Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters











Database
Language
Publication year range
1.
Circulation ; 126(21): 2491-501, 2012 Nov 20.
Article in English | MEDLINE | ID: mdl-23081990

ABSTRACT

BACKGROUND: Secretoneurin is a neuropeptide located in nerve fibers along blood vessels, is upregulated by hypoxia, and induces angiogenesis. We tested the hypothesis that secretoneurin gene therapy exerts beneficial effects in a rat model of myocardial infarction and evaluated the mechanism of action on coronary endothelial cells. METHODS AND RESULTS: In vivo secretoneurin improved left ventricular function, inhibited remodeling, and reduced scar formation. In the infarct border zone, secretoneurin induced coronary angiogenesis, as shown by increased density of capillaries and arteries. In vitro secretoneurin induced capillary tubes, stimulated proliferation, inhibited apoptosis, and activated Akt and extracellular signal-regulated kinase in coronary endothelial cells. Effects were abrogated by a vascular endothelial growth factor (VEGF) antibody, and secretoneurin stimulated VEGF receptors in these cells. Secretoneurin furthermore increased binding of VEGF to endothelial cells, and binding was blocked by heparinase, indicating that secretoneurin stimulates binding of VEGF to heparan sulfate proteoglycan binding sites. Additionally, secretoneurin increased binding of VEGF to its coreceptor neuropilin-1. In endothelial cells, secretoneurin also stimulated fibroblast growth factor receptor-3 and insulin-like growth factor-1 receptor, and in coronary vascular smooth muscle cells, we observed stimulation of VEGF receptor-1 and fibroblast growth factor receptor-3. Exposure of cardiac myocytes to hypoxia and ischemic heart after myocardial infarction revealed increased secretoneurin messenger RNA and protein. CONCLUSIONS: Our data show that secretoneurin acts as an endogenous stimulator of VEGF signaling in coronary endothelial cells by enhancing binding of VEGF to low-affinity binding sites and neuropilin-1 and stimulates further growth factor receptors like fibroblast growth factor receptor-3. Our in vivo findings indicate that secretoneurin may be a promising therapeutic tool in ischemic heart disease.


Subject(s)
Disease Models, Animal , Endothelium, Vascular/drug effects , Myocardial Infarction/drug therapy , Neovascularization, Physiologic/drug effects , Neuropeptides/administration & dosage , Secretogranin II/administration & dosage , Vascular Endothelial Growth Factor A/physiology , Animals , Coronary Vessels/drug effects , Coronary Vessels/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Genetic Therapy/methods , Humans , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Neovascularization, Physiologic/physiology , Neuropeptides/genetics , Plasmids/administration & dosage , Plasmids/genetics , RNA, Messenger/administration & dosage , RNA, Messenger/genetics , Rats , Secretogranin II/genetics , Signal Transduction/physiology
2.
Peptides ; 37(2): 252-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22884536

ABSTRACT

In this study, we investigated whether the proangiogenic neuropeptides secretoneurin (SN), substance P (SP), and neuropeptide Y (NPY) contribute to the development of abnormal neovascularization in the oxygen-induced retinopathy (OIR) model in mice. By exposing litters of C57Bl/6N mice to 75% oxygen from postnatal day 7 (P7) until postnatal day 11 (P11) and then returning them to normoxic conditions, retinal ischemia and subsequent neovascularization on the retinal surface were induced. Retinae were dissected on P9, P11, P12-P14, P16 and P20, and the concentrations of SN, SP, NPY and VEGF determined by radioimmunoassay or ELISA. The levels of SN and SP increased in controls from P9 until P16 and from P9 until P14, respectively, whereas the levels of NPY were high at P9 and decreased thereafter until P20, suggesting that NPY may participate in the development of the retina. However, dipeptidyl peptidase IV (DPPIV) and the NPY-Y2 receptor were not detectable in the immature retina indicating that NPY is not involved in the physiological vascularization in the retina. Compared to controls, OIR had no effect on the levels of SN, whereas levels of both SP and NPY slightly decreased during hyperoxia. Normalization of the levels of SP, and to a more pronounced extent of NPY, was significantly delayed during relative hypoxia. This clearly indicates that these three neuropeptides are not involved in the pathogenesis of neovascularization in OIR. Moreover, since there were no differences in the expression of two vessel markers in the retina of NPY knockout mice versus controls at P14, NPY is also not involved in the delayed development of the intermediate and deep vascular plexus in the retina in this animal model.


