Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters











Database
Language
Publication year range
1.
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
2.
Cardiovasc Res ; 90(3): 529-37, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21245057

ABSTRACT

AIMS: Parathyroid hormone (PTH) has been shown to promote stem cell mobilization into peripheral blood. Moreover, PTH treatment after myocardial infarction (MI) improved survival and myocardial function associated with enhanced homing of bone marrow-derived stem cells (BMCs). To unravel the molecular mechanisms of PTH-mediated stem cell trafficking, we analysed wild-type (wt) and green fluorescent protein (GFP)-transgenic mice after MI with respect to the pivotal stromal cell-derived factor-1 (SDF-1)/chemokine receptor type 4 (CXCR4) axis. METHODS AND RESULTS: WT and GFP-transgenic mice (C57BL/6J) were infarcted by coronary artery ligation and PTH (80 µg/kg/day) was injected for 6 days afterwards. Number of BMCs was analysed by flow cytometry. SDF-1 protein levels and activity of dipeptidyl peptidase-IV (DPP-IV) were investigated by ELISA and activity assay. Functional analyses were performed at day 30 after MI. PTH-treated animals revealed an enhanced homing of CXCR4(+) BMCs associated with an increased protein level of the corresponding homing factor SDF-1 in the ischaemic heart. In vitro and in vivo, PTH inhibited the activity of DPP-IV, which cleaves and inactivates SDF-1. Functionally, PTH significantly improved myocardial function after MI. Both stem cell homing as well as functional recovery were reversed by the CXCR4 antagonist AMD3100. CONCLUSION: In summary, PTH is a DPP-IV inhibitor leading to an increased cardiac SDF-1 level, which enhances recruitment of CXCR4(+) BMCs into the ischaemic heart associated with attenuated ischaemic cardiomyopathy. Since PTH is already clinically used our findings may have direct impact on the initiation of studies in patients with ischaemic disorders.


Subject(s)
Chemokine CXCL12/metabolism , Dipeptidyl Peptidase 4/metabolism , Hematopoietic Stem Cell Mobilization/methods , Myocardial Ischemia/therapy , Parathyroid Hormone/pharmacology , Receptors, CXCR4/metabolism , Adult Stem Cells/drug effects , Adult Stem Cells/metabolism , Adult Stem Cells/pathology , Animals , Benzylamines , Cardiotonic Agents/pharmacology , Cyclams , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Heterocyclic Compounds/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/prevention & control , Protease Inhibitors/pharmacology , Receptors, CXCR4/antagonists & inhibitors , Regenerative Medicine/methods
3.
Am J Physiol Heart Circ Physiol ; 298(5): H1466-71, 2010 May.
Article in English | MEDLINE | ID: mdl-20207820

ABSTRACT

Mobilization of stem cells by granulocyte colony-stimulating factor (G-CSF) was shown to have protective effects after myocardial infarction (MI); however, clinical trials failed to be effective. In search for alternative cytokines, parathyroid hormone (PTH) was recently shown to promote cardiac repair by enhanced neovascularization and cell survival. To compare the impact of the two cytokines G-CSF and PTH on myocardial perfusion, mice were noninvasively and repetitively investigated by pinhole single-photon emission computed tomography (SPECT) after MI. Mobilization and homing of bone marrow-derived stem cells (BMCs) was analyzed by fluorescence-activated cell sorter (FACS) analysis. Mice (C57BL/6J) were infarcted by left anterior descending artery ligation. PTH (80 mug/kg) and G-CSF (100 mug/kg) were injected for 5 days. Perfusion defects were determined by (99m)Tc-sestamibi SPECT at days 6 and 30 after MI. The number of BMCs characterized by Lin(-)/Sca-1(+)/c-kit(+) cells in peripheral blood and heart was analyzed by FACS. Both G-CSF and PTH treatment resulted in an augmented mobilization of BMCs in the peripheral blood. Contrary to G-CSF and controls, PTH and the combination showed significant migration of BMCs in ischemic myocardium associated with a significant reduction of perfusion defects from day 6 to day 30. A combination of both cytokines had no additional effects on migration and perfusion. In our preclinical model, SPECT analyses revealed the functional potential of PTH reducing size of infarction together with an enhanced homing of BMCs to the myocardium in contrast to G-CSF. A combination of both cytokines did not improve the functional outcome, suggesting clinical applications of PTH in ischemic heart diseases.


