ArticlePDF AvailableLiterature Review

Thiazolidinediones and risk for atherosclerosis: Pleiotropic effects of PPARγ agonism

SAGE Publications Inc
Diabetes and Vascular Disease Research
Authors:

Abstract

Despite advances in the development of anti-hyper-glycaemic drugs and a greater focus on cardiovascular risk modification for patients with diabetes, cardiovascular disease remains the most common complication of type 2 diabetes. Since their initial availability in 1997, the thiazolidinediones have become one of the most commonly prescribed classes of medications for type 2 diabetes. In addition to glucose control, the thiazolidinediones have a number of pleiotropic effects on myriad traditional and non-traditional risk factors for cardiovascular disease, and hold promise with regard to modification of cardiovascular risk. In a recently reported large-scale clinical trial, pioglitazone was associated with improved cardiovascular outcomes in patients with type 2 diabetes and prevalent atherosclerotic disease. In this review, we summarise the experimental, pre-clinical and clinical data regarding the effects of the thiazolidinediones on cardiovascular risk factors and clinical outcomes.
VOLUME 3 ISSUE 2 .SEPTEMBER 2006 65
Abstract
Despite advances in the development of anti-hyper-
glycaemic drugs and a greater focus on cardiovas-
cular risk modification for patients with diabetes,
cardiovascular disease remains the most common com-
plication of type 2 diabetes. Since their initial availability
in 1997, the thiazolidinediones have become one of the
most commonly prescribed classes of medications for
type 2 diabetes. In addition to glucose control, the thia-
zolidinediones have a number of pleiotropic effects on
myriad traditional and non-traditional risk factors for car-
diovascular disease, and hold promise with regard to
modification of cardiovascular risk. In a recently reported
large-scale clinical trial, pioglitazone was associated with
improved cardiovascular outcomes in patients with type
2 diabetes and prevalent atherosclerotic disease.
In this review, we summarise the experimental, pre-
clinical and clinical data regarding the effects of the thia-
zolidinediones on cardiovascular risk factors and clinical
outcomes.
Diabetes Vasc Dis Res 2006;3:65–71
Key words: thiazolidinedione, pioglitazone, rosiglitazone,
troglitazone, peroxisome proliferator-activated receptor,
inflammation, atherosclerosis.
Introduction
A global epidemic of diabetes mellitus (DM) has emerged,
with the World Health Organization (WHO) estimating a
global prevalence of more than 300 million people with DM
by the year 2025 (more than double the 1995 prevalence).1
This projection may well be an underestimate based on
2004 data from the Centers for Disease Control (CDC)
Behavior Risk Factor Surveillance System – these data pro-
vide a 2004 DM prevalence estimate of 20.7 million adults
in the US with diagnosed DM, a level that was originally pro-
jected to be reached in the year 2010. Moreover, as many
as one-third of patients with type 2 DM, which represents
more than 90% of DM worldwide, remain undiagnosed,2,3
and these estimates do not take into account individuals
who have impaired glucose tolerance (IGT), the metabolic
forerunner to DM.
Cardiovascular disease (CVD) is the most common com-
plication of type 2 DM, accounting for approximately 80%
of deaths.4Therefore, it is imperative to consider cardiovas-
cular risk modification as a key focus for the treatment of
patients with DM, both in clinical decision-making and ther-
apeutic drug development. This review summarises the
effects of the thiazolidinedione (TZD) class of medications
on a number of experimental, pre-clinical and clinical mea-
sures of atherosclerotic cardiovascular risk and disease, and
the potential for TZDs to influence cardiovascular outcomes
among patients with type 2 DM. The latter hypothesis is
being tested in a number of large-scale randomised clinical
outcomes trials.
The thiazolidinediones
The TZD class of drugs, which currently includes rosiglitazone
(Avandia®) and pioglitazone (Actos®), has emerged as an effec-
tive treatment of hyperglycaemia associated with type 2 DM,
alone or in combination with other oral hypoglycaemic med-
ications and with insulin. The first TZD, ciglitazone, was iden-
tified in a compound-screening programme for lipid lowering
and shares structural homology to the fibric acid derivatives,
which are agonists of the PPARαreceptor. While ciglitazone
had only modest effects on lipid parameters in animal mod-
els, it was noted to have unexpected glucose-lowering effects.
This observation led to subsequent development of the TZD
class of compounds for glucose control in the treatment of
type 2 DM. Troglitazone was the first to achieve regulatory
approval, despite poor understanding of the mechanism of
action. Troglitazone was approved for clinical use in the US in
1994, and pioglitazone and rosiglitazone in 1997; troglita-
zone was withdrawn from the market in 1997 due to idio-
syncratic hepatotoxicity, an adverse effect that has not been
observed with the other TZDs.
During the mid-1990s, the TZDs were discovered to be
potent and selective agonists for peroxisome proliferator-
activated receptor γ(PPARγ).5,6 The PPARs, which include
PPARα, PPARγand PPARδ(also referred to as PPARβ), are
members of the nuclear hormone receptor superfamily of
REVIEW
Thiazolidinediones and risk for
atherosclerosis: pleiotropic effects of
PPARγagonism
CHETAN B PATEL, JAMES A DE LEMOS, KATHLEEN L WYNE, DARREN K MCGUIRE
Department of Internal Medicine, Duke University Medical Center,
Durham, NC, US.
Chetan B Patel, Fellow, Division of Cardiovascular Medicine
Department of Internal Medicine, University of Texas Southwestern
Medical Center, Dallas, Texas, US.
James A de Lemos, Associate Professor of Medicine
Kathleen L Wyne, Assistant Professor of Medicine
Darren K McGuire, Assistant Professor of Medicine
Correspondence to: Dr Darren K McGuire
UT-Southwestern Cardiology, 5323 Harry Hines Blvd, Dallas, TX 75390-
9047, US.
Tel: +1 214 645 7500; Fax: +1 214 645 7501
E-mail: darren.mcguire@utsouthwestern.edu
66 DIABETES AND VASCULAR DISEASE RESEARCH
ligand-activated transcription factors, which also includes the
retinoic acid receptor (RXR) and the steroid and thyroid hor-
mone receptors.7Interaction of the PPARs with natural or
synthetic agonist ligands, with or without co-activation with
RXR, induces receptor binding to DNA response elements
(PPAR-response element [PPRE]),8and regulates a number of
physiological actions through transcriptional activation and
repression.9The transcriptional regulation associated with
PPARγprimarily involves adipogenesis, as well as glucose
and lipid metabolism.7
As the target of TZD medications, PPARγis the most
extensively studied of the PPAR subtypes. The receptor is
primarily expressed in adipose tissue, where it regulates adi-
pogenesis, the process of differentiation from pluripotent
stem cells to mature metabolically active adipocytes, and
glucose and lipid homeostasis.10 It has also been identified in
liver, skeletal muscle, cardiac muscle, renal cortex, colonic
epithelium, vascular endothelial cells, renal tubular cells,
renal collecting duct and macrophages.11-14 Although fairly
consistent tissue expression of PPARγhas been observed
across animal species, there are some differences between
the distribution in animals and humans,15 making it impera-
tive that effects observed in animal models be verified in
humans.
Since their introduction, rosiglitazone and pioglitazone
have quickly become among the most widely prescribed
medications for the treatment of hyperglycaemia associated
with DM, accounting for about 20% of prescriptions for oral
hypoglycaemic medications in the US. Data are rapidly
emerging that suggest diverse effects of the TZDs on a num-
ber of intermediate biomarkers and risk factors associated
with cardiovascular disease (table 1). These include effects
on conventional cardiovascular risk factors such as dyslipi-
daemia and hypertension, and modulation of endothelial
reactivity and the inflammatory cascade implicated in the
earliest stages of atherosclerosis.16
Adipogenesis
PPARγis a critical transcription factor in the regulation of
adipocyte differentiation. Through their effects on PPARγ,
TZDs modulate adipocyte differentiation, increase the num-
ber of insulin-sensitive small adipocytes,17 and lead to a
transfer of fat distribution from visceral to subcutaneous
depots – a pattern that has been associated with lower car-
diovascular risk.18,19 By potentiating the effects of insulin,
TZDs inhibit lipolysis and up-regulate expression of free fatty
acid transporters in adipocytes (fatty acid transport protein
[FATP]-1; CD36),20 thereby reducing circulating free fatty
acids, which have been implicated in induction of insulin
resistance in liver and skeletal muscle.6Therefore, the net
effect of PPARγactivation is an increased mass of small,
insulin-sensitive subcutaneous adipocytes and a larger reser-
voir for free fatty acid deposition. The long-term conse-
quences of this increased fat mass remain unclear, but the
improvement in insulin sensitivity associated with TZDs and
the redistribution of fat from the solid organs and the viscer-
al fat depot to the subcutaneous fat depot support the pos-
sibility of decreased cardiovascular risk despite increasing
adiposity, a hypothesis that requires further investigation.
