Anda di halaman 1dari 10

Diabetes and Vascular

Disease Research
http://dvr.sagepub.com/

Review article: Platelet abnormalities in diabetes mellitus


Jos Luis Ferreiro, Joan Antoni Gmez-Hospital and Dominick J Angiolillo
Diabetes and Vascular Disease Research 2010 7: 251 originally published online 4 October 2010
DOI: 10.1177/1479164110383994
The online version of this article can be found at:
http://dvr.sagepub.com/content/7/4/251

Published by:
http://www.sagepublications.com

Additional services and information for Diabetes and Vascular Disease Research can be found at:
Email Alerts: http://dvr.sagepub.com/cgi/alerts
Subscriptions: http://dvr.sagepub.com/subscriptions
Reprints: http://www.sagepub.com/journalsReprints.nav
Permissions: http://www.sagepub.com/journalsPermissions.nav
Citations: http://dvr.sagepub.com/content/7/4/251.refs.html

>> Version of Record - Nov 29, 2010


OnlineFirst Version of Record - Oct 13, 2010
OnlineFirst Version of Record - Oct 4, 2010
What is This?

Downloaded from dvr.sagepub.com by guest on January 14, 2014

Review article

Platelet abnormalities in diabetes mellitus

Diabetes & Vascular Disease Research


7(4) 251259
The Author(s) 2010
Reprints and permission: sagepub.
co.uk/journalsPermissions.nav
DOI: 10.1177/1479164110383994
dvr.sagepub.com

Jos Luis Ferreiro1,2, Joan Antoni Gmez-Hospital1 and


Dominick J Angiolillo3

Abstract
Patients with diabetes mellitus (DM) have accelerated atherosclerosis, which is the main underlying factor contributing
to the high risk of atherothrombotic events in these patients. Atherothrombotic complications are the leading cause of
morbidity and mortality in patients with DM. Among factors contributing to the prothrombotic condition which characterise patients with DM, platelet hyperreactivity plays a pivotal role. Platelets of DM patients are characterised by
dysregulation of several signalling pathways leading to intensified adhesion, activation and aggregation. Multiple mechanisms
are involved in platelet dysfunction of patients with DM, which can be categorised as follows: a) hyperglycaemia, b) insulin
deficiency and resistance, c) associated metabolic conditions, and d) other cellular abnormalities.The present manuscript aims
to provide an overview on the current status of knowledge on platelet abnormalities that characterise patients with DM.
Keywords
Cardiovascular disease, diabetes mellitus, platelets

Introduction: the diabetic platelet


Accelerated atherosclerosis is the main underlying factor
contributing to the high risk of atherothrombotic events in
patients with diabetes mellitus (DM). Cardiovascular disease, particularly coronary artery disease (CAD), often presenting as an acute coronary syndrome (ACS), is the leading
cause of morbidity and mortality in patients with DM.1 Of
note, DM patients without any history of CAD have the
same cardiac mortality risk as non-DM patients with a history of myocardial infarction (MI).2 In addition, cardiovascular disease has also worse prognosis in patients with DM
as they have a higher risk of complications and recurrent
atherothrombotic events than non-DM patients.3 In fact,
DM is a strong independent predictor of short- and longterm recurrent ischaemic events, including mortality, in an
ACS scenario.4,5 Further, the concomitant presence of cardiovascular risk factors and comorbidities that negatively
impact the outcomes of ACS is higher in DM patients.6
Several factors contribute to the prothrombotic condition that characterises patients with DM, such as the following: increased coagulation, impaired fibrinolysis,
endothelial dysfunction and platelet hyperreactivity.7-9 The
latter is of particular interest, since platelets play a key role
in the formation, development and sustainment of thrombi,
which are platelet-driven processes.10 Platelets of patients
with DM are characterised by dysregulation of several
signalling pathways, and have been proven to be hyperreactive with intensified adhesion, activation and aggregation.7,11-14 Such a hyperreactive platelet phenotype may

contribute to the higher proportion of DM patients with


inadequate response to antiplatelet agents compared with
non-DM subjects.15,16 The role of antiplatelet agents in
patients with DM will be developed in detail elsewhere in
this supplement.
Multiple mechanisms caused by metabolic and cellular
abnormalities have been suggested to play a role in the
increased platelet reactivity observed in patients with DM.
These mechanisms can be grouped together into the following aetiopathogenic categories: a) hyperglycaemia, b) insulin deficiency and resistance, c) associated metabolic
conditions, and d) other cellular abnormalities (Figure 1).17
The aim of this paper is to provide an overview on the
current status of knowledge on platelet abnormalities which
characterise DM patients and to analyse the evidence
regarding their implications in platelet reactivity.

IDIBELL-Hospital Universitari de Bellvitge, Department of Cardiology,


Interventional Cardiology Unit, LHospitalet de Llobregat, Barcelona,
Spain
2
IDIBELL-Hosptal Universitari de Bellvitge, Cardiovascular Research Lab,
LHospitalet de Llobregat, Barcelona, Spain
3
University of Florida College of Medicine-Jacksonville, Jacksonville,
Florida, USA
1

Corresponding author:
Dominick J Angiolillo, University of Florida College of MedicineJacksonville, 655 West 8th Street, Jacksonville, Florida, 32209, USA.
Email: dominick.angiolillo@jax.ufl.edu

Downloaded from dvr.sagepub.com by guest on January 14, 2014

252

Diabetes and Vascular Disease Research 7(4)

Figure 1. Mechanisms contributing to platelet dysfunction in patients with diabetes mellitus.


Platelet dysfunction in patients with diabetes mellitus (DM) is caused by several mechanisms, such as those due to hyperglycaemia,
insulin deficiency, associated metabolic conditions, and other cellular abnormalities. Hyperglycaemia may increase platelet reactivity
by glycating platelet surface proteins (impairing membrane fluidity and therefore increasing platelet adhesion), activating protein
kinase C (a mediator of platelet activation), inducing P-selectin (a surface adhesion protein) expression, and due to osmotic effect.
Insulin deficiency also plays an important role in platelet dysfunction by different mechanisms. Some of them have been suggested
to be IRS-dependent, such as the increased intracellular calcium concentration leading to enhanced platelet degranulation and
aggregation, while other factors are not dependent of IRS, such as the impaired response to NO and PGI2, which enhances platelet
reactivity. Metabolic conditions frequently associated with DM may per se play a role on platelet hyperreactivity, including obesity,
dyslipidaemia and enhanced systemic inflammation. Obesity, in addition to being associated with insulin resistance, contributes
to platelet dysfunction, mostly in terms of adhesion and activation, due to mechanisms such as increased cytosolic calcium
concentration and augmented oxidative stress. Abnormalities of lipidic profile, particularly hypertriglyceridaemia, also affect
platelet reactivity by different mechanisms, which include inducing endothelial dysfunction. Endothelial dysfunction is another
characteristic feature of DM, which enhances platelet reactivity by decreasing the production of NO and PGI2 and contributes to
the prothrombotic state through an increased production of TF. Patients with DM present other platelet abnormalities that can
enhance platelet adhesion and activation, such as the following: increased expression of surface proteins (P-selectin and GP IIb/IIIa),
augmented cytosolic calcium concentration, upregulation of P2Y12 signalling, increased platelet turnover, and oxidative stress, which
provokes an overproduction of reactive oxygen and nitrogen species.
ADP: adenosine diphosphate, Ca++: calcium, GP: glycoprotein, IRS: insulin receptor substrate, NO: nitric oxide, PGI2: prostacycline, PKC: protein kinase
C, ROS/NOS: reactive oxygen and nitrogen species, TF: tissue factor.
Reproduced from Ferreiro JL and Angiolillo DJ. Diabetes and anti-platelet therapy in acute coronary syndrome. Circulation 2010 (in press).17

Downloaded from dvr.sagepub.com by guest on January 14, 2014

253

Ferreiro et al.

Hyperglycaemia
As per the definition of the American Diabetes Association,
Diabetes is a group of metabolic diseases characterised by
hyperglycaemia resulting from defects in insulin secretion,
insulin action, or both.18 Hyperglycaemia not only defines
DM but also plays an independent and important role in the
blood abnormalities leading to a prothrombotic state in DM
patients.19,20
Platelets from patients with type-2 DM have increased
expression of adhesion molecules. A study by Eibl and colleagues showed that platelets from DM patients have a
greater expression of platelet activation markers compared
with an age-matched non-diabetic control group.21 After
improving metabolic control during 3months, a significant
decrease in expression of platelet activation markers
(CD31, CD49b, CD62P and CD63) was noted. Further, a
significant correlation between CD62P, CD63 and HbA1C
levels observed. These findings suggest that improvement
of glycaemic control may have a beneficial effect on platelet adhesion and subsequent activation.21 Expression of
platelet surface receptors is also increased in DM patients.
In particular, expression of glycoprotein (GP) Ib and GP
IIb/IIIa, which respectively mediate binding to von
Willebrand factor and platelet-fibrin interaction, is augmented in DM patients.13,14,16
Acute hyperglycaemia results in increased platelet activation, as documented by elevated levels of surface adhesion molecules such as P-selectin22 and soluble markers of
platelet activation (e.g. soluble P-selectin and CD40 ligand).23,24 Further, levels of HbA1C and fasting glucose have
been correlated to P-selectin expression in type-2 DM
patients undergoing coronary angioplasty,25 which suggests
again that an improved glycaemic control may reduce
platelet reactivity.
In line with the above mentioned laboratory findings,
there are some clinical data that support the concept that
glucose-lowering therapy to improve metabolic control is
beneficial in DM patients with atherothrombotic complications. In the setting of ACS, the DIGAMI (Diabetes
Mellitus, Insulin Glucose Infusion in Acute Myocardial
Infarction) trial randomised patients with DM presenting
with an acute MI to intensive glucose-lowering treatment
(standard treatment plus insulinglucose infusion for 24 h
followed by multidose insulin therapy) or standard treatment. Mortality in the intensive treatment group was significantly lower than in the control group (33% vs. 44%;
RR=0.72 (95% confidence interval 0.550.92); p=0.011)
after 3.4years of follow-up.26 No differences in mortality or
morbidity (non-fatal reinfarction or stroke) were found in
the DIGAMI-2 trial among three different glucose-lowering strategies (group 1: glucoseinsulin infusion titrated by
glucose levels for at least 24 h followed by insulin-based
long-term glucose control; group 2: glucoseinsulin infusion titrated by glucose levels for at least 24hours followed

by standard glucose control; and group 3: routine metabolic


control according to local practice).27 Glucose-lowering
levels were similar among the three study groups, suggesting that the benefit of decreasing glucose levels is independent of the way it is achieved. However, the optimal
blood glucose level remains unknown. In fact, an excessive lowering of glucose has been proven to be harmful in
the ACCORD (Action to Control Cardiovascular Risk in
Diabetes Study Group) trial, which randomised DM patients
to receive an intensive glucose-lowering regimen (targeting
a glycated haemoglobin level <6.0%) or a standard regimen
(targeting a glycated haemoglobin level 7.07.9%). The
trial was interrupted prematurely after 3.5years of followup due to an increased mortality in the intensive therapy
group.28 The latter is in line with the results of the NICESUGAR (Normoglycemia in Intensive Care Evaluation
Survival Using Glucose Algorithm Regulation) trial that
randomised patients admitted to an intensive care unit to
undergo either intensive glucose control (target blood glucose range 4.56.0mmol/l) or conventional glucose control
(target of 10.0 mmol/l or less), in which mortality was
higher in the intensive glucose control group.29
Several mechanisms have been proposed to contribute to
the increased platelet reactivity caused by hyperglycaemia,
including the following: a) non-enzymatic glycation of
platelet membrane proteins that decreases membrane fluidity, which may increase platelet adhesion;30,31 b) osmotic
effect of glucose that activates platelet GP IIb/IIIa and
P-selectin expression;32 c) activation of protein kinase C, a
mediator of platelet activation;33 and d) glycation of circulating low-density lipoproteins (LDL), which increases
intracellular calcium concentration and nitric oxide (NO)
production.34 Although a description of abnormalities inducing a prothrombotic state in DM other than those which are
platelet specific goes beyond the scope of this paper, it is
worth mentioning that hyperglycaemia also promotes
thrombosis through plasmatic components. In particular,
hyperglycaemia activates coagulation by raising concentrations of procoagulant factors (e.g. tissue factor, von
Willebrand factor)35,36 and inhibits fibrinolysis by increasing
concentrations of plasminogen activator inhibitor.9

