Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Imaging ; 132014.
Article in English | MEDLINE | ID: mdl-25249170

ABSTRACT

[68Ga]Annexin A5 positron emission tomography (PET) reveals the externalization of phosphatidylserine as a surrogate marker for apoptosis. We tested this technique for therapy monitoring in a murine model of myocardial infarction (MI) including parathyroid hormone (PTH) treatment. MI was induced in mice, and they were assigned to the saline or the PTH group. On day 2, they received [68Ga]annexin A5 PET or histofluorescence TUNEL staining. Mice had 2-deoxy-2-[18F]fluoro-d-glucose (FDG)-PET examinations on days 6 and 30 for calculation of the left ventricular ejection fraction and infarct area. [68Ga]Annexin A5 uptake was 7.4 ± 1.3 %ID/g within the infarction for the controls and 4.5 ± 1.9 %ID/g for the PTH group (p  =  .013). TUNEL staining revealed significantly more apoptotic cells in the infarct area on day 2 in the controls (64 ± 9%) compared to the treatment group (52 ± 4%; p  =  .045). FDG-PET revealed a significant decrease in infarct size in the treatment group and an increase in the controls. Examinations of left ventricular ejection fraction on days 6 and 30 did not reveal treatment effects. [68Ga]Annexin A5 PET can detect the effects of PTH treatment as a marker of apoptosis 2 days after MI; ex vivo examination confirmed significant rescue of myocardiocytes. FDG-PET showed a small but significant reduction in infarct size but no functional improvement.


Subject(s)
Annexin A5 , Enzyme Inhibitors , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/drug therapy , Parathyroid Hormone/administration & dosage , Animals , Annexin A5/chemistry , Apoptosis , Enzyme Inhibitors/chemistry , Fluorodeoxyglucose F18/pharmacokinetics , Gallium Radioisotopes/pharmacokinetics , Male , Mice , Mice, Inbred C57BL , Parathyroid Hormone/therapeutic use , Positron-Emission Tomography/methods , Radiopharmaceuticals/pharmacokinetics , Stroke Volume/drug effects
2.
Magn Reson Imaging ; 32(10): 1259-65, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25171818

ABSTRACT

INTRODUCTION: Cardiac magnetic resonance (CMR) is a unique method to determine regional and local aortic stiffness parameters. Although various methods have been validated, there are no data in patients after acute ST-segment elevation myocardial infarction (STEMI). In the present study we assessed the feasibility of different CMR derived measures of aortic stiffness in patients after first acute STEMI for the first time. METHODS: CMR derived aortic pulse wave velocity (PWV) determined by the regional transit-time (PWVTT) and local flow-area (PWVQA) method as well as local distensibility coefficients (DCs) was analyzed in 22 healthy young volunteers and 28 patients with recent acute STEMI. RESULTS: PWVTT and DC of the ascending aorta differed significantly between healthy subjects and STEMI patients (all p<0.001). PWVQA at thoracic levels of aorta was not different between groups (p>0.520) and did not correlate with age (p>0.149) and PWVTT (p>0.310). Intra- and interobserver variability was high for PWVTT (r=0.970, p<0.001 and r=0.920, p<0.001), acceptable for DC (all r>0. 809, p<0.001 and all r>0.510, p<0.001) but low for thoracic PWVQA (all r<0.330 and all r<0.372). CONCLUSION: PWVTT and local DC are robust methods for the assessment of aortic stiffness in patients after acute STEMI.


Subject(s)
Aorta/pathology , Blood Flow Velocity , Myocardial Infarction/pathology , Myocardium/pathology , Vascular Stiffness , Adult , Age Factors , Aged , Case-Control Studies , Electrocardiography , Female , Healthy Volunteers , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Observer Variation , Pulsatile Flow , Pulse Wave Analysis , Young Adult
3.
PLoS One ; 9(4): e94615, 2014.
Article in English | MEDLINE | ID: mdl-24747816

ABSTRACT

Accurate assessment of left ventricular function in rodent models is essential for the evaluation of new therapeutic approaches for cardiac diseases. In our study, we provide new insights regarding the role of a 1.5 Tesla (T) magnetic resonance imaging (MRI) device and different anesthetic regimens on data validity. As dedicated small animal MRI and echocardiographic devices are not broadly available, we evaluated whether monitoring cardiac function in small rodents with a clinical 1.5 T MRI device is feasible. On a clinical electrocardiogram (ECG) synchronized 1.5 T MRI scanner we therefore studied cardiac function parameters of mice with chronic virus-induced cardiomyopathy. Thus, reduced left ventricular ejection fraction (LVEF) could be verified compared to healthy controls. However, our results showed a high variability. First, anesthesia with medetomidine, midazolam and fentanyl (MMF) led to depressed cardiac function parameters and more variability than isoflurane gas inhalation anesthesia, especially at high concentrations. Furthermore, calculation of an average ejection fraction value from sequenced scans significantly reduced the variance of the results. To sum up, we introduce the clinical 1.5 T MRI device as a new tool for effective analysis of left ventricular function in mice with cardiomyopathy. Besides, we suggest isoflurane gas inhalation anesthesia at high concentrations for variance reduction and recommend calculation of an average ejection fraction value from multiple sequenced MRI scans to provide valid data and a solid basis for further clinical testing.


Subject(s)
Anesthetics/pharmacology , Cardiomyopathies/physiopathology , Heart Function Tests/methods , Heart/drug effects , Heart/physiopathology , Magnetic Resonance Imaging , Animals , Dose-Response Relationship, Drug , Heart Function Tests/instrumentation , Male , Mice , Stroke Volume/drug effects
5.
Angiogenesis ; 17(1): 27-36, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23918206

ABSTRACT

Diabetic foot ulcers represent a therapeutic problem of high clinical relevance. Reduced vascular supply, neuropathy and diminished expression of growth factors strongly contribute to wound healing impairment in diabetes. Secretoneurin, an angiogenic neuropeptide, has been shown to improve tissue perfusion in different animal models by increasing the amount of vessels in affected areas. Therefore, topical secretoneurin gene therapy was tested in a full thickness wound healing model in diabetic db/db mice. Secretoneurin significantly accelerated wound closure in these mice and immunohistochemistry revealed higher capillary and arteriole density in the wounded area compared to control mice. In-vitro, the mechanism of action of secretoneurin on human dermal microvascular endothelial cells was evaluated in normal and diabetic cells. Secretoneurin shows positive effects on in vitro angiogenesis, proliferation and apoptosis of these cells in a basic fibroblast growth factor dependent manner. A small molecular weight inhibitor revealed fibroblast growth factor receptor 3 as the main receptor for secretoneurin mediated effects. Additionally, we could identify heparan-sulfates as important co-factor of secretoneurin induced binding of basic fibroblast growth factor to human dermal endothelial cells. We suggest topical secretoneurin plasmid therapy as new tool for delayed wound healing in patients suffering from diabetes.


Subject(s)
Diabetic Foot , Fibroblast Growth Factor 2/metabolism , Genetic Therapy , Heparitin Sulfate/metabolism , Neuropeptides/biosynthesis , Proteoglycans/metabolism , Secretogranin II/biosynthesis , Wound Healing , Administration, Topical , Animals , Cells, Cultured , Diabetic Foot/genetics , Diabetic Foot/metabolism , Diabetic Foot/pathology , Diabetic Foot/therapy , Fibroblast Growth Factor 2/genetics , Heparitin Sulfate/genetics , Humans , Mice , Mice, Mutant Strains , Plasmids/genetics , Plasmids/pharmacology , Proteoglycans/genetics
6.
Artif Organs ; 38(3): 231-4, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24147881

ABSTRACT

In this retrospective study, we investigated the impact of preconditioning of the right ventricle with the calcium sensitizer levosimendan immediately before left ventricular assist device (LVAD) implantation on outcome and survival. Nine consecutive LVAD patients (seven suffering from dilative cardiomyopathy and two from ischemic cardiomyopathy) with echocardiographic and invasive evidence of right heart insufficiency received levosimendan with 0.1 µg/kg body weight/min for 24 h before implantation of the assist device (seven HeartWare and two Jarvik 2000). Administration of levosimendan was safe and had not to be discontinued in any patient. We observed no relevant side effects. Twelve-month survival after implantation of the LVAD was 89% representing a superior outcome compared with the fifth INTERMACS registry data with 75% survival. Two temporary extracorporeal membrane-oxygenation implantations were necessary due to intraoperative right ventricular dysfunction. Only one patient died 5 weeks after LVAD implantation of multiorgan failure, five patients were successfully transplanted, and three patients underwent LVAD implantation for destination therapy. Levosimendan might improve clinical outcome and survival when used as pretreatment in patients with right heart insufficiency prior to LVAD implantation. However, we recommend a larger controlled trial in the future to confirm our preliminary results.


Subject(s)
Cardiomyopathies/surgery , Cardiotonic Agents/therapeutic use , Heart Failure/surgery , Heart-Assist Devices , Hydrazones/therapeutic use , Pyridazines/therapeutic use , Aged , Cardiomyopathies/drug therapy , Female , Heart Failure/drug therapy , Humans , Male , Middle Aged , Retrospective Studies , Simendan , Treatment Outcome
7.
PLoS One ; 8(9): e74029, 2013.
Article in English | MEDLINE | ID: mdl-24086307

ABSTRACT

Deficient angiogenesis after ischemia may contribute to worse outcome of peripheral arterial disease in patients with diabetes mellitus. Based on our previous work where we demonstrated that Secretoneurin (SN) is up-regulated under hypoxic conditions and enhances angiogenesis, we analyzed the therapeutic potential of SN gene therapy using a model of severe hind limb ischemia in streptozotocin-induced diabetic mice (STZ-DM). After induction of hind limb ischemia, blood flow was assessed by means of laser Doppler perfusion imaging (LDPI) and increased blood perfusion in the SN-treated animal group was observed. These results were complemented by the clinical observation of reduced necrosis and by an increased number of capillaries and arterioles in the SN-treated animal group. In vitro, we found that SN is capable of promoting proliferation and chemotaxis and reduces apoptosis in HUVECs cultured under hyperglycemic conditions. Additionally, SN activated ERK, eNOS and especially AKT as well as EGF-receptor in hyperglycemic HUVECs. In conclusion, we show that SN gene therapy improves post-ischemic neovascularization in diabetic mice through stimulation of angiogenesis and arteriogenesis indicating a possible therapeutic role of this factor in ischemia-related diseases in diabetic patients.


Subject(s)
Diabetes Mellitus, Type 1/therapy , Genetic Therapy , Neovascularization, Physiologic , Neuropeptides/genetics , Regional Blood Flow , Secretogranin II/genetics , Animals , Cells, Cultured , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/physiopathology , Diabetes Mellitus, Experimental/therapy , Diabetes Mellitus, Type 1/complications , Diabetes Mellitus, Type 1/physiopathology , Endothelium, Vascular/cytology , Humans , Ischemia/complications , Ischemia/therapy , Mice , Streptozocin
8.
J Clin Invest ; 2013 Jul 01.
Article in English | MEDLINE | ID: mdl-23863629

ABSTRACT

Septic shock is characterized by increased vascular permeability and hypotension despite increased cardiac output. Numerous vasoactive cytokines are upregulated during sepsis, including angiopoietin 2 (ANG2), which increases vascular permeability. Here we report that mice engineered to inducibly overexpress ANG2 in the endothelium developed sepsis-like hemodynamic alterations, including systemic hypotension, increased cardiac output, and dilatory cardiomyopathy. Conversely, mice with cardiomyocyte-restricted ANG2 overexpression failed to develop hemodynamic alterations. Interestingly, the hemodynamic alterations associated with endothelial-specific overexpression of ANG2 and the loss of capillary-associated pericytes were reversed by intravenous injections of adeno-associated viruses (AAVs) transducing cDNA for angiopoietin 1, a TIE2 ligand that antagonizes ANG2, or AAVs encoding PDGFB, a chemoattractant for pericytes. To confirm the role of ANG2 in sepsis, we i.p. injected LPS into C57BL/6J mice, which rapidly developed hypotension, acute pericyte loss, and increased vascular permeability. Importantly, ANG2 antibody treatment attenuated LPS-induced hemodynamic alterations and reduced the mortality rate at 36 hours from 95% to 61%. These data indicate that ANG2-mediated microvascular disintegration contributes to septic shock and that inhibition of the ANG2/TIE2 interaction during sepsis is a potential therapeutic target.

9.
Int J Cardiol ; 168(4): 3359-69, 2013 Oct 09.
Article in English | MEDLINE | ID: mdl-23669105

ABSTRACT

BACKGROUND: Medical stimulation of endogenous progenitor cell circulation may serve as a new therapeutic tool for treatment of acute myocardial infarction. We analyzed the effects of antidiabetic gliptins plus GCSF (granulocyte colony stimulating factor) on myocardial regeneration after myocardial infarction in a mouse model. METHODS AND RESULTS: After surgical LAD-ligation (left anterior descending artery), Sitagliptin/Vildagliptin was applied yielding sufficient blood levels verified by mass spectrometry and significantly reducing activity of dipeptidyl peptidase (DPP) IV. GCSF or saline was administered intraperitoneally for 6 days. We assessed stem cell mobilization and homing (flow cytometry), infarct size (histology), neovascularization and cellular proliferation (immunohistology), heart function (Millar tip catheterization) and survival (Kaplan-Meier-curves). Gliptins±GCSF administration increased mobilization and cardiac homing of bone-marrow derived stem cells by stabilization of cardiac SDF1 (stromal cell-derived factor). For Sitagliptin, it could be shown that resident cardiac stem cells were stimulated, neovascularization was enhanced and cardiac remodeling was reduced. These effects finally improved myocardial function and increased survival for both gliptins. Although gliptins as a mono therapy lead to remarkable effects in a dose dependent manner and were superior to G-CSF mono-therapy, dual application of GCSF and gliptins revealed the best results. Since both gliptins yielded comparable effects concerning stem cell homing, cardiac function and survival, we suggest a class-effect of DPP-IV-inhibitors. CONCLUSIONS: Thus, gliptins+GCSF and in high concentrations even as mono therapy have beneficial effects on cardiac regeneration after myocardial infarction beyond its anti-diabetic potential.


Subject(s)
Adamantane/analogs & derivatives , Granulocyte Colony-Stimulating Factor/administration & dosage , Heart/drug effects , Hypoglycemic Agents/administration & dosage , Myocardial Infarction/drug therapy , Nitriles/administration & dosage , Pyrazines/administration & dosage , Pyrrolidines/administration & dosage , Triazoles/administration & dosage , Adamantane/administration & dosage , Animals , Cell Survival/drug effects , Cell Survival/physiology , Drug Therapy, Combination , Heart/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Sitagliptin Phosphate , Vildagliptin
10.
Cardiovasc Res ; 97(3): 454-63, 2013 Mar 01.
Article in English | MEDLINE | ID: mdl-23241315

ABSTRACT

AIMS: Future cardiac repair strategies will require a profound understanding of the principles underlying cardiovascular differentiation. Owing to its extracorporal and rapid development, Xenopus laevis provides an ideal experimental system to address these issues in vivo. Whereas mammalian MesP1 is currently regarded as the earliest marker for the cardiovascular system, several MesP1-related factors from Xenopus-mespa, mespb, and mespo-have been assigned only to somitogenesis so far. We, therefore, analysed these genes comparatively for potential contributions to cardiogenesis. METHODS AND RESULTS: RNA in situ hybridizations revealed a novel anterior expression domain exclusively occupied by mespa during gastrulation, which precedes the prospective heart field. Correspondingly, when overexpressed mespa most strongly induced cardiac markers in vivo as well as ex vivo. Transference to murine embryonic stem (ES) cells and subsequent FACS analyses for Flk-1 and Troponin I confirmed the high potential of mespa as a cardiac inducer. In vivo, Morpholino-based knockdown of mespa protein led to a dramatic loss of pro-cardiac and sarcomeric markers, which could be rescued either by mespa itself or human MesP1, but neither by mespb nor mespo. Epistatic analysis positioned mespa upstream of mespo and mespb, and revealed positive autoregulation for mespa at the time of its induction. CONCLUSIONS: Our findings contribute to the understanding of conserved events initiating vertebrate cardiogenesis. We identify mespa as functional amphibian homologue of mammalian MesP1. These results will enable the dissection of cardiac specification from the very beginning in the highly versatile Xenopus system.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/physiology , Cell Differentiation/physiology , Heart/embryology , Myocytes, Cardiac/cytology , Xenopus Proteins/physiology , Xenopus laevis/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Gene Expression Regulation, Developmental/drug effects , Heart/physiology , Mice , Models, Animal , Morpholinos/pharmacology , Myocytes, Cardiac/physiology , Xenopus Proteins/genetics , Xenopus laevis/physiology
11.
Circulation ; 124(25): 2892-902, 2011 Dec 20.
Article in English | MEDLINE | ID: mdl-22144571

ABSTRACT

BACKGROUND: Excessive formation of reactive oxygen species contributes to tissue injury and functional deterioration after myocardial ischemia/reperfusion. Especially, mitochondrial reactive oxygen species are capable of opening the mitochondrial permeability transition pore, a harmful event in cardiac ischemia/reperfusion. Thioredoxins are key players in the cardiac defense against oxidative stress. Mutations in the mitochondrial thioredoxin reductase (thioredoxin reductase-2, Txnrd2) gene have been recently identified to cause dilated cardiomyopathy in patients. Here, we investigated whether mitochondrial thioredoxin reductase is protective against myocardial ischemia/reperfusion injury. METHODS AND RESULTS: In mice, α-MHC-restricted Cre-mediated Txnrd2 deficiency, induced by tamoxifen (Txnrd2-/-ic), aggravated systolic dysfunction and cardiomyocyte cell death after ischemia (90 minutes) and reperfusion (24 hours). Txnrd2-/-ic was accompanied by a loss of mitochondrial integrity and function, which was resolved on pretreatment with the reactive oxygen species scavenger N-acetylcysteine and the mitochondrial permeability transition pore blocker cyclosporin A. Likewise, Txnrd2 deletion in embryonic endothelial precursor cells and embryonic stem cell-derived cardiomyocytes, as well as introduction of Txnrd2-shRNA into adult HL-1 cardiomyocytes, increased cell death on hypoxia and reoxygenation, unless N-acetylcysteine was coadministered. CONCLUSIONS: We report that Txnrd2 exerts a crucial function during postischemic reperfusion via thiol regeneration. The efficacy of cyclosporin A in cardiac Txnrd2 deficiency may indicate a role for Txnrd2 in reducing mitochondrial reactive oxygen species, thereby preventing opening of the mitochondrial permeability transition pore.


Subject(s)
Mitochondria/enzymology , Myocardial Reperfusion Injury/metabolism , Oxidative Stress/physiology , Sulfhydryl Compounds/metabolism , Thioredoxin Reductase 2/metabolism , Acetylcysteine/pharmacology , Animals , Cell Death/drug effects , Cell Death/physiology , Cells, Cultured , Cyclosporine/pharmacology , Embryonic Stem Cells/cytology , Endothelial Cells/cytology , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Gene Expression Regulation, Enzymologic/physiology , Hematopoietic Stem Cells/cytology , Mice , Mice, Knockout , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocytes, Cardiac/cytology , Oxidative Stress/drug effects , Thioredoxin Reductase 1/genetics , Thioredoxin Reductase 1/metabolism , Thioredoxin Reductase 2/genetics
12.
Stem Cell Res ; 7(3): 244-55, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21752744

ABSTRACT

BACKGROUND: G-CSF based stem cell mobilization and stabilization of cardiac SDF-1 by DPP-IV-inhibition (dual stem cell therapy) improve heart function and survival after myocardial infarction. However, it is barely understood whether this new approach acts specifically through the SDF-1/CXCR4 axis, stimulation of resident cardiac stem cells and improved myocardial perfusion. Therefore, we aimed to clarify the role of the SDF1/CXCR4 axis with respect to the benefits of a dual stem cell based therapy. METHODOLOGY/PRINCIPAL FINDINGS: After surgically induced ligation of the LAD, SDF-1/CXCR4 interactions were specifically blocked by the CXCR4 receptor antagonist AMD3100 in G-CSF and Diprotin A treated C57BL/6 mice. G-CSF+DipA treated and non-treated animals served as controls. Because AMD3100 is known to mobilize bone marrow derived stem cells (BMCs) in high concentrations, the optimal dosage (1.25mg per kg body weight) sufficient to block CXCR4 without stimulating mobilization was established. AMD3100 treatment of G-CSF and Diprotin A stimulated mice significantly decreased myocardial homing of circulating stem cells (FACS analysis) and inverted the beneficial effects of (i) cardiac remodeling (histological analyses), (ii) heart function (Millar tip catheterization) and (iii) survival (Kaplan-Meier curves). G-CSF treatment in combination with DPP-IV inhibition enhanced neovascularization at the infarct border zone which was related to an improved myocardial blood flow as measured by SPECT. Moreover, dual stem cell treatment effectively stimulated the pool of resident cardiac stem cells (FACS) which was reversed by AMD3100 treatment. CONCLUSIONS/SIGNIFICANCE: Our data give final proof that homing through the SDF-1/CXCR-4 axis is essential for the success of dual stem cell therapy.


Subject(s)
Chemokine CXCL12/metabolism , Hematopoietic Stem Cell Mobilization , Myocardial Infarction/therapy , Receptors, CXCR4/metabolism , Stem Cell Transplantation , Animals , Antigens, CD34/metabolism , Benzylamines , Chemokine CXCL12/antagonists & inhibitors , Cyclams , Dipeptidyl Peptidase 4/metabolism , Dose-Response Relationship, Drug , Granulocyte Colony-Stimulating Factor/pharmacology , Heart Function Tests/drug effects , Heterocyclic Compounds/pharmacology , Leukocyte Common Antigens/metabolism , Mice , Models, Biological , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/drug effects , Oligopeptides/pharmacology , Perfusion , Proto-Oncogene Proteins c-kit/metabolism , Receptors, CXCR4/antagonists & inhibitors , Survival Analysis
13.
Expert Rev Med Devices ; 8(2): 209-25, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21381911

ABSTRACT

Ischemic heart diseases are the leading cause of death in the Western world. With increasing numbers of patients surviving their acute myocardial infarction owing to effective heart catheter techniques and intensive care treatment, congestive heart failure has become an increasing health concern. With therapeutic options for the prevention and treatment of ischemic heart disease being limited at present, huge efforts have been made in the field of stem cell research to try to establish new approaches for myocardial tissue regeneration. Owing to their pronounced differentiation potential, pluripotent stem cells seem to represent the most promising cell source for future engineering of myocardial replacement tissue. However, several crucial hurdles regarding cell yield and purity of the cultured cardiovascular progenitor cells have still not been overcome to facilitate a clinical application today. By contrast, plenty of adult stem and progenitor cells have already been well characterized and investigated in human disease. However, all of these heterogeneous cell lines primarily seem to work in a paracrine manner on ischemic myocardial tissue, rather than transdifferentiating into contractile cardiomyocytes. This article will focus on the production, application and present limitations of stem cells potentially applicable for myocardial repair.


Subject(s)
Myocardial Ischemia/pathology , Myocardial Ischemia/therapy , Myocardium/pathology , Stem Cell Transplantation , Stem Cells/cytology , Wound Healing , Adult Stem Cells/cytology , Adult Stem Cells/transplantation , Bone Marrow Cells/cytology , Cell Line , Humans , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/transplantation , Tissue Engineering
15.
Exp Hematol ; 38(11): 1105-14, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20621157

ABSTRACT

OBJECTIVE: Clinical studies suggest that transplantation of total bone marrow (BM) after myocardial infarction (MI) is feasible and potentially effective. However, focusing on a defined BM-derived stem cell type may enable a more specific and optimized treatment. Multilineage differentiation potential makes BM-derived multipotent adult progenitor cells (MAPCs) a promising stem cell pool for regenerative purposes. We analyzed the cardioregenerative potential of human MAPCs in a murine model of myocardial infarction. MATERIALS AND METHODS: Human MAPCs were selected by negative depletion of CD45(+)/glycophorin(+) BM cells and plated on fibronectin-coated dishes. In vitro, stem cells were analyzed by reverse transcription polymerase chain reaction. In vivo, we transplanted human MAPCs (5 × 10(5)) by intramyocardial injection after MI in severe combined immunodeficient (SCID) beige mice. Six and 30 days after the surgical procedure, pressure-volume relationships were investigated in vivo. Heart tissues were analyzed immunohistochemically. RESULTS: Reverse transcription polymerase chain reaction experiments on early human MAPC passages evidenced an expression of Oct-4, a stem cell marker indicating pluripotency. In later passages, cardiac markers (Nkx2.5, GATA4, MLC-2v, MLC-2a, ANP, cTnT, cTnI,) and smooth muscle cell markers (SMA, SM22α) were expressed. Transplantation of human MAPCs into the ischemic border zone after MI resulted in an improved cardiac function at day 6 (ejection fraction, 26% vs 20%) and day 30 (ejection fraction, 30% vs 23%). Confirmation of human MAPC marker vimentin in immunohistochemistry demonstrated that human MAPC integrated in the peri-infarct region. The proliferation marker Ki67 was absent in immunohistochemistry and teratoma formation was not found, indicating no tumorous potential of transplanted human MAPCs in the tumor-sensitive SCID model. CONCLUSIONS: Transplantation of human MAPCs after MI ameliorates myocardial function, which may be explained by trophic effects of human MAPCs. Lack of evidence of tumorous potential in the tumor-sensitive SCID model indicates that human MAPCs may deliver an effective and safe stem cell pool for potential treatment of ischemic heart disease.


Subject(s)
Adult Stem Cells/cytology , Multipotent Stem Cells/cytology , Myocardial Infarction/surgery , Regeneration/physiology , Stem Cell Transplantation/methods , Actins/genetics , Adult , Adult Stem Cells/metabolism , Animals , Cells, Cultured , Coculture Techniques , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Expression Profiling , HLA Antigens/metabolism , Hemodynamics , Humans , Immunohistochemistry , Male , Mice , Mice, SCID , Multipotent Stem Cells/metabolism , Muscle, Smooth/chemistry , Myocardial Infarction/physiopathology , Myocardium/metabolism , Myocardium/pathology , Octamer Transcription Factor-3/genetics , Reverse Transcriptase Polymerase Chain Reaction , Troponin I/genetics , Troponin T/genetics
16.
Int J Cardiol ; 145(2): 282-284, 2010 Nov 19.
Article in English | MEDLINE | ID: mdl-20051299

ABSTRACT

AIMS: Our pre-clinical studies demonstrated that G-CSF based stem cell mobilization in combination with genetic or pharmaceutical CD26/DPP-IV inhibition after acute myocardial infarction leads to improved cardiac homing of stem cells, enhanced heart function and increased survival. Thereupon, we initiated a phase III, multi-centre, randomised, placebo-controlled efficacy and safety study (n=100) analyzing the effect of combined application of G-CSF and Sitagliptin, which is a clinically admitted, anti-diabetic DPP-IV-inhibitor, after acute myocardial infarction ("SITAGRAMI-Trial"; EudraCT Number: 2007-003941-34). METHODS: The primary objective of the study is to assess myocardial regeneration by improved myocardial homing of mobilized stem cells, as measured by cardiac function using MRI analysis. In this paper, we report on the study design and a planned first interim-analysis on safety issues without unblinding. RESULTS: During the first 6 weeks of follow-up, only two major adverse cardiac events occurred (one de novo stenosis and one instent-restenosis) in the first 36 patients. Presumably, they were not related to any study medication. No other side effects like headache, bone pain, hypoglycaemias etc. were observed. Furthermore, no myocardial infarction or death occurred in any patient. Thus, the rate of serious adverse events lay within the expected range. CONCLUSIONS: Our data demonstrate that the combined application of Sitagliptin and G-CSF seems to be safe on the short term and feasible after acute myocardial infarction and may represent a new therapeutic option in future.


Subject(s)
Granulocyte Colony-Stimulating Factor/administration & dosage , Granulocyte Colony-Stimulating Factor/adverse effects , Myocardial Infarction/drug therapy , Pyrazines/administration & dosage , Pyrazines/adverse effects , Triazoles/administration & dosage , Triazoles/adverse effects , Adult , Aged , Aged, 80 and over , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Male , Middle Aged , Myocardial Infarction/physiopathology , Sitagliptin Phosphate , Treatment Outcome
17.
Int J Cardiol ; 144(3): 399-404, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-19477537

ABSTRACT

AIMS: The aims of this trial were to investigate the effect of granulocyte colony-stimulating factor (G-CSF) on left-ventricular ejection fraction and event-free survival in patients suffering from sub-acute myocardial infarction (STEMI). METHODS: We enrolled 44 patients suffering from sub-acute STEMI with late revascularization achieved by percutaneous coronary intervention (PCI). Patients were randomized to receive either G-CSF (Filgrastim) at a dose of 10 µg/kg body weight/day subcutaneously or placebo. Changes of global and regional cardiac function from baseline (1 week after PCI) over 1 and 3 months to 12 months of follow-up were analyzed by magnetic resonance imaging. RESULTS: Ejection fraction improved in G-CSF treated patients from 41.1±11.9% to 47.1±11.9% (3 months) and decreased slightly to 45.7±15.1% after 1 year. Ejection fraction also improved in the placebo group from 43.8±9.0% to 49.5±11.8% (3 months) and decreased slightly to 42.9±15.4% after 1 year (1 year MRI follow-up was performed in 23 out initial 44 patients). There was no significant difference between the two groups at any time point. Other parameters such as infarct size, myocardial perfusion, left ventricular end-diastolic and end-systolic volumes were not different between the two groups. Event-free survival of such as death, (re) myocardial infarction or acute coronary syndromes, coronary artery bypass grafting and target lesion revascularization was not significantly different between both groups. CONCLUSIONS: G-CSF administration after sub-acute STEMI is feasible and safe but does not improve myocardial function or survival when used as a single substance.


Subject(s)
Angioplasty, Balloon, Coronary , Granulocyte Colony-Stimulating Factor/therapeutic use , Heart Conduction System/physiopathology , Myocardial Infarction/physiopathology , Myocardial Infarction/therapy , Ventricular Function, Left/drug effects , Aged , Double-Blind Method , Female , Filgrastim , Follow-Up Studies , Granulocyte Colony-Stimulating Factor/administration & dosage , Humans , Injections, Subcutaneous , Magnetic Resonance Imaging , Male , Middle Aged , Myocardial Infarction/diagnosis , Recombinant Proteins , Stroke Volume/drug effects , Treatment Outcome
18.
J Surg Res ; 151(1): 153-62, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18621399

ABSTRACT

BACKGROUND: Therapeutic angiogenesis has become a key technology in experimental and clinical medicine. Only few data are available on the effects of timing and targeting of therapeutic proteins after cell-based gene transfer. This work investigates such effects after temporary expression of vascular endothelial growth factor 165 (VEGF(165)), the most commonly used angiogenic protein for therapeutic purposes. METHODS: We established a cell-based gene-transfer model using fibroblasts to temporarily produce VEGF(165). Cells were implanted into 40 rats. Protein expression and angiogenic effects were measured by PCR, immunohistology, and microangiography. To determine an improvement for survival of ischemically challenged tissue, cells were implanted in an ischemic flap model at different locations and time points. RESULTS: After implantation of modified cells, a temporary increase was found in the target tissue for VEGF(165), endothelial cell counts, and capillary network formations. Four wk later, histological alterations in the target tissue area were not different from controls. Implantation of modified cells into flap plus wound margin 1 wk before surgery showed significant improvement of tissue survival demonstrated by planimetric measurements and blood vessels counting in the target tissue. CONCLUSION: In our model, temporary expression of VEGF(165) induces therapeutically relevant angiogenesis and improves blood supply only if applied 1 wk before ischemia. It is essential to include the surrounding area for induction of angiogenesis in this model. In contrast, the angiogenic effects are not effective in the target area and its surrounding tissue, if therapeutic gene expression is started during onset of ischemia or 2 wk before ischemia in this model.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation/physiology , Genetic Therapy/methods , Ischemia/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adenoviridae , Animals , Cell Proliferation , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Female , Fibroblasts/cytology , Gene Transfer Techniques , Ischemia/pathology , Models, Animal , Neovascularization, Physiologic/physiology , Rats , Rats, Sprague-Dawley , Surgical Flaps/blood supply , Time Factors , Transfection , Vascular Endothelial Growth Factor A/genetics
19.
Exp Hematol ; 36(6): 703-9, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18346835

ABSTRACT

OBJECTIVE: Granulocyte colony-stimulating factor (G-CSF) improves myocardial function after infarction in vivo. Placebo-controlled clinical studies failed to show beneficial effects on myocardial function. Recent data demonstrate that the time point of treatment initiation may be crucial for the efficacy of G-CSF. We investigated the influence of the timing of G-CSF treatment on myocardial function and perfusion in a subgroup study of the G-CSF-ST Elevation Myocardial Infarction trial. MATERIALS AND METHODS: Patients with late revascularized myocardial infarction (n = 44) were treated with either G-CSF or placebo over 5 days after successful percutaneous coronary intervention (PCI). Of the G-CSF group, 13 patients had received G-CSF early treatment started within 24 hours after PCI (mean: 16 +/- 6 hours). In 10 patients, G-CSF was initiated late (>24 hours after PCI, mean: 49 +/- 26 hours). Global and regional myocardial function and perfusion were assessed from baseline to 3 months after PCI using magnetic resonance imaging in 37 patients who completed magnetic resonance follow-up. RESULTS: G-CSF was safe when used early or late after PCI. Early G-CSF administration resulted in significantly improved perfusion at rest 1 month after PCI when compared to placebo (Up-slope, signal intensity 1.2 [0.4-1.8] vs 0.6 [0.1-1.3], p = 0.03). Timing of G-CSF had no influence on global and regional function. CONCLUSION: This post-hoc analysis indicates that timing of G-CSF after myocardial infarction does not improve myocardial function but myocardial perfusion if the cytokine is given early. This urges the need to investigate alternative dosage regimens or combination with novel therapeutics promoting mobilization and homing.


Subject(s)
Granulocyte Colony-Stimulating Factor/therapeutic use , Myocardial Infarction/drug therapy , Aged , Drug Administration Schedule , Female , Granulocyte Colony-Stimulating Factor/administration & dosage , Hematopoietic Stem Cell Mobilization , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Myocardial Infarction/pathology , Myocardial Revascularization , Stem Cell Transplantation , Treatment Outcome
20.
Respir Physiol Neurobiol ; 160(2): 232-8, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18054528

ABSTRACT

We wanted to test if a sojourn at moderate altitude can activate circulation of adult progenitor cells in healthy individuals. Thus, we investigated 11 healthy volunteers, who spent 1-week at 1700 m (Oberlech, Austria,) simulating an active holiday. We measured circulating CD34(+) progenitor cell populations by flow cytometry and cytokines (using ELISA) in peripheral blood at baseline (500 m) and at the end of the sojourn. Extent of physical activity was documented via armband. CD34(+)CXCR-4(+) cells significantly increased in peripheral blood after the sojourn. CD34(+)CD31(+) and CD34(+)CD133(+) cells were upregulated in trend. Levels of SDF-1, G-CSF and VEGF decreased in trend whereas erythropoietin and SCF remained equal. Progenitor cells and degree of daily physical exercise did not correlate. We present the first study showing that exposure to moderate altitude with physical activity leads to increased levels of circulating progenitor cells. This effect may be due to hypoxia and/or physical activity.


Subject(s)
Adaptation, Physiological/physiology , Adult Stem Cells/metabolism , Altitude , Antigens, CD34/metabolism , Exercise/physiology , Adult , Adult Stem Cells/cytology , Female , Humans , Lymphocyte Subsets/metabolism , Male , Receptors, CXCR4/metabolism , Reference Values , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...