Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Anal Chem ; 87(17): 8858-66, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26260548

ABSTRACT

Swiftness, reliability, and sensitivity of live bacteria detection in drinking water are key issues for human safety. The most widespread used indicator of live bacteria is a caged form of carboxyfluorescein in which 3' and 6' hydroxyl groups are masked as acetate esters (CFDA). This derivatization altogether abolishes fluorescein fluorescence and renders the molecule prone to passive diffusion through bacterial membranes. Once in the cytoplasm, acetate groups from CFDA are removed by bacterial hydrolases and fluorescence develops, rendering live but not dead cells detectable. Yet the reagent, carboxyfluorescein diacetate, still possesses a free carboxyl group whose ionization constant is such that the majority of the probe is charged at physiological pH. This unfavors probe permeation through membranes. Here, we prepare several chemical modifications of the carboxyl moiety of CFDA, in order to neutralize its charge and improve its passive diffusion through membranes. We show that the ethylamido derivative of the 5-carboxyl group from 5-carboxy-fluorescein diacetate or from Oregon green diacetate or from Oregon green diacetoxymethylester are stable molecules in biological media, penetrate into bacterial cells and are metabolized into fluorescent species. Only live bacteria are revealed since bleached samples are not labeled. Other derivatives with modification of the 5-carboxyl group with an ester group or with a thiourea-based moiety were almost inefficient probes. The most interesting probe, triembarine (5-ethylaminocarboxy-oregon green, 3',6'diacetoxymethyl ester) leads to 6-10 times more sensitive detection of bacteria as compared to CFDA. Addition of contrast agents (trypan blue or brilliant blue R) improve the signal-to-noise ratio by quenching extracellular fluorescence while bromophenol blue quenches both intracellular and extracellular fluorescence, allowing standardization of detections.


Subject(s)
Bacteria/isolation & purification , Fluoresceins/chemistry , Fluorescent Dyes/chemistry , Microbial Viability , Bacillus subtilis/isolation & purification , Chryseobacterium/isolation & purification , Enterobacter cloacae/isolation & purification , Escherichia coli/isolation & purification , Pseudomonas aeruginosa/isolation & purification
2.
Am J Physiol Heart Circ Physiol ; 298(3): H844-52, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20023120

ABSTRACT

Prokineticins are secreted peptides that activate two G protein-coupled receptors: PKR1 and PKR2. Prokineticins induce angiogenesis and fenestration, but the cognate receptors involved in these functions are unknown. We hypothesized a role for prokineticin receptor signaling pathways and expression profiles in determining the selective effects of prokineticins on coronary endothelial cells (H5V). Activation of the PKR1/MAPK/Akt signaling pathway stimulates proliferation, migration, and angiogenesis in H5V cells, in which PKR1 predominates over PKR2. PKR1 was colocalized with Galpha(11) and was internalized following the stimulation of these cells with prokineticin-2. Knock down of PKR1 or Galpha(11) expression in H5V cells effectively inhibited prokineticin-2-induced vessel formation and MAPK/Akt activation, indicating a role for PKR1/Galpha(11) in this process. However, in conditions in which PKR2 predominated over PKR1, these cells displayed a fenestrated endothelial cell phenotype. H5V cells overexpressing PKR2 displayed large numbers of multivesicular bodies and caveolar clusters and a disruption of the distribution of zonula occluden-1 tight junction protein. Prokineticin-2 induced the colocalization of PKR2 with Galpha(12), and activated Galpha(12), which bound to zonula occluden-1 to trigger the degradation of this protein in these cells. Prokineticin-2 induced the formation of vessel-like structures by human aortic endothelial cells expressing only PKR1, and disorganized the tight junctions in human hepatic sinusoidal endothelial cells expressing only PKR2, confirming the divergent roles of these receptors. Our findings show the functional characteristics of coronary endothelial cells depend on the expression of PKR1 and PKR2 levels and the divergent signaling pathways used by these receptors.