Insulin deficiency and resistance


The vast majority of DM patients can be assigned to two
broad aetiopathogenetic categories. Type-1 DM, which
accounts for only 510% of cases, results from a cellularmediated autoimmune destruction of the

-
cells of the pancreas, leading to an absolute deficiency of insulin secretion.
Type-2 DM, which accounts for around 9095% of DM
individuals, is caused by a combination of resistance to insulin action and an inadequate compensatory insulin secretory
response, usually having relative (rather than absolute) insulin deficiency.18

Downloaded from dvr.sagepub.com by guest on January 14, 2014

254

Diabetes and Vascular Disease Research 7(4)

Deficient insulin action is the cardinal factor for development of DM and clearly contributes to platelet dysfunction.37 Both insulin receptors (IR) and insulin-like growth
factor-1 (IGF-1) receptors are expressed in platelets.38,39
Among other effects in platelets, insulin increases surface
expression of adenylate cyclase-linked prostacyclin (PGI2)
receptor and induces the release of plasminogen activator.40,41 However, IR expression is relatively low as the
majority of its subunits heterodimerise with those of IGF-1
receptor to form insulin/IGF-1 receptor hybrids, which
eagerly bind IGF-1, but not insulin.39 IGF-1 is present in
alpha granules of platelets and high levels of its functional
receptor are expressed on the platelet surface, which may
contribute to the amplification of platelet responses. The
pathways through which IGF-1 may modulate platelet
function, however, are currently not fully elucidated. IGF-1
stimulation of platelets results in dose-dependent phosphorylation of the IGF receptor and in tyrosine phosphorylation of insulin receptor substrate-1 (IRS-1) and IRS-2,
stimulating their subsequent binding with the p85 subunit
of phosphoinositide-3 kinase (PI3K), leading to phosphorylation of protein kinase B, which is involved in several
cellular responses to insulin and IGF-1, including modulation of platelet reactivity.42
Several anomalies in insulin-mediated signalling pathways, which can be classified as IRS-dependent and independent factors, have been suggested to contribute to the
hampered or abolished platelet-inhibitory effect observed in
patients with insulin resistance.37,43 Among IRS-dependent
factors, insulin resistance causes an increase in intracellular
calcium concentration that leads to augmented platelet
degranulation and aggregation.44 The exact mechanism by
which the increase in calcium concentration is generated
is not yet entirely known and a number of hypotheses
have been suggested, such as IRS binding to the calciumATPase of the sarcoplasmic reticulum in an insulinregulated fashion,45 or via inhibition of cyclic adenosine
monophosphate formation by the inhibitory G-protein of
adenylyl cyclase, Gi.46 Among IRS-independent pathways
involved in platelet dysfunction due to insulin resistance,
the role of reduced platelet sensitivity to NO and PGI2 is
noteworthy, since both molecules released by the endothelium retard platelet activation; thus, an impaired response is
associated with enhanced platelet reactivity.47,48 Further, a
recent study found correlation between improvement of
insulin sensitivity, measured by the homeostasis model
assessment (HOMA) of insulin resistance, and changes of
platelet function.49
The importance of insulin resistance in platelet hyperreactivity characteristic of patients with DM is highlighted by
recent studies with thiazolidinediones in which this group
of insulin sensitisers has shown a beneficial effect on platelet function. For instance, rosiglitazone therapy has been
associated in some preliminary studies with improved

sensitivity to NO and partial normalisation of intracellular


calcium concentrations in DM patients,50 as well as with
reduced P-selectin expression in non-DM patients with
CAD.51 There are controversial data regarding the suggested
benefit of insulin-sensitiser therapy over insulin-providing
therapy in terms of atherosclerosis progression and cardiovascular outcomes. The PERISCOPE (Pioglitazone Effect
on Regression of Intravascular Sonographic Coronary
Obstruction Prospective Evaluation) trial was a prospective,
randomised, multicentre, double-blind clinical trial that
compared the effect of pioglitazone, an insulin sensitiser,
versus glimepiride, an insulin secretagogue, on progression
of coronary atherosclerosis measured by intravascular
ultrasonography in patients with type-2 DM and CAD.52
Pioglitazone therapy was associated with a significantly
lower rate of atherosclerosis progression at 18 months,
when compared with glimepiride treatment. In a substudy
of the SYMPHONY (Sibrafiban vs Aspirin to Yield
Maximum Protection From Ischemic Heart Events Postacute Coronary Syndromes) and SYMPHONY-2 trials performed in DM patients after an ACS, insulin-sensitiser
therapy with biguanide and/or thiazolidinedione was associated with a significantly lower rate of cardiovascular outcomes at 90days compared with insulin-providing treatment
with insulin and/or sulfonylurea.53 Although these studies
emphasised the important role of insulin resistance in progression of atherothrombotic disease in DM patients, these
results have not been confirmed in the recently published
APPROACH (Assessment on the Prevention of Progression
by Rosiglitazone on Atherosclerosis in Diabetes Patients
With Cardiovascular History) trial.54 In this study, the thiazolinedione rosiglitazone did not significantly decrease the
primary end point of progression of coronary atherosclerosis, measured by percent atheroma volume, more than glipizide, an insulin secretagogue, in patients with type-2 DM
and CAD.54