Subject(s)
Hyperoxia , Neuropeptide Y/analysis , Neuropeptide Y/metabolism , Neuropeptides/analysis , Retinal Neovascularization/metabolism , Retinal Neovascularization/pathology , Secretogranin II/analysis , Substance P/analysis , Animals , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Mice , Mice, Inbred C57BL , Mice, Knockout , Neuropeptide Y/deficiency , Radioimmunoassay , Retina/chemistry
3.
Circ Res ; 107(11): 1326-35, 2010 Nov 26.
Article in English | MEDLINE | ID: mdl-20930149

ABSTRACT

RATIONALE: The neuropeptide catestatin is an endogenous nicotinic cholinergic antagonist that acts as a pleiotropic hormone. OBJECTIVE: Catestatin shares several functions with angiogenic factors. We therefore reasoned that catestatin induces growth of new blood vessels. METHODS AND RESULTS: Catestatin induced migration, proliferation, and antiapoptosis in endothelial cells and exerted capillary tube formation in vitro in a Matrigel assay, and such effects were mediated via G protein, mitogen-activated protein kinase, and Akt. Catestatin-induced endothelial cell functions are further mediated by basic fibroblast growth factor, as shown by blockade of effects by a neutralizing fibroblast growth factor antibody. Furthermore, catestatin released basic fibroblast growth factor from endothelial cells and stimulated fibroblast growth factor signaling. In addition to its function on endothelial cells, catestatin also exerted effects on endothelial progenitor cells and vascular smooth muscle cells. In vivo, catestatin induced angiogenesis in the mouse cornea neovascularization assay and increased blood perfusion and number of capillaries in the hindlimb ischemia model. In addition to angiogenesis, catestatin increased density of arterioles/arteries and incorporation of endothelial progenitor cells in the hindlimb ischemia model, indicating induction of arteriogenesis and postnatal vasculogenesis. CONCLUSION: We conclude that catestatin acts as a novel angiogenic cytokine via a basic fibroblast growth factor-dependent mechanism.


Subject(s)
Angiogenic Proteins/physiology , Chromogranin A/physiology , Cytokines/physiology , Fibroblast Growth Factor 2/physiology , Neovascularization, Physiologic/physiology , Neuropeptides/physiology , Peptide Fragments/physiology , Animals , Cell Movement/physiology , Cells, Cultured , Endothelium, Vascular/physiology , Humans , Male , Mice , Mice, Inbred C57BL
4.
Circ Res ; 105(10): 994-1002, 2009 Nov 06.
Article in English | MEDLINE | ID: mdl-19797703

ABSTRACT

RATIONALE: The neuropeptide secretoneurin induces angiogenesis and postnatal vasculogenesis and is upregulated by hypoxia in skeletal muscle cells. OBJECTIVE: We sought to investigate the effects of secretoneurin on therapeutic angiogenesis. METHODS AND RESULTS: We generated a secretoneurin gene therapy vector. In the mouse hindlimb ischemia model secretoneurin gene therapy by intramuscular plasmid injection significantly increased secretoneurin content of injected muscles, improved functional parameters, reduced tissue necrosis, and restored blood perfusion. Increased muscular density of capillaries and arterioles/arteries demonstrates the capability of secretoneurin gene therapy to induce therapeutic angiogenesis and arteriogenesis. Furthermore, recruitment of endothelial progenitor cells was enhanced by secretoneurin gene therapy consistent with induction of postnatal vasculogenesis. Additionally, secretoneurin was able to activate nitric oxide synthase in endothelial cells and inhibition of nitric oxide inhibited secretoneurin-induced effects on chemotaxis and capillary tube formation in vitro. In vivo, secretoneurin induced nitric oxide production and inhibition of nitric oxide attenuated secretoneurin-induced effects on blood perfusion, angiogenesis, arteriogenesis, and vasculogenesis. Secretoneurin also induced upregulation of basic fibroblast growth factor and platelet-derived growth factor-B in endothelial cells. CONCLUSIONS: In summary, our data indicate that gene therapy with secretoneurin induces therapeutic angiogenesis, arteriogenesis, and vasculogenesis in the hindlimb ischemia model by a nitric oxide-dependent mechanism.


Subject(s)
Angiogenesis Inducing Agents/metabolism , Cytokines/biosynthesis , Genetic Therapy , Ischemia/therapy , Neovascularization, Physiologic , Neuropeptides/biosynthesis , Nitric Oxide/metabolism , Secretogranin II/biosynthesis , Animals , Cytokines/genetics , Disease Models, Animal , Endothelial Cells/metabolism , Hindlimb/blood supply , Hindlimb/metabolism , Humans , Ischemia/genetics , Ischemia/metabolism , Mice , Neuropeptides/genetics , Nitric Oxide Synthase Type III/biosynthesis , Secretogranin II/genetics , Stem Cells/metabolism
5.
Eur J Pharmacol ; 598(1-3): 104-11, 2008 Nov 19.
Article in English | MEDLINE | ID: mdl-18834877

ABSTRACT

Several members of the neuropeptide family exert chemotactic actions on blood monocytes consistent with neurogenic inflammation. Furthermore, chromogranin A (CgA) containing Alzheimer plaques are characterized by extensive microglia activation and such activation induces neuronal damage. We therefore hypothesized that the catecholamine release inhibitory peptide catestatin (hCgA(352-372)) would induce directed monocyte migration. We demonstrate that catestatin dose-dependently stimulates chemotaxis of human peripheral blood monocytes, exhibiting its maximal effect at a concentration of 1 nM comparable to the established chemoattractant formylated peptide Met-Leu-Phe (fMLP). The naturally occurring catestatin variants differed in their chemotactic property insofar as that the Pro370Leu variant was even more potent than wild type, whereas the Gly364Ser variant was less effective. Specificity of this effect was shown by inhibition of catestatin-induced chemotaxis by a specific neutralizing antibody. In addition, catestatin mediated effect was blocked by dimethylsphingosine and treatment with endothelial differentiation gene (Edg)-1 and Edg-3 antisense RNA as well as by incubation with pertussis toxin and genistein indicating involvement of tyrosine kinase receptor-, G-protein- and sphingosine-1-phosphate signaling. Catestatin also stimulated Akt- and extracellular signal related kinase (ERK)-phosphorylation and catestatin-induced chemotaxis was blocked by blockers of phosphoinositide-3 (PI-3) kinase and nitric oxide as well as by inhibition of the mitogen-activated protein kinases (MAPK) system indicating involvement of these signal transduction pathways. In summary, our data indicate that catestatin induces monocyte chemotaxis by activation of a variety of signal transduction pathways suggesting a role of this peptide as an inflammatory cytokine.


Subject(s)
Chromogranin A/pharmacology , Monocytes/physiology , Peptide Fragments/pharmacology , Signal Transduction/drug effects , Blotting, Western , Cell Movement/drug effects , Chemotaxis, Leukocyte/drug effects , Chromogranin A/genetics , Enzyme Activation/drug effects , Genistein/pharmacology , Humans , Neutrophil Infiltration/drug effects , Peptide Fragments/genetics , Pertussis Toxin/pharmacology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, G-Protein-Coupled/drug effects , Transfection
6.
FASEB J ; 21(11): 2906-17, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17504977

ABSTRACT

Expression of angiogenic cytokines like vascular endothelial growth factor is enhanced by hypoxia. We tested the hypothesis that decreased oxygen levels up-regulate the angiogenic factor secretoneurin. In vivo, muscle cells of mouse ischemic hind limbs showed increased secretoneurin expression, and inhibition of secretoneurin by a neutralizing antibody impaired the angiogenic response in this ischemia model. In a mouse soft tissue model of hypoxia, secretoneurin was increased in subcutaneous muscle fibers. In vitro, secretoneurin mRNA and protein were up-regulated in L6 myoblast cells after exposure to low oxygen levels. The hypoxia-dependent regulation of secretoneurin was tissue specific and was not observed in endothelial cells, vascular smooth muscle cells, or AtT20 pituitary tumor cells. The hypoxia-dependent induction of secretoneurin in L6 myoblasts is regulated by hypoxia-inducible factor-1alpha, since inhibition of this factor using si-RNA inhibited up-regulation of secretoneurin. Induction of secretoneurin by hypoxia was dependent on basic fibroblast growth factor in vivo and in vitro, and inhibition of this regulation by heparinase suggests an involvement of low-affinity basic fibroblast growth factor binding sites. In summary, our data show that the angiogenic cytokine secretoneurin is up-regulated by hypoxia in muscle cells by hypoxia-inducible factor-1alpha- and basic fibroblast growth factor-dependent mechanisms.


Subject(s)
Cell Hypoxia , Fibroblast Growth Factor 2/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myoblasts/metabolism , Neuropeptides/metabolism , Secretogranin II/metabolism , Signal Transduction , Animals , Blotting, Western , Cells, Cultured , DNA Primers/chemistry , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Extremities/surgery , Fluorescent Antibody Technique , Ischemia/metabolism , Ischemia/pathology , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/blood supply , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/metabolism , NAD/metabolism , Neovascularization, Physiologic , Pituitary Neoplasms/blood supply , Pituitary Neoplasms/metabolism , Pituitary Neoplasms/pathology , Polymerase Chain Reaction , Proprotein Convertases/metabolism , RNA, Small Interfering/pharmacology , Radioimmunoassay , Rats , Skin/metabolism , Transfection , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL