Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Transfus Med Hemother ; 50(5): 456-468, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37899992

ABSTRACT

Introduction: Blood product transfusion retains a critical role in the supportive care of patients with acute myeloid leukemia (AML). Whereas previous studies have shown increased transfusion dependency to portend inferior outcome, predictive factors of an increased transfusion burden and the prognostic impact of transfusion support have not been assessed recently. Methods/Patients: We performed a retrospective analysis on a recent cohort of patients given intensive induction chemotherapy in 2014-2022. Results: The analysis comprised 180 patients with a median age of 57 years with 80% designated as de novo AML. Fifty-four patients (31%) were FLT3-ITD mutated, and 73 patients (42%) harbored NPM1. Favorable risk and intermediate risk ELN 2017 patients accounted for 43% and 34% of patients, respectively. The median number of red blood cell (RBC) and platelet units given during induction were 9 and 7 units, respectively. Seventeen patients (9%) received cryoprecipitate, and fresh frozen plasma (FFP) was given to 12 patients (7%). Lower initial hemoglobin and platelet levels were predictive of increased use of RBC (p < 0.0001) and platelet transfusions (p < 0.0001). FFP was significantly associated with induction related mortality (42% vs. 5%; p < 0.0001) and with FLT3-ITD (72% vs. 28%; p = 0.004). Blood group AB experienced improved mean overall survival compared to blood group O patients (4.1 years vs. 2.8 years; p = 0.025). In multivariate analysis, increased number of FFP (hazard ratio [HR], 4.23; 95% confidence interval [CI], 2.1-8.6; p < 0.001) and RBC units (HR, 1.8; 95% CI, 1.2-2.8; p = 0.008) given was associated with inferior survival. Conclusion: Transfusion needs during induction crucially impact the clinical trajectory of AML patients.

2.
Transfus Apher Sci ; 62(5): 103793, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37661488
3.
Cell Rep ; 37(11): 110114, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34883043

ABSTRACT

Messenger RNA-based vaccines against COVID-19 induce a robust anti-SARS-CoV-2 antibody response with potent viral neutralization activity. Antibody effector functions are determined by their constant region subclasses and by their glycosylation patterns, but their role in vaccine efficacy is unclear. Moreover, whether vaccination induces antibodies similar to those in patients with COVID-19 remains unknown. We analyze BNT162b2 vaccine-induced IgG subclass distribution and Fc glycosylation patterns and their potential to drive effector function via Fcγ receptors and complement pathways. We identify unique and dynamic pro-inflammatory Fc compositions that are distinct from those in patients with COVID-19 and convalescents. Vaccine-induced anti-Spike IgG is characterized by distinct Fab- and Fc-mediated functions between different age groups and in comparison to antibodies generated during natural viral infection. These data highlight the heterogeneity of Fc responses to SARS-CoV-2 infection and vaccination and suggest that they support long-lasting protection differently.


Subject(s)
COVID-19/immunology , Glycosylation/drug effects , SARS-CoV-2/immunology , Adult , Aged , Antibodies, Viral/immunology , BNT162 Vaccine/immunology , COVID-19 Vaccines/metabolism , Female , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Israel/epidemiology , Male , Middle Aged , Spike Glycoprotein, Coronavirus/immunology , Vaccination/methods , Vaccine Efficacy , Vaccines, Synthetic/immunology , Vaccines, Synthetic/metabolism , mRNA Vaccines/immunology , mRNA Vaccines/metabolism
4.
Isr Med Assoc J ; 23(10): 639-645, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34672446

