Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
Pediatr Res ; 73(4 Pt 1): 409-13, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23370410

ABSTRACT

BACKGROUND: Infantile hemangioma (IH) is the most frequent vascular tumor of early childhood. Recently, propranolol, a nonselective ß1- and ß2-adrenoceptor inhibitor, was introduced into the therapy of severe proliferating IH with excellent results. However, the underlying mechanism of action of propranolol is still unclear. METHODS: We performed immunohistochemistry for cluster of differentiation 31 (CD31), D2-40, glucose transporter-1 (GLUT-1), and Ki67 in order to characterize 21 vascular anomalies (nine IH, seven venous malformations (VMs), and five lymphatic malformations (LMs)). Furthermore, we analyzed the expression of ß1-, ß2-, and ß3-adrenoceptor mRNA in these specimens as well as in hemangioma-derived stem cells by quantitative real-time PCR (qPCR). RESULTS: We show that the expression of ß1-adrenoceptor mRNA is 10.7-fold higher in IH independent of the proliferative or regressive phase as well as 2.5-fold higher in hemangioma-derived stem cells as compared with ß2-adrenoceptor mRNA. In LM, the expression of ß2-adrenoceptor mRNA was ninefold higher than that of ß1-adrenoceptor mRNA. VM showed low expression levels of all ß-adrenoceptor mRNAs, and ß3-adrenoceptor mRNA was hardly detectable in any specimens examined. CONCLUSION: These results provide the first evidence of distinctions between IH and vascular malformations with regard to ß-adrenoceptor subtype mRNA levels.


Subject(s)
Hemangioma, Capillary/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Syndromes, Hereditary/genetics , RNA, Messenger/metabolism , Receptors, Adrenergic, beta-1/genetics , Vascular Malformations/genetics , Adolescent , Antibodies, Monoclonal, Murine-Derived , Cells, Cultured , Child , Child, Preschool , Genetic Markers , Glucose Transporter Type 1/metabolism , Hemangioma, Capillary/metabolism , Hemangioma, Capillary/pathology , Humans , Immunohistochemistry , Infant , Infant, Newborn , Ki-67 Antigen/metabolism , Neoplastic Stem Cells/pathology , Neoplastic Syndromes, Hereditary/metabolism , Neoplastic Syndromes, Hereditary/pathology , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-3/genetics , Reverse Transcriptase Polymerase Chain Reaction , Vascular Malformations/metabolism , Vascular Malformations/pathology
2.
J Biol Chem ; 285(44): 34213-9, 2010 Oct 29.
Article in English | MEDLINE | ID: mdl-20729197

ABSTRACT

α(2)-Adrenoceptors belong to the family of adrenergic receptors, which regulate the neuronal release of norepinephrine as part of a negative feedback loop. Among the α(2)-adrenoceptors, the α(2B)-subtype may also influence developmental signaling pathways involved in angiogenesis of the placenta. Thus, the aim of the present study was to determine whether α(2B)-adrenoceptors are also involved in other developmental processes beyond placental angiogenesis. Ablation of α(2B)-adrenoceptors led to lethality of mutant mice during the first hours after birth. Despite normal breathing and drinking behavior, mutant mice developed cyanosis, which could be traced back to a defect in lung morphology with significantly reduced alveolar volume and thickened interalveolar septi. In α(2B)-deficient lungs and in isolated alveolar type II cells, expression of sonic hedgehog (SHH) was significantly increased, resulting in mesenchymal proliferation. In vitro α(2B)-adrenoreceptor stimulation suppressed expression of sonic hedgehog and the cell cycle genes cyclin D1 and Ki67. In vivo inhibition of enhanced SHH signaling by the smoothened antagonist cyclopamine partially rescued perinatal lethality, lung morphology, and altered gene expression in mutant mice. Thus, α(2B)-adrenoceptors in lung epithelia play an important role in suppressing sonic hedgehog-mediated proliferation of mesenchymal cells and thus prevent respiratory failure.


