Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Surg Technol Int ; 36: 234-238, 2020 May 28.
Article in English | MEDLINE | ID: mdl-32297970

ABSTRACT

AIM: Dilatation of the aortic root (AR) after aortic valve surgery associated with ascending aorta replacement (AVS+AAR) is an important concern in long-term follow-up (FU). This study aimed to identify factors that may be associated with this late complication. MATERIAL AND METHODS: Two-hundred-sixteen consecutive patients (150 males and 66 females) who underwent AVS+AAR from June 2009 to April 2018 at a single center were retrospectively analyzed. The mean trans-thoracic echocardiographic (TTE) FU was 44.9 ± 22.2 months. An increase of 10% in AR size compared to the pre-operative baseline was the outcome variable. The Student t-test or chi-square test was used as appropriate. For a survival analysis, a Kaplan-Meier curve was computed and a log-rank p-value was calculated. Cox's regression model was used to assess the predictive value of variables over time. A p-value < 0.05 was significant. RESULTS: No significant differences were observed among patients who underwent aortic valve repair and those who underwent aortic valve replacement (log-rank = 0.917). In patients who underwent valve replacement, AR enlargement was associated with the difference between the diameter of the prosthetic valve and the diameter of the straight vascular prosthesis (OR 0.87, P = 0.024). Based on the difference in diameter between vascular and valve prosthesis, we arbitrarily classified the patients into two groups: a small group (S) (n = 52), in which the difference was ≤ 5 mm, and a large group (L) (n = 34), in which the difference was > 5 mm. Significant AR enlargement was observed in 30.8 % of the S group and 14.7 % of the L group (log-rank = 0.026). A difference of more than 5 mm between the aortic valve prosthesis and the vascular prosthesis protected against AR enlargement in the long-term FU (OR 12.31, P = 0.033), even after adjusting for age and sex (OR 00.32, P = 0.043). CONCLUSION: According to our findings, a difference between the size of the aortic valve prosthesis and the vascular prosthesis of less than or equal to 5 mm was the only factor that increased the risk of AR enlargement after AVS+AAR in long-term FU.


Subject(s)
Aortic Valve , Heart Valve Prosthesis Implantation , Aortic Valve/surgery , Female , Humans , Male , Retrospective Studies , Risk Factors , Treatment Outcome
2.
J Pharmacol Exp Ther ; 364(2): 323-331, 2018 02.
Article in English | MEDLINE | ID: mdl-29222131

ABSTRACT

Cumulative doses of doxorubicin and other antitumor anthracyclines may cause heart failure (HF). Cardiotoxicity is determined by cardiac exposure to anthracyclines and to more toxic secondary alcohol metabolites that are formed inside cardiomyocytes or diffuse from the bloodstream. Concerns exist that HF might be caused by cumulative anthracycline doses that were thought to be safe. Patients with gain-of-function polymorphism of carbonyl reductase 3 (CBR3), which converts anthracyclines to secondary alcohol metabolites, would be at a higher risk of HF. Recently, a pharmacokinetic model was developed that simulated clinical exposure of human myocardium to anthracyclines and incorporated simulations of CBR3 polymorphism. It was shown that HF risk could occur after lower doxorubicin doses than previously reported, particularly for patients with CBR3 polymorphism. In this study, we show that also daunorubicin and idarubicin, but not epirubicin, might cause HF after reportedly safe cumulative doses. CBR3 polymorphism increased HF risk from daunorubicin and idarubicin to a greater extent as compared with doxorubicin. This was caused by daunorubicin and idarubicin forming higher levels of toxic metabolites in human myocardium; moreover, daunorubicin and idarubicin metabolites diffused from plasma and accumulated in cardiac tissue, whereas doxorubicin metabolite did not. CBR3 polymorphism did not aggravate HF risk from epirubicin, which was caused by the very low levels of formation of its toxic metabolite. These results support concerns about HF risk from low-dose anthracycline, characterize the analog specificity of HF risk, and illuminate the role of secondary alcohol metabolites.


Subject(s)
Alcohols/metabolism , Anthracyclines/adverse effects , Anthracyclines/pharmacokinetics , Heart Failure/chemically induced , Heart Failure/metabolism , Heart/drug effects , Myocardium/metabolism , Adult , Aged , Aged, 80 and over , Alcohol Oxidoreductases/metabolism , Conotoxins , Dose-Response Relationship, Drug , Female , Humans , Male , Middle Aged
3.
J Pharmacol Exp Ther ; 362(2): 263-270, 2017 08.
Article in English | MEDLINE | ID: mdl-28559479

ABSTRACT

The antitumor anthracycline, doxorubicin (DOX), can cause heart failure (HF) upon cumulative administration. Lowering the cumulative dose of DOX proved useful to minimize HF risk, and, yet, there is a growing concern that HF might occur after doses that were thought to be safe. Clinical trials that prospectively address such concerns are lacking. Because HF risk correlates with cardiac exposure to DOX, cumulative doses associated with HF risk were re-explored by modeling the accumulation of anthracycline pools in human myocardium. Ex vivo myocardial samples were used in vitro to simulate DOX rapid infusions. The accumulation of anthracycline pools was measured and incorporated into equations from which a risk versus dose curve was obtained. The experimental curve identified a 5% risk dose that was congruent with a previously reported clinical value (380 versus 400 mg/m2, respectively); however, 1-2% risk occurred after lower doses than reported. Simulations of gain-of-function polymorphism of carbonyl reductase 3, which converts DOX to its poorly diffusible alcohol metabolite, doxorubicinol (DOXOL), expanded anthracycline pools and caused 5% or 1-2% risk doses to decrease to 330 or 180-230 mg DOX/m2, respectively. These data show there is no safe dose of DOX. Diminishing cardiac exposure to circulating DOX may represent a cardioprotective strategy. We show that DOX slow infusions or liposomal DOX, which reduce cardiac exposure to DOX, caused formation of smaller anthracycline pools, did not generate DOXOL, increased the 5% risk dose to 750-800 mg/m2, and prevented HF risk aggravation by carbonyl reductase polymorphism.


Subject(s)
Antibiotics, Antineoplastic/toxicity , Doxorubicin/toxicity , Heart Failure/chemically induced , Heart Failure/metabolism , Myocardium/metabolism , Adult , Aged , Aged, 80 and over , Antibiotics, Antineoplastic/metabolism , Dose-Response Relationship, Drug , Doxorubicin/metabolism , Female , Humans , Male , Middle Aged , Organ Culture Techniques , Risk Factors
4.
Biochim Biophys Acta ; 1848(10 Pt B): 2727-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25596534

ABSTRACT

Antitumor drugs have long been known to introduce a measurable risk of cardiovascular events. Cardio-Oncology is the discipline that builds on collaboration between cardiologists and oncologists and aims at screening, preventing or minimizing such a risk. Overt concern about "possible" cardiovascular toxicity might expose cancer patients to the risk of tumor undertreatment and poor oncologic outcome. Careful analysis of risk:benefit balance is therefore central to the management of patients exposed to potentially cardiotoxic drugs. Concomitant or sequential management of cardiac and cancer therapies should also be tailored to the following strengths and weaknesses: i) molecular mechanisms and clinical correlates of cardiotoxicity have been characterized to some extent for anthracyclines but not for other chemotherapeutics or new generation "targeted" drugs, ii) anthracyclines and targeted drugs cause different mechanisms of cardiotoxicity (type I versus type II), and this classification should guide strategies of primary or secondary prevention, iii) with anthracyclines and nonanthracycline chemotherapeutics, cardiovascular events may occur on treatment as well as years or decades after completing chemotherapy, iv) some patients may be predisposed to a higher risk of cardiac events but there is a lack of prospective studies that characterized optimal genetic tests and pharmacologic measures to minimize excess risk, v) clinical toxicity may be preceded by asymptomatic systolic and/or diastolic dysfunction that necessitates innovative mechanism-based pharmacologic treatment, and vi) patient-tailored pharmacologic correction of comorbidities is important for both primary and secondary prevention. Active collaboration of physicians with laboratory scientists is much needed for improving management of cardiovascular sequelae of antitumor therapy. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Cardiotonic Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Disease Management , Neoplasms/drug therapy , Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Cardiotoxins/therapeutic use , Cardiovascular Diseases/complications , Cardiovascular Diseases/pathology , Drug Administration Schedule , Enzyme Inhibitors/therapeutic use , Humans , Neoplasms/complications , Neoplasms/pathology , Risk Assessment
5.
J Pharmacol Exp Ther ; 344(2): 467-78, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23192654

ABSTRACT

Cardiotoxicity from the antitumor anthracycline doxorubicin correlates with doxorubicin cardiac levels, redox activation to superoxide anion (O(2)(.-)) and hydrogen peroxide (H(2)O(2)), and formation of the long-lived secondary alcohol metabolite doxorubicinol. Cardiotoxicity may first manifest during salvage therapy with other drugs, such as the anthracenedione mitoxantrone. Minimal evidence for cardiotoxicity in anthracycline-pretreated patients with refractory-relapsed non-Hodgkin lymphoma was observed with the novel anthracenedione pixantrone. We characterized whether pixantrone and mitoxantrone caused different effects on doxorubicin levels, redox activation, and doxorubicinol formation. Pixantrone and mitoxantrone were probed in a validated ex vivo human myocardial strip model that was either doxorubicin-naïve or preliminarily subjected to doxorubicin loading and washouts to mimic doxorubicin treatment and elimination in the clinical setting. In doxorubicin-naïve strips, pixantrone showed higher uptake than mitoxantrone; however, neither drug formed O(2)(.-) or H(2)O(2). In doxorubicin-pretreated strips, neither pixantrone nor mitoxantrone altered the distribution and clearance of residual doxorubicin. Mitoxantrone showed an unchanged uptake and lacked effects on doxorubicin levels, but synergized with doxorubicin to form more O(2)(.-) and H(2)O(2), as evidenced by O(2)(.-)-dependent inactivation of mitochondrial aconitase or mitoxantrone oxidation by H(2)O(2)-activated peroxidases. In contrast, pixantrone uptake was reduced by prior doxorubicin exposure; moreover, pixantrone lacked redox synergism with doxorubicin, and formed an N-dealkylated product that inhibited metabolism of residual doxorubicin to doxorubicinol. Redox inactivity and inhibition of doxorubicinol formation correlate with the cardiac safety of pixantrone in doxorubicin-pretreated patients. Redox inactivity in the face of high cardiac uptake suggests that pixantrone might also be safe in doxorubicin-naïve patients.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Doxorubicin/analogs & derivatives , Heart/drug effects , Isoquinolines/pharmacology , Mitoxantrone/pharmacology , Myocardium/metabolism , Aged , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Biotransformation , Chromatography, High Pressure Liquid , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Drug Synergism , Female , Humans , Hydrogen Peroxide/metabolism , In Vitro Techniques , Isoquinolines/administration & dosage , Isoquinolines/pharmacokinetics , Male , Mitoxantrone/administration & dosage , Mitoxantrone/pharmacokinetics , Molecular Structure , Oxidation-Reduction , Superoxides/metabolism
6.
J Pharmacol Exp Ther ; 341(2): 464-73, 2012 May.
Article in English | MEDLINE | ID: mdl-22338033

ABSTRACT

Antitumor anthracyclines such as doxorubicin and epirubicin are known to cause cardiotoxicity that correlates with anthracycline accumulation in the heart. The anthracycline amrubicin [(7S,9S)-9-acetyl-9-amino-7-[(2-deoxy-ß-d-erythro-pentopyranosyl)oxy]-7,8,9,10-tetrahydro-6,11-dihydroxy-5,12-napthacenedione hydrochloride] has not shown cardiotoxicity in laboratory animals or patients in approved or investigational settings; therefore, we conducted preclinical work to characterize whether amrubicin attained lower levels than doxorubicin or epirubicin in the heart. Anthracyclines were evaluated in ex vivo human myocardial strips incubated in plasma to which anthracycline concentrations of 3 or 10 µM were added. Four-hour incubations were performed to characterize myocardial anthracycline accumulation derived from anthracycline uptake in equilibrium with anthracycline clearance. Short-term incubations followed by multiple washouts were performed to obtain independent measurements of anthracycline uptake or clearance. In comparison with doxorubicin or epirubicin, amrubicin attained very low levels in the soluble and membrane fractions of human myocardial strips. This occurred at both 3 and 10 µM anthracycline concentrations and was caused primarily by a highly favorable clearance of amrubicin. Amrubicin clearance was facilitated by formation and elimination of sizeable levels of 9-deaminoamrubicin and 9-deaminoamrubicinol. Amrubicin clearance was not mediated by P glycoprotein or other drug efflux pumps, as judged from the lack of effect of verapamil on the partitioning of amrubicin and its deaminated metabolites across myocardial strips and plasma. Limited accumulation of amrubicin in an ex vivo human myocardial strip model may therefore correlate with the improved cardiac tolerability observed with the use of amrubicin in preclinical or clinical settings.


Subject(s)
Anthracyclines/pharmacokinetics , Antineoplastic Agents/pharmacokinetics , Myocardium/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Adult , Aged , Aged, 80 and over , Anthracyclines/pharmacology , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacokinetics , Doxorubicin/pharmacology , Epirubicin/pharmacokinetics , Epirubicin/pharmacology , Female , Humans , In Vitro Techniques , Male , Middle Aged , Verapamil/pharmacology
7.
J Pharmacol Exp Ther ; 341(2): 474-83, 2012 May.
Article in English | MEDLINE | ID: mdl-22338034

ABSTRACT

Anthracycline-related cardiotoxicity correlates with cardiac anthracycline accumulation and bioactivation to secondary alcohol metabolites or reactive oxygen species (ROS), such as superoxide anion (O2·â») and hydrogen peroxide H2O2). We reported that in an ex vivo human myocardial strip model, 3 or 10 µM amrubicin [(7S,9S)-9-acetyl-9-amino-7-[(2-deoxy-ß-D-erythro-pentopyranosyl)oxy]-7,8,9,10-tetrahydro-6,11-dihydroxy-5,12-napthacenedione hydrochloride] accumulated to a lower level compared with equimolar doxorubicin or epirubicin (J Pharmacol Exp Ther 341:464-473, 2012). We have characterized how amrubicin converted to ROS or secondary alcohol metabolite in comparison with doxorubicin (that formed both toxic species) or epirubicin (that lacked ROS formation and showed an impaired conversion to alcohol metabolite). Amrubicin and doxorubicin partitioned to mitochondria and caused similar elevations of H2O2, but the mechanisms of H2O2 formation were different. Amrubicin produced H2O2 by enzymatic reduction-oxidation of its quinone moiety, whereas doxorubicin acted by inducing mitochondrial uncoupling. Moreover, mitochondrial aconitase assays showed that 3 µM amrubicin caused an O2·â»-dependent reversible inactivation, whereas doxorubicin always caused an irreversible inactivation. Low concentrations of amrubicin therefore proved similar to epirubicin in sparing mitochondrial aconitase from irreversible inactivation. The soluble fraction of human myocardial strips converted doxorubicin and epirubicin to secondary alcohol metabolites that irreversibly inactivated cytoplasmic aconitase; in contrast, strips exposed to amrubicin failed to generate its secondary alcohol metabolite, amrubicinol, and only occasionally exhibited an irreversible inactivation of cytoplasmic aconitase. This was caused by competing pathways that favored formation and complete or near-to-complete elimination of 9-deaminoamrubicinol. These results characterize amrubicin metabolic advantages over doxorubicin and epirubicin, which may correlate with amrubicin cardiac safety in preclinical or clinical settings.


Subject(s)
Anthracyclines/metabolism , Anthracyclines/pharmacokinetics , Doxorubicin/metabolism , Doxorubicin/pharmacokinetics , Epirubicin/metabolism , Epirubicin/pharmacokinetics , Myocardium/metabolism , Aconitate Hydratase/metabolism , Alcohols/metabolism , Anthracyclines/pharmacology , Antineoplastic Agents/metabolism , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/pharmacology , Cytoplasm/metabolism , Doxorubicin/pharmacology , Epirubicin/pharmacology , Humans , Hydrogen Peroxide/metabolism , Mitochondria/metabolism , Oxidation-Reduction/drug effects , Reactive Oxygen Species/metabolism , Troponin I/metabolism
8.
J Cell Mol Med ; 15(5): 1096-108, 2011 May.
Article in English | MEDLINE | ID: mdl-20518852

ABSTRACT

Myocardial infarction and its consequences represent one of the most demanding challenges in cell therapy and regenerative medicine. Transfer of skeletal myoblasts into decompensated hearts has been performed through intramyocardial injection. However, the achievements of both cardiomyocyte differentiation and precise integration of the injected cells into the myocardial wall, in order to augment synchronized contractility and avoid potentially life-threatening alterations in the electrical conduction of the heart, still remain a major target to be pursued. Recently, granulocytes colony-stimulating factor (G-CSF) fuelled the interest of researchers for its direct effect on cardiomyocytes, inhibiting both apoptosis and remodelling in the failing heart and protecting from ventricular arrhythmias through the up-regulation of connexin 43 (Cx43). We propose a tissue engineering approach concerning the fabrication of an electrospun cardiac graft functionalized with G-CSF, in order to provide the correct signalling sequence to orientate myoblast differentiation and exert important systemic and local effects, positively modulating the infarction microenvironment. Poly-(L-lactide) electrospun scaffolds were seeded with C2C12 murine skeletal myoblast for 48 hrs. Biological assays demonstrated the induction of Cx43 expression along with morphostructural changes resulting in cell elongation and appearance of cellular junctions resembling the usual cardiomyocyte arrangement at the ultrastructural level. The possibility of fabricating extracellular matrix-mimicking scaffolds able to promote myoblast pre-commitment towards myocardiocyte lineage and mitigate the hazardous environment of the damaged myocardium represents an interesting strategy in cardiac tissue engineering.


Subject(s)
Cell Differentiation , Granulocyte Colony-Stimulating Factor/therapeutic use , Myoblasts, Skeletal/transplantation , Stem Cell Transplantation/methods , Stem Cells/metabolism , Animals , Cell Proliferation , Connexin 43/biosynthesis , Connexin 43/genetics , Granulocyte Colony-Stimulating Factor/metabolism , Mice , Microscopy, Electron, Transmission , Myocardial Infarction/therapy , Polyesters/therapeutic use , Stem Cells/cytology , Tissue Engineering , Tissue Scaffolds
9.
J Card Surg ; 26(6): 632-3, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21923639

ABSTRACT

We report a case of a 69-year-old female with a previous Bentall procedure who developed a right coronary ostial aneurysm. The aneurysm was excluded with a Dacron patch and the right internal mammary artery was used to restore flow to the coronary artery.


Subject(s)
Aortic Valve Insufficiency/surgery , Blood Vessel Prosthesis Implantation/methods , Coronary Aneurysm/etiology , Heart Valve Prosthesis Implantation/adverse effects , Aged , Coronary Aneurysm/diagnostic imaging , Coronary Aneurysm/surgery , Coronary Angiography , Diagnosis, Differential , Female , Humans
10.
J Cardiovasc Pharmacol ; 55(1): 30-8, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19834333

ABSTRACT

Endothelial progenitor cells (EPCs) are a subtype of hematopoietic stem cells, which contribute to the repair of injured endothelium. Treatment with atorvastatin has been shown to increase EPC count in patients with coronary artery disease. Therefore, we investigated whether atorvastatin augments the number of EPCs after cardiopulmonary bypass (CPB) surgery. We conducted a randomized double-blind, placebo-controlled, 2-way crossover trial in 50 patients undergoing elective coronary surgery. Patients received either 3-week treatment with atorvastatin or placebo. EPCs were quantitated by flow cytometric phenotyping on blood samples. Levels of interleukin, IL-6 and IL-8; tumor necrosis factor alpha; SDF-1alpha; granulocyte colony-stimulating factor; and vascular endothelial growth factor were determined at recruitment, preoperatively, post-CPB, and 6, 12, and 24 hours postoperatively. The atorvastatin group showed a significantly higher amount of EPCs both pre- and postoperatively compared with the placebo, with a >4-fold increase compared with the baseline values. CPB induced an increase in all cytokines, but the levels of proinflammatory cytokines were significantly lower in the atorvastatin group (P < 0.05). Statin did not affect levels of SDF-1alpha, granulocyte colony-stimulating factor, and vascular endothelial growth factor. However, no correlation was found between plasma levels of any cytokine and number of EPCs, with the exception of SDF-1alpha. Pretreatment with atorvastatin significantly increases the amount of EPCs after CPB, by a mechanism independent of plasma levels of cytokines and cholesterol.


Subject(s)
Anticholesteremic Agents/pharmacology , Cardiopulmonary Bypass , Hematopoietic Stem Cells/drug effects , Heptanoic Acids/pharmacology , Pyrroles/pharmacology , Aged , Atorvastatin , Cholesterol/blood , Cross-Over Studies , Cytokines/drug effects , Cytokines/metabolism , Double-Blind Method , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Humans , Male , Middle Aged , Time Factors
11.
J Pharmacol Exp Ther ; 329(1): 175-84, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19144686

ABSTRACT

Secondary alcohol metabolites and reactive oxygen species mediate cardiomyopathy induced by cumulative doses of antitumor anthracyclines, such as doxorubicin and epirubicin. Epirubicin exhibits a defective conversion to both toxic species, thereby inducing cardiotoxicity at doses higher than equiactive to doxorubicin; however, the gain in cardiac tolerability seems to be marginal compared with the magnitude of the metabolic defects of epirubicin. Cardiomyopathy may occur independent of toxic metabolites if a given anthracycline tends to accumulate in the heart; therefore, we characterized whether epirubicin showed an unusual accumulation in human myocardial strips incubated in plasma. Epirubicin exhibited a higher uptake and reached myocardial levels 2 times higher than those of doxorubicin. Epirubicin also showed a unique metabolization to doxorubicinolone, the product of epirubicin deglycosidation and carbonyl reduction. In diffusing from the strips to plasma, doxorubicinolone caused membrane permeation effects that augmented epirubicin elimination. Experiments with purified doxorubicinolone showed that the efflux of 1 mol doxorubicinolone promoted the concomitant elimination of as many as approximately 40 mol epirubicin. Doxorubicinolone could also diffuse from plasma back to the strips, causing a permeation effect that promoted epirubicin reuptake; however, this reverse process was slower and less potent. On balance, doxorubicinolone efflux diminished the epirubicin to doxorubicin accumulation ratio to approximately 1.5. These results suggest that the cardiac tolerability of epirubicin is limited by its accumulation in the heart and that such accumulation would be even higher in the absence of doxorubicinolone formation and efflux. These results may also serve guidelines for developing noncardiotoxic anthracyclines.


Subject(s)
Antibiotics, Antineoplastic/metabolism , Doxorubicin/metabolism , Epirubicin/metabolism , Myocardium/metabolism , Naphthacenes/metabolism , Aged , Antibiotics, Antineoplastic/pharmacokinetics , Biotransformation , Chromatography, High Pressure Liquid , Doxorubicin/pharmacokinetics , Epirubicin/pharmacokinetics , Humans , Naphthacenes/pharmacokinetics , Permeability
12.
Diabetes Metab Res Rev ; 25(5): 420-6, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19405075

ABSTRACT

BACKGROUND: Diabetic patients exhibit an increased risk of saphenous graft occlusion after coronary bypass. Advanced glycation end products (AGEs) are ubiquitous signalling proteins that are associated with vascular and neurological complication of diabetes. The aim of this study is to verify whether AGE levels may promote endothelial cell alterations responsible for vein graft failure. METHODS: Segments of saphenous vein were obtained from both normal people and diabetic patients (HbA(1c) < 6.0%) at the time of coronary surgery. Cultured endothelial cells were incubated in the absence/presence of AGEs (2 and 20 microM), and mRNA and protein for both receptor of AGEs (RAGE) and peroxisome proliferator-activated receptors-gamma (PPAR-gamma) were analysed by real-time polymerised chain reaction (PCR) and Western blot analysis. In the same fashion, the cell release of reactive oxygen species (ROS) was estimated in the absence/presence of AGEs by spectrofluorimetric analysis. Finally, neutrophil-endothelial adhesion was evaluated in saphenous vein segments with and without the addition of AGEs. RESULTS: AGEs activated in a dose-dependent manner the expression of RAGE and inhibited PPAR-gamma expression in endothelial cells as testified by both reverse transcription-PCR (RT-PCR) and Western blot analysis. Stimulation of cultured endothelial cells with AGEs significantly enhanced intracellular ROS formation in a dose-dependent manner. Finally, neutrophil-endothelial adhesion was significantly increased after incubation of control veins with AGEs. CONCLUSIONS: These findings indicate that even in diabetic patients with HbA(1c) < 6.0%, elevated serum levels of AGE determine a sort of a pro-thrombotic state, providing a common mechanism that could explain the increased rate of vein graft occlusion in this population.


Subject(s)
Diabetes Mellitus, Type 2/blood , Endothelial Cells/metabolism , Glycation End Products, Advanced/adverse effects , PPAR gamma/metabolism , Receptors, Immunologic/drug effects , Blood Glucose/metabolism , Case-Control Studies , Cell Adhesion/physiology , Cells, Cultured , Coronary Artery Disease/blood , Coronary Artery Disease/complications , Coronary Artery Disease/surgery , Diabetes Mellitus, Type 2/complications , Diabetic Angiopathies/blood , Diabetic Angiopathies/complications , Diabetic Angiopathies/surgery , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Glycation End Products, Advanced/administration & dosage , Glycation End Products, Advanced/blood , Graft Survival , Humans , Neutrophils , RNA, Messenger/analysis , Reactive Oxygen Species/metabolism , Receptor for Advanced Glycation End Products , Reference Values , Saphenous Vein/cytology , Saphenous Vein/transplantation
13.
Circulation ; 114(14): 1455-61, 2006 Oct 03.
Article in English | MEDLINE | ID: mdl-17000910

ABSTRACT

BACKGROUND: Atrial fibrillation (AF) after cardiac surgery is associated with increased risk of complications, length of stay, and cost of care. Observational evidence suggests that patients who have undergone previous statin therapy have a lower incidence of postoperative AF. We tested this observation in a randomized, controlled trial. METHODS AND RESULTS: Two hundred patients undergoing elective cardiac surgery with cardiopulmonary bypass, without previous statin treatment or history of AF, were enrolled. Patients were randomized to atorvastatin (40 mg/d, n=101) or placebo (n=99) starting 7 days before operation. The primary end point was incidence of postoperative AF; secondary end points were length of stay, 30-day major adverse cardiac and cerebrovascular events, and postoperative C-reactive protein (CRP) variations. Atorvastatin significantly reduced the incidence of AF versus placebo (35% versus 57%, P=0.003). Accordingly, length of stay was longer in the placebo versus atorvastatin arm (6.9+/-1.4 versus 6.3+/-1.2 days, P=0.001). Peak CRP levels were lower in patients without AF (P=0.01), irrespective of randomization assignment. Multivariable analysis showed that atorvastatin treatment conferred a 61% reduction in risk of AF (odds ratio 0.39, 95% confidence interval 0.18 to 0.85, P=0.017), whereas high postoperative CRP levels were associated with increased risk (odds ratio 2.0, 95% confidence interval 1.2 to 7.0, P=0.01). The incidence of major adverse cardiac and cerebrovascular events at 30 days was similar in the 2 arms. CONCLUSIONS: Treatment with atorvastatin 40 mg/d, initiated 7 days before surgery, significantly reduces the incidence of postoperative AF after elective cardiac surgery with cardiopulmonary bypass and shortens hospital stay. These results may influence practice patterns with regard to adjuvant pharmacological therapy before cardiac surgery.


Subject(s)
Atrial Fibrillation/drug therapy , Cardiac Surgical Procedures/adverse effects , Heptanoic Acids/administration & dosage , Pyrroles/administration & dosage , Aged , Atorvastatin , Atrial Fibrillation/etiology , Atrial Fibrillation/prevention & control , C-Reactive Protein/analysis , Cerebrovascular Disorders/etiology , Female , Heart Diseases/etiology , Humans , Length of Stay , Male , Middle Aged , Postoperative Complications/drug therapy , Postoperative Complications/prevention & control
14.
Clin Ther ; 29(8): 1706-17, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17919551

ABSTRACT

BACKGROUND: Abnormal erythrocyte deformability can cause severe complications during cardiopulmonary bypass (CPB) surgery, including both hemolysis and perfusion abnormalities. OBJECTIVES: The goals of this study were to evaluate changes in erythrocyte membrane fluidity and lipid peroxidation during CPB and to examine the effect of simvastatin treatment on these parameters. METHODS: Patients undergoing cardiac surgery involving CPB were selected and randomized to receive either simvastatin 40 mg/d or placebo for 3 weeks before surgery. Three blood samples were obtained at different times during surgery for analysis of erythrocyte membrane fluidity, anion permeability, and lipid peroxidation. Erythrocyte ghosts were prepared and incubated with a lipophilic fluorescent probe (diphenyl-hexatriene), and fluorescence anisotropy was evaluated by spectrophotofluorimetric assay as a measure of membrane fluidity. Anion permeability was evaluated by the specific absorption of methemoglobin (CM) at 590 and 635 nm after treatment of heparinized blood with NaNO2. The formation of thiobarbituric acid-reactive substances was evaluated as an index of lipid peroxidation. Aspartate transaminase and lactate dehydrogenase were also measured as indices of hemolysis. RESULTS: Forty patients met the inclusion criteria (20 simvastatin, 20 placebo). Their characteristics differed significantly at baseline only in terms of the lipid profile; the statin group had higher levels of high-density lipoprotein cholesterol (P = 0.01) and lower levels of low-density lipoprotein cholesterol (P = 0.001) than the placebo group. CPB was found to significantly modify characteristics of the erythrocyte membrane. Compared with preoperative values, CPB induced decreases in both mean (SD) erythrocyte membrane fluidity and anion permeability (preoperative CM: 0.69 [0.02]; 24-hour postoperative CM: 0.18 [0.02]; P < 0.001) and an increase in mean (SD) membrane lipid peroxidation (preoperative malonyl dialdehyde [MDA]: 0.21 [0.01] nmol/mL; postoperative MDA: 0.10 [0.02] nmol/mL; P < 0.001). Treatment with simvastatin was associated with a significant reduction in mean (SD) membrane lipid peroxidation both preoperatively and at 24 hours postoperatively compared with placebo (preoperative MDA: 0.07 [0.01] vs 0.10 [0.02] nmol/mL, respectively; P < 0.05; postoperative MDA: 0.10 [0.04] vs 0.21 [0.01] nmol/mL; P < 0.05). In addition, statin treatment was associated with significant increases in anion permeability preoperatively and postoperatively compared with placebo (preoperative CM: 0.79 [0.01] vs 0.69 [0.02]; P < 0.01; 24-hour postoperative CM: 0.30 [0.01] vs 0.18 [0.02]; P < 0.01). CONCLUSION: The results of this study suggest that among these patients undergoing CPB surgery, use of simvastatin for 3 weeks before the surgery had significant beneficial effects on erythrocyte membrane fluidity, lipid peroxidation, and anion permeability.


Subject(s)
Cardiopulmonary Bypass/adverse effects , Coronary Artery Bypass , Coronary Artery Disease/drug therapy , Erythrocyte Membrane/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Membrane Fluidity/drug effects , Oxidative Stress/drug effects , Premedication , Simvastatin/therapeutic use , Aged , Cell Membrane Permeability/drug effects , Coronary Artery Disease/blood , Coronary Artery Disease/surgery , Double-Blind Method , Erythrocyte Deformability/drug effects , Erythrocyte Membrane/metabolism , Female , Hemolysis/drug effects , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Lipid Peroxidation/drug effects , Male , Middle Aged , Postoperative Complications/prevention & control , Simvastatin/pharmacology , Spectrometry, Fluorescence , Time Factors , Treatment Outcome
15.
Eur J Cardiothorac Surg ; 32(4): 667-8, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17651979

ABSTRACT

We report a case of a 76-year-old man complaining of dysphagia for solid food associated with a weight loss following an off-pump double vessel coronary artery revascularization. Multislice spiral computed tomography showed a 6 cm x 2.5 cm solid formation next to the posterior wall of the left atrium, adjacent to the pericardium and the right anterolateral side of the esophagus. The mass was confirmed to be an intramural esophageal hematoma by endoscopic ultrasound-guided fine-needle aspiration cytology. Injury to esophagus during the placement of deep pericardial sutures and postoperative infusion of heparin are claimed to be causes of this complication.


Subject(s)
Coronary Artery Bypass, Off-Pump/adverse effects , Coronary Disease/surgery , Deglutition Disorders/etiology , Esophageal Diseases/complications , Hematoma/complications , Aged , Esophageal Diseases/diagnosis , Hematoma/diagnosis , Humans , Male , Suture Techniques , Treatment Outcome , Weight Loss
16.
J Geriatr Cardiol ; 14(11): 662-668, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29321796

ABSTRACT

Heart rate variability (HRV) refers to the variations between consecutive heartbeats, which depend on the continuous modulation of the sympathetic and parasympathetic branches of the autonomic nervous system. HRV has been shown to be effective as a predictor of risk after myocardial infarction and an early warning sign of diabetic neuropathy, and in the cardiology setting is now recognized to be a useful tool for risk-stratification after hospital admission and after discharge. Recent evidences suggest that HRV analysis might predict complications even in patients undergoing cardiac surgery, and the present review summarizes the importance of HRV analysis in adult cardiac surgery and the perspectives for HRV use in current clinical practice. Although future larger studies are warranted before HRV can be included into daily clinical practice in adult cardiac surgery, HRV is a novel tool which might detect autonomic instability in the early postoperative phase and during hospital stay, thus predicting or prompt-diagnosing many of the post-operative complications.

17.
J Cardiovasc Med (Hagerstown) ; 18(12): 976-982, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28787317

ABSTRACT

AIMS: Statins are a widely recognized weapon in the primary and secondary prevention of coronary artery disease for their pleiotropic effects. However, recent reports from the cerebrovascular and pharmacological literature are insinuating concerns about a potential increase in the haemorrhagic risk among statin users.The effect of statins in postoperative bleeding should be carefully investigated in major cardiac surgery that exposes per se to risk of bleeding. METHODS: In this retrospective cohort study, we evaluated 441 patients who received atorvastatin until surgery and 213 patients who had never been treated with statins, undergoing elective primary isolated on-pump coronary artery bypass grafting. Postoperative bleedings, blood products use and complications were monitored during hospitalization. RESULTS: Preoperative and intraoperative variables were similar between groups. Early and overall postoperative bleedings were reduced among statin users, who had lower C-reactive protein values in the first postoperative day. Atorvastatin carries a strong protective effect against major bleedings, with a propensity score-adjusted odds ratio of 0.28 (P < 0.01). Also, blood products use for statin-treated patients was lower compared with controls, with fewer transfused patients and fewer red-packed cells units per transfused patient. CONCLUSION: Preoperative atorvastatin use is associated with reduced risk of bleeding and blood products use after coronary artery bypass grafting, likely due to a reduction in the postoperative inflammatory response. Statin continuation at the highest tolerable dose should be encouraged before cardiac surgery. The preoperative use of statins in cardiac surgery as 'bleeding-preventers' might have profound clinical implications.


Subject(s)
Atorvastatin/therapeutic use , Coronary Artery Bypass , Coronary Artery Disease/therapy , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Postoperative Complications/prevention & control , Postoperative Hemorrhage/prevention & control , Aged , Aged, 80 and over , Atorvastatin/administration & dosage , C-Reactive Protein/metabolism , Female , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage , Italy/epidemiology , Logistic Models , Male , Middle Aged , Preoperative Care , Propensity Score , Retrospective Studies
18.
J Am Coll Cardiol ; 67(20): 2334-2346, 2016 May 24.
Article in English | MEDLINE | ID: mdl-27199056

ABSTRACT

BACKGROUND: Guidelines recommend surgery for patients with severe ischemic mitral regurgitation (MR). Nonrandomized studies suggest that subvalvular repair is associated with longer survival, but randomized studies are lacking. OBJECTIVES: This study sought to investigate the benefit of papillary muscle surgery on long-term clinical outcomes of patients with ischemic MR. METHODS: Ninety-six patients with severe ischemic MR were randomized to either undersizing restrictive mitral annuloplasty (RA) or papillary muscle approximation with undersizing restrictive mitral annuloplasty (PMA) associated with complete surgical myocardial revascularization. The primary endpoint was change in left ventricular end-diastolic diameter (LVEDD) after 5 years, measured as the absolute difference from baseline, which was evaluated by paired Student t tests. Secondary endpoints included changes in echocardiographic parameters, overall mortality, the composite cardiac endpoint (major adverse cardiac and cerebrovascular events [MACCE]), and quality of life (QOL) during the 5-year follow-up. RESULTS: At 5 years, mean LVEDD was 56.5 ± 5.7 mm with PMA versus 60.6 ± 4.6 mm with RA (mean change from baseline -5.8 ± 4.1 mm and -0.2 ± 2.3 mm, respectively; p < 0.001). Ejection fraction was 44.1 ± 6% in the PMA group versus 39.9 ± 3.9% in the RA group (mean change from baseline 8.8 ± 5.9% and 2.5 ± 4.3%, respectively; p < 0.001). There was no statistically significant difference in mortality at 5 years, but freedom from MACCE favored PMA in the last year of follow-up. PMA significantly reduced tenting height, tenting area, and interpapillary distance soon after surgery and for the long-term, and significantly lowered moderate-to-severe MR recurrence. No differences were found in QOL measures. CONCLUSIONS: Compared with RA only, PMA exerted a long-term beneficial effect on left ventricular remodeling and more effectively restored the mitral valve geometric configuration in ischemic MR, which improved long-term cardiac outcomes, but did not produce differences in overall mortality and QOL.


Subject(s)
Mitral Valve Annuloplasty/methods , Mitral Valve Insufficiency/surgery , Myocardial Ischemia/etiology , Papillary Muscles/surgery , Coronary Artery Bypass , Echocardiography , Female , Follow-Up Studies , Heart Failure/etiology , Heart Ventricles/diagnostic imaging , Humans , Italy/epidemiology , Male , Middle Aged , Mitral Valve Annuloplasty/instrumentation , Mitral Valve Insufficiency/mortality , Myocardial Ischemia/surgery , Patient Readmission/statistics & numerical data , Prospective Studies , Quality of Life , Recurrence , Reoperation/statistics & numerical data , Stroke Volume , Ventricular Remodeling
19.
Clin Ther ; 27(9): 1411-9, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16291414

ABSTRACT

BACKGROUND: Plasma concentrations of intercellular adhesion molecule-1 [ICAM-1], vascular cell adhesion molecule-1 [VCAM-1], and E-selectin may increase after percutaneous coronary intervention (PCI). OBJECTIVE: The aim of this study was to assess whether anti-inflammatory treatment with steroid-eluting stents influenced concentrations of adhesion molecules after PCI in patients with unstable coronary syndromes. METHODS: Consecutive patients with unstable coronary syndromes who were prospectively assigned to undergo implantation of a dexamethasone-eluting stent were compared with a control group of consecutive historical patients who received a similar non-drugeluting stent in the period immediately preceding availability of the steroid-eluting stents. Circulating concentrations of adhesion molecules were measured before the procedure and at 6 and 24 hours after implantation. RESULTS: The study included 50 patients who received the dexamethasone-eluting stent (41 men, 9 women; mean [SD] age, 60 [9] years) and 50 historical controls who received the non-drug-eluting stent (40 men, 10 women; mean age, 63 [11] years). Baseline values for the adhesion molecules were similar in the 2 groups: in patients who received the dexamethasone-eluting stent, mean (SD) values were 210 (41) ng/mL for ICAM-1, 637 (119) ng/mL for VCAM-1, and 46 (9) ng/mL for E-selectin; in the control group, the corresponding values were 218 (42), 618 (140), and 43 (10) ng/mL. Circulating concentrations at 24 hours were significantly lower in patients receiving the drug-eluting stent compared with controls for both ICAM-1 (279 [49] vs 338 [60] ng/mL, respectively; P = 0.001) and VCAM-1 (772 [163] vs 896 [207] ng/mL; P = 0.004). There was no significant between-group difference in E-selectin concentrations at 24 hours (60 [10] vs 56 [11] ng/mL). CONCLUSION: In this study, dexamethasone-eluting stents reduced plasma concentrations of ICAM-1 and VCAM-1 after PCI in patients with unstable coronary syndromes compared with historical controls.


Subject(s)
Angioplasty, Balloon, Coronary , Anti-Inflammatory Agents/therapeutic use , Cell Adhesion Molecules/blood , Coronary Artery Disease/drug therapy , Coronary Disease/drug therapy , Dexamethasone/therapeutic use , Analysis of Variance , Angina, Unstable/blood , Angina, Unstable/therapy , Anti-Inflammatory Agents/administration & dosage , Coronary Artery Disease/blood , Coronary Disease/blood , Coronary Disease/therapy , Dexamethasone/administration & dosage , Female , Humans , Intercellular Adhesion Molecule-1/blood , Male , Myocardial Infarction/blood , Myocardial Infarction/therapy , Stents , Syndrome , Time Factors , Vascular Cell Adhesion Molecule-1/blood , White People
20.
J Heart Valve Dis ; 14(2): 268-70, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15792191

ABSTRACT

The case is reported of a patient with a symptomatic thrombophilic status and a positivity for both direct and indirect antiglobulin tests who underwent mitral valve replacement. Because of the antiglobulin test positivity, the patient was enrolled in a preoperative autologous blood donation program in order to avoid exposure to allogenic blood. The operation was successful, and the postoperative course uneventful; the patient was discharged on postoperative day 6 and prescribed oral anticoagulation. This case suggests that a safe valve replacement can be carried out even in the presence of a symptomatic thrombophilic preoperative status.


Subject(s)
Heart Valve Prosthesis , Mitral Valve Insufficiency/complications , Mitral Valve Insufficiency/surgery , Thrombophilia/complications , Adult , Coombs Test , Humans , Male , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL