197. doi:
advanced atherosclerosis, most likely by potentiating 10.1160/TH14-09-0790
11. Doddapattar P, Gandhi C, Prakash P, Dhanesha N, Grumbach IM,
NF-κB p65-mediated inflammation.11 Dailey ME, Lentz SR, Chauhan AK. Fibronectin splicing variants con-
In conclusion, our results provide novel insights taining extra domain A promote atherosclerosis in mice through toll-like
into the role of different pools of Fn-EDA at different receptor 4. Arterioscler Thromb Vasc Biol. 2015;35:2391–2400. doi:
10.1161/ATVBAHA.115.306474
stages of atherogenesis. This study further supports 12. Tan MH, Sun Z, Opitz SL, Schmidt TE, Peters JH, George EL. Deletion of
the notion that therapeutically targeting Fn-EDA may the alternatively spliced fibronectin EIIIA domain in mice reduces athero-
have the potential for the prevention and treatment sclerosis. Blood. 2004;104:11–18. doi: 10.1182/blood-2003-09-3363
13. Yu M, Ortega CA, Si K, Molinaro R, Schoen FJ, Leitao RFC, Xu X,
of atherosclerosis in patients at high risk for coronary Mahmoudi M, Ahn S, Liu J, et al. Nanoparticles targeting extra domain B of
heart disease. fibronectin-specific to the atherosclerotic lesion types III, IV, and V-enhance
plaque detection and cargo delivery. Theranostics. 2018;8:6008–6024. doi:
10.7150/thno.24365
ARTICLE INFORMATION 14. Al-Yafeai Z, Yurdagul A Jr, Peretik JM, Alfaidi M, Murphy PA, Orr AW.
Endothelial FN (Fibronectin) deposition by α5β1 integrins drives athero-
Received September 20, 2019; accepted May 11, 2020. genic inflammation. Arterioscler Thromb Vasc Biol. 2018;38:2601–2614.
doi: 10.1161/ATVBAHA.118.311705
Downloaded from http://ahajournals.org by on May 31, 2020

Affiliation 15. Jain M, Dhanesha N, Doddapattar P, Chorawala MR, Nayak MK,


From the Division of Hematology/Oncology, Department of Internal Medicine, Cornelissen A, Guo L, Finn AV, Lentz SR, Chauhan AK. Smooth mus-
University of Iowa, Iowa City. cle cell-specific fibronectin-EDA mediates phenotypic switching
and neointimal hyperplasia. J Clin Invest. 2020;130:295–314. doi:
Sources of Funding 10.1172/JCI124708
The AKC lab is supported by grants from the National Institutes of Health grants 16. Dhanesha N, Chorawala MR, Jain M, Bhalla A, Thedens D, Nayak M,
(R35HL139926, R01NS109910, and U01NS113388) and by Established In- Doddapattar P, Chauhan AK. Fn-EDA (Fibronectin Containing Extra Domain
vestigator Award 18EIA33900009 from American Heart Association. P. Dodda- A) in the plasma, but not endothelial cells, exacerbates stroke outcome
pattar is supported by a postdoctoral grant from the American Heart Association by promoting thrombo-inflammation. Stroke. 2019;50:1201–1209. doi:
(18POST33960179), and N. Dhanesha is supported by the American Society of 10.1161/STROKEAHA.118.023697
Hematology Scholar award. 17. Muro AF, Chauhan AK, Gajovic S, Iaconcig A, Porro F, Stanta G, Baralle FE.
Regulated splicing of the fibronectin EDA exon is essential for proper skin
Disclosures wound healing and normal lifespan. J Cell Biol. 2003;162:149–160. doi:
None. 10.1083/jcb.200212079
18. Doddapattar P, Dhanesha N, Chorawala MR, Tinsman C, Jain M, Nayak MK,
Staber JM, Chauhan AK. Endothelial cell-derived von willebrand factor, but
not platelet-derived, promotes atherosclerosis in apolipoprotein E-defi-
REFERENCES cient mice. Arterioscler Thromb Vasc Biol. 2018;38:520–528. doi:
1. Raines EW. The extracellular matrix can regulate vascular cell migration, 10.1161/ATVBAHA.117.309918
proliferation, and survival: relationships to vascular disease. Int J Exp Pathol. 19. Ovchinnikova O, Robertson AK, Wågsäter D, Folco EJ, Hyry M, Myllyharju J,
2000;81:173–182. doi: 10.1046/j.1365-2613.2000.00155.x Eriksson P, Libby P, Hansson GK. T-cell activation leads to reduced col-
2. Shekhonin BV, Domogatsky SP, Idelson GL, Koteliansky VE, Rukosuev VS. lagen maturation in atherosclerotic plaques of apoe(-/-) mice. Am J Pathol.
Relative distribution of fibronectin and type I, III, IV, V collagens in normal 2009;174:693–700. doi: 10.2353/ajpath.2009.080561
and atherosclerotic intima of human arteries. Atherosclerosis. 1987;67:9– 20. Gandhi C, Khan MM, Lentz SR, Chauhan AK. ADAMTS13 reduces vascular
16. doi: 10.1016/0021-9150(87)90259-0 inflammation and the development of early atherosclerosis in mice. Blood.
3. Pedretti M, Rancic Z, Soltermann A, Herzog BA, Schliemann C, 2012;119:2385–2391. doi: 10.1182/blood-2011-09-376202
Lachat M, Neri D, Kaufmann PA. Comparative immunohistochemical stain- 21. Boring L, Gosling J, Cleary M, Charo IF. Decreased lesion formation in
ing of atherosclerotic plaques using F16, F8 and L19: three clinical- CCR2-/- mice reveals a role for chemokines in the initiation of atheroscle-
grade fully human antibodies. Atherosclerosis. 2010;208:382–389. doi: rosis. Nature. 1998;394:894–897. doi: 10.1038/29788
10.1016/j.atherosclerosis.2009.07.043 22. Sharrett AR. Serum cholesterol levels and atherosclerosis. Coron Artery Dis.
4. Hynes RO. The evolution of metazoan extracellular matrix. J Cell Biol. 1993;4:867–870. doi: 10.1097/00019501-199310000-00005
2012;196:671–679. doi: 10.1083/jcb.201109041 23. Chait A, Wight TN. Interaction of native and modified low-density lipopro-
5. Dubin D, Peters JH, Brown LF, Logan B, Kent KC, Berse B, teins with extracellular matrix. Curr Opin Lipidol. 2000;11:457–463. doi:
Berven S, Cercek B, Sharifi BG, Pratt RE. Balloon catheterization induced 10.1097/00041433-200010000-00003
arterial expression of embryonic fibronectins. Arterioscler Thromb Vasc Biol. 24. Yurdagul A Jr, Sulzmaier FJ, Chen XL, Pattillo CB, Schlaepfer DD, Orr AW.
1995;15:1958–1967. doi: 10.1161/01.atv.15.11.1958 Oxidized LDL induces FAK-dependent RSK signaling to drive NF-κB

Arterioscler Thromb Vasc Biol. 2020;40:00–00. DOI: 10.1161/ATVBAHA.120.314459 July 2020   9


Doddapattar et al Role of SMC vs Endothelial Fn-EDA in Atherogenesis

activation and VCAM-1 expression. J Cell Sci. 2016;129:1580–1591. doi: 28. Warnatsch A, Ioannou M, Wang Q, Papayannopoulos V. Inflamma-
Original Research - VB

10.1242/jcs.182097 tion. Neutrophil extracellular traps license macrophages for cyto-


25. Mambole A, Bigot S, Baruch D, Lesavre P, Halbwachs-Mecarelli L. Human kine production in atherosclerosis. Science. 2015;349:316–320. doi:
neutrophil integrin alpha9beta1: up-regulation by cell activation and synergy 10.1126/science.aaa8064
with beta2 integrins during adhesion to endothelium under flow. J Leukoc 29. Qu D, Wang L, Huo M, Song W, Lau CW, Xu J, Xu A, Yao X, Chiu JJ,
Biol. 2010;88:321–327. doi: 10.1189/jlb.1009704 Tian XY, et al. Focal tlr4 activation mediates disturbed flow-induced endo-
26. Drechsler M, Megens RT, van Zandvoort M, Weber C, Soehnlein O. Hyper- thelial inflammation. Cardiovasc Res. 2019;
lipidemia-triggered neutrophilia promotes early atherosclerosis. Circulation. 30. Raines EW, Ferri N. Thematic review series: The immune system and
2010;122:1837–1845. doi: 10.1161/CIRCULATIONAHA.110.961714 atherogenesis. Cytokines affecting endothelial and smooth mus-
27. Soehnlein O. Multiple roles for neutrophils in atherosclerosis. Circ Res. cle cells in vascular disease. J Lipid Res. 2005;46:1081–1092. doi:
2012;110:875–888. doi: 10.1161/CIRCRESAHA.111.257535 10.1194/jlr.R500004-JLR200
Downloaded from http://ahajournals.org by on May 31, 2020

10   July 2020 Arterioscler Thromb Vasc Biol. 2020;40:00–00. DOI: 10.1161/ATVBAHA.120.314459

You might also like