ABSTRACT

BACKGROUND: Extra peritoneal packing (EPP) is a quick and highly effective method to control pelvic hemorrhage. OBJECTIVES: To determine whether EPP can be as safely and efficiently performed in the emergency department (ED) as in the operating room (OR). METHODS: Retrospective study of 29 patients who underwent EPP in the ED or OR in two trauma centers in Israel 2008-2018. RESULTS: Our study included 29 patients, 13 in the ED-EPP group and 16 in the OR-EPP group. The mean injury severity score (ISS) was 34.9 ± 11.8. Following EPP, hemodynamic stability was successfully achieved in 25 of 29 patients (86.2%). A raise in the mean arterial pressure (MAP) with a median of 25 mmHg (mean 30.0 ± 27.5, P < 0.001) was documented. All patients who did not achieve hemodynamic stability after EPP had multiple sources of bleeding or fatal head injury and eventually succumbed. Patients who underwent EPP in the ED showed higher change in MAP (P = 0.0458). The overall mortality rate was 27.5% (8/29) with no difference between the OR and ED-EPP. No differences were found between ED and OR-EPP in the amount of transfused blood products, surgical site infections, and length of stay in the hospital. However, patients who underwent ED-EPP were more prone to develop deep vein thrombosis (DVT): 50% (5/10) vs. 9% (1/11) in ED and OR-EPP groups respectively (P = 0.038). CONCLUSIONS: EPP is equally effective when performed in the ED or OR with similar surgical site infection rates but higher incidence of DVT.


Subject(s)
Exsanguination , Fractures, Bone , Hemostasis, Surgical , Pelvis , Postoperative Complications , Surgical Wound Infection , Venous Thrombosis , Blood Pressure Determination/methods , Emergency Service, Hospital/statistics & numerical data , Exsanguination/diagnosis , Exsanguination/etiology , Exsanguination/mortality , Exsanguination/surgery , Female , Fractures, Bone/complications , Fractures, Bone/diagnosis , Fractures, Bone/surgery , Hemostasis, Surgical/adverse effects , Hemostasis, Surgical/instrumentation , Hemostasis, Surgical/methods , Humans , Injury Severity Score , Israel/epidemiology , Male , Middle Aged , Outcome and Process Assessment, Health Care , Pelvis/diagnostic imaging , Pelvis/injuries , Postoperative Complications/diagnosis , Postoperative Complications/epidemiology , Retrospective Studies , Surgical Wound Infection/diagnosis , Surgical Wound Infection/etiology , Trauma Centers/statistics & numerical data , Venous Thrombosis/diagnosis , Venous Thrombosis/etiology
5.
J Am Coll Cardiol ; 60(11): 990-1000, 2012 Sep 11.
Article in English | MEDLINE | ID: mdl-22749309

ABSTRACT

OBJECTIVES: The goal of this study was to establish a patient-specific human-induced pluripotent stem cells (hiPSCs) model of catecholaminergic polymorphic ventricular tachycardia (CPVT). BACKGROUND: CPVT is a familial arrhythmogenic syndrome characterized by abnormal calcium (Ca(2+)) handling, ventricular arrhythmias, and sudden cardiac death. METHODS: Dermal fibroblasts were obtained from a CPVT patient due to the M4109R heterozygous point RYR2 mutation and reprogrammed to generate the CPVT-hiPSCs. The patient-specific hiPSCs were coaxed to differentiate into the cardiac lineage and compared with healthy control hiPSCs-derived cardiomyocytes (hiPSCs-CMs). RESULTS: Intracellular electrophysiological recordings demonstrated the development of delayed afterdepolarizations in 69% of the CPVT-hiPSCs-CMs compared with 11% in healthy control cardiomyocytes. Adrenergic stimulation by isoproterenol (1 µM) or forskolin (5 µM) increased the frequency and magnitude of afterdepolarizations and also led to development of triggered activity in the CPVT-hiPSCs-CMs. In contrast, flecainide (10 µM) and thapsigargin (10 µM) eliminated all afterdepolarizations in these cells. The latter finding suggests an important role for internal Ca(2+) stores in the pathogenesis of delayed afterdepolarizations. Laser-confocal Ca(2+) imaging revealed significant whole-cell [Ca(2+)] transient irregularities (frequent local and large-storage Ca(2+)-release events, broad and double-humped transients, and triggered activity) in the CPVT cardiomyocytes that worsened with adrenergic stimulation and Ca(2+) overload and improved with beta-blockers. Store-overload-induced Ca(2+) release was also identified in the hiPSCs-CMs and the threshold for such events was significantly reduced in the CPVT cells. CONCLUSIONS: This study highlights the potential of hiPSCs for studying inherited arrhythmogenic syndromes, in general, and CPVT specifically. As such, it represents a promising paradigm to study disease mechanisms, optimize patient care, and aid in the development of new therapies.


