Vasomotor dysfunction after cardiac surgery

13
Review Vasomotor dysfunction after cardiac surgery Marc Ruel a,b,1 , Tanveer A. Khan a,1 , Pierre Voisine a,1 , Cesario Bianchi a,1 , Frank W. Sellke a, * ,1 a Division of Cardiothoracic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA b Division of Cardiac Surgery, University of Ottawa, Ottawa, Ontario, Canada Received 24 February 2004; received in revised form 28 June 2004; accepted 23 July 2004; Available online 1 September 2004 Summary Cardiopulmonary bypass and cardioplegic arrest, which allow for support of the circulation and stabilization of the heart during cardiac procedures, are still used for the vast majority of cardiac operations worldwide. However, in addition to a well-recognized systemic inflammatory response, cardiopulmonary bypass and cardioplegic arrest elicit complex, multifactorial vasomotor disturbances that vary according to the affected organ bed, with reduced vascular resistances in the skeletal muscle and peripheral circulation, and increased propensity to spasm in the cardiac, pulmonary, mesenteric and cerebral vascular beds. This article outlines the nature, mechanistic basis, and clinical correlates of the vasomotor alterations encountered in patients undergoing cardiac surgery using cardiopulmonary bypass and cardioplegic arrest. q 2004 Elsevier B.V. All rights reserved. Keywords: Cardioplegia; Cardiopulmonary bypass; Endothelial function; Vascular tone 1. Introduction Continued improvements in cardiopulmonary bypass (CPB) and myocardial protection techniques over the last four decades have made cardiac surgical procedures feasible and safe for the vast majority of patients. Nevertheless, modern CPB and cardioplegic arrest techniques remain associated with an acute transcriptional response of hundreds of genes that primarily affects the largest organ of the body, the endothelium, as well as other constituents of blood vessels [1]. The resultant vasomotor disturbances may clinically manifest as reduced peripheral vascular resist- ances in the skeletal muscle bed and, conversely, increased propensity to spasm in the coronary, pulmonary, mesenteric, and cerebral circulations. Abnormal vascular permeability and secondary tissue edema may in turn contribute to malperfusion and dysfunction of the heart, lungs, brain, kidneys, gastrointestinal tract, and other organs [2–5]. These phenomena are most striking after cardiac operations for patients in cardiogenic shock [6,7] or in patients for whom CPB support of more than 80 min is needed [8–10]. Cardioplegia, whether blood or crystalloid, is also intrinsically associated with functional changes in the coronary vasculature that add to the effects of CPB. This may be observed after even routine cardiac cases, with up to 8% of patients having coronary artery spasm manifested by temporary ST segment elevations on ECG after surgery [11–13], and an even greater proportion exhibiting myo- cardial contractile dysfunction that usually peaks 4–6 h postoperatively [14]. These phenomena may be seen in virtually any patient regardless of age, and irrespective of the presence of atherosclerotic coronary artery disease or other risk factors for endothelial dysfunction [15]. Mechanisms that mediate microvascular alterations after CPB and cardioplegic arrest include activation of the complement system, leukocyte-mediated cytokine release, as well as increases in oxidative stress and disturbances in calcium homeostasis that result from ischemia-reperfusion [3] (Fig. 1). These mechanisms lead to an increased local concentration of nitric oxide from upregulation of inducible nitric oxide synthase, and to the release of inflammatory substances such as thromboxane A 2 and inducible cycloox- ygenase from various types of cells, which result in alterations of vasomotor regulation, endothelial integrity, European Journal of Cardio-thoracic Surgery 26 (2004) 1002–1014 www.elsevier.com/locate/ejcts 1010-7940/$ - see front matter q 2004 Elsevier B.V. All rights reserved. doi:10.1016/j.ejcts.2004.07.040 * Corresponding author. Tel.: C1-617-632-8385; fax: C1-617-632- 8287. E-mail address: [email protected] (F.W. Sellke). 1 The authors have no relationship to disclose pertaining to this research.

Transcript of Vasomotor dysfunction after cardiac surgery

Review

Vasomotor dysfunction after cardiac surgery

Marc Ruela,b,1, Tanveer A. Khana,1, Pierre Voisinea,1, Cesario Bianchia,1, Frank W. Sellkea,*,1

aDivision of Cardiothoracic Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USAbDivision of Cardiac Surgery, University of Ottawa, Ottawa, Ontario, Canada

Received 24 February 2004; received in revised form 28 June 2004; accepted 23 July 2004; Available online 1 September 2004

Summary

Cardiopulmonary bypass and cardioplegic arrest, which allow for support of the circulation and stabilization of the heart during cardiac

procedures, are still used for the vast majority of cardiac operations worldwide. However, in addition to a well-recognized systemic

inflammatory response, cardiopulmonary bypass and cardioplegic arrest elicit complex, multifactorial vasomotor disturbances that vary

according to the affected organ bed, with reduced vascular resistances in the skeletal muscle and peripheral circulation, and increased

propensity to spasm in the cardiac, pulmonary, mesenteric and cerebral vascular beds. This article outlines the nature, mechanistic basis, and

clinical correlates of the vasomotor alterations encountered in patients undergoing cardiac surgery using cardiopulmonary bypass and

cardioplegic arrest.

q 2004 Elsevier B.V. All rights reserved.

Keywords: Cardioplegia; Cardiopulmonary bypass; Endothelial function; Vascular tone

1. Introduction

Continued improvements in cardiopulmonary bypass

(CPB) and myocardial protection techniques over the last

four decades have made cardiac surgical procedures feasible

and safe for the vast majority of patients. Nevertheless,

modern CPB and cardioplegic arrest techniques remain

associated with an acute transcriptional response of

hundreds of genes that primarily affects the largest organ

of the body, the endothelium, as well as other constituents of

blood vessels [1]. The resultant vasomotor disturbances may

clinically manifest as reduced peripheral vascular resist-

ances in the skeletal muscle bed and, conversely, increased

propensity to spasm in the coronary, pulmonary, mesenteric,

and cerebral circulations. Abnormal vascular permeability

and secondary tissue edema may in turn contribute to

malperfusion and dysfunction of the heart, lungs, brain,

kidneys, gastrointestinal tract, and other organs [2–5]. These

phenomena are most striking after cardiac operations for

1010-7940/$ - see front matter q 2004 Elsevier B.V. All rights reserved.

doi:10.1016/j.ejcts.2004.07.040

* Corresponding author. Tel.: C1-617-632-8385; fax: C1-617-632-

8287.

E-mail address: [email protected] (F.W. Sellke).1The authors have no relationship to disclose pertaining to this research.

patients in cardiogenic shock [6,7] or in patients for whom

CPB support of more than 80 min is needed [8–10].

Cardioplegia, whether blood or crystalloid, is

also intrinsically associated with functional changes in

the coronary vasculature that add to the effects of CPB. This

may be observed after even routine cardiac cases, with up to

8% of patients having coronary artery spasm manifested by

temporary ST segment elevations on ECG after surgery

[11–13], and an even greater proportion exhibiting myo-

cardial contractile dysfunction that usually peaks 4–6 h

postoperatively [14]. These phenomena may be seen in

virtually any patient regardless of age, and irrespective of

the presence of atherosclerotic coronary artery disease or

other risk factors for endothelial dysfunction [15].

Mechanisms that mediate microvascular alterations after

CPB and cardioplegic arrest include activation of the

complement system, leukocyte-mediated cytokine release,

as well as increases in oxidative stress and disturbances in

calcium homeostasis that result from ischemia-reperfusion

[3] (Fig. 1). These mechanisms lead to an increased local

concentration of nitric oxide from upregulation of inducible

nitric oxide synthase, and to the release of inflammatory

substances such as thromboxane A2 and inducible cycloox-

ygenase from various types of cells, which result in

alterations of vasomotor regulation, endothelial integrity,

European Journal of Cardio-thoracic Surgery 26 (2004) 1002–1014

www.elsevier.com/locate/ejcts

Fig. 1. Pathophysiology of vasomotor dysfunction after cardiopulmonary bypass and cardioplegic arrest.

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–1014 1003

and vascular permeability. Since these processes constitute

sine qua non consequences of CPB and cardioplegic arrest

and ultimately impact on the convalescence of cardiac

surgical patients, surgeons and other clinicians caring for

patients undergoing cardiac surgery may benefit from a

better understanding of these alterations, which thereby

constitutes the aim of this article.

2. The central role of nitric oxide

Cardiopulmonary bypass and cardioplegic arrest result in

vasomotor dysfunction through impairments of endothelial

as well as medial vascular responses [16–19], each of which

is separately addressed below. Common to both types of

impairment is an altered concentration of nitric oxide (NO),

a gas produced in healthy endothelial cells by the activation

of constitutive nitric oxide synthase (eNOS), and in a wide

variety of cells such as activated endothelial cells,

inflammatory cells and macrophages, cardiomyocytes,

interstitial cells and vascular smooth muscle cells by the

inducible form of nitric oxide synthase (iNOS).

2.1. eNOS

Physiologic roles of eNOS, which is responsible for the

endothelial production of NO via the conversion of L-

arginine to L-citrulline, include endothelium-dependent

vasorelaxation through the endothelial-mediated activation

of guanylate cyclase, inhibition of leukocyte adhesion, and

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–10141004

attenuation of platelet activation. In addition to these

endothelial effects, eNOS also regulates tone in the vascular

smooth muscle, to which the endothelium signals, and thus

affects medial vasodilatory responses [20].

eNOS activity is decreased after CPB and cardioplegic

arrest as a result of changes in cell membrane potential

[21–23], substrate and cofactor depletion [5,24], alterations

in the concentration or compartmentalization of

intracellular calcium [25,26], and injury to cell membranes,

associated regulatory enzymes, or ion pumps [19].

Following reperfusion after cardioplegic arrest, increased

breakdown of bioavailable NO occurs from increased

oxidative stress secondary to the generation of oxygen-

derived free-radicals [27], and production is further

impaired by exposure of the endothelium to fragments of

activated complement [28,29], activated neutrophils, and

macrophages [3].

2.2. iNOS

On the other hand the inducible form of NOS, iNOS, is

found in increased quantities in the myocardium after

cardioplegic arrest [30,31], and to a lesser extent in other

organs after CPB [32–36]. In contrast to the low

concentrations of NO produced by eNOS which inhibit

adhesion molecule expression, cytokine synthesis, and

leukocyte adhesion, the large amounts of NO generated by

iNOS under stressed local conditions can be toxic and pro-

inflammatory [37], since the excess NO reacts spon-

taneously with the reactive oxygen radicals released under

inflammatory or postischemic conditions by stressed

endothelial cells and activated leukocytes in order to form

peroxynitrite, which may cause cell apoptosis, cell necrosis,

and circulatory shock [38].

3. Endothelial responses

3.1. Complement activation

Activation of the alternative complement pathway occurs

during CPB as soon as blood gets in contact with

components of the extracorporeal circuit [39]. In addition,

cardioplegic arrest activates the classical complement

pathway, which may cause direct endothelial and cardio-

myocyte injury [40]. The anaphylotoxins C3a, C4a and C5a

are released, which promote neutrophil chemotaxis and

adherence, augment the myocardial inflammatory response,

and cause tissue edema [39,41]. Complement fragments like

C5b-9, known as ‘terminal membrane attack complexes’,

further impair endothelial function by causing direct cell

membrane injury and promoting neutrophil chemotaxis

[28]. Complement activation also causes upregulation of

adhesion molecules and increased generation of oxygen-

derived free radicals [42]. Experimentally, exposure of

isolated myocardial microvessels to zymosan-induced

complement-activated serum reduces NO-mediated endo-

thelium-dependent relaxation [28,29,43], suggesting that

activated complement intrinsically causes endothelial

injury, i.e. in tissues isolated from other blood constituents.

In the clinical setting, complement activation may be

partially inhibited by the administration of heparin [44,45],

by the use of heparin-coated circuits [46,47], by systemic

cooling on CPB [44,48], and by the use of complement

inhibitors such as pexelizumab [49] (Table 1).

3.2. Leukocytes and cytokines

3.2.1. Leukocyte activation

Activated leukocytes, through the release of oxygen-

derived free radicals, proteolytic enzymes, and inflamma-

tory cytokines, have been implicated in myocardial and

endothelial damage after ischemia [50], cardioplegia

[31,51], and CPB alone [41]. Focal leukocyte-endothelial

adherence has been identified on electron microscopy

following cardioplegic arrest and reperfusion [17], and

improved myocardial perfusion and recovery of function

have been observed if leukocyte-depleted blood is used to

reperfuse hearts after cardioplegic arrest in animals [51].

Similarly, improved endothelial and vascular smooth

muscle function have been demonstrated when monoclonal

antibodies to adhesion molecules are administered prior to

reperfusion [31,52].

Leukocytes also mediate detrimental myocardial and

systemic effects in response to CPB alone, regardless of the

occurrence of the cardiac-specific ischemia/reperfusion

injury attributed to cardioplegic arrest. For instance, the

expression of selectins is increased after the start of CPB [1,

53], and this process initiates neutrophil rolling, adherence

to the endothelium, and transmigration across the vascular

wall [54]. Granulocyte apoptosis is also decreased after

CPB, effectively prolonging the functional lifespan of

neutrophils [55]. Neutrophil and macrophage infiltration

has been documented in myocardial [56], pulmonary [57],

and mesenteric [41] tissues after CPB, and may clinically

correlate with the degree of post-CPB renal impairment [58].

3.2.2. Cytokine release

Increased circulating levels of tumor necrosis factor-a(TNF-a), interleukin (IL)-6, IL-8, and other cytokines

liberated during CPB have been shown to directly increase

the permeability of blood vessels, mediate inflammation,

and increase the expression of iNOS [30,59–63]. The

expression of vascular endothelial growth factor (VEGF), a

potent vasodilator and inducer of vascular permeability, and

that of its receptor VEGFR-1 are also upregulated after

cardioplegic arrest and reperfusion, resulting in increased

coronary microvascular relaxation responses [64,65]. On

the other hand, responses to another endothelium-dependent

vasodilator, ADP, are unchanged after cardioplegic arrest,

suggesting that the upregulation of VEGF receptors on the

coronary endothelium is selective and may play a role in

Table 1

Overview of vasomotor disturbances due to cardiopulmonary bypass and cardioplegia

Mechanism Site affected Effect Interventions and selected clinical trials

Alternative and

classical complement

activation

Systemic and

myocardial

endothelial cells

Neutrophil chemotaxis

and adherence,

free- radical production

Complement inhibitors. Intravenous Pexelizumab (a C5 complement

inhibitor)!24 h during and after CABG was effective in reducing the

incidence of the composite end-point of death or perioperative

myocardial infarction in 3088 patients [49]

Full systemic heparinization and heparin-coated circuits. Decrease

serum complement complex levels, CK-MB release and systemic

edema in patients after CPB [44–47]

Systemic cooling. Decreases C3a/C5a and elastase release [44,134],

with however no significant difference between 28 and 34 8C [48]

Leukocyte activation

and cytokine release

Systemic and

myocardial endothelial

and medial smooth

muscle cells

Free-radicals,

proteolytic enzymes,

and inflammatory

cytokines production

Leukocyte-depleted cardioplegic solutions. Were effective in

decreasing troponin T release and the incidence of postoperative low

cardiac output in a trial of 160 patients [135]

Antibodies to adhesion molecules. No clinical data. Anti-P-selectin and

anti-CD18 monoclonal antibodies appeared efficacious in reducing lung

and myocardial injury in animals [52,136–138]

Modified ultrafiltration. Decreases systemic cytokine and adhesion

molecules concentrations after CPB, with however no clinically

detectable effect [139]

Free radical formation Myocardial endothelial

cells

Endothelial cell

membrane injury

Blood cardioplegia. Limits superoxide production [140], warm being

potentially better than cold [141]

Free radical scavengers. N-acetylcysteine added to crystalloid

cardioplegia decreases myeloperoxidase and leukocyte activation, but

with no clinical impact [70]. Similarly, the addition of deferoxamine, an

iron chelator, to cardioplegia reduces release of superoxide and lipid

peroxidation products only [142]. Mannitol was ineffective in a small

study [143]

Prostaglandin

production

Systemic, pulmonary,

and myocardial

endothelial cells

Vasoconstriction,

increased permeability

Aspirin. Preoperative use may protect from protamine-induced

pulmonary vasoconstriction [144]. Data sparse with respect to possible

edema reduction

Steroids. Dextamethasone 1 mg/kg IV at induction may limit

extravascular fluid accumulation, inhibit pulmonary vasoconstriction

[145], and 1.5 g IV methylprednisolone over 24 h improves pulmonary

(controversial [146]) and myocardial function, and decreases CK-MB

release [130]

Intracellular calcium

accumulation

Systemic and

myocardial endothelial

and medial smooth

muscle cells

Increased basal

vascular tone and

propensity to spasm

Low calcium blood cardioplegia. Clinical data sparse. Animal data

suggest optimal calcium concentration of crystalloid solution is between

0.4 and 1.2 mmol/l [89,90]. Optimal concentration in warm blood is

undetermined [91]. In human neonates, the addition of nicardipine, a

calcium-channel blocker, to crystalloid cardioplegia may reduce

myocardial injury [147], although nifedipine was not effective

in adults [148]

Magnesium supplementation. 5-10 mmol/l added to cold blood

cardioplegia may be associated with less ATP depletion [149], less

troponin release and possibly improved performance in humans [150].

Magnesium cardioplegia may also decrease the incidence of atrial

fibrillation by increasing serum magnesium levels [97]

Increased iNOS, ERK,

and MAPK expressions

Systemic and

myocardial endothelial

and medial smooth

muscle cells

Increased permeability,

vasodilatation,

maldistribution

of perfusion

Improvements in CPB circuit biocompatibility. No clinical trial has yet

examined the possible impact of HCC or SMA circuits on iNOS, ERK

and MAPK

Increased peroxynitirite

production, with

resultant cell necrosis

and apoptosis

Peroxynitrite (iNOS degradation product) scavenging. The addition of

glutatione to crystalloid cardioplegia decreased neutrophil adhesion and

improved cardiac function a dog model [151]. Clinical data lacking

Minimization of CPB duration. Animal or observational clinical data

only (clinical RCT setting may be unethical)

Avoidance of alpha-adrenergic agonists. Animal or observational

clinical data only (clinical RCT setting may be unethical)

HSP70 induction with IV glutamine. Small animal data only [103]

CABG, coronary artery bypass grafting; CK-MB, creatine kinase MB isoenzyme; CPB, cardiopulmonary bypass; HCC, heparin-coated circuits; HSP, heat

shock protein; IV, intravenous; RCT, randomized controlled trial; SMA, surface modifying additive.

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–1014 1005

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–10141006

mediating perioperative increases in myocardial vascular

permeability.

3.3. Ischemia-reperfusion

3.3.1. Free-radicals

Oxidative stresses resulting from ischemia and reperfu-

sion damage not only myocardial endothelial cells after

anoxia and cardioplegia, but also affect systemic, non-

cardiac cells during CPB, largely due to the synthesis of

superoxide anions and hydroxyl radicals, and the interaction

between superoxide anions and NO leading to peroxynitrite

free-radicals [66–68]. In the myocardium, strategies to

decrease oxidative stress associated with cardioplegic arrest

have also been examined; for instance, the addition to

cardioplegic solutions of manganese superoxide dismutase

or deferoxamine, which respectively neutralize superoxide

and hydroxyl free-radicals, has been shown to better

preserve coronary endothelium-dependent relaxation after

cardioplegic arrest in swine [18]. The antioxidant N-

acetylcysteine has been evaluated as a cardioplegia additive

and has resulted in impressive effects in dogs [69] but no

clinically demonstrable benefit a small series of patients

[70]. The well-recognized free-radical scavenging proper-

ties of blood, particularly if warm or tepid, constitute some

of the reasons for its widespread preference over crystalloid

solutions in the composition of cardioplegia, with however

little clinically demonstrable benefit in elective, stable

patients [71–74].

The myocardium can also be protected from ischemia-

reperfusion by inhibiting ‘futile’ cell cycles such as the

poly(ADP-ribose) polymerase nuclear enzyme pathway,

which is overinduced by ischemia and cardioplegic arrest,

consumes ATP, increases the oxygen debt, results in cellular

dysfunction, and yet has little known homeostatic role. In a

recent study in pigs, Khan et al. showed that an intravenous

poly(ADP-ribose) polymerase inhibitor improved myocar-

dial perfusion, reduced the extent of infarction, and

improved cardiac function after regional ischemia and

cardioplegia-CPB [75]. However, no report on the use of

such a strategy in humans is yet available.

Few studies have examined the role of interventions

destined at decreasing systemic oxidative stress during

clinical CPB; in this regard, it is possible that angiotensin-

converting enzyme inhibitors, angiotensin-1 blockers, and

statin drugs could play a role in improving systemic and

myocardial outcomes [76–79], although clinical studies

involving angiotensin-converting enzyme inhibitors have so

far been disappointing [80,81].

3.3.2. Cyclooxygenase

Cardioplegia-reperfusion enhances the myocardial

expression of the inducible isoform of cyclooxygenase

(COX-2), an enzyme involved in prostaglandin synthesis

from the conversion of arachidonic acid. Cyclooxygenase is

implicated in the inflammatory response of diseases such as

rheumatoid arthritis and cancer. In the cardiac and systemic

vasculatures, prostaglandins have either vasodilatory or

vasoconstrictive actions, and their effects on the regulation

of vascular permeability changes are comparable and

synergistic to that of NO. CPB and cardioplegia both result

in the increased expression of COX-2 as well as other

predominantly constrictive prostanglandins, whose overall

effect is vasoconstriction of atrial and ventricular micro-

vessels [82–84]. However, no report is yet available of the

possibly beneficial effects of specific COX-2 inhibitors on

the myocardial and systemic vasculature of patients

operated using cardiopulmonary bypass and cardioplegic

arrest.

3.3.3. Calcium homeostasis

Cardioplegic arrest is associated with cellular depolariz-

ation, calcium (Ca2C) influx through voltage-dependent

Ca2C channels, release of Ca2C from intracellular Ca2C

stores, and increases in intracellular Ca2C concentration, all

of which mediate reperfusion injury. Transsarcolemmal

Ca2C influx via NaC–Ca2C exchange also plays an

important role in ischemia/reperfusion-mediated intracellu-

lar Ca2C accumulation [23,85]. These processes may be

exacerbated by the use of crystalloid over blood cardiople-

gic solutions, as crystalloid-based approaches may increase

myocardial hypoxia during cardioplegic arrest [86,87] and

cause higher accumulations of intracellular Ca2C.

Elevated intracellular Ca2C concentrations also alter the

Ca2C sensitivity of the vascular smooth muscle contractile

apparatus, which is regulated by a myosin light chain kinase

whose activity is in turn governed by Ca2C-calmodulin-

mediated phosphorylation [88]. Consequently, Ca2C over-

loading during cardioplegia constitutes a trigger for the

enhancement of basal vascular tone and agonist-induced

microvascular contraction or spasm after reperfusion,

providing an additional rationale for the use of low Ca2C

concentrations in cardioplegic solutions at a concentration

between 0.4 and 1.2 mmol/l [89,90], with the possible

exception of continuous warm blood cardioplegic tech-

niques [91]. Complete calcium depletion of the cardioplegia

solution should be avoided, as it may paradoxically lead to

increased myocardial injury and necrosis [89,92].

3.3.4. Magnesium

Following crystalloid cardioplegia and reperfusion,

contractile myocardial microvascular responses are

enhanced, whereas intrinsic myogenic contractile responses

are diminished [19]. Supplementation of cardioplegic

solutions with magnesium (Mg2C) at a concentration of

at least 5.0 mM preserves these agonist-induced and

myogenic responses [93]. While the exact causes of this

phenomenon remain unclear, higher local Mg2C concen-

trations may confer protection against endothelial dysfunc-

tion [94], or against alterations of the contractile properties

of vascular smooth muscle by displacing Ca2C from

calcium channels binding sites and hyperpolarizing

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–1014 1007

sarcolemmal membranes, thus inhibiting Ca2C entry into

cells [95]. Extracellular Mg2C may also act by raising

intracellular Mg2C concentration, thereby reducing the

release of Ca2C from the sarcoplasmic reticulum [12,51], or

by diminishing the depletion of ATP stores and preserving

the intracellular homeostasis of smooth muscle cells. Since

Mg2C supplementation may translate into endothelial

protection and reduce the incidence of coronary vascular

spasm after cardioplegia, its addition to cardioplegic

solutions at a concentration of 5.0 mEq/l or more appears

desirable [90,96]. This was recently shown in a clinical trial

to decrease requirements for internal defibrillation and

temporary epicardial pacing intraoperatively, as well as

decrease the incidence of new postoperative atrial fibrilla-

tion in urgent CABG patients operated using warm blood

cardioplegia [97].

3.4. Apoptosis and heat-shock proteins

Cardioplegic arrest and cardiopulmonary bypass are

intrinsically associated with apoptosis of endothelial and

other vascular cells in the myocardial and systemic vascular

beds of animals and humans [1,98,99]. This is mediated

by a variety of transcriptional genes such as c-FOS and

JUN-b, which are significantly upregulated in peripheral

and myocardial tissues during cardiopulmonary bypass

and cardioplegic arrest [1]. Protection against apoptosis,

necrosis, and detrimental iNOS/peroxynitrite activity may

be afforded by the spontaneously increased production of

cytoplasmic heat shock proteins (HSP) 70, 72, and 73 in

endothelial cells and cardiomyocytes [100–102]. Upregula-

tion of these heat shock proteins could theoretically afford

protection against apoptosis and necrosis. In rats exposed to

CPB, the systemic administration of glutamine, an inducer

of HSP70, resulted in lower plasma concentrations of

interleukin-6 and interleukin-8, preserved eNOS activity,

and attenuated iNOS activity [103]. No such data is yet

available in humans.

Fig. 2. Skeletal muscle expression of activated ERK 1/2 before and after

cardiopulmonary bypass. Immunohistochemistry shows dark staining

localized to peripheral arterioles (black arrows) with decreased expression

after cardiopulmonary bypass (CPB) of activated ERK 1/2, a major

mediator of vascular tone and of the response to alpha-adrenergic agents.

(Adapted from Khan et al. [98]; with permission).

4. Medial responses

Blood flow is regulated in vascular medial smooth

muscle by metabolic, myogenic, and autonomic mechan-

isms, the latter of which is separately discussed below.

Metabolic regulation of vascular medial tone after CPB and

cardioplegic arrest is altered in the peripheral vasculature by

the systemic release of vasoactive substances during CPB,

and in the myocardial circulation by the abnormally

elevated proportion of open potassium channels during

cardioplegia [104]. Myogenic regulation, i.e. the intrinsic

property of medial smooth muscle to regulate vascular

resistance in response to changes in transmural pressure, is

actually preserved in coronary microvessels after CPB and

cardioplegia, but its pressure-diameter relationship is

shifted upward and normalized only in the presence of a

NO antagonist, thus consistent with an increased release of

NO from iNOS [105].

4.1. Autonomic responses

Autonomic medial regulation of peripheral and myocar-

dial arterioles is on the other hand markedly impaired by

CPB. This mechanism, in conjunction with the increased

circulating levels of vasodilator substances and cytokines,

explains much of the decrease in peripheral vascular

resistance frequently observed as patients are being

separated from CPB and thereafter [30,106,107], the

magnitude of which appears to correlate with the duration

of CPB [10]. As previously mentioned, the systemic

induction of iNOS as well as other vasoactive substances

such as bradykinin and VEGF also contributes to this

vasoplegic syndrome [31,64] and to the reductions in

peripheral vascular resistances and increases in vascular

permeability that contribute to the marked generalized

edema frequently observed after cardiac surgery, all with

the potential of culminating in organ malperfusion [85].

Changes in the tissue activity and expression of

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–10141008

regulatory enzymes such as MAP kinases (MAPK) are now

also being recognized as major mediators of the complex

autonomic vascular responses observed after CPB. In the

coronary and pulmonary circulations, CPB leads to

enhanced vasoconstriction partly as a result of the rapid

induction of ERK1/2 [16,108,109], a subset of MAPK

involved in a variety of signal transduction mechanisms

triggered by a broad range of effectors including TNF-a,

which itself is elevated during CPB and returns to basal

levels after 24 h [110]. ERK1/2 and MAPK in turn induce

interleukin-6 (IL-6) production [111], leading to a cascade

of inflammatory events that include leukocytosis, thrombo-

sis and lymphocyte activation [112]. On the other hand,

recent evidence indicates that ERK1/2 activity is actually

decreased in the non-cardiac, systemic circulation after

CPB, and that this is associated with reduced contractile

responses of peripheral arterioles (Fig. 2) [113]. These

findings implicate MAPK as a major mediator of the site-

specific vasomotor dysfunction observed after CPB, with a

decreased ERK1/2 activity in the peripheral vasculature

associated with a decreased vascular tone, and an increased

ERK1/2 activity in the myocardial circulation associated

with an increased propensity to coronary spasm.

5. Clinical implications

The most frequent clinical scenario resulting from the

adverse endothelial and medial responses to CPB and

cardioplegic arrest described above can be summarized as

follows: (1) decreased basal tone and decreased alpha-

adrenergic microvascular responses in the peripheral/skele-

tal muscle vascular beds [31,64,113], with propensity

to distributive organ malperfusion, (2) initially decreased

coronary vascular tone during cardioplegia [104] followed

by decreased endothelial-mediated relaxation and

increased propensity to spasm [83,84], modifiable in part

by the composition of the cardioplegic solution [18,90,94],

(3) increased vasocontractile responses in the pulmonary

and mesenteric microcirculations [29,32,41] predisposing to

the development of pulmonary shunt and mesenteric

ischemia, respectively, particularly when vasoactive drugs

are administered to regulate blood pressure after CPB [114],

and (4) impaired but as of this writing still incompletely

characterized endothelium-mediated relaxation responses in

the cerebral microvasculature [115] (Fig. 1).

Several approaches introduced over the years to increase

the safety of CPB may have their clinical benefit explained

at least in part by their propensity to limit vasomotor

disturbances after cardiac surgery. For instance, the

increased expression of COX-2 and enhanced contractile

response of coronary arterioles to serotonin after CPB and

cardioplegia [83,84] suggest that the improvements in

coronary bypass graft patency obtained from the periopera-

tive administration of acetylsalicylic acid may not only

derive from its effects on prevention of platelet aggregation

and thrombus formation [116–118], but also from

the prevention of coronary spasm and preservation of

microvascular flow as a result of COX-2 inhibition.

The advantages of blood over crystalloid cardioplegia in

emergency or high-risk cases may result from the inhibitory

effects of blood on oxygen-derived free radical generation,

improved coronary endothelial oxygenation, enhanced

buffering capacity from histidine and other blood proteins,

and better preservation of the morphology of coronary

endothelial cells [3,82]. Antiproteases such as aprotinin, in

addition to inhibiting fibrinolysis and decreasing blood loss

after cardiopulmonary bypass, are also potent inhibitors of

the bradykinin and kallikrein systems [81,119] and may

confer additional clinical benefit by non-specifically redu-

cing the systemic inflammatory response to CPB [120,121],

although the magnitude of this effect remains controversial.

Finally, the inhalational administration of low concen-

trations of NO at 20 ppm for 8 h was recently associated

with decreased myocardial troponin release [122], but it is

reasonable to speculate that high-dose systemic NO

administration, in parallel with the effects of excessive

local production of NO and peroxynitrite from increased

iNOS activity during CPB, might ultimately prove

detrimental.

Other strategies are also available to help prevent

systemic and coronary vasomotor dysfunction after CPB

and cardioplegic arrest (Table 1). Mild systemic cooling and

the administration of heparin, both of which decrease

complement activation [44,45], the utilization of blood-

based cardioplegic solutions [82,123,124], magnesium

supplementation [93] and moderate calcium depletion [15]

of cardioplegic solutions all have beneficial effects on

endothelium-dependent relaxation, myogenic contraction,

responses to adrenergic agonists, and other indices of

myocardial vascular health after CPB and ischemia-

reperfusion. It should be remembered, however, that several

of these strategies may prove beneficial for emergency or

high-risk patients only, as improved outcomes related to

their use during routine operations has not been demon-

strated [125,126].

One standard of practice in need of further research and

foreseeable change, in light of the knowledge gained in

understanding the pathophysiology of vasomotor dysfunc-

tion after CPB, is the routine use of alpha-adrenergic

agonists for the treatment of hemodynamic instability

associated with post-CPB distributive shock. Since periph-

eral arterioles show decreased alpha-adrenergic responsive-

ness after CPB and that these responses are conversely

increased in the mesenteric and coronary circulations, the

widespread use of alpha-adrenergic agonists may constitute

a suboptimal approach for cardiac surgical patients with

vasoplegic syndromes, as these agents can worsen splanch-

nic and cerebral malperfusion, increase pulmonary shunt,

and favor the development of coronary spasm. More

specifically targeted to the underlying cause of vasoplegia

are iNOS/guanylate cyclase inhibitors such as methylene

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–1014 1009

blue, with which preliminary clinical experience in 54

patients treated a 2 mg/kg intravenous infusion adminis-

tered over 20 min demonstrated relative safety and potential

effectiveness [127]. Other promising approaches in post-

CPB vasoplegic syndromes include ‘protecting’ the mesen-

teric circulation against its exacerbated contractile

responses to alpha-agonists agents with an intravenous

infusion of the eNOS/nitric oxide substrate L-arginine [5].

Several interventions have not delivered on the promises

that experimental data suggested with respect to their

clinical use. This may be related to the falsely exaggerated

effects of CPB in animal models compared to humans (due,

for instance, to less advanced circuits, larger priming to

circulatory volume ratios, lack of a dedicated perfusionist,

non-sterile conditions, larger amounts of embolic material,

etc.), to the rarity and lack of sensitivity of adverse clinical

outcomes, to the large potential for confounding biological

noises such as baseline endothelial function and immuno-

logic response in patients, and to the difficulty in a priori

obtaining a proper treatment effect estimate in order to

perform an adequate sample size calculation prior to

launching these small trials. The clinical use of glucocorti-

costeroids on CPB constitutes one example of disappointing

clinical results, since these agents, despite their theoretical

role in blocking the effects of inflammatory cytokines and

the expression of iNOS and COX-2, have repeatedly not

resulted in appreciable clinical benefit [128,129], with the

possible exception of decreased creatine kinase release and

a reduction in the incidence of postoperative atrial

fibrillation in two recent double-blind randomized con-

trolled trials [130] (Rubens FD et al.; in press). Also

controversial has been the role of inhibiting neutrophil

infiltration with a monoclonal antibody to C5a, which

experimentally results in improved endothelial-dependent

relaxation but no demonstrable benefit on myocardial,

pulmonary, or mesenteric functional recovery [31,131,132],

until one clinical trial demonstrated a dose-dependent

inhibition of the generation of complement byproducts, a

reduction in leukocyte activation, a 40% reduction in

creatine kinase-MB release, a 80% reduction in new

cognitive deficits, and a significant reduction in post-

operative blood loss [133]. More research is needed to

further examine the clinical impact of these and other

mechanistically based interventions on the clinical out-

comes of emergency, high-risk, and perhaps even routine

cardiac surgical patients operated using CPB and cardio-

plegic arrest.

6. Conclusion

Cardiopulmonary bypass and cardioplegia are indispen-

sable tools of the cardiac surgical armamentarium that are

still used for the majority of cardiac operations and will

remain selectively needed for decades to come. However,

these modalities constitute a double-edged sword that lead

to formidable multi-systemic microvascular derangements.

Although most of the pathophysiologic contributors to these

disturbances have been identified and their clinical impact

reasonably well elucidated, therapeutic options to circum-

vent them remain limited. Consequently, the development

and evaluation of new therapeutic modalities oriented at

limiting or eliminating the consequences of vasomotor

dysfunction after cardiac surgery should remain a main

focus of applied cardiac surgical research, as we surgeons

aim at continuously improving the results and safety of the

surgical treatment of heart disease for our patients.

References

[1] Ruel M, Bianchi C, Khan TA, Xu S, Liddicoat JR, Voisine P,

Araujo E, Lyon H, Kohane IS, Libermann TA, Sellke FW. Gene

expression profile after cardiopulmonary bypass and cardioplegic

arrest. J Thorac Cardiovasc Surg 2003;126:1521–30.

[2] Long C, Hu X, Zhang J, Xiu R, Guan Y. Changes of microvascular

vasomotion and oxygen metabolism during cooling and rewarming

period of cardiopulmonary bypass. J Extra Corpor Technol 2003;35:

13–16.

[3] Sellke FW, Boyle Jr EM, Verrier ED. Endothelial cell injury in

cardiovascular surgery: the pathophysiology of vasomotor dysfunc-

tion. Ann Thorac Surg 1996;62:1222–8.

[4] Edmunds Jr LH. Inflammatory response to cardiopulmonary bypass.

Ann Thorac Surg 1998;66:S12–S16 discussion S25–8.

[5] Andrasi TB, Soos P, Bakos G, Stumpf N, Blazovics A, Hagl S,

Szabo G. L-arginine protects the mesenteric vascular circulation

against cardiopulmonary bypass-induced vascular dysfunction.

Surgery 2003;134:72–9.

[6] Locker C, Mohr R, Paz Y, Kramer A, Lev-Ran O, Pevni D, Shapira I.

Myocardial revascularization for acute myocardial infarction:

benefits and drawbacks of avoiding cardiopulmonary bypass. Ann

Thorac Surg 2003;76:771–6 discussion 776–7.

[7] Sergeant P, Meyns B, Wouters P, Demeyere R, Lauwers P. Long-

term outcome after coronary artery bypass grafting in cardiogenic

shock or cardiopulmonary resuscitation. J Thorac Cardiovasc Surg

2003;126:1279–86.

[8] Boldt J, Brenner T, Lehmann A, Suttner SW, Kumle B, Isgro F. Is

kidney function altered by the duration of cardiopulmonary bypass?

Ann Thorac Surg 2003;75:906–12.

[9] Kumle B, Boldt J, Suttner SW, Piper SN, Lehmann A, Blome M.

Influence of prolonged cardiopulmonary bypass times on splanchnic

perfusion and markers of splanchnic organ function. Ann Thorac

Surg 2003;75:1558–64.

[10] Carrel T, Englberger L, Mohacsi P, Neidhart P, Schmidli J. Low

systemic vascular resistance after cardiopulmonary bypass: inci-

dence, etiology, and clinical importance. J Card Surg 2000;15:

347–53.

[11] Buxton AE, Hirshfeld Jr JW, Untereker WJ, Goldberg S,

Harken AH, Stephenson LW, Edie RN. Perioperative coronary

arterial spasm: long-term follow-up. Am J Cardiol 1982;50:444–51.

[12] Lockerman ZS, Rose DM, Cunningham Jr JN, Lichstein E.

Postoperative ST-segment elevation in coronary artery bypass

surgery. Chest 1986;89:647–51.

[13] Skarvan K, Graedel E, Hasse J, Stulz P, Pfisterer M. Coronary artery

spasms after coronary artery bypass surgery. Anesthesiology 1984;

61:323–7.

[14] Royster RL. Myocardial dysfunction following cardiopulmonary

bypass: recovery patterns, predictors of inotropic need, theoretical

concepts of inotropic administration. J Cardiothorac Vasc Anesth

1993;7:19–25.

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–10141010

[15] Wernovsky G, Wypij D, Jonas RA, Mayer Jr JE, Hanley FL,

Hickey PR, Walsh AZ, Chang AC, Castaneda AR, Newburger JW,

Wessel DL. Postoperative course and hemodynamic profile after

the arterial switch operation in neonates and infants. A comparison

of low-flow cardiopulmonary bypass and circulatory arrest. Circula-

tion 1995;92:2226–35.

[16] Khan TA, Bianchi C, Ruel M, Voisine P, Li J, Liddicoat JR,

Sellke FW. Mitogen-activated protein kinase inhibition and

cardioplegia-cardiopulmonary bypass reduce coronary myogenic

tone. Circulation 2003;108(Suppl 1):II348–II353.

[17] Sellke FW, Shafique T, Schoen FJ, Weintraub RM. Impaired

endothelium-dependent coronary microvascular relaxation after cold

potassium cardioplegia and reperfusion. J Thorac Cardiovasc Surg

1993;105:52–8.

[18] Sellke FW, Shafique T, Ely DL, Weintraub RM. Coronary

endothelial injury after cardiopulmonary bypass and ischemic

cardioplegia is mediated by oxygen-derived free radicals. Circula-

tion 1993;88:II395–II400.

[19] Sellke FW, Friedman M, Dai HB, Shafique T, Schoen FJ,

Weintraub RM, Johnson RG. Mechanisms causing coronary

microvascular dysfunction following crystalloid cardioplegia and

reperfusion. Cardiovasc Res 1993;27:1925–32.

[20] Beckman JS, Beckman TW, Chen J, Marshall PA, Freeman BA.

Apparent hydroxyl radical production by peroxynitrite: implications

for endothelial injury from nitric oxide and superoxide. Proc Natl

Acad Sci USA 1990;87:1620–4.

[21] Engelman DT, Watanabe M, Engelman RM, Rousou JA, Flack

3rd JE, Deaton DW, Das DK. Constitutive nitric oxide release is

impaired after ischemia and reperfusion. J Thorac Cardiovasc Surg

1995;110:1047–53.

[22] Busse R, Luckhoff A, Pohl U. Role of membrane potential in the

synthesis and release of EDRF. In: Rubanyi GM, editor. Endothelial

regulation of vasomotor tone. New York: Marcel Dekker; 1992. p.

105–20.

[23] He GW, Yang CQ. Hyperkalemia alters endothelium-dependent

relaxation through non-nitric oxide and noncyclooxygenase path-

way: a mechanism for coronary dysfunction due to cardioplegia. Ann

Thorac Surg 1996;61:1394–9.

[24] Cakir O, Oruc A, Eren S, Buyukbayram H, Erdinc L, Eren N. Does

sodium nitroprusside reduce lung injury under cardiopulmonary

bypass? Eur J Cardiothorac Surg 2003;23:1040–5.

[25] Cavallo MJ, Dorman BH, Spinale FG, Roy RC. Myocyte contractile

responsiveness after hypothermic, hyperkalemic cardioplegic arrest.

Disparity between exogenous calcium and beta- adrenergic stimu-

lation. Anesthesiology 1995;82:926–39.

[26] Meldrum DR, Cleveland Jr JC, Sheridan BC, Rowland RT,

Banerjee A, Harken AH. Cardiac surgical implications of calcium

dyshomeostasis in the heart. Ann Thorac Surg 1996;61:1273–80.

[27] Kukreja RC, Hess ML. The oxygen free radical system: from

equations through membrane-protein interactions to cardiovascular

injury and protection. Cardiovasc Res 1992;26:641–55.

[28] Stahl GL, Reenstra WR, Frendl G. Complement-mediated loss

of endothelium-dependent relaxation of porcine coronary arteries.

Role of the terminal membrane attack complex. Circ Res 1995;76:

575–83.

[29] Friedman M, Wang SY, Stahl GL, Johnson RG, Sellke FW. Altered

beta-adrenergic and cholinergic pulmonary vascular responses after

total cardiopulmonary bypass. J Appl Physiol 1995;79:1998–2006.

[30] de Vera ME, Shapiro RA, Nussler AK, Mudgett JS, Simmons RL,

Morris Jr SM, Billiar TR, Geller DA. Transcriptional regulation of

human inducible nitric oxide synthase (NOS2) gene by cytokines:

initial analysis of the human NOS2 promoter. Proc Natl Acad Sci

USA 1996;93:1054–9.

[31] Tofukuji M, Stahl GL, Agah A, Metais C, Simons M, Sellke FW.

Anti-C5a monoclonal antibody reduces cardiopulmonary bypass and

cardioplegia-induced coronary endothelial dysfunction. J Thorac

Cardiovasc Surg 1998;116:1060–8.

[32] Sato K, Li J, Metais C, Bianchi C, Sellke F. Increased pulmonary

vascular contraction to serotonin after cardiopulmonary bypass: role

of cyclooxygenase. J Surg Res 2000;90:138–43.

[33] Tofukuji M, Stahl GL, Metais C, Tomita M, Agah A, Bianchi C,

Fink MP, Sellke FW. Mesenteric dysfunction after cardiopulmonary

bypass: role of complement C5a. Ann Thorac Surg 2000;69:

799–807.

[34] Mayers I, Hurst T, Radomski A, Johnson D, Fricker S, Bridger G,

Cameron B, Darkes M, Radomski MW. Increased matrix metallo-

proteinase activity after canine cardiopulmonary bypass is sup-

pressed by a nitric oxide scavenger. J Thorac Cardiovasc Surg 2003;

125:661–8.

[35] Hayashi Y, Sawa Y, Fukuyama N, Nakazawa H, Matsuda H.

Inducible nitric oxide production is an adaptation to cardiopulmon-

ary bypass-induced inflammatory response. Ann Thorac Surg 2001;

72:149–55.

[36] Hill GE, Springall DR, Robbins RA. Aprotinin is associated with a

decrease in nitric oxide production during cardiopulmonary bypass.

Surgery 1997;121:449–55.

[37] Guzik TJ, Korbut R, Adamek-Guzik T. Nitric oxide and superoxide

in inflammation and immune regulation. J Physiol Pharmacol 2003;

54:469–87.

[38] Becker BF, Kupatt C, Massoudy P, Zahler S. Reactive oxygen

species and nitric oxide in myocardial ischemia and reperfusion. Z

Kardiol 2000;89(Suppl 9):IX/88–IX/91.

[39] Chenoweth DE, Cooper SW, Hugli TE, Stewart RW, Blackstone EH,

Kirklin JW. Complement activation during cardiopulmonary bypass:

evidence for generation of C3a and C5a anaphylatoxins. N Engl

J Med 1981;304:497–503.

[40] Pinckard RN, Olson MS, Giclas PC, Terry R, Boyer JT,

O’Rourke RA. Consumption of classical complement components

by heart subcellular membranes in vitro and in patients after acute

myocardial infarction. J Clin Invest 1975;56:740–50.

[41] Sellke FW, Tofukuji M, Stahl GL, Tomita M, Fink MP. Effect of C5a

inhibition on mesenteric dysfunction after cardiopulmonary bypass

(abstract). Shock 1998;9.

[42] Adler S, Baker PJ, Johnson RJ, Ochi RF, Pritzl P, Couser WG.

Complement membrane attack complex stimulates production of

reactive oxygen metabolites by cultured rat mesangial cells. J Clin

Invest 1986;77:762–7.

[43] Stamler A, Wang SY, Li J, Thurer RL, Schoen FJ, Sellke FW.

Moderate hypothermia reduces cardiopulmonary bypass-induced

impairment of cerebrovascular responses to platelet products. Ann

Thorac Surg 1996;62:191–8.

[44] Moore Jr FD, Warner KG, Assousa S, Valeri CR, Khuri SF. The

effects of complement activation during cardiopulmonary bypass.

Attenuation by hypothermia, heparin, and hemodilution. Ann Surg

1988;208:95–103.

[45] Edens RE, Linhardt RJ, Bell CS, Weiler JM. Heparin and derivatized

heparin inhibit zymosan and cobra venom factor activation of

complement in serum. Immunopharmacology 1994;27:145–53.

[46] Belboul A, Lofgren C, Storm C, Jungbeck M. Heparin-coated

circuits reduce occult myocardial damage during CPB: a random-

ized, single blind clinical trial. Eur J Cardiothorac Surg 2000;17:

580–6.

[47] Jansen PG, te Velthuis H, Huybregts RA, Paulus R, Bulder ER, van

der Spoel HI, Bezemer PD, Slaats EH, Eijsman L, Wildevuur CR.

Reduced complement activation and improved postoperative

performance after cardiopulmonary bypass with heparin-coated

circuits. J Thorac Cardiovasc Surg 1995;110:829–34.

[48] Lindholm L, Bengtsson A, Hansdottir V, Lundqvist M, Rosengren L,

Jeppsson A. Regional oxygenation and systemic inflammatory

response during cardiopulmonary bypass: influence of temperature

and blood flow variations. J Cardiothorac Vasc Anesth 2003;17:

182–7.

[49] Verrier ED, Shernan SK, Taylor KM, Van de Werf F, Newman MF,

Chen JC, Carrier M, Haverich A, Malloy KJ, Adams PX, Todaro TG,

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–1014 1011

Mojcik CF, Rollins SA, Levy JH. Terminal complement blockade

with pexelizumab during coronary artery bypass graft surgery

requiring cardiopulmonary bypass: a randomized trial. J Am Med

Assoc 2004;291:2319–27.

[50] Dreyer WJ, Michael LH, West MS, Smith CW, Rothlein R,

Rossen RD, Anderson DC, Entman ML. Neutrophil accumulation

in ischemic canine myocardium. Insights into time course,

distribution, and mechanism of localization during early reperfusion.

Circulation 1991;84:400–11.

[51] Kawata H, Sawatari K, Mayer Jr JE. Evidence for the role of

neutrophils in reperfusion injury after cold cardioplegic ischemia in

neonatal lambs. J Thorac Cardiovasc Surg 1992;103:908–17

discussion 917–8.

[52] Kawata H, Aoki M, Hickey PR, Mayer Jr JE. Effect of antibody

to leukocyte adhesion molecule CD18 on recovery of neonatal

lamb hearts after 2 h of cold ischemia. Circulation 1992;86:

II364–II370.

[53] Chen YF, Tsai WC, Lin CC, Lee CS, Huang CH, Pan PC, Chen ML,

Huang YS. Leukocyte depletion attenuates expression of neutrophil

adhesion molecules during cardiopulmonary bypass in human

beings. J Thorac Cardiovasc Surg 2002;123:218–24.

[54] Mulligan MS, Polley MJ, Bayer RJ, Nunn MF, Paulson JC,

Ward PA. Neutrophil-dependent acute lung injury. Requirement

for P-selectin (GMP-140). J Clin Invest 1992;90:1600–7.

[55] Chello M, Mastroroberto P, Quirino A, Cuda G, Perticone F,

Cirillo F, Covino E. Inhibition of neutrophil apoptosis after coronary

bypass operation with cardiopulmonary bypass. Ann Thorac Surg

2002;73:123–9 discussion 129–30.

[56] Vakeva AP, Agah A, Rollins SA, Matis LA, Li L, Stahl GL.

Myocardial infarction and apoptosis after myocardial ischemia and

reperfusion: role of the terminal complement components and

inhibition by anti-C5 therapy. Circulation 1998;97:2259–67.

[57] Friedman M, Wang SY, Sellke FW, Cohn WE, Weintraub RM,

Johnson RG. Neutrophil adhesion blockade with NPC 15669

decreases pulmonary injury after total cardiopulmonary bypass.

J Thorac Cardiovasc Surg 1996;111:460–8.

[58] Rinder CS, Fontes M, Mathew JP, Rinder HM, Smith BR. Neutrophil

CD11b upregulation during cardiopulmonary bypass is associated

with postoperative renal injury. Ann Thorac Surg 2003;75:899–905.

[59] Butler J, Chong GL, Baigrie RJ, Pillai R, Westaby S, Rocker GM.

Cytokine responses to cardiopulmonary bypass with membrane and

bubble oxygenation. Ann Thorac Surg 1992;53:833–8.

[60] Dauber IM, Parsons PE, Welsh CH, Giclas PC, Whitman GJ,

Wheeler GS, Horwitz LD, Weil JV. Peripheral bypass-induced

pulmonary and coronary vascular injury. Association with increased

levels of tumor necrosis factor. Circulation 1993;88:726–35.

[61] Downing SW, Edmunds Jr LH. Release of vasoactive substances

during cardiopulmonary bypass. Ann Thorac Surg 1992;54:1236–43.

[62] Wan S, LeClerc JL, Vincent JL. Cytokine responses to cardiopul-

monary bypass: lessons learned from cardiac transplantation. Ann

Thorac Surg 1997;63:269–76.

[63] Borgermann J, Friedrich I, Flohe S, Spillner J, Majetschak M,

Kuss O, Sablotzki A, Feldt T, Reidemeister JC, Schade FU.

Tumor necrosis factor-alpha production in whole blood after

cardiopulmonary bypass: downregulation caused by circulating

cytokine-inhibitory activities. J Thorac Cardiovasc Surg 2002;

124:608–17.

[64] Tofukuji M, Metais C, Li J, Franklin A, Simons M, Sellke FW.

Myocardial VEGF expression after cardiopulmonary bypass

and cardioplegia. Circulation 1998;98:II242–II246 discussion

II247–8.

[65] Sellke FW, Wang SY, Stamler A, Lopez JJ, Li J, Simons M.

Enhanced microvascular relaxations to VEGF and bFGF in

chronically ischemic porcine myocardium. Am J Physiol 1996;

271:H713–H720.

[66] Clermont G, Vergely C, Jazayeri S, Lahet JJ, Goudeau JJ, Lecour S,

David M, Rochette L, Girard C. Systemic free radical activation is

a major event involved in myocardial oxidative stress related to

cardiopulmonary bypass. Anesthesiology 2002;96:80–7.

[67] Ochoa JJ, Vilchez MJ, Ibanez S, Huertas JR, Palacio MA, Munoz-

Hoyos A. Oxidative stress is evident in erythrocytes as well as

plasma in patients undergoing heart surgery involving cardiopul-

monary bypass. Free Radic Res 2003;37:11–17.

[68] Ulus AT, Aksoyek A, Ozkan M, Katircioglu SF, Basu S.

Cardiopulmonary bypass as a cause of free radical-induced oxidative

stress and enhanced blood-borne isoprostanes in humans. Free Radic

Biol Med 2003;34:911–7.

[69] Fischer UM, Cox Jr CS, Allen SJ, Stewart RH, Mehlhorn U,

Laine GA. The antioxidant N-acetylcysteine preserves myocardial

function and diminishes oxidative stress after cardioplegic arrest.

J Thorac Cardiovasc Surg 2003;126:1483–8.

[70] Vento A, Nemlander A, Aittomaki J, Salo J, Karhunen J, Ramo OJ.

N-Acetylcysteine as an additive to crystalloid cardioplegia increased

oxidative stress capacity in CABG patients. Scand Cardiovasc J

2003;37:349–55.

[71] Biagioli B, Borrelli E, Maccherini M, Bellomo G, Lisi G,

Giomarelli P, Sani G, Toscano M. Reduction of oxidative stress

does not affect recovery of myocardial function: warm continuous

versus cold intermittent blood cardioplegia. Heart 1997;77:465–73.

[72] Young JN, Choy IO, Silva NK, Obayashi DY, Barkan HE.

Antegrade cold blood cardioplegia is not demonstrably advan-

tageous over cold crystalloid cardioplegia in surgery for congenital

heart disease. J Thorac Cardiovasc Surg 1997;114:1002–8 discus-

sion 1008–9.

[73] Elwatidy AM, Fadalah MA, Bukhari EA, Aljubair KA, Syed A,

Ashmeg AK, Alfagih MR. Antegrade crystalloid cardioplegia vs

antegrade/retrograde cold and tepid blood cardioplegia in CABG.

Ann Thorac Surg 1999;68:447–53.

[74] Jacquet LM, Noirhomme PH, Van Dyck MJ, El Khoury GA,

Matta AJ, Goenen MJ, Dion RA. Randomized trial of intermittent

antegrade warm blood versus cold crystalloid cardioplegia. Ann

Thorac Surg 1999;67:471–7.

[75] Khan TA, Ruel M, Bianchi C, Voisine P, Komjati K, Szabo C,

Sellke FW. Poly(ADP-ribose) polymerase inhibition improves

postischemic myocardial function after cardioplegia-cardiopulmon-

ary bypass. J Am Coll Surg 2003;197:270–7.

[76] Maack C, Kartes T, Kilter H, Schafers HJ, Nickenig G, Bohm M,

Laufs U. Oxygen free radical release in human failing myocardium is

associated with increased activity of rac1-GTPase and represents a

target for statin treatment. Circulation 2003;108:1567–74.

[77] Hamilton CA, Miller WH, Al-Benna S, Brosnan MJ, Drummond RS,

McBride MW, Dominiczak AF. Strategies to reduce oxididative

stress in cardiovascular disease. Clin Sci (Lond) 2004.

[78] Chello M, Mastroroberto P, Patti G, D’Ambrosio A, Morichetti MC,

Di Sciascio G, Covino E. Simvastatin attenuates leucocyte–

endothelial interactions after coronary revascularisation with

cardiopulmonary bypass. Heart 2003;89:538–43.

[79] Lazar HL, Bao Y, Zhang Y, Bernard SA. Pretreatment with statins

enhances myocardial protection during coronary revascularization.

J Thorac Cardiovasc Surg 2003;125:1037–42.

[80] Walter T, Helber U, Bail D, Heller W, Hoffmeister HM. Influence of

ACE inhibition on myocardial damage, the Kallikrein–Kinin system

and hemostasis during cardiopulmonary bypass surgery. Thorac

Cardiovasc Surg 2002;50:150–4.

[81] Campbell DJ, Dixon B, Kladis A, Kemme M, Santamaria JD.

Activation of the kallikrein–kinin system by cardiopulmonary

bypass in humans. Am J Physiol Regul Integr Comp Physiol 2001;

281:R1059–R1070.