Subject(s)
Endothelium, Vascular/physiology , Neovascularization, Physiologic/physiology , Receptors, G-Protein-Coupled/physiology , Receptors, Peptide/physiology , Animals , Cell Line , Cell Movement , Cell Proliferation , Cells, Cultured , Endothelium, Vascular/cytology , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Humans , Membrane Proteins/metabolism , Mice , Mitogen-Activated Protein Kinase Kinases/physiology , Models, Animal , Phosphoproteins/metabolism , Proto-Oncogene Proteins c-akt/physiology , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Signal Transduction/physiology , Zonula Occludens-1 Protein
3.
Cardiovasc Res ; 81(1): 28-37, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-18806277

ABSTRACT

AIMS: Prokineticins are small secreted bioactive molecules. They exert their biological activity by binding to two G protein-coupled receptors. Previously, we have shown that the overexpression of prokineticin receptor-1 (PKR1) in transgenic (TG) mouse hearts induced neovascularization. Since PKR1 and PKR2 are 85% identical and expressed in cardiovascular tissues, we hypothesized that PKR2 may also contribute to cardiomyocyte growth and vascularization. METHODS AND RESULTS: We have generated TG mice overexpressing PKR2 in cardiomyocytes. TG mice exhibit increased hypertrophic gene expression and heart-to-body weight ratio accompanied by an increased length of cardiomyocytes at the age of 12 weeks. Increased left ventricular end-systolic and diastolic diameters without cardiac dysfunction at the age of 24 weeks indicate that TG mice have an eccentric hypertrophy with compensated cardiac function. Quantitative morphological analysis showed that TG hearts have a normal microvessel density and number of branch points. However, they exhibit increased abnormal endothelial cell shape and ultrastructure, changed cellular distribution of a tight junction protein zona occludens-1 (ZO-1), and vascular leakage in heart without a rise of angiogenic factor levels at early and late age. The application of media conditioned by H9c2 cardioblast cells overexpressing PKR2 significantly induced impaired ZO-1 localization in H5V endothelial cells, mimicking the TG model. CONCLUSION: These findings provide the first genetic evidence that cardiomyocyte PKR2 signalling leads to eccentric hypertrophy in an autocrine regulation and impaired endothelial integrity in a paracrine regulation without inducing angiogenesis. These TG mice may provide a new genetic model for heart diseases.


Subject(s)
Capillary Leak Syndrome/etiology , Cardiomegaly/etiology , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Arterioles/pathology , Arterioles/physiopathology , Blood Pressure/physiology , Capillary Leak Syndrome/metabolism , Capillary Leak Syndrome/pathology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cell Membrane Permeability/physiology , Cells, Cultured , Coronary Vessels/pathology , Coronary Vessels/physiopathology , Disease Models, Animal , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Heart Rate/physiology , Mice , Mice, Transgenic , Myocardium/pathology , Myocytes, Cardiac/pathology , Myocytes, Cardiac/ultrastructure , Receptors, G-Protein-Coupled/genetics , Signal Transduction/physiology
4.
Arterioscler Thromb Vasc Biol ; 28(5): 841-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18421008