Subject(s)
Bone Marrow Transplantation/physiology , Coronary Circulation/drug effects , Granulocyte Colony-Stimulating Factor/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Infarction/physiopathology , Parathyroid Hormone/therapeutic use , Animals , Cytokines/metabolism , Flow Cytometry , In Vitro Techniques , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/diagnostic imaging , Myocardium/cytology , Myocardium/pathology , Perfusion , Radiopharmaceuticals , Technetium Tc 99m Sestamibi , Tomography, Emission-Computed, Single-Photon , Treatment Outcome
4.
Mol Imaging Biol ; 12(4): 427-34, 2010 Aug.
Article in English | MEDLINE | ID: mdl-19937392

ABSTRACT

INTRODUCTION: The aim of this study is to evaluate a non-invasive method for measuring myocardial perfusion defect size in mice using a clinical single-photon emission computed tomography system equipped with pinhole collimators (pinhole SPECT). MATERIALS AND METHODS: Thirty days after ligation of the left anterior descending coronary artery, 13 mice (C57BL/6J) were imaged following intravenous injection of 370 MBq [99mTc]sestamibi. Eight control mice without myocardial infarction were likewise investigated. Image quality optimization had been achieved by repeated scanning of a multiple point phantom, with varying zoom factors, number of projection angles, and pinhole diameter. Volumetric sampling was used to generate polar maps, in which intensity was normalized to that of a standard septal region of interest (ROI), which was set at 100%. Receiver operating characteristic analyses were performed to define an optimal threshold as compared to histologically measured defect sizes, which were considered as gold standard. RESULTS: A spatial resolution of 1.9 mm was achieved using a pinhole diameter of 0.5 mm, a zoom factor of 2, and 6 degrees projection angles. Histological results were best reproduced by a 60% threshold relative to the septal reference ROI. By applying this threshold, SPECT perfusion defect sizes revealed very high correlation to the histological results (R(2) = 0.867) with excellent intra- and interobserver reproducibility (intraclass correlation coefficients of 0.84 and 0.82). CONCLUSIONS: We achieved a spatial resolution of 1.9 mm in myocardial perfusion imaging in mice using a clinical SPECT system mounted with pinhole collimators. Compared to a histological gold standard, the infarct sizes were accurately estimated, indicating that this method shows promise to monitor experimental cardiac interventions in mice.


Subject(s)
Myocardial Infarction/diagnosis , Myocardial Infarction/pathology , Perfusion/instrumentation , Tomography, Emission-Computed, Single-Photon/instrumentation , Tomography, Emission-Computed, Single-Photon/methods , Animals , Feasibility Studies , Mice , Phantoms, Imaging
5.
Cell Stem Cell ; 4(4): 313-23, 2009 Apr 03.
Article in English | MEDLINE | ID: mdl-19341621

ABSTRACT

Ischemic cardiomyopathy is one of the main causes of death, which may be prevented by stem cell-based therapies. SDF-1alpha is the major chemokine attracting stem cells to the heart. Since SDF-1alpha is cleaved and inactivated by CD26/dipeptidylpeptidase IV (DPP-IV), we established a therapeutic concept--applicable to ischemic disorders in general--by combining genetic and pharmacologic inhibition of DPP-IV with G-CSF-mediated stem cell mobilization after myocardial infarction in mice. This approach leads to (1) decreased myocardial DPP-IV activity, (2) increased myocardial homing of circulating CXCR-4+ stem cells, (3) reduced cardiac remodeling, and (4) improved heart function and survival. Indeed, CD26 depletion promoted posttranslational stabilization of active SDF-1alpha in heart lysates and preserved the cardiac SDF-1-CXCR4 homing axis. Therefore, we propose pharmacological DPP-IV inhibition and G-CSF-based stem cell mobilization as a therapeutic concept for future stem cell trials after myocardial infarction.


Subject(s)
Chemokine CXCL12/metabolism , Dipeptidyl-Peptidase IV Inhibitors , Granulocyte Colony-Stimulating Factor/therapeutic use , Heart/drug effects , Hematopoietic Stem Cells/physiology , Myocardial Infarction/drug therapy , Receptors, CXCR4/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Dipeptidyl Peptidase 4/genetics , Dipeptidyl Peptidase 4/metabolism , Granulocyte Colony-Stimulating Factor/pharmacology , Heart/physiology , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/drug effects , Kaplan-Meier Estimate , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/drug effects , Neovascularization, Physiologic/physiology
6.
FASEB J ; 23(2): 351-61, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18827024

ABSTRACT

Mobilization of bone marrow-derived stem cells (BMCs) was shown to have protective effects after myocardial infarction (MI). However, the classical mobilizing agent, granulocyte-colony stimulating factor (G-CSF) relapsed after revealing an impaired homing capacity. In the search for superior cytokines, erythropoietin (EPO) appears to be a promising agent. Therefore, we analyzed in a murine model of surgically induced MI the influence of EPO treatment on survival and functional parameters as well as BMC mobilization, homing, and effect on resident cardiac stem cells (CSCs). Human EPO was injected intraperitoneally after ligation of the left anterior descendens (LAD) for 3 days with a total dose of 5000 IU/kg 6 and 30 days after MI, and pressure volume relationships were investigated in vivo. Cardiac tissues were analyzed by histology. To show the effect on BMCs and CSCs, FACS analyses were performed. Homing factors were analyzed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and ELISA. EPO-treated animals showed a significant improvement of survival post-MI (62 vs. 36%). At days 6 and 30, all hemodynamic parameters associated with attenuated remodeling, enhanced neovascularization, and diminished apoptotic cells in the peri-infarct area were improved. BMC subpopulations (CD31(+), c-kit(+), and Sca-1(+) cells) were mobilized, and homing of Sca-1(+) and CXCR4(+) BMCs toward an SDF-1 gradient into the ischemic myocardium was enhanced. However, there was no beneficial effect on CSCs. We have shown that EPO application after MI shows cardioprotective effects. This may be explained by mobilization of BMCs, which are homing via the CXCR-4/SDF-1 axis. However, EPO has no beneficial effects on resident CSCs. Therefore, new treatment regimes using EPO together with other agents may combine complementary beneficial effects preventing ischemic cardiomyopathy.


Subject(s)
Bone Marrow Cells/cytology , Cardiomegaly/drug therapy , Chemokine CXCL12/metabolism , Erythropoietin/therapeutic use , Myocardial Infarction/drug therapy , Receptors, CXCR4/metabolism , Stem Cells/metabolism , Animals , Antigens, CD34/metabolism , Cardiomegaly/complications , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Movement , Erythropoietin/administration & dosage , Female , Immunohistochemistry , Leukocyte Common Antigens/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/complications , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Stem Cells/cytology , Survival Rate , Time Factors
7.
Exp Hematol ; 36(6): 695-702, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18346841

ABSTRACT

OBJECTIVE: Besides its classical function in the field of autologous and allogenic stem cell transplantation, granulocyte colony-stimulating factor (G-CSF) was shown to have protective effects after myocardial infarction (MI) by mobilization of bone marrow-derived progenitor cells (BMCs) and in addition by activation of multiple signaling pathways. In the present study, we focused on the impact of G-CSF on migration of BMCs and the impact on resident cardiac cells after MI. MATERIALS AND METHODS: Mice (C57BL/6J) were sublethally irradiated, and BM from green fluorescent protein (GFP)-transgenic mice was transplanted. Coronary artery ligation was performed 10 weeks later. G-CSF (100 microg/kg) was daily injected for 6 days. Subpopulations of enhanced GFP(+) cells in peripheral blood, bone marrow, and heart were characterized by flow cytometry. Growth factor expression in the heart was analyzed by quantitative real-time polymerase chain reaction. Perfusion was investigated in vivo by gated single photon emission computed tomography (SPECT). RESULTS: G-CSF-treated animals revealed a reduced migration of c-kit(+) and CXCR-4(+) BMCs associated with decreased expression levels of the corresponding growth factors, namely stem cell factor and stromal-derived factor-1 alpha in ischemic myocardium. In contrast, the number of resident cardiac Sca-1(+) cells was significantly increased. However, SPECT-perfusion showed no differences in infarct size between G-CSF-treated and control animals 6 days after MI. CONCLUSION: Our study shows that G-CSF treatment after MI reduces migration capacity of BMCs into ischemic tissue, but increases the number of resident cardiac cells. To optimize homing capacity a combination of G-CSF with other agents may optimize cytokine therapy after MI.


Subject(s)
Bone Marrow Transplantation/methods , Granulocyte Colony-Stimulating Factor/therapeutic use , Myocardial Infarction/drug therapy , Myocardial Infarction/surgery , Animals , DNA Primers , Disease Models, Animal , Flow Cytometry , Genes, Reporter , Green Fluorescent Proteins/analysis , Green Fluorescent Proteins/genetics , Hematopoietic Stem Cell Mobilization , Mice , Mice, Inbred C57BL , Mice, Transgenic , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
8.
Cardiovasc Res ; 77(4): 722-31, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18055578

ABSTRACT

AIMS: An ongoing concept is that stem cells have the potential to regenerate the injured myocardium. In addition to direct vasorelaxing effects on the vasculature, which are mediated by an increased cAMP production leading to a decreased calcium influx in smooth muscle cells, parathyroid hormone (PTH) was recently shown to facilitate stem cell mobilization. Therefore, we analysed in a murine model of experimental myocardial infarction (MI) the influence of PTH treatment on survival, functional parameters, stem cell migration, and expression of vascular endothelial growth factor A (VEGF-A). METHODS AND RESULTS: Mice (C57BL/6) were treated with PTH (80 microg/kg/d) for up to 14 days after coronary artery ligation. Functional and immunohistochemical analyses were performed at days 6 and 30 after MI. Stem cells and VEGF expression in the myocardium were analysed by FACS and qRT-PCR at day 2 after MI. PTH-treated animals revealed a significant improvement of post-MI survival and myocardial function that was related to a subsequent reduction of left ventricular wall thinning and scar extension. Infarcted hearts of PTH-treated mice revealed increased numbers of CD45(+)/CD34(+) progenitor cells as well as an upregulation of VEGF-A mRNA associated with increased neovascularization and cell survival. CONCLUSIONS: PTH application after MI increases migration of angiogenic CD45(+)/CD34(+) progenitor cells to the ischaemic heart, which may attenuate ischaemic cardiomyopathy. As PTH is already used in patients with osteoporosis, our findings may have a direct impact on the initiation of clinical studies in patients with ischaemic heart disease.


Subject(s)
Cardiovascular Agents/pharmacology , Myocardial Infarction/drug therapy , Myocardium/pathology , Neovascularization, Physiologic/drug effects , Parathyroid Hormone/pharmacology , Stem Cells/drug effects , Animals , Antigens, CD34/analysis , Apoptosis/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Disease Models, Animal , Flow Cytometry , Immunohistochemistry , Leukocyte Common Antigens/analysis , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Paracrine Communication , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/immunology , Stem Cells/pathology , Time Factors , Up-Regulation , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL