Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Basic Res Cardiol ; 110(1): 456, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25480109

ABSTRACT

The ß1-adrenergic antagonist metoprolol improves cardiac function in animals and patients with chronic heart failure, isolated mitral regurgitation (MR), and ischemic heart disease, though the molecular mechanisms remain incompletely understood. Metoprolol has been reported to upregulate cardiac expression of ß3-adrenergic receptors (ß3AR) in animal models. Myocardial ß3AR signaling via neuronal nitric oxide synthase (nNOS) activation has recently emerged as a cardioprotective pathway. We tested whether chronic ß1-adrenergic blockade with metoprolol enhances myocardial ß3AR coupling with nitric oxide-stimulated cyclic guanosine monophosphate (ß3AR/NO-cGMP) signaling in the MR-induced, volume-overloaded heart. We compared the expression, distribution, and inducible activation of ß3AR/NO-cGMP signaling proteins within myocardial membrane microdomains in dogs (canines) with surgically induced MR, those also treated with metoprolol succinate (MR+ßB), and unoperated controls. ß3AR mRNA transcripts, normalized to housekeeping gene RPLP1, increased 4.4 × 10(3)- and 3.2 × 10(2)-fold in MR and MR+ßB hearts, respectively, compared to Control. Cardiac ß3AR expression was increased 1.4- and nearly twofold in MR and MR+ßB, respectively, compared to Control. ß3AR was detected within caveolae-enriched lipid rafts (Cav3(+)LR) and heavy density, non-lipid raft membrane (NLR) across all groups. However, in vitro selective ß3AR stimulation with BRL37344 (BRL) triggered cGMP production within only NLR of MR+ßB. BRL induced Ser (1412) phosphorylation of nNOS within NLR of MR+ßB, but not Control or MR, consistent with detection of NLR-specific ß3AR/NO-cGMP coupling. Treatment with metoprolol prevented MR-associated oxidation of NO biosensor soluble guanylyl cyclase (sGC) within NLR. Metoprolol therapy also prevented MR-induced relocalization of sGCß1 subunit away from caveolae, suggesting preserved NO-sGC-cGMP signaling, albeit without coupling to ß3AR, within MR+ßB caveolae. Chronic ß1-blockade is associated with myocardial ß3AR/NO-cGMP coupling in a microdomain-specific fashion. Our canine study suggests that microdomain-targeted enhancement of myocardial ß3AR/NO-cGMP signaling may explain, in part, ß1-adrenergic antagonist-mediated preservation of cardiac function in the volume-overloaded heart.


Subject(s)
Adrenergic beta-1 Receptor Antagonists/pharmacology , Cyclic GMP/physiology , Mitral Valve Insufficiency/drug therapy , Nitric Oxide/physiology , Receptors, Adrenergic, beta-3/physiology , Signal Transduction/physiology , Adrenergic beta-1 Receptor Antagonists/therapeutic use , Animals , Chronic Disease , Dogs , Ethanolamines/pharmacology , Guanylate Cyclase/metabolism , Membrane Microdomains/drug effects , Membrane Microdomains/physiology , Metoprolol/pharmacology , Mitral Valve Insufficiency/physiopathology , Nitric Oxide Synthase Type I/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Ventricular Function, Left
2.
J Thorac Cardiovasc Surg ; 143(4): 962-6, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22264415

ABSTRACT

OBJECTIVES: Ventricular remodeling after myocardial infarction begins with massive extracellular matrix deposition and resultant fibrosis. This loss of functional tissue and stiffening of myocardial elastic and contractile elements starts the vicious cycle of mechanical inefficiency, adverse remodeling, and eventual heart failure. We hypothesized that stromal cell-derived factor 1α (SDF-1α) therapy to microrevascularize ischemic myocardium would rescue salvageable peri-infarct tissue and subsequently improve myocardial elasticity. METHODS: Immediately after left anterior descending coronary artery ligation, mice were randomly assigned to receive peri-infarct injection of either saline solution or SDF-1α. After 6 weeks, animals were killed and samples were taken from the peri-infarct border zone and the infarct scar, as well as from the left ventricle of noninfarcted control mice. Determination of tissues' elastic moduli was carried out by mechanical testing in an atomic force microscope. RESULTS: SDF-1α-treated peri-infarct tissue most closely approximated the elasticity of normal ventricle and was significantly more elastic than saline-treated peri-infarct myocardium (109 ± 22.9 kPa vs 295 ± 42.3 kPa; P < .0001). Myocardial scar, the strength of which depends on matrix deposition from vasculature at the peri-infarct edge, was stiffer in SDF-1α-treated animals than in controls (804 ± 102.2 kPa vs 144 ± 27.5 kPa; P < .0001). CONCLUSIONS: Direct quantification of myocardial elastic properties demonstrates the ability of SDF-1α to re-engineer evolving myocardial infarct and peri-infarct tissues. By increasing elasticity of the ischemic and dysfunctional peri-infarct border zone and bolstering the weak, aneurysm-prone scar, SDF-1α therapy may confer a mechanical advantage to resist adverse remodeling after infarction.


Subject(s)
Angiogenesis Inducing Agents/pharmacology , Chemokine CXCL12/pharmacology , Elasticity Imaging Techniques/methods , Heart Ventricles/drug effects , Microscopy, Atomic Force , Myocardial Infarction/drug therapy , Myocardium/pathology , Neovascularization, Physiologic/drug effects , Animals , Biomechanical Phenomena , Disease Models, Animal , Elastic Modulus , Fibrosis , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Male , Mice , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Time Factors , Ventricular Remodeling/drug effects
3.
Ann Thorac Surg ; 92(4): 1501-3, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21958802

ABSTRACT

We report transventricular mitral valve operations in 2 patients with severe mitral regurgitation and postinfarction left ventricular rupture and pseudoaneurysm. The first patient had direct papillary muscle involvement necessitating replacement of the mitral valve. The second patient had indirect mitral involvement allowing for placement of an atrial mitral annuloplasty ring via the left ventricle. Both patients showed no mitral valve regurgitation after replacement or repair and had uneventful postoperative recoveries. These cases demonstrate a feasible, alternative, transventricular approach to mitral valve replacement and repair.


Subject(s)
Cardiac Catheterization/methods , Heart Ventricles/surgery , Mitral Valve Annuloplasty/methods , Mitral Valve Insufficiency/surgery , Aged , Female , Follow-Up Studies , Humans , Male , Middle Aged , Mitral Valve Insufficiency/diagnosis , Severity of Illness Index
4.
Circulation ; 124(11 Suppl): S18-26, 2011 Sep 13.
Article in English | MEDLINE | ID: mdl-21911811

ABSTRACT

BACKGROUND: Experimentally, exogenous administration of recombinant stromal cell-derived factor-1α (SDF) enhances neovasculogenesis and cardiac function after myocardial infarction. Smaller analogs of SDF may provide translational advantages including enhanced stability and function, ease of synthesis, lower cost, and potential modulated delivery via engineered biomaterials. In this study, computational protein design was used to create a more efficient evolution of the native SDF protein. METHODS AND RESULTS: Protein structure modeling was used to engineer an SDF polypeptide analog (engineered SDF analog [ESA]) that splices the N-terminus (activation and binding) and C-terminus (extracellular stabilization) with a diproline segment designed to limit the conformational flexibility of the peptide backbone and retain the relative orientation of these segments observed in the native structure of SDF. Endothelial progenitor cells (EPCs) in ESA gradient, assayed by Boyden chamber, showed significantly increased migration compared with both SDF and control gradients. EPC receptor activation was evaluated by quantification of phosphorylated AKT, and cells treated with ESA yielded significantly greater phosphorylated AKT levels than SDF and control cells. Angiogenic growth factor assays revealed a distinct increase in angiopoietin-1 expression in the ESA- and SDF-treated hearts. In addition, CD-1 mice (n=30) underwent ligation of the left anterior descending coronary artery and peri-infarct intramyocardial injection of ESA, SDF-1α, or saline. At 2 weeks, echocardiography demonstrated a significant gain in ejection fraction, cardiac output, stroke volume, and fractional area change in mice treated with ESA compared with controls. CONCLUSIONS: Compared with native SDF, a novel engineered SDF polypeptide analog (ESA) more efficiently induces EPC migration and improves post-myocardial infarction cardiac function and thus offers a more clinically translatable neovasculogenic therapy.


Subject(s)
Angiogenic Proteins/chemistry , Angiogenic Proteins/pharmacology , Chemokine CXCL12/chemistry , Chemokine CXCL12/pharmacology , Computational Biology/methods , Neovascularization, Physiologic/drug effects , Protein Engineering/methods , Angiogenic Proteins/therapeutic use , Animals , Cardiac Output/drug effects , Cardiac Output/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cells, Cultured , Chemokine CXCL12/therapeutic use , Coronary Vessels/drug effects , Coronary Vessels/physiology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Male , Mice , Mice, Inbred Strains , Models, Animal , Myocardial Infarction/physiopathology , Myocardial Infarction/prevention & control , Neovascularization, Physiologic/physiology , Rats , Rats, Wistar , Stem Cells/cytology , Stem Cells/drug effects , Stroke Volume/drug effects , Stroke Volume/physiology
5.
J Appl Physiol (1985) ; 110(5): 1460-5, 2011 May.
Article in English | MEDLINE | ID: mdl-21292844

ABSTRACT

This study evaluates a therapy for infarct modulation and acute myocardial rescue and utilizes a novel technique to measure local myocardial oxygenation in vivo. Bone marrow-derived endothelial progenitor cells (EPCs) were targeted to the heart with peri-infarct intramyocardial injection of the potent EPC chemokine stromal cell-derived factor 1α (SDF). Myocardial oxygen pressure was assessed using a noninvasive, real-time optical technique for measuring oxygen pressures within microvasculature based on the oxygen-dependent quenching of the phosphorescence of Oxyphor G3. Myocardial infarction was induced in male Wistar rats (n = 15) through left anterior descending coronary artery ligation. At the time of infarction, animals were randomized into two groups: saline control (n = 8) and treatment with SDF (n = 7). After 48 h, the animals underwent repeat thoracotomy and 20 µl of the phosphor Oxyphor G3 was injected into three areas (peri-infarct myocardium, myocardial scar, and remote left hindlimb muscle). Measurements of the oxygen distribution within the tissue were then made in vivo by applying the end of a light guide to the beating heart. Compared with controls, animals in the SDF group exhibited a significantly decreased percentage of hypoxic (defined as oxygen pressure ≤ 15.0 Torr) peri-infarct myocardium (9.7 ± 6.7% vs. 21.8 ± 11.9%, P = 0.017). The peak oxygen pressures in the peri-infarct region of the animals in the SDF group were significantly higher than the saline controls (39.5 ± 36.7 vs. 9.2 ± 8.6 Torr, P = 0.02). This strategy for targeting EPCs to vulnerable peri-infarct myocardium via the potent chemokine SDF-1α significantly decreased the degree of hypoxia in peri-infarct myocardium as measured in vivo by phosphorescence quenching. This effect could potentially mitigate the vicious cycle of myocyte death, myocardial fibrosis, progressive ventricular dilatation, and eventual heart failure seen after acute myocardial infarction.


Subject(s)
Chemokine CXCL12/therapeutic use , Luminescent Measurements/methods , Metalloporphyrins/pharmacokinetics , Myocardial Infarction/drug therapy , Myocardial Infarction/metabolism , Myocardium/metabolism , Oxygen/metabolism , Angiogenesis Inducing Agents/therapeutic use , Animals , Male , Myocardial Infarction/pathology , Rats , Rats, Wistar , Tissue Distribution , Treatment Outcome
6.
J Thorac Cardiovasc Surg ; 140(5): 1174-80, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20951261

ABSTRACT

OBJECTIVES: Stromal cell-derived factor (SDF)-1α is a potent endogenous endothelial progenitor cell (EPC) chemokine and key angiogenic precursor. Recombinant SDF-1α has been demonstrated to improve neovasculogenesis and cardiac function after myocardial infarction (MI) but SDF-1α is a bulky protein with a short half-life. Small peptide analogs might provide translational advantages, including ease of synthesis, low manufacturing costs, and the potential to control delivery within tissues using engineered biomaterials. We hypothesized that a minimized peptide analog of SDF-1α, designed by splicing the N-terminus (activation and binding) and C-terminus (extracellular stabilization) with a truncated amino acid linker, would induce EPC migration and preserve ventricular function after MI. METHODS: EPC migration was first determined in vitro using a Boyden chamber assay. For in vivo analysis, male rats (n = 48) underwent left anterior descending coronary artery ligation. At infarction, the rats were randomized into 4 groups and received peri-infarct intramyocardial injections of saline, 3 µg/kg of SDF-1α, 3 µg/kg of spliced SDF analog, or 6 µg/kg spliced SDF analog. After 4 weeks, the rats underwent closed chest pressure volume conductance catheter analysis. RESULTS: EPCs showed significantly increased migration when placed in both a recombinant SDF-1α and spliced SDF analog gradient. The rats treated with spliced SDF analog at MI demonstrated a significant dose-dependent improvement in end-diastolic pressure, stroke volume, ejection fraction, cardiac output, and stroke work compared with the control rats. CONCLUSIONS: A spliced peptide analog of SDF-1α containing both the N- and C- termini of the native protein induced EPC migration, improved ventricular function after acute MI, and provided translational advantages compared with recombinant human SDF-1α.


Subject(s)
Cardiotonic Agents/pharmacology , Cell Movement/drug effects , Chemokine CXCL12/pharmacology , Endothelial Cells/drug effects , Myocardial Infarction/drug therapy , Peptides/pharmacology , Stem Cells/drug effects , Ventricular Function, Left/drug effects , Animals , Cardiac Output/drug effects , Cardiotonic Agents/administration & dosage , Cardiotonic Agents/chemical synthesis , Cells, Cultured , Chemokine CXCL12/administration & dosage , Chemokine CXCL12/chemistry , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Hemodynamics/drug effects , Humans , Injections, Intralesional , Male , Myocardial Infarction/physiopathology , Peptides/administration & dosage , Peptides/chemical synthesis , Protein Conformation , Protein Engineering , Protein Structure, Tertiary , Rats , Rats, Inbred Lew , Rats, Transgenic , Rats, Wistar , Recombinant Proteins/administration & dosage , Stem Cells/metabolism , Stroke Volume/drug effects , Time Factors
7.
Circulation ; 122(11 Suppl): S107-17, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20837901

ABSTRACT

BACKGROUND: Myocardial ischemia causes cardiomyocyte death, adverse ventricular remodeling, and ventricular dysfunction. Endothelial progenitor cells (EPCs) have been shown to ameliorate this process, particularly when activated with stromal cell-derived factor-1α (SDF), known to be the most potent EPC chemokine. We hypothesized that implantation of a tissue-engineered extracellular matrix (ECM) scaffold seeded with EPCs primed with SDF could induce borderzone neovasculogenesis, prevent adverse geometric remodeling, and preserve ventricular function after myocardial infarction. METHODS AND RESULTS: Lewis rats (n=82) underwent left anterior descending artery ligation to induce myocardial infarction. EPCs were isolated, characterized, and cultured on a vitronectin/collagen scaffold and primed with SDF to generate the activated EPC matrix (EPCM). EPCM was sutured to the anterolateral left ventricular wall, which included the region of ischemia. Control animals received sutures but no EPCM. Additional groups underwent application of the ECM alone, ECM primed with SDF (ECM+SDF), and ECM seeded with EPCs but not primed with SDF (ECM+SDF). At 4 weeks, borderzone myocardial tissue demonstrated increased levels of vascular endothelial growth factor in the EPCM group. When compared to controls, Vessel density as assessed by immunohistochemical microscopy was significantly increased in the EPCM group (4.1 versus 6.2 vessels/high-powered field; P<0.001), and microvascular perfusion measured by lectin microangiography was enhanced 4-fold (0.7% versus 2.7% vessel volume/section volume; P=0.04). Comparisons to additional groups also showed a significantly improved vasculogenic response in the EPCM group. Ventricular geometry and scar fraction assessed by digital planimetric analysis of sectioned hearts exhibited significantly preserved left ventricular internal diameter (9.7 mm versus 8.6 mm; P=0.005) and decreased infarct scar formation expressed as percent of total section area (16% versus 7%; P=0.002) when compared with all other groups. In addition, EPCM animals showed a significant preservation of function as measured by echocardiography, pressure-volume conductance, and Doppler flow. CONCLUSIONS: Extracellular matrix seeded with EPCs primed with SDF induces borderzone neovasculogenesis, attenuates adverse ventricular remodeling, and preserves ventricular function after myocardial infarction.


Subject(s)
Chemokine CXCL12/pharmacology , Endothelial Cells/cytology , Extracellular Matrix , Myocardial Infarction/therapy , Neovascularization, Physiologic/drug effects , Stem Cells/cytology , Tissue Engineering/methods , Animals , Cells, Cultured , Collagen , Disease Models, Animal , Humans , Male , Myocardial Infarction/pathology , Myocytes, Cardiac/pathology , Rats , Rats, Inbred Lew , Ventricular Dysfunction/pathology , Ventricular Dysfunction/therapy , Vitronectin
8.
Heart Lung Circ ; 19(11): 644-54, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20719564

ABSTRACT

PURPOSE: Post-myocardial infarction heart failure is a major health concern with limited therapy. Molecular revascularisation utilising granulocyte-macrophage colony stimulating factor (GMCSF) mediated endothelial progenitor cell (EPC) upregulation and stromal cell derived factor-1α (SDF) mediated myocardial EPC chemokinesis, may prevent myocardial loss and adverse remodelling. Vasculogenesis, viability, and haemodynamic improvements following therapy were investigated. PROCEDURES: Lewis rats (n=91) underwent LAD ligation and received either intramyocardial SDF and subcutaneous GMCSF or saline injections at the time of infarction. Molecular and haemodynamic assessments were performed at pre-determined time points following ligation. FINDINGS: SDF/GMCSF therapy upregulated EPC density as shown by flow cytometry (0.12±0.02% vs. 0.06±0.01% circulating lymphocytes, p=0.005), 48hours following infarction. A marked increase in perfusion was evident eight weeks after therapy, utilising confocal angiography (5.02±1.7×10(-2)µm(3)blood/µm(3)myocardial tissue vs. 2.03±0.710(-2)µm(3)blood/µm(3)myocardial tissue, p=0.00004). Planimetric analysis demonstrated preservation of wall thickness (0.98±0.09mm vs. 0.67±0.06mm, p=0.003) and ventricular diameter (7.81±0.99mm vs. 9.41±1.1mm, p=0.03). Improved haemodynamic function was evidenced by echocardiography and PV analysis (ejection fraction: 56.4±18.1% vs. 25.3±15.6%, p=0.001; pre-load adjusted maximal power: 6.6±2.6mW/µl(2) vs. 2.7±1.4mW/µl(2), p=0.01). CONCLUSION: Neovasculogenic therapy with GMCSF-mediated EPC upregulation and SDF-mediated EPC chemokinesis maybe an effective therapy for infarct modulation and preservation of myocardial function following acute myocardial infarction.


Subject(s)
Chemokine CXCL12/therapeutic use , Endothelial Cells/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Heart/drug effects , Hemodynamics/drug effects , Myocardial Infarction/drug therapy , Neovascularization, Physiologic/drug effects , Ventricular Function, Left/drug effects , Animals , Apoptosis , Chemokine CXCL12/pharmacology , Echocardiography , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Heart Ventricles/anatomy & histology , Hematopoietic Stem Cells/physiology , Male , Models, Animal , Myocardium/pathology , Rats , Rats, Inbred Lew , Up-Regulation
9.
J Thorac Cardiovasc Surg ; 140(3): 667-76, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20363480

ABSTRACT

OBJECTIVE: Microvascular malperfusion after myocardial infarction leads to infarct expansion, adverse remodeling, and functional impairment. Native reparative mechanisms exist but are inadequate to vascularize ischemic myocardium. We hypothesized that a 3-dimensional human fibroblast culture (3DFC) functions as a sustained source of angiogenic cytokines, thereby augmenting native angiogenesis and limiting adverse effects of myocardial ischemia. METHODS: Lewis rats underwent ligation of the left anterior descending coronary artery to induce heart failure; experimental animals received a 3DFC scaffold to the ischemic region. Border-zone tissue was analyzed for the presence of human fibroblast surface protein, vascular endothelial growth factor, and hepatocyte growth factor. Cardiac function was assessed with echocardiography and pressure-volume conductance. Hearts underwent immunohistochemical analysis of angiogenesis by co-localization of platelet endothelial cell adhesion molecule and alpha smooth muscle actin and by digital analysis of ventricular geometry. Microvascular angiography was performed with fluorescein-labeled lectin to assess perfusion. RESULTS: Immunoblotting confirmed the presence of human fibroblast surface protein in rats receiving 3DFC, indicating survival of transplanted cells. Increased expression of vascular endothelial growth factor and hepatocyte growth factor in experimental rats confirmed elution by the 3DFC. Microvasculature expressing platelet endothelial cell adhesion molecule/alpha smooth muscle actin was increased in infarct and border-zone regions of rats receiving 3DFC. Microvascular perfusion was also improved in infarct and border-zone regions in these rats. Rats receiving 3DFC had increased wall thickness, smaller infarct area, and smaller infarct fraction. Echocardiography and pressure-volume measurements showed that cardiac function was preserved in these rats. CONCLUSIONS: Application of a bioengineered 3DFC augments native angiogenesis through delivery of angiogenic cytokines to ischemic myocardium. This yields improved microvascular perfusion, limits infarct progression and adverse remodeling, and improves ventricular function.


Subject(s)
Angiogenic Proteins/metabolism , Coronary Circulation , Fibroblasts/transplantation , Myocardial Infarction/surgery , Neovascularization, Physiologic , Tissue Engineering/methods , Tissue Scaffolds , Ventricular Function, Left , Ventricular Remodeling , Animals , Cell Survival , Cells, Cultured , Disease Models, Animal , Echocardiography , Fibroblasts/metabolism , Hemodynamics , Hepatocyte Growth Factor/metabolism , Humans , Male , Microcirculation , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/pathology , Rats , Rats, Inbred Lew , Time Factors , Vascular Endothelial Growth Factor A/metabolism
10.
J Thorac Cardiovasc Surg ; 137(4): 971-7, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19327526

ABSTRACT

OBJECTIVE: It is generally accepted that patients who require biventricular assist device support have poorer outcomes than those requiring isolated left ventricular assist device support. However, it is unknown how the timing of biventricular assist device insertion affects outcomes. We hypothesized that planned biventricular assist device insertion improves survival compared with delayed conversion of left ventricular assist device support to biventricular assist device support. METHODS: We reviewed and compared outcomes of 266 patients undergoing left ventricular assist device or biventricular assist device placement at the University of Pennsylvania from April 1995 to June 2007. We subdivided patients receiving biventricular assist devices into planned biventricular assist device (P-BiVAD) and delayed biventricular assist device (D-BiVAD) groups based on the timing of right ventricular assist device insertion. We defined the D-BiVAD group as any failure of isolated left ventricular assist device support. RESULTS: Of 266 patients who received left ventricular assist devices, 99 (37%) required biventricular assist device support. We compared preoperative characteristics, successful bridging to transplantation, survival to hospital discharge, and Kaplan-Meier 1-year survival between the P-BiVAD (n = 71) and D-BiVAD (n = 28) groups. Preoperative comparison showed that patients who ultimately require biventricular support have similar preoperative status. Left ventricular assist device (n = 167) outcomes in all categories exceeded both P-BiVAD and D-BiVAD group outcomes. Furthermore, patients in the P-BiVAD group had superior survival to discharge than patients in the D-BiVAD group (51% vs 29%, P < .05). One-year and long-term Kaplan-Meier survival distribution confirmed this finding. There was also a trend toward improved bridging to transplantation in the P-BiVAD (n = 55) versus D-BiVAD (n = 22) groups (65% vs 45%, P = .10). CONCLUSION: When patients at high risk for failure of isolated left ventricular assist device support are identified, proceeding directly to biventricular assist device implantation is advised because early institution of biventricular support results in dramatic improvement in survival.


Subject(s)
Heart Failure/therapy , Heart-Assist Devices , Adult , Female , Heart Failure/complications , Humans , Male , Middle Aged , Retrospective Studies , Survival Analysis , Time Factors , Treatment Outcome
11.
J Heart Lung Transplant ; 27(12): 1286-92, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19059108

ABSTRACT

BACKGROUND: Right ventricular (RV) failure after left ventricular assist device (LVAD) placement is a serious complication and is difficult to predict. In the era of destination therapy and the total artificial heart, predicting post-LVAD RV failure requiring mechanical support is extremely important. METHODS: We reviewed patient characteristics, laboratory values and hemodynamic data from 266 patients who underwent LVAD placement at the University of Pennsylvania from April 1995 to June 2007. RESULTS: Of 266 LVAD recipients, 99 required RV assist device (BiVAD) placement (37%). We compared 36 parameters between LVAD (n = 167) and BiVAD patients (n = 99) to determine pre-operative risk factors for RV assist device (RVAD) need. By univariate analysis, 23 variables showed statistically significant differences between the two groups (p < or = 0.05). By multivariate logistic regression, cardiac index < or =2.2 liters/min/m(2) (odds ratio [OR] 5.7), RV stroke work index < or =0.25 mm Hg . liter/m(2) (OR 5.1), severe pre-operative RV dysfunction (OR 5.0), pre-operative creatinine > or =1.9 mg/dl (OR 4.8), previous cardiac surgery (OR 4.5) and systolic blood pressure < or =96 mm Hg (OR 2.9) were the best predictors of RVAD need. CONCLUSIONS: The most significant predictors for RVAD need were cardiac index, RV stroke work index, severe pre-operative RV dysfunction, creatinine, previous cardiac surgery and systolic blood pressure. Using these data, we constructed an algorithm that can predict which LVAD patients will require RVAD with >80% sensitivity and specificity.


Subject(s)
Heart Failure/physiopathology , Heart Transplantation , Heart-Assist Devices , Ventricular Dysfunction, Left/surgery , Ventricular Dysfunction, Right/surgery , Adult , Aged , Blood Pressure , Diabetes Complications/epidemiology , Equipment Design , Female , Heart Failure/surgery , Heart-Assist Devices/adverse effects , Hemodynamics , Humans , Male , Middle Aged , Predictive Value of Tests , Pulmonary Disease, Chronic Obstructive/epidemiology , Retrospective Studies , Risk Assessment , Risk Factors , Ventricular Dysfunction, Left/epidemiology , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Right/physiopathology , Ventricular Function, Right
12.
J Thorac Cardiovasc Surg ; 135(2): 283-91, 291.e1; discussion 291, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18242252

ABSTRACT

OBJECTIVE: A significant number of patients have coronary artery disease that is not amenable to traditional revascularization. Prospective, randomized clinical trials have demonstrated therapeutic benefits with transmyocardial laser revascularization in this cohort. The molecular mechanisms underlying this therapy, however, are poorly understood. The focus of this study was evaluation of the proposed vasculogenic mechanisms involved in transmyocardial laser revascularization. METHODS: Male Yorkshire pigs (30-35 kg, n = 25) underwent left thoracotomy and placement of ameroid constrictors around the proximal left circumflex coronary artery. During the next 4 weeks, a well-defined region of myocardial ischemia developed, and the animals underwent a redo left thoracotomy. The animals were randomly assigned to sham treatment (thoracotomy only, control, n = 11) or transmyocardial laser revascularization of hibernating myocardium with a holmium:yttrium-aluminum-garnet laser (n = 14). After an additional 4 weeks, the animals underwent median sternotomy, echocardiographic analysis of wall motion, and hemodynamic analysis with an ascending aortic flow probe and pulmonary artery catheter. The hearts were explanted for molecular analysis. RESULTS: Molecular analysis demonstrated statistically significant increases in the proangiogenic proteins nuclear factor kappaB (42 +/- 27 intensity units vs 591 +/- 383 intensity units, P = .03) and angiopoietin 1 (0 +/- 0 intensity units vs 241 +/- 87 intensity units, P = .003) relative to sham control values with transmyocardial laser revascularization within the ischemic myocardium. There were also increases in vasculogenesis (18.8 +/- 8.7 vessels/high-power field vs 31.4 +/- 10.2 vessels/high-power field, P = .02), and perfusion (0.028 +/- 0.009 microm3 blood/microm3 tissue vs 0.044 +/- 0.004 microm3 blood/microm3 tissue, P = .01). Enhanced myocardial viability was demonstrated by increased myofilament density (40.7 +/- 8.5 cardiomyocytes/high-power field vs 50.8 +/- 7.5 cardiomyocytes/high-power field, P = .03). Regional myocardial function within the treated territory demonstrated augmented contractility. Global hemodynamic function was significantly improved relative to the control group with transmyocardial laser revascularization (cardiac output 2.1 +/- 0.2 L/min vs 2.7 +/- 0.2 L/min, P = .007, mixed venous oxygen saturation 64.7% +/- 3.6% vs 76.1% +/- 3.4%, P = .008). CONCLUSION: Transmyocardial laser revascularization with the holmium-YAG laser enhances perfusion, with resultant improvement in myocardial contractility.


Subject(s)
Coronary Circulation/physiology , Laser Therapy , Myocardial Ischemia/surgery , Myocardial Revascularization/methods , Animals , Chemokines/metabolism , Disease Models, Animal , Flow Cytometry , Hemodynamics/physiology , Lasers, Solid-State , Male , Myocardial Contraction/physiology , Neovascularization, Physiologic , Probability , Random Allocation , Reference Values , Sensitivity and Specificity , Swine , Up-Regulation , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...