scispace - formally typeset
Open AccessJournal ArticleDOI

The SGLT-2 Inhibitor Dapagliflozin Has a Therapeutic Effect on Atherosclerosis in Diabetic ApoE−/− Mice

Reads0
Chats0
TLDR
Dapagliflozin may be of therapeutic potential for diabetic atherosclerosis induced by high-fat diet, and these benefits may depend on the inhibitory effect on the secretion of IL-1β by macrophages via the ROS-NLRP3-caspase-1 pathway.
Abstract
Background. Our study aimed to observe the effect of sodium glucose cotransporter-2 (SGLT2) inhibitor dapagliflozin on diabetic atherosclerosis and investigate the subsequent mechanism. Methods. Aortic atherosclerosis was induced in streptozotocin induced diabetic ApoE−/− mice by feeding with high-fat diet, and dapagliflozin was administrated intragastrically for 12 weeks as treatment. Effects of dapagliflozin on indices of glucose and fat metabolism, IL-1β, IL-18, NLRP3 protein levels, and the reactive oxygen species (ROS) were measured. The atherosclerosis was evaluated by oil red O and hematoxylin-eosin staining. The effects of dapagliflozin on the IL-1β production in culturing primary macrophages of wild type and NLRP3−/− knockout mice were investigated for mechanism analyses. Results. Dapagliflozin treatment showed favorable effects on glucose and fat metabolism, partially reversed the formation of atherosclerosis, inhibited macrophage infiltration, and enhanced the stability of lesion. Also, reduced production of IL-1β, IL-18, NLRP3 protein, and mitochondrial ROS in the aortic tissues was detected with dapagliflozin treatment. In vitro, NLRP3 inflammasome was activated by hyperglucose and hyperlipid through ROS pathway. Conclusions. Dapagliflozin may be of therapeutic potential for diabetic atherosclerosis induced by high-fat diet, and these benefits may depend on the inhibitory effect on the secretion of IL-1β by macrophages via the ROS-NLRP3-caspase-1 pathway.

read more

Content maybe subject to copyright    Report

Research A rticle
The SGLT-2 Inhibitor Dapagliflozin Has a Therapeutic
Effect on Atherosclerosis in Diabetic ApoE
−/−
Mice
Weiling Leng,
1
Xinshou Ouyang,
2
Xiaotian Lei,
1
Mingxia Wu,
1
Liu Chen,
1
Qinan Wu,
1
Wuquan Deng,
1
and Ziwen Liang
1
1
Department of Endocrinology, e First Aliated Hospital of ird Military Medical University, Chongqing 400038, China
2
Department of Internal Medicine, Section of Digestive Diseases, Yale University of Medicine, New Haven, CT 06520, USA
Correspondence should be addressed to Ziwen Liang; ziwenliang@.com
Received August ; Revised  November ; Accepted December 
Academic Editor: Anshu Agrawal
Copyright ©  Weiling Leng et al. is is an open access article distributed under the Creative Commons Attribution License,
which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.
Background. Our study aimed to observe the eect of sodium glucose cotransporter- (SGLT) inhibitor dapagliozin on diabetic
atherosclerosis and investigate the subsequent mechanism. Methods. Aortic atherosclerosis was induced in streptozotocin induced
diabetic ApoE
−/−
mice by feeding with high-fat diet, and dapagliozin was administrated intragastrically for  weeks as treatment.
Eects of dapagliozin on indices of glucose and fat metabolism, IL-𝛽, IL-, NLRP protein levels, and the reactive oxygen species
(ROS) were measured. e atherosclerosis was evaluated by oil red O and hematoxylin-eosin staining. e eects of dapagliozin
on the IL-𝛽 production in culturing primary macrophages of wild type and NLRP
−/−
knockout mice were investigated for
mechanism analyses. Results. Dapagliozin treatment showed favorable eects on glucose and fat metabolism, partially reversed
the formation of atherosclerosis, inhibited macrophage inltration, and enhanced the stability of lesion. Also, reduced production
of IL-𝛽, IL-, NLRP protein, and mitochondrial ROS in the aortic tissues was detected with dapagliozin treatment. In vitro,
NLRP inammasome was activated by hyperglucose and hyperlipid through ROS pathway. Conclusions. Dapagliozin may be of
therapeutic potential for diabeti c atherosclerosis induced by high-fat diet, and these benets may depend on the inhibitory eect
on the secretion of IL-𝛽 by macrophages via the ROS-NLRP-caspase- pathway.
1. Introduction
Previous evidence from epidemiological studies has indicated
that cardiovascular diseases (CVDs) have become the most
serious complications of diabetes mellitus (DM), which is the
leading cause of mortality and morbidity for DM patients.
It has been well understood that pathophysiological changes
in patients with DM, mainly the toxicity of hig h blood
glucose to the endothelium and other cells of the vessels, may
lead to the pathogenesis of atherosclerosis and subsequent
CVDs. Although the key pathophysiological mechanisms
underlying the so-called diabetic atherosclerosis were not
fully determined, chronic inammatory response, and asso-
ciated lipid deposition, macrophage inltration and smooth
muscle cell proliferation in DM have been suggested to play
important roles [–].
Recently, interleukin-𝛽 (IL-𝛽)andinterleukin-
(IL-) have been recognized as two inammatory
cytokines which contribute to the development of diabetic
atherosclerosis [], and the underlying mechanisms
downstream of these cytokines have been e valuated. e
inherent immunity has been linked to the activation
of inammatory response of the body. e inherent
immunity recognizes damage-associated molecular patterns
(DAMPs) and pathogen-associated molecular patterns
(PAMPs) via pattern recognition receptors (PRRs) and
thus excites the inammatory reaction and starts the
defense mechanism []. NLRP, a NOD-like receptor
andalsoanintracellularPRR,hasbeensuggestedto
play a key role during the above pathophysiological
process. e NLRP can be activated by several dierent
exogenous and endogenous stimulation signals to form a
multiprotein complex known as NLRP inammasome.
Previous studies indicated that ligand-binding NLRP
can promote the formation of inammasome and activate
caspase-, eventually facilitating the maturation and
Hindawi Publishing Corporation
Mediators of Inflammation
Volume 2016, Article ID 6305735, 13 pages
http://dx.doi.org/10.1155/2016/6305735

Mediators of Inammation
secretion of pro-IL-𝛽 and pro-IL- [, ]. Re cent studies
have conrmed that NLRP inammasome is involved
in inammatory response during the pathogenesis of
atherosclerosis. Indeed, activation of NLRP inammasome
in macrophages has been involved in the cholesterol
crystals formation in the pathogenesis of atherosclerosis [].
Moreover, silence of NLRP gene has been found to delay
the progression of atherosclerosis in mice []. Although
IL-𝛽 [, ] and IL- [] have been suggested to be
proatherosclerosis, whether they functions via regulation of
NLRP inammasome remains to be determined.
Sodium glucose cotransporters- (SGLT-) inhibitors are
a new class of antidiabetic medications targeting against renal
glucose reabsorption. D apagliozin, as a SGLT- inhibitor,
was marketed in . e glucose lowering eect of
dapagliozin has been conrmed in many randomized con-
trolled clinical trials which showed distinguished lowering
eects of dapagliozin for glycosylated hemoglobin (HbAc),
fasting blood glucose, and postprandial blood glucose [, ].
Moreover, besides the antidiabetic eect, dapagliozin also
seemed to be cardioprotective, by lowering blood pressure
[], reducing body weight [], triglyceride and uric acid
[, ], and improving insulin resistance []. Recently,
a few studies indicated that SGLT- inhibitors may exert
their cardiometabolic benets via anti-inammatory eects
[, ]. However, the overall eects of dapagliozin on
atherosclerosis in DM and the potential benets involved,
for example, their eects on IL-𝛽 and IL- cytokines
and NLRP inammasome systems, have not been eval-
uated. Randomized controlled clinical t rials have proven
that dapagliozin can be used alone to treat the patients
with type diabetes mellitus and it has good eects in
decreasing glycosylated hemoglobin (HbAc), fasting blood
glucose, and postprandial blood glucose [, ]. Interestingly,
dapagliozin can improve cardiovascular diseas es in diabetes
mellitus by slightly lowering blood pressure [], reducing
body weight [], triglyceride and uric acid [, ], and
improving insulin resistance []. In addition, there are trials
showing that SGLT- inhibitors can reduce inammatory
markers in the serum and organs an d thus decrease the degree
of arteriosclerosis in the diabetic animal mo dels [, ].
However, the eects of SGLT - inhibitors on inammatory
markers, including NLRP inammasome and IL-𝛽,IL-
cytokines have not been completely claried yet. It is unclear
that whether SGLT- inhibitors can aect atherosclerosis
progression or not. Accumulated knowledge ab out eect of
dapagliozin on atherosclerosis and its underlying mecha-
nisms can bring us more information about its safety and
practicability in clinic.
e aims of the present study were to explore the eects
of dapagliozin on aortic atherosclerosis in diabetic versus
nondiabetic ApoE
−/−
mice and detect the possible underlying
mechanism, particularly their inuence on the ROS-NLRP-
caspase- pathway.
2. Methods
2.1. Mice. e spontaneous atherosclerotic lesions in
apolipoprotein E knockout (ApoE
−/−
)miceaerthe
hypercholesterolemia have been widely used as a mouse
model of atherosclerosis in previous studies [].
Consistently, we used this atherosclerotic mouse model
in our study. Briey,  male ApoE
−/−
mice ( weeks old,
Beijing HFK Bioscience Co., Ltd.) and the male CBL/J
mice ( weeks old, e Experimental Animal Center of
the ird Military Medical University) were all bred in the
animal rooms of the First Aliated Hospital of the ird
Military Medical University. e specic conditions were as
follows: a SPF environment, temperature (–
C), relative
humidity (%), and  h light/ h dark cycle. e animal
cages and drinking bottles were disinfected regularly, the
beddings were sterilized in an autoclave, and the animal
rooms were disinfected with ultraviolet lamps perio dica lly.
Aer one-week accommodation, the ApoE
−/−
mice were fed
with a high-fat diet (HFD, general diet .% + fat (oil)
% + custard powder % + cholesterol .%) to induce
atherosclerosis of the aorta, while the CBL/J mice were fed
with a general diet. e animal diets were purchased from the
Experimental Animal Center of the ird Aliated Hospital
of the ird Military Medical University. Male ApoE
−/−
mice ( weeks old,  to  g) were intraperitoneally
injected STZ (Sigma-Aldrich, St. Louis, MO) at the dose
of  mg/kg dissolved in  mM citrate buer (pH.)
to induce DM, while the controls received buered saline
alone. Aer one week, we measured the blood glucose levels
of all mice using a glucometer (Abbott Diabetes Care Inc.
Optium Xceed) by tail vein puncture blood s ampling. Aer
weeks, the mice with blood glucose > . mmol/l served
as diabetic group according to the previous studies [], and
the non-STZ-injected ApoE
−/−
mice served as nondiabetic
group. e diabetic and nondiabetic mice were randomly
assigned to treatment group (𝑛=12) and control group
(𝑛=8) separately. e mice in treatment group received
intragastrically dapagliozin (AstraZeneca) . mg/kg/d
for -week treatment, while CBL/J mice and mice in
control group were intragastrically given vehicle . mg/kg/ d.
All animals received human care and all study protocols
were approved by the Institutional Animal Care and Use
Committee of the ird Military Medical University before
performance.
2.2. Metabolic Measurement. Aer intraperitoneal injection
ofSTZ,thebloodglucoseandbodyweightofmicewere
measured weekly. A t an age of weeks, all mice were subject
to – h fasting and then anesthetized by intraperitoneal
injection of % chloral hydrate. ereaer, the blood was
collected via orbital vein and then centrifuged (
C,  r
×  min) to separate the serum. A part of serum was used
to detect the serum concentration of total cholesterol (TCH),
triglyceride (TG), high density lipoprotein cholesterol (HDL-
c), low density lipoprotein cholesterol (LDL-c), and free
fatty acid (FFA) at Clinical Laboratory of the First Aliated
HospitaloftheirdMilitaryMedicalUniversity,andtherest
were preserved at 
Cforthemeasurementsoftheserum
levels of NLRP, IL-𝛽, and IL- with ELISA kits (Shanghai
Jinglai Biotech, shanghai, China) according to t he methods
and procedures provided by the reagent manufacturer .

Mediators of Inammation
2.3. Quantication of Atherosclerotic Lesion Area. Aer blood
collection via orbital vein, the aorta of the mice was dissected
with fat removed under a surgery microscope. e aortic
rootwasembeddedwithOCTandthenmadeinto𝜇m
frozen sections. ereaer, the sections were dried, soaked in
% isopropanol for  min, stained with oil red O (Sigma-
Aldrich, O, USA) working uid ( : ) for  min, dif-
ferentiatedinisopropanolforstilltheinterstitiumwas
clear,andthenwashedwithwater.eobtainedsectionswere
counter-stained with Mayer hematoxylin, dierentiated in %
Hcl-alcohol for  s, blued with running tap water, carefully
dried with lter papers, and mounted with glycogelatin. e
upper segments of aortas were xed in % paraformaldehyde
solution, embedded in paran, and sectioned at 𝜇m. Aer
dehydration, sections were stained with hematoxylin-eosin
(HE, Beyotime, Shanghai, China). Aer removal of adjacent
fat, the upper segments of thoracic aorta and abdominal
aorta were atly placed on the slides, dierentiated in %
isopropanol for  min, stained in oil red O working uid
(:) for h, dierentiated in % isopropanol for -
times till the background color became white, and then
photographed. e area of lesion in the sections of aorta
and aortic root was calculated using Image-pro plus .
(MediaCybernetics, Inc.).
2.4. Immu nouor escent Staining. e frozen sections of aor -
tic root were xed with ice-acetone for  min, added with
.%TritonX-tomakecellmembranepermeabilized,
and blocked with % BSA at room temperature for h. en
they were blocked at
C overnight with primary antibodies
(MOMA- antibody : , Abcam; 𝛼-SMA antibody : ,
Sigma), and incubated at room temperature for h using
secondary antibodies ( :  and : ) protecting from
light for coloration by antigen-antibody binding. Finally they
were counterstained for nuclei with DAPI and mounted with
the uorescer. Macrophages and smooth muscle actin were
observed under confocal uorescence microscope (Zeiss
LSM ) and the positively stained macrophages and
smooth muscle cells were automatically analyzed and quan-
tiedonImage-proplus..
2.5. ROS Assay. To investigate aortic atherosclerotic
lesions ROS levels, isolated arteries were lo aded with DHE
( 𝜇mol/L) for  minutes at 
C. e ROS were observed
under confocal uorescence microscope (Zeiss LSM ),
andDHEturnreduorescentuponoxidation.
2.6. Isolation and Culture Macrophages from Bone Marrow.
WT and NLRP
−/−
KO male mice bone marrow-derived
macrophages (BMDMs) were obtained by dierentiating
bone marrow. Briey, bone marrow cells were ushed out of
femurs and tibias and the remaining cells were maintained in
dierent medium. BMDMs were maintained in macrophage
dierentiating medium (DMEM and M-CSF ( ng/mL,
Sigma)) for days. e medium was supplemented with
% FBS (Hyclone Laboratories), m M L-glutamine, peni-
cillin ( U/mL), and streptomycin ( 𝜇g/mL). To test
the direct action of dapagliozin on macrophages, the WT