Associated metabolic conditions


Several metabolic conditions associated with type-2 DM
may contribute to the enhanced platelet reactivity observed
in these patients, including obesity, dyslipidaemia and
increased systemic inflammation.
Obesity is a common feature in patients with type-2 DM,
and obesity itself may be associated with some degree of
insulin resistance, which has relevant implications for platelet reactivity as described above. Nevertheless, other factors
present in obese subjects may account for platelet dysfunction, which include: a) elevated platelet count and high
mean platelet volume,55 which is related with platelet reactivity and has prognostic implications in atherothrombotic
processes such as stroke and ACS;56 b) higher serum leptin
concentration, which is associated with increased platelet
aggregability;57 c) greater cytosolic calcium concentration,58

Downloaded from dvr.sagepub.com by guest on January 14, 2014

255

Ferreiro et al.
which also boosts platelet reactivity; and d) increased oxidative stress.59 Overall, these metabolic abnormalities
caused by obesity ultimately lead to enhanced platelet reactivity.60,61 Russo et al. recently observed in a population of
subjects affected with central obesity that diet-induced
weight loss restored the sensitivity to NO and PGI2 and
reduced platelet activation.49 Of note, response to antiplatelet drugs, in particular to clopidogrel, is impaired as well in
subjects with elevated body mass index.62-64
Patients with type-2 DM are prone to have dyslipidaemia,
typically manifested by elevated triglycerides and low highdensity lipoprotein (HDL) cholesterol. Hypertriglyceridaemia
is a typical manifestation that is known to induce higher
platelet activation.65
The role of triglyceride-rich very-low-density lipoprotein (VLDL) particles is not limited to platelet activation
and also affects coagulation cascade and impairs fibrinolysis, thus predisposing to atherothrombosis.66 The suggested
mechanism to explain the effects of triglycerides on platelet
function is mediated by the apoE content of VLDL particles
and their interaction with the platelet LDL receptor.67 Low
levels of HDL are associated with endothelial dysfunction,68 which increases the atherothrombotic risk in DM
patients. Further, intravenous infusion of reconstituted
HDL rapidly normalises endothelial function by increasing
NO bioavailability in hypercholesterolaemic patients.69 In
line with these findings, an interesting study recently
reported by Calkin and colleagues showed that administration of reconstituted HDL caused a reduction of platelet
aggregation in DM subjects by promoting cholesterol
efflux from platelets.70
DM is also associated with systemic inflammation.
Indeed, DM patients show higher concentrations of inflammatory, platelet activation and coagulation markers (soluble CD40 ligand, soluble P-selectin, interleukin-6, and
tissue factor) than healthy subjects.71 Further, expression of
FcgammaRIIA receptor, which is enhanced in platelets of
DM patients and contributes to platelet activation,72 has
been suggested to be modulated by inflammation.73

Other cellular abnormalities


In this section, the roles of other platelet anomalies common
in DM patients that may account for the global hyperreactivity status, including dysregulation of calcium metabolism,
oxidative stress, upregulation of P2Y12 signalling pathway
and accelerated platelet turnover, are discussed.
Impaired regulation of calcium metabolism that leads to
increased intracellular calcium levels is a major feature of
platelets from patients with DM. The mechanisms involved
in calcium signalling abnormalities present in DM are not
fully elucidated. Factors that have been proposed to play a
role in impaired calcium homeostasis and, therefore, in
platelet hyperreactivity are: a) excessive influx of calcium

through the sodium/calcium exchanger;74 b) changes in the


activity of calcium ATPases;75 c) impaired sensitivity to
insulin, which decreases sarcoplasmic endoplasmic reticulum calcium-ATPase (SERCA-2);37,50 and d) augmented
oxidative stress, which enhances calcium signalling due to
increased formation of superoxide anions and reduced
nitric oxide production.76 The final result of this altered
calcium homeostasis is an augmented concentration of
cytosolic calcium, which leads to enhanced platelet reactivity and aggregation.37,77
Modulation of platelet or vascular redox status and the
presence of reactive oxygen species can impair platelet
function and may be prothrombotic.78 DM is associated
with oxidative stress, in particular with an overproduction
of reactive oxygen and nitrogen species, as well as reduced
platelet antioxidant levels.79,80 The excessive generation of
potent oxidants, such as superoxide anions and hydrogen
peroxide, increases platelet activation.78,81 An increase in
reactive oxygen species in addition to chronic hyperglycaemia enhances production of advanced glycation end products (AGEs).82 These glycated proteins may contribute to
the development of atherosclerotic complications by activation of the receptor for AGEs (RAGE)83 and by enhancing platelet aggregation through the serotonin receptor.84
Further, endothelial function is impaired also by oxidative
stress through a reduction in production of NO and PGI2.85
This is of particular interest in patients with DM since their
platelets have diminished sensitivity to the actions of these
vasodilator molecules produced by the endothelium.47,48
The platelet adenosine diphosphate (ADP) P2Y12 receptor signalling pathway has been suggested to be upregulated in diabetic platelets, in particular those with type-2
DM.86 This suppresses cAMP concentration and, in addition to a lower responsiveness to insulin, leads to increased
adhesion, aggregation, and procoagulant activity.87 Another
abnormality in platelet surface molecules is the increased
expression of surface proteins such as P-selectin and glycoproteins Ib and IIb/IIIa, which are integrins that mediate
adhesion.60,88
An accelerated platelet turnover represented by the presence of a higher number of reticulated platelets has been
observed in patients with DM.91 Reticulated platelets are
larger and more sensitive, resulting in platelet hyperreactivity and lower response to antiplatelet therapies including
aspirin and clopidogrel.90,91 In addition, platelets of DM
patients are larger, and thus hyperreactive, as platelet size
correlates with greater platelet reactivity measured by
aggregation and total release of granular content.92

