Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Circ Res ; 124(2): 243-255, 2019 01 18.
Article in English | MEDLINE | ID: mdl-30582450

ABSTRACT

RATIONALE: Endothelial colony forming cells (ECFCs) or late blood outgrowth endothelial cells can be isolated from human cord or peripheral blood, display properties of endothelial progenitors, home into ischemic tissues and support neovascularization in ischemic disease models. OBJECTIVE: To assess the functions of CYTL1 (cytokine-like 1), a factor we found preferentially produced by ECFCs, in regard of vessel formation. METHODS AND RESULTS: We show by transcriptomic analysis that ECFCs are distinguished from endothelial cells of the vessel wall by production of high amounts of CYTL1. Modulation of expression demonstrates that the factor confers increased angiogenic sprouting capabilities to ECFCs and can also trigger sprouting of mature endothelial cells. The data further display that CYTL1 can be induced by hypoxia and that it functions largely independent of VEGF-A (vascular endothelial growth factor-A). By recombinant production of CYTL1 we confirm that the peptide is indeed a strong proangiogenic factor and induces sprouting in cellular assays and functional vessel formation in animal models comparable to VEGF-A. Mass spectroscopy corroborates that CYTL1 is specifically O-glycosylated on 2 neighboring threonines in the C-terminal part and this modification is important for its proangiogenic bioactivity. Further analyses show that the factor does not upregulate proinflammatory genes and strongly induces several metallothionein genes encoding anti-inflammatory and antiapoptotic proteins. CONCLUSIONS: We conclude that CYTL1 can mediate proangiogenic functions ascribed to endothelial progenitors such as ECFCs in vivo and may be a candidate to support vessel formation and tissue regeneration in ischemic pathologies.


Subject(s)
Angiogenic Proteins/metabolism , Autocrine Communication , Blood Proteins/metabolism , Corneal Neovascularization , Cytokines/metabolism , Endothelial Progenitor Cells/metabolism , Human Umbilical Vein Endothelial Cells/metabolism , Neovascularization, Physiologic , Paracrine Communication , Angiogenic Proteins/genetics , Animals , Blood Proteins/genetics , Cell Hypoxia , Cytokines/genetics , Disease Models, Animal , Female , Glycosylation , HEK293 Cells , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Male , Mice, Inbred C57BL , Mice, SCID , Secretory Pathway , Signal Transduction , Spheroids, Cellular , Vascular Endothelial Growth Factor A/metabolism
2.
Front Immunol ; 8: 535, 2017.
Article in English | MEDLINE | ID: mdl-28555134

ABSTRACT

We identified ZNF683/HOBIT as the most highly upregulated transcription factor gene during ex vivo differentiation of human CD34+ cord blood progenitor cells to CD56+ natural killer (NK) cells. ZNF683/HOBIT mRNA was preferentially expressed in NK cells compared to other human peripheral blood lymphocytes and monocytes. During ex vivo differentiation, ZNF683/HOBIT mRNA started to increase shortly after addition of IL-15 and further accumulated in parallel to the generation of CD56+ NK cells. shRNA-mediated knockdown of ZNF683/HOBIT resulted in a substantial reduction of CD56-CD14- NK-cell progenitors and the following generation of CD56+ NK cells was largely abrogated. The few CD56+ NK cells, which escaped the developmental inhibition in the ZNF683/HOBIT knockdown cultures, displayed normal levels of NKG2A and KIR receptors. Functional analyses of these cells showed no differences in degranulation capacity from control cultures. However, the proportion of IFN-γ-producing cells appeared to be increased upon ZNF683/HOBIT knockdown. These results indicate a key role of ZNF683/HOBIT for the differentiation of the human NK-cell lineage and further suggest a potential negative control on IFN-γ production in more mature human NK cells.