Subject(s)
Hedgehog Proteins/metabolism , Lung Diseases/metabolism , Receptors, Adrenergic, alpha-2/metabolism , Animals , Cyclin D1/biosynthesis , Disease Models, Animal , Epithelium/metabolism , Heterozygote , Ki-67 Antigen/biosynthesis , Lung/metabolism , Lung/pathology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Neurotransmitter Agents/metabolism , Receptors, Adrenergic, alpha-2/deficiency , Veratrum Alkaloids/pharmacology
3.
Mol Pharmacol ; 75(5): 1160-70, 2009 May.
Article in English | MEDLINE | ID: mdl-19251826

ABSTRACT

Alpha(2)-adrenoceptors mediate diverse functions of the sympathetic system and are targets for the treatment of cardiovascular disease, depression, pain, glaucoma, and sympathetic activation during opioid withdrawal. To determine whether alpha(2)-adrenoceptors on adrenergic neurons or alpha(2)-adrenoceptors on nonadrenergic neurons mediate the physiological and pharmacological responses of alpha(2)-agonists, we used the dopamine beta-hydroxylase (Dbh) promoter to drive expression of alpha(2A)-adrenoceptors exclusively in noradrenergic and adrenergic cells of transgenic mice. Dbh-alpha(2A) transgenic mice were crossed with double knockout mice lacking both alpha(2A)- and alpha(2C)-receptors to generate lines with selective expression of alpha(2A)-autoreceptors in adrenergic cells. These mice were subjected to a comprehensive phenotype analysis and compared with wild-type mice, which express alpha(2A)- and alpha(2C)-receptors in both adrenergic and nonadrenergic cells, and alpha(2A)/alpha(2C) double-knockout mice, which do not express these receptors in any cell type. We were surprised to find that only a few functions previously ascribed to alpha(2)-adrenoceptors were mediated by receptors on adrenergic neurons, including feedback inhibition of norepinephrine release from sympathetic nerves and spontaneous locomotor activity. Other agonist effects, including analgesia, hypothermia, sedation, and anesthetic-sparing, were mediated by alpha(2)-receptors in nonadrenergic cells. In dopamine beta-hydroxylase knockout mice lacking norepinephrine, the alpha(2)-agonist medetomidine still induced a loss of the righting reflex, confirming that the sedative effect of alpha(2)-adrenoceptor stimulation is not mediated via autoreceptor-mediated inhibition of norepinephrine release. The present study paves the way for a revision of the current view of the alpha(2)-adrenergic receptors, and it provides important new considerations for future drug development.


Subject(s)
Receptors, Adrenergic, alpha-2/physiology , Adrenergic alpha-Agonists/pharmacology , Analgesics/pharmacology , Anesthetics/pharmacology , Animals , Body Temperature/drug effects , Hypnotics and Sedatives/pharmacology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Motor Activity/drug effects , Norepinephrine/metabolism , Receptors, Adrenergic, alpha-2/genetics , Transgenes
4.
Circ Res ; 101(7): 682-91, 2007 Sep 28.
Article in English | MEDLINE | ID: mdl-17673674

ABSTRACT

Alpha2-adrenoceptors are essential presynaptic regulators of norepinephrine release from sympathetic nerves. Previous studies in mice with targeted deletions in the 3 alpha2-adrenoceptor genes have indicated that these receptors are essential for embryonic development. In the present study, we searched for the alpha2-adrenoceptor subtype(s) involved in placental development and its molecular mechanism using mice carrying targeted deletions in alpha2-adrenoceptor genes. Congenic alpha2B-adrenoceptor-deficient mice (Adra2b-/-) developed a defect in fetal and maternal vessel formation in the placenta labyrinth at embryonic day 10.5. This defect was accompanied by reduced endothelial cell proliferation and decreased extracellular signal-regulated kinase 1/2 phosphorylation levels in Adra2b-/- as compared with Adra2b+/+ placentae. Microarray analysis of wild-type and mutant placentae (maternal genotype Adra2b+/-) revealed 179 genes, which were significantly up- or downregulated >1.5-fold in alpha2B-deficient placentae. The type 1 receptor for vascular endothelial growth factor (Flt1), which is coexpressed with alpha2B-adrenoceptors in spongiotrophoblast and giant cells of the placenta, was found to be 2.3-fold upregulated in alpha2B-deficient placentae. Neutralization of Flt1 and its soluble splice variant sFlt1 by a specific antibody in vivo prevented the vascular defect in alpha2B-deficient placentae at embryonic day 10.5. Thus, alpha2B-adrenoceptors are essential to suppress antiangiogenic (s)Flt1 in spongiotrophoblasts to control the coordinated formation of a vascular labyrinth of fetal and maternal blood vessels in the murine placenta during development.


Subject(s)
Neovascularization, Pathologic/metabolism , Placenta/blood supply , Placenta/metabolism , Receptors, Adrenergic, alpha-2/deficiency , Receptors, Adrenergic, alpha-2/metabolism , Up-Regulation/physiology , Vascular Endothelial Growth Factor Receptor-1/biosynthesis , Animals , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Receptors, Adrenergic, alpha-2/genetics , Solubility , Vascular Endothelial Growth Factor Receptor-1/genetics , Vascular Endothelial Growth Factor Receptor-1/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...