ABSTRACT

OBJECTIVE: Identification of novel factors that contribute to myocardial repair and collateral vessel growth hold promise for treatment of heart diseases. We have shown that transient prokineticin receptor-1 (PKR1) gene transfer protects the heart against myocardial infarction in a mouse model. Here, we investigated the role of excessive PKR1 signaling in heart. METHODS AND RESULTS: Transgenic mice overexpressing PKR1 in cardiomyocytes displayed no spontaneous abnormalities in cardiomyocytes but showed an increased number of epicardial-derived progenitor cells (EPDCs), capillary density, and coronary arterioles. Coculturing EPDCs with H9c2 cardiomyoblasts overexpressing PKR1 promotes EPDC differentiation into endothelial and smooth muscle cells, mimicking our transgenic model. Overexpressing PKR1 in H9c2 cardiomyoblasts or in transgenic hearts upregulated prokineticin-2 levels. Exogenous prokineticin-2 induces significant outgrowth from neonatal and adult epicardial explants, promoting EPDC differentiation. These prokineticin-2 effects were abolished in cardiac explants from mice with PKR1-null mutation. Reduced capillary density and prokineticin-2 levels in PKR1-null mutant hearts supports the hypothesis of an autocrine/paracrine loop between PKR1 and prokineticin-2. CONCLUSIONS: Cardiomyocyte-PKR1 signaling upregulates its own ligand prokineticin-2 that acts as a paracrine factor, triggering EPDCs proliferation/differentiation. This study provides a novel insight for possible therapeutic strategies aiming at restoring pluripotency of adult EPDCs to promote neovasculogenesis by induction of cardiomyocyte PKR1 signaling.


Subject(s)
Cell Differentiation/physiology , Neovascularization, Physiologic/physiology , Pericardium/pathology , Protein Kinase C/metabolism , Stem Cells/pathology , Animals , Cell Movement/physiology , Cell Proliferation , Coculture Techniques , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Gastrointestinal Hormones/genetics , Gastrointestinal Hormones/metabolism , Mice , Mice, Transgenic , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Neuropeptides/genetics , Neuropeptides/metabolism , Pericardium/metabolism , Protein Kinase C/genetics , Signal Transduction/genetics , Signal Transduction/physiology , Stem Cells/metabolism , Up-Regulation
5.
FASEB J ; 21(11): 2980-93, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17442730

ABSTRACT

Prokineticins are potent angiogenic factors that bind to two G protein-coupled receptors to initiate their biological effects. We hypothesize that prokineticin receptor-1 (PKR1/GPR73) signaling may contribute to cardiomyocyte survival or repair in myocardial infarction. Since we showed that prokineticin-2 and PKR1 are expressed in adult mouse heart and cardiac cells, we investigated the role of prokineticin-2 on capillary endothelial cell and cardiomyocyte function. In cultured cardiac endothelial cells, prokineticin-2 or overexpression of PKR1 induces vessel-like formation without increasing VEGF levels. In cardiomyocytes and H9c2 cells, prokineticin-2 or overexpressing PKR1 activates Akt to protect cardiomyocytes against oxidative stress. The survival and angiogenesis promoting effects of prokineticin-2 in cardiac cells were completely reversed by siRNA-PKR1, indicating PKR1 involvement. We thus, further investigated whether intramyocardial gene transfer of DNA encoding PKR1 may rescue the myocardium against myocardial infarction in mouse model. Transient PKR1 gene transfer after coronary ligation reduces mortality and preserves left ventricular function by promoting neovascularization and protecting cardiomyocytes without altering VEGF levels. In human end-stage failing heart samples, reduced PKR1 and prokineticin-2 transcripts and protein levels implicate a more important role for prokineticin-2/PKR1 signaling in heart. Our results suggest that PKR1 may represent a novel therapeutic target to limit myocardial injury following ischemic events.


Subject(s)
Heart/physiology , Myocardial Ischemia/prevention & control , Myocytes, Cardiac/metabolism , Neovascularization, Pathologic , Receptors, G-Protein-Coupled/metabolism , Vascular Endothelial Growth Factor A/metabolism , Adenoviridae/genetics , Animals , Apoptosis , Cell Hypoxia , Cells, Cultured , Embryo, Mammalian , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Genetic Therapy , In Situ Hybridization , Male , Mice , Mice, Inbred C57BL , Myocardial Infarction/metabolism , Myocardial Infarction/prevention & control , Myocardial Ischemia/metabolism , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/cytology , Proto-Oncogene Proteins c-akt/metabolism , RNA Probes , RNA, Small Interfering/pharmacology , Rats , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, Peptide/genetics , Receptors, Peptide/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...