Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 193
Filter
1.
Curr Res Transl Med ; 64(2): 97-106, 2016.
Article in English | MEDLINE | ID: mdl-27316393

ABSTRACT

Stem cell-based therapy is currently tested in several trials of chronic heart failure. The main question is to determine how its implementation could be extended to standard clinical practice. To answer this question, it is helpful to capitalize on the three main lessons drawn from the accumulated experience, both in the laboratory and in the clinics. Regarding the cell type, the best outcomes seem to be achieved by cells the phenotype of which closely matches that of the target tissue. This argues in favor of the use of cardiac-committed cells among which the pluripotent stem cell-derived cardiac progeny is particularly attractive. Regarding the mechanism of action, there has been a major paradigm shift whereby cells are no longer expected to structurally integrate within the recipient myocardium but rather to release biomolecules that foster endogenous repair processes. This implies to focus on early cell retention, rather than on sustained cell survival, so that the cells reside in the target tissue long enough and in sufficient amounts to deliver the factors underpinning their action. Biomaterials are here critical adjuncts to optimize this residency time. Furthermore, the paracrine hypothesis gives more flexibility for using allogeneic cells in that targeting an only transient engraftment requires to delay, and no longer to avoid, rejection, which, in turn, should simplify immunomodulation regimens. Regarding manufacturing, a broad dissemination of cardiac cell therapy requires the development of automated systems allowing to yield highly reproducible cell products. This further emphasizes the interest of allogeneic cells because of their suitability for industrially-relevant and cost-effective scale-up and quality control procedures. At the end, definite confirmation that the effects of cells can be recapitulated by the factors they secrete could lead to acellular therapies whereby factors alone (possibly clustered in extracellular vesicles) would be delivered to the patient. The production process of these cell-derived biologics would then be closer to that of a pharmaceutical compound, which could streamline the manufacturing and regulatory paths and thereby facilitate an expended clinical use.


Subject(s)
Heart Failure/therapy , Stem Cell Transplantation , Allografts , Animals , Cell Separation/methods , Embryonic Stem Cells/transplantation , Extracellular Vesicles/transplantation , Graft Survival , Humans , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/therapeutic use , Mice , MicroRNAs/physiology , MicroRNAs/therapeutic use , Myocardium/cytology , Myocytes, Cardiac/transplantation , Paracrine Communication , Pluripotent Stem Cells/transplantation , Randomized Controlled Trials as Topic , Stem Cell Transplantation/adverse effects , Stem Cell Transplantation/economics , Stem Cell Transplantation/methods , Stem Cells/metabolism , Teratoma/etiology , Teratoma/prevention & control , Tissue Engineering
2.
Am J Transplant ; 16(2): 454-67, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26492394

ABSTRACT

Implantation of embryonic stem cells (ESCs) and their differentiated derivatives into allogeneic hosts triggers an immune response that represents a hurdle to clinical application. We established in autoimmunity and in transplantation that CD3 antibody therapy induces a state of immune tolerance. Promising results have been obtained with CD3 antibodies in the clinic. In this study, we tested whether this strategy can prolong the survival of undifferentiated ESCs and their differentiated derivatives in histoincompatible hosts. Recipients of either mouse ESC-derived embryoid bodies (EBs) or cardiac progenitors received a single short tolerogenic regimen of CD3 antibody. In immunocompetent mice, allogeneic EBs and cardiac progenitors were rejected within 20-25 days. Recipients treated with CD3 antibody showed long-term survival of implanted cardiac progenitors or EBs. In due course, EBs became teratomas, the growth of which was self-limited. Regulatory CD4(+)FoxP3(+) T cells and signaling through the PD1/PDL1 pathway played key roles in the CD3 antibody therapeutic effect. Gene profiling emphasized the importance of TGF-ß and the inhibitory T cell coreceptor Tim3 to the observed effect. These results demonstrate that CD3 antibody administered alone promotes prolonged survival of allogeneic ESC derivatives and thus could prove useful for enhancing cell engraftment in the absence of chronic immunosuppression.


Subject(s)
Antibodies, Monoclonal/pharmacology , CD3 Complex/immunology , Embryoid Bodies/immunology , Embryonic Stem Cells/immunology , Graft Rejection/immunology , Immune Tolerance/immunology , T-Lymphocytes/immunology , Animals , Cell Differentiation , Embryoid Bodies/drug effects , Embryonic Stem Cells/drug effects , Gene Expression Profiling , Graft Survival , Immune Tolerance/drug effects , Immunosuppression Therapy , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes/drug effects , Transplantation, Homologous
4.
J Thorac Cardiovasc Surg ; 131(2): 352-6, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16434264

ABSTRACT

OBJECTIVE: We sought to evaluate the effects of pexelizumab, a C5 complement inhibitor, on death and myocardial infarction in patients undergoing combined aortic valve replacement and coronary artery bypass grafting surgery. METHODS: The Pexelizumab for Reduction in Myocardial Infarction and Mortality in Coronary Artery Bypass Graft surgery trial, a phase III prospective, randomized, double-blind, placebo-controlled study, enrolled 3099 patients at 205 centers. The primary end point was the composite of death, myocardial infarction, or both at postoperative day 30 in patients undergoing coronary artery bypass grafting without valve surgery. Postoperative myocardial infarction was defined as a creatine kinase MB fraction value of 100 ng/mL or greater, Q-wave myocardial infarction with a creatine kinase MB fraction value of 70 ng/mL or greater, or new Q-wave evidence of myocardial infarction by postoperative day 30. Because patients undergoing coronary artery bypass grafting with a valve procedure were not included in the primary population, separate analysis of death and myocardial infarction was conducted in 218 patients undergoing combined aortic valve replacement and coronary artery bypass grafting surgery. RESULTS: Of the 353 patients randomized to any valve procedure, 106 (61%) underwent combined aortic valve replacement and coronary artery bypass grafting in the pexelizumab treatment group compared with 112 (63%) patients in the placebo group. Coronary artery bypass grafting was performed with 1 or more internal thoracic artery grafts in 139 (64%) patients and with 1 or more saphenous vein grafts in 179 (82%) patients. There were 4 (3.8%) deaths in the pexelizumab group versus 11 (9.9%) in the placebo group by postoperative day 30 and 6 (5.7%) deaths in the active group versus 16 (14.4%) in the placebo group by postoperative day 180 (P =.107 and P =.043, respectively, Fisher exact test). The incidence of myocardial infarction 30 days after surgical intervention was identical in the 2 groups, but the study was not designed to detect differences in this cohort of patients. CONCLUSIONS: Inhibition of complement activation by pexelizumab resulted in a decreased mortality at 180 days among 218 patients who underwent combined aortic valve replacement and coronary artery bypass grafting surgery. Additional studies are warranted to confirm this decrease in mortality with pexelizumab in combined aortic valve replacement and coronary artery bypass grafting procedures.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Aortic Valve/surgery , Complement Inactivating Agents/therapeutic use , Coronary Artery Bypass/mortality , Heart Valve Prosthesis Implantation/mortality , Myocardial Infarction/prevention & control , Aged , Antibodies, Monoclonal, Humanized , Cardiopulmonary Bypass , Coronary Artery Bypass/adverse effects , Double-Blind Method , Female , Heart Valve Prosthesis Implantation/adverse effects , Humans , Male , Myocardial Infarction/etiology , Postoperative Complications , Single-Chain Antibodies
5.
Arch Mal Coeur Vaiss ; 98(6): 649-54, 2005 Jun.
Article in French | MEDLINE | ID: mdl-16007819

ABSTRACT

Cell therapy in cardiology is already a reality, as evidenced by the number of ongoing clinical trials. These studies entail administration of either skeletal myoblasts in patients with severe ischemic left ventricular dysfunction or of bone marrow-derived cells in patients with acute myocardial infarction and in whom cell therapy is an adjunct to a percutaneous revascularization procedure. The techniques of preparation, expansion and storage of myoblasts are now quite effective. The problem is simpler for bone marrow cells as in most studies, the procedure is limited to an iliac crest biopsy followed by reinjection of the crude, unfractionated bone marrow, as routinely done in clinical haematology since many years. The results of these studies are not yet fully available. Some of them have been enthusiastically reported to be positive but should be interpreted cautiously because of the usually small sample sizes and the common lack of randomisation and double-blind assessment of outcomes. Thus, the fact that cell therapy has now become a reality should not lead to underscore the yet unsettled fundamental issue, i.e., the ability of this novel mode of therapy to truly regenerate areas of necrotic myocardium and restore function in once akinetic territories. From this standpoint, cell therapy is still a dream. Since the beginning, it has been clear that myoblasts were exclusively committed to differentiate into myotubes, without any evidence for a phenotypic conversion into cardiomyocytes. Although the debate is more controversial for bone marrow cells, the reliance on accurate genetic methods of cell tracking has led to increasingly challenge the purported plasticity of these cells. This by no means implies that cell therapy does not exert beneficial effects that could be mediated by alternate mechanisms like limitation of remodelling of paracrine effects. The basic point is that neither skeletal myoblasts nor bone marrow cells fulfill the major criteria required for a true cardiac regeneration: a coupling of the grafted cells with those of the recipient myocardium and the subsequent generation of a contractile force. It is therefore critical to go on exploring other paths, among which embryonic stem cells are particularly attractive.


Subject(s)
Heart Failure/therapy , Myoblasts/transplantation , Stem Cell Transplantation/trends , Bone Marrow Cells , Cell Culture Techniques , Clinical Trials as Topic , Humans , Myocardium/cytology , Phenotype , Ventricular Function, Left , Ventricular Remodeling
6.
Arch Mal Coeur Vaiss ; 98(6): 637-48, 2005 Jun.
Article in French | MEDLINE | ID: mdl-16007818

ABSTRACT

Despite the improvement in revascularisation techniques, coronary artery disease remains the principal aetiology of cardiac failure in developed countries. The therapeutic management of cardiac failure has been improved over recent years, yet cardiac failure is still associated with significant morbidity and mortality. As cardiac transplantation lacks donors, techniques that allow myocardial regeneration represent an attractive alternative. To date, several types of cells are under study and are suitable for implantation into infarcted myocardium (myoblasts, medullary stem cells...). Following good preclinical study results, the first human cell therapy trials, using the intramyocardial route, have begun, in the course of aorto-coronary bypass surgery in patients with chronic ischaemic cardiopathy and little altered left ventricular function, and then in those with ventricular dysfunction. Different modes of administration of these cell therapy products are under study and could be envisaged in clinical situations such as just after infarction in order to improve ventricular remodelling with an intracoronary injection technique. As for every new treatment, there are numerous problems to resolve, from understanding the relevant mechanisms of cellular transplantation, to the secondary effects that it could entail. Nevertheless, cardiac cellular transplantation is expanding rapidly and with the evolution of techniques it allows a glimpse of a new field of treatment for cardiac failure.


Subject(s)
Cell Transplantation/methods , Cell Transplantation/trends , Coronary Artery Disease/therapy , Myocardial Ischemia/therapy , Clinical Trials as Topic , Humans , Myocardium/cytology , Stem Cell Transplantation , Ventricular Dysfunction, Left , Ventricular Remodeling
9.
Presse Med ; 31(33): 1569-76, 2002 Oct 12.
Article in French | MEDLINE | ID: mdl-12422486

ABSTRACT

THE DEVELOPMENT OF CELL THERAPY: The stakes in the management of heart failure have become such that new therapeutic strategies have to be developed. Among the cell, molecular and genetic approaches aimed at reinforcing the deficient heart muscle by restoring its functional potential, cell therapy is the favored option in clinical application perspectives. IN THE FIELD OF ISCHEMIC HEART FAILURE: All the experimental data have shown that implantation of contractile cells in the post-infarction areas led to improved cardiac function. INTERESTING PRELIMINARY RESULTS: For ethical and immuno-biological reasons, the successful transplantation of autologous skeletal myoblasts has led our team to conduct a phase I clinical trial. Although the results of this study are preliminary with regard to cardiac function, they suggest the validity of the cell transplantation concept and allow one to hope that this new treatment method will have its place among the therapeutic arms of heart failure.


Subject(s)
Heart Failure/therapy , Myoblasts, Skeletal/transplantation , Myocardium/pathology , Myocytes, Cardiac/physiology , Humans
12.
Arch Mal Coeur Vaiss ; 95(12): 1219-25, 2002 Dec.
Article in English | MEDLINE | ID: mdl-12611044

ABSTRACT

Post-ischemic heart failure is becoming a major issue for public health in occidental countries and therapeutical options are limited. Therefore cell transplantation was developed as an alternative strategy to improve cardiac structure and function. This review describes the multiple cell types and clinical trials considered for use in this indication. The transplantation of fetal or neonatal cardiomyocytes has proven to be functionally successful, but ethical as well as technical reasons make their clinical use limited. Recent reports, however, suggested that adult autologous cardiomyocytes could be prepared from stem cells present in various mesenchymal tissues. Alternatively, endothelial progenitors originating from bone marrow or peripheral blood could promote the neoangiogenesis within the scar tissue. Finally, the transplantation of skeletal muscle cells (SMC) in the infarcted area improved myocardial function, in correlation with the development of skeletal muscle tissue in various animal models. The latter results paved the way for the development of a first phase I clinical trial of SMC transplantation in patients with severe ischemic heart failure. It required the scale-up of human cell production according to Good Manufacturing Procedures, it started in June 2000 in Paris and was terminated in November 2001, and it was followed by several others. The results were encouraging and prompted the onset of a blinded, multicentric phase II clinical trial for SMC transplantation. Meanwhile, clinical trials also evaluate the safety and efficacy of various cells types originating from the bone marrow.


Subject(s)
Cell Transplantation , Heart Failure/therapy , Myocardial Ischemia/complications , Neovascularization, Physiologic , Bone Marrow Transplantation , Clinical Trials as Topic , Humans , Muscle, Skeletal/cytology , Myocardium/cytology , Peripheral Blood Stem Cell Transplantation
13.
Hypertension ; 38(6): 1413-5, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11751727

ABSTRACT

Intramyocardial cell grafting aims to limit the consequences of the loss of contractile function of a damaged left ventricle. Its functional efficacy is suggested by a wealth of experimental data using multiple evaluation techniques in different animal species. Intramyocardial injections of cultured fetal cardiomyocytes after infarction increase the ejection fraction. Cultured autologous skeletal myoblasts, which do not raise immunologic, ethical, tumorigenesis, or donor availability problems, improve ventricular function to a similar extent. The presence of connexin-43 is demonstrated between fetal (but not myoblast) grafted cells and host myocytes. Thus, the mechanisms of this beneficial effect (direct systolic effect, paracrine factors, passive girdling effect, and decrease in wall stress) remain controversial. These encouraging results have opened the way to the first clinical trial in patients with low ejection fractions, akinetic and nonviable postinfarction scars, and indications for coronary artery bypasses in remote, viable, and ischemic areas. Large-scale cell expansion allows a yield of >10(9) myoblasts from a single human muscular biopsy. Cultured autologous myoblasts are directly administered by multiple injections within and around the infarcted area during open-chest surgery. Preliminary postoperative observations show an improvement in ejection fraction, reappearance of a systolic thickening of the grafted scars, and a new-onset metabolic viability within this area. Thus, this new procedure might become a useful adjunct to current treatments of severe ischemic heart failure.


Subject(s)
Myocardial Ischemia/surgery , Myocardium/cytology , Animals , Cell Differentiation , Cell Transplantation , Humans , Myocardial Contraction , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Regeneration
14.
Circulation ; 104(12 Suppl 1): I223-8, 2001 Sep 18.
Article in English | MEDLINE | ID: mdl-11568060

ABSTRACT

BACKGROUND: There is compelling experimental evidence that autologous skeletal muscle (SM) cell transplantation improves postinfarction cardiac function. This study assessed whether this benefit is still manifested in the clinically relevant setting of a treatment by ACE inhibitors. METHODS AND RESULTS: A myocardial infarction was created in 99 rats by coronary artery ligation. They were divided into 4 groups. Two groups did not receive any drug and were intramyocardially injected 7 days after the infarct with either culture medium alone (control rats, n=16) or autologous SM cells (2.3x10(6) myoblasts) previously expanded ex vivo for 7 days (myoblasts, n=24). Two other groups received the ACE inhibitor perindoprilat (1 mg. kg(-1). d(-1)), started the day of the infarct and continued uninterruptedly thereafter, and underwent time-matched procedures, that is, they were intramyocardially injected at 7 days after infarction with either culture medium alone (ACE inhibitors, n=22) or autologous SM cells (2.5x10(6) myoblasts) previously expanded ex vivo for 7 days (ACE inhibitors+myoblasts, n=37). Left ventricular function was assessed by 2D echocardiography. At the end of the 2-month study, left ventricular ejection fraction (%, mean+/-SEM) was increased in all groups (myoblasts, 37.4+/-1.2; ACE inhibitors, 31.6+/-1.7; ACE inhibitors+myoblasts, 43.9+/-1.4) compared with that in control rats (19.8+/-0.7) (P<0.0001). The improvement in ejection fraction was similar in the ACE inhibitor and the myoblast groups (31.6+/-1.7 versus 37.4+/-1.2, P=0.0636). However, in the ACE inhibitor+myoblast group, this improvement was greater than that seen in hearts receiving either treatment alone (43.9+/-1.4 versus 31.6+/-1.7 in the ACE inhibitor group and 43.9+/-1.4. versus 37.4+/-1.2 in the myoblast group, P<0.0001 and P=0.0084, respectively). CONCLUSIONS: These data provide further support for the clinical relevance of autologous SM cell transplantation in that its cardioprotective effects are additive to those observed with ACE inhibitors.


Subject(s)
Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Indoles/therapeutic use , Muscle, Skeletal/transplantation , Myocardial Infarction/therapy , Animals , Cell Count , Disease Models, Animal , Echocardiography , Heart Ventricles/diagnostic imaging , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Immunohistochemistry , Male , Muscle, Skeletal/cytology , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/physiopathology , Rats , Rats, Wistar , Stroke Volume/drug effects , Transplantation, Autologous , Treatment Outcome , Ventricular Function, Left/drug effects
15.
Ann Thorac Surg ; 72(1): 107-12; discussion 112-3, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11465162

ABSTRACT

BACKGROUND: Cardiac harvest teams are usually committed to immediately transfer the explanted donor heart into its cold storage solution. We tested the opposite hypothesis that a brief prestorage episode of heat-enhanced ischemic preconditioning could be protective. METHODS: Fifty-three isolated isovolumic rat hearts underwent 4 hours of cold (4 degrees C) storage in the Celsior preservation solution and 2 hours of reperfusion. Control hearts were immediately immersed after arrest. In the 3 treated groups, 2 customized thermal probes were first applied onto the left ventricular free wall of the explanted heart at 22 degrees C, 37 degrees C or 42.5 degrees C for 15 minutes before immersion. Each of the selected temperatures were monitored at the probe-tissue interface by a thermocouple. RESULTS: Whereas base line end-diastolic pressure was set at = 8 mm Hg in all groups, it increased during reperfusion (mean +/- SEM) to 28+/-3, 27+/-3, 17+/-1, and 18+/-2 mm Hg in control, 22 degrees C, 37 degrees C and 42.5 degrees C-heated hearts, respectively (37 degrees C and 42.5 degrees C: p < 0.05 versus controls and 22 degrees C). Slopes of pressure-volume curves featured similar patterns. Likewise, reperfusion dP/dT (mm Hg/s(-1)) was significantly lower in control and 22 degrees C hearts (1,119+/-114 and 1,076+/-125, respectively) than in those undergoing prestorage heating to 37 degrees C and 42.5 degrees C (1,545+/-109 and 1,719+/-111, p < 0.05 and p < 0.01 versus controls and 22 degrees C, respectively). Western blot analysis of LV samples did not demonstrate any upregulation of HSP 72 in either group. Conversely, the involvement of preconditioning was evidenced by the loss of protection in the 42.5 degrees C-heated hearts when, in 2 additional groups, the storage solution was supplemented with either the protein kinase C and tyrosine kinase inhibitors chelerythrine (5 micromol/L) and genistein (50 micromol/L) or the mitochondrial K(ATP) channel inhibitor 5-hydroxydecanoate (200 micromol/L). CONCLUSIONS: A brief period of postexplant ischemia with enhancement by topical heating ("backtable preconditioning") could be a simple and effective means of improving the functional recovery of heart transplants.


Subject(s)
Heart Transplantation , Ischemic Preconditioning , Myocardial Contraction/physiology , Organ Preservation , Animals , Diastole/physiology , HSP72 Heat-Shock Proteins , Heat-Shock Proteins/metabolism , Heating , Male , Rats , Rats, Wistar , Systole/physiology , Ventricular Function, Left/physiology
16.
Arch Mal Coeur Vaiss ; 94(3): 180-2, 2001 Mar.
Article in French | MEDLINE | ID: mdl-11338251

ABSTRACT

The authors report the first intramyocardial transplantation of autologous skeletal myoblasts in a patient with severe ischaemic cardiac failure. The encouraging result after eight months' follow-up underlines the potential of this new approach.


Subject(s)
Cardiac Output, Low/therapy , Cell Transplantation , Muscle, Skeletal/cytology , Muscle, Skeletal/transplantation , Myocardial Ischemia/complications , Aged , Cardiac Output, Low/pathology , Electrocardiography , Humans , Male , Transplantation, Autologous , Treatment Outcome
17.
Ann Thorac Surg ; 71(3): 844-50; discussion 850-1, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11269463

ABSTRACT

BACKGROUND: This study assessed the extent to which the initial degree of functional impairment and the number of injected cells may influence the functional improvement provided by autologous skeletal myoblast transplantation into infarcted myocardium. METHODS: One week after left coronary artery ligation, 44 rats received into the infarcted scar, autologous skeletal myoblasts expanded in vitro for 7 days (mean, 3.5 x 10(6), n = 21), or culture medium alone (controls, n = 23). Left ventricular function was assessed by two-dimensional echocardiography. RESULTS: When transplanted hearts were stratified according to their baseline ejection fraction, a significant improvement occurred at 2 months in the less than 25% (from 21.4% to 37%), 25% to 35% (from 29% to 43.8%), and in the 35% to 40% (from 37.2% to 41.7%) groups, compared to controls (p = 0.048, 0.0057, and 0.034, respectively), but not in the more than 40% stratum. A significant linear relationship was found between the improvement in ejection fraction and the number of injected myoblasts, both at 1 and 2 months after transplantation (p < 0.0001). CONCLUSIONS: Autologous myoblast transplantation is functionally effective over a wide range of postinfarct ejection fractions, including in the sickest hearts provided that they are injected with a sufficiently high number of cells.


Subject(s)
Cell Transplantation , Muscle, Skeletal/cytology , Myocardial Infarction/therapy , Animals , Male , Rats , Rats, Wistar , Treatment Outcome
18.
Lancet ; 357(9252): 279-80, 2001 Jan 27.
Article in English | MEDLINE | ID: mdl-11214133

ABSTRACT

Intramyocardial skeletal muscle transplantation has been shown experimentally to improve heart function after infarction. We report success with this procedure in a patient with severe ischaemic heart failure. We implanted autologous skeletal myoblasts into the postinfarction scar during coronary artery bypass grafting of remote myocardial areas. 5 months later, there was evidence of contraction and viability in the grafted scar on echocardiography and positron emission tomography. Although this result is encouraging, it requires validation by additional studies.


Subject(s)
Blastomeres/transplantation , Cardiac Output, Low/surgery , Muscle, Skeletal/cytology , Myocardial Infarction/complications , Aged , Cardiac Output, Low/etiology , Cardiac Output, Low/physiopathology , Echocardiography , Heart/diagnostic imaging , Humans , Male , Tomography, Emission-Computed , Transplantation, Autologous
19.
Eur J Cardiothorac Surg ; 19(1): 108-10, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11163575

ABSTRACT

Determining cerebral blood flow during circulatory arrest in patients undergoing surgery for aortic aneurysms has been traditionally based on body weight. We report the use of per-aortic antegrade cerebral perfusion regulated by perfusion pressure using a triple lumen cardioplegia catheter thus optimising cerebral flow.


Subject(s)
Aortic Aneurysm, Thoracic/surgery , Blood Flow Velocity/physiology , Blood Pressure/physiology , Brain/blood supply , Catheters, Indwelling , Heart Arrest, Induced/instrumentation , Blood Vessel Prosthesis Implantation , Cardiopulmonary Bypass/instrumentation , Humans
20.
J Thorac Cardiovasc Surg ; 121(1): 155-62, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11135172

ABSTRACT

OBJECTIVE: This study was designed to compare ischemic preconditioning with opening of mitochondrial adenosine triphosphate-sensitive potassium channels and Na(+)/H(+) exchange inhibition in an isolated heart model of cold storage, simulating the situation of cardiac allografts. METHODS: Sixty-seven isolated isovolumic buffer-perfused rat hearts were arrested with and stored in Celsior solution (Imtix-Sangstat) at 4 degrees C for 4 hours before a 2-hour reperfusion. Group I hearts served as controls and were arrested with and stored in Celsior solution. In group II, hearts were preconditioned by two 5-minute episodes of global ischemia, each separated by 5 minutes of reperfusion before arrest with Celsior solution. Group III hearts were arrested with and stored in Celsior solution supplemented with 100 micromol/L of the mitochondrial adenosine triphosphate-sensitive potassium channel opener diazoxide. In group IV, hearts received an infusion of diazoxide (30 micromol/L) during the first 15 minutes of reperfusion. Group V hearts underwent a protocol combining both interventions used in groups III and IV. In group VI, hearts were arrested with and stored in Celsior solution supplemented with 1 micromol/L of the Na(+)/H(+) exchange inhibitor cariporide. Group VII hearts received an infusion of cariporide (1 micromol/L) during the first 15 minutes of reperfusion. In group VIII, hearts underwent a protocol combining both interventions used in groups VI and VII. Group IX hearts were ischemically preconditioned as in group II, and sustained Na(+)/H(+) exchange inhibition during both storage and early reperfusion was used as in group VIII. RESULTS: On the basis of comparisons of postischemic left ventricular contractility and diastolic function, coronary flow, total creatine kinase leakage, and myocardial water content, values indicative of improved protection were obtained by combining ischemic preconditioning with Na(+)/H(+) exchange inhibition by cariporide given during storage and initial reperfusion. The endothelium-dependent vasodilatory postischemic responses to 5-hydroxytryptamine or acetylcholine and endothelium-independent responses to papaverine were not affected by these interventions. CONCLUSIONS: These data suggest that cardioprotection conferred by the Na(+)/H(+) exchange inhibitor cariporide is additive to that of ischemic preconditioning and might effectively contribute to improve donor heart preservation during cardiac transplantation.


Subject(s)
Adenosine Triphosphate/agonists , Heart Transplantation , Ischemic Preconditioning, Myocardial/methods , Mitochondria, Heart/metabolism , Myocardial Ischemia/prevention & control , Potassium Channels/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Anti-Arrhythmia Agents/pharmacology , Coronary Circulation/drug effects , Creatine Kinase/metabolism , Diazoxide/pharmacology , Disaccharides/pharmacology , Electrolytes/pharmacology , Glutamates/pharmacology , Glutathione/pharmacology , Guanidines/pharmacology , Heart Arrest, Induced/methods , Heart Transplantation/adverse effects , Histidine/pharmacology , In Vitro Techniques , Male , Mannitol/pharmacology , Mitochondria, Heart/drug effects , Myocardial Contraction/drug effects , Myocardial Ischemia/etiology , Myocardial Ischemia/metabolism , Myocardial Ischemia/physiopathology , Organ Preservation/methods , Organ Preservation Solutions/pharmacology , Potassium Channels/metabolism , Rats , Rats, Wistar , Sodium-Potassium-Exchanging ATPase/metabolism , Sulfones/pharmacology , Transplantation, Homologous , Vasodilator Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...