macrophageswereplatedintothesterile-wellplate,
stimulated with LPS ( ng/mL, Sigma) for h, and then,
respectively, cultured with . M palmitate (PA, Sigma), hig h
glucose (HG, mmol/L), and PA+HG DMEM medium
for  h. Finally the medium was added with dapagliozin
(. 𝜇M) and new-prepared ATP ( mM) for  min. en
we collected the supernatant of cell culture to assay the
content of IL-𝛽 with ELISA kits (Shanghai Jinglai Biotech,
shanghai, China). To test the action pathway of PA or HG on
IL-𝛽 production, the WT and NLRP
−/−
KO macrophages
were plated into -well plate, stimulated with LPS for h, and
then, respectively, cultured with . M PA, HG ( mmol/L),
and PA+HG DMEM medium for  h and collected cell
lysates to test NLRP and caspase- expression with Western
blot.
2.7. Western Blot. e total aorta tissue was homogenized.
e proteins of tissues and cell lysates were then separated
by SDS-PAGE and further blotted using following specic
antibodies: (anti-NLRP antibody : , anti-ASC antibody
: , anti-caspase- antibody : , anti-IL-𝛽 antibody
: , and anti-IL- antibody : ) (Novus biological,
Littleton, Co., USA). e membranes were scanned with
Typhoon (Pharmacia, USA) and quantitated using Quality
One. e experiments were repeated for times.
2.8. Statistical Analysis. All data were presented as means ±
standard deviation (M ± SD). e nonparametric rank sum
test or analysis of variance (ANOVA) was applied to evaluate
the dierences among the groups. A 𝑝 < 0.05 indicates a
statistically signicant dierence. All statistical analyses were
performed on SPSS (., Inc., Chicago, IL, USA).
3. Results
3.1. Changes of Metabolic Parameters aer Dapagliozin Treat-
ment. e fasting body weight, blood glucose, and blood
lipid levels of mice in each group measured during the
study were shown in Table . No signicant changes of body
weight were detected in DM or non-DM ApoE
−/−
mice t hat
received dapagliozin. However, the levels of blood glucose,
TCH, TG, and FFA of diabetic ApoE
−/−
mice were markedly
increased as compared with nondiabetic ApoE
−/−
mice. More
importantly, aer treatment with dapagliozin, the fasting
blood glucose decreased by % (𝑝 < 0.01)intheDM
ApoE
−/−
mice (𝑝 < 0.05), wh ile HDL-c, TC, and LDL-c levels
were merely aected (Figure ).
3.2. Changes of Serum NLRP3 Inammasome, IL-1𝛽,and
IL-18 aer Dapagliozin Treatment. As shown in Table ,
compared with those of the non-DM mice, DM mice had
signicantly increased serum levels of NLRP (𝑝 < 0.01),
IL-𝛽 (𝑝 < 0.05), and IL- (𝑝 < 0.05). Moreover, serum
IL-𝛽 and IL- levels in nondiabetic control group were
also signicantly increased as compared with those of the
CBL/J group (𝑝 < 0.01). More importantly, aer -week
treatment with dapagliozin in DM mice, the serum levels of
NLRP (𝑝 < 0.01), IL-𝛽 (𝑝 < 0.05), and IL- (𝑝 < 0.05)
proteins were all signicantly reduced, while NLRP and IL-
 levels were not signicantly aected (Figure ).

Mediators of Inammation
DM+dapa
DM+con
NDM+dapa
NDM+con
C57
0
10
20
30
Glucose (mmol/l)
17
18
19
20
21
22
23
24
25
26
27
28
16
Weeks age
(a)
DM+dapa
DM+con
NDM+dapa
NDM+con
C57
20
25
30
35
Weight (g)
17
18
19
20
21
22
23
24
25
26
27
28
16
Weeks age
(b)
##
##
##
∗∗
DM+dapa
DM+con
NDM+dapa
NDM+con
C57
0
10
20
30
Glucose (mmol/l)
16 w
28 w
(c)
DM+dapa
DM+con
NDM+dapa
NDM+con
C57
0
10
20
30
40
Weight (g)
16 w
28 w
(d)
∗∗
∗∗
DM+dapa
DM+con
NDM+dapa
NDM+con
C57
0
10
20
30
40
50
TCH (mmol/l)
(e)
∗∗∗∗
DM+dapa
DM+con
NDM+dapa
NDM+con
C57
0
1
2
3
TG (mmol/l)
(f)
F : Continued.

Mediators of Inammation
C57
DM+con
DM+dapa
NDM+con
NDM+dapa
0
1
2
3
4
5
HDL-C (mmol/l)
(g)
∗∗
C57
DM+con
DM+dapa
NDM+con
NDM+dapa
0
10
20
30
40
LDL-C (mmol/l)
(h)
∗∗
C57
DM+con
DM+dapa
NDM+con
NDM+dapa
0.0
0.5
1.0
1.5
2.0
FFA (mmol/l)
(i)
F : Changes of metabolic parameters in mice from each group. (a) and (c) Compared with non-DM group, the blood glucose in DM
group was signicantly increased; and dapagliozin treatment of  wks signicantly decreased blood glucose in both groups; (b) and (d) no
signicant changes of body weight were detected before and aer dapagliozin treatment in each group; (e), (g), and (h) the levels of cholesterol
were signicantly increased in the ApoE
−/−
mice fed with a high-fat diet, part icularly in DM ApoE
−/−
mice. Dapagliozin treatment did not
reduce the levels of TCH, HDL-c, or LDL-c; (f) and (i): dapagliozin treatment decreased the levels of TG and FFA. , 𝑝 < 0.05; ∗∗, 𝑝 < 0.01;
, 𝑝 < 0.01.
T : Data on body weight, fasting blood glucose, and blood lipid prole in all groups.
DM+dapa DM+con NDM+dapa NDM+con C
BW (g) . ± . . ± . . ± . . ± . . ± .
FBG (mmol/l) 13.69 ± 2.58
##
. ± . 9.17 ± 2.28
. ± . . ± .
TC (mmol/l) . ± . . ± . . ± . . ± . 2.48 ± 0.41
&&
TG (mmol/l) 0.87 ± 0.22
##
. ± . . ± . . ± . . ± .
HDL-c (mmol/l) . ± . . ± . . ± . . ± . . ± .
LDL-c (mmol/l) . ± . . ± . . ± . . ± . 0.45 ± 0.14
&&
FFA (mmol/l) 1.02 ± 0.22
#
. ± . . ± . . ± . . ± .
Diabetic mice that received dapagliozin (DM+dapa), diabetic mice that received vehicle (DM+con), nondiabetic mice that received dapagliozin
(NDM+dapa), nondiabetic mice that received vehicle (NDM+con), and wild-type (C/BL) mice were measured. BW: body weight; FBG: fasting blood
glucose; TC: total cholesterol; TG: triglycerides; HDL-c: HDL cholesterol; LDL-c: LDL cholesterol; FFA: free fat acid; values are expressed as mean ± SD, per
group. #, 𝑝 < 0.05; ##, 𝑝 < 0.01 versus DM+con; , 𝑝 < 0.05 versus NDM+con; &&, 𝑝 < 0.01 versus NDM+con.

Citations
More filters
Journal ArticleDOI

Mechanisms of Cardiovascular Benefits of Sodium Glucose Co-Transporter 2 (SGLT2) Inhibitors: A State-of-the-Art Review

TL;DR: The potential mechanisms by which SGLT2 inhibitors exert their beneficial effects are discussed, including beneficial effects on cardiac energy metabolism, reducing inflammation, improving kidney function, and increasing erythropoiesis.
Journal ArticleDOI

Vascular Inflammation and Oxidative Stress: Major Triggers for Cardiovascular Disease

TL;DR: The value of immunomodulation for the treatment of cardiovascular disease was recently supported by large-scale clinical trials demonstrating reduced cardiovascular mortality in patients with established atherosclerotic disease when treated by highly specific anti-inflammatory therapies.
Journal ArticleDOI

Reactive oxygen species (ROS) in macrophage activation and function in diabetes.

TL;DR: The role of ROS in macrophage polarization is dissected, how ROS production links metabolism and inflammation in diabetes and its complications is analyzed, and the contribution of ROS to the crosstalk between macrophages and endothelial cells in diabetic complications is discussed.
Journal ArticleDOI

SGLT2 inhibition via dapagliflozin improves generalized vascular dysfunction and alters the gut microbiota in type 2 diabetic mice

TL;DR: Dapagliflozin treatment improves arterial stiffness, endothelial dysfunction and vascular smooth muscle dysfunction, and subtly alters microbiota composition in type 2 diabetic mice, and may represent an important mechanism underlying the cardiovascular benefits of SGLT2i treatment.
Journal ArticleDOI

Oxidative Stress and NLRP3-Inflammasome Activity as Significant Drivers of Diabetic Cardiovascular Complications: Therapeutic Implications.

TL;DR: A review of the latest findings suggesting not only a key role for inflammatory pathways underpinning metabolic and haemodynamic dysfunction in diabetes, but furthermore that these perturbations are driven by activation of the NOD-like receptor family pyrin domain containing 3 (NLRP3) inflammasome.
References
More filters
Journal ArticleDOI

Inflammation, Atherosclerosis, and Coronary Artery Disease

TL;DR: The evidence is recounted that atherosclerosis, the main cause of CAD, is an inflammatory disease in which immune mechanisms interact with metabolic risk factors to initiate, propagate, and activate lesions in the arterial tree.
Journal ArticleDOI

Inflammation and Atherosclerosis

TL;DR: New insights into inflammation in atherosclerosis not only increase the understanding of this disease, but also have practical clinical applications in risk stratification and targeting of therapy for this scourge of growing worldwide importance.
Journal ArticleDOI

Pattern Recognition Receptors and Inflammation

TL;DR: The role of PRRs, their signaling pathways, and how they control inflammatory responses are discussed.
Journal ArticleDOI

Inflammation and atherosclerosis.

TL;DR: New insights are provided on inflammatory processes involved in atherosclerosis development provide important links between risk factors and the mechanisms of atherogenesis and a major challenge for future research is to implement these new insights in order to improve strategies for prediction, prevention and treatment of cardiovascular events.
Related Papers (5)