Conclusions
Patients with DM have accelerated atherosclerosis and
an elevated risk of recurrent atherothrombotic events.
Among the factors that contribute to the prothrombotic

Downloaded from dvr.sagepub.com by guest on January 14, 2014

256

Diabetes and Vascular Disease Research 7(4)

Key messages

 therothrombotic events are more frequent and are


A
associated with worse prognosis in patients with DM
compared with non-DM patients.
Platelet hyperreactivity is a characteristic feature of DM
and plays a pivotal role in its overall prothrombotic state.
Several mechanisms contribute to platelet dysfunction
and affect the adhesion, activation and, aggregation phases
of platelet-mediated thrombosis.

status characterising these patients, abnormalities in platelet function play a pivotal role. These platelet abnormalities
ultimately result in a hyperreactive platelet phenotype. The
understanding of the molecular mechanisms leading to
increased platelet reactivity in patients with DM may set
the basis for targeted antiplatelet treatment strategies in this
high-risk cohort.
Funding
This work was supported by an unrestricted educational
grant from Eli Lilly and Daiichi Sankyo to University of
Sheffield to fund the activities of the European Platelet
Academy.
Conflict of interest
Dominick J Angiolillo (corresponding author) reports
receiving: honoraria for lectures from Bristol Myers
Squibb; Sanofi-Aventis; Eli Lilly Co; Daiichi Sankyo, Inc;
consulting fees from Bristol Myers Squibb; SanofiAventis; Eli Lilly Co; Daiichi Sankyo, Inc.; The Medicines
Company; Portola; Novartis; Medicure; Accumetrics;
Arena Pharmaceuticals; Astra Zeneca; Merck; research
grants from GlaxoSmithKline; Otsuka; Eli Lilly Co;
Daiichi Sankyo, Inc., The Medicines Company; Portola;
Accumetrics; Schering-Plough; Astra-Zeneca; Eisai. Jos
Luis Ferreiro has no conflicts of interest to report. Joan
Antoni Gmez-Hospital has no conflicts of interest to
report.
References
1. Webster MW and Scott RS. What cardiologists need to know
about diabetes. Lancet 1997; 350(Suppl 1): SI2328.
2. Haffner SM, Lehto S, Rnnemaa T, Pyrl K and Laakso
M. Mortality from coronary heart disease in subjects with
type 2 diabetes and in nondiabetic subjects with and without prior myocardial infarction. N Engl J Med 1998; 339:
229234.
3. Lscher TF, Creager MA, Beckman JA and Cosentino F.
Diabetes and vascular disease: pathophysiology, clinical
consequences, and medical therapy: Part II. Circulation
2003; 108: 16551661.

4. Malmberg K, Yusuf S, Gerstein HC, Brown J, Zhao F, Hunt D,


et al. Impact of diabetes on long-term prognosis in patients with
unstable angina and non-Q-wave myocardial infarction: results
of the OASIS (Organization to Assess Strategies for Ischemic
Syndromes) Registry. Circulation 2000; 102: 10141019.
5. Roffi M, Chew DP, Mukherjee D, Bhatt DL, White JA,
Heeschen C, et al. Platelet glycoprotein IIb/IIIa inhibitors
reduce mortality in diabetic patients with non-ST-segmentelevation acute coronary syndromes. Circulation 2001; 104:
27672771.
6. Brogan GX Jr, Peterson ED, Mulgund J, Bhatt DL, Ohman
EM, Gibler WB, et al. Treatment disparities in the care of
patients with and without diabetes presenting with non-STsegment elevation acute coronary syndromes. Diabetes Care
2006; 29: 914.
7. Creager MA, Lscher TF, Cosentino F and Beckman JA.
Diabetes and vascular disease: pathophysiology, clinical
consequences, and medical therapy: Part I. Circulation 2003;
108: 15271532.
8. Osende JI, Badimon JJ, Fuster V, et al. Blood thrombogenicity in type 2 diabetes mellitus patients is associated with glycemic control. J Am Coll Cardiol 2001; 38: 13071312.
9. Takada Y, Urano T, Watanabe I, Taminato A, Yoshimi T
and Takada A. Changes in fibrinolytic parameters in male
patients with type 2 (non-insulin-dependent) diabetes mellitus. Thromb Res 1993; 71: 405415.
10. Dav G and Patrono C. Platelet activation and atherothrombosis. N Engl J Med 2007; 357: 24822494.
11. Stratmann B and Tschoepe D. Pathobiology and cell interactions of platelets in diabetes. Diab Vasc Dis Res 2005; 2:
1623.
12. Angiolillo DJ, Fernandez-Ortiz A, Bernardo E, et al. Platelet
function profiles in patients with type 2 diabetes and coronary artery disease on combined aspirin and clopidogrel
treatment. Diabetes 2005; 54: 24302435.
13. Vinik AI, Erbas T, Park TS, Nolan R and Pittenger GL.
Platelet dysfunction in type 2 diabetes. Diabetes Care 2001;
24: 14761485.
14. Ferroni P, Basili S, Falco A and Dav G. Platelet activation
in type 2 diabetes mellitus. J Thromb Haemost 2004; 2:
12821291.
15. Angiolillo DJ. Antiplatelet therapy in diabetes: efficacy and
limitations of current treatment strategies and future directions. Diabetes Care 2009; 32: 531540.
16. Natarajan A, Zaman AG and Marshall SM. Platelet hyperactivity in type 2 diabetes: role of antiplatelet agents. Diab Vasc
Dis Res 2008; 5: 138144.
17. Ferreiro JL and Angiolillo DJ. Diabetes and anti-platelet therapy in acute coronary syndrome. Circulation 2010 (in press).
18. American Diabetes Association. Diagnosis and classification
of diabetes mellitus. Diabetes Care 2010; 33: S62S69.
19. Schneider DJ. Factors contributing to increased platelet
reactivity in people with diabetes. Diabetes Care 2009; 32:
525527.

Downloaded from dvr.sagepub.com by guest on January 14, 2014

257

Ferreiro et al.
20. Lemkes BA, Hermanides J, Devries JH, Holleman F, Meijers
JC and Hoekstra JB. Hyperglycaemia, a prothrombotic factor? J Thromb Haemost 2010 [Epub ahead of print].
21. Eibl N, Krugluger W, Streit G, Schrattbauer K, Hopmeier P
and Schernthaner G. Improved metabolic control decreases
platelet activation markers in patients with type-2 diabetes.
Eur J Clin Invest 2004; 34: 205209.
22. Gresele P, Guglielmini G, De Angelis M, et al. Acute, shortterm hyperglycemia enhances shear stress-induced platelet
activation in patients with type II diabetes mellitus. J Am
Coll Cardiol 2003; 41: 10131020.
23. Yngen M, Ostenson CG, Li N, Hjemdahl P and Walln
NH. Acute hyperglycemia increases soluble P-selectin in
male patients with mild diabetes mellitus. Blood Coagul
Fibrinolysis 2001; 12: 109116.
24. Undas A, Wiek I, Stpien E, Zmudka K and Tracz W.
Hyperglycemia is associated with enhanced thrombin formation, platelet activation, and fibrin clot resistance to lysis in
patients with acute coronary syndrome. Diabetes Care 2008;
31: 15901595.
25. Yngen M, Norhammar A, Hjemdahl P and Walln NH.
Effects of improved metabolic control on platelet reactivity
in patients with type 2 diabetes mellitus following coronary
angioplasty. Diab Vasc Dis Res 2006; 3: 5256.
26. Malmberg K. Prospective randomised study of intensive
insulin treatment on long-term survival after acute myocardial infarction in patients with diabetes mellitus. DIGAMI
(Diabetes Mellitus, Insulin Glucose Infusion in Acute
Myocardial Infarction) Study Group. Br Med J 1997; 314:
15121515.
27. Malmberg K, Rydn L, Wedel H, et al. DIGAMI 2
Investigators. Intense metabolic control by means of insulin
in patients with diabetes mellitus and acute myocardial infarction (DIGAMI 2): effects on mortality and morbidity. Eur
Heart J 2005; 26: 650661.
28. Gerstein HC, Miller ME, Byington RP, et al. Effects of intensive glucose lowering in type 2 diabetes. N Engl J Med 2008;
358: 25452559.
29. Finfer S, Chittock DR, Su SY, et al. NICE-SUGAR Study
Investigators. Intensive versus conventional glucose control
in critically ill patients. N Engl J Med 2009; 360: 12831297.
30. Winocour PD, Watala C, Perry DW and Kinlough-Rathbone
RL. Decreased platelet membrane fluidity due to glycation
or acetylation of membrane proteins. Thromb Haemost 1992;
68: 577582.
31. Watala C, Golaski J, Boncler MA, Pietrucha T and
Gwodziski K. Membrane lipid fluidity of blood platelets: a
common denominator that underlies the opposing actions of
various agents that affect platelet activation in whole blood.
Platelets 1998; 9: 315327.
32. Keating FK, Sobel BE and Schneider DJ. Effects of increased
concentrations of glucose on platelet reactivity in healthy
subjects and in patients with and without diabetes mellitus.
Am J Cardiol 2003; 92: 13621365.

33. Assert R, Scherk G, Bumbure A, Pirags V, Schatz H and


Pfeiffer AF. Regulation of protein kinase C by short-term
hyperglycaemia in human platelets in vivo and in vitro.
Diabetologia 2001; 44: 188195.
34. Ferretti G, Rabini RA, Bacchetti T, et al. Glycated lowdensity lipoproteins modify platelet properties: a compositional and functional study. J Clin Endocrinol Metab 2002;
87: 21802184.
35. Kessler L, Wiesel ML, Attali P, Mossard JM, Cazenave JP
and Pinget M. Von Willebrand factor in diabetic angiopathy.
Diabetes Metab 1998; 24: 327336.
36. Boden G and Rao AK. Effects of hyperglycemia and hyperinsulinemia on the tissue factor pathway of blood coagulation. Curr Diab Rep 2007; 7: 223227.
37. Randriamboavonjy V and Fleming I. Insulin, insulin resistance, and platelet signaling in diabetes. Diabetes Care 2009;
32: 528530.
38. Hajek AS and Joist JH. Platelet insulin receptor. Methods
Enzymol 1992; 215: 398403.
39. Hunter RW and Hers I. Insulin/IGF-1 hybrid receptor
expression on human platelets: consequences for the effect
of insulin on platelet function. J Thromb Haemost 2009; 7:
21232130.
40. Kahn NN. Insulin-induced expression of prostacyclin receptors on platelets is mediated through ADP-ribosylation of Gi
alpha protein. Life Sci 1998; 63: 20312038.
41. Kahn NN, Bauman WA and Sinha AK. Insulin-induced
release of plasminogen activator from human blood platelets.
Am J Physiol 1995; 268: H117124.
42. Hers I. Insulin-like growth factor-1 potentiates platelet activation via the IRS/PI3Kalpha pathway. Blood 2007; 110:
42434252.
43. Westerbacka J, Yki-Jrvinen H, Turpeinen A, et al. Inhibition
of platelet-collagen interaction: an in vivo action of insulin abolished by insulin resistance in obesity. Arterioscler
Thromb Vasc Biol 2002; 22: 167172.
44. Ishida M, Ishida T, Ono N, et al. Effects of insulin on calcium
metabolism and platelet aggregation. Hypertension 1996; 28:
209212.
45. Algenstaedt P, Antonetti DA, Yaffe MB and Kahn CR.
Insulin receptor substrate proteins create a link between the
tyrosine phosphorylation cascade and the Ca2+-ATPases in
muscle and heart. J Biol Chem 1997; 272: 2369623702.
46. Ferreira IA, Eybrechts KL, Mocking AI, Kroner C and
Akkerman JW. IRS-1 mediates inhibition of Ca2+ mobilization by insulin via the inhibitory G-protein Gi. J Biol Chem
2004; 279: 32543264.
47. Betteridge DJ, El Tahir KE, Reckless JP and Williams KI.
Platelets from diabetic subjects show diminished sensitivity
to prostacyclin. Eur J Clin Invest 1982; 12: 395398.
48. Anfossi G, Mularoni EM, Burzacca S, et al. Platelet resistance to nitrates in obesity and obese NIDDM, and normal platelet sensitivity to both insulin and nitrates in lean
NIDDM. Diabetes Care 1998; 21: 121126.

Downloaded from dvr.sagepub.com by guest on January 14, 2014

258

Diabetes and Vascular Disease Research 7(4)

49. Russo I, Traversa M, Bonomo K, et al. In central obesity,


weight loss restores platelet sensitivity to nitric oxide and
prostacyclin. Obesity (Silver Spring) 2010; 18: 788797.
50. Randriamboavonjy V, Pistrosch F, Blck B, et al. Platelet
sarcoplasmic endoplasmic reticulum Ca2+-ATPase and mucalpain activity are altered in type 2 diabetes mellitus and
restored by rosiglitazone. Circulation 2008; 117: 5260.
51. Sidhu JS, Cowan D, Tooze JA and Kaski JC. Peroxisome
proliferator-activated receptor-gamma agonist rosiglitazone
reduces circulating platelet activity in patients without diabetes mellitus who have coronary artery disease. Am Heart J
2004; 147: e25.
52. Nissen SE, Nicholls SJ, Wolski K, et al. PERISCOPE
Investigators. Comparison of pioglitazone vs glimepiride on
progression of coronary atherosclerosis in patients with type
2 diabetes: the PERISCOPE randomized controlled trial.
JAMA 2008; 299: 15611573.
53. McGuire DK, Newby LK, Bhapkar MV, et al. SYMPHONY
and 2nd SYMPHONY Investigators. Association of diabetes
mellitus and glycemic control strategies with clinical outcomes after acute coronary syndromes. Am Heart J 2004;
147: 246252.
54. Gerstein HC, Ratner RE, Cannon CP, et al. APPROACH
Study Group. Effect of rosiglitazone on progression of coronary atherosclerosis in patients with type 2 diabetes mellitus
and coronary artery disease: the assessment on the prevention of progression by rosiglitazone on atherosclerosis in diabetes patients with cardiovascular history trial. Circulation
2010; 121: 11761187.
55. Muscari A, De Pascalis S, Cenni A, et al. Determinants of
mean platelet volume (MPV) in an elderly population: relevance of body fat, blood glucose and ischaemic electrocardiographic changes. Thromb Haemost 2008; 99: 10791084.
56. Vizioli L, Muscari S and Muscari A. The relationship of
mean platelet volume with the risk and prognosis of cardiovascular diseases. Int J Clin Pract 2009; 63: 15091515.
57. Sugiyama C, Ishizawa M, Kajita K, et al. Platelet aggregation in obese and diabetic subjects: association with leptin
level. Platelets 2007; 18: 128134.
58. Scherrer U, Nussberger J, Torriani S, et al. Effect of weight
reduction in moderately overweight patients on recorded
ambulatory blood pressure and free cytosolic platelet calcium. Circulation 1991; 83: 552558.
59. Anfossi G, Russo I and Trovati M. Platelet dysfunction in central obesity. Nutr Metab Cardiovasc Dis 2009; 19: 440449.
60. Schneider DJ, Hardison RM, Lopes N, Sobel BE, Brooks
MM and the Pro-Thrombosis Ancillary Study Group. Asso
ciation between increased platelet P-selectin expression and
obesity in patients with type 2 diabetes: a BARI 2D (Bypass
Angioplasty Revascularization Investigation 2 Diabetes)
substudy. Diabetes Care 2009; 32: 944949.
61. Murakami T, Horigome H, Tanaka K, Nakata Y, Ohkawara K,
Katayama Y et al. Impact of weight reduction on production

62.

63.

64.

65.

66.

67.

68.

69.

70.

71.

72.

73.

of platelet-derived microparticles and fibrinolytic parameters


in obesity. Thromb Res 2007; 119: 4553.
Angiolillo DJ, Fernndez-Ortiz A, Bernardo E, et al. Platelet
aggregation according to body mass index in patients undergoing coronary stenting: should clopidogrel loading-dose be
weight adjusted? J Invasive Cardiol 2004; 16: 169174.
Sibbing D, von Beckerath O, Schmig A, Kastrati A and von
Beckerath N. Impact of body mass index on platelet aggregation after administration of a high loading dose of 600 mg of
clopidogrel before percutaneous coronary intervention. Am J
Cardiol 2007; 100: 203205.
Hochholzer W, Trenk D, Fromm MF, et al.
Impact of cytochrome P450 2C19 loss-of-function polymorphism and of
major demographic characteristics on residual platelet function after loading and maintenance treatment with clopidogrel in patients undergoing elective coronary stent placement.
J Am Coll Cardiol 2010; 55: 24272434.
de Man FH, Nieuwland R, van der Laarse A, et al. Activated
platelets in patients with severe hypertriglyceridemia: effects
of triglyceride-lowering therapy. Atherosclerosis 2000; 152:
407414.
Olufadi R and Byrne CD. Effects of VLDL and remnant particles on platelets. Pathophysiol Haemost Thromb 2006; 35:
281291.
Pedreo J, Hurt-Camejo E, Wiklund O, Badimn L and
Masana L. Platelet function in patients with familial hypertriglyceridemia: evidence that platelet reactivity is modulated
by apolipoprotein E content of very-low-density lipoprotein
particles. Metabolism 2000; 49: 942949.
Kuhn FE, Mohler ER, Satler LF, Reagan K, Lu DY and
Rackley CE. Effects of high-density lipoprotein on acetylcholine-induced coronary vasoreactivity. Am J Cardiol 1991;
68: 14251430.
Spieker LE, Sudano I, Hrlimann D, et al.
High-density lipoprotein restores endothelial function in hypercholesterolemic
men. Circulation 2002; 105: 13991402.
Calkin AC, Drew BG, Ono A, et al. Reconstituted highdensity lipoprotein attenuates platelet function in individuals with type 2 diabetes mellitus by promoting cholesterol
efflux. Circulation 2009; 120: 20952104.
Lim HS, Blann AD and Lip GY. Soluble CD40 ligand, soluble P-selectin, interleukin-6, and tissue factor in diabetes
mellitus: relationships to cardiovascular disease and risk factor intervention. Circulation 2004; 109: 25242528.
Calverley DC, Hacker MR, Loda KA, et al. Increased platelet Fc receptor expression as a potential contributing cause of
platelet hypersensitivity to collagen in diabetes mellitus. Br J
Haematol 2003; 121: 139142.
Belostocki K, Pricop L, Redecha PB, et al. Infliximab treatment shifts the balance between stimulatory and inhibitory Fcgamma receptor type II isoforms on neutrophils in
patients with rheumatoid arthritis. Arthritis Rheum 2008; 58:
384388.

Downloaded from dvr.sagepub.com by guest on January 14, 2014

259

Ferreiro et al.
74. Li Y, Woo V and Bose R. Platelet hyperactivity and abnormal
Ca(2+) homeostasis in diabetes mellitus. Am J Physiol Heart
Circ Physiol 2001; 280: H14801489.
75. Mazzanti L, Rabini RA, Faloia E, Fumelli P, Bertoli E and
De Pirro R. Altered cellular Ca2+ and Na+ transport in diabetes mellitus. Diabetes 1990; 39: 850854.
76. Schaeffer G, Wascher TC, Kostner GM and Graier WF.
Alterations in platelet Ca2+ signalling in diabetic patients is
due to increased formation of superoxide anions and reduced
nitric oxide production. Diabetologia 1999; 42: 167176.
77. Ishii H, Umeda F, Hashimoto T and Nawata H. Increased
intracellular calcium mobilization in platelets from patients
with type 2 (non-insulin-dependent) diabetes mellitus.
Diabetologia 1991; 34: 332336.
78. Freedman JE. Oxidative stress and platelets. Arterioscler
Thromb Vasc Biol 2008; 28: s1116.
79. Seghieri G, Di Simplicio P, Anichini R, et al. Platelet antioxidant enzymes in insulin-dependent diabetes mellitus. Clin
Chim Acta 2001; 309: 1923.
80. Jardn I, Redondo PC, Salido GM, Pariente JA and Rosado
JA. Endogenously generated reactive oxygen species
reduce PMCA activity in platelets from patients with noninsulin-dependent diabetes mellitus. Platelets 2006; 17:
283288.
81. Redondo PC, Jardin I, Hernndez-Cruz JM, Pariente JA,
Salido GM and Rosado JA. Hydrogen peroxide and peroxynitrite enhance Ca2+ mobilization and aggregation in
platelets from type 2 diabetic patients. Biochem Biophys Res
Commun 2005; 333: 794802.
82. Nishikawa T, Edelstein D, Du XL, et al.
Normalizing mitochondrial superoxide production blocks three pathways of
hyperglycaemic damage. Nature 2000; 404: 787790.
83. Schmidt AM, Yan SD, Wautier JL and Stern D. Activation of
receptor for advanced glycation end products: a mechanism

84.

85.

86.

87.

88.

89.

90.

91.

92.

for chronic vascular dysfunction in diabetic vasculopathy


and atherosclerosis. Circ Res 1999; 84: 489497.
Hasegawa Y, Suehiro A, Higasa S, Namba M and Kakishita E.
Enhancing effect of advanced glycation end products on
serotonin-induced platelet aggregation in patients with diabetes mellitus. Thromb Res 2002; 107: 319323.
Schfer A and Bauersachs J. Endothelial dysfunction, impaired
endogenous platelet inhibition and platelet activation in diabetes and atherosclerosis. Curr Vasc Pharmacol 2008; 6: 5260.
Matsuno H, Tokuda H, Ishisaki A, Zhou Y, Kitajima Y and
Kozawa O. P2Y12 receptors play a significant role in the
development of platelet microaggregation in patients with
diabetes. J Clin Endocrinol Metab 2005; 90: 920927.
Ferreira IA, Mocking AI, Feijge MA, et al. Platelet inhibition
by insulin is absent in type 2 diabetes mellitus. Arterioscler
Thromb Vasc Biol 2006; 26: 417422.
Tschoepe D, Roesen P, Kaufmann L, et al. Evidence for
abnormal platelet glycoprotein expression in diabetes mellitus. Eur J Clin Invest 1990; 20: 166170.
DiMinno G, Silver MJ, Cerbone AM and Murphy S. Trial
of repeated low-dose aspirin in diabetic angiopathy. Blood
1986; 68: 886891.
Guthikonda S, Lev EI, Patel R, DeLao T, Bergeron AL,
Dong JF, et al. Reticulated platelets and uninhibited COX-1
and COX-2 decrease the antiplatelet effects of aspirin.
J Thromb Haemost 2007; 5: 490496.
Guthikonda S, Alviar CL, Vaduganathan M, Arikan M, Tellez
A, DeLao T, et al. Role of reticulated platelets and platelet size
heterogeneity on platelet activity after dual antiplatelet therapy
with aspirin and clopidogrel in patients with stable coronary
artery disease. J Am Coll Cardiol 2008; 52: 743749.
Tschoepe D, Roesen P, Esser J, et al. Large

platelets circulate in an activated state in diabetes mellitus. Semin Thromb


Hemost 1991; 17: 433438.

Downloaded from dvr.sagepub.com by guest on January 14, 2014

Anda mungkin juga menyukai