[82] Sellke FW, Shafique T, Johnson RG, Dai HB, Banitt PF, Schoen FJ,

Weintraub RM. Blood and albumin cardioplegia preserve endo-

thelium-dependent microvascular responses. Ann Thorac Surg 1993;

55:977–85.

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–10141012

[83] Metais C, Li J, Simons M, Sellke FW. Serotonin-induced coronary

contraction increases after blood cardioplegia-reperfusion: role of

COX-2 expression. Circulation 1999;100:II328–II334.

[84] Metais C, Bianchi C, Li J, Simons M, Sellke FW. Serotonin-induced

human coronary microvascular contraction during acute myocardial

ischemia is blocked by COX-2 inhibition. Basic Res Cardiol 2001;

96:59–67.

[85] Yuan Y, Granger HJ, Zawieja DC, DeFily DV, Chilian WM.

Histamine increases venular permeability via a phospholipase C-NO

synthase-guanylate cyclase cascade. Am J Physiol 1993;264:

H1734–H1739.

[86] Ibrahim MF, Venn GE, Young CP, Chambers DJ. A clinical

comparative study between crystalloid and blood-based St Thomas’

hospital cardioplegic solution. Eur J Cardiothorac Surg 1999;15:

75–83.

[87] Quinlan GJ, Westerman ST, Mumby S, Pepper JR, Gutteridge JM.

Plasma hypoxanthine levels during crystalloid and blood cardiople-

gias: warm blood cardioplegia increases hypoxanthine levels with a

greater risk of oxidative stress. J Cardiovasc Surg (Torino) 1999;40:

65–9.

[88] Pfitzer G. Invited review: regulation of myosin phosphorylation in

smooth muscle. J Appl Physiol 2001;91:497–503.

[89] Yamamoto F, Braimbridge MV, Hearse DJ. Calcium and cardiople-

gia. The optimal calcium content for the St Thomas’ Hospital

cardioplegic solution. J Thorac Cardiovasc Surg 1984;87:908–12.

[90] Kronon MT, Allen BS, Hernan J, Halldorsson AO, Rahman S,

Buckberg GD, Wang T, Ilbawi MN. Superiority of magnesium

cardioplegia in neonatal myocardial protection. Ann Thorac Surg

1999;68:2285–91 discussion 2291–2.

[91] Nakamura Y, Kuroda H, Takemoto N, Ohgi S, Mori T. Risk of low

calcium and high magnesium in continuous warm hyperkalemic

cardioplegia. Ann Thorac Surg 1999;68:1295–301.

[92] Rebeyka IM, Axford-Gatley RA, Bush BG, del Nido PJ, Mickle DA,

Romaschin AD, Wilson GJ. Calcium paradox in an in vivo model of

multidose cardioplegia and moderate hypothermia. Prevention with

diltiazem or trace calcium levels. J Thorac Cardiovasc Surg 1990;99:

475–83.

[93] Matsuda N, Tofukuji M, Morgan KG, Sellke FW. Coronary

microvascular protection with mg2C: effects on intracellular

calcium regulation and vascular function. Am J Physiol 1999;276:

H1124–H1130.

[94] Tofukuji M, Stamler A, Li J, Franklin A, Wang SY, Hariawala MD,

Sellke FW. Effects of magnesium cardioplegia on regulation of the

porcine coronary circulation. J Surg Res 1997;69:233–9.

[95] Cartier R, Pellerin M, Hollmann C, Pelletier LC. Effects of pressure

and duration of hyperkalemic infusions on endothelial function. Ann

Thorac Surg 1993;55:700–5.

[96] Tsukube T, McCully JD, Faulk EA, Federman M, LoCicero 3rd J,

Krukenkamp IB, Levitsky S. Magnesium cardioplegia reduces

cytosolic and nuclear calcium and DNA fragmentation in the

senescent myocardium. Ann Thorac Surg 1994;58:1005–11.

[97] Yeatman M, Caputo M, Narayan P, Lotto AA, Ascione R, Bryan AJ,

Angelini GD. Magnesium-supplemented warm blood cardioplegia in

patients undergoing coronary artery revascularization. Ann Thorac

Surg 2002;73:112–8.

[98] Aebert H, Kirchner S, Keyser A, Birnbaum DE, Holler E,

Andreesen R, Eissner G. Endothelial apoptosis is induced by

serum of patients after cardiopulmonary bypass. Eur J Cardiothorac

Surg 2000;18:589–93.

[99] Yeh CH, Wang YC, Wu YC, Chu JJ, Lin PJ. Continuous tepid

blood cardioplegia can preserve coronary endothelium and amelio-

rate the occurrence of cardiomyocyte apoptosis. Chest 2003;123:

1647–54.

[100] Aebert H, Cornelius T, Birnbaum DE, Siegel AV, Riegger GA,

Schunkert H. Induction of early immediate genes and programmed

cell death following cardioplegic arrest in human hearts. Eur

J Cardiothorac Surg 1997;12:261–7.

[101] Qing M, Vazquez-Jimenez JF, Schumacher K, Bhardwaj RS,

Klosterhalfen B, Minkenberg R, Messmer BJ, von Bernuth G,

Seghaye MC. Moderate hypothermia during cardiopulmonary

bypass increases intramyocardial synthesis of heat shock protein

72. J Thorac Cardiovasc Surg 2002;124:724–31.

[102] Demidov ON, Tyrenko VV, Svistov AS, Komarova YY,

Karpishenko AI, Margulis BA, Shevchenko YL. Heat shock proteins

in cardiosurgery patients. Eur J Cardiothorac Surg 1999;16:444–9.

[103] Hayashi Y, Sawa Y, Fukuyama N, Nakazawa H, Matsuda H.

Preoperative glutamine administration induces heat-shock protein 70

expression and attenuates cardiopulmonary bypass-induced inflam-

matory response by regulating nitric oxide synthase activity.

Circulation 2002;106:2601–7.

[104] Wang SY, Friedman M, Johnson RG, Zeind AJ, Sellke FW.

Adenosine triphosphate-sensitive KC channels mediate postcardio-

plegia coronary hyperemia. J Thorac Cardiovasc Surg 1995;110:

1073–82.

[105] Wang SY, Friedman M, Franklin A, Sellke FW. Myogenic reactivity

of coronary resistance arteries after cardiopulmonary bypass and

hyperkalemic cardioplegia. Circulation 1995;92:1590–6.

[106] Wang SY, Stamler A, Li J, Johnson RG, Sellke FW. Decreased

myogenic reactivity in skeletal muscle arterioles after hypothermic

cardiopulmonary bypass. J Surg Res 1997;69:40–4.

[107] Wang SY, Friedman M, Johnson RG, Weintraub RM, Sellke FW.

Adrenergic regulation of coronary microcirculation after extracor-

poreal circulation and crystalloid cardioplegia. Am J Physiol 1994;

267:H2462–H2470.

[108] Araujo EG, Bianchi C, Sato K, Faro R, Li XA, Sellke FW.

Inactivation of the MEK/ERK pathway in the myocardium during

cardiopulmonary bypass. J Thorac Cardiovasc Surg 2001;121:

773–81.

[109] Khan TA, Bianchi C, Araujo EG, Ruel M, Voisine P, Sellke FW.

Activation of pulmonary mitogen-activated protein kinases during

cardiopulmonary bypass. J Surg Res 2003;115:56–62.

[110] Marano CW, Garulacan LA, Laughlin KV, Igidbashian L, Trace C,

Goldman SM, Sutter FP, Reichard Jr GA, Mullin JM. Plasma

concentrations of soluble tumor necrosis factor receptor I and tumor

necrosis factor during cardiopulmonary bypass. Ann Thorac Surg

2000;70:1313–8.

[111] Hayashi Y, Sawa Y, Nishimura M, Tojo SJ, Fukuyama N,

Nakazawa H, Matsuda H. P-selectin participates in cardiopulmonary

bypass-induced inflammatory response in association with nitric

oxide and peroxynitrite production. J Thorac Cardiovasc Surg 2000;

120:558–65.

[112] Van den Berghe G. Novel insights into the neuroendocrinology of

critical illness. Eur J Endocrinol 2000;143:1–13.

[113] Khan TA, Bianchi C, Araujo EG, Ruel M, Voisine P, Li J,

Liddicoat JR, Sellke FW. Cardiopulmonary bypass reduces periph-

eral microvascular contractile function by inhibition of mitogen-

activated protein kinase activity. Surgery 2003;134:247–54.

[114] Allen KB, Salam AA, Lumsden AB. Acute mesenteric ischemia after

cardiopulmonary bypass. J Vasc Surg 1992;16:391–5 discussion

395–6.

[115] Cooper WA, Duarte IG, Thourani VH, Nakamura M, Wang NP,

Brown 3rd WM, Gott JP, Vinten-Johansen J, Guyton RA. Hypother-

mic circulatory arrest causes multisystem vascular endothelial

dysfunction and apoptosis. Ann Thorac Surg 2000;69:696–702

discussion 703.

[116] Chesebro JH, Clements IP, Fuster V, Elveback LR, Smith HC,

Bardsley WT, Frye RL, Holmes Jr DR, Vlietstra RE, Pluth JR,

Wallace RB, Puga FJ, Orszulak TA, Piehler JM, Schaff HV,

Danielson GK. A platelet-inhibitor-drug trial in coronary-artery

bypass operations: benefit of perioperative dipyridamole and aspirin

therapy on early postoperative vein-graft patency. N Engl J Med

1982;307:73–8.

[117] Gavaghan TP, Gebski V, Baron DW. Immediate postoperative

aspirin improves vein graft patency early and late after coronary

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–1014 1013

artery bypass graft surgery. A placebo-controlled, randomized study.

Circulation 1991;83:1526–33.

[118] Mangano DT. Aspirin and mortality from coronary bypass surgery.

N Engl J Med 2002;347:1309–17.

[119] Mojcik CF, Levy JH. Aprotinin and the systemic inflammatory

response after cardiopulmonary bypass. Ann Thorac Surg 2001;71:

745–54.

[120] Hill GE, Alonso A, Spurzem JR, Stammers AH, Robbins RA.

Aprotinin and methylprednisolone equally blunt cardiopulmonary

bypass-induced inflammation in humans. J Thorac Cardiovasc Surg

1995;110:1658–62.

[121] Schmartz D, Tabardel Y, Preiser JC, Barvais L, d’Hollander A,

Duchateau J, Vincent JL. Does aprotinin influence the inflammatory

response to cardiopulmonary bypass in patients? J Thorac Cardio-

vasc Surg 2003;125:184–90.

[122] Gianetti J, Del Sarto P, Bevilacqua S, Vassalle C, De Filippis R,

Kacila M, Farneti PA, Clerico A, Glauber M, Biagini A. Sup-

plemental nitric oxide and its effect on myocardial injury and

function in patients undergoing cardiac surgery with extracorporeal

circulation. J Thorac Cardiovasc Surg 2004;127:44–50.

[123] Wang SY, Stamler A, Tofukuji M, Deuson TE, Sellke FW. Effects of

blood and crystalloid cardioplegia on adrenergic and myogenic

vascular mechanisms. Ann Thorac Surg 1997;63:41–9.

[124] Murphy CO, Pan C, Gott JP, Guyton RA. Microvascular reactivity

after crystalloid, cold blood, and warm blood cardioplegic arrest.

Ann Thorac Surg 1995;60:1021–7.

[125] Shapira N, Kirsh M, Jochim K, Behrendt DM. Comparison of

the effect of blood cardioplegia to crystalloid cardioplegia on

myocardial contractility in man. J Thorac Cardiovasc Surg 1980;80:

647–55.

[126] Shanewise JS, Kosinski AS, Coto JA, Jones EL. Prospective,

randomized trial comparing blood and oxygenated crystalloid

cardioplegia in reoperative coronary artery bypass grafting.

J Thorac Cardiovasc Surg 1998;115:1166–71.

[127] Leyh RG, Kofidis T, Struber M, Fischer S, Knobloch K,

Wachsmann B, Hagl C, Simon AR, Haverich A. Methylene blue:

the drug of choice for catecholamine-refractory vasoplegia after

cardiopulmonary bypass? J Thorac Cardiovasc Surg 2003;125:

1426–31.

[128] Boscoe MJ, Yewdall VM, Thompson MA, Cameron JS. Comp-

lement activation during cardiopulmonary bypass: quantitative study

of effects of methylprednisolone and pulsatile flow. Br Med J (Clin

Res Ed) 1983;287:1747–50.

[129] Andersen LW, Baek L, Thomsen BS, Rasmussen JP. Effect of

methylprednisolone on endotoxemia and complement activation

during cardiac surgery. J Cardiothorac Anesth 1989;3:544–9.

[130] Giomarelli P, Scolletta S, Borrelli E, Biagioli B. Myocardial and

lung injury after cardiopulmonary bypass: role of interleukin (IL)-

10. Ann Thorac Surg 2003;76:117–23.

[131] Park KW, Tofukuji M, Metais C, Comunale ME, Dai HB, Simons M,

Stahl GL, Agah A, Sellke FW. Attenuation of endothelium-

dependent dilation of pig pulmonary arterioles after cardiopulmon-

ary bypass is prevented by monoclonal antibody to complement C5a.

Anesth Analg 1999;89:42–8.

[132] Tofukuji M, Stahl GL, Metais C, Tomita M, Agah A, Bianchi C,

Fink MP, Sellke FW. Mesenteric dysfunction after cardiopulmonary

bypass: role of complement C5a. Ann Thorac Surg 2000;69:

799–807.

[133] Fitch JC, Rollins S, Matis L, Alford B, Aranki S, Collard CD,

Dewar M, Elefteriades J, Hines R, Kopf G, Kraker P, Li L, O’Hara R,

Rinder C, Rinder H, Shaw R, Smith B, Stahl G, Shernan SK.

Pharmacology and biological efficacy of a recombinant, humanized,

single-chain antibody C5 complement inhibitor in patients under-

going coronary artery bypass graft surgery with cardiopulmonary

bypass. Circulation 1999;100:2499–506.

[134] Menasche P, Peynet J, Haeffner-Cavaillon N, Carreno MP, de

Chaumaray T, Dillisse V, Faris B, Piwnica A, Bloch G, Tedgui A.

Influence of temperature on neutrophil trafficking during clinical

cardiopulmonary bypass. Circulation 1995;92:II334–II340.

[135] Palatianos GM, Balentine G, Papadakis EG, Triantafillou CD,

Vassili MI, Lidoriki A, Dinopoulos A, Astras GM. Neutrophil

depletion reduces myocardial reperfusion morbidity. Ann Thorac

Surg 2004;77:956–61.

[136] Hayashi Y, Sawa Y, Nishimura M, Tojo SJ, Ichikawa H, Satoh H,

Yamaguchi T, Suhara H, Ohtake S, Matsuda H. P-selectin

monoclonal antibody may attenuate the whole body inflammatory

response induced by cardiopulmonary bypass. Asaio J 2000;46:

334–7.

[137] Aoki M, Jonas RA, Nomura F, Kawata H, Hickey PR. Anti-CD18

attenuates deleterious effects of cardiopulmonary bypass and

hypothermic circulatory arrest in piglets. J Card Surg 1995;10:

407–17.

[138] Mayers I, Hurst T, Johnson D, Cujec B, Ang LC, Thomson D,

Fox JA, Blank GS, Saxena A, Richardson JS. Anti-CD18 antibodies

improve cardiac function following cardiopulmonary bypass in dogs.

J Crit Care 1996;11:189–96.

[139] Grunenfelder J, Zund G, Schoeberlein A, Maly FE, Schurr U,

Guntli S, Fischer K, Turina M. Modified ultrafiltration lowers

adhesion molecule and cytokine levels after cardiopulmonary bypass

without clinical relevance in adults. Eur J Cardiothorac Surg 2000;

17:77–83.

[140] Kalawski R, Balinski M, Bugajski P, Wysocki H, Olszewski R,

Siminiak T. Stimulation of neutrophil activation during coronary

artery bypass grafting: comparison of crystalloid and blood

cardioplegia. Ann Thorac Surg 2001;71:827–31.

[141] Mezzetti A, Calafiore AM, Lapenna D, Deslauriers R, Tian G,

Salerno TA, Verna AM, Bosco G, Pierdomenico SD, Caccurullo F.

Intermittent antegrade warm cardioplegia reduces oxidative stress

and improves metabolism of the ischemic-reperfused human

myocardium. J Thorac Cardiovasc Surg 1995;109:787–95.

[142] Drossos G, Lazou A, Panagopoulos P, Westaby S. Deferoxamine

cardioplegia reduces superoxide radical production in human

myocardium. Ann Thorac Surg 1995;59:169–72.

[143] Larsen M, Webb G, Kennington S, Kelleher N, Sheppard J, Kuo J,

Unsworth-White J. Mannitol in cardioplegia as an oxygen free

radical scavenger measured by malondialdehyde. Perfusion 2002;17:

51–5.

[144] Comunale ME, Maslow A, Robertson LK, Haering JM,

Mashikian JS, Lowenstein E. Effect of site of venous protamine

administration, previously alleged risk factors, and preoperative use

of aspirin on acute protamine-induced pulmonary vasoconstriction.

J Cardiothorac Vasc Anesth 2003;17:309–13.

[145] von Spiegel T, Giannaris S, Wietasch GJ, Schroeder S, Buhre W,

Schorn B, Hoeft A. Effects of dexamethasone on intravascular and

extravascular fluid balance in patients undergoing coronary bypass

surgery with cardiopulmonary bypass. Anesthesiology 2002;96:

827–34.

[146] Chaney MA, Nikolov MP, Blakeman B, Bakhos M, Slogoff S.

Pulmonary effects of methylprednisolone in patients undergoing

coronary artery bypass grafting and early tracheal extubation. Anesth

Analg 1998;87:27–33.

[147] Mori F, Miyamoto M, Tsuboi H, Noda H, Esato K. Clinical trial of

nicardipine cardioplegia in pediatric cardiac surgery. Ann Thorac

Surg 1990;49:413–7 discussion 417-8.

[148] Flameng W, De Meyere R, Daenen W, Sergeant P, Ngalikpima V,

Geboers J, Suy R, Stalpaert G. Nifedipine as an adjunct to St Thomas’

Hospital cardioplegia. A double-blind, placebo-controlled, random-

ized clinical trial. J Thorac Cardiovasc Surg 1986;91:723–31.

[149] Caputo M, Bryan AJ, Calafiore AM, Suleiman MS, Angelini GD.

Intermittent antegrade hyperkalaemic warm blood cardioplegia

M. Ruel et al. / European Journal of Cardio-thoracic Surgery 26 (2004) 1002–10141014

supplemented with magnesium prevents myocardial substrate

derangement in patients undergoing coronary artery bypass surgery.

Eur J Cardiothorac Surg 1998;14:596–601.

[150] Ji B, Feng Z, Liu J, Long C. Myocardial protection related to

magnesium content of cold blood hyperkalemic cardioplegic

solutions in CABG. J Extra Corpor Technol 2002;34:107–10.

[151] Nakamura M, Thourani VH, Ronson RS, Velez DA, Ma XL,

Katzmark S, Robinson J, Schmarkey LS, Zhao ZQ, Wang NP,

Guyton RA, Vinten-Johansen J. Glutathione reverses endothelial

damage from peroxynitrite, the byproduct of nitric oxide degra-

dation, in crystalloid cardioplegia. Circulation 2000;102:

III332–III338.