Subject(s)
Induced Pluripotent Stem Cells , Myocytes, Cardiac/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Tachycardia, Ventricular/physiopathology , Arrhythmias, Cardiac/genetics , Calcium/metabolism , Electrophysiologic Techniques, Cardiac , Gene Expression , Humans , Models, Cardiovascular , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism
6.
Biochem J ; 438(2): 337-47, 2011 Sep 01.
Article in English | MEDLINE | ID: mdl-21635224

ABSTRACT

The nuclear protein PARP-1 [poly(ADP-ribose) polymerase-1] is activated in cardiomyocytes exposed to hypoxia causing DNA breaks. Unlike this stress-induced PARP-1 activation, our results provide evidence for Ca(2+)-induced PARP-1 activation in contracting newborn cardiomyocytes treated with growth factors and hormones that increased their contraction rate, induced intracellular Ca(2+) mobilization and its rhythmical and transient translocation into the nucleus. Furthermore, activated PARP-1 up-regulated the activity of phosphorylated ERK (extracellular-signal-regulated kinase) in the nucleus, promoting expression of the Elk1 target gene c-fos. Up-regulation of the transcription factor c-Fos/GATA-4 promoted ANF (atrial natriuretic factor) expression. Given that expression of ANF is known to be implicated in morphological changes, growth and development of cardiomyocytes, these results outline a PARP-1-dependent signal transduction mechanism that links contraction rate and Ca(2+) mobilization with the expression of genes underlying morphological changes in cardiomyocytes.


Subject(s)
Atrial Natriuretic Factor/metabolism , Calcium/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/enzymology , Poly(ADP-ribose) Polymerases/metabolism , Angiotensin II/pharmacology , Animals , Animals, Newborn , Atrial Natriuretic Factor/genetics , Calcium/metabolism , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , GATA4 Transcription Factor/metabolism , Gene Expression Regulation/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Myocardial Contraction/drug effects , Phosphorylation/drug effects , Poly (ADP-Ribose) Polymerase-1 , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Proto-Oncogene Proteins c-fos/genetics , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Sprague-Dawley , Ribose/metabolism
7.
Cell Transplant ; 18(3): 275-82, 2009.
Article in English | MEDLINE | ID: mdl-19558776

ABSTRACT

Tissue engineering holds the promise of providing new solutions for heart transplant shortages and pediatric heart transplantation. The aim of this study was to evaluate the ability of a peritoneal-generated, tissue-engineered cardiac patch to replace damaged myocardium in a heterotopic heart transplant model. Fetal cardiac cells (1 x 10(6)/scaffold) from syngeneic Lewis rats were seeded into highly porous alginate scaffolds. The cell constructs were cultured in vitro for 4 days and then they were implanted into the rat peritoneal cavity for 1 week. During this time the peritoneal-implanted patches were vascularized and populated with myofibroblasts. They were harvested and their performance in an infrarenal heterotopic abdominal heart transplantation model was examined (n = 15). After transplantation and before reperfusion of the donor heart, a 5-mm left (n = 6) or right (n = 9) ventriculotomy was performed and the patch was sutured onto the donor heart to repair the defect. Echocardiographical studies carried out 1-2 weeks after transplantation showed normal LV function in seven of the eight hearts studied. After 1 month, visual examination of the grafted patch revealed no aneurysmal dilatation. Microscopic examination revealed, in most of the cardiac patches, a complete disappearance of the scaffold and its replacement by a consistent tissue composed of myofibroblasts embedded in collagen bundles. The cardiac patch was enriched with a relatively large number of infiltrating blood vessels. In conclusion, cardiac patches generated in the peritoneum were developed into consistent tissue patches with properties to seal and correct myocardial defects. Our study also offers a viable rat model for screening and evaluating new concepts in cardiac reconstruction and engineering.


Subject(s)
Heart Transplantation/methods , Models, Animal , Myocytes, Cardiac/cytology , Peritoneum/cytology , Transplantation, Heterotopic/methods , Animals , Biomarkers/metabolism , Echocardiography , Myocytes, Cardiac/ultrastructure , Peritoneum/blood supply , Rats , Rats, Inbred Lew , Tissue Engineering
8.
Circulation ; 117(11): 1388-96, 2008 Mar 18.
Article in English | MEDLINE | ID: mdl-18316487

ABSTRACT

BACKGROUND: Adverse cardiac remodeling and progression of heart failure after myocardial infarction are associated with excessive and continuous damage to the extracellular matrix. We hypothesized that injection of in situ-forming alginate hydrogel into recent and old infarcts would provide a temporary scaffold and attenuate adverse cardiac remodeling and dysfunction. METHODS AND RESULTS: We developed a novel absorbable biomaterial composed of calcium-crosslinked alginate solution, which displays low viscosity and, after injection into the infarct, undergoes phase transition into hydrogel. To determine the outcome of the biomaterial after injection, calcium-crosslinked biotin-labeled alginate was injected into the infarct 7 days after anterior myocardial infarction in rat. Serial histology studies showed in situ formation of alginate hydrogel implant, which occupied up to 50% of the scar area. The biomaterial was replaced by connective tissue within 6 weeks. Serial echocardiography studies before and 60 days after injection showed that injection of alginate biomaterial into recent (7 days) infarct increased scar thickness and attenuated left ventricular systolic and diastolic dilatation and dysfunction. These beneficial effects were comparable and sometimes superior to those achieved by neonatal cardiomyocyte transplantation. Moreover, injection of alginate biomaterial into old myocardial infarction (60 days) increased scar thickness and improved systolic and diastolic dysfunction. CONCLUSIONS: We show for the first time that injection of in situ-forming, bioabsorbable alginate hydrogel is an effective acellular strategy that prevents adverse cardiac remodeling and dysfunction in recent and old myocardial infarctions in rat.


Subject(s)
Alginates/therapeutic use , Biocompatible Materials/therapeutic use , Hydrogels/therapeutic use , Hypertrophy, Left Ventricular/prevention & control , Myocardial Infarction/drug therapy , Ventricular Dysfunction, Left/prevention & control , Ventricular Remodeling/drug effects , Alginates/administration & dosage , Alginates/chemistry , Alginates/pharmacology , Animals , Biocompatible Materials/administration & dosage , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Cicatrix/diagnostic imaging , Cicatrix/pathology , Drug Administration Schedule , Drug Evaluation, Preclinical , Glucuronic Acid/administration & dosage , Glucuronic Acid/chemistry , Glucuronic Acid/pharmacology , Glucuronic Acid/therapeutic use , Hexuronic Acids/administration & dosage , Hexuronic Acids/chemistry , Hexuronic Acids/pharmacology , Hexuronic Acids/therapeutic use , Hydrogels/administration & dosage , Hydrogels/pharmacology , Hypertrophy, Left Ventricular/diagnostic imaging , Hypertrophy, Left Ventricular/etiology , Injections, Intralesional , Male , Materials Testing , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Myocytes, Cardiac/transplantation , Random Allocation , Rats , Rats, Sprague-Dawley , Ultrasonography , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Dysfunction, Left/etiology , Viscosity
9.
Circulation ; 116(11 Suppl): I38-45, 2007 Sep 11.
Article in English | MEDLINE | ID: mdl-17846324

ABSTRACT

BACKGROUND: Cell labeling with superparamagnetic iron oxide (SPIO) nanoparticles enables noninvasive MRI and tracking of transplanted stem cells. We sought to determine whether mesenchymal stem cell (MSC) outcome is affected by SPIO labeling in a rat model of myocardial infarction. METHODS AND RESULTS: Rat MSCs were labeled with SPIO (ferumoxides; Endorem; Guerbet, Villepinte, France). By trypan-blue exclusion assay, almost 100% of the cells remained viable after labeling. Seven days after MI, rats were randomized to injections of 2x10(6) SPIO-labeled MSCs, 2x10(6) unlabeled MSCs, or saline. Labeled cells were visualized in the infarcted myocardium as large black spots by serial MRI studies throughout the 4-week follow-up. The presence of labeled cells was confirmed by iron staining and real-time polymerase chain reaction on postmortem specimens. At 4 weeks after transplantation, the site of cell injection was infiltrated by inflammatory cells. Costaining for iron and ED1 (resident macrophage marker) showed that the iron-positive cells were cardiac macrophages. By real-time polymerase chain reaction, the Y-chromosome-specific SRY DNA of MSCs from male donors was not detected in infarcted hearts of female recipients. Serial echocardiography studies at baseline and 4 weeks after cell transplantation showed that both unlabeled and labeled MSCs attenuated progressive left ventricular dilatation and dysfunction compared with controls. CONCLUSIONS: At 4 weeks after transplantation of SPIO-labeled MSCs, the transplanted cells are not present in the scar and the enhanced MRI signals arise from cardiac macrophages that engulfed the SPIO nanoparticles. However, both labeled and unlabeled cells attenuate left ventricular dilatation and dysfunction after myocardial infarction.


Subject(s)
Ferric Compounds , Mesenchymal Stem Cell Transplantation/methods , Myocardial Infarction/surgery , Staining and Labeling/methods , Animals , Echocardiography/methods , Ferric Compounds/analysis , Magnetics , Mesenchymal Stem Cells/chemistry , Myocardial Infarction/diagnostic imaging , Myocardium/chemistry , Myocardium/pathology , Nanoparticles/analysis , Radiography , Rats , Rats, Sprague-Dawley , Treatment Outcome , Ventricular Remodeling/physiology
10.
Heart ; 93(10): 1278-84, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17566061

ABSTRACT

OBJECTIVE: To test the hypothesis that human embryonic stem cells (hESCs) can be guided to form new myocardium by transplantation into the normal or infarcted heart, and to assess the influence of hESC-derived cardiomyocytes (hESCMs) on cardiac function in a rat model of myocardial infarction (MI). METHODS: Undifferentiated hESCs (0.5-1x10(6)), human embryoid bodies (hEBs) (4-8 days; 0.5-1x10(6)), 0.1 mm pieces of embryonic stem-derived beating myocardial tissue, and phosphate-buffered saline (control) were injected into the normal or infarcted myocardium of athymic nude rats (n = 58) by direct injection into the muscle or into preimplanted three-dimensional alginate scaffold. By 2-4 weeks after transplantation, heart sections were examined to detect the human cells and differentiation with fluorescent in situ hybridisation, using DNA probes specific for human sex chromosomes and HLA-DR or HLA-ABC immunostaining. RESULTS: Microscopic examination showed transplanted human cells in the normal, and to a lesser extent in the infarcted myocardium (7/7 vs 2/6; p<0.05). The transplanted hESCs and hEBs rarely created new vessels and did not form new myocardium. Transplantation of hESCM tissue into normal heart produced islands of disorganised myofibres, fibrosis and, in a single case, a teratoma. However, transplantation of hESCMs into the infarcted myocardium did prevent post-MI dysfunction and scar thinning. CONCLUSIONS: Undifferentiated hESCs and hEBs are not directed to form new myocardium after transplantation into normal or infarcted heart and may create teratoma. Nevertheless, this study shows that hESC-derived cardiomyocyte transplantation can attenuate post-MI scar thinning and left ventricular dysfunction.


Subject(s)
Embryonic Stem Cells/transplantation , Myocardial Infarction/therapy , Myocytes, Cardiac/transplantation , Stem Cell Transplantation/methods , Animals , Cell Line , Echocardiography , Female , Humans , Immunohistochemistry , In Situ Hybridization, Fluorescence , Myocardial Contraction , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Rats , Rats, Nude , Ventricular Remodeling
11.
Stem Cells ; 24(3): 772-80, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16195418

ABSTRACT

The use of adult stem cells for myocardial tissue repair might be limited in elderly and sick people because their cells are depleted and exhausted. The present study was conducted to explore the potential of human umbilical cord blood (UCB) CD133+ progenitor cells for myocardial tissue repair in a model of extensive myocardial infarction (MI). CD133+ progenitor cells were isolated from newborn UCB. Cells (1.2-2 x 10(6)) or saline (control) was infused intravenously 7 days after permanent coronary artery ligation in athymic nude rats. Left ventricular (LV) function was assessed before and 1 month after infusion by echocardiography. Tracking of human cells was performed by fluorescent in situ hybridization for human X and Y chromosomes or by immunostaining for HLA-DR or HLA-ABC. One month after delivery, LV fractional shortening improved by 42 +/- 17% in cell-treated hearts and decreased by 39 +/- 10% in controls (p = .001). Anterior wall thickness decreased significantly in controls but not in treated hearts. Microscopic examination revealed that the UCB cells were able to migrate, colonize, and survive in the infarcted myocardium. Human cells were identified near vessel walls and LV cavity and were occasionally incorporated into endothelial cells in six of nine cell-treated animals but not in controls. Scar tissue from cell-treated animals was significantly populated with autologous myofibroblasts as indicated by colocalization of HLA-DR and alpha-smooth muscle actin staining. In conclusion, the present work suggests that, after MI, intravenous delivery of human UCB-derived CD133+ cells can produce functional recovery by preventing scar thinning and LV systolic dilatation.


Subject(s)
Antigens, CD , Fetal Blood/cytology , Glycoproteins , Myocardial Infarction/therapy , Peptides , Recovery of Function , Stem Cell Transplantation , Stem Cells/cytology , AC133 Antigen , Animals , Humans , Myocardial Infarction/pathology , Rats , Rats, Nude
12.
Technol Cancer Res Treat ; 4(6): 699-705, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16292891

ABSTRACT

In this study we perform in vitro irreversible electroporation (IRE) experiments with human hepatocarcinoma cells (HepG2) to investigate IRE as a new technique for undesirable tissue ablation. Irreversible electroporation (IRE) is the irreversible permeabilization of the cell membrane through the application of microsecond through millisecond electrical pulses. Until now IRE was studied only as an undesirable condition during the use of reversible electroporation in gene therapy and electrochemotherapy. There was a possibility that the IRE ablation domain is mostly superimposed on the electrical pulses induced Joule heating thermal ablation domain. This study demonstrates that there is a real and substantial domain of electrical parameters for IRE ablation of cancer that is distinct from the thermal domain and which results in complete cancer cell ablation. Experiments show that the application of 1500 V/cm in three sets of ten pulses of 300 microseconds each can produce complete cancer cell ablation. We also find that the use of multiple pulses appears to be more effective for cancer cell ablation than the application of the same energy in one single pulse.


Subject(s)
Carcinoma, Hepatocellular/therapy , Cell Membrane Permeability , Electric Stimulation Therapy , Electroporation , Liver Neoplasms/therapy , Carcinoma, Hepatocellular/pathology , Cell Survival , Humans , Liver Neoplasms/pathology
13.
Methods Mol Med ; 112: 205-21, 2005.
Article in English | MEDLINE | ID: mdl-16010019

ABSTRACT

We review an experimental protocol for investigating concepts and methods for myocardial repair using fetal cardiomyocyte transplantation. We describe methods of cell isolation, culture, labeling, and assessment of the influence of the engrafted cells on left ventricular remodeling and function.


Subject(s)
Cell Transplantation , Fetal Heart/cytology , Fetal Tissue Transplantation , Myocardial Infarction/surgery , Myocytes, Cardiac/cytology , Animals , Cells, Cultured , Myocytes, Cardiac/metabolism , Rats , Rats, Inbred Strains
14.
Ann Thorac Surg ; 77(5): 1648-55, 2004 May.
Article in English | MEDLINE | ID: mdl-15111159

ABSTRACT

BACKGROUND: Arctic fish survive subzero temperatures by producing a family of antifreeze proteins (AFPs) that noncolligatively lower the freezing temperature of their body fluids. We report 24-hour storage of mammalian hearts for transplantation at subzero temperatures using AFPs derived from arctic fish. METHODS: Forty-two heterotopic transplantations were performed in isoimmune Sprague-Dawley rats. Harvested hearts were retrogradely infused with cold 4 degrees C University of Wisconsin (UW) solution and were preserved in a specialized cooling bath at two target temperatures, 4 degrees C and -1.3 degrees C for 12,18, and 24 hours (6 experiments/group). Preservation solutions were UW alone for the 4 degrees C group, and UW with 15 mg/mL AFP III for the -1.3 degrees C group. After hypothermic storage the hearts were heterotopically transplanted into isoimmune rats. Viability was assessed and graded on a scale of 0 to 6 (0 = no contractions to 6 = excellent contractions). Transplanted hearts were then fixed in vivo and were subject to electron microscopy and histopathologic examination. RESULTS: None of the hearts preserved at -1.3 degrees C in UW/AFP III solution froze. All control hearts preserved at -1.3 degrees C without AFP protection froze and died at reperfusion. Viability of hearts preserved at -1.3 degrees C in UW/AFP III solution was significantly better after 18 hours of preservation, 30 and 60 minutes after reperfusion (median, 5 versus 3 and 6 versus 3, respectively; p < 0.05) and after 24 hours of preservation 30 and 60 minutes after reperfusion (median, 4.5 versus 1.5 and 5 versus 2, respectively; p < 0.05). Histologic and electron microscopy studies demonstrated better myocyte structure and mitochondrial integrity preservation with UW/AFP III solution. CONCLUSIONS: Antifreeze proteins prevent freezing in subzero cryopreservation of mammalian hearts for transplantation. Subzero preservation prolongs ischemic times and improves posttransplant viability.


Subject(s)
Antifreeze Proteins, Type III/therapeutic use , Heart Transplantation , Organ Preservation Solutions , Organ Preservation/methods , Transplantation, Heterotopic , Adenosine/therapeutic use , Allopurinol/therapeutic use , Animals , Cardioplegic Solutions/therapeutic use , Glutathione/therapeutic use , Insulin/therapeutic use , Raffinose/therapeutic use , Rats , Rats, Sprague-Dawley
15.
Circulation ; 108(7): 863-8, 2003 Aug 19.
Article in English | MEDLINE | ID: mdl-12900340

ABSTRACT

BACKGROUND: Systemic delivery of bone marrow-derived mesenchymal stem cells (BM-MSCs) is an attractive approach for myocardial repair. We aimed to test this strategy in a rat model after myocardial infarction (MI). METHODS AND RESULTS: BM-MSCs were obtained from rat bone marrow, expanded in vitro to a purity of >50%, and labeled with 99mTc exametazime, fluorescent dye, LacZ marker gene, or bromodeoxyuridine. Rats were subjected to MI by transient coronary artery occlusion or to sham MI. 99mTc-labeled cells (4x10(6)) were transfused into the left ventricular cavity of MI rats either at 2 or 10 to 14 days after MI and were compared with sham-MI rats or MI rats treated with intravenous infusion. Gamma camera imaging and isolated organ counting 4 hours after intravenous infusion revealed uptake of the 99mTc-labeled cells mainly in the lungs, with significantly smaller amounts in the liver, heart, and spleen. Delivery by left ventricular cavity infusion resulted in drastically lower lung uptake, better uptake in the heart, and specifically higher uptake in infarcted compared with sham-MI hearts. Histological examination at 1 week after infusion identified labeled cells either in the infarcted or border zone but not in remote viable myocardium or sham-MI hearts. Labeled cells were also identified in the lung, liver, spleen, and bone marrow. CONCLUSIONS: Systemic intravenous delivery of BM-MSCs to rats after MI, although feasible, is limited by entrapment of the donor cells in the lungs. Direct left ventricular cavity infusion enhances migration and colonization of the cells preferentially to the ischemic myocardium.


Subject(s)
Bone Marrow Cells , Cell Movement , Mesoderm/transplantation , Myocardial Infarction/therapy , Stem Cell Transplantation , Animals , Bone Marrow Cells/cytology , Cardiac Catheterization , Cell Separation , Cells, Cultured , Disease Models, Animal , Feasibility Studies , Female , Gamma Cameras , Heart/diagnostic imaging , Heart/physiopathology , Lung/blood supply , Lung/cytology , Mesoderm/cytology , Myocardial Infarction/diagnostic imaging , Organ Specificity , Radionuclide Imaging , Rats , Rats, Sprague-Dawley , Technetium , Treatment Outcome , Ventricular Function
16.
Circulation ; 106(12 Suppl 1): I125-30, 2002 Sep 24.
Article in English | MEDLINE | ID: mdl-12354721

ABSTRACT

BACKGROUND: The muscle-specific MyoD family of transcription factors function as master genes that are able to prompt myogenesis in a variety of cells. The purpose of our study was to determine whether MyoD could induce primary cardiac fibroblasts, isolated from infarcted myocardium or pericardium, to undergo myogenic conversion in a clinically relevant approach. METHODS AND RESULTS: Primary rat fibroblasts from 7-day-old infarcted myocardium or normal pericardium were transfected by an E1/E3-deleted adenoviral vector carrying both a human MyoD cDNA driven by a CMV promoter and a green fluorescent protein (GFP) reporter gene driven by a second CMV promoter. Expression of MyoD caused myogenic differentiation of cultured fibroblasts, as defined by elongation and fusion into multinucleated myotubes, typical cross striation as identified by electron microscopy, and positive immunostaining for sarcomeric actin, fast myosin heavy chain (MHC), and actinin. The myogenic cells (1.5x10(6)) were transplanted into the infarcted myocardium 7 days after coronary artery occlusion. By 1 month after transplantation, the converted fibroblasts gave rise to a cluster of myogenic cells that in a few hearts occupied a large part of the scar with positive immunostaining for the myogenic proteins fast-MHC and sarcomeric actin. A few cells expressed the gap junction protein connexin 43 in a disorganized manner. There was no positive staining in the control hearts treated with injections of untreated fibroblasts or culture medium. CONCLUSIONS: Our work shows that it is possible to exploit the unique capacity of MyoD to activate myogenesis in fibroblasts ex vivo and to create a vast source of autologous myogenic cells for transplantation.


Subject(s)
Adenoviridae/genetics , Fibroblasts/transplantation , MyoD Protein/genetics , Myocardium/cytology , Animals , Cardiomyoplasty , Cell Differentiation , Fibroblasts/cytology , Genetic Vectors , Heart/physiology , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Myocardium/ultrastructure , Rats , Rats, Sprague-Dawley , Wound Healing
SELECTION OF CITATIONS
SEARCH DETAIL
...