Lipid metabolism
Dyslipidaemia is common among patients with DM: it is
characterised by high triglycerides, low high-density lipopro-
tein (HDL) cholesterol, and modest elevations of low-densi-
ty lipoprotein (LDL) cholesterol with increased fractions of
small, dense LDL particles, all of which are associated with
increased cardiovascular risk.21 Both rosiglitazone and piogli-
tazone are associated with increases in HDL and LDL cho-
lesterol concentrations, shifting the LDL particle profile
toward larger, more buoyant particles so resulting in little
change in LDL particle concentration despite increased LDL
mass. A recent head-to-head clinical trial showed that piogli-
tazone decreased and rosiglitazone increased triglyceride
concentrations; the net lipid effect was a slightly lower
LDL/HDL ratio with pioglitazone and little change with
rosiglitazone.22 It remains unclear whether the lipoprotein
effects are an indirect result of improved glycaemic control,
a direct effect of the TZDs or a combination, but with iden-
tical glucose control achieved in both groups of the head-to-
head trial,22 it appears that the lipid effects of the TZDs
extend beyond glycaemic modulation.
The clinical relevance of these TZD lipid effects remains
uncertain, especially in the context of recommendations to
treat all patients with type 2 DM who are at increased car-
diovascular risk with either a statin or a fibrate. These treat-
REVIEW
Table 1. Pleiotropic effects of thiazolidinediones (TZDs) on
cardiovascular parameters
Lipids Inflammation
/ Triglycerides IL-6, TNF-α, CRP, MMP-9
HDL-C and LDL-C I-CAM; V-CAM
/ - LDL/HDL ratio
/ - TG/HDL ratio
Buoyancy of LDL particles MCP-1, sCD40L
Coagulation Ventricular performance
PAI-1 / - Systolic function
Fibrinogen / - Cardiac output
Platelet aggregation / - LVH
Proinsulin Diastolic function
Infarct size
Vascular effects Fat distribution
Blood pressure Visceral fat
Intima-media thickness Subcutaneous fat
Brachial artery reactivity Fat content of liver
Coronary flow reserve
Vascular permeability
Other
Intravascular volume
Microalbuminuria
Key: HDL = high-density lipoprotein; LDL = low-density lipoprotein;
TG= triglycerides; PAI-1= plasminogen activator inhibitor;
IL = interleukin; TNF = tumour necrosis factor; CRP = C-reactive
protein; MMP = matrix metalloproteinase; I-ICAM = intercellular
adhesion molecule; V-CAM = vascular cell adhesion molecule;
MCP = monocyte-chemoattractant protein; sCD40L = soluble CD40
ligand; LVH = left ventricular hypertrophy
VOLUME 3 ISSUE 2 .SEPTEMBER 2006 67
ments have been specifically excluded in most studies
assessing the lipid effects of TZDs reported to date.
Hypertension
In animal models, TZDs attenuate the pressor response to
norepinephrine and angiotensin II and prevent the develop-
ment of hypertension.23-26 The mechanism of these observa-
tions remains unclear, but appears to be independent of the
metabolic effects of PPARγagonism. One possible mecha-
nism is blockade of voltage-gated (L-type) calcium channels,
the target of action of dihydropyridine calcium channel
blockers and a property of all TZDs.26 The TZDs also
increase expression of vascular endothelial growth factor
(VEGF) and inhibit expression of endothelin (ET)-1,27 both of
which may contribute to their favourable influence on blood
pressure. In patients with DM with and without hyperten-
sion, TZDs have been shown to reduce blood pressure,
although the changes tend to be modest (1–3 mmHg).28,29
Vascular effects
Endothelial function
Endothelial dysfunction, a precursor to atherosclerotic dis-
ease, is favourably affected by TZDs.30-31 One proposed
mechanism for this observation involves augmentation of
insulin-dependent endothelial nitric oxide (eNO) release,
resulting in enhanced endothelium-dependent vasodila-
tion.32,33 Support for this theory comes from experimental
data showing an increase in eNO production with troglita-
zone treatment in a bovine aortic endothelial cell prepara-
tion,30 and reversal of angiotensin II effects on endothelial
dysfunction in mice treated with pioglitazone or rosiglita-
zone.34 In controlled human studies, both troglitazone and
pioglitazone have been shown to increase forearm blood
flow in response to acetylcholine in patients with type 2
DM,35,36 while rosiglitazone improved coronary flow reserve
in response to cold-pressor stimulus.37
Inflammation
Data continue to accumulate supporting a potential causal
relationship between inflammation and the development
and progression of atherosclerosis.38 TZDs have been shown
to inhibit the initial inflammatory cascade in ex vivo prepa-
rations of human endothelial cells,39,40 and to inhibit the
expression of adhesion molecules on endothelial cells.41 The
TZDs also exhibit direct effects on monocytes/macrophages,
with reduced expression of inflammatory cytokines.42 In
human studies, TZDs decrease circulating markers of inflam-
mation, including TNFα, C-reactive protein, monocyte-
chemoattractant protein (MCP)-1, matrix metalloproteinase
(MMP)-9, soluble CD40 ligand (sCD40L) and white blood
cell count.43-45
The TZDs also have a potentially pro-atherogenic effect
via the up-regulation of expression of the macrophage free
fatty acid scavenger receptor CD36, which increases uptake
of oxidised LDL and may increase development of foam
cells that are directly implicated in plaque formation and
progression. However, this potentially deleterious effect may
be countered by up-regulation of ABCA-1, a transporter that
facilitates cholesterol efflux from macrophages.46,47
In summary, the TZDs have divergent effects on parame-
ters of inflammation, but most data support a fairly robust
anti-inflammatory effect associated with these drugs.16 The
net influence of inflammatory modulation of the TZDs on
atherosclerosis disease development and clinical manifesta-
tions remains to be defined.
Atherosclerosis development and progression
In ex-vivo preparations of animal and human vascular smooth
muscle and endothelial cells, TZDs are potent inhibitors of
cellular migration and proliferation, important contributors to
atherosclerosis.48,49 In the LDL-receptor null mouse model of
atherosclerosis, troglitazone and rosiglitazone attenuated aor-
tic atherosclerotic disease progression in mice fed both high-
fat and high-fructose diets.50,51 In the clinical setting, TZDs
attenuate the progression of carotid intima media thickness
(IMT) and decrease restenosis following percutaneous coro-
nary intervention in patients with and without diabetes;52-57
these differences were independent of glycaemic control or
changes in lipid concentrations.53,57 The accumulated data
from animal and human investigations support the potential
efficacy of the TZDs on atherosclerotic disease development
and progression and on cardiovascular risk modification
among patients with type 2 DM.
Acute coronary syndromes
Diabetes is associated with a 2–4-fold increased risk for
developing acute coronary complications,58 including unsta-
ble angina and myocardial infarction, and with increased risk
for subsequent cardiovascular morbidity and mortality com-
pared with patients without diabetes.59 Increased plaque vul-
nerability among patients with DM and coronary disease
may account for this increased risk, with a pro-inflammato-
ry state and perturbations of the coagulation system as
potential contributors. Evidence of a pro-thrombotic state
associated with DM is increased circulating plasminogen
activator inhibitor 1 (PAI-1) and sCD40L, both of them asso-
ciated with pro-coagulant effects and inflammation.60-63
Exposure to TZDs decreases PAI-1 expression in ex vivo
human endothelial cell and adipocyte preparations,64 and is
associated with decreased plasma levels of PAI-1 in obese
patients who do not have diabetes.65 Compared with place-
bo, rosiglitazone treatment is associated with decreased cir-
culating sCD40L in patients with DM,45 whereas pioglitazone
had no effect on sCD40L levels compared with glimepiri-
de.66 Likewise, measures of platelet activity dropped in a
controlled study of patients with stable coronary artery dis-
ease but without diabetes who were treated with rosiglita-
zone.67
Once a plaque becomes unstable and an acute coronary
ischaemic event occurs, TZDs may further influence the
ischaemic complications favourably. In several animal mod-
els of myocardial ischaemia/reperfusion, PPARγagonism
with either natural ligand (15d PGJ2) or with TZDs resulted
in dose-dependent decreases in myocardial infarct size and
improved parameters of cardiac performance.68-70 In the
studies by Khandoudi and Liu, rosiglitazone treatment inhib-
ited activation of the mitogen-activated protein (MAP) kinase
and the Jun NH2-terminal kinase (JNK) pathways, which play
REVIEW
68 DIABETES AND VASCULAR DISEASE RESEARCH
a key role in cardiac myocyte apoptosis. This observation
suggests that the decrease in infarct size may at least in part
be due to decreased apoptosis in response to ischaemic
injury. Whether similar mechanisms are present in humans
has yet to be investigated.
Cardiovascular clinical outcomes trials
Based on the accumulated data suggesting potential direct
cardiovascular benefits of the TZDs, a number of large-scale
randomised controlled trials have been initiated to assess the
effect of rosiglitazone and pioglitazone on major adverse
cardiovascular clinical outcomes (table 2). While most of
these studies are 2–4 years from completion, the
PROspective pioglitAzone Clinical Trial in macroVascular
Events (PROactive) study results were recently reported. This
was the first large-scale randomised clinical outcomes study
to evaluate the effect of a TZD on secondary prevention of
major adverse cardiovascular outcomes.29
The PROactive study was a double-blind, placebo-con-
trolled study that enrolled patients with type 2 DM and
prevalent atherosclerotic vascular disease. Participants were
randomised to treatment with pioglitazone versus placebo,
added to existing therapy, with mean follow-up of three
years.29,71 Pioglitazone was initiated at 15 mg daily, with
monthly increases to the target dose of 45 mg daily that was
continued throughout the study. The primary end point was
the time to the first occurrence of a composite of seven
major adverse cardiovascular events, including spontaneous
clinical events (all-cause mortality, myocardial infarction
[MI], cerebrovascular accident [CVA] or acute coronary syn-
drome [ACS]) and clinically-driven vascular events (coronary
or leg revascularisation or leg amputation). A prospectively
determined hierarchical series of secondary end points
included, in order: the time to the first occurrence of a com-
posite of death, MI and CVA (the 'principal' secondary end
point); cumulative cardiovascular death; and time to first
occurrence of each of the primary composite component
end points.
The study included 5,238 participants who were
enrolled between May 2001 and April 2002 from 321 cen-
tres in 19 European countries. Mean treatment duration was
2.9 years. Despite accumulating more than 1,000 primary
end point events (514 in the pioglitazone arm versus 572 in
the placebo arm), the study failed to demonstrate a statisti-
cally significant difference in the primary end point between
the treatment groups, but there was a trend toward a 10%
reduction (23.5% vs. 21%; p=0.095) favouring pioglitazone.
Treatment with pioglitazone did significantly reduce the
prospectively identified principal secondary end point, the
time to occurrence of the first event of a composite of all-
cause mortality/MI/stroke (14.4% vs. 12.3%; HR 0.84;
p=0.027). Therefore, the authors concluded that pioglita-
zone reduced the composite of all-cause mortality, non-fatal
MI and stroke in patients with type 2 DM who have a high
risk of macrovascular events.29
This conclusion is further supported when considering all
accumulated cardiovascular events in the study, which num-
bered more than 1,700. In this comparison, 803 events
occurred in the group allocated to pioglitazone treatment
compared with 900 events in the placebo group, represent-
ing a 10.8% relative reduction (statistical testing of this com-
parison was not reported). These data are the first to suggest
that addition of a TZD to the medical regimen of patients
with type 2 DM and macrovascular disease may prevent fur-
ther cardiovascular morbidity and mortality.
Congestive heart failure
While most of the CV effects of TZDs appear favourable,
there remains a concern with regard to the propensity for
these drugs to cause peripheral oedema and to precipitate
REVIEW
Table 2. Randomised clinical trials underway to evaluate the effect of TZDs on cardiovascular clinical outcomes
Study Objective Treatments Size Length 10end points 20end points
IRIS Prevent CV Pio vs. Plac 3,136 3 years CVA or MI CVA, ACS, new DM,
events mortality
RECORD Prevent CV Met+Rsg; 4,000 6 years Time to CV Individual CV events
events Rsg+SU; event composite and glycaemic
Met+SU parameters
BARI-2D Prevent death IP (ins+/-SU) 3,000 5 years All-cause mortality CV events
and CV vs. IS (TZD Quality of life
events +/- Met)
DREAM Prevent Rsg vs. plac; 4,000 3 years New DM or death MACE;
IGT DM Ram vs. plac Microvascular
and death end points
Key: CV = cardiovascular; Pio = pioglitazone; Plac = placebo; MI = myocardial infarction; CVA = cerebrovascular accident; ACS = acute coronary
syndromes; DM = diabetes mellitus; IGT = impaired glucose tolerance; Met = metformin; Rsg = rosiglitazone; SU = sulfonylurea; IP = insulin-
providing therapy; Ins = insulin; IS = insulin-sensitising therapy; Ram = ramipril; MACE = major adverse cardiovascular events; IRIS = Insulin Resistance
Intervention after Stroke: effect of pioglitazone versus placebo on subsequent stroke and myocardial infarction in population with prior stroke and insulin
resistance but without DM; RECORD = Rosiglitazone Evaluated for Cardiac Outcomes and Regulation of glycaemia in DM; BARI-2D = Bypass
Angioplasty Revascularisation Intervention in type 2 DM; DREAM = Diabetes Reduction Assessment with rosiglitazone and ramipril Medication
VOLUME 3 ISSUE 2 .SEPTEMBER 2006 69
or exacerbate congestive heart failure (CHF).72 The clinical
relevance of these observations remains unclear but has
prompted inclusion in the product labelling for both rosigli-
tazone and pioglitazone of a caution against the use of TZDs
for patients with, or at high risk of developing, CHF.73,74 These
warnings are further supported by a position statement from
a joint panel of the American Diabetes Association (ADA)
and the American Heart Association (AHA).75
Several lines of evidence suggest that although plasma
volume expansion occurs with the TZDs,74,76,77 and some
patients develop signs and symptoms of CHF, direct cardiac
toxicity does not result from administration of these drugs. In
animal models, PPARγagonists improve contractility and sys-
tolic performance,78-81 enhance diastolic performance,79-82
and decrease cardiac hypertrophy independent of loading
conditions.83-85 Similarly, randomised controlled trials of
patients with DM have demonstrated no untoward effects
on cardiac performance, assessed using echocardiography,
and some trends toward improved function associated with
longer-term TZD therapy.78,86
The mechanism contributing to peripheral oedema and
incremental risk for CHF is most likely to be alteration of
sodium handling resulting in net volume retention.14 In mice,
PPARγexpression was identified in endothelial cells lining
the renal collecting duct (CD), and a mouse model with CD-
specific knockout of PPARγwas resistant to rosiglitazone-
induced increases in body weight and plasma volume
expansion (dietary sodium intake was unchanged). These
provocative findings demonstrate a mechanism of PPARγ-
dependent sodium retention and require confirmation in
humans.
In the placebo-controlled PROactive study,29 pioglitazone
was associated with a 2% absolute increase in hospitalisation
for incident CHF compared with placebo. Although there
were too few CHF mortality events to assess the effect rigor-
ously, there was no evident increase in risk with pioglitazone
therapy: the risk was 1% in both groups. Similarly, among
16,417 Medicare beneficiaries with DM discharged with a
primary diagnosis of CHF, the use of TZDs was associated
with a significant 13% reduced adjusted odds for mortality,
despite a statistically significant 6% increased rate of rehos-
pitalisation for CHF.87 In another observational study evalu-
ating TZD use in 111 patients with CHF, TZDs were associ-
ated with an increased rate of peripheral oedema, but there
was no change in New York Heart Association (NYHA) class
or echocardiographic severity of cardiac dysfunction,88 and
the fluid retention was reversible with drug withdrawal or
dose reduction.
The mechanisms and clinical relevance of peripheral
oedema and incident CHF resulting from TZD use remain to
be defined. Until further data are available, the cautionary
guidelines from the product manufacturers, AHA and ADA
should guide clinical decision-making with regard to the
judicious use of TZDs in patients with impaired left ventric-
ular function and CHF. For Class I and II CHF, TZDs may be
used with close observation for signs and symptoms of CHF,
with dose reduction or drug discontinuation if such findings
are observed; TZDs are presently not recommended for use
in patients with Class III or IV CHF.
Conclusions
The TZDs are safe and effective in the treatment of
hyperglycaemia associated with type 2 DM as an adjunct to
therapeutic lifestyle modification, as monotherapy or in
combination with existing therapies. The apparent
pleiotropic cardiovascular effects of these medications have
fostered optimism regarding their use to improve cardiovas-
cular outcomes among this high-risk population of patients,
and the PROactive study results demonstrate the efficacy of
pioglitazone in a population at high risk for cardiovascular
events. These accumulated data provide support for use of a
thiazolidinedione as an 'evidence-based' strategy for the
treatment of DM patients with increased cardiovascular risk.
Despite their efficacy in glucose control and cardiovas-
cular risk modification, a caution remains with regard to the
propensity for TZDs to precipitate or exacerbate CHF, and
improved understanding of the mechanism(s) and relevance
of the CHF signal remain important research objectives.
Conflicts of interest
DKM has received honoraria (Speakers Bureau) from Takeda
Pharmaceuticals and has research grant support from
GlaxoSmithKline. KLW has received honoraria (Speakers
Bureau) and research grant support from GlaxoSmithKline.
CBP and JADL have no conflicts of interest to declare.
References
1. King H, Aubert RE, Herman WH. Global burden of diabetes, 1995-
2025: prevalence, numerical estimates, and projections. Diabetes Care
1998;21:1414-31.
2. Saydah SH, Loria CM, Eberhardt MS, Brancati FL. Subclinical states of
glucose intolerance and risk of death in the US. Diabetes Care 2001;
24:447-53.
3. Taubert G, Winkelmann BR, Schleiffer T et al. Prevalence, predictors,
and consequences of unrecognized diabetes mellitus in 3266 patients
scheduled for coronary angiography. Am Heart J 2003;145:285-91.
4. American Diabetes Association. Consensus statement: role of cardio-
vascular risk factors in prevention and treatment of macrovascular dis-
ease in diabetes. Diabetes Care 1993;16:72-8.
5. Lehmann JM, Moore LB, Smith-Oliver TA, Wilkison WO, Willson TM,
Kliewer SA. An antidiabetic thiazolidinedione is a high affinity ligand for
peroxisome proliferator-activated receptor gamma (PPAR gamma). J
Biol Chem 1995;270:12953-6.
6. Spiegelman BM. PPAR-gamma: adipogenic regulator and thiazolidine-
dione receptor. Diabetes 1998;47:507-14.
7. Willson TM, Brown PJ, Sternbach DD, Henke BR. The PPARs: from
orphan receptors to drug discovery. J Med Chem 2000;43:527-50.
8. Kliewer SA, Sundseth SS, Jones SA et al. Fatty acids and eicosanoids
regulate gene expression through direct interactions with peroxisome
proliferator-activated receptors alpha and gamma. Proc Natl Acad Sci
USA 1997;94:4318-23.
9. Yki-Jarvinen H. Thiazolidinediones. N Engl J Med 2004;351:1106-18.
10. Olefsky JM. Treatment of insulin resistance with peroxisome pro-
liferator-activated receptor gamma agonists. J Clin Invest 2000;106:
467-72.
11. Marx N. PPARgamma and vascular inflammation: adding another piece
to the puzzle. Circ Res 2002;91:373-4.
12. Plutzky J. Peroxisome proliferator-activated receptors in endothelial cell
biology. Curr Opin Lipidol 2001;12:511-18.
13. Lowell BB. PPARgamma: an essential regulator of adipogenesis and
modulator of fat cell function. Cell 1999;99:239-42.
14. Zhang H, Zhang A, Kohan DE, Nelson RD, Gonzalez FJ, Yang T.
Collecting duct-specific deletion of peroxisome proliferator-activated
receptor gamma blocks thiazolidinedione-induced fluid retention. Proc
Natl Acad Sci USA 2005;102:9406-11.
15. Desvergne B, Wahli W. Peroxisome proliferator-activated receptors:
nuclear control of metabolism. Endocr Rev 1999;20:649-88.
16. Plutzky J. Medicine. PPARs as therapeutic targets: reverse cardiology?
Science 2003;302:406-07.
17. Okuno A, Tamemoto H, Tobe K et al. Troglitazone increases the num-
REVIEW
70 DIABETES AND VASCULAR DISEASE RESEARCH
ber of small adipocytes without the change of white adipose tissue mass
in obese Zucker rats. J Clin Invest 1998;101:1354-61.
18. Adams M, Montague CT, Prins JB et al. Activators of peroxisome prolif-
erator-activated receptor gamma have depot-specific effects on human
preadipocyte differentiation. J Clin Invest 1997;100:3149-53.
19. Kissebah AH, Krakower GR. Regional adiposity and morbidity. Physiol
Rev 1994;74:761-811.
20. Martin G, Schoonjans K, Lefebvre AM, Staels B, Auwerx J. Coordinate
regulation of the expression of the fatty acid transport protein and acyl-
CoA synthetase genes by PPARalpha and PPARgamma activators. J Biol
Chem 1997;272:28210-17.
21. St-Pierre AC, Cantin B, Dagenais GR et al. Low-density lipoprotein sub-
fractions and the long-term risk of ischemic heart disease in men. 13-
year follow-up data from the Quebec Cardiovascular Study.
Arterioscler Thromb Vasc Biol 2005;25:553-9.
22. Goldberg RB, Kendall DM, Deeg MA et al. A comparison of lipid and
glycemic effects of pioglitazone and rosiglitazone in patients with type
2 diabetes and dyslipidemia. Diabetes Care 2005;28:1547-54.
23. Yoshioka S, Nishino H, Shiraki T et al. Antihypertensive effects of CS-
045 treatment in obese Zucker rats. Metabolism 1993;42:75-80.
24. Kaufman LN, Peterson MM, DeGrange LM. Pioglitazone attenuates
diet-induced hypertension in rats. Metabolism 1995;44:1105-09.
25. Zhang HY, Reddy SR, Kotchen TA. Antihypertensive effect of pioglita-
zone is not invariably associated with increased insulin sensitivity.
Hypertension 1994;24:106-10.
26. Zhang F, Sowers JR, Ram JL, Standley PR, Peuler JD. Effects of pioglita-
zone on calcium channels in vascular smooth muscle. Hypertension
1994;24:170-5.
27. Iglarz M, Touyz RM, Amiri F, Lavoie MF, Diep QN, Schiffrin EL. Effect of
peroxisome proliferator-activated receptor-alpha and -gamma activa-
tors on vascular remodeling in endothelin-dependent hypertension.
Arterioscler Thromb Vasc Biol 2003;23:45-51.
28. Chiquette E, Ramirez G, Defronzo R. A meta-analysis comparing the
effect of thiazolidinediones on cardiovascular risk factors. Arch Intern
Med 2004;164:2097-104.
29. Dormandy JA, Charbonnel B, Eckland DJ et al. Secondary prevention
of macrovascular events in patients with type 2 diabetes in the
PROactive Study (PROspective pioglitAzone Clinical Trial In
macroVascular Events): a randomised controlled trial. Lancet 2005;
366:1279-89.
30. Cho DH, Choi YJ, Jo SA, Jo I. Nitric oxide production and regulation of
endothelial nitric-oxide synthase phosphorylation by prolonged treat-
ment with troglitazone: evidence for involvement of peroxisome prolif-
erator-activated receptor (PPAR) gamma-dependent and PPARgamma-
independent signaling pathways. J Biol Chem 2004;279:2499-506.
31. Ryan MJ, Didion SP, Mathur S, Faraci FM, Sigmund CD. PPAR(gamma)
agonist rosiglitazone improves vascular function and lowers blood pres-
sure in hypertensive transgenic mice. Hypertension 2004;43:661-6.
32. Scherrer U, Sartori C. Insulin as a vascular and sympathoexcitatory hor-
mone: implications for blood pressure regulation, insulin sensitivity, and
cardiovascular morbidity. Circulation 1997;96:4104-13.
33. Kotchen TA, Zhang HY, Reddy S, Hoffmann RG. Effect of pioglitazone
on vascular reactivity in vivo and in vitro. Am J Physiol 1996;270:R660-
6.
34. Diep QN, Amiri F, Touyz RM et al. PPARalpha activator effects on Ang
II-induced vascular oxidative stress and inflammation. Hypertension
2002;40:866-71.
35. Caballero AE, Saouaf R, Lim SC et al. The effects of troglitazone, an
insulin-sensitizing agent, on the endothelial function in early and late
type 2 diabetes: a placebo-controlled randomized clinical trial.
Metabolism 2003;52:173-80.
36. Forst T, Lubben G, Hohberg C et al. Influence of glucose control and
improvement of insulin resistance on microvascular blood flow and
endothelial function in patients with diabetes mellitus type 2.
Microcirculation 2005;12:543-50.
37. Plummer EV, Lawson M, Domb A, Chen K, Gerkin R, Reaven PD.
Effects of treatment with rosiglitazone on myocardial blood flow in type
2 diabetes. Diabetes 2002;51:A161(Abstract 653-P).
38. Libby P, Ridker PM, Maseri A. Inflammation and atherosclerosis.
Circulation 2002;105:1135-43.
39. Marx N, Mach F, Sauty A et al. Peroxisome proliferator-activated recep-
tor-gamma activators inhibit IFN-gamma-induced expression of the T
cell-active CXC chemokines IP-10, Mig, and I-TAC in human endothe-
lial cells. J Immunol 2000;164:6503-08.
40. Marx N, Kehrle B, Kohlhammer K et al. PPAR activators as antiinflam-
matory mediators in human T lymphocytes: implications for athero-
sclerosis and transplantation-associated arteriosclerosis. Circ Res
2002;90:703-10.
41. Pasceri V, Wu HD, Willerson JT, Yeh ET. Modulation of vascular inflam-
mation in vitro and in vivo by peroxisome proliferator-activated recep-
tor-gamma activators. Circulation 2000;101:235-8.
42. Marx N, Sukhova G, Murphy C, Libby P, Plutzky J. Macrophages in
human atheroma contain PPARgamma: differentiation-dependent per-
oxisomal proliferator-activated receptor gamma(PPARgamma) expres-
sion and reduction of MMP-9 activity through PPARgamma activation
in mononuclear phagocytes in vitro. Am J Pathol 1998;153:17-23.
43. Mohanty P, Aljada A, Ghanim H et al. Evidence for a potent antiin-
flammatory effect of rosiglitazone. J Clin Endocrinol Metab 2004;89:
2728-35.
44. Haffner SM, Greenberg AS, Weston WM, Chen H, Williams K, Freed
MI. Effect of rosiglitazone treatment on nontraditional markers of car-
diovascular disease in patients with type 2 diabetes mellitus. Circulation
2002;106:679-84.
45. Marx N, Imhof A, Froehlich J et al. Effect of rosiglitazone treatment on
soluble CD40L in patients with type 2 diabetes and coronary artery dis-
ease. Circulation 2003;107:1954-7.
46. Chinetti G, Lestavel S, Bocher V et al. PPAR-alpha and PPAR-gamma
activators induce cholesterol removal from human macrophage foam
cells through stimulation of the ABCA1 pathway. PG - 53-8. Nat Med
2001;7:23-4.
47. Chen Z, Ishibashi S, Perrey S et al. Troglitazone inhibits atherosclerosis
in apolipoprotein E-knockout mice: pleiotropic effects on CD36
expression and HDL. Arterioscler Thromb Vasc Biol 2001;21:372-7.
48. Marx N, Schonbeck U, Lazar MA, Libby P, Plutzky J. Peroxisome prolif-
erator-activated receptor gamma activators inhibit gene expression and
migration in human vascular smooth muscle cells. Circ Res 1998;83:
1097-103.
49. Fukunaga Y, Itoh H, Doi K et al. Thiazolidinediones, peroxisome prolif-
erator-activated receptor gamma agonists, regulate endothelial cell
growth and secretion of vasoactive peptides. Atherosclerosis 2001;
158:113-19.
50. Collins AR, Meehan WP, Kintscher U et al. Troglitazone inhibits forma-
tion of early atherosclerotic lesions in diabetic and nondiabetic low
density lipoprotein receptor-deficient mice. Arterioscler Thromb Vasc
Biol 2001;21:365-71.
51. Li AC, Brown KK, Silvestre MJ, Willson TM, Palinski W, Glass CK.
Peroxisome proliferator-activated receptor gamma ligands inhibit
development of atherosclerosis in LDL receptor-deficient mice. J Clin
Invest 2000;106:523-31.
52. Koshiyama H, Shimono D, Kuwamura N, Minamikawa J, Nakamura Y.
Rapid communication: inhibitory effect of pioglitazone on carotid arte-
rial wall thickness in type 2 diabetes. J Clin Endocrinol Metab 2001;86:
3452-6.
53. Sidhu JS, Kaposzta Z, Markus HS, Kaski JC. Effect of rosiglitazone on
common carotid intima-media thickness progression in coronary artery
disease patients without diabetes mellitus. Arterioscler Thromb Vasc
Biol 2004;24:930-4.
54. Takagi T, Yamamuro A, Tamita K et al. Pioglitazone reduces neointimal
tissue proliferation after coronary stent implantation in patients with
type 2 diabetes mellitus: an intravascular ultrasound scanning study. Am
Heart J 2003;146:E5.
55. Takagi T, Akasaka T, Yamamuro A et al. Troglitazone reduces neointimal
tissue proliferation after coronary stent implantation in patients with
non-insulin dependent diabetes mellitus: a serial intravascular ultra-
sound study. J Am Coll Cardiol 2000;36:1529-35.
56. Choi D, Kim SK, Choi SH et al. Preventative effects of rosiglitazone on
restenosis after coronary stent implantation in patients with type 2 dia-
betes. Diabetes Care 2004;27:2654-60.
57. Marx N, Wohrle J, Nusser T et al. Pioglitazone reduces neointima vol-
ume after coronary stent implantation: a randomized, placebo-con-
trolled, double-blind trial in nondiabetic patients. Circulation 2005;
112:2792-8.
58. Haffner SM, Lehto S, Ronnemaa T, Pyorala K, Laakso M. Mortality from
coronary heart disease in subjects with type 2 diabetes and in nondia-
betic subjects with and without prior myocardial infarction. N Engl J
Med 1998;339:229-34.
59. McGuire DK, Emanuelsson H, Granger CB et al. Influence of diabetes
mellitus on clinical outcomes across the spectrum of acute coronary
syndromes. Findings from the GUSTO-IIb study. GUSTO IIb
Investigators. Eur Heart J 2000;21:1750-8.
60. Jokl R, Colwell JA. Arterial thrombosis and atherosclerosis in diabetes.
Diabetes Reviews 1997;5:316-30.
61.
Sobel BE. The potential influence of insulin and plasminogen activator
inhibitor type 1 on the formation of vulnerable atherosclerotic plaques
associated with type 2 diabetes. Proc Assoc Am Phys 1999;111:313-18.
62. Uemura S, Matsushita H, Li W et al. Diabetes mellitus enhances vas-
cular matrix metalloproteinase activity: role of oxidative stress. Circ Res
2001;88:1291-8.
REVIEW
VOLUME 3 ISSUE 2 .SEPTEMBER 2006 71
REVIEW
63. Ford ES. Body mass index, diabetes, and C-reactive protein among US.
adults. Diabetes Care 1999;22:1971-7.
64. Zirlik A, Leugers A, Lohrmann J et al. Direct attenuation of plasmino-
gen activator inhibitor type-1 expression in human adipose tissue by
thiazolidinediones. Thromb Haemost 2004;91:674-82.
65. Ghanim H, Garg R, Aljada A et al. Suppression of nuclear factor-
kappaB and stimulation of inhibitor kappaB by troglitazone: evidence
for an anti-inflammatory effect and a potential antiatherosclerotic effect
in the obese. J Clin Endocrinol Metab 2001;86:1306-12.
66. Pfutzner A, Marx N, Lubben G et al. Improvement of cardiovascular
risk markers by pioglitazone is independent from glycemic control:
results from the pioneer study. J Am Coll Cardiol 2005;45:1925-31.
67. Sidhu JS, Cowan D, Tooze JA, Kaski JC. Peroxisome proliferator-activat-
ed receptor-gamma agonist rosiglitazone reduces circulating platelet
activity in patients without diabetes mellitus who have coronary artery
disease. Am Heart J 2004;147:e25.
68. Yue Tl TL, Chen J, Bao W et al. In vivo myocardial protection from
ischemia/reperfusion injury by the peroxisome proliferator-activated
receptor-gamma agonist rosiglitazone. Circulation 2001;104:2588-94.
69. Khandoudi N, Delerive P, Berrebi-Bertrand I, Buckingham RE, Staels B,
Bril A. Rosiglitazone, a peroxisome proliferator-activated receptor-
gamma, inhibits the Jun NH(2)-terminal kinase/activating protein 1
pathway and protects the heart from ischemia/reperfusion injury.
Diabetes 2002;51:1507-14.
70. Liu HR, Tao L, Gao E et al. Anti-apoptotic effects of rosiglitazone in
hypercholesterolemic rabbits subjected to myocardial ischemia and
reperfusion. Cardiovasc Res 2004;62:135-44.
71. Charbonnel B, Dormandy J, Erdmann E, Massi-Benedetti M, Skene A.
The prospective pioglitazone clinical trial in macrovascular events
(PROactive): can pioglitazone reduce cardiovascular events in dia-
betes? Study design and baseline characteristics of 5238 patients.
Diabetes Care 2004;27:1647-53.
72. Patel C, Wyne KL, McGuire DK. Thiazolidinediones, peripheral oede-
ma and congestive heart failure: what is the evidence? Diabetes Vasc
Dis Res 2005;2:61-6.
73. Actos [prescribing Information]. Lincolnshire, IL: Takeda
Pharmaceuticals, 2005.
74. SmithKline Beecham. Avandia (rosiglitazone): summary of product
characteristics. Welwyn Garden City: SmithKline Beecham UK, 2000.
75. Nesto RW, Bell D, Bonow RO et al. Thiazolidinedione use, fluid reten-
tion, and congestive heart failure: a consensus statement from the
American Heart Association and American Diabetes Association.
Circulation 2003;108:2941-8.
76. Rezulin (troglitazone) [package insert]. Morris Plains, NJ: Parke-Davis,
Division of Warner-Lambert Co; July 1998.
77. Takeda. Pioglitazone: summary of product characteristics. High
Wycombe: Takeda UK, 2000.
78. Ghazzi MN, Perez JE, Antonucci TK et al. Cardiac and glycemic bene-
fits of troglitazone treatment in NIDDM. The Troglitazone Study Group.
Diabetes 1997;46:433-9.
79. Furuse Y, Ogino K, Shimoyama M, Sasaki N, Hisatome I. Ca(2+)-sensi-
tizing effect is involved in the positive inotropic effect of troglitazone. Br
J Pharmacol 2001;133:1307-13.
80. Shimoyama M, Ogino K, Tanaka Y, Ikeda T, Hisatome I. Hemodynamic
basis for the acute cardiac effects of troglitazone in isolated perfused rat
hearts. Diabetes 1999;48:609-15.
81. Shimabukuro M, Higa S, Shinzato T, Nagamine F, Komiya I, Takasu N.
Cardioprotective effects of troglitazone in streptozotocin-induced dia-
betic rats. Metabolism 1996;45:1168-73.
82. Yao L, Mizushige K, Noma T, Murakami K, Ohmori K, Matsuo H.
Improvement of left ventricular diastolic dynamics in prediabetic stage
of a type II diabetic rat model after troglitazone treatment. Angiology
2001;52:53-7.
83. Asakawa M, Takano H, Nagai T et al. Peroxisome proliferator-activated
receptor gamma plays a critical role in inhibition of cardiac hypertro-
phy in vitro and in vivo. Circulation 2002;105:1240-6.
84. Yamamoto K, Ohki R, Lee RT, Ikeda U, Shimada K. Peroxisome prolif-
erator-activated receptor gamma activators inhibit cardiac hypertrophy
in cardiac myocytes. Circulation 2001;104:1670-5.
85. Tsuji T, Mizushige K, Noma T et al. Pioglitazone improves left ventricu-
lar diastolic function and decreases collagen accumulation in predia-
betic stage of a type II diabetic rat. J Cardiovasc Pharmacol 2001;38:
868-74.
86. St John Sutton M, Rendell M, Dandona P et al. A comparison of the
effects of rosiglitazone and glyburide on cardiovascular function and
glycemic control in patients with type 2 diabetes. Diabetes Care 2002;
25:2058-64.
87. Masoudi FA, Inzucchi SE, Wang Y, Havranek EP, Foody JM, Krumholz
HM. Thiazolidinediones, metformin, and outcomes in older patients
with diabetes and heart failure: an observational study. Circulation
2005;111:583-90.
88. Tang WH, Francis GS, Hoogwerf BJ, Young JB. Fluid retention after ini-
tiation of thiazolidinedione therapy in diabetic patients with established
chronic heart failure. J Am Coll Cardiol 2003;41:1394-8.
Erratum: Effects of pioglitazone on lipid and lipoprotein
profiles in patients with type 2 diabetes and dyslipidaemia
after treatment conversion from rosiglitazone while continuing
stable statin therapy
PAULOS BERHANU, MARK S KIPNES, MEHMOOD A KHAN, ALFONSO T PEREZ, STUART F KUPFER,
ROBERT G SPANHEIMER, SELESHI DEMISSIE, PENNY R FLECK Diabetes Vasc Dis Res 2006;3:39–44
In the above article which was published in the May 2006 issue of the journal, the incorrect conversion factor
was inadvertently used for the conversion of triglyceride levels from mg/dL to mmol/L. Also the SI unit for
HsCRP is mg/L instead of mmol/L, as provided in table 1.
This has now been corrected and appears in the correct form on the website, www.dvdres.com
... Thiazolidinediones are used for glucose control in type 2 diabetes. Regarding atherosclerosis, they have been shown to have several beneficial pleiotropic effects on cardiovascular risk factors, namely improvement of insulin sensitivity, redistribution of fat from visceral to subcutaneous depot, reduction of blood pressure, and improvement of endothelial function [101]. ...
... Might trigger psoriasis (especially beta-blockers, less likely ACE inhibitors) Unknown effect on PASI Suppression of vasoconstriction, smooth muscle proliferation, connective tissue synthesis, and chemotaxis of monocytes by angiotensin II [8,88] Reduction in MACE [8] Aspirin Insufficient data Possible induction or exacerbation of psoriasis-insufficient data Antithrombotic effect [8] Used in the secondary prevention of atherosclerosis; Lowers the risk of nonfatal myocardial infarction and ischemic stroke [89] No reduction in all-cause or cardiovascular mortality [8,89] Anti-inflammatory action [95,113] ↓ IL-1β [93,94,97,98] Possible decrease in PASI [95] Improvement of insulin resistance [113] ↓ LDL and total cholesterol [113] Improvement of endothelial dysfunction [113] Improvement of function of vascular smooth muscle cells [113] Inhibition of cardiac remodeling [113] Reduction in MACE in patients with pre-diabetes [114] GLP-1 receptor agonists Anti-inflammatory action by ↓ IL-17 [98][99][100] Decrease in PASI [98][99][100] ↓ blood pressure [93] ↓ body weight [93] Improvement of dyslipidemia [93] ↑ NO [93] Inhibits of adhesion and procoagulant factors [96,97] Reduction of MACE in patients with type 2 diabetes [103] Thiazolidinediones Anti-inflammatory effects: Reduction of lymphocyte migration [115] Reduction of macrophage activation [115] Decrease in IL-17 expression [115] Modulation of monocyte cytokine secretion [7,95,115] Increase in PASI [7,95] Improvement of insulin sensitivity [101] Redistribution of fat from visceral to subcutaneous depot [101] ↓ blood pressure [101] Improvement of endothelial function [101] Reduction of MACE in patients with type 2 diabetes [101,107] SGLT-2 inhibitors Unknown-no data Insufficient data ...
... Might trigger psoriasis (especially beta-blockers, less likely ACE inhibitors) Unknown effect on PASI Suppression of vasoconstriction, smooth muscle proliferation, connective tissue synthesis, and chemotaxis of monocytes by angiotensin II [8,88] Reduction in MACE [8] Aspirin Insufficient data Possible induction or exacerbation of psoriasis-insufficient data Antithrombotic effect [8] Used in the secondary prevention of atherosclerosis; Lowers the risk of nonfatal myocardial infarction and ischemic stroke [89] No reduction in all-cause or cardiovascular mortality [8,89] Anti-inflammatory action [95,113] ↓ IL-1β [93,94,97,98] Possible decrease in PASI [95] Improvement of insulin resistance [113] ↓ LDL and total cholesterol [113] Improvement of endothelial dysfunction [113] Improvement of function of vascular smooth muscle cells [113] Inhibition of cardiac remodeling [113] Reduction in MACE in patients with pre-diabetes [114] GLP-1 receptor agonists Anti-inflammatory action by ↓ IL-17 [98][99][100] Decrease in PASI [98][99][100] ↓ blood pressure [93] ↓ body weight [93] Improvement of dyslipidemia [93] ↑ NO [93] Inhibits of adhesion and procoagulant factors [96,97] Reduction of MACE in patients with type 2 diabetes [103] Thiazolidinediones Anti-inflammatory effects: Reduction of lymphocyte migration [115] Reduction of macrophage activation [115] Decrease in IL-17 expression [115] Modulation of monocyte cytokine secretion [7,95,115] Increase in PASI [7,95] Improvement of insulin sensitivity [101] Redistribution of fat from visceral to subcutaneous depot [101] ↓ blood pressure [101] Improvement of endothelial function [101] Reduction of MACE in patients with type 2 diabetes [101,107] SGLT-2 inhibitors Unknown-no data Insufficient data ...
Article
Full-text available
Psoriasis is a chronic systemic inflammatory disease. Due to systemic inflammation, it is associated with many comorbidities. Among them, cardiovascular diseases represent the most common causes of morbidity and mortality in this population. Therefore, physicians treating patients with psoriasis should keep in mind that, as important as the treatment of psoriasis, awareness of cardiovascular risk deserves additional attention. Thus, in parallel with psoriasis treatment, a car-diovascular risk assessment must also be performed and addressed accordingly. In addition to encouraging non-pharmacologic strategies for a healthy lifestyle, physicians should be familiar with different pharmacologic options that can target psoriasis and reduce cardiovascular risk. In the present article, we present the pathophysiological mechanisms of the psoriasis and cardiometabolic interplay, our view on the interaction of psoriasis and cardiovascular disease, review the athero-sclerotic effect of therapeutic options used in psoriasis, and vice versa, i.e., what the effect of medications used in the prevention of atherosclerosis could be on psoriasis.
... TZDs, the insulin-sensitizing agents, are known to have a strong protective effect on atherosclerosis-driven events such as cardiac or cerebrovascular disease [21]. Zhang et al. [6] conducted a meta-analysis of 130,854 diabetic patients in seven studies to evaluate TZD's effect on AF development. ...
Article
Full-text available
Background and Objectives: Type 2 diabetes (T2D) is an independent risk factor for the development of atrial fibrillation (AF). Sodium-glucose cotransporter-2 inhibitors (SGLT-2i) have recently been shown to decrease the incidence of AF through several mechanisms, including the reduction of atrial dilatation via diuresis and the lowering of body weight. In observational studies of diabetic patients, the use of thiazolidinedione (TZD) was found to have a protective effect on new-onset AF. In this study, we aimed to compare the effect of SGLT-2i and TZD on the risk of AF in patients with T2D. Methods: We enrolled 69,122 patients newly prescribed SGLT-2i and 94,262 patients prescribed TZD from January 2014 to December 2018, using the Korean National Health Insurance Service database. We compared new-onset AF events (hospitalizations and outpatient events) in SGLT-2i and TZD groups after having taken medication for greater than 90 days. Results: During a mean follow-up of 1.8 years, 397 (0.72%) new-onset AF events occurred in the SGLT-2i group and 432 (0.79%) events in the TZD group following propensity score matching (each group n = 54,993). The hazard ratio (HR) of AF was 0.918 (95% confidence interval: 0.783–1.076, p = 0.29) in SGLT-2i-treated patients compared with TZD-treated patients. Conclusions: In this study, the risk of new-onset AF is comparable in patients treated with SGLT-2i and TZD in T2D. Either SGLT-2i or TZD would be a reasonable choice for T2D patients who are at risk for AF.
... Glitazones may also protect arterial wall properties. Experimental studies have shown that rosiglitazone increases NO-mediated flow-dependent dilation of coronary arterioles by reducing vascular superoxide production which mediates increased production of nitric oxide (NO) and reduction of endothelin-1 levels [178,179]. Accordingly, thiazolidinediones may improve large artery elastic properties and reduces pulse wave velocity (PWV) [180]. ...
Article
Full-text available
Heart failure (HF) is a common cardiac syndrome, whose pathophysiology involves complex mechanisms, some of which remain unknown. Diabetes mellitus (DM) constitutes not only a glucose metabolic disorder accompanied by insulin resistance but also a risk factor for cardiovascular disease and HF. During the last years though emerging data set up, a bidirectional interrelationship between these two entities. In the case of DM impaired calcium homeostasis, free fatty acid metabolism, redox state, and advance glycation end products may accelerate cardiac dysfunction. On the other hand, when HF exists, hypoperfusion of the liver and pancreas, b-blocker and diuretic treatment, and autonomic nervous system dysfunction may cause impairment of glucose metabolism. These molecular pathways may be used as therapeutic targets for novel antidiabetic agents. Peroxisome proliferator-activated receptors (PPARs) not only improve insulin resistance and glucose and lipid metabolism but also manifest a diversity of actions directly or indirectly associated with systolic or diastolic performance of left ventricle and symptoms of HF. Interestingly, they may beneficially affect remodeling of the left ventricle, fibrosis, and diastolic performance but they may cause impaired water handing, sodium retention, and decompensation of HF which should be taken into consideration in the management of patients with DM. In this review article, we present the pathophysiological data linking HF with DM and we focus on the molecular mechanisms of PPARs agonists in left ventricle systolic and diastolic performance providing useful insights in the molecular mechanism of this class of metabolically active regiments.
... Since PPARγ activation upregulates the regulators of cholesterol efflux in macrophages [3,4] and reduces inflammation in atheroma by inhibiting nuclear factor-kappa B activity [4][5][6][7][8], this pathway could be an attractive target for treating atherosclerosis. While a few reports have demonstrated that PPARγ agonists could stabilize high-risk plaques [9][10][11][12], PPARγ agonist therapy for atherosclerosis is still challenging due to adverse systemic effects including weight gain, edema, congestive heart failure, and bone fracture [13][14][15]. ...
Article
Full-text available
Rationale: Atherosclerotic plaque is a chronic inflammatory disorder involving lipid accumulation within arterial walls. In particular, macrophages mediate plaque progression and rupture. While PPARγ agonist is known to have favorable pleiotropic effects on atherogenesis, its clinical application has been very limited due to undesirable systemic effects. We hypothesized that the specific delivery of a PPARγ agonist to inflamed plaques could reduce plaque burden and inflammation without systemic adverse effects. Methods: Herein, we newly developed a macrophage mannose receptor (MMR)-targeted biocompatible nanocarrier loaded with lobeglitazone (MMR-Lobe), which is able to specifically activate PPARγ pathways within inflamed high-risk plaques, and investigated its anti-atherogenic and anti-inflammatory effects both in in vitro and in vivo experiments. Results: MMR-Lobe had a high affinity to macrophage foam cells, and it could efficiently promote cholesterol efflux via LXRα-, ABCA1, and ABCG1 dependent pathways, and inhibit plaque protease expression. Using in vivo serial optical imaging of carotid artery, MMR-Lobe markedly reduced both plaque burden and inflammation in atherogenic mice without undesirable systemic effects. Comprehensive analysis of en face aorta by ex vivo imaging and immunostaining well corroborated the in vivo findings. Conclusion: MMR-Lobe was able to activate PPARγ pathways within high-risk plaques and effectively reduce both plaque burden and inflammation. This novel targetable PPARγ activation in macrophages could be a promising therapeutic strategy for high-risk plaques.
... Thiazolidinediones (TZDs) act by binding to "nuclear peroxisome proliferator activated receptor-gamma" (PPAR-), which is chiefly expressed in fatty tissue, and mediate their effects by activating the transcription of the genes that influence adipocyte differentiation as well as glucose and lipid metabolism [21][22][23]. TZDs decrease the triglyceride concentration in -cells, leading to improved -cell function. TZDs, apart from their direct effect on fatty tissue, might influence the release of adipocyte-derived signal factors that determine the insulin sensitivity of muscles, such as FFAs, adiponectin, leptin, and tumor necrosis factor-alpha (TNF-). ...
Article
Full-text available
Aim: To investigate the efficacy of combined therapy of insulin and rosiglitazone on metabolic and inflammatory parameters, insulin sensitivity, and adipocytokine levels in patients with type 1 diabetes mellitus (type 1 DM). Material and methods: A total of 61 adults with type 1 DM were randomly and prospectively assigned in open-label fashion to take insulin and rosiglitazone 4 mg/day (n = 30) or insulin alone (n = 31) for a period of 18 weeks while undergoing insulin therapy without acute metabolic complications. Results: Combination therapy did not significantly improve metabolic and inflammatory parameters, insulin sensitivity, and adiponectin levels. While leptin and resistin levels decreased in both groups (group 1: resistin 6.96 ± 3.06 to 4.99 ± 2.64, P = 0.006; leptin 25.8 ± 17.6 to 20.1 ± 12.55, P = 0.006; group 2: resistin 7.16 ± 2.30 to 5.57 ± 2.48, P = 0.031; leptin 16.72 ± 16.1 to 14.0 ± 13.4, P = 0.007) Hgb and fibrinogen levels decreased only in group 1 (Hgb 13.72 ± 1.98 to 13.16 ± 1.98, P = 0.015, and fibrinogen 4.00 ± 1.08 to 3.46 ± 0.90, P = 0.002). Patients in both groups showed weight gain and the incidence of hypoglycemia was not lower. Discussion: The diverse favorable effects of TZDs were not fully experienced in patients with type 1 DM. These results are suggesting that insulin sensitizing and anti-inflammatory characteristics of TZDs were likely to be more pronounced in patients who were not totally devoid of endogenous insulin secretion.
Article
Full-text available
Hypertension characterized by an elevated blood pressure is a cardiovascular disease that afflicts greater than one in every three adults worldwide. Nuclear receptors are large superfamily of DNA-binding transcription factors that target genes to regulate metabolic and cardiovascular function. Drugs have been developed for nuclear receptors such as peroxisome proliferator-activated receptors (PPARα and PPARγ) and farnesoid X receptor (FXR). PPARα, PPARγ, and FXR agonists are used clinically to treat lipid disorders and metabolic diseases. Evidence from clinical studies and animal hypertension models have demonstrated that PPARα, PPARγ, and FXR agonism can lower blood pressure and decrease end organ damage which could be useful for the treatment of hypertension in patients with metabolic diseases. Unfortunately, PPAR and FXR agonists have unwanted clinical side effects. There have been recent developments to limit side effects for PPAR and FXR agonists. Combining PPAR and FXR agonism with soluble epoxide hydrolase (sEH) inhibition or Takeda G protein receptor 5 (TGR5) agonism has been demonstrated in preclinical studies to have actions that would decrease clinical side effects. In addition, these dual modulating drugs have been demonstrated in preclinical studies to have blood pressure lowering, anti-fibrotic, and anti-inflammatory actions. There is now an opportunity to thoroughly test these novel dual modulators in animal models of hypertension associated with metabolic diseases. In particular, these newly developed dual modulating PPAR and FXR drugs could be beneficial for the treatment of metabolic diseases, organ fibrosis, and hypertension.
Article
Full-text available
Endothelial dysfunction and core illnesses are now among the top causes of death on a global scale. Atherogenesis is a prominent risk factor for adverse vascular blockage diseases. People who have hyperglycemia and hyperlipidemia become more prone to developing cerebro-cardiovascular disease and lower limb disorders. Additionally, diabetic metabolic syndrome additionally alters the extracellular signal-regulated kinase and immunosignal pathways, as well as the activation of vascular endothelial growth factor, nuclear factors and cell migration all of which contribute to pathogenesis. The creation of fibrous and fatty lesions in the intimal layer of an artery is referred to as atherogenesis associated vascular problems. In this study, atherogenic lipid particles, markers and inflammatory signaling pathways related to hyperglycemia and hyperlipidemia complications of atherosclerosis are discussed which will aid in the development of a new strategy to treat the macro-vascular complication for future research and treatment.
Article
Despite the successful development of new therapies for the treatment of type 2 diabetes, such as glucagon-like peptide-1 (GLP-1) receptor agonists and sodium-glucose cotransporter-2 inhibitors, the search for novel treatment options that can provide better glycaemic control and at reduce complications is a continuous effort. The present Review aims to present an overview of novel targets and mechanisms and focuses on glucose-lowering effects guiding this search and developments. We discuss not only novel developments of insulin therapy (eg, so-called smart insulin preparation with a glucose-dependent mode of action), but also a group of drug classes for which extensive research efforts have not been rewarded with obvious clinical impact. We discuss the potential clinical use of the salutary adipokine adiponectin and the hepatokine fibroblast growth factor (FGF) 21, among others. A GLP-1 peptide receptor agonist (semaglutide) is now available for oral absorption, and small molecules activating GLP-1 receptors appear on the horizon. Bariatric surgery and its accompanying changes in the gut hormonal milieu offer a background for unimolecular peptides interacting with two or more receptors (for GLP-1, glucose-dependent insulinotropic polypeptide, glucagon, and peptide YY) and provide more substantial glycaemic control and bodyweight reduction compared with selective GLP-1 receptor agonists. These and additional approaches will help expand the toolbox of effective medications needed for optimising the treatment of well delineated subgroups of type 2 diabetes or help develop personalised approaches for glucose-lowering drugs based on individual characteristics of our patients.
Article
Atherosclerosis is now the major cause of mortality and morbidity worldwide. Formation of macrophage-derived foam cells is a hallmark of atherosclerosis, which is regulated by cholesterol uptake, intracellular metabolism, and efflux. PPARγ-LXRα-ABCA1/ABCG1 pathway plays an important part in regulating cholesterol efflux and this pathway could be a promising target for treating atherosclerosis. However, due to undesirable systemic effects, PPARγ agonist therapy for atherosclerosis remains challenging. Many traditional Chinese medicine has been well accepted and applied in atherosclerosis treatment. Yin-xing-tong-mai decoction (YXTMD) has been applied for treating atherosclerosis for decades. However, the mechanism remains to be explored. Here, we showed that YXTMD effectively attenuated atherosclerosis in ApoE-/- mice. YXTMD increased cholesterol efflux of foam cell by upregulation of ABCA1 and ABCG1 in vivo and in vitro. Through bioinformatic analysis and experimental validation, we found that PPARγ was an important downstream effector of YXTMD in macrophages. Reduction of PPARγ significantly decreased LXRα, ABCA1, and ABCG1 expression in macrophages, with reduced cholesterol efflux. In conclusion, these findings confirmed that YXTMD attenuated atherosclerosis by activating the PPARγ-LXRα- ABCA1/ABCG1 pathway to enhance cholesterol efflux.
Article
Thiazolidinediones (or glitazones) are a new class of oral hypoglycemizing drugs which act by reducing insulin resistance in adipose tissue, skeletal muscle and liver in type 2 diabetes mellitus. Drugs currently on the market belonging to this family are rosiglitazone and pioglitazone. A specific advantage of this class of drugs is the ability to maintain the hypoglycemizing effect and an acceptable control long term, thus avoiding the introduction of insulin therapy. The action of glitazones goes beyond the control of hyperglycaemia and is expressed specifically in the ability to modify other cardiovascular risk factors. The improvement in insulin sensitivity induced by thiazolidinediones could in fact translate into a more favourable cardiovascular risk profile in patients with type 2 diabetes. This work takes into consideration several studies on the tolerability of these drugs, highlighting the differences and the points in common and evaluating their long term efficacy and safety. According to these studies, thiazolidinediones are generally well-tolerated drugs, both in monotherapy and in combination with other oral antidiabetic drugs.
Article
Full-text available
This study was conducted to evaluate whether treatment of normal and diabetic rat hearts with rosiglitazone, a high-affinity ligand of the peroxisome proliferator-activated receptor-gamma (PPAR-gamma) used for the treatment of type 2 diabetes, improves postischemic functional recovery. The effects of acute rosiglitazone administration were investigated using working hearts isolated from normal rat or rats diabetic for 4 weeks after streptozotocin (STZ) injection. Hearts were subjected to 30 min of normothermic, zero-flow ischemia followed by 30-min reperfusion. Rosiglitazone (1 mumol/l) administered before ischemia had no effect on cardiac function during baseline perfusion, but it significantly improved aortic flow during reperfusion in both normal and diabetic hearts. In a chronic protocol in which rosiglitazone was given by daily gavage (10 mumol/kg body wt) immediately after STZ injection, rosiglitazone also prevented postischemic injury and significantly improved functional recovery. Using Western immunoblotting, it was demonstrated that the acute cardioprotective effect of rosiglitazone is associated with an inhibition of Jun NH2-terminal kinase phosphorylation in both normal and diabetic rat hearts. Furthermore, rosiglitazone also inhibited activating protein-1 DNA-binding activity. These data, demonstrating that rosiglitazone limits postischemic injury in isolated hearts, suggest an important function for PPAR-gamma in the heart.
Article
Enhancement of multiple thrombogenic constituents, including platelet activation, hypercoagulation, impaired fibrinolysis, abnormal hemorheology, and altered endothelial metabolism, has been found in diabetes. Specific thrombogenic abnormalities in diabetes and their association with the development of macrovascular disease are reviewed in this article. Proposed pathogenetic mechanisms underlying the prothrombotic tendency in diabetes include not only changes of insulin, glucose, and lipoprotein metabolism, but also modified immune and inflammatory defenses and genetic variability. Many hemostatic disorders in diabetes, however, are also known to be a consequence of established vascular lesions. Standard diabetes management can help to correct the prothrombotic imbalance in diabetes, although additional antithrombotic therapy may be necessary. Meta-analysis of antiplatelet trials supports the use of low-dose aspirin in all diabetic individuals with high vascular risk. New clinical trials may justify other approaches, such as using new antiplatelet agents, specific anticoagulants, antioxidants, drugs to lower fibrinogen and reduce overproduction of plasminogen activator inhibitors, or agents that stimulate endothelium-associated fibrinolysis. Future research may identify different candidate genes for atherosclerosis within the hemostatic system and expand evolving vascular gene therapy.
Article
Atherosclerosis, formerly considered a bland lipid storage disease, actually involves an ongoing inflammatory response. Recent advances in basic science have established a fundamental role for inflammation in mediating all stages of this disease from initiation through progression and, ultimately, the thrombotic complications of atherosclerosis. These new findings provide important links between risk factors and the mechanisms of atherogenesis. Clinical studies have shown that this emerging biology of inflammation in atherosclerosis applies directly to human patients. Elevation in markers of inflammation predicts outcomes of patients with acute coronary syndromes, independently of myocardial damage. In addition, low-grade chronic inflammation, as indicated by levels of the inflammatory marker C-reactive protein, prospectively defines risk of atherosclerotic complications, thus adding to prognostic information provided by traditional risk factors. Moreover, certain treatments that reduce coronary risk also limit inflammation. In the case of lipid lowering with statins, this anti-inflammatory effect does not appear to correlate with reduction in low-density lipoprotein levels. These new insights into inflammation in atherosclerosis not only increase our understanding of this disease, but also have practical clinical applications in risk stratification and targeting of therapy for this scourge of growing worldwide importance.
Article
OBJECTIVES We sought to define the characteristics of fluid retention after thiazolidinedione (TZD) initiation in patients with established heart failure (HF). BACKGROUND Fluid retention associated with the use of TZD is commonly attributed to exacerbation of HF, which has led to the proscription of these potentially useful agents in patients with chronic HF. METHODS We examined 111 consecutive diabetic patients with chronic systolic HF who were treated with TZD from January 1999 to June 2001. A retrospective chart review was performed to determine the incidence of fluid retention in this cohort. Physical signs of fluid retention were compared between TZD users and an age- and gender-matched control group of diabetic, non-TZD users with chronic HF who had fluid retention. Baseline clinical and echocardiographic data were compared between TZD users with and without fluid retention. RESULTS Nineteen TZD users (17.1%) developed fluid retention, which reversed after drug withdrawal and presented predominantly as peripheral and not central edema. Comparing patients in the upper and lower tertiles of weight gain, more female patients and insulin users developed TZD-related fluid retention. However, there were no differences in the baseline New York Heart Association functional class or echocardiographic severity of cardiac dysfunction. CONCLUSIONS Although fluid retention after treatment with TZD in diabetic patients with chronic systolic HF occurs, the mechanism is undefined. Fluid retention related to TZD tends to be peripheral and is usually reversible after drug withdrawal. No direct association between the risk of fluid retention and the baseline degree of severity of HF was observed. (C) 2003 by the American College of Cardiology Foundation.
Article
Objective— Peroxisome proliferator–activated receptors (PPARs) may modulate in vitro the vascular production of vasoactive peptides such as endothelin-1 (ET-1). Thus, we investigated in vivo the interaction between PPARs and ET-1 in deoxycorticosterone acetate (DOCA)–salt rats that overexpress vascular ET-1. Methods and Results— Unilaterally nephrectomized 16-week-old Sprague-Dawley rats (Uni-Nx) were divided into 4 groups (n=6 each): control group, DOCA-salt group, DOCA-salt+PPAR-γ activator (rosiglitazone, 5 mg · kg⁻¹ · d⁻¹), or DOCA-salt+PPAR-α activator (fenofibrate, 100 mg · kg⁻¹ · d⁻¹). Systolic blood pressure was significantly increased in the DOCA-salt group (240±11 vs 121±2 mm Hg in Uni-Nx, P<0.01). Progression of hypertension was partially prevented by coadministration of rosiglitazone (172±3 mm Hg vs DOCA-salt, P<0.05) but not by fenofibrate. Both PPAR activators abrogated the increase in prepro-ET-1 mRNA content in the mesenteric vasculature of DOCA-salt rats. The media-to-lumen ratio was increased in DOCA-salt rats (10.3±0.9% vs 4.9±0.5% in Uni-Nx rats, P<0.01). Rosiglitazone and fenofibrate prevented the hypertrophic remodeling observed in DOCA-salt rats without affecting vascular stiffness. Rosiglitazone but not fenofibrate prevented endothelial dysfunction in pressurized mesenteric arteries. Finally, both rosiglitazone and fenofibrate prevented the vascular increase in superoxide anion production induced in DOCA-salt animals. Conclusions— PPAR-α and -γ activators were able to modulate endogenous production of ET-1 and had beneficial vascular effects in endothelin-dependent hypertension.
Article
Mononuclear phagocytes play an important role in atherosclerosis and its sequela plaque rupture in part by their secretion of matrix metalloproteinases (MMPs), including MMP-9. Peroxisomal proliferator-activated receptor γ (PPARγ), a transcription factor in the nuclear receptor superfamily, regulates gene expression in response to various activators, including 15-deoxy-Δ12,14-prostaglandin J2 and the antidiabetic agent troglitazone. The role of PPARγ in human atherosclerosis is unexplored. We report here that monocytes/macrophages in human atherosclerotic lesions (n = 12) express immunostainable PPARγ. Normal artery specimens (n = 6) reveal minimal immunoreactive PPARγ. Human monocytes and monocyte-derived macrophages cultured for 6 days in 5% human serum expressed PPARγ mRNA and protein by reverse transcription-polymerase chain reaction and Western blotting, respectively. In addition, PPARγ mRNA expression in U937 cells increased during phorbol 12-myristate 13 acetate-induced differentiation. Stimulation of PPARγ with troglitazone or 15-deoxy-Δ12,14-prostaglandin J2 in human monocyte-derived macrophages inhibited MMP-9 gelatinolytic activity in a concentration-dependent fashion as revealed by zymography. This inhibition correlates with decreased MMP-9 secretion as determined by Western blotting. Thus, PPARγ is present in macrophages in human atherosclerotic lesions and may regulate expression and activity of MMP-9, an enzyme implicated in plaque rupture. PPARγ is likely to be an important regulator of monocyte/macrophage function with relevance for human atherosclerotic disease.