3.
PLoS One ; 9(1): e87131, 2014.
Article in English | MEDLINE | ID: mdl-24498025

ABSTRACT

The possibility to modulate ex vivo human NK cell differentiation towards specific phenotypes will contribute to a better understanding of NK cell differentiation and facilitate tailored production of NK cells for immunotherapy. In this study, we show that addition of a specific low dose of IL-12 to an ex vivo NK cell differentiation system from cord blood CD34(+) stem cells will result in significantly increased proportions of cells with expression of CD62L as well as KIRs and CD16 which are preferentially expressed on mature CD56(dim) peripheral blood NK cells. In addition, the cells displayed decreased expression of receptors such as CCR6 and CXCR3, which are typically expressed to a lower extent by CD56(dim) than CD56(bright) peripheral blood NK cells. The increased number of CD62L and KIR positive cells prevailed in a population of CD33(+)NKG2A(+) NK cells, supporting that maturation occurs via this subtype. Among a series of transcription factors tested we found Gata3 and TOX to be significantly downregulated, whereas ID3 was upregulated in the IL-12-modulated ex vivo NK cells, implicating these factors in the observed changes. Importantly, the cells differentiated in the presence of IL-12 showed enhanced cytokine production and cytolytic activity against MHC class I negative and positive targets. Moreover, in line with the enhanced CD16 expression, these cells exhibited improved antibody-dependent cellular cytotoxicity for B-cell leukemia target cells in the presence of the clinically applied antibody rituximab. Altogether, these data provide evidence that IL-12 directs human ex vivo NK cell differentiation towards more mature NK cells with improved properties for potential cancer therapies.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/immunology , Cell Differentiation/immunology , Interleukin-2/immunology , Killer Cells, Natural/immunology , Antibodies, Monoclonal, Murine-Derived/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibody-Dependent Cell Cytotoxicity/drug effects , Antigens, CD34/immunology , Antigens, CD34/metabolism , Antineoplastic Agents/immunology , Antineoplastic Agents/pharmacology , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line, Tumor , Cells, Cultured , Dose-Response Relationship, Drug , Fetal Blood/cytology , Fetal Blood/immunology , Fetal Blood/metabolism , Flow Cytometry , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/immunology , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , High Mobility Group Proteins/genetics , High Mobility Group Proteins/immunology , Humans , Immunotherapy, Adoptive/methods , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/immunology , Interleukin-2/pharmacology , K562 Cells , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , L-Selectin/immunology , L-Selectin/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Receptors, CCR6/immunology , Receptors, CCR6/metabolism , Receptors, CXCR3/immunology , Receptors, CXCR3/metabolism , Receptors, IgG/immunology , Receptors, IgG/metabolism , Receptors, KIR/immunology , Receptors, KIR/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rituximab
4.
Stem Cells Dev ; 21(16): 2926-38, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22571679

ABSTRACT

Ex vivo differentiation systems of natural killer (NK) cells from CD34+ hematopoietic stem cells are of potential importance for adjuvant immunotherapy of cancer. Here, we analyzed ex vivo differentiation of NK cells from cord blood-derived CD34+ stem cells by gene expression profiling, real-time RT-PCR, flow cytometry, and functional analysis. Additionally, we compared the identified characteristics to peripheral blood (PB) CD56(bright) and CD56(dim) NK cells. The data show sequential expression of CD56 and the CD94 and NKG2 receptor chains during ex vivo NK cell development, resulting finally in the expression of a range of genes with partial characteristics of CD56(bright) and CD56(dim) NK cells from PB. Expression of characteristic NK cell receptors and cytotoxic genes was mainly found within the predominant ex vivo generated population of NKG2A+ NK cells, indicating the importance of NKG2A expression during NK cell differentiation and maturation. Furthermore, despite distinct phenotypic characteristics, the detailed analysis of cytolytic genes expressed within the ex vivo differentiated NK cells revealed a pattern close to CD56(dim) NK cells. In line with this finding, ex vivo generated NK cells displayed potent cytotoxicity. This supports that the ex vivo differentiation system faithfully reproduces major steps of the differentiation of NK cells from their progenitors, constitutes an excellent model to study NK cell differentiation, and is valuable to generate large-scale NK cells appropriate for immunotherapy.


Subject(s)
Cell Culture Techniques/methods , Cytotoxicity, Immunologic/genetics , Gene Expression Profiling , Gene Expression Regulation , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Receptors, Natural Killer Cell/genetics , Adult , Antibody-Dependent Cell Cytotoxicity/genetics , Biomarkers/metabolism , CD56 Antigen/metabolism , Cell Differentiation/genetics , Cell Proliferation , Cells, Cultured , Flow Cytometry , Humans , Interferon-gamma/biosynthesis , Monocytes/cytology , Monocytes/metabolism , Phenotype , Receptors, Natural Killer Cell/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Time Factors , Up-Regulation/genetics
5.
Interact Cardiovasc Thorac Surg ; 14(1): 120-1, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22108919

ABSTRACT

Acute traumatic transection of the aorta (ATAT) is a devastating event. Transluminal endovascular aortic repair (TEVAR) has meanwhile become an excellent alternative for such lesions. A 27-year old woman sustained a multiple trauma in a car accident including ATAT which was treated by aortic stent graft placement. Six years after the endovascular repair, our patient had only noticed, just shortly after an annual computed tomography (CT) check-up, that she was pregnant and in the 9th week of gestation. The CT was considered as unproblematic for the foetus since direct radiation of the uterus had been avoided. Our patient clearly preferred a vaginal delivery over a caesarean section. The pregnancy was uneventful with uncomplicated vaginal delivery at term. This case shows that vaginal delivery is possible if the stent graft is in regular position without signs of endoleaks and the rest of the aorta is free from dissection or aneurysm formation. It indicates that TEVAR is a sound and durable treatment modality in ATATs and that aortic stent grafts can resist increased intravascular volume and elevated aortic pressure levels as encountered in pregnancy and during vaginal delivery. This particular case also shows that it is possible to respect a patient's right of self-determination.


Subject(s)
Aorta, Thoracic/injuries , Aortic Aneurysm, Thoracic/surgery , Aortic Dissection/surgery , Blood Vessel Prosthesis , Delivery, Obstetric , Pregnancy Complications, Cardiovascular/surgery , Stents , Acute Disease , Adult , Aortic Dissection/diagnostic imaging , Aortic Dissection/etiology , Aorta, Thoracic/diagnostic imaging , Aorta, Thoracic/surgery , Aortic Aneurysm, Thoracic/diagnostic imaging , Aortic Aneurysm, Thoracic/etiology , Female , Follow-Up Studies , Humans , Infant, Newborn , Male , Pregnancy , Pregnancy Complications, Cardiovascular/diagnostic imaging , Thoracic Injuries/complications , Thoracic Injuries/diagnosis , Time Factors , Tomography, X-Ray Computed
6.
J Thorac Cardiovasc Surg ; 142(3): 687-96, 696.e1-2, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21388641

ABSTRACT

OBJECTIVES: Although adequate numbers of hematopoietic progenitor cells reside in the human bone marrow, the extent of endogenous neovascularization after myocardial infarction remains insufficient. The aim of this study was to identify the role of the CXC chemokine receptor 4/stromal cell-derived factor 1 axis in the mobilization and homing of hematopoietic progenitor cells in the ischemic heart. METHODS: Human bone marrow-derived hematopoietic progenitor cells or saline were injected systemically into athymic nude rats 48 hours after myocardial infarction. Myocardial and bone marrow expression of stromal cell-derived factor 1 and chemotaxis of hematopoietic progenitor cells were measured in vitro in the presence or absence of stromal cell-derived factor 1. The role of the CXC chemokine receptor 4/stromal cell-derived factor 1 axis was investigated by means of antibody blockade or systemic administration of granulocyte colony-stimulating factor. Morphologic analysis included measurement of the infarct area, capillary density, and apoptosis, whereas left ventricular function was measured by means of echocardiographic analysis. RESULTS: Expression of postinfarct stromal cell-derived factor 1 was increased by 67% in the bone marrow and decreased by 43% in myocardium. Disruption of bone marrow stromal cell-derived factor 1/CXC chemokine receptor 4 interactions by antibody blockade resulted in a redirection of human hematopoietic progenitor cells from the bone marrow to the ischemic heart and augmented neovascularization and cardiomyocyte survival. Similarly, systemic administration of granulocyte colony-stimulating factor to block CXC chemokine receptor 4/stromal cell-derived factor 1 interaction resulted in increased mobilization and homing of hematopoietic progenitor cells to the ischemic heart, which translated to augmented myocardial neovascularization, prevention of apoptosis, and improved cardiac function. CONCLUSIONS: Bone marrow stromal cell-derived factor 1 upregulation after myocardial ischemia prevents mobilization of endogenous hematopoietic progenitor cells. We provide evidence that disruption of stromal cell-derived factor 1/CXC chemokine receptor 4 interactions allows redirection of hematopoietic progenitor cells to ischemic myocardium and enhances recovery of left ventricular function.


Subject(s)
Chemokine CXCL12/metabolism , Hematopoietic Stem Cells/physiology , Myocardial Infarction/physiopathology , Myocytes, Cardiac/physiology , Receptors, CXCR4/metabolism , Ventricular Remodeling/physiology , Animals , Apoptosis/physiology , Bone Marrow/metabolism , Cell Survival , Chemotaxis/physiology , Coronary Circulation/physiology , Down-Regulation/physiology , Granulocyte Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cell Mobilization , Myocardial Infarction/pathology , Neovascularization, Physiologic/physiology , Rats , Rats, Nude , Recovery of Function/physiology , Tissue Culture Techniques , Ventricular Function, Left/physiology
7.
Circ J ; 74(1): 188-94, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19926917

ABSTRACT

BACKGROUND: Umbilical cord blood (UCB) is a source of human hematopoietic precursor cells (HPCs), a stem cell (SC) type that has been used in several trials for myocardial repair. A certain minimal number of cells is required for measurable regeneration and a major challenge of SC-based regenerative therapy constitutes ex-vivo expansion of the primitive cell compartment. The aim of this study was to investigate the ex-vivo expansion potential of UCB-derived HPCs and the ability of these expanded cells to migrate to the site of damage and improve ventricular function in a rodent model of myocardial infarction (MI). METHODS AND RESULTS: UCB-derived HPCs, defined by coexpression of CD133 and CD34, were expanded using various cytokine combinations. MI was induced by left anterior descending artery ligation in nude rats. Cells were injected intravenously 2 days after infarction. The combination of SC factor, thrombopoietin, flt3-ligand and interleukin-6 was found to be the most effective for inducing proliferation of HPCs. The migratory capacity of expanded HPCs was similar to that of non-expanded HPCs and improvement of ejection fraction was significant in both groups, with a relative increase of >60%. CONCLUSIONS: UCB-derived HPCs can be reproducibly expanded ex-vivo and retain their potential to improve cardiac function post-MI. (Circ J 2010; 74: 188 - 194).


Subject(s)
Cord Blood Stem Cell Transplantation , Heart/physiology , Myocardial Infarction/physiopathology , Regeneration/physiology , Animals , Cell Movement/drug effects , Cell Movement/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Coronary Vessels/physiopathology , Disease Models, Animal , Heart/drug effects , Humans , Interleukins/pharmacology , Ligation , Male , Rats , Rats, Inbred Strains
8.
Interact Cardiovasc Thorac Surg ; 7(2): 249-55, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17925321

ABSTRACT

OBJECTIVES: We previously reported that combined transplantation of skeletal myoblasts and AC-133+ cells leads to improved left ventricular function, reduced infarct size and myocardial apoptosis in a model of chronic ischemia. The aim of this study is to elucidate on the possible mechanisms and to assess new implications in increasing cell therapy efficacy in chronic ischemia. METHODS: Heart failure was induced by LAD-ligation in nude rats. (a) Homologous skeletal myoblasts (SM), (b) human derived AC-133+ cells (SC), (c) combination of both cells (Comb) and (d) culture medium (CM) were injected in the infarct and peri-infarct area, respectively, four weeks after infarction. Cell engraftment was detected by fluorescence microscopy and confirmed by immunohistochemical techniques. Cardiac gene expression levels of VEFG-A, cardiac troponin, ACTA2, SDF-1, TGF-beta-1, were assessed by RT-PCR. RESULTS: Both cell types were detected in the injection areas four weeks after cell transplantation. Double cell therapy led to increased cell engraftment (SM: 52+/-13/mm(2), SC: 45+/-8 in the combination group vs. SM: 31+/-9 and 23+/-7 in the monotherapy groups, P=0.007). This effect was confirmed using PCR. Apoptotic index among engrafted cells was significantly lower in the Comb group (Comb: 0.53+/-0.12 for myoblasts and 0.34+/-0.09 for SC, vs. SM: 0.76+/-0.19 and SC: 0.63+/-0.16, P=0.013). Expression of cardiac troponin was higher in the combination group in the peri-infarct area. Evaluation of capillary density revealed increased angiogenesis in the combination group (Comb: 12.3+/-2.3, SM: 5.2+/-1.2, SC: 8.3+/-1.8, P=0.002). Neoangiogenesis was associated with higher levels of VEGF-A and TGF-beta in the injection areas as detected by RT-PCR. The higher SDF-1 expression in the injected areas implies an increased secretion of chemoattractants by the injected cells, which suggests that the effect of combined cell transplantation is mainly associated with paracrine mechanisms. CONCLUSIONS: The mechanism of functional improvement after combined transplantation of skeletal myoblasts and AC-133+ progenitors in ischemic heart failure is mainly associated with increased angiogenesis based on paracrine factors, which leads to improved survival and lower apoptosis rates of the injected cells.


Subject(s)
Apoptosis , Endothelial Cells/transplantation , Heart Failure/surgery , Myoblasts, Skeletal/transplantation , Myocardial Infarction/surgery , Myocardial Ischemia/complications , Myocardium/pathology , Neovascularization, Physiologic , Stem Cell Transplantation , AC133 Antigen , Actinin/metabolism , Animals , Antigens, CD/analysis , Cell Survival , Cells, Cultured , Chemokine CXCL12/metabolism , Disease Models, Animal , Endothelial Cells/immunology , Glycoproteins/analysis , Heart Failure/etiology , Heart Failure/pathology , Heart Failure/physiopathology , Humans , Male , Myocardial Infarction/etiology , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardial Ischemia/surgery , Myocardium/metabolism , Paracrine Communication , Peptides/analysis , RNA, Messenger/metabolism , Rats , Rats, Inbred F344 , Rats, Nude , Time Factors , Transforming Growth Factor beta1/metabolism , Troponin T/metabolism , Vascular Endothelial Growth Factor A/metabolism
9.
Transpl Int ; 20(9): 731-46, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17555531

ABSTRACT

Despite many advances in cardiovascular medicine, heart failure (HF) remains the leading cause of death in developed countries affecting at least 10 million people in Western Europe alone. The poor long-term prognosis of HF patients, and immense public health implications has fuelled interest in finding new therapeutic modalities. Recent observations of the beneficial effect of stem cells on the damaged heart in animal experiments have generated tremendous excitement and stimulated clinical studies suggesting that this approach is feasible, safe, and potentially effective in humans. Cell-based myocardial regeneration is currently explored for a wide range of cardiac disease states, including acute and chronic ischemic myocardial damage, cardiomyopathy and as biological heart pacemakers. The aim of the present manuscript is to review the work that has been done to establish the role of stem cells in cardiac repair, give an update on the clinical trials performed so far, as well as to discuss critically the controversies, challenges and future surrounding this novel therapeutic concept.


Subject(s)
Heart Diseases/surgery , Heart/physiopathology , Regeneration , Stem Cell Transplantation , Animals , Clinical Trials as Topic , Humans , Stem Cell Transplantation/methods , Stem Cell Transplantation/trends
10.
J Thorac Cardiovasc Surg ; 132(6): 1321-8, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17140950

ABSTRACT

OBJECTIVES: Cellular cardiomyoplasty using skeletal myoblasts or angiopoietic progenitor cells offers a promising approach for the treatment of ischemic heart failure. Although several studies have shown encouraging results in acute myocardial infarction, the efficacy of cell therapy using skeletal myoblasts and angiopoietic progenitor cells in chronic ischemic heart disease remains undetermined. METHODS: Ischemic heart failure was induced by left anterior descending coronary artery ligation in nude rats: (1) Culture medium, (2) homologous skeletal myoblasts (SM), (3) human AC-133+ cells (SC), and (4) both skeletal myoblasts and AC-133+ cells (Comb) were injected in the infarct (SM) and peri-infarct area (SC) 4 weeks after infarction. Assessment of myocardial function included echocardiography 4 weeks after cell delivery. Histology was based on quantification of myocardial fibrosis, apoptosis, and capillary density. RESULTS: Left ventricular dilatation was attenuated and ejection fraction improved significantly after cell transplantation (SM: 59.4% +/- 8.8%, SC: 60.3% +/- 6.6%, Comb: 68.2% +/- 5.6% vs control: 41.5% +/- 7.4%, P = .0013). Quantification of scar tissue showed a significant reduction of infarct area in cell-treated animals (SM: 22.3% +/- 9.1%, SC: 19.8% +/- 7.6%, Comb: 13.2% +/- 5.8% vs controls: 36.5% +/- 8.2%, P = .008). Improvement of myocardial function was associated with reduced apoptotic index (SM: 3.2% +/- 0.9%, SC: 3.1% +/- 0.6%, Comb: 1.8% +/- 0.8% vs controls: 10.3% +/- 1.6%, P = .0002) and increased vascular density (SM: 5.2 +/- 1.2, SC: 8.3 +/- 1.8, Comb: 12.3 +/- 2.3, controls: 1.9 +/- 0.3, all capillary vessels/high-power field, P = .007) in animals after cellular cardiomyoplasty. CONCLUSIONS: Combined transplantation of skeletal myoblasts and angiopoietic progenitor cells results in ventricular function improvement, reduction of scar size and myocardial apoptosis, and increased neoangiogenesis in chronic ischemia. Clinical studies are warranted to prove this new therapeutic concept.


Subject(s)
Apoptosis , Heart Failure/pathology , Heart Failure/surgery , Myoblasts, Skeletal/transplantation , Myocardial Infarction/pathology , Myocardial Infarction/surgery , Myocardial Ischemia/pathology , Myocardial Ischemia/surgery , Neovascularization, Physiologic , Stem Cell Transplantation , Animals , Chronic Disease , Male , Rats
11.
J Heart Lung Transplant ; 22(3): 250-7, 2003 Mar.
Article in English | MEDLINE | ID: mdl-12633691

ABSTRACT

BACKGROUND: Cytomegalovirus (CMV) has emerged as the most important pathogen to affect the post-operative course after heart transplantation. We performed a retrospective analysis to evaluate the efficiency of CMV hyperimmune globulin (CMVIG) prophylaxis in preventing CMV disease in aggressively immunosuppressed patients after heart transplantation. METHODS: We studied 377 heart transplant recipients who received quadruple-immunosuppressive therapy and CMVIG as sole CMV prophylaxis. The study population was categorized into 4 groups according to donor and recipient CMV serology at the time of transplantation (D+/R+, D+/R-, D-/R+, D-/R-) and was monitored for CMV immediate early antigen in peripheral blood cells, in urine sediments, and in throat washings; for the presence of serum CMV immunoglobulin M and CMV immunoglobulin G; and for clinical evidence of CMV-related symptoms. In addition, we compared the incidence of cardiac allograft vasculopathy and infection among the groups. RESULTS: During the first 5 years after transplantation, CMV disease developed in 79 patients (20.96%). Comparison among the groups showed significantly increased risk for CMV disease in allograft recipients of organs from seropositive donors (D+, 27.31%; D-, 11.33%; p = 0.0003). We observed 6 CMV-associated deaths, all in CMV-antibody-negative recipients. Additionally CMV-positive recipients had a greater incidence of cardiac allograft vasculopathy (p = 0.048), and a greater overall infection rate (p = 0.0034). CONCLUSIONS: Cytomegalovirus hyperimmune globulin administration prevents CMV disease and infection in aggressively immunosuppressed heart transplant recipients. Because fatal CMV disease in CMV-negative recipients of organs from seropositive donors could not be prevented with CMVIG alone, we recommend the additional use of prophylactic ganciclovir in this CMV-mismatched population.


Subject(s)
Cytomegalovirus Infections/prevention & control , Heart Transplantation , Immunization, Passive , Immunoglobulins/administration & dosage , Antiviral Agents/therapeutic use , Cytomegalovirus Infections/drug therapy , Female , Follow-Up Studies , Ganciclovir/therapeutic use , Humans , Immunoglobulins, Intravenous , Immunosuppression Therapy , Immunosuppressive Agents/therapeutic use , Male , Middle Aged , Retrospective Studies , Time Factors , Transplantation, Homologous
12.
Ann Thorac Surg ; 73(6): 1843-8, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12078779

ABSTRACT

BACKGROUND: The purpose of this study was to evaluate the significance of aortic rupture on clinical outcome in patients after aortic repair for acute type A dissection. METHODS: One hundred and twenty patients underwent aortic operations with resection of the intimal tear and open distal anastomosis. Median age was 60 years (range 16 to 87); 78 were male. Thirty-six patients had only ascending aortic replacement, 82 had hemiarch repair, and 2 had the entire arch replaced. Retrograde cerebral perfusion was utilized in 66 patients (53%). Rupture defined as free blood in the pericardial space was present in 60 patients (50%). Univariate and multivariate analyses were performed to assess the risk factors for mortality and neurologic dysfunction. RESULTS: Overall hospital mortality rate was 24.2% +/- 4.0% (+/- 70% confidence level) but did not differ between patients with aortic rupture or without (p = 0.83). The incidence of permanent neurologic dysfunction was 9.4% overall, 10.5% with rupture and 8.3% without rupture (p = 0.75). Multivariate analysis revealed absence of retrograde cerebral perfusion and any postoperative complication as statistically significant indicators for in-hospital mortality (p < 0.05). Overall 1- and 5-year survival was 85.3% and 33.7%; among discharged patients, survival in the nonruptured group was 89% and 37%, versus 81% and 31% in the ruptured group (p = 0.01). CONCLUSIONS: Aortic rupture at the time of surgery does not increase the risk of hospital mortality or permanent neurologic complications in patients with acute type A dissections. However, aortic rupture at the time of surgery does influence long-term survival.


Subject(s)
Aortic Aneurysm, Thoracic/surgery , Aortic Dissection/surgery , Aortic Rupture/surgery , Acute Disease , Adolescent , Adult , Aged , Aged, 80 and over , Aortic Dissection/mortality , Aortic Aneurysm, Thoracic/mortality , Aortic Rupture/mortality , Female , Humans , Male , Middle Aged , Nervous System Diseases/etiology , Retrospective Studies , Risk Factors , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...